1
|
Díaz-Santiago E, Moya-García AA, Pérez-García J, Yahyaoui R, Orengo C, Pazos F, Perkins JR, Ranea JAG. Better understanding the phenotypic effects of drugs through shared targets in genetic disease networks. Front Pharmacol 2025; 15:1470931. [PMID: 39911831 PMCID: PMC11794328 DOI: 10.3389/fphar.2024.1470931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/26/2024] [Accepted: 12/12/2024] [Indexed: 02/07/2025] Open
Abstract
Introduction Most drugs fail during development and there is a clear and unmet need for approaches to better understand mechanistically how drugs exert both their intended and adverse effects. Gaining traction in this field is the use of disease data linking genes with pathological phenotypes and combining this with drugtarget interaction data. Methods We introduce methodology to associate drugs with effects, both intended and adverse, using a tripartite network approach that combines drug-target and target-phenotype data, in which targets can be represented as proteins and protein domains. Results We were able to detect associations for over 140,000 ChEMBL drugs and 3,800 phenotypes, represented as Human Phenotype Ontology (HPO) terms. The overlap of these results with the SIDER databases of known drug side effects was up to 10 times higher than random, depending on the target type, disease database and score threshold used. In terms of overlap with drug-phenotype pairs extracted from the literature, the performance of our methodology was up to 17.47 times greater than random. The top results include phenotype-drug associations that represent intended effects, particularly for cancers such as chronic myelogenous leukemia, which was linked with nilotinib. They also include adverse side effects, such as blurred vision being linked with tetracaine. Discussion This work represents an important advance in our understanding of how drugs cause intended and adverse side effects through their action on disease causing genes and has potential applications for drug development and repositioning.
Collapse
Affiliation(s)
- Elena Díaz-Santiago
- Department of Molecular Biology and Biochemistry, University of Malaga, Malaga, Spain
| | | | - Jesús Pérez-García
- Department of Molecular Biology and Biochemistry, University of Malaga, Malaga, Spain
| | - Raquel Yahyaoui
- Laboratory of Inherited Metabolic Diseases and Newborn Screening, Malaga Regional University Hospital, Malaga, Spain
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Malaga, Spain
| | - Christine Orengo
- Department of Structural and Molecular Biology, University College London, London, United Kingdom
| | - Florencio Pazos
- Computational Systems Biology Group, Systems Biology Department, National Centre for Biotechnology (CNB-CSIC), Madrid, Spain
| | - James R. Perkins
- Department of Molecular Biology and Biochemistry, University of Malaga, Malaga, Spain
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Malaga, Spain
- CIBER de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain
| | - Juan A. G. Ranea
- Department of Molecular Biology and Biochemistry, University of Malaga, Malaga, Spain
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Malaga, Spain
- CIBER de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain
- Spanish National Bioinformatics Institute (INB/ELIXIR-ES), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| |
Collapse
|
2
|
Attri M, Raghav A, Sinha J. Revolutionising Neurological Therapeutics: Investigating Drug Repurposing Strategies. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2025; 24:115-131. [PMID: 39323347 DOI: 10.2174/0118715273329531240911075309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 05/09/2024] [Revised: 07/08/2024] [Accepted: 07/15/2024] [Indexed: 09/27/2024]
Abstract
Repurposing drugs (DR) has become a viable approach to hasten the search for cures for neurodegenerative diseases (NDs). This review examines different off-target and on-target drug discovery techniques and how they might be used to find possible treatments for non-diagnostic depressions. Off-target strategies look at the known or unknown side effects of currently approved drugs for repositioning, whereas on-target strategies connect disease pathways to targets that can be treated with drugs. The review highlights the potential of experimental and computational methodologies, such as machine learning, proteomic techniques, network and genomics-based approaches, and in silico screening, in uncovering new drug-disease correlations. It also looks at difficulties and failed attempts at drug repurposing for NDs, highlighting the necessity of exact and standardised procedures to increase success rates. This review's objectives are to address the purpose of drug repurposing in human disorders, particularly neurological diseases, and to provide an overview of repurposing candidates that are presently undergoing clinical trials for neurological conditions, along with any possible causes and early findings. We then include a list of drug repurposing strategies, restrictions, and difficulties for upcoming research.
Collapse
Affiliation(s)
- Meenakshi Attri
- School of Medical & Allied Sciences, K.R. Mangalam University, Gurugram, Haryana 122103, India
| | - Asha Raghav
- Department of Pharmaceutics, School of Health Sciences, Sushant University, Gurugram, Haryana 122003, India
| | - Jyoti Sinha
- Department of Pharmaceutics, School of Health Sciences, Sushant University, Gurugram, Haryana 122003, India
| |
Collapse
|
3
|
Verma C, Jain K, Saini A, Mani I, Singh V. Exploring the potential of drug repurposing for treating depression. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2024; 207:79-105. [PMID: 38942546 DOI: 10.1016/bs.pmbts.2024.03.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 06/30/2024]
Abstract
Researchers are interested in drug repurposing or drug repositioning of existing pharmaceuticals because of rising costs and slower rates of new medication development. Other investigations that authorized these treatments used data from experimental research and off-label drug use. More research into the causes of depression could lead to more effective pharmaceutical repurposing efforts. In addition to the loss of neurotransmitters like serotonin and adrenaline, inflammation, inadequate blood flow, and neurotoxins are now thought to be plausible mechanisms. Because of these other mechanisms, repurposing drugs has resulted for treatment-resistant depression. This chapter focuses on therapeutic alternatives and their effectiveness in drug repositioning. Atypical antipsychotics, central nervous system stimulants, and neurotransmitter antagonists have investigated for possible repurposing. Nonetheless, extensive research is required to ensure their formulation, effectiveness, and regulatory compliance.
Collapse
Affiliation(s)
- Chaitenya Verma
- Department of Pathology, Ohio State University, Columbus, OH, United States
| | - Kritika Jain
- Department of Microbiology, Institute of Home Economics, University of Delhi, New Delhi, India
| | - Ashok Saini
- Department of Microbiology, Institute of Home Economics, University of Delhi, New Delhi, India
| | - Indra Mani
- Department of Microbiology, Gargi College, University of Delhi, New Delhi, India.
| | - Vijai Singh
- Department of Biosciences, School of Science, Indrashil University, Rajpur, Mehsana, India.
| |
Collapse
|
4
|
Waseem T, Rajput TA, Mushtaq MS, Babar MM, Rajadas J. Computational biology approaches for drug repurposing. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2024; 205:91-109. [PMID: 38789189 DOI: 10.1016/bs.pmbts.2024.03.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 05/26/2024]
Abstract
The drug discovery and development (DDD) process greatly relies on the data available in various forms to generate hypotheses for novel drug design. The complex and heterogeneous nature of biological data makes it difficult to utilize or gather meaningful information as such. Computational biology techniques have provided us with opportunities to better understand biological systems through refining and organizing large amounts of data into actionable and systematic purviews. The drug repurposing approach has been utilized to overcome the expansive time periods and costs associated with traditional drug development. It deals with discovering new uses of already approved drugs that have an established safety and efficacy profile, thereby, requiring them to go through fewer development phases. Thus, drug repurposing through computational biology provides a systematic approach to drug development and overcomes the constraints of traditional processes. The current chapter covers the basics, approaches and tools of computational biology that can be employed to effectively develop repurposing profile of already approved drug molecules.
Collapse
Affiliation(s)
- Tanya Waseem
- Shifa College of Pharmaceutical Sciences, Shifa Tameer-e-Millat University, Islamabad, Pakistan
| | - Tausif Ahmed Rajput
- Shifa College of Pharmaceutical Sciences, Shifa Tameer-e-Millat University, Islamabad, Pakistan
| | | | - Mustafeez Mujtaba Babar
- Shifa College of Pharmaceutical Sciences, Shifa Tameer-e-Millat University, Islamabad, Pakistan; Advanced Drug Delivery and Regenerative Biomaterials Laboratory, Cardiovascular Institute and Pulmonary and Critical Care Medicine, Stanford University School of Medicine, Stanford University, Palo Alto, CA, United States.
| | - Jayakumar Rajadas
- Advanced Drug Delivery and Regenerative Biomaterials Laboratory, Cardiovascular Institute and Pulmonary and Critical Care Medicine, Stanford University School of Medicine, Stanford University, Palo Alto, CA, United States
| |
Collapse
|
5
|
Huang MS, Han JC, Lin PY, You YT, Tsai RTH, Hsu WL. Surveying biomedical relation extraction: a critical examination of current datasets and the proposal of a new resource. Brief Bioinform 2024; 25:bbae132. [PMID: 38609331 PMCID: PMC11014787 DOI: 10.1093/bib/bbae132] [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] [Academic Contribution Register] [Received: 06/16/2023] [Revised: 11/06/2023] [Accepted: 03/02/2023] [Indexed: 04/14/2024] Open
Abstract
Natural language processing (NLP) has become an essential technique in various fields, offering a wide range of possibilities for analyzing data and developing diverse NLP tasks. In the biomedical domain, understanding the complex relationships between compounds and proteins is critical, especially in the context of signal transduction and biochemical pathways. Among these relationships, protein-protein interactions (PPIs) are of particular interest, given their potential to trigger a variety of biological reactions. To improve the ability to predict PPI events, we propose the protein event detection dataset (PEDD), which comprises 6823 abstracts, 39 488 sentences and 182 937 gene pairs. Our PEDD dataset has been utilized in the AI CUP Biomedical Paper Analysis competition, where systems are challenged to predict 12 different relation types. In this paper, we review the state-of-the-art relation extraction research and provide an overview of the PEDD's compilation process. Furthermore, we present the results of the PPI extraction competition and evaluate several language models' performances on the PEDD. This paper's outcomes will provide a valuable roadmap for future studies on protein event detection in NLP. By addressing this critical challenge, we hope to enable breakthroughs in drug discovery and enhance our understanding of the molecular mechanisms underlying various diseases.
Collapse
Affiliation(s)
- Ming-Siang Huang
- Intelligent Agent Systems Laboratory, Department of Computer Science and Information Engineering, Asia University, New Taipei City, Taiwan
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
- Department of Computer Science and Information Engineering, College of Information and Electrical Engineering, Asia University, Taichung, Taiwan
| | - Jen-Chieh Han
- Intelligent Information Service Research Laboratory, Department of Computer Science and Information Engineering, National Central University, Taoyuan, Taiwan
| | - Pei-Yen Lin
- Intelligent Agent Systems Laboratory, Department of Computer Science and Information Engineering, Asia University, New Taipei City, Taiwan
| | - Yu-Ting You
- Intelligent Agent Systems Laboratory, Department of Computer Science and Information Engineering, Asia University, New Taipei City, Taiwan
| | - Richard Tzong-Han Tsai
- Intelligent Information Service Research Laboratory, Department of Computer Science and Information Engineering, National Central University, Taoyuan, Taiwan
- Center for Geographic Information Science, Research Center for Humanities and Social Sciences, Academia Sinica, Taipei, Taiwan
| | - Wen-Lian Hsu
- Intelligent Agent Systems Laboratory, Department of Computer Science and Information Engineering, Asia University, New Taipei City, Taiwan
- Department of Computer Science and Information Engineering, College of Information and Electrical Engineering, Asia University, Taichung, Taiwan
| |
Collapse
|
6
|
Luo H, Zhu C, Wang J, Zhang G, Luo J, Yan C. Prediction of drug-disease associations based on reinforcement symmetric metric learning and graph convolution network. Front Pharmacol 2024; 15:1337764. [PMID: 38384286 PMCID: PMC10879308 DOI: 10.3389/fphar.2024.1337764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/13/2023] [Accepted: 01/18/2024] [Indexed: 02/23/2024] Open
Abstract
Accurately identifying novel indications for drugs is crucial in drug research and discovery. Traditional drug discovery is costly and time-consuming. Computational drug repositioning can provide an effective strategy for discovering potential drug-disease associations. However, the known experimentally verified drug-disease associations is relatively sparse, which may affect the prediction performance of the computational drug repositioning methods. Moreover, while the existing drug-disease prediction method based on metric learning algorithm has achieved better performance, it simply learns features of drugs and diseases only from the drug-centered perspective, and cannot comprehensively model the latent features of drugs and diseases. In this study, we propose a novel drug repositioning method named RSML-GCN, which applies graph convolutional network and reinforcement symmetric metric learning to predict potential drug-disease associations. RSML-GCN first constructs a drug-disease heterogeneous network by integrating the association and feature information of drugs and diseases. Then, the graph convolutional network (GCN) is applied to complement the drug-disease association information. Finally, reinforcement symmetric metric learning with adaptive margin is designed to learn the latent vector representation of drugs and diseases. Based on the learned latent vector representation, the novel drug-disease associations can be identified by the metric function. Comprehensive experiments on benchmark datasets demonstrated the superior prediction performance of RSML-GCN for drug repositioning.
