1
|
Li Y, Yang Y, Tong Z, Wang Y, Mi Q, Bai M, Liang G, Li B, Shu K. A comparative benchmarking and evaluation framework for heterogeneous network-based drug repositioning methods. Brief Bioinform 2024; 25:bbae172. [PMID: 38647153 PMCID: PMC11033846 DOI: 10.1093/bib/bbae172] [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: 12/10/2023] [Revised: 02/25/2024] [Accepted: 04/02/2024] [Indexed: 04/25/2024] Open
Abstract
Computational drug repositioning, which involves identifying new indications for existing drugs, is an increasingly attractive research area due to its advantages in reducing both overall cost and development time. As a result, a growing number of computational drug repositioning methods have emerged. Heterogeneous network-based drug repositioning methods have been shown to outperform other approaches. However, there is a dearth of systematic evaluation studies of these methods, encompassing performance, scalability and usability, as well as a standardized process for evaluating new methods. Additionally, previous studies have only compared several methods, with conflicting results. In this context, we conducted a systematic benchmarking study of 28 heterogeneous network-based drug repositioning methods on 11 existing datasets. We developed a comprehensive framework to evaluate their performance, scalability and usability. Our study revealed that methods such as HGIMC, ITRPCA and BNNR exhibit the best overall performance, as they rely on matrix completion or factorization. HINGRL, MLMC, ITRPCA and HGIMC demonstrate the best performance, while NMFDR, GROBMC and SCPMF display superior scalability. For usability, HGIMC, DRHGCN and BNNR are the top performers. Building on these findings, we developed an online tool called HN-DREP (http://hn-drep.lyhbio.com/) to facilitate researchers in viewing all the detailed evaluation results and selecting the appropriate method. HN-DREP also provides an external drug repositioning prediction service for a specific disease or drug by integrating predictions from all methods. Furthermore, we have released a Snakemake workflow named HN-DRES (https://github.com/lyhbio/HN-DRES) to facilitate benchmarking and support the extension of new methods into the field.
Collapse
Affiliation(s)
- Yinghong Li
- Chongqing Key Laboratory of Big Data for Bio Intelligence, Chongqing University of Posts and Telecommunications, Chongqing 400065, P. R. China
| | - Yinqi Yang
- Chongqing Key Laboratory of Big Data for Bio Intelligence, Chongqing University of Posts and Telecommunications, Chongqing 400065, P. R. China
| | - Zhuohao Tong
- Chongqing Key Laboratory of Big Data for Bio Intelligence, Chongqing University of Posts and Telecommunications, Chongqing 400065, P. R. China
| | - Yu Wang
- Chongqing Key Laboratory of Big Data for Bio Intelligence, Chongqing University of Posts and Telecommunications, Chongqing 400065, P. R. China
| | - Qin Mi
- Chongqing Key Laboratory of Big Data for Bio Intelligence, Chongqing University of Posts and Telecommunications, Chongqing 400065, P. R. China
| | - Mingze Bai
- Chongqing Key Laboratory of Big Data for Bio Intelligence, Chongqing University of Posts and Telecommunications, Chongqing 400065, P. R. China
| | - Guizhao Liang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing, 400044, P. R. China
| | - Bo Li
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, P. R. China
| | - Kunxian Shu
- Chongqing Key Laboratory of Big Data for Bio Intelligence, Chongqing University of Posts and Telecommunications, Chongqing 400065, P. R. China
| |
Collapse
|
2
|
Lei S, Lei X, Chen M, Pan Y. Drug Repositioning Based on Deep Sparse Autoencoder and Drug-Disease Similarity. Interdiscip Sci 2024; 16:160-175. [PMID: 38103130 DOI: 10.1007/s12539-023-00593-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 11/03/2023] [Accepted: 11/06/2023] [Indexed: 12/17/2023]
Abstract
Drug repositioning is critical to drug development. Previous drug repositioning methods mainly constructed drug-disease heterogeneous networks to extract drug-disease features. However, these methods faced difficulty when we are using structurally simple models to deal with complex heterogeneous networks. Therefore, in this study, the researchers introduced a drug repositioning method named DRDSA. The method utilizes a deep sparse autoencoder and integrates drug-disease similarities. First, the researchers constructed a drug-disease feature network by incorporating information from drug chemical structure, disease semantic data, and existing known drug-disease associations. Then, we learned the low-dimensional representation of the feature network using a deep sparse autoencoder. Finally, we utilized a deep neural network to make predictions on new drug-disease associations based on the feature representation. The experimental results show that our proposed method has achieved optimal results on all four benchmark datasets, especially on the CTD dataset where AUC and AUPR reached 0.9619 and 0.9676, respectively, outperforming other baseline methods. In the case study, the researchers predicted the top ten antiviral drugs for COVID-19. Remarkably, six out of these predictions were subsequently validated by other literature sources.
Collapse
Affiliation(s)
- Song Lei
- School of Computer Science, Shaanxi Normal University, Xi'an, 710119, China
| | - Xiujuan Lei
- School of Computer Science, Shaanxi Normal University, Xi'an, 710119, China.
| | - Ming Chen
- College of Information Science and Engineering, Hunan Normal University, Changsha, 410081, China
| | - Yi Pan
- Faculty of Computer Science and Control Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
- Shenzhen Key Laboratory of Intelligent Bioinformatics, Shenzhen Institute of Advanced Technology, Shenzhen, 518055, China.
