1
|
Glajzner P, Bernat A, Jasińska-Stroschein M. Improving the treatment of bacterial infections caused by multidrug-resistant bacteria through drug repositioning. Front Pharmacol 2024; 15:1397602. [PMID: 38910882 PMCID: PMC11193365 DOI: 10.3389/fphar.2024.1397602] [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: 03/07/2024] [Accepted: 05/20/2024] [Indexed: 06/25/2024] Open
Abstract
Drug repurposing (repositioning) is a dynamically-developing area in the search for effective therapy of infectious diseases. Repositioning existing drugs with a well-known pharmacological and toxicological profile is an attractive method for quickly discovering new therapeutic indications. The off-label use of drugs for infectious diseases requires much less capital and time, and can hasten progress in the development of new antimicrobial drugs, including antibiotics. The use of drug repositioning in searching for new therapeutic options has brought promising results for many viral infectious diseases, such as Ebola, ZIKA, Dengue, and HCV. This review describes the most favorable results for repositioned drugs for the treatment of bacterial infections. It comprises publications from various databases including PubMed and Web of Science published from 2015 to 2023. The following search keywords/strings were used: drug repositioning and/or repurposing and/or antibacterial activity and/or infectious diseases. Treatment options for infections caused by multidrug-resistant bacteria were taken into account, including methicillin-resistant staphylococci, multidrug-resistant Mycobacterium tuberculosis, or carbapenem-resistant bacteria from the Enterobacteriaceae family. It analyses the safety profiles of the included drugs and their synergistic combinations with antibiotics and discusses the potential of antibacterial drugs with antiparasitic, anticancer, antipsychotic effects, and those used in metabolic diseases. Drug repositioning may be an effective response to public health threats related to the spread of multidrug-resistant bacterial strains and the growing antibiotic resistance of microorganisms.
Collapse
Affiliation(s)
- Paulina Glajzner
- Department of Biopharmacy, Faculty of Pharmacy, Medical University of Lodz, Łódź, Poland
| | | | | |
Collapse
|
2
|
Jamir E, Sarma H, Priyadarsinee L, Kiewhuo K, Nagamani S, Sastry GN. Polypharmacology guided drug repositioning approach for SARS-CoV2. PLoS One 2023; 18:e0289890. [PMID: 37556478 PMCID: PMC10411734 DOI: 10.1371/journal.pone.0289890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 07/27/2023] [Indexed: 08/11/2023] Open
Abstract
Drug repurposing has emerged as an important strategy and it has a great potential in identifying therapeutic applications for COVID-19. An extensive virtual screening of 4193 FDA approved drugs has been carried out against 24 proteins of SARS-CoV2 (NSP1-10 and NSP12-16, envelope, membrane, nucleoprotein, spike, ORF3a, ORF6, ORF7a, ORF8, and ORF9b). The drugs were classified into top 10 and bottom 10 drugs based on the docking scores followed by the distribution of their therapeutic indications. As a result, the top 10 drugs were found to have therapeutic indications for cancer, pain, neurological disorders, and viral and bacterial diseases. As drug resistance is one of the major challenges in antiviral drug discovery, polypharmacology and network pharmacology approaches were employed in the study to identify drugs interacting with multiple targets and drugs such as dihydroergotamine, ergotamine, bisdequalinium chloride, midostaurin, temoporfin, tirilazad, and venetoclax were identified among the multi-targeting drugs. Further, a pathway analysis of the genes related to the multi-targeting drugs was carried which provides insight into the mechanism of drugs and identifying targetable genes and biological pathways involved in SARS-CoV2.