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
| | - Chunli Zhu
- School of Computer and Information Engineering, Henan University, Kaifeng, China
- Henan Key Laboratory of Big Data Analysis and Processing, Henan University, Kaifeng, China
| | - Jianlin Wang
- School of Computer and Information Engineering, Henan University, Kaifeng, China
- Henan Key Laboratory of Big Data Analysis and Processing, Henan University, Kaifeng, 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
| | - Junwei Luo
- School of Computer and Information Engineering, Henan University, Kaifeng, China
| | - Chaokun Yan
- School of Computer and Information Engineering, Henan University, Kaifeng, China
- Henan Key Laboratory of Big Data Analysis and Processing, Henan University, Kaifeng, China
| |
Collapse
|
7
|
Kumar P, Sheokand D, Grewal A, Saini V, Kumar A. Clinical side-effects based drug repositioning for anti-epileptic activity. J Biomol Struct Dyn 2024; 42:1443-1454. [PMID: 37042987 DOI: 10.1080/07391102.2023.2199874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/06/2022] [Accepted: 04/01/2023] [Indexed: 04/13/2023]
Abstract
Several generations of anti-epileptic drugs (AEDs) are available but have several associated side effects apart from a limited success rate. Drug repositioning strategies have gained importance in the last two decades owing to lower failure rates and economic burden. Drugs with similar side effect profiles may share a common mechanism of action and thus can be linked to other disease treatments. The present study was carried out to identify the newly approved drug candidate(s) as AEDs using clinical side-effects drug repositioning strategy. The clinical side effect similarity of drugs available in the SIDER v4.1 database was estimated against common side effects of 5 major marketed AEDs, using the 'dplyr' package library in the R. Further drugs were filtered based on Blood Brain Barrier permeability prediction and FDA-approval status. Molecular docking studies were performed for selected 26 hits (drugs) against previously identified epilepsy target receptors: Voltage-gated sodium channel α2 (Nav1.2), GABA receptor α1-β1 (GABAr α1-β1), and Voltage-gated calcium channel α-1 G (Cav3.1). Only 2 drugs (Ziprasidone and Paroxetine) showed better binding affinities against studied epilepsy receptors Nav1.2, GABAr α1-β1, and Cav3.1, than their corresponding standard AEDs, i.e. Carbamazepine, Clonazepam, and Pregabalin, respectively. Ziprasidone reportedly showed seizure-like symptoms in ∼3% of patients and was hence omitted from further study. The MDS study of docked complexes of Paroxetine with selected epilepsy target receptors showed stable RMSD values and better interaction energies. The study reveals Paroxetine as a potential candidate to be repurposed for 1st line epileptic seizure medication.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Pawan Kumar
- Toxicology and Computational Biology Group, Centre for Bioinformatics, Maharshi Dayanand University, Rohtak, India
| | - Deepak Sheokand
- Toxicology and Computational Biology Group, Centre for Bioinformatics, Maharshi Dayanand University, Rohtak, India
| | - Annu Grewal
- Toxicology and Computational Biology Group, Centre for Bioinformatics, Maharshi Dayanand University, Rohtak, India
| | - Vandana Saini
- Toxicology and Computational Biology Group, Centre for Bioinformatics, Maharshi Dayanand University, Rohtak, India
| | - Ajit Kumar
- Toxicology and Computational Biology Group, Centre for Bioinformatics, Maharshi Dayanand University, Rohtak, India
| |
Collapse
|
8
|
Liu Y, Sang G, Liu Z, Pan Y, Cheng J, Zhang Y. MPTN: A message-passing transformer network for drug repurposing from knowledge graph. Comput Biol Med 2024; 168:107800. [PMID: 38043469 DOI: 10.1016/j.compbiomed.2023.107800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/15/2023] [Revised: 11/09/2023] [Accepted: 11/29/2023] [Indexed: 12/05/2023]
Abstract
Drug repurposing (DR) based on knowledge graphs (KGs) is challenging, which uses knowledge graph reasoning models to predict new therapeutic pathways for existing drugs. With the rapid development of computing technology and the growing availability of validated biomedical data, various knowledge graph-based methods have been widely used to analyze and process complex and novel data to discover new indications for given drugs. However, existing methods need to be improved in extracting semantic information from contextual triples of biomedical entities. In this study, we propose a message-passing transformer network named MPTN based on knowledge graph for drug repurposing. Firstly, CompGCN is used as precoder to jointly aggregate entity and relation embeddings. Then, to fully capture the semantic information of entity context triples, the message propagating transformer module is designed. The module integrates the transformer into the message passing mechanism and incorporates the attention weight information of computing entity context triples into the entity embedding to update the entity embedding. Next, the residual connection is introduced to retain information as much as possible and improve prediction accuracy. Finally, MPTN utilizes the InteractE module as the decoder to obtain heterogeneous feature interactions in entity and relation representations and predict new pathways for drug treatment. Experiments on two datasets show that the model is superior to the existing knowledge graph embedding (KGE) learning methods.
Collapse
Affiliation(s)
- Yuanxin Liu
- School of Information Science and Technology, Dalian Maritime University, Dalian, 116026, Liaoning, China
| | - Guoming Sang
- School of Information Science and Technology, Dalian Maritime University, Dalian, 116026, Liaoning, China
| | - Zhi Liu
- School of Information Science and Technology, Dalian Maritime University, Dalian, 116026, Liaoning, China
| | - Yilin Pan
- School of Artificial Intelligence, Dalian Maritime University, Dalian, 116026, Liaoning, China
| | - Junkai Cheng
- School of Information Science and Technology, Dalian Maritime University, Dalian, 116026, Liaoning, China
| | - Yijia Zhang
- School of Information Science and Technology, Dalian Maritime University, Dalian, 116026, Liaoning, China.
| |
Collapse
|
9
|
Chakkittukandiyil A, Chakraborty S, Kothandan R, Rymbai E, Muthu SK, Vasu S, Sajini DV, Sugumar D, Mohammad ZB, Jayaram S, Rajagopal K, Ramachandran V, Selvaraj D. Side effects based network construction and drug repositioning of ropinirole as a potential molecule for Alzheimer's disease: an in-silico, in-vitro, and in-vivo study. J Biomol Struct Dyn 2023; 42:10785-10799. [PMID: 37723871 DOI: 10.1080/07391102.2023.2258968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/22/2022] [Accepted: 09/08/2023] [Indexed: 09/20/2023]
Abstract
Alzheimer's disease (AD) is the leading cause of dementia in older adults. Drug repositioning is a process of finding new therapeutic applications for existing drugs. One of the methods in drug repositioning is to use the side-effect profile of a drug to identify a new therapeutic indication. The drugs with similar side-effects may act on similar biological targets and could affect the same biochemical process. In this study, we explored the Food and Drug Administration-approved drugs using PROMISCUOUS database to find those that have adverse effects profile comparable with the ligands being studied or used to treat AD. Here, we found that the ropinirole, a dopamine receptor agonist, shared a maximum number of side-effects with the drugs proven beneficial for treating AD. Furthermore, molecular modelling demonstrated that ropinirole exhibited strong binding affinity (-9.313 kcal/mol) and best ligand efficiency (0.49) with sigma-1 receptor. Here, we observed that the quaternary amino group of ropinirole is essential for binding with sigma-1 receptor. Molecular dynamic simulation indicated that the movement of the carboxy-terminal helices (α4/α5) could play a major role in the receptor's physiological functions. The neurotoxicity induced by Aβ25-35 in SH-SY5Y cells was reduced by ropinirole at concentrations 10, 30, and 50 µM. The effect on spatial learning and memory was examined in mice with Aβ25-35 induced memory deficit using the radial arm maze. Ropinirole (10 and 20 mg/kg) significantly improved the short and long-term memories in the radial arm maze test. Our results suggest that ropinirole has the potential to be repositioned for AD treatment.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Amritha Chakkittukandiyil
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamil Nadu, India
| | - Saurav Chakraborty
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamil Nadu, India
| | - Ram Kothandan
- Bioinformatics Laboratory, Department of Biotechnology, Kumaraguru College of Technology, Coimbatore, Tamil Nadu, India
| | - Emdormi Rymbai
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamil Nadu, India
| | - Santhosh Kumar Muthu
- Department of Biochemistry, Kongunadu Arts and Science College, GN Mills, Coimbatore, Tamil Nadu, India
| | - Soumya Vasu
- Department of Pharmaceutical Chemistry, Sri Ramachandra Institute of Higher Education & Research, Porur, Chennai, Tamil Nadu, India
| | - Deepak Vasudevan Sajini
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamil Nadu, India
| | - Deepa Sugumar
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamil Nadu, India
| | - Zubair Baba Mohammad
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamil Nadu, India
| | - Saravanan Jayaram
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamil Nadu, India
| | - Kalirajan Rajagopal
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamil Nadu, India
| | - Vadivelan Ramachandran
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamil Nadu, India
| | - Divakar Selvaraj
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamil Nadu, India
| |
Collapse
|
10
|
Rymbai E, Sugumar D, Krishnamurthy PT, Selvaraj D, Vasu S, Priya S, Jayaram S. A preliminary study to identify existing drugs for potential repurposing in breast cancer based on side effect profile. Drug Res (Stuttg) 2023. [PMID: 36878466 DOI: 10.1055/a-2011-5662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 03/08/2023]
Abstract
Breast cancer is the most commonly diagnosed cancer and the second leading cause of cancer-related death in women after lung cancer. The present study aims to identify potential drug candidates using the PROMISCUOUS database for breast cancer based on side effect profile and then proceed with in silico and in vitro studies. PROMISCUOUS database was used to construct a group of drugs that share maximum side effects with letrozole. Based on the existing literature, ropinirole, risperidone, pregabalin, and gabapentin were selected for in silico and in vitro studies. The molecular docking was carried out using AUTODOCK 4.2.6. MCF-7 cell line was used to evaluate the anti-cancer activity of the selected drugs. PROMISCUOUS database revealed that as many as 23 existing drugs shared between 62 and 79 side-effects with letrozole. From docking result, we found that, ropinirole showed a good binding affinity (-7.7 kcal/mol) against aromatase compared to letrozole (-7.1 kcal/mol) which was followed by gabapentin (-6.4 kcal/mol), pregabalin (-5.7 kcal/mol) and risperidone (-5.1 kcal/mol). From the in vitro results, ropinirole and risperidone showed good anti-cancer activity of IC50 with 40.85±11.02 μg/ml and 43.10±9.58 μg/ml cell viability. Based on this study results and existing literature we conclude that risperidone, pregabalin, and gabapentin are not ideal candidates for repurposing in breast cancer but ropinirole could be an excellent choice for repurposing in breast cancer after further studies.
Collapse
Affiliation(s)
- Emdormi Rymbai
- JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, Tamil Nadu, India
| | - Deepa Sugumar
- JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, Tamil Nadu, India
| | | | - Divakar Selvaraj
- JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, Tamil Nadu, India
| | - Soumya Vasu
- Faculty of Pharmacy, Sri Ramachandra Institute of Higher Education and Research, Porur, Chennai, Tamil Nadu, India
| | - Shiva Priya
- JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, Tamil Nadu, India
| | - Saravanan Jayaram
- JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, Tamil Nadu, India
| |
Collapse
|
11
|
Miura S, Sawada R, Yorita A, Kida H, Kamada T, Yamanishi Y. A trial of topiramate for patients with hereditary spinocerebellar ataxia. Clin Case Rep 2023; 11:e6980. [PMID: 36855409 PMCID: PMC9968455 DOI: 10.1002/ccr3.6980] [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] [Academic Contribution Register] [Received: 09/29/2022] [Revised: 12/24/2022] [Accepted: 02/06/2023] [Indexed: 02/27/2023] Open
Abstract
In an open pilot trial, six patients with various hereditary forms of spinocerebellar ataxia (SCA) were assigned to topiramate (50 mg/day) for 24 weeks. Four patients completed the protocol without adverse events. Of these four patients, topiramate was effective for three patients. Some patients with SCA could respond to treatment with topiramate.
Collapse
Affiliation(s)
- Shiroh Miura
- Department of Neurology and Geriatric MedicineEhime University Graduate School of MedicineToonEhimeJapan
| | - Ryusuke Sawada
- Department of PharmacologyOkayama University Graduate School of Medicine, Dentistry and Pharmaceutical SciencesKita‐kuOkayamaJapan
| | - Akiko Yorita
- Division of Respirology, Neurology and Rheumatology, Department of MedicineKurume University School of MedicineKurumeFukuokaJapan
| | - Hiroshi Kida
- Department of AnatomyFukuoka University School of MedicineJonan‐kuFukuokaJapan
| | - Takashi Kamada
- Department of NeurologyFukuoka Sanno HospitalSawara‐kuFukuokaJapan
| | - Yoshihiro Yamanishi
- Department of Bioscience and Bioinformatics, Faculty of Computer Science and Systems EngineeringKyushu Institute of TechnologyIizukaFukuokaJapan
| |
Collapse
|
12
|
Sarojamma V, Gupta MK, Shaik JB, Vadde R. Old drugs and new opportunities—Drug repurposing in colon cancer prevention. COMPUTATIONAL METHODS IN DRUG DISCOVERY AND REPURPOSING FOR CANCER THERAPY 2023:223-235. [DOI: 10.1016/b978-0-443-15280-1.00010-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 09/06/2023]
|
13
|
Taweel B, Marson AG, Mirza N. A systems medicine strategy to predict the efficacy of drugs for monogenic epilepsies. Epilepsia 2022; 63:3125-3133. [PMID: 36196775 PMCID: PMC10092251 DOI: 10.1111/epi.17429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/19/2022] [Revised: 10/03/2022] [Accepted: 10/03/2022] [Indexed: 01/11/2023]
Abstract
OBJECTIVE Monogenic epilepsies are rare but often severe. Because of their rarity, they are neglected by traditional drug developers. Hence, many lack effective treatments. Treatments for a disease can be discovered more quickly and economically by computationally predicting drugs that can be repurposed for it. We aimed to create a computational method to predict the efficacy of drugs for monogenic epilepsies, and to use the method to predict drugs for Dravet syndrome, as (1) it is the archetypal monogenic catastrophic epilepsy; (2) few antiseizure medications are efficacious in Dravet syndrome; and (3) predicting the effect of drugs on Dravet syndrome is challenging, because Dravet syndrome is typically caused by an SCN1A mutation, but some antiseizure medications that are efficacious in Dravet syndrome do not affect SCN1A, and some antiseizure medications that affect SCN1A aggravate seizures in Dravet syndrome. METHODS We have devised a computational method to predict drugs that could be repurposed for a monogenic epilepsy, based on a combined measure of drugs' effects upon (1) the function of the disease's causal gene and other genes predicted to influence its phenotype, (2) the transcriptomic dysregulation induced by the casual gene mutation, and (3) clinical phenotypes. RESULTS Our method correctly predicts drugs that are more effective, less effective, ineffective, and aggravating for seizures in people with Dravet syndrome. Our method correctly predicts the positive "hits" from large-scale screening of compounds in an animal model of Dravet syndrome. We predict the relative efficacy of 1462 drugs. At least 38 drugs are ranked higher than one or more of the antiseizure drugs currently used for Dravet syndrome and have existing evidence of antiseizure efficacy in animal models. SIGNIFICANCE Our predictions are a novel resource for identifying new treatments for seizures in Dravet syndrome, and our method can be adapted for other monogenic epilepsies.