| |
Collapse
|
3
|
Cüvitoğlu A, Isik Z. Network neighborhood operates as a drug repositioning method for cancer treatment. PeerJ 2023; 11:e15624. [PMID: 37456868 PMCID: PMC10340098 DOI: 10.7717/peerj.15624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 06/01/2023] [Indexed: 07/18/2023] Open
Abstract
Computational drug repositioning approaches are important, as they cost less compared to the traditional drug development processes. This study proposes a novel network-based drug repositioning approach, which computes similarities between disease-causing genes and drug-affected genes in a network topology to suggest candidate drugs with highest similarity scores. This new method aims to identify better treatment options by integrating systems biology approaches. It uses a protein-protein interaction network that is the main topology to compute a similarity score between candidate drugs and disease-causing genes. The disease-causing genes were mapped on this network structure. Transcriptome profiles of drug candidates were taken from the LINCS project and mapped individually on the network structure. The similarity of these two networks was calculated by different network neighborhood metrics, including Adamic-Adar, PageRank and neighborhood scoring. The proposed approach identifies the best candidates by choosing the drugs with significant similarity scores. The method was experimented on melanoma, colorectal, and prostate cancers. Several candidate drugs were predicted by applying AUC values of 0.6 or higher. Some of the predictions were approved by clinical phase trials or other in-vivo studies found in literature. The proposed drug repositioning approach would suggest better treatment options with integration of functional information between genes and transcriptome level effects of drug perturbations and diseases.
Collapse
Affiliation(s)
- Ali Cüvitoğlu
- The Graduate School of Natural and Applied Sciences, Dokuz Eylül University, Izmir, Turkiye
| | - Zerrin Isik
- Computer Engineering Department, Engineering Faculty, Dokuz Eylül University, Izmir, Turkiye
| |
Collapse
|
4
|
Ahmed F, Samantasinghar A, Manzoor Soomro A, Kim S, Hyun Choi K. A systematic review of computational approaches to understand cancer biology for informed drug repurposing. J Biomed Inform 2023; 142:104373. [PMID: 37120047 DOI: 10.1016/j.jbi.2023.104373] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/25/2023] [Accepted: 04/23/2023] [Indexed: 05/01/2023]
Abstract
Cancer is the second leading cause of death globally, trailing only heart disease. In the United States alone, 1.9 million new cancer cases and 609,360 deaths were recorded for 2022. Unfortunately, the success rate for new cancer drug development remains less than 10%, making the disease particularly challenging. This low success rate is largely attributed to the complex and poorly understood nature of cancer etiology. Therefore, it is critical to find alternative approaches to understanding cancer biology and developing effective treatments. One such approach is drug repurposing, which offers a shorter drug development timeline and lower costs while increasing the likelihood of success. In this review, we provide a comprehensive analysis of computational approaches for understanding cancer biology, including systems biology, multi-omics, and pathway analysis. Additionally, we examine the use of these methods for drug repurposing in cancer, including the databases and tools that are used for cancer research. Finally, we present case studies of drug repurposing, discussing their limitations and offering recommendations for future research in this area.
Collapse
Affiliation(s)
- Faheem Ahmed
- Department of Mechatronics Engineering, Jeju National University, Republic of Korea
| | | | | | - Sejong Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea; Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea.
| | - Kyung Hyun Choi
- Department of Mechatronics Engineering, Jeju National University, Republic of Korea.
| |
Collapse
|
5
|
Casillas-Espinosa PM, Anderson A, Harutyunyan A, Li C, Lee J, Braine EL, Brady RD, Sun M, Huang C, Barlow CK, Shah AD, Schittenhelm RB, Mychasiuk R, Jones NC, Shultz SR, O'Brien TJ. Disease-modifying effects of sodium selenate in a model of drug-resistant, temporal lobe epilepsy. eLife 2023; 12:e78877. [PMID: 36892461 PMCID: PMC10208637 DOI: 10.7554/elife.78877] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 03/08/2023] [Indexed: 03/10/2023] Open
Abstract
There are no pharmacological disease-modifying treatments with an enduring effect to mitigate the seizures and comorbidities of established chronic temporal lobe epilepsy (TLE). This study aimed to evaluate for disease modifying effects of sodium selenate treatment in the chronically epileptic rat post-status epilepticus (SE) model of drug-resistant TLE. Wistar rats underwent kainic acid-induced SE or sham. Ten-weeks post-SE, animals received sodium selenate, levetiracetam, or vehicle subcutaneousinfusion continuously for 4 weeks. To evaluate the effects of the treatments, one week of continuous video-EEG was acquired before, during, and 4, 8 weeks post-treatment, followed by behavioral tests. Targeted and untargeted proteomics and metabolomics were performed on post-mortem brain tissue to identify potential pathways associated with modified disease outcomes. Telomere length was investigated as a novel surrogate marker of epilepsy disease severity in our current study. The results showed that sodium selenate treatment was associated with mitigation of measures of disease severity at 8 weeks post-treatment cessation; reducing the number of spontaneous seizures (p< 0.05), cognitive dysfunction (p< 0.05), and sensorimotor deficits (p< 0.01). Moreover, selenate treatment was associated with increased protein phosphatase 2A (PP2A) expression, reduced hyperphosphorylated tau, and reversed telomere length shortening (p< 0.05). Network medicine integration of multi-omics/pre-clinical outcomes identified protein-metabolite modules positively correlated with TLE. Our results provide evidence that treatment with sodium selenate results in a sustained disease-modifying effect in chronically epileptic rats in the post-KA SE model of TLE, including improved comorbid learning and memory deficits.