Collapse
Affiliation(s)
- Esther Jamir
- Advanced Computation and Data Sciences Division, CSIR–North East Institute of Science and Technology, Jorhat, Assam, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Himakshi Sarma
- Advanced Computation and Data Sciences Division, CSIR–North East Institute of Science and Technology, Jorhat, Assam, India
| | - Lipsa Priyadarsinee
- Advanced Computation and Data Sciences Division, CSIR–North East Institute of Science and Technology, Jorhat, Assam, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Kikrusenuo Kiewhuo
- Advanced Computation and Data Sciences Division, CSIR–North East Institute of Science and Technology, Jorhat, Assam, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Selvaraman Nagamani
- Advanced Computation and Data Sciences Division, CSIR–North East Institute of Science and Technology, Jorhat, Assam, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - G. Narahari Sastry
- Advanced Computation and Data Sciences Division, CSIR–North East Institute of Science and Technology, Jorhat, Assam, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
3
|
Torricelli F, Sauta E, Manicardi V, Mandato VD, Palicelli A, Ciarrocchi A, Manzotti G. An Innovative Drug Repurposing Approach to Restrain Endometrial Cancer Metastatization. Cells 2023; 12:794. [PMID: 36899930 PMCID: PMC10001006 DOI: 10.3390/cells12050794] [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] [Received: 01/13/2023] [Revised: 02/22/2023] [Accepted: 02/28/2023] [Indexed: 03/06/2023] Open
Abstract
BACKGROUND Endometrial cancer (EC) is the most common gynecologic tumor and the world's fourth most common cancer in women. Most patients respond to first-line treatments and have a low risk of recurrence, but refractory patients, and those with metastatic cancer at diagnosis, remain with no treatment options. Drug repurposing aims to discover new clinical indications for existing drugs with known safety profiles. It provides ready-to-use new therapeutic options for highly aggressive tumors for which standard protocols are ineffective, such as high-risk EC. METHODS Here, we aimed at defining new therapeutic opportunities for high-risk EC using an innovative and integrated computational drug repurposing approach. RESULTS We compared gene-expression profiles, from publicly available databases, of metastatic and non-metastatic EC patients being metastatization the most severe feature of EC aggressiveness. A comprehensive analysis of transcriptomic data through a two-arm approach was applied to obtain a robust prediction of drug candidates. CONCLUSIONS Some of the identified therapeutic agents are already successfully used in clinical practice to treat other types of tumors. This highlights the potential to repurpose them for EC and, therefore, the reliability of the proposed approach.
Collapse
Affiliation(s)
- Federica Torricelli
- Laboratory of Translational Research, Azienda USL-IRCCS di Reggio Emilia, Viale Risorgimento 80, 42123 Reggio Emilia, Italy
| | - Elisabetta Sauta
- Laboratory of Translational Research, Azienda USL-IRCCS di Reggio Emilia, Viale Risorgimento 80, 42123 Reggio Emilia, Italy
- Department of Electrical, Computer and Biomedical Engineering, University of Pavia, 27100 Pavia, Italy
| | - Veronica Manicardi
- Laboratory of Translational Research, Azienda USL-IRCCS di Reggio Emilia, Viale Risorgimento 80, 42123 Reggio Emilia, Italy
- Department of Electrical, Computer and Biomedical Engineering, University of Pavia, 27100 Pavia, Italy
- Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Vincenzo Dario Mandato
- Unit of Obstetrics and Gynaecology, Azienda USL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy
| | - Andrea Palicelli
- Pathology Unit, Department of Oncology and Advanced Technologies, Azienda USL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy
| | - Alessia Ciarrocchi
- Laboratory of Translational Research, Azienda USL-IRCCS di Reggio Emilia, Viale Risorgimento 80, 42123 Reggio Emilia, Italy
| | - Gloria Manzotti
- Laboratory of Translational Research, Azienda USL-IRCCS di Reggio Emilia, Viale Risorgimento 80, 42123 Reggio Emilia, Italy
| |
Collapse
|
4
|
Yu H, Guo Y, Chen J, Chen X, Jia P, Zhao Z. Rewired Pathways and Disrupted Pathway Crosstalk in Schizophrenia Transcriptomes by Multiple Differential Coexpression Methods. Genes (Basel) 2021; 12:665. [PMID: 33946654 PMCID: PMC8146818 DOI: 10.3390/genes12050665] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 04/16/2021] [Accepted: 04/27/2021] [Indexed: 02/03/2023] Open
Abstract
Transcriptomic studies of mental disorders using the human brain tissues have been limited, and gene expression signatures in schizophrenia (SCZ) remain elusive. In this study, we applied three differential co-expression methods to analyze five transcriptomic datasets (three RNA-Seq and two microarray datasets) derived from SCZ and matched normal postmortem brain samples. We aimed to uncover biological pathways where internal correlation structure was rewired or inter-coordination was disrupted in SCZ. In total, we identified 60 rewired pathways, many of which were related to neurotransmitter, synapse, immune, and cell adhesion. We found the hub genes, which were on the center of rewired pathways, were highly mutually consistent among the five datasets. The combinatory list of 92 hub genes was generally multi-functional, suggesting their complex and dynamic roles in SCZ pathophysiology. In our constructed pathway crosstalk network, we found "Clostridium neurotoxicity" and "signaling events mediated by focal adhesion kinase" had the highest interactions. We further identified disconnected gene links underlying the disrupted pathway crosstalk. Among them, four gene pairs (PAK1:SYT1, PAK1:RFC5, DCTN1:STX1A, and GRIA1:MAP2K4) were normally correlated in universal contexts. In summary, we systematically identified rewired pathways, disrupted pathway crosstalk circuits, and critical genes and gene links in schizophrenia transcriptomes.