Collapse
Affiliation(s)
- Basel Taweel
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular, and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Anthony G Marson
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular, and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Nasir Mirza
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular, and Integrative Biology, University of Liverpool, Liverpool, UK
| |
Collapse
|
14
|
Huang W, Li Z, Kang Y, Ye X, Feng W. Drug Repositioning Based on the Enhanced Message Passing and Hypergraph Convolutional Networks. Biomolecules 2022; 12:1666. [PMID: 36359016 PMCID: PMC9687543 DOI: 10.3390/biom12111666] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/12/2022] [Revised: 11/03/2022] [Accepted: 11/08/2022] [Indexed: 10/17/2023] Open
Abstract
Drug repositioning, an important method of drug development, is utilized to discover investigational drugs beyond the originally approved indications, expand the application scope of drugs, and reduce the cost of drug development. With the emergence of increasingly drug-disease-related biological networks, the challenge still remains to effectively fuse biological entity data and accurately achieve drug-disease repositioning. This paper proposes a new drug repositioning method named EMPHCN based on enhanced message passing and hypergraph convolutional networks (HGCN). It firstly constructs the homogeneous multi-view information with multiple drug similarity features and then extracts the intra-domain embedding of drugs through the combination of HGCN and channel attention mechanism. Secondly, inter-domain information of known drug-disease associations is extracted by graph convolutional networks combining node and edge embedding (NEEGCN), and a heterogeneous network composed of drugs, proteins and diseases is built as an important auxiliary to enhance the inter-domain message passing of drugs and diseases. Besides, the intra-domain embedding of diseases is also extracted through HGCN. Ultimately, intra-domain and inter-domain embeddings of drugs and diseases are integrated as the final embedding for calculating the drug-disease correlation matrix. Through 10-fold cross-validation on some benchmark datasets, we find that the AUPR of EMPHCN reaches 0.593 (T1) and 0.526 (T2), respectively, and the AUC achieves 0.887 (T1) and 0.961 (T2) respectively, which shows that EMPHCN has an advantage over other state-of-the-art prediction methods. Concerning the new disease association prediction, the AUC of EMPHCN through the five-fold cross-validation reaches 0.806 (T1) and 0.845 (T2), which are 4.3% (T1) and 4.0% (T2) higher than the second best existing methods, respectively. In the case study, EMPHCN also achieves satisfactory results in real drug repositioning for breast carcinoma and Parkinson's disease.
Collapse
Affiliation(s)
- Weihong Huang
- School of Informatics Science and Technology, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Zhong Li
- School of Informatics Science and Technology, Zhejiang Sci-Tech University, Hangzhou 310018, China
- Zhejiang Province Key Laboratory of Smart Management & Application of Modern Agricultural Resources, School of Information Engineering, Huzhou University, Huzhou 313000, China
| | - Yanlei Kang
- Zhejiang Province Key Laboratory of Smart Management & Application of Modern Agricultural Resources, School of Information Engineering, Huzhou University, Huzhou 313000, China
| | - Xinghuo Ye
- Zhejiang Province Key Laboratory of Smart Management & Application of Modern Agricultural Resources, School of Information Engineering, Huzhou University, Huzhou 313000, China
| | - Wenming Feng
- Department of General Surgery, The First Affiliated Hospital of Huzhou University, Huzhou 313000, China
| |
Collapse
|
15
|
Kakoti BB, Bezbaruah R, Ahmed N. Therapeutic drug repositioning with special emphasis on neurodegenerative diseases: Threats and issues. Front Pharmacol 2022; 13:1007315. [PMID: 36263141 PMCID: PMC9574100 DOI: 10.3389/fphar.2022.1007315] [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] [Academic Contribution Register] [Received: 07/30/2022] [Accepted: 09/12/2022] [Indexed: 11/21/2022] Open
Abstract
Drug repositioning or repurposing is the process of discovering leading-edge indications for authorized or declined/abandoned molecules for use in different diseases. This approach revitalizes the traditional drug discovery method by revealing new therapeutic applications for existing drugs. There are numerous studies available that highlight the triumph of several drugs as repurposed therapeutics. For example, sildenafil to aspirin, thalidomide to adalimumab, and so on. Millions of people worldwide are affected by neurodegenerative diseases. According to a 2021 report, the Alzheimer's disease Association estimates that 6.2 million Americans are detected with Alzheimer's disease. By 2030, approximately 1.2 million people in the United States possibly acquire Parkinson's disease. Drugs that act on a single molecular target benefit people suffering from neurodegenerative diseases. Current pharmacological approaches, on the other hand, are constrained in their capacity to unquestionably alter the course of the disease and provide patients with inadequate and momentary benefits. Drug repositioning-based approaches appear to be very pertinent, expense- and time-reducing strategies for the enhancement of medicinal opportunities for such diseases in the current era. Kinase inhibitors, for example, which were developed for various oncology indications, demonstrated significant neuroprotective effects in neurodegenerative diseases. This review expounds on the classical and recent examples of drug repositioning at various stages of drug development, with a special focus on neurodegenerative disorders and the aspects of threats and issues viz. the regulatory, scientific, and economic aspects.
Collapse
Affiliation(s)
- Bibhuti Bhusan Kakoti
- Department of Pharmaceutical Sciences, Faculty of Science and Engineering, Dibrugarh University, Dibrugarh, India
| | | | | |
Collapse
|
16
|
Song Y, Cui H, Zhang T, Yang T, Li X, Xuan P. Prediction of Drug-Related Diseases Through Integrating Pairwise Attributes and Neighbor Topological Structures. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2022; 19:2963-2974. [PMID: 34133286 DOI: 10.1109/tcbb.2021.3089692] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 06/12/2023]
Abstract
Identifying new disease indications for the approved drugs can help reduce the cost and time of drug development. Most of the recent methods focus on exploiting the various information related to drugs and diseases for predicting the candidate drug-disease associations. However, the previous methods failed to deeply integrate the neighborhood topological structure and the node attributes of an interested drug-disease node pair. We propose a new prediction method, ANPred, to learn and integrate pairwise attribute information and neighbor topology information from the similarities and associations related to drugs and diseases. First, a bi-layer heterogeneous network with intra-layer and inter-layer connections is established to combine the drug similarities, the disease similarities, and the drug-disease associations. Second, the embedding of a pair of drug and disease is constructed based on integrating multiple biological premises about drugs and diseases. The learning framework based on multi-layer convolutional neural networks is designed to learn the attribute representation of the pair of drug and disease nodes from its embedding. The sequences composed of neighbor nodes are formed based on random walk on the heterogeneous network. A framework based on fully-connected autoencoder and skip-gram module is constructed to learn the neighbor topological representations of nodes. The cross-validation results indicate the performance of ANPred is superior to several state-of-the-art methods. The case studies on 5 drugs further confirm the ability of ANPred in discovering the potential drug-disease association candidates.
Collapse
|
17
|
Zhu Q, Qu C, Liu R, Vatas G, Clough A, Nguyễn ÐT, Sid E, Mathé E, Xu Y. Rare disease-based scientific annotation knowledge graph. Front Artif Intell 2022; 5:932665. [PMID: 36034595 PMCID: PMC9403737 DOI: 10.3389/frai.2022.932665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/30/2022] [Accepted: 07/15/2022] [Indexed: 11/13/2022] Open
Abstract
Rare diseases (RDs) are naturally associated with a low prevalence rate, which raises a big challenge due to there being less data available for supporting preclinical and clinical studies. There has been a vast improvement in our understanding of RD, largely owing to advanced big data analytic approaches in genetics/genomics. Consequently, a large volume of RD-related publications has been accumulated in recent years, which offers opportunities to utilize these publications for accessing the full spectrum of the scientific research and supporting further investigation in RD. In this study, we systematically analyzed, semantically annotated, and scientifically categorized RD-related PubMed articles, and integrated those semantic annotations in a knowledge graph (KG), which is hosted in Neo4j based on a predefined data model. With the successful demonstration of scientific contribution in RD via the case studies performed by exploring this KG, we propose to extend the current effort by expanding more RD-related publications and more other types of resources as a next step.
Collapse
Affiliation(s)
- Qian Zhu
- Division of Pre-clinical Innovation, National Center for Advancing Translational Sciences, Rockville, MD, United States
- *Correspondence: Qian Zhu
| | - Chunxu Qu
- Division of Rare Diseases Research Innovation, National Center for Advancing Translational Sciences, Bethesda, MD, United States
| | - Ruizheng Liu
- Division of Rare Diseases Research Innovation, National Center for Advancing Translational Sciences, Bethesda, MD, United States
| | | | | | | | - Eric Sid
- Division of Rare Diseases Research Innovation, National Center for Advancing Translational Sciences, Bethesda, MD, United States
| | - Ewy Mathé
- Division of Pre-clinical Innovation, National Center for Advancing Translational Sciences, Rockville, MD, United States
| | - Yanji Xu
- Division of Rare Diseases Research Innovation, National Center for Advancing Translational Sciences, Bethesda, MD, United States
| |
Collapse
|
18
|
Cuesta AM, Gallardo-Vara E, Casado-Vela J, Recio-Poveda L, Botella LM, Albiñana V. The Role of Propranolol as a Repurposed Drug in Rare Vascular Diseases. Int J Mol Sci 2022; 23:ijms23084217. [PMID: 35457036 PMCID: PMC9025921 DOI: 10.3390/ijms23084217] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/25/2022] [Revised: 04/08/2022] [Accepted: 04/09/2022] [Indexed: 01/27/2023] Open
Abstract
Rare Diseases (RD) are defined by their prevalence in less than 5 in 10,000 of the general population. Considered individually, each RD may seem insignificant, but together they add up to more than 7000 different diseases. Research in RD is not attractive for pharmaceutical companies since it is unlikely to recover development costs for medicines aimed to small numbers of patients. Since most of these diseases are life threatening, this fact underscores the urgent need for treatments. Drug repurposing consists of identifying new uses for approved drugs outside the scope of the original medical indication. It is an alternative option in drug development and represents a viable and risk-managed strategy to develop for RDs. In 2008, the “off label” therapeutic benefits of propranolol were described in the benign tumor Infantile Hemangioma. Propranolol, initially prescribed for high blood pressure, irregular heart rate, essential tremor, and anxiety, has, in the last decade, shown increasing evidence of its antiangiogenic, pro-apoptotic, vasoconstrictor and anti-inflammatory properties in different RDs, including vascular or oncological pathologies. This review highlights the finished and ongoing trials in which propranolol has arisen as a good repurposing drug for improving the health condition in RDs.
Collapse
Affiliation(s)
- Angel M. Cuesta
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad Complutense de Madrid, 28040 Madrid, Spain;
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Unidad 707, 28029 Madrid, Spain;
| | - Eunate Gallardo-Vara
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, 300 George Street, New Haven, CT 06511, USA;
| | - Juan Casado-Vela
- Facultad de Ciencias Experimentales, Universidad Francisco de Vitoria, Pozuelo, 28223 Madrid, Spain;
- Departamento de Bioingeniería, Escuela Politécnica Superior, Universidad Carlos III de Madrid, Av. de la Universidad, 30, 28911 Madrid, Spain
| | - Lucía Recio-Poveda
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Unidad 707, 28029 Madrid, Spain;
- Centro de Investigaciones Biológicas Margaritas Salas, 28040 Madrid, Spain
| | - Luisa-María Botella
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Unidad 707, 28029 Madrid, Spain;
- Centro de Investigaciones Biológicas Margaritas Salas, 28040 Madrid, Spain
- Correspondence: (L.-M.B.); (V.A.)
| | - Virginia Albiñana
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Unidad 707, 28029 Madrid, Spain;
- Centro de Investigaciones Biológicas Margaritas Salas, 28040 Madrid, Spain
- Correspondence: (L.-M.B.); (V.A.)
| |
Collapse
|
19
|
Lee S, Jeon S, Kim HS. A Study on Methodologies of Drug Repositioning Using Biomedical Big Data: A Focus on Diabetes Mellitus. Endocrinol Metab (Seoul) 2022; 37:195-207. [PMID: 35413782 PMCID: PMC9081315 DOI: 10.3803/enm.2022.1404] [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] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 01/11/2022] [Accepted: 03/21/2022] [Indexed: 11/11/2022] Open
Abstract
Drug repositioning is a strategy for identifying new applications of an existing drug that has been previously proven to be safe. Based on several examples of drug repositioning, we aimed to determine the methodologies and relevant steps associated with drug repositioning that should be pursued in the future. Reports on drug repositioning, retrieved from PubMed from January 2011 to December 2020, were classified based on an analysis of the methodology and reviewed by experts. Among various drug repositioning methods, the network-based approach was the most common (38.0%, 186/490 cases), followed by machine learning/deep learningbased (34.3%, 168/490 cases), text mining-based (7.1%, 35/490 cases), semantic-based (5.3%, 26/490 cases), and others (15.3%, 75/490 cases). Although drug repositioning offers several advantages, its implementation is curtailed by the need for prior, conclusive clinical proof. This approach requires the construction of various databases, and a deep understanding of the process underlying repositioning is quintessential. An in-depth understanding of drug repositioning could reduce the time, cost, and risks inherent to early drug development, providing reliable scientific evidence. Furthermore, regarding patient safety, drug repurposing might allow the discovery of new relationships between drugs and diseases.
