1
|
Calzetta L, Pistocchini E, Gholamalishahi S, Grugni L, Cazzola M, Rogliani P. Novel drug discovery strategies for chronic obstructive pulmonary disease: the latest developments. Expert Opin Drug Discov 2025; 20:683-692. [PMID: 40223433 DOI: 10.1080/17460441.2025.2490251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 04/03/2025] [Indexed: 04/15/2025]
Abstract
INTRODUCTION The journey from initial drug discovery to approval for respiratory diseases typically spans approximately 10.4 years and cost over $2.8 billion. This intricate process involves five stages: target identification, therapeutic molecule discovery, preclinical testing, clinical trials, and regulatory approval. AREAS COVERED This review examines novel drug discovery strategies for chronic obstructive pulmonary disease (COPD), focusing on advanced in vitro models that replicate human lung conditions for accurate drug testing according to the following search string: discovery AND strategy AND COPD. It explores targeted molecular therapies, structure-based drug design, and drug repurposing approaches facilitated by computational analysis. The significance of personalized medicine in tailoring treatments for diverse COPDs is emphasized, highlighting the complexity of the disease and the necessity of these innovative methodologies to improve therapeutic outcomes. EXPERT OPINION COPD remains a challenging area, with a significant unmet medical need. Despite previous efforts, few effective therapies exist. Innovative in vitro models, targeted molecular therapies, and drug repurposing strategies are showing promise. Emphasizing advanced preclinical models and repurposing existing drugs could transform treatment paradigms, promoting more effective therapies for complex diseases like COPD. These innovations hold potential for enhancing drug discovery efficiency, leading to personalized and precision medicine approaches.
Collapse
Affiliation(s)
- Luigino Calzetta
- Respiratory Disease and Lung Function Unit, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Elena Pistocchini
- Unit of Respiratory Medicine, Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Shima Gholamalishahi
- Unit of Respiratory Medicine, Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Lucia Grugni
- Unit of Respiratory Medicine, Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Mario Cazzola
- Unit of Respiratory Medicine, Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Paola Rogliani
- Unit of Respiratory Medicine, Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy
| |
Collapse
|
2
|
Yu SI, Chang YK, Sheu ML, Tseng YH. Thyroiditis and Thyroid Cancer: Bioinformatics Analysis of Gene Expression Data. In Vivo 2024; 38:2205-2213. [PMID: 39187346 PMCID: PMC11363792 DOI: 10.21873/invivo.13684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 06/19/2024] [Accepted: 07/03/2024] [Indexed: 08/28/2024]
Abstract
BACKGROUND/AIM Hashimoto thyroiditis (HT) association with thyroid lymphoma is well established; however, the association with papillary thyroid cancer (PTC) is still unclear. Thyroid cancer incidence has shown an increasing trend in recent years. It is characterized by slow growth, making it generally amenable to successful treatment. MATERIALS AND METHODS We aimed to identify genes considered as promising biomarkers of the progression from thyroiditis to thyroid cancer in public gene expression datasets. RESULTS We identified 70 differentially expressed genes (DEGs) and used them to prioritize biological risk genes for thyroiditis and thyroid cancer. Statistics and a scoring system based on six functional annotations of significant biological impact identified four genes of interest: CXCR4, IL6ST, PPARG and TP53. Kaplan-Meier plots were used to assess the expression levels related to overall survival. Furthermore, a manual bibliographic search was carried out for each gene, and a protein-protein interaction (PPI) network was built to verify their known associations. CONCLUSION The results showed that all four genes (CXCR4, IL6ST, PPARG, TP53) were highly relevant to thyroiditis and thyroid cancer, thus making them worthy of further investigation to understand their relationship with these two diseases.
Collapse
Affiliation(s)
- Szu-I Yu
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan, R.O.C
- Department of Medical Research, Tungs' Taichung MetroHarbor Hospital, Taichung, Taiwan, R.O.C
| | - Yu-Kang Chang
- Department of Medical Research, Tungs' Taichung MetroHarbor Hospital, Taichung, Taiwan, R.O.C
| | - Meei-Ling Sheu
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan, R.O.C.;
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan, R.O.C
- Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, Taiwan, R.O.C
| | - Yao-Hsien Tseng
- Department of Endocrinology and Metabolism, Tungs' Taichung MetroHarbor Hospital, Taichung, Taiwan, R.O.C.
| |
Collapse
|
3
|
Jiménez A, Merino MJ, Parras J, Zazo S. Explainable drug repurposing via path based knowledge graph completion. Sci Rep 2024; 14:16587. [PMID: 39025897 PMCID: PMC11258358 DOI: 10.1038/s41598-024-67163-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 07/09/2024] [Indexed: 07/20/2024] Open
Abstract
Drug repurposing aims to find new therapeutic applications for existing drugs in the pharmaceutical market, leading to significant savings in time and cost. The use of artificial intelligence and knowledge graphs to propose repurposing candidates facilitates the process, as large amounts of data can be processed. However, it is important to pay attention to the explainability needed to validate the predictions. We propose a general architecture to understand several explainable methods for graph completion based on knowledge graphs and design our own architecture for drug repurposing. We present XG4Repo (eXplainable Graphs for Repurposing), a framework that takes advantage of the connectivity of any biomedical knowledge graph to link compounds to the diseases they can treat. Our method allows methapaths of different types and lengths, which are automatically generated and optimised based on data. XG4Repo focuses on providing meaningful explanations to the predictions, which are based on paths from compounds to diseases. These paths include nodes such as genes, pathways, side effects, or anatomies, so they provide information about the targets and other characteristics of the biomedical mechanism that link compounds and diseases. Paths make predictions interpretable for experts who can validate them and use them in further research on drug repurposing. We also describe three use cases where we analyse new uses for Epirubicin, Paclitaxel, and Predinisone and present the paths that support the predictions.
Collapse
Affiliation(s)
- Ana Jiménez
- Information Processing and Telecommunications Center, Universidad Politécnica de Madrid, ETSI Telecomunicación, Avda. Complutense, 30, 28040, Madrid, Spain
| | - María José Merino
- Information Processing and Telecommunications Center, Universidad Politécnica de Madrid, ETSI Telecomunicación, Avda. Complutense, 30, 28040, Madrid, Spain
| | - Juan Parras
- Information Processing and Telecommunications Center, Universidad Politécnica de Madrid, ETSI Telecomunicación, Avda. Complutense, 30, 28040, Madrid, Spain.