Collapse
Affiliation(s)
- Pablo M Casillas-Espinosa
- Department of Medicine, The Royal Melbourne Hospital, The University of MelbourneMelbourneAustralia
- Department of Neuroscience, Central Clinical School, Monash UniversityMelbourneAustralia
- Monash Proteomics & Metabolomics Facility and Monash Biomedicine Discovery Institute, Monash UniversityClayton, VictoriaAustralia
| | - Alison Anderson
- Department of Medicine, The Royal Melbourne Hospital, The University of MelbourneMelbourneAustralia
- Department of Neuroscience, Central Clinical School, Monash UniversityMelbourneAustralia
| | - Anna Harutyunyan
- Department of Medicine, The Royal Melbourne Hospital, The University of MelbourneMelbourneAustralia
- Department of Neuroscience, Central Clinical School, Monash UniversityMelbourneAustralia
| | - Crystal Li
- Department of Neuroscience, Central Clinical School, Monash UniversityMelbourneAustralia
| | - Jiyoon Lee
- Department of Medicine, The Royal Melbourne Hospital, The University of MelbourneMelbourneAustralia
| | - Emma L Braine
- Department of Medicine, The Royal Melbourne Hospital, The University of MelbourneMelbourneAustralia
- Department of Neuroscience, Central Clinical School, Monash UniversityMelbourneAustralia
| | - Rhys D Brady
- Department of Medicine, The Royal Melbourne Hospital, The University of MelbourneMelbourneAustralia
- Department of Neuroscience, Central Clinical School, Monash UniversityMelbourneAustralia
| | - Mujun Sun
- Department of Neuroscience, Central Clinical School, Monash UniversityMelbourneAustralia
| | - Cheng Huang
- Department of Neurology, The Alfred Hospital, Commercial Road,Melbourne, VictoriaAustralia
| | - Christopher K Barlow
- Department of Neurology, The Alfred Hospital, Commercial Road,Melbourne, VictoriaAustralia
| | - Anup D Shah
- Department of Neurology, The Alfred Hospital, Commercial Road,Melbourne, VictoriaAustralia
| | - Ralf B Schittenhelm
- Department of Neurology, The Alfred Hospital, Commercial Road,Melbourne, VictoriaAustralia
| | - Richelle Mychasiuk
- Department of Neuroscience, Central Clinical School, Monash UniversityMelbourneAustralia
| | - Nigel C Jones
- Department of Medicine, The Royal Melbourne Hospital, The University of MelbourneMelbourneAustralia
- Department of Neuroscience, Central Clinical School, Monash UniversityMelbourneAustralia
| | - Sandy R Shultz
- Department of Medicine, The Royal Melbourne Hospital, The University of MelbourneMelbourneAustralia
- Department of Neuroscience, Central Clinical School, Monash UniversityMelbourneAustralia
| | - Terence J O'Brien
- Department of Medicine, The Royal Melbourne Hospital, The University of MelbourneMelbourneAustralia
- Department of Neuroscience, Central Clinical School, Monash UniversityMelbourneAustralia
- Monash Proteomics & Metabolomics Facility and Monash Biomedicine Discovery Institute, Monash UniversityClayton, VictoriaAustralia
| |
Collapse
|
6
|
Nayak SS, Sundararajan V. Robust anti-inflammatory activity of genistein against neutrophil elastase: a microsecond molecular dynamics simulation study. J Biomol Struct Dyn 2023; 41:11612-11628. [PMID: 36705087 DOI: 10.1080/07391102.2023.2170919] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 12/26/2022] [Indexed: 01/28/2023]
Abstract
Human Neutrophil Elastase (HNE) is one of the major causes of tissue destruction in numerous chronic and inflammatory disorders and has been reported as a therapeutic target for inflammatory diseases. Overexpression of this enzyme plays a critical role in the pathogenesis of rheumatoid arthritis (RA). The focus of this study is to identify potent natural inhibitors that could target the active site of the HNE through the use of computational methods. The molecular structure of small molecules was retrieved from several natural compound databases. This was followed by structure-based virtual screening, molecular docking, ADMET property predictions and molecular dynamic simulation studies to screen potential HNE inhibitors. In total, 1881 natural compounds were extracted and subjected to molecular docking studies, and 10 compounds were found to have good interactions, exhibiting the best docking scores. Genistein showed higher binding efficacy (-10.28 Kcal/mol) to HNE in comparison to other natural compounds. The conformational stability of the docked complex of the ELANE gene (HNE) with genistein was assessed using 1-microsecond molecular dynamic simulation (MDs), which reliably revealed the unique stereochemical alteration of the complex, indicating its conformational stability and flexibility. Alterations in the enzyme structure upon complex formation were further characterized through clustering analysis and linear interaction energy (LIE) calculation. The outcomes of this research propose novel potential candidates against target HNE.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Smruti Sudha Nayak
- Department of Bio-Sciences, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamilnadu, India
| | - Vino Sundararajan
- Department of Bio-Sciences, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamilnadu, India
| |
Collapse
|
7
|
Qin S, Li W, Yu H, Xu M, Li C, Fu L, Sun S, He Y, Lv J, He W, Chen L. Guiding Drug Repositioning for Cancers Based on Drug Similarity Networks. Int J Mol Sci 2023; 24:ijms24032244. [PMID: 36768566 PMCID: PMC9917231 DOI: 10.3390/ijms24032244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 01/05/2023] [Accepted: 01/16/2023] [Indexed: 01/24/2023] Open
Abstract
Drug repositioning aims to discover novel clinical benefits of existing drugs, is an effective way to develop drugs for complex diseases such as cancer and may facilitate the process of traditional drug development. Meanwhile, network-based computational biology approaches, which allow the integration of information from different aspects to understand the relationships between biomolecules, has been successfully applied to drug repurposing. In this work, we developed a new strategy for network-based drug repositioning against cancer. Combining the mechanism of action and clinical efficacy of the drugs, a cancer-related drug similarity network was constructed, and the correlation score of each drug with a specific cancer was quantified. The top 5% of scoring drugs were reviewed for stability and druggable potential to identify potential repositionable drugs. Of the 11 potentially repurposable drugs for non-small cell lung cancer (NSCLC), 10 were confirmed by clinical trial articles and databases. The targets of these drugs were significantly enriched in cancer-related pathways and significantly associated with the prognosis of NSCLC. In light of the successful application of our approach to colorectal cancer as well, it provides an effective clue and valuable perspective for drug repurposing in cancer.