Collapse
Affiliation(s)
- Suehyun Lee
- Department of Biomedical Informatics, Konyang University College of Medicine, Daejeon, Korea
- Health Care Data Science Center, Konyang University Hospital, Daejeon, Korea
| | - Seongwoo Jeon
- Health Care Data Science Center, Konyang University Hospital, Daejeon, Korea
| | - Hun-Sung Kim
- Department of Medical Informatics, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Corresponding author: Hun-Sung Kim Department of Medical Informatics, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Korea Tel: +82-2-2258-8262, Fax: +82-2-2258-8297, E-mail:
| |
Collapse
|
20
|
Qin L, Wang J, Wu Z, Li W, Liu G, Tang Y. Drug Repurposing for Newly Emerged Diseases via Network-Based Inference on A Gene-Disease-Drug Network. Mol Inform 2022; 41:e2200001. [PMID: 35338586 DOI: 10.1002/minf.202200001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/05/2022] [Accepted: 03/25/2022] [Indexed: 11/06/2022]
Abstract
Identification of disease-drug associations is an effective strategy for drug repurposing, especially in searching old drugs for newly emerged diseases like COVID-19. In this study, we put forward a network-based method named NEDNBI to predict disease-drug associations based on a gene-disease-drug tripartite network, which could be applied in drug repurposing. The novelty of our method lies in the fact that no negative data are required, and new disease could be added into the disease-drug network with gene as the bridge. The comprehensive evaluation results showed that the proposed method had good performance, with AUC value 0.948 ± 0.009 for 10-fold cross validation. In a case study, 8 of the 20 predicted old drugs have been tested clinically for the treatment of COVID-19, which illustrated the usefulness of our method in drug repurposing. The source code and data of the method are available at https://github.com/Qli97/NEDNBI.
Collapse
Affiliation(s)
- Li Qin
- East China University of Science and Technology School of Pharmacy, CHINA
| | - Jiye Wang
- East China University of Science and Technology School of Pharmacy, CHINA
| | - Zengrui Wu
- East China University of Science and Technology, CHINA
| | | | - Guixia Liu
- East China University of Science and Technology, CHINA
| | - Yun Tang
- East China University of Science and Technology, CHINA
| |
Collapse
|
21
|
Bahmad HF, Demus T, Moubarak MM, Daher D, Alvarez Moreno JC, Polit F, Lopez O, Merhe A, Abou-Kheir W, Nieder AM, Poppiti R, Omarzai Y. Overcoming Drug Resistance in Advanced Prostate Cancer by Drug Repurposing. Med Sci (Basel) 2022; 10:medsci10010015. [PMID: 35225948 PMCID: PMC8883996 DOI: 10.3390/medsci10010015] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/22/2022] [Revised: 02/12/2022] [Accepted: 02/16/2022] [Indexed: 12/12/2022] Open
Abstract
Prostate cancer (PCa) is the second most common cancer in men. Common treatments include active surveillance, surgery, or radiation. Androgen deprivation therapy and chemotherapy are usually reserved for advanced disease or biochemical recurrence, such as castration-resistant prostate cancer (CRPC), but they are not considered curative because PCa cells eventually develop drug resistance. The latter is achieved through various cellular mechanisms that ultimately circumvent the pharmaceutical’s mode of action. The need for novel therapeutic approaches is necessary under these circumstances. An alternative way to treat PCa is by repurposing of existing drugs that were initially intended for other conditions. By extrapolating the effects of previously approved drugs to the intracellular processes of PCa, treatment options will expand. In addition, drug repurposing is cost-effective and efficient because it utilizes drugs that have already demonstrated safety and efficacy. This review catalogues the drugs that can be repurposed for PCa in preclinical studies as well as clinical trials.
Collapse
Affiliation(s)
- Hisham F. Bahmad
- Arkadi M. Rywlin M.D. Department of Pathology and Laboratory Medicine, Mount Sinai Medical Center, Miami Beach, FL 33140, USA; (J.C.A.M.); (F.P.); (R.P.); (Y.O.)
- Correspondence: or ; Tel.: +1-786-961-0216
| | - Timothy Demus
- Division of Urology, Columbia University, Mount Sinai Medical Center, Miami Beach, FL 33140, USA; (T.D.); (A.M.N.)
| | - Maya M. Moubarak
- Department of Anatomy, Cell Biology, and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107-2020, Lebanon; (M.M.M.); (W.A.-K.)
- CNRS, IBGC, UMR5095, Universite de Bordeaux, F-33000 Bordeaux, France
| | - Darine Daher
- Faculty of Medicine, American University of Beirut, Beirut 1107-2020, Lebanon;
| | - Juan Carlos Alvarez Moreno
- Arkadi M. Rywlin M.D. Department of Pathology and Laboratory Medicine, Mount Sinai Medical Center, Miami Beach, FL 33140, USA; (J.C.A.M.); (F.P.); (R.P.); (Y.O.)
| | - Francesca Polit
- Arkadi M. Rywlin M.D. Department of Pathology and Laboratory Medicine, Mount Sinai Medical Center, Miami Beach, FL 33140, USA; (J.C.A.M.); (F.P.); (R.P.); (Y.O.)
| | - Olga Lopez
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA;
| | - Ali Merhe
- Department of Urology, Jackson Memorial Hospital, University of Miami, Leonard M. Miller School of Medicine, Miami, FL 33136, USA;
| | - Wassim Abou-Kheir
- Department of Anatomy, Cell Biology, and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107-2020, Lebanon; (M.M.M.); (W.A.-K.)
| | - Alan M. Nieder
- Division of Urology, Columbia University, Mount Sinai Medical Center, Miami Beach, FL 33140, USA; (T.D.); (A.M.N.)
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA;
| | - Robert Poppiti
- Arkadi M. Rywlin M.D. Department of Pathology and Laboratory Medicine, Mount Sinai Medical Center, Miami Beach, FL 33140, USA; (J.C.A.M.); (F.P.); (R.P.); (Y.O.)
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA;
| | - Yumna Omarzai
- Arkadi M. Rywlin M.D. Department of Pathology and Laboratory Medicine, Mount Sinai Medical Center, Miami Beach, FL 33140, USA; (J.C.A.M.); (F.P.); (R.P.); (Y.O.)
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA;
| |
Collapse
|
22
|
Chyr J, Gong H, Zhou X. DOTA: Deep Learning Optimal Transport Approach to Advance Drug Repositioning for Alzheimer's Disease. Biomolecules 2022; 12:196. [PMID: 35204697 PMCID: PMC8961573 DOI: 10.3390/biom12020196] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/28/2021] [Revised: 01/16/2022] [Accepted: 01/22/2022] [Indexed: 02/04/2023] Open
Abstract
Alzheimer's disease (AD) is the leading cause of age-related dementia, affecting over 5 million people in the United States and incurring a substantial global healthcare cost. Unfortunately, current treatments are only palliative and do not cure AD. There is an urgent need to develop novel anti-AD therapies; however, drug discovery is a time-consuming, expensive, and high-risk process. Drug repositioning, on the other hand, is an attractive approach to identify drugs for AD treatment. Thus, we developed a novel deep learning method called DOTA (Drug repositioning approach using Optimal Transport for Alzheimer's disease) to repurpose effective FDA-approved drugs for AD. Specifically, DOTA consists of two major autoencoders: (1) a multi-modal autoencoder to integrate heterogeneous drug information and (2) a Wasserstein variational autoencoder to identify effective AD drugs. Using our approach, we predict that antipsychotic drugs with circadian effects, such as quetiapine, aripiprazole, risperidone, suvorexant, brexpiprazole, olanzapine, and trazadone, will have efficacious effects in AD patients. These drugs target important brain receptors involved in memory, learning, and cognition, including serotonin 5-HT2A, dopamine D2, and orexin receptors. In summary, DOTA repositions promising drugs that target important biological pathways and are predicted to improve patient cognition, circadian rhythms, and AD pathogenesis.
Collapse
Affiliation(s)
- Jacqueline Chyr
- Center for Computational Systems Medicine, School of Biomedical Informatics, University of Texas Health Science Center, Houston, TX 77030, USA;
| | - Haoran Gong
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu 610041, China;
| | - Xiaobo Zhou
- Center for Computational Systems Medicine, School of Biomedical Informatics, University of Texas Health Science Center, Houston, TX 77030, USA;
| |
Collapse
|
23
|
Lu L, Qin J, Chen J, Wu H, Zhao Q, Miyano S, Zhang Y, Yu H, Li C. DDIT: An Online Predictor for Multiple Clinical Phenotypic Drug-Disease Associations. Front Pharmacol 2022; 12:772026. [PMID: 35126114 PMCID: PMC8809407 DOI: 10.3389/fphar.2021.772026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/07/2021] [Accepted: 11/19/2021] [Indexed: 12/20/2022] Open
Abstract
Background: Drug repurposing provides an effective method for high-speed, low-risk drug development. Clinical phenotype-based screening exceeded target-based approaches in discovering first-in-class small-molecule drugs. However, most of these approaches predict only binary phenotypic associations between drugs and diseases; the types of drug and diseases have not been well exploited. Principally, the clinical phenotypes of a known drug can be divided into indications (Is), side effects (SEs), and contraindications (CIs). Incorporating these different clinical phenotypes of drug–disease associations (DDAs) can improve the prediction accuracy of the DDAs. Methods: We develop Drug Disease Interaction Type (DDIT), a user-friendly online predictor that supports drug repositioning by submitting known Is, SEs, and CIs for a target drug of interest. The dataset for Is, SEs, and CIs was extracted from PREDICT, SIDER, and MED-RT, respectively. To unify the names of the drugs and diseases, we mapped their names to the Unified Medical Language System (UMLS) ontology using Rest API. We then integrated multiple clinical phenotypes into a conditional restricted Boltzmann machine (RBM) enabling the identification of different phenotypes of drug–disease associations, including the prediction of as yet unknown DDAs in the input. Results: By 10-fold cross-validation, we demonstrate that DDIT can effectively capture the latent features of the drug–disease association network and represents over 0.217 and over 0.072 improvement in AUC and AUPR, respectively, for predicting the clinical phenotypes of DDAs compared with the classic K-nearest neighbors method (KNN, including drug-based KNN and disease-based KNN), Random Forest, and XGBoost. By conducting leave-one-drug-class-out cross-validation, the AUC and AUPR of DDIT demonstrated an improvement of 0.135 in AUC and 0.075 in AUPR compared to any of the other four methods. Within the top 10 predicted indications, side effects, and contraindications, 7/10, 9/10, and 9/10 hit known drug–disease associations. Overall, DDIT is a useful tool for predicting multiple clinical phenotypic types of drug–disease associations.
Collapse
Affiliation(s)
- Lu Lu
- Department of Human Genetics, Department of Ultrasound and Women’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiale Qin
- Department of Human Genetics, Department of Ultrasound and Women’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Hangzhou, China
| | - Jiandong Chen
- School of Public Health, Undergraduate School of Zhejiang University, Hangzhou, China
| | - Hao Wu
- Department of Human Genetics, Department of Ultrasound and Women’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qiang Zhao
- Department of Human Genetics, Department of Ultrasound and Women’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Satoru Miyano
- M&D Data Science Center, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yaozhong Zhang
- The Institute of Medical Science, the University of Tokyo, Tokyo, Japan
- *Correspondence: Yaozhong Zhang, ; Hua Yu, ; Chen Li,
| | - Hua Yu
- Department of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou, China
- *Correspondence: Yaozhong Zhang, ; Hua Yu, ; Chen Li,
| | - Chen Li
- Department of Human Genetics, Department of Ultrasound and Women’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Hangzhou, China
- Department of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou, China
- *Correspondence: Yaozhong Zhang, ; Hua Yu, ; Chen Li,
| |
Collapse
|
24
|
Schuler J, Falls Z, Mangione W, Hudson ML, Bruggemann L, Samudrala R. Evaluating the performance of drug-repurposing technologies. Drug Discov Today 2022; 27:49-64. [PMID: 34400352 PMCID: PMC10014214 DOI: 10.1016/j.drudis.2021.08.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/17/2021] [Revised: 06/20/2021] [Accepted: 08/08/2021] [Indexed: 01/22/2023]
Abstract
Drug-repurposing technologies are growing in number and maturing. However, comparisons to each other and to reality are hindered because of a lack of consensus with respect to performance evaluation. Such comparability is necessary to determine scientific merit and to ensure that only meaningful predictions from repurposing technologies carry through to further validation and eventual patient use. Here, we review and compare performance evaluation measures for these technologies using version 2 of our shotgun repurposing Computational Analysis of Novel Drug Opportunities (CANDO) platform to illustrate their benefits, drawbacks, and limitations. Understanding and using different performance evaluation metrics ensures robust cross-platform comparability, enabling us to continue to strive toward optimal repurposing by decreasing the time and cost of drug discovery and development.
Collapse
Affiliation(s)
- James Schuler
- Department of Biomedical Informatics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA.
| | - Zackary Falls
- Department of Biomedical Informatics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - William Mangione
- Department of Biomedical Informatics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Matthew L Hudson
- Department of Biomedical Informatics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Liana Bruggemann
- Department of Biomedical Informatics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Ram Samudrala
- Department of Biomedical Informatics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA.
| |
Collapse
|
25
|
AI-powered drug repurposing for developing COVID-19 treatments. REFERENCE MODULE IN BIOMEDICAL SCIENCES 2022. [PMCID: PMC8865759 DOI: 10.1016/b978-0-12-824010-6.00005-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Academic Contribution Register] [Indexed: 11/17/2022]
Abstract
Emerging infectious diseases are an ever-present threat to public health, and COVID-19 is the most recent example. There is an urgent need to develop a robust framework to combat the disease with safe and effective therapeutic options. Compared to de novo drug discovery, drug repurposing may offer a lower-cost and faster drug discovery paradigm to explore potential treatment options of existing drugs. This chapter elucidates the advantages of artificial intelligence (AI) in enhancing the drug repurposing process from a data science perspective, using COVID-19 as an example. First, we elaborate on how AI-powered drug repurposing benefits from the accumulated data and knowledge of COVID-19 natural history and pathogenesis. Second, we summarize the pros and cons of AI-powered drug repurposing strategies to facilitate fit-for-purpose selection. Finally, we outline challenges of AI-powered drug repurposing from a regulatory perspective and suggest some potential solutions. AI-powered drug purposing is promising for emerging treatments for COVID-19 infection. Accumulated biological data profiles facilitate AI-based drug repurposing efforts for development of COVID-19 therapies. The ‘fit-for-purpose selection of AI-powered drug repurposing strategies is key to uncovering hidden information among drugs, targets, and diseases. Efforts from different stakeholders boost the adoption of AI-powered drug repurposing in the regulatory setting.