| | - Santiago Zazo
- Information Processing and Telecommunications Center, Universidad Politécnica de Madrid, ETSI Telecomunicación, Avda. Complutense, 30, 28040, Madrid, Spain
| |
Collapse
|
4
|
Satish KS, Saraswathy GR, Ritesh G, Saravanan KS, Krishnan A, Bhargava J, Ushnaa K, Dsouza PL. Exploring cutting-edge strategies for drug repurposing in female cancers - An insight into the tools of the trade. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2024; 207:355-415. [PMID: 38942544 DOI: 10.1016/bs.pmbts.2024.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/30/2024]
Abstract
Female cancers, which include breast and gynaecological cancers, represent a significant global health burden for women. Despite advancements in research pertinent to unearthing crucial pathological characteristics of these cancers, challenges persist in discovering potential therapeutic strategies. This is further exacerbated by economic burdens associated with de novo drug discovery and clinical intricacies such as development of drug resistance and metastasis. Drug repurposing, an innovative approach leveraging existing FDA-approved drugs for new indications, presents a promising avenue to expedite therapeutic development. Computational techniques, including virtual screening and analysis of drug-target-disease relationships, enable the identification of potential candidate drugs. Integration of diverse data types, such as omics and clinical information, enhances the precision and efficacy of drug repurposing strategies. Experimental approaches, including high-throughput screening assays, in vitro, and in vivo models, complement computational methods, facilitating the validation of repurposed drugs. This review highlights various target mining strategies based on analysis of differential gene expression, weighted gene co-expression, protein-protein interaction network, and host-pathogen interaction, among others. To unearth drug candidates, the technicalities of leveraging information from databases such as DrugBank, STITCH, LINCS, and ChEMBL, among others are discussed. Further in silico validation techniques encompassing molecular docking, pharmacophore modelling, molecular dynamic simulations, and ADMET analysis are elaborated. Overall, this review delves into the exploration of individual case studies to offer a wide perspective of the ever-evolving field of drug repurposing, emphasizing the multifaceted approaches and methodologies employed for the same to confront female cancers.
Collapse
Affiliation(s)
- Kshreeraja S Satish
- Department of Pharmacy Practice, Faculty of Pharmacy, M.S. Ramaiah University of Applied Sciences, Bangalore, Karnataka, India
| | - Ganesan Rajalekshmi Saraswathy
- Department of Pharmacy Practice, Faculty of Pharmacy, M.S. Ramaiah University of Applied Sciences, Bangalore, Karnataka, India.
| | - Giri Ritesh
- Department of Pharmacy Practice, Faculty of Pharmacy, M.S. Ramaiah University of Applied Sciences, Bangalore, Karnataka, India
| | - Kamatchi Sundara Saravanan
- Department of Pharmacognosy, Faculty of Pharmacy, M.S. Ramaiah University of Applied Sciences, Bangalore, Karnataka, India
| | - Aarti Krishnan
- Department of Pharmacy Practice, Faculty of Pharmacy, M.S. Ramaiah University of Applied Sciences, Bangalore, Karnataka, India
| | - Janhavi Bhargava
- Department of Pharmacy Practice, Faculty of Pharmacy, M.S. Ramaiah University of Applied Sciences, Bangalore, Karnataka, India
| | - Kuri Ushnaa
- Department of Pharmacy Practice, Faculty of Pharmacy, M.S. Ramaiah University of Applied Sciences, Bangalore, Karnataka, India
| | - Prizvan Lawrence Dsouza
- Department of Pharmacy Practice, Faculty of Pharmacy, M.S. Ramaiah University of Applied Sciences, Bangalore, Karnataka, India
| |
Collapse
|
5
|
Shukla H, John D, Banerjee S, Tiwari AK. Drug repurposing for neurodegenerative diseases. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2024; 207:249-319. [PMID: 38942541 DOI: 10.1016/bs.pmbts.2024.03.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/30/2024]
Abstract
Neurodegenerative diseases (NDDs) are neuronal problems that include the brain and spinal cord and result in loss of sensory and motor dysfunction. Common NDDs include Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), Multiple Sclerosis (MS), and Amyotrophic Lateral Sclerosis (ALS) etc. The occurrence of these diseases increases with age and is one of the challenging problems among elderly people. Though, several scientific research has demonstrated the key pathologies associated with NDDs still the underlying mechanisms and molecular details are not well understood and need to be explored and this poses a lack of effective treatments for NDDs. Several lines of evidence have shown that NDDs have a high prevalence and affect more than a billion individuals globally but still, researchers need to work forward in identifying the best therapeutic target for NDDs. Thus, several researchers are working in the directions to find potential therapeutic targets to alter the disease pathology and treat the diseases. Several steps have been taken to identify the early detection of the disease and drug repurposing for effective treatment of NDDs. Moreover, it is logical that current medications are being evaluated for their efficacy in treating such disorders; therefore, drug repurposing would be an efficient, safe, and cost-effective way in finding out better medication. In the current manuscript we discussed the utilization of drugs that have been repurposed for the treatment of AD, PD, HD, MS, and ALS.
Collapse
Affiliation(s)
- Halak Shukla
- Department of Biotechnology and Bioengineering, Institute of Advanced Research (IAR), Gandhinagar, Gujarat, India
| | - Diana John
- Department of Biotechnology and Bioengineering, Institute of Advanced Research (IAR), Gandhinagar, Gujarat, India
| | - Shuvomoy Banerjee
- Department of Biotechnology and Bioengineering, Institute of Advanced Research (IAR), Gandhinagar, Gujarat, India
| | - Anand Krishna Tiwari
- Genetics and Developmental Biology Laboratory, Department of Biotechnology and Bioengineering, Institute of Advanced Research (IAR), Gandhinagar, Gujarat, India.
| |
Collapse
|
6
|
Karampuri A, Kundur S, Perugu S. Exploratory drug discovery in breast cancer patients: A multimodal deep learning approach to identify novel drug candidates targeting RTK signaling. Comput Biol Med 2024; 174:108433. [PMID: 38642491 DOI: 10.1016/j.compbiomed.2024.108433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/04/2024] [Accepted: 04/07/2024] [Indexed: 04/22/2024]
Abstract
Breast cancer, a highly formidable and diverse malignancy predominantly affecting women globally, poses a significant threat due to its intricate genetic variability, rendering it challenging to diagnose accurately. Various therapies such as immunotherapy, radiotherapy, and diverse chemotherapy approaches like drug repurposing and combination therapy are widely used depending on cancer subtype and metastasis severity. Our study revolves around an innovative drug discovery strategy targeting potential drug candidates specific to RTK signalling, a prominently targeted receptor class in cancer. To accomplish this, we have developed a multimodal deep neural network (MM-DNN) based QSAR model integrating omics datasets to elucidate genomic, proteomic expression data, and drug responses, validated rigorously. The results showcase an R2 value of 0.917 and an RMSE value of 0.312, affirming the model's commendable predictive capabilities. Structural analogs of drug molecules specific to RTK signalling were sourced from the PubChem database, followed by meticulous screening to eliminate dissimilar compounds. Leveraging the MM-DNN-based QSAR model, we predicted the biological activity of these molecules, subsequently clustering them into three distinct groups. Feature importance analysis was performed. Consequently, we successfully identified prime drug candidates tailored for each potential downstream regulatory protein within the RTK signalling pathway. This method makes the early stages of drug development faster by removing inactive compounds, providing a hopeful path in combating breast cancer.