Collapse
Affiliation(s)
- Shimei Qin
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Wan Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Hongzheng Yu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Manyi Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Chao Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Lei Fu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Shibin Sun
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Yuehan He
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Junjie Lv
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Weiming He
- Institute of Opto-Electronics, Harbin Institute of Technology, Harbin 150001, China
| | - Lina Chen
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
- Correspondence: ; Tel.: +86-451-8667-4768
| |
Collapse
|
8
|
Zhang ML, Zhao BW, Su XR, He YZ, Yang Y, Hu L. RLFDDA: a meta-path based graph representation learning model for drug-disease association prediction. BMC Bioinformatics 2022; 23:516. [PMID: 36456957 PMCID: PMC9713188 DOI: 10.1186/s12859-022-05069-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 11/21/2022] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Drug repositioning is a very important task that provides critical information for exploring the potential efficacy of drugs. Yet developing computational models that can effectively predict drug-disease associations (DDAs) is still a challenging task. Previous studies suggest that the accuracy of DDA prediction can be improved by integrating different types of biological features. But how to conduct an effective integration remains a challenging problem for accurately discovering new indications for approved drugs. METHODS In this paper, we propose a novel meta-path based graph representation learning model, namely RLFDDA, to predict potential DDAs on heterogeneous biological networks. RLFDDA first calculates drug-drug similarities and disease-disease similarities as the intrinsic biological features of drugs and diseases. A heterogeneous network is then constructed by integrating DDAs, disease-protein associations and drug-protein associations. With such a network, RLFDDA adopts a meta-path random walk model to learn the latent representations of drugs and diseases, which are concatenated to construct joint representations of drug-disease associations. As the last step, we employ the random forest classifier to predict potential DDAs with their joint representations. RESULTS To demonstrate the effectiveness of RLFDDA, we have conducted a series of experiments on two benchmark datasets by following a ten-fold cross-validation scheme. The results show that RLFDDA yields the best performance in terms of AUC and F1-score when compared with several state-of-the-art DDAs prediction models. We have also conducted a case study on two common diseases, i.e., paclitaxel and lung tumors, and found that 7 out of top-10 diseases and 8 out of top-10 drugs have already been validated for paclitaxel and lung tumors respectively with literature evidence. Hence, the promising performance of RLFDDA may provide a new perspective for novel DDAs discovery over heterogeneous networks.
Collapse
Affiliation(s)
- Meng-Long Zhang
- The Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi, China
- University of Chinese Academy of Sciences, Beijing, China
- Xinjiang Laboratory of Minority Speech and Language Information Processing, Urumqi, China
| | - Bo-Wei Zhao
- The Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi, China
- University of Chinese Academy of Sciences, Beijing, China
- Xinjiang Laboratory of Minority Speech and Language Information Processing, Urumqi, China
| | - Xiao-Rui Su
- The Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi, China
- University of Chinese Academy of Sciences, Beijing, China
- Xinjiang Laboratory of Minority Speech and Language Information Processing, Urumqi, China
| | - Yi-Zhou He
- School of Computer Science and Technology, Wuhan University of Technology, Wuhan, China
| | - Yue Yang
- School of Computer Science and Technology, Wuhan University of Technology, Wuhan, China
| | - Lun Hu
- The Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi, China
- University of Chinese Academy of Sciences, Beijing, China
- Xinjiang Laboratory of Minority Speech and Language Information Processing, Urumqi, China
| |
Collapse
|
9
|
A comprehensive review of Artificial Intelligence and Network based approaches to drug repurposing in Covid-19. Biomed Pharmacother 2022; 153:113350. [PMID: 35777222 PMCID: PMC9236981 DOI: 10.1016/j.biopha.2022.113350] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/22/2022] [Accepted: 06/24/2022] [Indexed: 11/26/2022] Open
Abstract
Conventional drug discovery and development is tedious and time-taking process; because of which it has failed to keep the required pace to mitigate threats and cater demands of viral and re-occurring diseases, such as Covid-19. The main reasons of this delay in traditional drug development are: high attrition rates, extensive time requirements, and huge financial investment with significant risk. The effective solution to de novo drug discovery is drug repurposing. Previous studies have shown that the network-based approaches and analysis are versatile platform for repurposing as the network biology is used to model the interactions between variety of biological concepts. Herein, we provide a comprehensive background of machine learning and deep learning in drug repurposing while specifically focusing on the applications of network-based approach to drug repurposing in Covid-19, data sources, and tools used. Furthermore, use of network proximity, network diffusion, and AI on network-based drug repurposing for Covid-19 is well-explained. Finally, limitations of network-based approaches in general and specific to network are stated along with future recommendations for better network-based models.
Collapse
|
10
|
Sadeghi SS, Keyvanpour MR. An Analytical Review of Computational Drug Repurposing. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2021; 18:472-488. [PMID: 31403439 DOI: 10.1109/tcbb.2019.2933825] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Drug repurposing is a vital function in pharmaceutical fields and has gained popularity in recent years in both the pharmaceutical industry and research community. It refers to the process of discovering new uses and indications for existing or failed drugs. It is cost-effective and reliable in contrast to experimental drug discovery, which is a costly, time-consuming, and risky process and limited to a relatively small number of targets. Accordingly, a plethora of computational methodologies have been propounded to repurpose drugs on a large scale by utilizing available high throughput data. The available literature, however, lacks a contemporary and comprehensive analysis of the current computational drug repurposing methodologies. In this paper, we presented a systematic analysis of computational drug repurposing which consists of three main sections: Initially, we categorize the computational drug repurposing methods based on their technical approach and artificial intelligence perspective and discuss the strengths and weaknesses of various methods. Secondly, some general criteria are recommended to analyze our proposed categorization. In the third and final section, a qualitative comparison is made between each approach which is a guide to understanding their preference to one another. Further, this systematic analysis can help in the efficient selection and improvement of drug repurposing techniques based on the nature of computational methods implemented on biological resources.