Collapse
|
26
|
Yang B, Wang X, Dong D, Pan Y, Wu J, Liu J. Existing Drug Repurposing for Glioblastoma to Discover Candidate Drugs as a New a Approach. LETT DRUG DES DISCOV 2022. [DOI: 10.2174/1570180818666210509141735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/22/2022]
Abstract
Aims:
Repurposing of drugs has been hypothesized as a means of identifying novel
treatment methods for certain diseases.
Background:
Glioblastoma (GB) is an aggressive type of human cancer; the most effective treatment
for glioblastoma is chemotherapy, whereas, when repurposing drugs, a lot of time and money can be
saved.
Objective:
Repurposing of the existing drug may be used to discover candidate drugs for individualized
treatments of GB.
Method:
We used the bioinformatics method to obtain the candidate drugs. In addition, the drugs
were verified by MTT assay, Transwell® assays, TUNEL staining, and in vivo tumor formation experiments,
as well as statistical analysis.
Result:
We obtained 4 candidate drugs suitable for the treatment of glioma, camptothecin, doxorubicin,
daunorubicin and mitoxantrone, by the expression spectrum data IPAS algorithm analysis and
drug-pathway connectivity analysis. These validation experiments showed that camptothecin was
more effective in treating the GB, such as MTT assay, Transwell® assays, TUNEL staining, and in
vivo tumor formation.
Conclusion:
With regard to personalized treatment, this present study may be used to guide the research
of new drugs via verification experiments and tumor formation. The present study also provides
a guide to systematic, individualized drug discovery for complex diseases and may contribute
to the future application of individualized treatments.
Collapse
Affiliation(s)
- Bo Yang
- Department of Neurosurgery, Hangzhou Medical College Affiliated Lin’an People’s Hospital, The First People’s
Hospital of Hangzhou Lin’an District, Hangzhou, Zhejiang, 311300, China
| | - Xiande Wang
- Department of Neurosurgery, Hangzhou Medical College Affiliated Lin’an People’s Hospital, The First People’s
Hospital of Hangzhou Lin’an District, Hangzhou, Zhejiang, 311300, China
| | - Dong Dong
- Department of Neurosurgery, Hangzhou Medical College Affiliated Lin’an People’s Hospital, The First People’s
Hospital of Hangzhou Lin’an District, Hangzhou, Zhejiang, 311300, China
| | - Yunqing Pan
- Department of Neurosurgery, Hangzhou Medical College Affiliated Lin’an People’s Hospital, The First People’s
Hospital of Hangzhou Lin’an District, Hangzhou, Zhejiang, 311300, China
| | - Junhua Wu
- Department of Neurosurgery, Hangzhou Medical College Affiliated Lin’an People’s Hospital, The First People’s
Hospital of Hangzhou Lin’an District, Hangzhou, Zhejiang, 311300, China
| | - Jianjian Liu
- Department of Neurosurgery, Hangzhou Medical College Affiliated Lin’an People’s Hospital, The First People’s
Hospital of Hangzhou Lin’an District, Hangzhou, Zhejiang, 311300, China
| |
Collapse
|
27
|
Xiang H, Li A, Lin X. An Optimization Method for Drug-Target Interaction Prediction Based on RandSAS Strategy. LECTURE NOTES IN COMPUTER SCIENCE 2022:547-555. [DOI: 10.1007/978-3-031-13829-4_47] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 01/03/2025]
|
28
|
Yin Z, Wong STC. Artificial intelligence unifies knowledge and actions in drug repositioning. Emerg Top Life Sci 2021; 5:803-813. [PMID: 34881780 PMCID: PMC8923082 DOI: 10.1042/etls20210223] [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] [Academic Contribution Register] [Received: 10/11/2021] [Revised: 11/08/2021] [Accepted: 11/09/2021] [Indexed: 11/17/2022]
Abstract
Drug repositioning aims to reuse existing drugs, shelved drugs, or drug candidates that failed clinical trials for other medical indications. Its attraction is sprung from the reduction in risk associated with safety testing of new medications and the time to get a known drug into the clinics. Artificial Intelligence (AI) has been recently pursued to speed up drug repositioning and discovery. The essence of AI in drug repositioning is to unify the knowledge and actions, i.e. incorporating real-world and experimental data to map out the best way forward to identify effective therapeutics against a disease. In this review, we share positive expectations for the evolution of AI and drug repositioning and summarize the role of AI in several methods of drug repositioning.
Collapse
Affiliation(s)
- Zheng Yin
- Department of Systems Medicine and Bioengineering, Houston Methodist Cancer Center and Ting Tsung & Wei Fong Chao Center for BRAIN, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX 77030, U.S.A
| | - Stephen T C Wong
- Department of Systems Medicine and Bioengineering, Houston Methodist Cancer Center and Ting Tsung & Wei Fong Chao Center for BRAIN, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, TX 77030, U.S.A
| |
Collapse
|
29
|
Charvériat M, Darmoni SJ, Lafon V, Moore N, Bordet R, Veys J, Mouthon F. Use of real-world evidence in translational pharmacology research. Fundam Clin Pharmacol 2021; 36:230-236. [PMID: 34676579 DOI: 10.1111/fcp.12734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/17/2021] [Revised: 10/01/2021] [Accepted: 10/18/2021] [Indexed: 12/01/2022]
Abstract
Real-world evidence (RWE) refers to observational data gathered outside the formalism of randomized controlled trials, in real life situations, on marketed drugs. While clinical trials are the gold standards to demonstrate the efficacy and tolerability of a medicinal product, the generalizability of their results to actual use in real-life is limited by the biases induced by the very nature of clinical trials; indeed, the patients included in the trials may differ from actual users because of their concomitant diseases or treatments, or other factors excluding them from the trials. Clinical researchers and pharmaceutical industries have hence become increasingly interested in expanding and integrating RWE into clinical research, by capitalizing on the exponential growth in access to data from electronic health records, claims databases, electronic devices, software or mobile applications, registries embedded in clinical practice and social media. Meanwhile, applications of RWE may also be used for drug discovery and repurposing, for clinical developments and post-marketing studies. The aim of this review is to provide our opinion regarding the use of RWE in translational research, including non-clinical and clinical pharmacology research, at the different step of drugs development use.
Collapse
Affiliation(s)
| | - Stephan J Darmoni
- Department of BioMedical Informatics, Rouen University Hospital & LIMICS U1142 INSERM, Sorbonne University, Paris, France
| | | | | | - Régis Bordet
- INSERM, CHU Lille, University of Lille, Lille, France
| | | | | | | |
Collapse
|
30
|
Prieto Santamaría L, Ugarte Carro E, Díaz Uzquiano M, Menasalvas Ruiz E, Pérez Gallardo Y, Rodríguez-González A. A data-driven methodology towards evaluating the potential of drug repurposing hypotheses. Comput Struct Biotechnol J 2021; 19:4559-4573. [PMID: 34471499 PMCID: PMC8387760 DOI: 10.1016/j.csbj.2021.08.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/20/2021] [Revised: 07/08/2021] [Accepted: 08/03/2021] [Indexed: 12/14/2022] Open
Abstract
Drug repurposing has become a widely used strategy to accelerate the process of finding treatments. While classical de novo drug development involves high costs, risks, and time-consuming paths, drug repurposing allows to reuse already-existing and approved drugs for new indications. Numerous research has been carried out in this field, both in vitro and in silico. Computational drug repurposing methods make use of modern heterogeneous biomedical data to identify and prioritize new indications for old drugs. In the current paper, we present a new complete methodology to evaluate new potentially repurposable drugs based on disease-gene and disease-phenotype associations, identifying significant differences between repurposing and non-repurposing data. We have collected a set of known successful drug repurposing case studies from the literature and we have analysed their dissimilarities with other biomedical data not necessarily participating in repurposing processes. The information used has been obtained from the DISNET platform. We have performed three analyses (at the genetical, phenotypical, and categorization levels), to conclude that there is a statistically significant difference between actual repurposing-related information and non-repurposing data. The insights obtained could be relevant when suggesting new potential drug repurposing hypotheses.
Collapse
Key Words
- ACE, Angiotensin I Converting Enzyme
- AHR, Aryl Hydrocarbon Receptor
- ALK, Anaplastic Lymphoma Kinase
- API, Application Programming Interface
- CMap, Connectivity Map
- COX-2, Cyclooxygenase 2
- CUI, Concept Unique Identifier
- DISNET knowledge base
- DR, Drug Repurposing or Drug Repositioning
- DRD3, Dopamine Receptor D3
- Data integration
- Disease understanding
- Drug repositioning
- Drug repurposing
- Drug-disease validation
- ESR1, Estrogen Receptor 1
- ESR2, Estrogen Receptor 2
- FCGR2A, Fc Fragment Of IgG Receptor IIa
- FCGR3A, Fc Fragment Of IgG Receptor IIIa
- FCGR3B, Fc Fragment Of IgG Receptor IIIb
- GDA, Gene Disease Association
- ICD-10-CM, International Classification of Diseases, 10th revision, Clinical Modification
- ID, Identifier
- KDR, Kinase insert Domain Receptor
- LTα, Lymphotoxin alpha
- MeSH-PA, Medical Subject Headings – Pharmacological Action
- ND, New Disease
- NLM, National Library of Medicine
- OD, Original Disease
- PTGS2, Prostaglandin-endoperoxidase synthase 2
- SM, Supplementary Material
- SRD5A1, Steroid 5 Alpha-Reductase 1
- SRD5A2, Steroid 5 Alpha-Reductase 2
- TNFα, Tumour Necrosis Factor alpha
- UMLS, Unified Medical Language System
Collapse
Affiliation(s)
- Lucía Prieto Santamaría
- Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, 28660 Boadilla del Monte, Madrid, Spain.,ETS Ingenieros Informáticos, Universidad Politécnica de Madrid, 28660 Boadilla del Monte, Madrid, Spain.,Ezeris Networks Global Services S.L., 28028 Madrid, Spain
| | - Esther Ugarte Carro
- Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, 28660 Boadilla del Monte, Madrid, Spain
| | - Marina Díaz Uzquiano
- Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, 28660 Boadilla del Monte, Madrid, Spain
| | - Ernestina Menasalvas Ruiz
- Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, 28660 Boadilla del Monte, Madrid, Spain.,ETS Ingenieros Informáticos, Universidad Politécnica de Madrid, 28660 Boadilla del Monte, Madrid, Spain
| | | | - Alejandro Rodríguez-González
- Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, 28660 Boadilla del Monte, Madrid, Spain.,ETS Ingenieros Informáticos, Universidad Politécnica de Madrid, 28660 Boadilla del Monte, Madrid, Spain
| |
Collapse
|
31
|
Chong LC, Gandhi G, Lee JM, Yeo WWY, Choi SB. Drug Discovery of Spinal Muscular Atrophy (SMA) from the Computational Perspective: A Comprehensive Review. Int J Mol Sci 2021; 22:8962. [PMID: 34445667 PMCID: PMC8396480 DOI: 10.3390/ijms22168962] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/15/2021] [Accepted: 01/27/2021] [Indexed: 01/02/2023] Open
Abstract
Spinal muscular atrophy (SMA), one of the leading inherited causes of child mortality, is a rare neuromuscular disease arising from loss-of-function mutations of the survival motor neuron 1 (SMN1) gene, which encodes the SMN protein. When lacking the SMN protein in neurons, patients suffer from muscle weakness and atrophy, and in the severe cases, respiratory failure and death. Several therapeutic approaches show promise with human testing and three medications have been approved by the U.S. Food and Drug Administration (FDA) to date. Despite the shown promise of these approved therapies, there are some crucial limitations, one of the most important being the cost. The FDA-approved drugs are high-priced and are shortlisted among the most expensive treatments in the world. The price is still far beyond affordable and may serve as a burden for patients. The blooming of the biomedical data and advancement of computational approaches have opened new possibilities for SMA therapeutic development. This article highlights the present status of computationally aided approaches, including in silico drug repurposing, network driven drug discovery as well as artificial intelligence (AI)-assisted drug discovery, and discusses the future prospects.
Collapse
Affiliation(s)
- Li Chuin Chong
- Centre for Bioinformatics, School of Data Sciences, Perdana University, Suite 9.2, 9th Floor, Wisma Chase Perdana, Changkat Semantan, Kuala Lumpur 50490, Malaysia; (L.C.C.); (J.M.L.)
| | - Gayatri Gandhi
- Perdana University Graduate School of Medicine, Perdana University, Suite 9.2, 9th Floor, Wisma Chase Perdana, Changkat Semantan, Kuala Lumpur 50490, Malaysia; (G.G.); (W.W.Y.Y.)
| | - Jian Ming Lee
- Centre for Bioinformatics, School of Data Sciences, Perdana University, Suite 9.2, 9th Floor, Wisma Chase Perdana, Changkat Semantan, Kuala Lumpur 50490, Malaysia; (L.C.C.); (J.M.L.)
| | - Wendy Wai Yeng Yeo
- Perdana University Graduate School of Medicine, Perdana University, Suite 9.2, 9th Floor, Wisma Chase Perdana, Changkat Semantan, Kuala Lumpur 50490, Malaysia; (G.G.); (W.W.Y.Y.)
| | - Sy-Bing Choi
- Centre for Bioinformatics, School of Data Sciences, Perdana University, Suite 9.2, 9th Floor, Wisma Chase Perdana, Changkat Semantan, Kuala Lumpur 50490, Malaysia; (L.C.C.); (J.M.L.)
| |
Collapse
|
32
|
Drug Repurposing for the Management of Depression: Where Do We Stand Currently? Life (Basel) 2021; 11:life11080774. [PMID: 34440518 PMCID: PMC8398872 DOI: 10.3390/life11080774] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/29/2021] [Revised: 07/20/2021] [Accepted: 07/26/2021] [Indexed: 12/22/2022] Open
Abstract
A slow rate of new drug discovery and higher costs of new drug development attracted the attention of scientists and physicians for the repurposing and repositioning of old medications. Experimental studies and off-label use of drugs have helped drive data for further studies of approving these medications. A deeper understanding of the pathogenesis of depression encourages novel discoveries through drug repurposing and drug repositioning to treat depression. In addition to reducing neurotransmitters like epinephrine and serotonin, other mechanisms such as inflammation, insufficient blood supply, and neurotoxicants are now considered as the possible involved mechanisms. Considering the mentioned mechanisms has resulted in repurposed medications to treat treatment-resistant depression (TRD) as alternative approaches. This review aims to discuss the available treatments and their progress way during repositioning. Neurotransmitters’ antagonists, atypical antipsychotics, and CNS stimulants have been studied for the repurposing aims. However, they need proper studies in terms of formulation, matching with regulatory standards, and efficacy.