Collapse
Affiliation(s)
- Hui Yu
- Department of Internal Medicine, University of New Mexico, Albuquerque, NM 87131, USA; (H.Y.); (Y.G.)
| | - Yan Guo
- Department of Internal Medicine, University of New Mexico, Albuquerque, NM 87131, USA; (H.Y.); (Y.G.)
| | - Jingchun Chen
- Nevada Institute of Personalized Medicine, University of Nevada Las Vegas, Las Vegas, NV 89154, USA; (J.C.); (X.C.)
| | - Xiangning Chen
- Nevada Institute of Personalized Medicine, University of Nevada Las Vegas, Las Vegas, NV 89154, USA; (J.C.); (X.C.)
| | - Peilin Jia
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA;
| | - Zhongming Zhao
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA;
- Human Genetics Center, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN 37203, USA
| |
Collapse
|
5
|
Yu H, Chen D, Oyebamiji O, Zhao YY, Guo Y. Expression correlation attenuates within and between key signaling pathways in chronic kidney disease. BMC Med Genomics 2020; 13:134. [PMID: 32957963 PMCID: PMC7504859 DOI: 10.1186/s12920-020-00772-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Compared to the conventional differential expression approach, differential coexpression analysis represents a different yet complementary perspective into diseased transcriptomes. In particular, global loss of transcriptome correlation was previously observed in aging mice, and a most recent study found genetic and environmental perturbations on human subjects tended to cause universal attenuation of transcriptome coherence. While methodological progresses surrounding differential coexpression have helped with research on several human diseases, there has not been an investigation of coexpression disruptions in chronic kidney disease (CKD) yet. METHODS RNA-seq was performed on total RNAs of kidney tissue samples from 140 CKD patients. A combination of differential coexpression methods were employed to analyze the transcriptome transition in CKD from the early, mild phase to the late, severe kidney damage phase. RESULTS We discovered a global expression correlation attenuation in CKD progression, with pathway Regulation of nuclear SMAD2/3 signaling demonstrating the most remarkable intra-pathway correlation rewiring. Moreover, the pathway Signaling events mediated by focal adhesion kinase displayed significantly weakened crosstalk with seven pathways, including Regulation of nuclear SMAD2/3 signaling. Well-known relevant genes, such as ACTN4, were characterized with widespread correlation disassociation with partners from a wide array of signaling pathways. CONCLUSIONS Altogether, our analysis reported a global expression correlation attenuation within and between key signaling pathways in chronic kidney disease, and presented a list of vanishing hub genes and disrupted correlations within and between key signaling pathways, illuminating on the pathophysiological mechanisms of CKD progression.
Collapse
Affiliation(s)
- Hui Yu
- Department of Internal Medicine, University of New Mexico, Albuquerque, NM 87131 USA
| | - Danqian Chen
- Key Laboratory of Resource Biology and Biotechnology in Western China, School of Life Sciences, Northwest University, Xi’an, 710069 Shaanxi China
| | | | - Ying-Yong Zhao
- Key Laboratory of Resource Biology and Biotechnology in Western China, School of Life Sciences, Northwest University, Xi’an, 710069 Shaanxi China
| | - Yan Guo
- Department of Internal Medicine, University of New Mexico, Albuquerque, NM 87131 USA
| |
Collapse
|
6
|
Lau A, So HC. Turning genome-wide association study findings into opportunities for drug repositioning. Comput Struct Biotechnol J 2020; 18:1639-1650. [PMID: 32670504 PMCID: PMC7334463 DOI: 10.1016/j.csbj.2020.06.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 06/05/2020] [Accepted: 06/05/2020] [Indexed: 02/02/2023] Open
Abstract
Drug development is a very costly and lengthy process, while repositioned or repurposed drugs could be brought into clinical practice within a shorter time-frame and at a much reduced cost. Numerous computational approaches to drug repositioning have been developed, but methods utilizing genome-wide association studies (GWASs) data are less explored. The past decade has observed a massive growth in the amount of data from GWAS; the rich information contained in GWAS has great potential to guide drug repositioning or discovery. While multiple tools are available for finding the most relevant genes from GWAS hits, searching for top susceptibility genes is only one way to guide repositioning, which has its own limitations. Here we provide a comprehensive review of different computational approaches that employ GWAS data to guide drug repositioning. These methods include selecting top candidate genes from GWAS as drug targets, deducing drug candidates based on drug-drug and disease-disease similarities, searching for reversed expression profiles between drugs and diseases, pathway-based methods as well as approaches based on analysis of biological networks. Each method is illustrated with examples, and their respective strengths and limitations are discussed. We also discussed several areas for future research.