Collapse
|
11
|
Roy S, Dhaneshwar S, Bhasin B. Drug Repurposing: An Emerging Tool for Drug Reuse, Recycling and Discovery. Curr Drug Res Rev 2021; 13:101-119. [PMID: 33573567 DOI: 10.2174/2589977513666210211163711] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 09/07/2020] [Accepted: 10/26/2020] [Indexed: 11/22/2022]
Abstract
Drug repositioning or repurposing is a revolutionary breakthrough in drug development that focuses on rediscovering new uses for old therapeutic agents. Drug repositioning can be defined more precisely as the process of exploring new indications for an already approved drug while drug repurposing includes overall re-development approaches grounded in the identical chemical structure of the active drug moiety as in the original product. The repositioning approach accelerates the drug development process, curtails the cost and risk inherent to drug development. The strategy focuses on the polypharmacology of drugs to unlocks novel opportunities for logically designing more efficient therapeutic agents for unmet medical disorders. Drug repositioning also expresses certain regulatory challenges that hamper its further utilization. The review outlines the eminent role of drug repositioning in new drug discovery, methods to predict the molecular targets of a drug molecule, advantages that the strategy offers to the pharmaceutical industries, explaining how the industrial collaborations with academics can assist in the discovering more repositioning opportunities. The focus of the review is to highlight the latest applications of drug repositioning in various disorders. The review also includes a comparison of old and new therapeutic uses of repurposed drugs, assessing their novel mechanisms of action and pharmacological effects in the management of various disorders. Various restrictions and challenges that repurposed drugs come across during their development and regulatory phases are also highlighted.
Collapse
Affiliation(s)
- Supriya Roy
- Amity Institute of Pharmacy, Amity University Uttar Pradesh, Lucknow Campus, India
| | - Suneela Dhaneshwar
- Amity Institute of Pharmacy, Amity University Uttar Pradesh, Lucknow Campus, India
| | - Bhavya Bhasin
- Poona College of Pharmacy, Bharati Vidyapeeth University, Pune, India
| |
Collapse
|
12
|
Vanhaelen Q. Web-based Tools for Drug Repurposing: Successful Examples of Collaborative Research. Curr Med Chem 2021; 28:181-195. [PMID: 32003659 DOI: 10.2174/0929867327666200128111925] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 11/23/2019] [Accepted: 11/30/2019] [Indexed: 11/22/2022]
Abstract
Computational approaches have been proven to be complementary tools of interest in identifying potential candidates for drug repurposing. However, although the methods developed so far offer interesting opportunities and could contribute to solving issues faced by the pharmaceutical sector, they also come with their constraints. Indeed, specific challenges ranging from data access, standardization and integration to the implementation of reliable and coherent validation methods must be addressed to allow systematic use at a larger scale. In this mini-review, we cover computational tools recently developed for addressing some of these challenges. This includes specific databases providing accessibility to a large set of curated data with standardized annotations, web-based tools integrating flexible user interfaces to perform fast computational repurposing experiments and standardized datasets specifically annotated and balanced for validating new computational drug repurposing methods. Interestingly, these new databases combined with the increasing number of information about the outcomes of drug repurposing studies can be used to perform a meta-analysis to identify key properties associated with successful drug repurposing cases. This information could further be used to design estimation methods to compute a priori assessment of the repurposing possibilities.
Collapse
Affiliation(s)
- Quentin Vanhaelen
- Insilico Medicine, 307A, Core Building 1, 1 Science Park East Avenue, Hong Kong Science Park, Pak Shek Kok, Hong Kong
| |
Collapse
|
13
|
Luo H, Li M, Yang M, Wu FX, Li Y, Wang J. Biomedical data and computational models for drug repositioning: a comprehensive review. Brief Bioinform 2020; 22:1604-1619. [PMID: 32043521 DOI: 10.1093/bib/bbz176] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 12/07/2019] [Accepted: 12/26/2019] [Indexed: 12/16/2022] Open
Abstract
Drug repositioning can drastically decrease the cost and duration taken by traditional drug research and development while avoiding the occurrence of unforeseen adverse events. With the rapid advancement of high-throughput technologies and the explosion of various biological data and medical data, computational drug repositioning methods have been appealing and powerful techniques to systematically identify potential drug-target interactions and drug-disease interactions. In this review, we first summarize the available biomedical data and public databases related to drugs, diseases and targets. Then, we discuss existing drug repositioning approaches and group them based on their underlying computational models consisting of classical machine learning, network propagation, matrix factorization and completion, and deep learning based models. We also comprehensively analyze common standard data sets and evaluation metrics used in drug repositioning, and give a brief comparison of various prediction methods on the gold standard data sets. Finally, we conclude our review with a brief discussion on challenges in computational drug repositioning, which includes the problem of reducing the noise and incompleteness of biomedical data, the ensemble of various computation drug repositioning methods, the importance of designing reliable negative samples selection methods, new techniques dealing with the data sparseness problem, the construction of large-scale and comprehensive benchmark data sets and the analysis and explanation of the underlying mechanisms of predicted interactions.