Collapse
|
33
|
Abstract
Drug repositioning is a strategy to identify new uses for existing, approved, or research drugs that are outside the scope of its original medical indication. Drug repurposing is based on the fact that one drug can act on multiple targets or that two diseases can have molecular similarities, among others. Currently, thanks to the rapid advancement of high-performance technologies, a massive amount of biological and biomedical data is being generated. This allows the use of computational methods and models based on biological networks to develop new possibilities for drug repurposing. Therefore, here, we provide an in-depth review of the main applications of drug repositioning that have been carried out using biological network models. The goal of this review is to show the usefulness of these computational methods to predict associations and to find candidate drugs for repositioning in new indications of certain diseases.
Collapse
|
34
|
Unexpected beneficial effects of drugs: an analysis of cases in the Dutch spontaneous reporting system. Eur J Clin Pharmacol 2021; 77:1543-1551. [PMID: 33884456 DOI: 10.1007/s00228-021-03142-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/09/2020] [Accepted: 04/11/2021] [Indexed: 10/21/2022]
Abstract
INTRODUCTION Drug use is inherently related to both beneficial effects on health as well as the occurrence of risks. The beneficial effects may be related to efficacy, the treatment range of a product, or even to user-friendliness of a product. However, in addition to the occurrence of adverse drug reactions, a drug can also have an unexpected beneficial effect on a patient's health, not related to the indication for which the drug was used. The aim of this article is to characterize the reports of unexpected beneficial effects of drugs in the Dutch spontaneous reporting system. METHODS A descriptive analysis was used to gain insight in number of reports and drug classes responsible for unexpected beneficial effects of drugs. Grouping of positive side effects into classes was done by a conventional qualitative content analysis of the cases. RESULTS Four hundred nine reports which described unexpected beneficial effects of drugs were included, which mentioned 451 associations between suspected drugs and unexpected beneficial effects. There were 147 drug classes on the 4th ATC level involved. Content analysis of the reports gave rise to 22 categories of unexpected beneficial effects of drugs, including one "other category". DISCUSSION AND CONCLUSION: The analysis showed a diverse spectrum of reported reactions and drugs with some categories of unexpected beneficial effects of drugs mentioned multiple times for certain drug classes on the 4th ATC level. Most of these findings are consistent with the existing literature and knowledge on the pharmacological mechanism of the drugs in question. Coding harmonization would make it possible to study these effects in international databases.
Collapse
|
35
|
Jain P, Jain SK, Jain M. Harnessing Drug Repurposing for Exploration of New Diseases: An Insight to Strategies and Case Studies. Curr Mol Med 2021; 21:111-132. [PMID: 32560606 DOI: 10.2174/1566524020666200619125404] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/20/2020] [Revised: 05/12/2020] [Accepted: 05/13/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Traditional drug discovery is time consuming, costly, and risky process. Owing to the large investment, excessive attrition, and declined output, drug repurposing has become a blooming approach for the identification and development of new therapeutics. The method has gained momentum in the past few years and has resulted in many excellent discoveries. Industries are resurrecting the failed and shelved drugs to save time and cost. The process accounts for approximately 30% of the new US Food and Drug Administration approved drugs and vaccines in recent years. METHODS A systematic literature search using appropriate keywords were made to identify articles discussing the different strategies being adopted for repurposing and various drugs that have been/are being repurposed. RESULTS This review aims to describe the comprehensive data about the various strategies (Blinded search, computational approaches, and experimental approaches) used for the repurposing along with success case studies (treatment for orphan diseases, neglected tropical disease, neurodegenerative diseases, and drugs for pediatric population). It also inculcates an elaborated list of more than 100 drugs that have been repositioned, approaches adopted, and their present clinical status. We have also attempted to incorporate the different databases used for computational repurposing. CONCLUSION The data presented is proof that drug repurposing is a prolific approach circumventing the issues poised by conventional drug discovery approaches. It is a highly promising approach and when combined with sophisticated computational tools, it also carries high precision. The review would help researches in prioritizing the drugrepositioning method much needed to flourish the drug discovery research.
Collapse
Affiliation(s)
- Priti Jain
- Department of Pharmaceutical Chemistry and Computational Chemistry, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Dhule (425405) Maharashtra, India
| | - Shreyans K Jain
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, Uttar Pradesh, India
| | - Munendra Jain
- SVKM's Department of Sciences, Narsee Monjee Institute of Management Studies, Indore, Madhya Pradesh, India
| |
Collapse
|
36
|
Chen H, Zhang Z, Zhang J. In silico drug repositioning based on the integration of chemical, genomic and pharmacological spaces. BMC Bioinformatics 2021; 22:52. [PMID: 33557749 PMCID: PMC7868667 DOI: 10.1186/s12859-021-03988-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/10/2020] [Accepted: 01/25/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Drug repositioning refers to the identification of new indications for existing drugs. Drug-based inference methods for drug repositioning apply some unique features of drugs for new indication prediction. Complementary information is provided by these different features. It is therefore necessary to integrate these features for more accurate in silico drug repositioning. RESULTS In this study, we collect 3 different types of drug features (i.e., chemical, genomic and pharmacological spaces) from public databases. Similarities between drugs are separately calculated based on each of the features. We further develop a fusion method to combine the 3 similarity measurements. We test the inference abilities of the 4 similarity datasets in drug repositioning under the guilt-by-association principle. Leave-one-out cross-validations show the integrated similarity measurement IntegratedSim receives the best prediction performance, with the highest AUC value of 0.8451 and the highest AUPR value of 0.2201. Case studies demonstrate IntegratedSim produces the largest numbers of confirmed predictions in most cases. Moreover, we compare our integration method with 3 other similarity-fusion methods using the datasets in our study. Cross-validation results suggest our method improves the prediction accuracy in terms of AUC and AUPR values. CONCLUSIONS Our study suggests that the 3 drug features used in our manuscript are valuable information for drug repositioning. The comparative results indicate that integration of the 3 drug features would improve drug-disease association prediction. Our study provides a strategy for the fusion of different drug features for in silico drug repositioning.
Collapse
Affiliation(s)
- Hailin Chen
- School of Software, East China Jiaotong University, Nanchang, 330013 China
| | - Zuping Zhang
- School of Computer Science and Engineering, Central South University, Changsha, 410083 China
| | - Jingpu Zhang
- School of Computer and Data Science, Henan University of Urban Construction, Pingdingshan, 467000 China
| |
Collapse
|
37
|
Jarada TN, Rokne JG, Alhajj R. SNF-NN: computational method to predict drug-disease interactions using similarity network fusion and neural networks. BMC Bioinformatics 2021; 22:28. [PMID: 33482713 PMCID: PMC7821180 DOI: 10.1186/s12859-020-03950-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/12/2020] [Accepted: 12/22/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Drug repositioning is an emerging approach in pharmaceutical research for identifying novel therapeutic potentials for approved drugs and discover therapies for untreated diseases. Due to its time and cost efficiency, drug repositioning plays an instrumental role in optimizing the drug development process compared to the traditional de novo drug discovery process. Advances in the genomics, together with the enormous growth of large-scale publicly available data and the availability of high-performance computing capabilities, have further motivated the development of computational drug repositioning approaches. More recently, the rise of machine learning techniques, together with the availability of powerful computers, has made the area of computational drug repositioning an area of intense activities. RESULTS In this study, a novel framework SNF-NN based on deep learning is presented, where novel drug-disease interactions are predicted using drug-related similarity information, disease-related similarity information, and known drug-disease interactions. Heterogeneous similarity information related to drugs and disease is fed to the proposed framework in order to predict novel drug-disease interactions. SNF-NN uses similarity selection, similarity network fusion, and a highly tuned novel neural network model to predict new drug-disease interactions. The robustness of SNF-NN is evaluated by comparing its performance with nine baseline machine learning methods. The proposed framework outperforms all baseline methods ([Formula: see text] = 0.867, and [Formula: see text]=0.876) using stratified 10-fold cross-validation. To further demonstrate the reliability and robustness of SNF-NN, two datasets are used to fairly validate the proposed framework's performance against seven recent state-of-the-art methods for drug-disease interaction prediction. SNF-NN achieves remarkable performance in stratified 10-fold cross-validation with [Formula: see text] ranging from 0.879 to 0.931 and [Formula: see text] from 0.856 to 0.903. Moreover, the efficiency of SNF-NN is verified by validating predicted unknown drug-disease interactions against clinical trials and published studies. CONCLUSION In conclusion, computational drug repositioning research can significantly benefit from integrating similarity measures in heterogeneous networks and deep learning models for predicting novel drug-disease interactions. The data and implementation of SNF-NN are available at http://pages.cpsc.ucalgary.ca/ tnjarada/snf-nn.php .
Collapse
Affiliation(s)
- Tamer N Jarada
- Department of Computer Science, University of Calgary, Calgary, AB, Canada
| | - Jon G Rokne
- Department of Computer Science, University of Calgary, Calgary, AB, Canada
| | - Reda Alhajj
- Department of Computer Science, University of Calgary, Calgary, AB, Canada.
- Department of Computer Engineering, Istanbul Medipol University, Istanbul, Turkey.
- Department of Health Informatics, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
38
|
Chrétien B, Jourdan JP, Davis A, Fedrizzi S, Bureau R, Sassier M, Rochais C, Alexandre J, Lelong-Boulouard V, Dolladille C, Dallemagne P. Disproportionality analysis in VigiBase as a drug repositioning method for the discovery of potentially useful drugs in Alzheimer's disease. Br J Clin Pharmacol 2020; 87:2830-2837. [PMID: 33274491 DOI: 10.1111/bcp.14690] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/11/2020] [Revised: 11/09/2020] [Accepted: 11/28/2020] [Indexed: 12/12/2022] Open
Abstract
Drug repositioning aims to propose new indications for marketed drugs. Although several methods exist, the utility of pharmacovigilance databases for this purpose is unclear. We conducted a disproportionality analysis in the World Health Organization pharmacovigilance database VigiBase to identify potential anticholinesterase drug candidates for repositioning in Alzheimer's disease (AD). METHODS Disproportionality analysis is a validated method for detecting significant associations between drugs and adverse events (AEs) in pharmacovigilance databases. We applied this approach in VigiBase to establish the safety profile displayed by the anticholinesterase drugs used in AD and searched the database for drugs with similar safety profiles. The detected drugs with potential activity against acetylcholinesterase and butyrylcholinesterases (BuChEs) were then evaluated to confirm their anticholinesterase potential. RESULTS We identified 22 drugs with safety profiles similar to AD medicines. Among these drugs, 4 (clozapine, aripiprazole, sertraline and S-duloxetine) showed a human BuChE inhibition rate of over 70% at 10-5 M. Their human BuChE half maximal inhibitory concentration values were compatible with clinical anticholinesterase action in humans at their normal doses. The most active human BuChE inhibitor in our study was S-duloxetine, with a half maximal inhibitory concentration of 1.2 μM. Combined with its ability to inhibit serotonin (5-HT) reuptake, the use of this drug could represent a novel multitarget directed ligand therapeutic strategy for AD. CONCLUSION We identified 4 drugs with repositioning potential in AD using drug safety profiles derived from a pharmacovigilance database. This method could be useful for future drug repositioning efforts.