Collapse
Affiliation(s)
- Anush Karampuri
- Department of Biotechnology, National Institute of Technology, Warangal, 500604, India
| | - Sunitha Kundur
- Department of Biotechnology, National Institute of Technology, Warangal, 500604, India
| | - Shyam Perugu
- Department of Biotechnology, National Institute of Technology, Warangal, 500604, India.
| |
Collapse
|
7
|
Mishra A, Vasanthan M, Malliappan SP. Drug Repurposing: A Leading Strategy for New Threats and Targets. ACS Pharmacol Transl Sci 2024; 7:915-932. [PMID: 38633585 PMCID: PMC11019736 DOI: 10.1021/acsptsci.3c00361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 03/01/2024] [Accepted: 03/06/2024] [Indexed: 04/19/2024]
Abstract
Less than 6% of rare illnesses have an appropriate treatment option. Repurposed medications for new indications are a cost-effective and time-saving strategy that results in excellent success rates, which may significantly lower the risk associated with therapeutic development for rare illnesses. It is becoming a realistic alternative to repurposing "conventional" medications to treat joint and rare diseases considering the significant failure rates, high expenses, and sluggish stride of innovative medication advancement. This is due to delisted compounds, cheaper research fees, and faster development time frames. Repurposed drug competitors have been developed using strategic decisions based on data analysis, interpretation, and investigational approaches, but technical and regulatory restrictions must also be considered. Combining experimental and computational methodologies generates innovative new medicinal applications. It is a one-of-a-kind strategy for repurposing human-safe pharmaceuticals to treat uncommon and difficult-to-treat ailments. It is a very effective method for discovering and creating novel medications. Several pharmaceutical firms have developed novel therapies by repositioning old medications. Repurposing drugs is practical, cost-effective, and speedy and generally involves lower risks when compared to developing a new drug from the beginning.
Collapse
Affiliation(s)
- Ashish
Sriram Mishra
- Department
of Pharmaceutics, SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur, 603202, Tamil Nadu, India
| | - Manimaran Vasanthan
- Department
of Pharmaceutics, SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur, 603202, Tamil Nadu, India
| | - Sivakumar Ponnurengam Malliappan
- School
of Medicine and Pharmacy, Duy Tan University, Da Nang Vietnam, Institute
of Research and Development, Duy Tan University, Da Nang 550000, Vietnam
| |
Collapse
|
8
|
Pellegrini M. Advances in Network-Based Drug Repositioning. LECTURE NOTES IN COMPUTER SCIENCE 2024:99-114. [DOI: 10.1007/978-3-031-55248-9_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
9
|
Li X, Liao M, Wang B, Zan X, Huo Y, Liu Y, Bao Z, Xu P, Liu W. A drug repurposing method based on inhibition effect on gene regulatory network. Comput Struct Biotechnol J 2023; 21:4446-4455. [PMID: 37731599 PMCID: PMC10507583 DOI: 10.1016/j.csbj.2023.09.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 09/05/2023] [Accepted: 09/07/2023] [Indexed: 09/22/2023] Open
Abstract
Numerous computational drug repurposing methods have emerged as efficient alternatives to costly and time-consuming traditional drug discovery approaches. Some of these methods are based on the assumption that the candidate drug should have a reversal effect on disease-associated genes. However, such methods are not applicable in the case that there is limited overlap between disease-related genes and drug-perturbed genes. In this study, we proposed a novel Drug Repurposing method based on the Inhibition Effect on gene regulatory network (DRIE) to identify potential drugs for cancer treatment. DRIE integrated gene expression profile and gene regulatory network to calculate inhibition score by using the shortest path in the disease-specific network. The results on eleven datasets indicated the superior performance of DRIE when compared to other state-of-the-art methods. Case studies showed that our method effectively discovered novel drug-disease associations. Our findings demonstrated that the top-ranked drug candidates had been already validated by CTD database. Additionally, it clearly identified potential agents for three cancers (colorectal, breast, and lung cancer), which was beneficial when annotating drug-disease relationships in the CTD. This study proposed a novel framework for drug repurposing, which would be helpful for drug discovery and development.
Collapse
Affiliation(s)
- Xianbin Li
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, China
- School of Computer Science of Information Technology, Qiannan Normal University for Nationalities, Duyun, China
| | - Minzhen Liao
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, China
| | - Bing Wang
- School of Medicine, Southeast University, Nanjing, China
| | - Xiangzhen Zan
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, China
| | - Yanhao Huo
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, China
| | - Yue Liu
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, China
| | - Zhenshen Bao
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, China
- School of Computer Science of Information Technology, Qiannan Normal University for Nationalities, Duyun, China
| | - Peng Xu
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, China
- School of Computer Science of Information Technology, Qiannan Normal University for Nationalities, Duyun, China
| | - Wenbin Liu
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, China
| |
Collapse
|
10
|
Wang Y, Liu JX, Wang J, Shang J, Gao YL. A Graph Representation Approach Based on Light Gradient Boosting Machine for Predicting Drug-Disease Associations. J Comput Biol 2023; 30:937-947. [PMID: 37486669 DOI: 10.1089/cmb.2023.0078] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/25/2023] Open
Abstract
Determining the association between drug and disease is important in drug development. However, existing approaches for drug-disease associations (DDAs) prediction are too homogeneous in terms of feature extraction. Here, a novel graph representation approach based on light gradient boosting machine (GRLGB) is proposed for prediction of DDAs. After the introduction of the protein into a heterogeneous network, nodes features were extracted from two perspectives: network topology and biological knowledge. Finally, the GRLGB classifier was applied to predict potential DDAs. GRLGB achieved satisfactory results on Bdataset and Fdataset through 10-fold cross-validation. To further prove the reliability of the GRLGB, case studies involving anxiety disorders and clozapine were conducted. The results suggest that GRLGB can identify novel DDAs.
Collapse
Affiliation(s)
- Ying Wang
- School of Computer Science, Qufu Normal University, Rizhao, Shandong, China
| | - Jin-Xing Liu
- School of Computer Science, Qufu Normal University, Rizhao, Shandong, China
| | - Juan Wang
- School of Computer Science, Qufu Normal University, Rizhao, Shandong, China
| | - Junliang Shang
- School of Computer Science, Qufu Normal University, Rizhao, Shandong, China
| | - Ying-Lian Gao
- Qufu Normal University Library, Qufu Normal University, Rizhao, Shandong, China
| |
Collapse
|
11
|
Munj SA, Taz TA, Arslanturk S, Heath EI. Biomarker-driven drug repurposing on biologically similar cancers with DNA-repair deficiencies. Front Genet 2022; 13:1015531. [PMID: 36583025 PMCID: PMC9792769 DOI: 10.3389/fgene.2022.1015531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 11/15/2022] [Indexed: 12/15/2022] Open
Abstract
Similar molecular and genetic aberrations among diseases can lead to the discovery of jointly important treatment options across biologically similar diseases. Oncologists closely looked at several hormone-dependent cancers and identified remarkable pathological and molecular similarities in their DNA repair pathway abnormalities. Although deficiencies in Homologous Recombination (HR) pathway plays a significant role towards cancer progression, there could be other DNA-repair pathway deficiencies that requires careful investigation. In this paper, through a biomarker-driven drug repurposing model, we identified several potential drug candidates for breast and prostate cancer patients with DNA-repair deficiencies based on common specific biomarkers and irrespective of the organ the tumors originated from. Normalized discounted cumulative gain (NDCG) and sensitivity analysis were used to assess the performance of the drug repurposing model. Our results showed that Mitoxantrone and Genistein were among drugs with high therapeutic effects that significantly reverted the gene expression changes caused by the disease (FDR adjusted p-values for prostate cancer =1.225e-4 and 8.195e-8, respectively) for patients with deficiencies in their homologous recombination (HR) pathways. The proposed multi-cancer treatment framework, suitable for patients whose cancers had common specific biomarkers, has the potential to identify promising drug candidates by enriching the study population through the integration of multiple cancers and targeting patients who respond poorly to organ-specific treatments.