Collapse
Affiliation(s)
- Huimin Luo
- School of Computer Science and Engineering at Central South University
| | - Min Li
- School of Computer Science and Engineering at Central South University
| | - Mengyun Yang
- School of Computer Science and Engineering at Central South University
| | - Fang-Xiang Wu
- College of Engineering and the Department of Computer Science at University of Saskatchewan, Saskatoon, Canada
| | - Yaohang Li
- Department of Computer Science at Old Dominion University, Norfolk, USA
| | - Jianxin Wang
- School of Computer Science and Engineering at Central South University
| |
Collapse
|
14
|
Gupta V, Crudu A, Matsuoka Y, Ghosh S, Rozot R, Marat X, Jäger S, Kitano H, Breton L. Multi-dimensional computational pipeline for large-scale deep screening of compound effect assessment: an in silico case study on ageing-related compounds. NPJ Syst Biol Appl 2019; 5:42. [PMID: 31798962 PMCID: PMC6879499 DOI: 10.1038/s41540-019-0119-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 09/23/2019] [Indexed: 12/18/2022] Open
Abstract
Designing alternative approaches to efficiently screen chemicals on the efficacy landscape is a challenging yet indispensable task in the current compound profiling methods. Particularly, increasing regulatory restrictions underscore the need to develop advanced computational pipelines for efficacy assessment of chemical compounds as alternative means to reduce and/or replace in vivo experiments. Here, we present an innovative computational pipeline for large-scale assessment of chemical compounds by analysing and clustering chemical compounds on the basis of multiple dimensions-structural similarity, binding profiles and their network effects across pathways and molecular interaction maps-to generate testable hypotheses on the pharmacological landscapes as well as identify potential mechanisms of efficacy on phenomenological processes. Further, we elucidate the application of the pipeline on a screen of anti-ageing-related compounds to cluster the candidates based on their structure, docking profile and network effects on fundamental metabolic/molecular pathways associated with the cell vitality, highlighting emergent insights on compounds activities based on the multi-dimensional deep screen pipeline.
Collapse
Affiliation(s)
| | - Alina Crudu
- L’Oréal Research and Innovation, Aulnay-sous-Bois, France
| | | | | | - Roger Rozot
- L’Oréal Research and Innovation, Aulnay-sous-Bois, France
| | - Xavier Marat
- L’Oréal Research and Innovation, Aulnay-sous-Bois, France
| | - Sibylle Jäger
- L’Oréal Research and Innovation, Aulnay-sous-Bois, France
| | - Hiroaki Kitano
- The Systems Biology Institute, Tokyo, Japan
- Okinawa Institute of Science and Technology, Okinawa, Japan
| | - Lionel Breton
- L’Oréal Research and Innovation, Aulnay-sous-Bois, France
| |
Collapse
|
15
|
Kumar R, Harilal S, Gupta SV, Jose J, Thomas Parambi DG, Uddin MS, Shah MA, Mathew B. Exploring the new horizons of drug repurposing: A vital tool for turning hard work into smart work. Eur J Med Chem 2019; 182:111602. [PMID: 31421629 PMCID: PMC7127402 DOI: 10.1016/j.ejmech.2019.111602] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 08/07/2019] [Accepted: 08/07/2019] [Indexed: 02/07/2023]
Abstract
Drug discovery and development are long and financially taxing processes. On an average it takes 12-15 years and costs 1.2 billion USD for successful drug discovery and approval for clinical use. Many lead molecules are not developed further and their potential is not tapped to the fullest due to lack of resources or time constraints. In order for a drug to be approved by FDA for clinical use, it must have excellent therapeutic potential in the desired area of target with minimal toxicities as supported by both pre-clinical and clinical studies. The targeted clinical evaluations fail to explore other potential therapeutic applications of the candidate drug. Drug repurposing or repositioning is a fast and relatively cheap alternative to the lengthy and expensive de novo drug discovery and development. Drug repositioning utilizes the already available clinical trials data for toxicity and adverse effects, at the same time explores the drug's therapeutic potential for a different disease. This review addresses recent developments and future scope of drug repositioning strategy.
Collapse
Affiliation(s)
- Rajesh Kumar
- Department of Pharmacy, Kerala University of Health Sciences, Thrissur, Kerala, India
| | - Seetha Harilal
- Department of Pharmacy, Kerala University of Health Sciences, Thrissur, Kerala, India
| | - Sheeba Varghese Gupta
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, 33612, USA
| | - Jobin Jose
- Department of Pharmaceutics, NGSM Institute of Pharmaceutical Science, NITTE Deemed to be University, Manglore, 575018, India
| | - Della Grace Thomas Parambi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Sakaka, Al Jouf, 2014, Saudi Arabia
| | - Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh; Pharmakon Neuroscience Research Network, Dhaka, Bangladesh
| | - Muhammad Ajmal Shah
- Department of Pharmacogonosy, Faculty of Pharmaceutical Sciences, Government College University, Faisalabad, Pakistan
| | - Bijo Mathew
- Division of Drug Design and Medicinal Chemistry Research Lab, Department of Pharmaceutical Chemistry, Ahalia School of Pharmacy, Palakkad, 678557, Kerala, India.
| |
Collapse
|
16
|
Turanli B, Altay O, Borén J, Turkez H, Nielsen J, Uhlen M, Arga KY, Mardinoglu A. Systems biology based drug repositioning for development of cancer therapy. Semin Cancer Biol 2019; 68:47-58. [PMID: 31568815 DOI: 10.1016/j.semcancer.2019.09.020] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 09/23/2019] [Accepted: 09/24/2019] [Indexed: 01/20/2023]
Abstract
Drug repositioning is a powerful method that can assists the conventional drug discovery process by using existing drugs for treatment of a disease rather than its original indication. The first examples of repurposed drugs were discovered serendipitously, however data accumulated by high-throughput screenings and advancements in computational biology methods have paved the way for rational drug repositioning methods. As chemotherapeutic agents have notorious side effects that significantly reduce quality of life, drug repositioning promises repurposed noncancer drugs with little or tolerable adverse effects for cancer patients. Here, we review current drug-related data types and databases including some examples of web-based drug repositioning tools. Next, we describe systems biology approaches to be used in drug repositioning for effective cancer therapy. Finally, we highlight examples of mostly repurposed drugs for cancer treatment and provide an overview of future expectations in the field for development of effective treatment strategies.