Collapse
Affiliation(s)
- Alexandria Lau
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Hon-Cheong So
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research of Common Diseases, Kunming Zoology Institute of Zoology and The Chinese University of Hong Kong, Hong Kong SAR, China
- Department of Psychiatry, The Chinese University of Hong Kong, Hong Kong SAR, China
- Margaret K.L. Cheung Research Centre for Management of Parkinsonism, The Chinese University of Hong Kong, Hong Kong SAR, China
- Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
- Brain and Mind Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
- Hong Kong Branch of the Chinese Academy of Sciences Center for Excellence in Animal Evolution and Genetics, The Chinese University of Hong Kong, Hong Kong SAR, China
- Corresponding author at: School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
7
|
|
8
|
Karaman B, Sippl W. Computational Drug Repurposing: Current Trends. Curr Med Chem 2019; 26:5389-5409. [DOI: 10.2174/0929867325666180530100332] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 05/06/2018] [Accepted: 05/14/2018] [Indexed: 01/31/2023]
Abstract
:
Biomedical discovery has been reshaped upon the exploding digitization of data
which can be retrieved from a number of sources, ranging from clinical pharmacology to
cheminformatics-driven databases. Now, supercomputing platforms and publicly available
resources such as biological, physicochemical, and clinical data, can all be integrated to construct
a detailed map of signaling pathways and drug mechanisms of action in relation to drug
candidates. Recent advancements in computer-aided data mining have facilitated analyses of
‘big data’ approaches and the discovery of new indications for pre-existing drugs has been
accelerated. Linking gene-phenotype associations to predict novel drug-disease signatures or
incorporating molecular structure information of drugs and protein targets with other kinds of
data derived from systems biology provide great potential to accelerate drug discovery and
improve the success of drug repurposing attempts. In this review, we highlight commonly
used computational drug repurposing strategies, including bioinformatics and cheminformatics
tools, to integrate large-scale data emerging from the systems biology, and consider both
the challenges and opportunities of using this approach. Moreover, we provide successful examples
and case studies that combined various in silico drug-repurposing strategies to predict
potential novel uses for known therapeutics.
Collapse
Affiliation(s)
- Berin Karaman
- Biruni University - Department of Pharmaceutical Chemistry, Istanbul, Turkey
| | - Wolfgang Sippl
- Martin-Luther University of Halle-Wittenberg - Institute of Pharmacy, Halle (Saale), Germany
| |
Collapse
|
9
|
Masoudi-Sobhanzadeh Y, Omidi Y, Amanlou M, Masoudi-Nejad A. DrugR+: A comprehensive relational database for drug repurposing, combination therapy, and replacement therapy. Comput Biol Med 2019; 109:254-262. [PMID: 31096089 DOI: 10.1016/j.compbiomed.2019.05.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 04/26/2019] [Accepted: 05/06/2019] [Indexed: 12/14/2022]
Abstract
Drug repurposing or repositioning, which introduces new applications of the existing drugs, is an emerging field in drug discovery scope. To enhance the success rate of the research and development (R&D) process in a cost- and time-effective manner, a number of pharmaceutical companies worldwide have made tremendous investments. Besides, many researchers have proposed various methods and databases for the repurposing of various drugs. However, there is not a proper and well-organized database available. To this end, for the first time, we developed a new database based on DrugBank and KEGG data, which is named "DrugR+". Our developed database provides some advantages relative to the DrugBank, and its interface supplies new capabilities for both single and synthetic repositioning of drugs. Moreover, it includes four new datasets which can be used for predicting drug-target interactions using supervised machine learning methods. As a case study, we introduced novel applications of some drugs and discussed the obtained results. A comparison of several machine learning methods on the generated datasets has also been reported in the Supplementary File. Having included several normalized tables, DrugR + has been organized to provide key information on data structures for the repurposing and combining applications of drugs. It provides the SQL query capability for professional users and an appropriate method with different options for unprofessional users. Additionally, DrugR + consists of repurposing service that accepts a drug and proposes a list of potential drugs for some usages. Taken all, DrugR+ is a free web-based database and accessible using (http://www.drugr.ir), which can be updated through a map-reduce parallel processing method to provide the most relevant information.