Collapse
Affiliation(s)
- Seeya Awadhut Munj
- Department of Computer Science, Wayne State University, Detroit, MI, United States
| | - Tasnimul Alam Taz
- Department of Computer Science, Wayne State University, Detroit, MI, United States
| | - Suzan Arslanturk
- Department of Computer Science, Wayne State University, Detroit, MI, United States,*Correspondence: Suzan Arslanturk,
| | - Elisabeth I. Heath
- Department of Oncology, Wayne State University, Detroit, MI, United States,Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Detroit, MI, United States
| |
Collapse
|
12
|
Chen C, Wang N, Wang B, Zhang Q, Hu Y, Cheng G, Tao S, Huang J, Wang C, Zhang Y. Network analysis-based strategy to investigate the protective effect of cepharanthine on rat acute respiratory distress syndrome. Front Pharmacol 2022; 13:1054339. [DOI: 10.3389/fphar.2022.1054339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 10/17/2022] [Indexed: 11/13/2022] Open
Abstract
Combined with Network Analysis (NA) and in vivo experimental methods, we explored and verified the mechanism of Cepharanthine (CEP) involved in the treatment of acute respiratory distress syndrome (ARDS). Potential targets of CEP were searched using the SwissTargetPrediction database. The pathogenic genes related to ARDS were obtained using the DisGeNET database. A protein-protein interaction network of common target genes of disease-compound was subsequently built and visualised. Functional enrichment analysis was performed through the Enrichr database. Finally, for in vivo experimental verification, we established an oleic acid-induced ARDS rat model, mainly through histological evaluation and the ELISA method to evaluate both the protective effect of CEP on ARDS and its effect on inflammation. A total of 100 genes were found to be CEP targeted genes, while 153 genes were found to be associated with ARDS. The PPI network was used to illustrate the link and purpose of the genes associated with CEP and ARDS, which contained 238 nodes and 2,333 links. GO and KEGG analyses indicated that inflammatory response and its related signalling pathways were closely associated with CEP-mediated ARDS treatment. Thus, a key CEP–gene–pathway-ARDS network was constructed through network analysis, including 152 nodes (5 targets and 6 pathways) and 744 links. The results of in vivo experiments showed that CEP could alleviate histopathological changes and pulmonary edema related to ARDS, in addition to reducing neutrophil infiltration and secretion of inflammatory cytokines, whilst increasing serum contents of ResolvinD1 and ResolvinE1. Thus, these effects enhance the anti-inflammatory responses. Thus, our results show that CEP can treat oleic acid-induced ARDS in rats via ResolvinE1 and ResolvinD1 signalling pathways that promote inflammation resolution, providing a new avenue to explore for the clinical treatment of ARDS.
Collapse
|
13
|
Gao Z, Ding P, Xu R. KG-Predict: A knowledge graph computational framework for drug repurposing. J Biomed Inform 2022; 132:104133. [PMID: 35840060 PMCID: PMC9595135 DOI: 10.1016/j.jbi.2022.104133] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 06/18/2022] [Accepted: 07/03/2022] [Indexed: 11/26/2022]
Abstract
The emergence of large-scale phenotypic, genetic, and other multi-model biochemical data has offered unprecedented opportunities for drug discovery including drug repurposing. Various knowledge graph-based methods have been developed to integrate and analyze complex and heterogeneous data sources to find new therapeutic applications for existing drugs. However, existing methods have limitations in modeling and capturing context-sensitive inter-relationships among tens of thousands of biomedical entities. In this paper, we developed KG-Predict: a knowledge graph computational framework for drug repurposing. We first integrated multiple types of entities and relations from various genotypic and phenotypic databases to construct a knowledge graph termed GP-KG. GP-KG was composed of 1,246,726 associations between 61,146 entities. KG-Predict then aggregated the heterogeneous topological and semantic information from GP-KG to learn low-dimensional representations of entities and relations, and further utilized these representations to infer new drug-disease interactions. In cross-validation experiments, KG-Predict achieved high performances [AUROC (the area under receiver operating characteristic) = 0.981, AUPR (the area under precision-recall) = 0.409 and MRR (the mean reciprocal rank) = 0.261], outperforming other state-of-art graph embedding methods. We applied KG-Predict in identifying novel repositioned candidate drugs for Alzheimer's disease (AD) and showed that KG-Predict prioritized both FDA-approved and active clinical trial anti-AD drugs among the top (AUROC = 0.868 and AUPR = 0.364).
Collapse
Affiliation(s)
- Zhenxiang Gao
- Center for Artificial Intelligence in Drug Discovery, School of Medicine, Case Western Reserve University, Cleveland, 44106 OH, USA.
| | - Pingjian Ding
- Center for Artificial Intelligence in Drug Discovery, School of Medicine, Case Western Reserve University, Cleveland, 44106 OH, USA.
| | - Rong Xu
- Center for Artificial Intelligence in Drug Discovery, School of Medicine, Case Western Reserve University, Cleveland, 44106 OH, USA.
| |
Collapse
|
14
|
Xu J, Meng Y, Peng L, Cai L, Tang X, Liang Y, Tian G, Yang J. Computational drug repositioning using similarity constrained weight regularization matrix factorization: A case of COVID-19. J Cell Mol Med 2022; 26:3772-3782. [PMID: 35644992 PMCID: PMC9258716 DOI: 10.1111/jcmm.17412] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 02/03/2022] [Accepted: 05/11/2022] [Indexed: 02/06/2023] Open
Abstract
Amid the COVID‐19 crisis, we put sizeable efforts to collect a high number of experimentally validated drug–virus association entries from literature by text mining and built a human drug–virus association database. To the best of our knowledge, it is the largest publicly available drug–virus database so far. Next, we develop a novel weight regularization matrix factorization approach, termed WRMF, for in silico drug repurposing by integrating three networks: the known drug–virus association network, the drug–drug chemical structure similarity network, and the virus–virus genomic sequencing similarity network. Specifically, WRMF adds a weight to each training sample for reducing the influence of negative samples (i.e. the drug–virus association is unassociated). A comparison on the curated drug–virus database shows that WRMF performs better than a few state‐of‐the‐art methods. In addition, we selected the other two different public datasets (i.e. Cdataset and HMDD V2.0) to assess WRMF's performance. The case study also demonstrated the accuracy and reliability of WRMF to infer potential drugs for the novel virus. In summary, we offer a useful tool including a novel drug–virus association database and a powerful method WRMF to repurpose potential drugs for new viruses.