Collapse
Affiliation(s)
- Beste Turanli
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm SE-17121, Sweden; Department of Bioengineering, Marmara University, Istanbul, Turkey; Department of Bioengineering, Istanbul Medeniyet University, Istanbul, Turkey
| | - Ozlem Altay
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm SE-17121, Sweden
| | - Jan Borén
- Department of Molecular and Clinical Medicine, University of Gothenburg and Sahlgrenska University Hospital Gothenburg, Sweden
| | - Hasan Turkez
- Department of Molecular Biology and Genetics, Erzurum Technical University, Erzurum 25240, Turkey
| | - Jens Nielsen
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Mathias Uhlen
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm SE-17121, Sweden
| | | | - Adil Mardinoglu
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm SE-17121, Sweden; Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London SE1 9RT, United Kingdom.
| |
Collapse
|
17
|
Lotfi Shahreza M, Ghadiri N, Mousavi SR, Varshosaz J, Green JR. A review of network-based approaches to drug repositioning. Brief Bioinform 2019; 19:878-892. [PMID: 28334136 DOI: 10.1093/bib/bbx017] [Citation(s) in RCA: 176] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Indexed: 01/17/2023] Open
Abstract
Experimental drug development is time-consuming, expensive and limited to a relatively small number of targets. However, recent studies show that repositioning of existing drugs can function more efficiently than de novo experimental drug development to minimize costs and risks. Previous studies have proven that network analysis is a versatile platform for this purpose, as the biological networks are used to model interactions between many different biological concepts. The present study is an attempt to review network-based methods in predicting drug targets for drug repositioning. For each method, the preferred type of data set is described, and their advantages and limitations are discussed. For each method, we seek to provide a brief description, as well as an evaluation based on its performance metrics.We conclude that integrating distinct and complementary data should be used because each type of data set reveals a unique aspect of information about an organism. We also suggest that applying a standard set of evaluation metrics and data sets would be essential in this fast-growing research domain.
Collapse
Affiliation(s)
- Maryam Lotfi Shahreza
- Department of Electrical and Computer Engineering, Isfahan University of Technology, Isfahan, Iran
| | - Nasser Ghadiri
- Department of Electrical and Computer Engineering, Isfahan University of Technology, Isfahan, Iran
| | | | - Jaleh Varshosaz
- Drug Delivery Systems Research Center of Isfahan University of Medical Sciences
| | - James R Green
- Department of Electrical and Computer Engineering, Isfahan University of Technology, Isfahan, Iran
| |
Collapse
|
18
|
Xue H, Li J, Xie H, Wang Y. Review of Drug Repositioning Approaches and Resources. Int J Biol Sci 2018; 14:1232-1244. [PMID: 30123072 PMCID: PMC6097480 DOI: 10.7150/ijbs.24612] [Citation(s) in RCA: 336] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Accepted: 06/12/2018] [Indexed: 12/23/2022] Open
Abstract
Drug discovery is a time-consuming, high-investment, and high-risk process in traditional drug development. Drug repositioning has become a popular strategy in recent years. Different from traditional drug development strategies, the strategy is efficient, economical and riskless. There are usually three kinds of approaches: computational approaches, biological experimental approaches, and mixed approaches, all of which are widely used in drug repositioning. In this paper, we reviewed computational approaches and highlighted their characteristics to provide references for researchers to develop more powerful approaches. At the same time, the important findings obtained using these approaches are listed. Furthermore, we summarized 76 important resources about drug repositioning. Finally, challenges and opportunities in drug repositioning are discussed from multiple perspectives, including technology, commercial models, patents and investment.
Collapse
Affiliation(s)
- Hanqing Xue
- School of Computer Science and Technology, Harbin Institute of Technology, 150001, Harbin, China
| | - Jie Li
- School of Computer Science and Technology, Harbin Institute of Technology, 150001, Harbin, China
| | - Haozhe Xie
- School of Computer Science and Technology, Harbin Institute of Technology, 150001, Harbin, China
| | - Yadong Wang
- School of Computer Science and Technology, Harbin Institute of Technology, 150001, Harbin, China
| |
Collapse
|
19
|
Vanhaelen Q, Mamoshina P, Aliper AM, Artemov A, Lezhnina K, Ozerov I, Labat I, Zhavoronkov A. Design of efficient computational workflows for in silico drug repurposing. Drug Discov Today 2016; 22:210-222. [PMID: 27693712 DOI: 10.1016/j.drudis.2016.09.019] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 08/26/2016] [Accepted: 09/21/2016] [Indexed: 12/22/2022]
Abstract
Here, we provide a comprehensive overview of the current status of in silico repurposing methods by establishing links between current technological trends, data availability and characteristics of the algorithms used in these methods. Using the case of the computational repurposing of fasudil as an alternative autophagy enhancer, we suggest a generic modular organization of a repurposing workflow. We also review 3D structure-based, similarity-based, inference-based and machine learning (ML)-based methods. We summarize the advantages and disadvantages of these methods to emphasize three current technical challenges. We finish by discussing current directions of research, including possibilities offered by new methods, such as deep learning.