Collapse
Affiliation(s)
- Basile Chrétien
- Department of Pharmacology, Caen University Hospital, Caen, F-14000, France.,Pharmacovigilance Regional Center, Caen University Hospital, Caen, F-14000, France
| | - Jean-Pierre Jourdan
- Department of Pharmacy, Caen University Hospital, Caen, F-14000, France.,Centre d'Etudes et de Recherche sur le Médicament de Normandie (CERMN), Normandie Univ, UNICAEN, Caen, F-14000, France
| | - Audrey Davis
- Centre d'Etudes et de Recherche sur le Médicament de Normandie (CERMN), Normandie Univ, UNICAEN, Caen, F-14000, France
| | - Sophie Fedrizzi
- Department of Pharmacology, Caen University Hospital, Caen, F-14000, France.,Pharmacovigilance Regional Center, Caen University Hospital, Caen, F-14000, France
| | - Ronan Bureau
- Centre d'Etudes et de Recherche sur le Médicament de Normandie (CERMN), Normandie Univ, UNICAEN, Caen, F-14000, France
| | - Marion Sassier
- Department of Pharmacology, Caen University Hospital, Caen, F-14000, France.,Pharmacovigilance Regional Center, Caen University Hospital, Caen, F-14000, France
| | - Christophe Rochais
- Centre d'Etudes et de Recherche sur le Médicament de Normandie (CERMN), Normandie Univ, UNICAEN, Caen, F-14000, France
| | - Joachim Alexandre
- Department of Pharmacology, Caen University Hospital, Caen, F-14000, France.,Pharmacovigilance Regional Center, Caen University Hospital, Caen, F-14000, France.,EA4650, Signalisation, électrophysiologie et imagerie des lésions d'ischémie-reperfusion myocardique, Normandie Univ, UNICAEN, Caen, 14000, France
| | - Véronique Lelong-Boulouard
- Department of Pharmacology, Caen University Hospital, Caen, F-14000, France.,INSERM UMR 1075, COMETE-MOBILITES "Vieillissement, Pathologie, Santé", Normandie Univ, UNICAEN, Caen, 14000, France
| | - Charles Dolladille
- Department of Pharmacology, Caen University Hospital, Caen, F-14000, France.,EA4650, Signalisation, électrophysiologie et imagerie des lésions d'ischémie-reperfusion myocardique, Normandie Univ, UNICAEN, Caen, 14000, France
| | - Patrick Dallemagne
- Centre d'Etudes et de Recherche sur le Médicament de Normandie (CERMN), Normandie Univ, UNICAEN, Caen, F-14000, France
| |
Collapse
|
39
|
Yang M, Huang L, Xu Y, Lu C, Wang J. Heterogeneous graph inference with matrix completion for computational drug repositioning. Bioinformatics 2020; 36:5456-5464. [PMID: 33331887 DOI: 10.1093/bioinformatics/btaa1024] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/09/2020] [Revised: 11/23/2020] [Accepted: 11/26/2020] [Indexed: 12/24/2022] Open
Abstract
MOTIVATION Emerging evidence presents that traditional drug discovery experiment is time-consuming and high costs. Computational drug repositioning plays a critical role in saving time and resources for drug research and discovery. Therefore, developing more accurate and efficient approaches is imperative. Heterogeneous graph inference is a classical method in computational drug repositioning, which not only has high convergence precision, but also has fast convergence speed. However, the method has not fully considered the sparsity of heterogeneous association network. In addition, rough similarity measure can reduce the performance in identifying drug-associated indications. RESULTS In this article, we propose a heterogeneous graph inference with matrix completion (HGIMC) method to predict potential indications for approved and novel drugs. First, we use a bounded matrix completion (BMC) model to prefill a part of the missing entries in original drug-disease association matrix. This step can add more positive and formative drug-disease edges between drug network and disease network. Second, Gaussian radial basis function (GRB) is employed to improve the drug and disease similarities since the performance of heterogeneous graph inference more relies on similarity measures. Next, based on the updated drug-disease associations and new similarity measures of drug and disease, we construct a novel heterogeneous drug-disease network. Finally, HGIMC utilizes the heterogeneous network to infer the scores of unknown association pairs, and then recommend the promising indications for drugs. To evaluate the performance of our method, HGIMC is compared with five state-of-the-art approaches of drug repositioning in the 10-fold cross-validation and de novo tests. As the numerical results shown, HGIMC not only achieves a better prediction performance, but also has an excellent computation efficiency. In addition, cases studies also confirm the effectiveness of our method in practical application. AVAILABILITY The HGIMC software is freely available at https://github.com/BioinformaticsCSU/HGIMC, https://hub.docker.com/repository/docker/yangmy84/hgimc, and http://doi.org/10.5281/zenodo.4285640. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Mengyun Yang
- The Hunan Provincial Key Lab of Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha, P.R.China.,School of Science, Shaoyang University, Shaoyang, P.R.China
| | - Lan Huang
- The Hunan Provincial Key Lab of Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha, P.R.China
| | - Yunpei Xu
- The Hunan Provincial Key Lab of Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha, P.R.China
| | - Chengqian Lu
- The Hunan Provincial Key Lab of Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha, P.R.China
| | - Jianxin Wang
- The Hunan Provincial Key Lab of Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha, P.R.China
| |
Collapse
|
40
|
Yu Z, Huang F, Zhao X, Xiao W, Zhang W. Predicting drug-disease associations through layer attention graph convolutional network. Brief Bioinform 2020; 22:5918381. [PMID: 33078832 DOI: 10.1093/bib/bbaa243] [Citation(s) in RCA: 164] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/29/2020] [Revised: 08/16/2020] [Accepted: 08/31/2020] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Determining drug-disease associations is an integral part in the process of drug development. However, the identification of drug-disease associations through wet experiments is costly and inefficient. Hence, the development of efficient and high-accuracy computational methods for predicting drug-disease associations is of great significance. RESULTS In this paper, we propose a novel computational method named as layer attention graph convolutional network (LAGCN) for the drug-disease association prediction. Specifically, LAGCN first integrates the known drug-disease associations, drug-drug similarities and disease-disease similarities into a heterogeneous network, and applies the graph convolution operation to the network to learn the embeddings of drugs and diseases. Second, LAGCN combines the embeddings from multiple graph convolution layers using an attention mechanism. Third, the unobserved drug-disease associations are scored based on the integrated embeddings. Evaluated by 5-fold cross-validations, LAGCN achieves an area under the precision-recall curve of 0.3168 and an area under the receiver-operating characteristic curve of 0.8750, which are better than the results of existing state-of-the-art prediction methods and baseline methods. The case study shows that LAGCN can discover novel associations that are not curated in our dataset. CONCLUSION LAGCN is a useful tool for predicting drug-disease associations. This study reveals that embeddings from different convolution layers can reflect the proximities of different orders, and combining the embeddings by the attention mechanism can improve the prediction performances.
Collapse
Affiliation(s)
- Zhouxin Yu
- College of Informatics, Huazhong Agricultural University
| | - Feng Huang
- College of Informatics, Huazhong Agricultural University
| | - Xiaohan Zhao
- College of Informatics, Huazhong Agricultural University
| | | | - Wen Zhang
- College of Informatics, Huazhong Agricultural University
| |
Collapse
|
41
|
He S, Wen Y, Yang X, Liu Z, Song X, Huang X, Bo X. PIMD: An Integrative Approach for Drug Repositioning Using Multiple Characterization Fusion. GENOMICS PROTEOMICS & BIOINFORMATICS 2020; 18:565-581. [PMID: 33075523 PMCID: PMC8377380 DOI: 10.1016/j.gpb.2018.10.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Academic Contribution Register] [Received: 07/09/2018] [Revised: 09/21/2018] [Accepted: 10/10/2018] [Indexed: 11/28/2022]
Abstract
The accumulation of various types of drug informatics data and computational approaches for drug repositioning can accelerate pharmaceutical research and development. However, the integration of multi-dimensional drug data for precision repositioning remains a pressing challenge. Here, we propose a systematic framework named PIMD to predict drug therapeutic properties by integrating multi-dimensional data for drug repositioning. In PIMD, drug similarity networks (DSNs) based on chemical, pharmacological, and clinical data are fused into an integrated DSN (iDSN) composed of many clusters. Rather than simple fusion, PIMD offers a systematic way to annotate clusters. Unexpected drugs within clusters and drug pairs with a high iDSN similarity score are therefore identified to predict novel therapeutic uses. PIMD provides new insights into the universality, individuality, and complementarity of different drug properties by evaluating the contribution of each property data. To test the performance of PIMD, we use chemical, pharmacological, and clinical properties to generate an iDSN. Analyses of the contributions of each drug property indicate that this iDSN was driven by all data types and performs better than other DSNs. Within the top 20 recommended drug pairs, 7 drugs have been reported to be repurposed. The source code for PIMD is available at https://github.com/Sepstar/PIMD/.
Collapse
Affiliation(s)
- Song He
- Department of Biotechnology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Yuqi Wen
- Department of Biotechnology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Xiaoxi Yang
- Department of Biotechnology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Zhen Liu
- Department of Biotechnology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Xinyu Song
- Department of Biotechnology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Xin Huang
- Department of Biotechnology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Xiaochen Bo
- Department of Biotechnology, Beijing Institute of Radiation Medicine, Beijing 100850, China.
| |
Collapse
|
42
|
Rymbai E, Sugumar D, Saravanan J, Divakar S. Ropinirole, a potential drug for systematic repositioning based on side effect profile for management and treatment of breast cancer. Med Hypotheses 2020; 144:110156. [PMID: 32763725 DOI: 10.1016/j.mehy.2020.110156] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/02/2020] [Revised: 07/18/2020] [Accepted: 07/29/2020] [Indexed: 01/11/2023]
Abstract
Drug repositioning offers two main advantages in drug discovery - the process is less tedious and less costly. In the past, many drugs like thalidomide and sildenafil were successfully repositioned but the process was entirely serendipitous. These days drug repositioning is widely accepted as an alternate method of drug discovery and the process is based on building a strong hypothesis guided by systematic computational and experimental methods. One of the methods used in drug repositioning is based on shared side effects by drugs of different pharmacological categories. This method rests on the principle that drugs that share side effects might also share common biological targets and therefore same pharmacological indications. Old drugs can be repositioned for new uses by identifying the shared side effects of existing drugs and by modulating their chemical structure if required. Breast cancer is the most common type of cancer in women and the second leading cause of death worldwide after lung cancer in both men and women. Letrozole, an aromatase inhibitor, is used in the treatment of advanced, recurrent and metastatic breast cancer in post-menopausal women. Identification of drugs that share side effects with letrozole might help us to identify a potential drug for repositioning in the treatment of breast cancer. Ropinirole, a dopaminergic agonist was found to share the maximum number of side effects with letrozole. Studies have proposed that dopaminergic agonists induce apoptosis in breast, colon, ovarian cancer cells and leukemia neuroblastoma. This is consistent with our hypothesis that ropinirole that shares the maximum number of side effects with letrozole might be effective in the management of breast cancer. This hypothesis was further validated by preliminary molecular docking and in-vitro cell-line studies.
Collapse
Affiliation(s)
- Emdormi Rymbai
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, The Nilgiris, Tamil Nadu, India
| | - Deepa Sugumar
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, The Nilgiris, Tamil Nadu, India
| | - J Saravanan
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, The Nilgiris, Tamil Nadu, India.
| | - S Divakar
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, The Nilgiris, Tamil Nadu, India
| |
Collapse
|
43
|
Jarada TN, Rokne JG, Alhajj R. A review of computational drug repositioning: strategies, approaches, opportunities, challenges, and directions. J Cheminform 2020; 12:46. [PMID: 33431024 PMCID: PMC7374666 DOI: 10.1186/s13321-020-00450-7] [Citation(s) in RCA: 170] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/19/2020] [Accepted: 07/13/2020] [Indexed: 01/13/2023] Open
Abstract
Drug repositioning is the process of identifying novel therapeutic potentials for existing drugs and discovering therapies for untreated diseases. Drug repositioning, therefore, plays an important role in optimizing the pre-clinical process of developing novel drugs by saving time and cost compared to the traditional de novo drug discovery processes. Since drug repositioning relies on data for existing drugs and diseases the enormous growth of publicly available large-scale biological, biomedical, and electronic health-related data along with the high-performance computing capabilities have accelerated the development of computational drug repositioning approaches. Multidisciplinary researchers and scientists have carried out numerous attempts, with different degrees of efficiency and success, to computationally study the potential of repositioning drugs to identify alternative drug indications. This study reviews recent advancements in the field of computational drug repositioning. First, we highlight different drug repositioning strategies and provide an overview of frequently used resources. Second, we summarize computational approaches that are extensively used in drug repositioning studies. Third, we present different computing and experimental models to validate computational methods. Fourth, we address prospective opportunities, including a few target areas. Finally, we discuss challenges and limitations encountered in computational drug repositioning and conclude with an outline of further research directions.
Collapse
Affiliation(s)
- Tamer N Jarada
- Department of Computer Science, University of Calgary, Calgary, Alberta, Canada
| | - Jon G Rokne
- Department of Computer Science, University of Calgary, Calgary, Alberta, Canada
| | - Reda Alhajj
- Department of Computer Science, University of Calgary, Calgary, Alberta, Canada.
- Department of Computer Engineering, Istanbul Medipol University, Istanbul, Turkey.
| |
Collapse
|
44
|
Zhu Y, Che C, Jin B, Zhang N, Su C, Wang F. Knowledge-driven drug repurposing using a comprehensive drug knowledge graph. Health Informatics J 2020; 26:2737-2750. [DOI: 10.1177/1460458220937101] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/13/2023]
Abstract
Due to the huge costs associated with new drug discovery and development, drug repurposing has become an important complement to the traditional de novo approach. With the increasing number of public databases and the rapid development of analytical methodologies, computational approaches have gained great momentum in the field of drug repurposing. In this study, we introduce an approach to knowledge-driven drug repurposing based on a comprehensive drug knowledge graph. We design and develop a drug knowledge graph by systematically integrating multiple drug knowledge bases. We describe path- and embedding-based data representation methods of transforming information in the drug knowledge graph into valuable inputs to allow machine learning models to predict drug repurposing candidates. The evaluation demonstrates that the knowledge-driven approach can produce high predictive results for known diabetes mellitus treatments by only using treatment information on other diseases. In addition, this approach supports exploratory investigation through the review of meta paths that connect drugs with diseases. This knowledge-driven approach is an effective drug repurposing strategy supporting large-scale prediction and the investigation of case studies.
Collapse
Affiliation(s)
| | | | - Bo Jin
- Dalian University of Technology, China
| | | | | | | |
Collapse
|
45
|
Durai P, Ko YJ, Pan CH, Park K. Evolutionary chemical binding similarity approach integrated with 3D-QSAR method for effective virtual screening. BMC Bioinformatics 2020; 21:309. [PMID: 32664863 PMCID: PMC7362480 DOI: 10.1186/s12859-020-03643-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/11/2020] [Accepted: 07/03/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Despite continued efforts using chemical similarity methods in virtual screening, currently developed approaches suffer from time-consuming multistep procedures and low success rates. We recently developed a machine learning-based chemical binding similarity model considering common structural features from molecules binding to the same, or evolutionarily related targets. The chemical binding similarity measures the resemblance of chemical compounds in terms of binding site similarity to better describe functional similarities that arise from target binding. In this study, we have shown how the chemical binding similarity could be used in virtual screening together with the conventional structure-based methods. RESULTS The chemical binding similarity, receptor-based pharmacophore, chemical structure similarity, and molecular docking methods were evaluated to identify an effective virtual screening procedure for desired target proteins. When we tested the chemical binding similarity method with test sets of 51 kinases, it outperformed the traditional structural similarity-based methods as well as structure-based methods, such as molecular docking and receptor-based pharmacophore modeling, in terms of finding active compounds. We further validated the results by performing virtual screening (using the chemical binding similarity and receptor-based pharmacophore methods) against a completely blind dataset for mitogen-activated protein kinase kinase 1 (MEK1), ephrin type-B receptor 4 (EPHB4) and wee1-like protein kinase (WEE1). The in vitro kinase binding assay confirmed that 6 out of 13 (46.2%) for MEK1 and 2 out of 12 (16.7%) for EPHB4 were newly identified only by the chemical binding similarity model. CONCLUSIONS We report that the virtual screening results could further be improved by combining the chemical binding similarity model with 3D-QSAR pharmacophore and molecular docking models. Not only the new inhibitors are identified in this study, but also many of the identified molecules have low structural similarity scores against already reported inhibitors and that show the revelation of novel scaffolds.