Collapse
Affiliation(s)
- Junlin Xu
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, China
| | - Yajie Meng
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, China
| | - Lihong Peng
- School of Computer Science, Hunan University of Technology, Zhuzhou, China
| | - Lijun Cai
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, China
| | - Xianfang Tang
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, China
| | | | - Geng Tian
- Geneis Beijing Co., Ltd., Beijing, China
| | | |
Collapse
|
15
|
Chavda VP, Kapadia C, Soni S, Prajapati R, Chauhan SC, Yallapu MM, Apostolopoulos V. A global picture: therapeutic perspectives for COVID-19. Immunotherapy 2022; 14:351-371. [PMID: 35187954 PMCID: PMC8884157 DOI: 10.2217/imt-2021-0168] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 01/19/2022] [Indexed: 02/06/2023] Open
Abstract
The COVID-19 pandemic is a lethal virus outbreak by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), which has severely affected human lives and the global economy. The most vital part of the research and development of therapeutic agents is to design drug products to manage COVID-19 efficiently. Numerous attempts have been in place to determine the optimal drug dose and combination of drugs to treat the disease on a global scale. This article documents the information available on SARS-CoV-2 and its life cycle, which will aid in the development of the potential treatment options. A consolidated summary of several natural and repurposed drugs to manage COVID-19 is depicted with summary of current vaccine development. People with high age, comorbity and concomitant illnesses such as overweight, metabolic disorders, pulmonary disease, coronary heart disease, renal failure, fatty liver and neoplastic disorders are more prone to create serious COVID-19 and its consequences. This article also presents an overview of post-COVID-19 complications in patients.
Collapse
Affiliation(s)
- Vivek P Chavda
- Department of Pharmaceutics & Pharmaceutical Technology, L.M. College of Pharmacy, Ahmedabad, Gujarat, 380009, India
- Department of Pharmaceutics, K B Institute of Pharmaceutical Education & Research, Kadi Sarva Vishwavidhyalaya, Gandhinagar, Gujarat, 382023, India
| | - Carron Kapadia
- Department of Pharmaceutics & Pharmaceutical Technology, L.M. College of Pharmacy, Ahmedabad, Gujarat, 380009, India
| | - Shailvi Soni
- Department of Pharmaceutics & Pharmaceutical Technology, L.M. College of Pharmacy, Ahmedabad, Gujarat, 380009, India
| | - Riddhi Prajapati
- Department of Pharmaceutics & Pharmaceutical Technology, L.M. College of Pharmacy, Ahmedabad, Gujarat, 380009, India
| | - Subhash C Chauhan
- Department of Immunology & Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78503, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78503, USA
| | - Murali M Yallapu
- Department of Immunology & Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78503, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78503, USA
| | - Vasso Apostolopoulos
- Institute for Health & Sport, Victoria University, Melbourne, VIC, 3030, Australia
| |
Collapse
|
16
|
Chavda VP, Kapadia C, Soni S, Prajapati R, Chauhan SC, Yallapu MM, Apostolopoulos V. A global picture: therapeutic perspectives for COVID-19. Immunotherapy 2022. [PMID: 35187954 DOI: 10.2217/imt-2021-0168.10.2217/imt-2021-0168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2023] Open
Abstract
The COVID-19 pandemic is a lethal virus outbreak by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), which has severely affected human lives and the global economy. The most vital part of the research and development of therapeutic agents is to design drug products to manage COVID-19 efficiently. Numerous attempts have been in place to determine the optimal drug dose and combination of drugs to treat the disease on a global scale. This article documents the information available on SARS-CoV-2 and its life cycle, which will aid in the development of the potential treatment options. A consolidated summary of several natural and repurposed drugs to manage COVID-19 is depicted with summary of current vaccine development. People with high age, comorbity and concomitant illnesses such as overweight, metabolic disorders, pulmonary disease, coronary heart disease, renal failure, fatty liver and neoplastic disorders are more prone to create serious COVID-19 and its consequences. This article also presents an overview of post-COVID-19 complications in patients.
Collapse
Affiliation(s)
- Vivek P Chavda
- Department of Pharmaceutics & Pharmaceutical Technology, L.M. College of Pharmacy, Ahmedabad, Gujarat, 380009, India
- Department of Pharmaceutics, K B Institute of Pharmaceutical Education & Research, Kadi Sarva Vishwavidhyalaya, Gandhinagar, Gujarat, 382023, India
| | - Carron Kapadia
- Department of Pharmaceutics & Pharmaceutical Technology, L.M. College of Pharmacy, Ahmedabad, Gujarat, 380009, India
| | - Shailvi Soni
- Department of Pharmaceutics & Pharmaceutical Technology, L.M. College of Pharmacy, Ahmedabad, Gujarat, 380009, India
| | - Riddhi Prajapati
- Department of Pharmaceutics & Pharmaceutical Technology, L.M. College of Pharmacy, Ahmedabad, Gujarat, 380009, India
| | - Subhash C Chauhan
- Department of Immunology & Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78503, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78503, USA
| | - Murali M Yallapu
- Department of Immunology & Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78503, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78503, USA
| | - Vasso Apostolopoulos
- Institute for Health & Sport, Victoria University, Melbourne, VIC, 3030, Australia
| |
Collapse
|
17
|
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] [Scholar 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
|
18
|
Kim HS. Drug Repositioning: Exploring New Indications for Existing Drug-Disease Relationships. Endocrinol Metab (Seoul) 2022; 37:62-64. [PMID: 35255602 PMCID: PMC8901967 DOI: 10.3803/enm.2022.1403] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 01/20/2022] [Indexed: 11/23/2022] Open
Affiliation(s)
- Hun-Sung Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul,
Korea
- Department of Medical Informatics, College of Medicine, The Catholic University of Korea, Seoul,
Korea
| |
Collapse
|
19
|
Sasidharan S, Sarkar N, Saudagar P. Discovery of compounds inhibiting SARS-COV-2 multi-targets. J Biomol Struct Dyn 2022; 41:2602-2617. [PMID: 34994297 DOI: 10.1080/07391102.2021.2025149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has become a pandemic that has devastated the lives of millions. Researchers around the world are relentlessly working in hopes of finding a cure. Even though the virus shares similarities with reported SARS-CoV and MERS-CoV at the genomic and proteomic level, efforts to repurpose already known drugs against SARS-CoV-2 have resulted ineffective. In this succinct review, we discuss the different potential targets in SARS-CoV-2 at both the genomic and proteomic levels. In addition, we analyze the compounds inhibiting individual target protein as well as multiple targets of SARS-CoV-2. ACE-2 receptor in humans has also been considered a target, keeping the role of the receptor in mind. The mechanism of action of these compounds has also been highlighted along with their clinical manifestation. Towards the end of the review, a brief note on the drugs currently in clinical trials and the current status of the vaccines are also examined. In conclusion, compounds targeting multiple targets of the virus hold the key in putting an end to the coronavirus malady.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Santanu Sasidharan
- Department of Biotechnology, National Institute of Technology, Warangal, Telangana, India
| | - Neellohit Sarkar
- Department of Biotechnology, National Institute of Technology, Warangal, Telangana, India
| | - Prakash Saudagar
- Department of Biotechnology, National Institute of Technology, Warangal, Telangana, India
| |
Collapse
|
20
|
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: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar 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
|
21
|
Golriz Khatami S, Mubeen S, Bharadhwaj VS, Kodamullil AT, Hofmann-Apitius M, Domingo-Fernández D. Using predictive machine learning models for drug response simulation by calibrating patient-specific pathway signatures. NPJ Syst Biol Appl 2021; 7:40. [PMID: 34707117 PMCID: PMC8551267 DOI: 10.1038/s41540-021-00199-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 09/21/2021] [Indexed: 11/21/2022] Open
Abstract
The utility of pathway signatures lies in their capability to determine whether a specific pathway or biological process is dysregulated in a given patient. These signatures have been widely used in machine learning (ML) methods for a variety of applications including precision medicine, drug repurposing, and drug discovery. In this work, we leverage highly predictive ML models for drug response simulation in individual patients by calibrating the pathway activity scores of disease samples. Using these ML models and an intuitive scoring algorithm to modify the signatures of patients, we evaluate whether a given sample that was formerly classified as diseased, could be predicted as normal following drug treatment simulation. We then use this technique as a proxy for the identification of potential drug candidates. Furthermore, we demonstrate the ability of our methodology to successfully identify approved and clinically investigated drugs for four different cancers, outperforming six comparable state-of-the-art methods. We also show how this approach can deconvolute a drugs' mechanism of action and propose combination therapies. Taken together, our methodology could be promising to support clinical decision-making in personalized medicine by simulating a drugs' effect on a given patient.