Collapse
Affiliation(s)
- Quentin Vanhaelen
- Insilico Medicine Inc., Johns Hopkins University, ETC, B301, MD 21218, USA.
| | - Polina Mamoshina
- Insilico Medicine Inc., Johns Hopkins University, ETC, B301, MD 21218, USA
| | - Alexander M Aliper
- Insilico Medicine Inc., Johns Hopkins University, ETC, B301, MD 21218, USA
| | - Artem Artemov
- Insilico Medicine Inc., Johns Hopkins University, ETC, B301, MD 21218, USA
| | - Ksenia Lezhnina
- Insilico Medicine Inc., Johns Hopkins University, ETC, B301, MD 21218, USA
| | - Ivan Ozerov
- Insilico Medicine Inc., Johns Hopkins University, ETC, B301, MD 21218, USA
| | - Ivan Labat
- BioTime Inc., 1010 Atlantic Avenue, 102, Alameda, CA 94501, USA
| | - Alex Zhavoronkov
- Insilico Medicine Inc., Johns Hopkins University, ETC, B301, MD 21218, USA
| |
Collapse
|
20
|
Langley G. Response to "Comment on 'Lessons from Toxicology: Developing a 21st-Century Paradigm for Medical Research'". ENVIRONMENTAL HEALTH PERSPECTIVES 2016; 124:A85. [PMID: 27135756 PMCID: PMC4858408 DOI: 10.1289/ehp.1611305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Affiliation(s)
- Gill Langley
- Address correspondence to G. Langley, 8 Crow Furlong, Hitchin, Hertfordshire, SG5 2HW, United Kingdom. E-mail:
| |
Collapse
|
21
|
Barneh F, Jafari M, Mirzaie M. Updates on drug-target network; facilitating polypharmacology and data integration by growth of DrugBank database. Brief Bioinform 2015; 17:1070-1080. [PMID: 26490381 DOI: 10.1093/bib/bbv094] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2015] [Revised: 09/16/2015] [Indexed: 02/07/2023] Open
Abstract
Network pharmacology elucidates the relationship between drugs and targets. As the identified targets for each drug increases, the corresponding drug-target network (DTN) evolves from solely reflection of the pharmaceutical industry trend to a portrait of polypharmacology. The aim of this study was to evaluate the potentials of DrugBank database in advancing systems pharmacology. We constructed and analyzed DTN from drugs and targets associations in the DrugBank 4.0 database. Our results showed that in bipartite DTN, increased ratio of identified targets for drugs augmented density and connectivity of drugs and targets and decreased modular structure. To clear up the details in the network structure, the DTNs were projected into two networks namely, drug similarity network (DSN) and target similarity network (TSN). In DSN, various classes of Food and Drug Administration-approved drugs with distinct therapeutic categories were linked together based on shared targets. Projected TSN also showed complexity because of promiscuity of the drugs. By including investigational drugs that are currently being tested in clinical trials, the networks manifested more connectivity and pictured the upcoming pharmacological space in the future years. Diverse biological processes and protein-protein interactions were manipulated by new drugs, which can extend possible target combinations. We conclude that network-based organization of DrugBank 4.0 data not only reveals the potential for repurposing of existing drugs, also allows generating novel predictions about drugs off-targets, drug-drug interactions and their side effects. Our results also encourage further effort for high-throughput identification of targets to build networks that can be integrated into disease networks.
Collapse
|
22
|
Systems Medicine: The Application of Systems Biology Approaches for Modern Medical Research and Drug Development. Mol Biol Int 2015; 2015:698169. [PMID: 26357572 PMCID: PMC4556074 DOI: 10.1155/2015/698169] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Revised: 07/27/2015] [Accepted: 07/29/2015] [Indexed: 12/21/2022] Open
Abstract
The exponential development of highly advanced scientific and medical research technologies throughout the past 30 years has arrived to the point where the high number of characterized molecular agents related to pathogenesis cannot be readily integrated or processed by conventional analytical approaches. Indeed, the realization that several moieties are signatures of disease has partly led to the increment of complex diseases being characterized. Scientists and clinicians can now investigate and analyse any individual dysregulations occurring within the genomic, transcriptomic, miRnomic, proteomic, and metabolomic levels thanks to currently available advanced technologies. However, there are drawbacks within this scientific brave new age in that only isolated molecular levels are individually investigated for their influence in affecting any particular health condition. Since their conception in 1992, systems biology/medicine focuses mainly on the perturbations of overall pathway kinetics for the consequent onset and/or deterioration of the investigated condition/s. Systems medicine approaches can therefore be employed for shedding light in multiple research scenarios, ultimately leading to the practical result of uncovering novel dynamic interaction networks that are critical for influencing the course of medical conditions. Consequently, systems medicine also serves to identify clinically important molecular targets for diagnostic and therapeutic measures against such a condition.
Collapse
|
23
|
Dharmaprakash A, Thandavarayan R, Joseph I, Thomas S. Development of broad-spectrum antibiofilm drugs: strategies and challenges. Future Microbiol 2015; 10:1035-48. [DOI: 10.2217/fmb.15.14] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
ABSTRACT The severity of many chronic bacterial infections is mainly due to the biofilm mode of life adapted by pathogenic bacteria. The bacteria in biofilm-stage exhibit high resistance to host immune responses and antimicrobials, which complicates the treatment process and results in life threatening conditions. Most of the chronic infections are polymicrobial in nature. In order to combat the polymicrobial biofilm infections and to increase the efficiency of antimicrobials, there is an urgent need for broad-spectrum antibiofilm drugs. This review discusses the clinical needs and current status of broad-spectrum antibiofilm drugs with special emphasis on prospective strategies and hurdles in the process of new drug discovery.
Collapse
Affiliation(s)
- Akhilandeswarre Dharmaprakash
- Cholera & Biofilm Research Laboratory, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram – 695 014, Kerala, India
| | | | - Iype Joseph
- Pathogen Biology Program, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram – 695 014, Kerala, India
| | - Sabu Thomas
- Cholera & Biofilm Research Laboratory, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram – 695 014, Kerala, India
| |
Collapse
|