Collapse
Affiliation(s)
- Prasannavenkatesh Durai
- Natural Product Informatics Research Center, KIST Gangneung Institute of Natural Products, Gangneung, 25451, Republic of Korea
| | - Young-Joon Ko
- Natural Product Informatics Research Center, KIST Gangneung Institute of Natural Products, Gangneung, 25451, Republic of Korea
- Department of Bioinformatics and Life Science, Soongsil University, Seoul, 06978, Republic of Korea
| | - Cheol-Ho Pan
- Natural Product Informatics Research Center, KIST Gangneung Institute of Natural Products, Gangneung, 25451, Republic of Korea
| | - Keunwan Park
- Natural Product Informatics Research Center, KIST Gangneung Institute of Natural Products, Gangneung, 25451, Republic of Korea.
| |
Collapse
|
46
|
Valli D, Gruszka AM, Alcalay M. Has Drug Repurposing Fulfilled its Promise in Acute Myeloid Leukaemia? J Clin Med 2020; 9:E1892. [PMID: 32560371 PMCID: PMC7356362 DOI: 10.3390/jcm9061892] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/11/2020] [Revised: 06/11/2020] [Accepted: 06/15/2020] [Indexed: 12/16/2022] Open
Abstract
Drug repurposing is a method of drug discovery that consists of finding a new therapeutic context for an old drug. Compound identification arises from screening of large libraries of active compounds, through interrogating databases of cell line gene expression response upon treatment or by merging several types of information concerning disease-drug relationships. Although, there is a general consensus on the potential and advantages of this drug discovery modality, at the practical level to-date no non-anti-cancer repurposed compounds have been introduced into standard acute myeloid leukaemia (AML) management, albeit that preclinical validation yielded several candidates. The review presents the state-of-the-art drug repurposing approach in AML and poses the question of what has to be done in order to take a full advantage of it, both at the stage of screening design and later when progressing from the preclinical to the clinical phases of drug development. We argue that improvements are needed to model and read-out systems as well as to screening technologies, but also to more funding and trust in drug repurposing strategies.
Collapse
Affiliation(s)
- Debora Valli
- Department of Experimental Oncology, Istituto Europeo di Oncologia IRCCS, Via Adamello 16, 20 139 Milan, Italy; (D.V.); (M.A.)
| | - Alicja M. Gruszka
- Department of Experimental Oncology, Istituto Europeo di Oncologia IRCCS, Via Adamello 16, 20 139 Milan, Italy; (D.V.); (M.A.)
| | - Myriam Alcalay
- Department of Experimental Oncology, Istituto Europeo di Oncologia IRCCS, Via Adamello 16, 20 139 Milan, Italy; (D.V.); (M.A.)
- Department of Oncology and Hemato-Oncology, University of Milan, Via Festa del Perdono 7, 20 122 Milan, Italy
| |
Collapse
|
47
|
Li X, Rousseau JF, Ding Y, Song M, Lu W. Understanding Drug Repurposing From the Perspective of Biomedical Entities and Their Evolution: Bibliographic Research Using Aspirin. JMIR Med Inform 2020; 8:e16739. [PMID: 32543442 PMCID: PMC7327595 DOI: 10.2196/16739] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/19/2019] [Revised: 01/08/2020] [Accepted: 03/31/2020] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Drug development is still a costly and time-consuming process with a low rate of success. Drug repurposing (DR) has attracted significant attention because of its significant advantages over traditional approaches in terms of development time, cost, and safety. Entitymetrics, defined as bibliometric indicators based on biomedical entities (eg, diseases, drugs, and genes) studied in the biomedical literature, make it possible for researchers to measure knowledge evolution and the transfer of drug research. OBJECTIVE The purpose of this study was to understand DR from the perspective of biomedical entities (diseases, drugs, and genes) and their evolution. METHODS In the work reported in this paper, we extended the bibliometric indicators of biomedical entities mentioned in PubMed to detect potential patterns of biomedical entities in various phases of drug research and investigate the factors driving DR. We used aspirin (acetylsalicylic acid) as the subject of the study since it can be repurposed for many applications. We propose 4 easy, transparent measures based on entitymetrics to investigate DR for aspirin: Popularity Index (P1), Promising Index (P2), Prestige Index (P3), and Collaboration Index (CI). RESULTS We found that the maxima of P1, P3, and CI are closely associated with the different repurposing phases of aspirin. These metrics enabled us to observe the way in which biomedical entities interacted with the drug during the various phases of DR and to analyze the potential driving factors for DR at the entity level. P1 and CI were indicative of the dynamic trends of a specific biomedical entity over a long time period, while P2 was more sensitive to immediate changes. P3 reflected the early signs of the practical value of biomedical entities and could be valuable for tracking the research frontiers of a drug. CONCLUSIONS In-depth studies of side effects and mechanisms, fierce market competition, and advanced life science technologies are driving factors for DR. This study showcases the way in which researchers can examine the evolution of DR using entitymetrics, an approach that can be valuable for enhancing decision making in the field of drug discovery and development.
Collapse
Affiliation(s)
- Xin Li
- Information Retrieval and Knowledge Mining Laboratory, School of Information Management, Wuhan University, Wuhan, China.,School of Informatics, Computing, and Engineering, Indiana University, Bloomington, IN, United States
| | - Justin F Rousseau
- Department of Population Health and Department of Neurology, Dell Medical School, The University of Texas at Austin, Austin, TX, United States
| | - Ying Ding
- School of Information, Dell Medical School, The University of Texas Austin, Austin, TX, United States
| | - Min Song
- Department of Library and Information Science, Yonsei University, Seoul, Republic of Korea
| | - Wei Lu
- Information Retrieval and Knowledge Mining Laboratory, School of Information Management, Wuhan University, Wuhan, China
| |
Collapse
|
48
|
Wu Y, Warner JL, Wang L, Jiang M, Xu J, Chen Q, Nian H, Dai Q, Du X, Yang P, Denny JC, Liu H, Xu H. Discovery of Noncancer Drug Effects on Survival in Electronic Health Records of Patients With Cancer: A New Paradigm for Drug Repurposing. JCO Clin Cancer Inform 2020; 3:1-9. [PMID: 31141421 PMCID: PMC6693869 DOI: 10.1200/cci.19.00001] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/22/2022] Open
Abstract
PURPOSE Drug development is becoming increasingly expensive and time consuming. Drug repurposing is one potential solution to accelerate drug discovery. However, limited research exists on the use of electronic health record (EHR) data for drug repurposing, and most published studies have been conducted in a hypothesis-driven manner that requires a predefined hypothesis about drugs and new indications. Whether EHRs can be used to detect drug repurposing signals is not clear. We want to demonstrate the feasibility of mining large, longitudinal EHRs for drug repurposing by detecting candidate noncancer drugs that can potentially be used for the treatment of cancer. PATIENTS AND METHODS By linking cancer registry data to EHRs, we identified 43,310 patients with cancer treated at Vanderbilt University Medical Center (VUMC) and 98,366 treated at the Mayo Clinic. We assessed the effect of 146 noncancer drugs on cancer survival using VUMC EHR data and sought to replicate significant associations (false discovery rate < .1) using the identical approach with Mayo Clinic EHR data. To evaluate replicated signals further, we reviewed the biomedical literature and clinical trials on cancers for corroborating evidence. RESULTS We identified 22 drugs from six drug classes (statins, proton pump inhibitors, angiotensin-converting enzyme inhibitors, β-blockers, nonsteroidal anti-inflammatory drugs, and α-1 blockers) associated with improved overall cancer survival (false discovery rate < .1) from VUMC; nine of the 22 drug associations were replicated at the Mayo Clinic. Literature and cancer clinical trial evaluations also showed very strong evidence to support the repurposing signals from EHRs. CONCLUSION Mining of EHRs for drug exposure–mediated survival signals is feasible and identifies potential candidates for antineoplastic repurposing. This study sets up a new model of mining EHRs for drug repurposing signals.
Collapse
Affiliation(s)
- Yonghui Wu
- The University of Texas Health Science Center at Houston, Houston, TX.,University of Florida, Gainesville, FL
| | | | | | - Min Jiang
- The University of Texas Health Science Center at Houston, Houston, TX
| | - Jun Xu
- The University of Texas Health Science Center at Houston, Houston, TX
| | - Qingxia Chen
- Vanderbilt University Medical Center, Nashville, TN
| | - Hui Nian
- Vanderbilt University Medical Center, Nashville, TN
| | - Qi Dai
- Vanderbilt University Medical Center, Nashville, TN.,Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN
| | - Xianglin Du
- The University of Texas Health Science Center at Houston, Houston, TX
| | | | | | | | - Hua Xu
- The University of Texas Health Science Center at Houston, Houston, TX
| |
Collapse
|
49
|
Machine-Learning Prediction of Oral Drug-Induced Liver Injury (DILI) via Multiple Features and Endpoints. BIOMED RESEARCH INTERNATIONAL 2020; 2020:4795140. [PMID: 32509859 PMCID: PMC7254069 DOI: 10.1155/2020/4795140] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Academic Contribution Register] [Received: 01/21/2020] [Accepted: 04/17/2020] [Indexed: 12/17/2022]
Abstract
Drug discovery is a costly process which usually takes more than 10 years and billions of dollars for one successful drug to enter the market. Despite all the safety tests, drugs may still cause adverse reactions and be restricted in use or even withdrawn from the market. Drug-induced liver injury (DILI) is one of the major adverse drug reactions, and computational models may be used to predict and reduce it. To assess the computational prediction performance of DILI, we curated DILI endpoints from three databases and prepared drug features including chemical descriptors, therapeutic classifications, gene expressions, and binding proteins. We trained machine-learning models to predict the various DILI endpoints using different drug features. Using the optimal feature sets, the top-performing models obtained areas under the receiver operating characteristic curve (AUC) around 0.8 for some DILI endpoints. We found that some features, including therapeutic classifications and proteins, have good prediction performance towards DILI. We also discovered that the severity of DILI endpoints as well as the selection of negative samples may significantly affect the prediction results. Overall, our study provided a comprehensive collection, curation, and prediction of DILI endpoints using various drug features, which may help the drug researchers to better understand and prevent DILI during the drug discovery process.
Collapse
|
50
|
Emon MA, Domingo-Fernández D, Hoyt CT, Hofmann-Apitius M. PS4DR: a multimodal workflow for identification and prioritization of drugs based on pathway signatures. BMC Bioinformatics 2020; 21:231. [PMID: 32503412 PMCID: PMC7275349 DOI: 10.1186/s12859-020-03568-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/30/2019] [Accepted: 05/28/2020] [Indexed: 12/21/2022] Open
Abstract
Background During the last decade, there has been a surge towards computational drug repositioning owing to constantly increasing -omics data in the biomedical research field. While numerous existing methods focus on the integration of heterogeneous data to propose candidate drugs, it is still challenging to substantiate their results with mechanistic insights of these candidate drugs. Therefore, there is a need for more innovative and efficient methods which can enable better integration of data and knowledge for drug repositioning. Results Here, we present a customizable workflow (PS4DR) which not only integrates high-throughput data such as genome-wide association study (GWAS) data and gene expression signatures from disease and drug perturbations but also takes pathway knowledge into consideration to predict drug candidates for repositioning. We have collected and integrated publicly available GWAS data and gene expression signatures for several diseases and hundreds of FDA-approved drugs or those under clinical trial in this study. Additionally, different pathway databases were used for mechanistic knowledge integration in the workflow. Using this systematic consolidation of data and knowledge, the workflow computes pathway signatures that assist in the prediction of new indications for approved and investigational drugs. Conclusion We showcase PS4DR with applications demonstrating how this tool can be used for repositioning and identifying new drugs as well as proposing drugs that can simulate disease dysregulations. We were able to validate our workflow by demonstrating its capability to predict FDA-approved drugs for their known indications for several diseases. Further, PS4DR returned many potential drug candidates for repositioning that were backed up by epidemiological evidence extracted from scientific literature. Source code is freely available at https://github.com/ps4dr/ps4dr.
Collapse
Affiliation(s)
- Mohammad Asif Emon
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (Fraunhofer SCAI), 53757, Sankt Augustin, Germany. .,Bonn-Aachen International Center for IT, Rheinische Friedrich-Wilhelms-Universität Bonn, 53117, Bonn, Germany.
| | - Daniel Domingo-Fernández
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (Fraunhofer SCAI), 53757, Sankt Augustin, Germany. .,Bonn-Aachen International Center for IT, Rheinische Friedrich-Wilhelms-Universität Bonn, 53117, Bonn, Germany.
| | - Charles Tapley Hoyt
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (Fraunhofer SCAI), 53757, Sankt Augustin, Germany.,Bonn-Aachen International Center for IT, Rheinische Friedrich-Wilhelms-Universität Bonn, 53117, Bonn, Germany
| | - Martin Hofmann-Apitius
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (Fraunhofer SCAI), 53757, Sankt Augustin, Germany.,Bonn-Aachen International Center for IT, Rheinische Friedrich-Wilhelms-Universität Bonn, 53117, Bonn, Germany
| |
Collapse
|