Collapse
Affiliation(s)
- Sepehr Golriz Khatami
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing, Sankt Augustin, 53757, Germany.
- Bonn-Aachen International Center for Information Technology (B-IT), University of Bonn, 53115, Bonn, Germany.
| | - Sarah Mubeen
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing, Sankt Augustin, 53757, Germany
- Bonn-Aachen International Center for Information Technology (B-IT), University of Bonn, 53115, Bonn, Germany
- Fraunhofer Center for Machine Learning, Sankt Augustin, Germany
| | - Vinay Srinivas Bharadhwaj
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing, Sankt Augustin, 53757, Germany
- Bonn-Aachen International Center for Information Technology (B-IT), University of Bonn, 53115, Bonn, Germany
| | - Alpha Tom Kodamullil
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing, Sankt Augustin, 53757, Germany
| | - Martin Hofmann-Apitius
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing, Sankt Augustin, 53757, Germany
- Bonn-Aachen International Center for Information Technology (B-IT), University of Bonn, 53115, Bonn, Germany
| | - Daniel Domingo-Fernández
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing, Sankt Augustin, 53757, Germany.
- Fraunhofer Center for Machine Learning, Sankt Augustin, Germany.
- Enveda Biosciences, Boulder, CO, 80301, USA.
| |
Collapse
|
22
|
Islam MM, Wang Y, Hu P. A Maximum Flow-Based Approach to Prioritize Drugs for Drug Repurposing of Chronic Diseases. Life (Basel) 2021; 11:1115. [PMID: 34832991 PMCID: PMC8625622 DOI: 10.3390/life11111115] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 10/12/2021] [Accepted: 10/18/2021] [Indexed: 12/28/2022] Open
Abstract
The discovery of new drugs is required in the time of global aging and increasing populations. Traditional drug development strategies are expensive, time-consuming, and have high risks. Thus, drug repurposing, which treats new/other diseases using existing drugs, has become a very admired tactic. It can also be referred to as the re-investigation of the existing drugs that failed to indicate the usefulness for the new diseases. Previously published literature used maximum flow approaches to identify new drug targets for drug-resistant infectious diseases but not for drug repurposing. Therefore, we are proposing a maximum flow-based protein-protein interactions (PPIs) network analysis approach to identify new drug targets (proteins) from the targets of the FDA (Food and Drug Administration) drugs and their associated drugs for chronic diseases (such as breast cancer, inflammatory bowel disease (IBD), and chronic obstructive pulmonary disease (COPD)) treatment. Experimental results showed that we have successfully turned the drug repurposing into a maximum flow problem. Our top candidates of drug repurposing, Guanidine, Dasatinib, and Phenethyl Isothiocyanate for breast cancer, IBD, and COPD were experimentally validated by other independent research as the potential candidate drugs for these diseases, respectively. This shows the usefulness of the proposed maximum flow approach for drug repurposing.
Collapse
Affiliation(s)
- Md. Mohaiminul Islam
- Department of Computer Science, University of Manitoba, Winnipeg, MB R3T 2N2, Canada; (M.M.I.); (Y.W.)
| | - Yang Wang
- Department of Computer Science, University of Manitoba, Winnipeg, MB R3T 2N2, Canada; (M.M.I.); (Y.W.)
| | - Pingzhao Hu
- Department of Computer Science, University of Manitoba, Winnipeg, MB R3T 2N2, Canada; (M.M.I.); (Y.W.)
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
- Department of Electrical Computer Engineering, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
- CancerCare Manitoba Research Institute, Winnipeg, MB R3T 2N2, Canada
| |
Collapse
|
23
|
Falvo P, Orecchioni S, Roma S, Raveane A, Bertolini F. Drug Repurposing in Oncology, an Attractive Opportunity for Novel Combinatorial Regimens. Curr Med Chem 2021; 28:2114-2136. [PMID: 33109033 DOI: 10.2174/0929867327999200817104912] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/21/2020] [Accepted: 05/26/2020] [Indexed: 11/22/2022]
Abstract
The costs of developing, validating and buying new drugs are dramatically increasing. On the other hand, sobering economies have difficulties in sustaining their healthcare systems, particularly in countries with an elderly population requiring increasing welfare. This conundrum requires immediate action, and a possible option is to study the large, already present arsenal of drugs approved and to use them for innovative therapies. This possibility is particularly interesting in oncology, where the complexity of the cancer genome dictates in most patients a multistep therapeutic approach. In this review, we discuss a) Computational approaches; b) preclinical models; c) currently ongoing or already published clinical trials in the drug repurposing field in oncology; and d) drug repurposing to overcome resistance to previous therapies.
Collapse
Affiliation(s)
- Paolo Falvo
- Laboratory of Hematology-Oncology, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Stefania Orecchioni
- Laboratory of Hematology-Oncology, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Stefania Roma
- Laboratory of Hematology-Oncology, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Alessandro Raveane
- Laboratory of Hematology-Oncology, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Francesco Bertolini
- Laboratory of Hematology-Oncology, European Institute of Oncology IRCCS, 20141 Milan, Italy
| |
Collapse
|
24
|
Cui C, Ding X, Wang D, Chen L, Xiao F, Xu T, Zheng M, Luo X, Jiang H, Chen K. Drug repurposing against breast cancer by integrating drug-exposure expression profiles and drug-drug links based on graph neural network. Bioinformatics 2021; 37:2930-2937. [PMID: 33739367 PMCID: PMC8479657 DOI: 10.1093/bioinformatics/btab191] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 03/16/2021] [Accepted: 03/18/2021] [Indexed: 02/02/2023] Open
Abstract
MOTIVATION Breast cancer is one of the leading causes of cancer deaths among women worldwide. It is necessary to develop new breast cancer drugs because of the shortcomings of existing therapies. The traditional discovery process is time-consuming and expensive. Repositioning of clinically approved drugs has emerged as a novel approach for breast cancer therapy. However, serendipitous or experiential repurposing cannot be used as a routine method. RESULTS In this study, we proposed a graph neural network model GraphRepur based on GraphSAGE for drug repurposing against breast cancer. GraphRepur integrated two major classes of computational methods, drug network-based and drug signature-based. The differentially expressed genes of disease, drug-exposure gene expression data and the drug-drug links information were collected. By extracting the drug signatures and topological structure information contained in the drug relationships, GraphRepur can predict new drugs for breast cancer, outperforming previous state-of-the-art approaches and some classic machine learning methods. The high-ranked drugs have indeed been reported as new uses for breast cancer treatment recently. AVAILABILITYAND IMPLEMENTATION The source code of our model and datasets are available at: https://github.com/cckamy/GraphRepur and https://figshare.com/articles/software/GraphRepur_Breast_Cancer_Drug_Repurposing/14220050. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Chen Cui
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaoyu Ding
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Dingyan Wang
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lifan Chen
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Fu Xiao
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | | | - Mingyue Zheng
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- To whom correspondence should be addressed. or
| | - Xiaomin Luo
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- To whom correspondence should be addressed. or
| | - Hualiang Jiang
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 200031, China
| | - Kaixian Chen
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 200031, China
| |
Collapse
|
25
|
Draghici S, Nguyen TM, Sonna LA, Ziraldo C, Vanciu R, Fadel R, Morrison A, Kenney RM, Alangaden G, Ramesh M, Mor G. COVID-19: disease pathways and gene expression changes predict methylprednisolone can improve outcome in severe cases. Bioinformatics 2021; 37:2691-2698. [PMID: 33693506 PMCID: PMC7989618 DOI: 10.1093/bioinformatics/btab163] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 02/15/2021] [Accepted: 03/04/2021] [Indexed: 11/27/2022] Open
Abstract
Motivation COVID-19 has several distinct clinical phases: a viral replication phase, an inflammatory phase, and in some patients, a hyper-inflammatory phase. High mortality is associated with patients developing cytokine storm syndrome. Treatment of hyper-inflammation in these patients using existing, approved therapies with proven safety profiles could address the immediate need to reduce mortality. Results We analyzed the changes in the gene expression, pathways and putative mechanisms induced by SARS-CoV2 in NHBE, and A549 cells, as well as COVID-19 lung vs. their respective controls. We used these changes to identify FDA approved drugs that could be repurposed to help COVID-19 patients with severe symptoms related to hyper-inflammation. We identified methylprednisolone (MP) as a potential leading therapy. The results were then confirmed in five independent validation data sets including Vero E6 cells, lung and intestinal organoids, as well as additional patient lung sample vs. their respective controls. Finally, the efficacy of MP was validated in an independent clinical study. Thirty-day all-cause mortality occurred at a significantly lower rate in the MP-treated group compared to control group (29.6% vs. 16.6%, p = 0.027). Clinical results confirmed the in silico prediction that MP could improve outcomes in severe cases of COVID-19. A low number needed to treat (NNT = 5) suggests MP may be more efficacious than dexamethasone or hydrocortisone. Availability iPathwayGuide is available at https://ipathwayguide.advaitabio.com/ Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Sorin Draghici
- Department of Computer Science, Wayne State University, Michigan 48202, USA.,Department of Obstetrics and Gynecology, Wayne State University, Michigan 48202, USA.,Department of Internal Medicine, Henry Ford Health System, USA
| | - Tuan-Minh Nguyen
- Department of Computer Science, Wayne State University, Michigan 48202, USA
| | | | | | | | - Raef Fadel
- Department of Internal Medicine, Henry Ford Health System, USA
| | | | | | | | - Mayur Ramesh
- Division of Infectious Diseases, Henry Ford Health System, USA
| | - Gil Mor
- Department of Obstetrics and Gynecology, Wayne State University, Michigan 48202, USA
| |
Collapse
|
26
|
Chen HG, Zhou XH. MNBDR: A Module Network Based Method for Drug Repositioning. Genes (Basel) 2020; 12:E25. [PMID: 33375395 PMCID: PMC7824496 DOI: 10.3390/genes12010025] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/21/2020] [Accepted: 12/24/2020] [Indexed: 11/17/2022] Open
Abstract
Drug repurposing/repositioning, which aims to find novel indications for existing drugs, contributes to reducing the time and cost for drug development. For the recent decade, gene expression profiles of drug stimulating samples have been successfully used in drug repurposing. However, most of the existing methods neglect the gene modules and the interactions among the modules, although the cross-talks among pathways are common in drug response. It is essential to develop a method that utilizes the cross-talks information to predict the reliable candidate associations. In this study, we developed MNBDR (Module Network Based Drug Repositioning), a novel method that based on module network to screen drugs. It integrated protein-protein interactions and gene expression profile of human, to predict drug candidates for diseases. Specifically, the MNBDR mined dense modules through protein-protein interaction (PPI) network and constructed a module network to reveal cross-talks among modules. Then, together with the module network, based on existing gene expression data set of drug stimulation samples and disease samples, we used random walk algorithms to capture essential modules in disease development and proposed a new indicator to screen potential drugs for a given disease. Results showed MNBDR could provide better performance than popular methods. Moreover, functional analysis of the essential modules in the network indicated our method could reveal biological mechanism in drug response.
Collapse
Affiliation(s)
| | - Xiong-Hui Zhou
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan 430070, China;
| |
Collapse
|
27
|
Yang M, Wu G, Zhao Q, Li Y, Wang J. Computational drug repositioning based on multi-similarities bilinear matrix factorization. Brief Bioinform 2020; 22:5956157. [PMID: 33147616 DOI: 10.1093/bib/bbaa267] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/31/2020] [Accepted: 09/16/2020] [Indexed: 12/13/2022] Open
Abstract
With the development of high-throughput technology and the accumulation of biomedical data, the prior information of biological entity can be calculated from different aspects. Specifically, drug-drug similarities can be measured from target profiles, drug-drug interaction and side effects. Similarly, different methods and data sources to calculate disease ontology can result in multiple measures of pairwise disease similarities. Therefore, in computational drug repositioning, developing a dynamic method to optimize the fusion process of multiple similarities is a crucial and challenging task. In this study, we propose a multi-similarities bilinear matrix factorization (MSBMF) method to predict promising drug-associated indications for existing and novel drugs. Instead of fusing multiple similarities into a single similarity matrix, we concatenate these similarity matrices of drug and disease, respectively. Applying matrix factorization methods, we decompose the drug-disease association matrix into a drug-feature matrix and a disease-feature matrix. At the same time, using these feature matrices as basis, we extract effective latent features representing the drug and disease similarity matrices to infer missing drug-disease associations. Moreover, these two factored matrices are constrained by non-negative factorization to ensure that the completed drug-disease association matrix is biologically interpretable. In addition, we numerically solve the MSBMF model by an efficient alternating direction method of multipliers algorithm. The computational experiment results show that MSBMF obtains higher prediction accuracy than the state-of-the-art drug repositioning methods in cross-validation experiments. Case studies also demonstrate the effectiveness of our proposed method in practical applications. Availability: The data and code of MSBMF are freely available at https://github.com/BioinformaticsCSU/MSBMF. Corresponding author: Jianxin Wang, School of Computer Science and Engineering, Central South University, Changsha, Hunan 410083, P. R. China. E-mail: jxwang@mail.csu.edu.cn Supplementary Data: Supplementary data are available online at https://academic.oup.com/bib.
Collapse
Affiliation(s)
- Mengyun Yang
- School of Computer Science and Engineering, Central South University, China
| | - Gaoyan Wu
- School of Computer Science and Engineering, Central South University, China
| | - Qichang Zhao
- School of Computer Science and Engineering, Central South University, China
| | | | - Jianxin Wang
- School of Computer Science and Engineering, Central South University, China
| |
Collapse
|
28
|
Mangione W, Falls Z, Chopra G, Samudrala R. cando.py: Open Source Software for Predictive Bioanalytics of Large Scale Drug-Protein-Disease Data. J Chem Inf Model 2020; 60:4131-4136. [PMID: 32515949 PMCID: PMC8098009 DOI: 10.1021/acs.jcim.0c00110] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Traditional drug discovery methods focus on optimizing the efficacy of a drug against a single biological target of interest for a specific disease. However, evidence supports the multitarget theory, i.e., drugs work by exerting their therapeutic effects via interaction with multiple biological targets, which have multiple phenotypic effects. Analytics of drug-protein interactions on a large proteomic scale provides insight into disease systems while also allowing for prediction of putative therapeutics against specific indications. We present a Python package for analysis of drug-proteome and drug-disease relationships implementing the Computational Analysis of Novel Drug Opportunities (CANDO) platform. The CANDO package allows for rapid drug similarity assessment, most notably via an in-house interaction scoring protocol where billions of drug-protein interactions are rapidly scored and the similarity of drug-proteome interaction signatures is calculated. The package also implements a variety of benchmarking protocols for shotgun drug discovery and repurposing, i.e., to determine how every known drug is related to every other in the context of the indications/diseases for which they are approved. Drug predictions are generated through consensus scoring of the most similar compounds to drugs known to treat a particular indication. Support for comparing and ranking novel chemical entities, as well as machine learning modules for both benchmarking and putative drug candidate prediction is also available. The CANDO Python package is available on GitHub at https://github.com/ram-compbio/CANDO, through the Conda Python package installer, and at http://compbio.org/software/.
Collapse
Affiliation(s)
- William Mangione
- Department of Biomedical Informatics, University at Buffalo, Buffalo, New York 14120, United States
| | - Zackary Falls
- Department of Biomedical Informatics, University at Buffalo, Buffalo, New York 14120, United States
| | - Gaurav Chopra
- Department of Chemistry, Purdue Institute for Drug Discovery, Integrated Data Science Institute, Purdue University, West Lafayette, Indiana 47907, United States
| | - Ram Samudrala
- Department of Biomedical Informatics, University at Buffalo, Buffalo, New York 14120, United States
| |
Collapse
|
29
|
Abstract
The current global pandemic COVID-19 caused by the SARS-CoV-2 virus has already inflicted insurmountable damage both to the human lives and global economy. There is an immediate need for identification of effective drugs to contain the disastrous virus outbreak. Global efforts are already underway at a war footing to identify the best drug combination to address the disease. In this review, an attempt has been made to understand the SARS-CoV-2 life cycle, and based on this information potential druggable targets against SARS-CoV-2 are summarized. Also, the strategies for ongoing and future drug discovery against the SARS-CoV-2 virus are outlined. Given the urgency to find a definitive cure, ongoing drug repurposing efforts being carried out by various organizations are also described. The unprecedented crisis requires extraordinary efforts from the scientific community to effectively address the issue and prevent further loss of human lives and health.
Collapse
Affiliation(s)
- Ambrish Saxena
- Indian Institute of Technology Tirupati, Tirupati, India
| |
Collapse
|
30
|
Mangione W, Falls Z, Melendy T, Chopra G, Samudrala R. Shotgun drug repurposing biotechnology to tackle epidemics and pandemics. Drug Discov Today 2020; 25:1126-1128. [PMID: 32405249 PMCID: PMC7217781 DOI: 10.1016/j.drudis.2020.05.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/03/2020] [Accepted: 05/05/2020] [Indexed: 12/14/2022]
Affiliation(s)
- William Mangione
- Department of Biomedical Informatics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, 14203, United States
| | - Zackary Falls
- Department of Biomedical Informatics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, 14203, United States
| | - Thomas Melendy
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, 14203, United States
| | - Gaurav Chopra
- Department of Chemistry, Purdue Institute for Drug Discovery, Integrated Data Science Institute, Purdue University, West Lafayette, IN, 47907, United States.
| | - Ram Samudrala
- Department of Biomedical Informatics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, 14203, United States.
| |
Collapse
|
31
|
Mangione W, Falls Z, Melendy T, Chopra G, Samudrala R. Shotgun drug repurposing biotechnology to tackle epidemics and pandemics. CHEMRXIV : THE PREPRINT SERVER FOR CHEMISTRY 2020:10.26434/chemrxiv.12045318.v2. [PMID: 32511286 PMCID: PMC7252447 DOI: 10.26434/chemrxiv.12045318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
In this manuscript we highlight consensus between the list of drugs currently in clinical trials to treat COVID-19, the worldwide pandemic caused by severe acute respiratory coronavirus 2 (SARS-CoV-2), and the list of predictions made using our shotgun drug discovery, repurposing, and design platform known as CANDO (Computational Analysis of Novel Drug Opportunities). We make the argument that increased funding and development for drug repurposing biotechnology like ours will help combat the inevitable pathogenic outbreaks of the future. <br>
Collapse
Affiliation(s)
- William Mangione
- Department of Biomedical Informatics, University at Buffalo, Buffalo, NY, 14120, United States
| | - Zackary Falls
- Department of Biomedical Informatics, University at Buffalo, Buffalo, NY, 14120, United States
| | - Thomas Melendy
- Department of Microbiology and Immunology, University at Buffalo, Buffalo, NY, 14120, United States
| | - Gaurav Chopra
- Department of Chemistry, Purdue Institute for Drug Discovery, Integrated Data Science Institute, Purdue University, West Lafayette, IN, 47907, United States
| | - Ram Samudrala
- Department of Biomedical Informatics, University at Buffalo, Buffalo, NY, 14120, United States
| |
Collapse
|
32
|
Satish Kumar K, Velayutham R, Roy KK. A systematic computational analysis of human matrix metalloproteinase 13 (MMP-13) crystal structures and structure-based identification of prospective drug candidates as MMP-13 inhibitors repurposable for osteoarthritis. J Biomol Struct Dyn 2019; 38:3074-3086. [PMID: 31378153 DOI: 10.1080/07391102.2019.1651221] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
| | - Ravichandiran Velayutham
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, India
| | - Kuldeep K. Roy
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, India
| |
Collapse
|