Collapse
Affiliation(s)
- Yosef Masoudi-Sobhanzadeh
- Laboratory of Systems Biology and Bioinformatics (LBB), Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Yadollah Omidi
- Research Center for Pharmaceutical Nanotechnology and Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Massoud Amanlou
- Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, 14176-53955, Iran
| | - Ali Masoudi-Nejad
- Laboratory of Systems Biology and Bioinformatics (LBB), Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran. http://LBB.ut.ac.ir
| |
Collapse
|
10
|
Matteucci C, Argaw-Denboba A, Balestrieri E, Giovinazzo A, Miele M, D'Agostini C, Pica F, Grelli S, Paci M, Mastino A, Sinibaldi Vallebona P, Garaci E, Tomino C. Deciphering cellular biological processes to clinical application: a new perspective for Tα1 treatment targeting multiple diseases. Expert Opin Biol Ther 2019; 18:23-31. [PMID: 30063863 DOI: 10.1080/14712598.2018.1474198] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
BACKGROUND Thymosin alpha 1 (Tα1) is a well-recognized immune response modulator in a wide range of disorders, particularly infections and cancer. The bioinformatic analysis of public databases allows drug repositioning, predicting a new potential area of clinical intervention. We aimed to decipher the cellular network induced by Tα1 treatment to confirm present use and identify new potential clinical applications. RESEARCH DESIGN AND METHODS We used the transcriptional profile of human peripheral blood mononuclear cells treated in vitro with Tα1 to perform the enrichment network analysis by the Metascape online tools and the disease enrichment analysis by the DAVID online tool. RESULTS Networked cellular responses reflected Tα1 regulated biological processes including immune and metabolic responses, response to compounds and oxidative stress, ion homeostasis, peroxisome biogenesis and drug metabolic process. Beyond cancer and infections, the analysis evidenced the association with disorders such as kidney chronic failure, diabetes, cardiovascular, chronic respiratory, neuropsychiatric, neurodegenerative and autoimmune diseases. CONCLUSIONS In addition to the known ability to promote immune response pathways, the network enrichment analysis demonstrated that Tα1 regulates cellular metabolic processes and oxidative stress response. Notable, the analysis highlighted the association with several diseases, suggesting new translational implication of Tα1 treatment in pathological conditions unexpected until now.
Collapse
Affiliation(s)
- Claudia Matteucci
- a Department of Experimental Medicine and Surgery , University of Rome "Tor Vergata" , Rome , Italy
| | - Ayele Argaw-Denboba
- a Department of Experimental Medicine and Surgery , University of Rome "Tor Vergata" , Rome , Italy
| | - Emanuela Balestrieri
- a Department of Experimental Medicine and Surgery , University of Rome "Tor Vergata" , Rome , Italy
| | - Alessandro Giovinazzo
- a Department of Experimental Medicine and Surgery , University of Rome "Tor Vergata" , Rome , Italy
| | - Martino Miele
- a Department of Experimental Medicine and Surgery , University of Rome "Tor Vergata" , Rome , Italy
| | - Cartesio D'Agostini
- a Department of Experimental Medicine and Surgery , University of Rome "Tor Vergata" , Rome , Italy
| | - Francesca Pica
- a Department of Experimental Medicine and Surgery , University of Rome "Tor Vergata" , Rome , Italy
| | - Sandro Grelli
- a Department of Experimental Medicine and Surgery , University of Rome "Tor Vergata" , Rome , Italy
| | - Maurizio Paci
- b Department of Chemical Sciences and Technologies , University of Rome "Tor Vergata" , Rome , Italy
| | - Antonio Mastino
- c Department of Chemical, Biological, Pharmaceutical and Environmental Sciences , University of Messina , Messina , Italy.,d National Research Council , Institute of Translational Pharmacology , Rome , Italy
| | - Paola Sinibaldi Vallebona
- a Department of Experimental Medicine and Surgery , University of Rome "Tor Vergata" , Rome , Italy.,d National Research Council , Institute of Translational Pharmacology , Rome , Italy
| | | | - Carlo Tomino
- e Università San Raffaele Pisana , Roma , Italy.,f IRCSS San Raffaele Pisana , Scientific Institute for Research, Hospitalization and Health Care , Roma , Italy
| |
Collapse
|
11
|
Wang YY, Cui C, Qi L, Yan H, Zhao XM. DrPOCS: Drug Repositioning Based on Projection Onto Convex Sets. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2019; 16:154-162. [PMID: 29993698 DOI: 10.1109/tcbb.2018.2830384] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Drug repositioning, i.e., identifying new indications for known drugs, has attracted a lot of attentions recently and is becoming an effective strategy in drug development. In literature, several computational approaches have been proposed to identify potential indications of old drugs based on various types of data sources. In this paper, by formulating the drug-disease associations as a low-rank matrix, we propose a novel method, namely DrPOCS, to identify candidate indications of old drugs based on projection onto convex sets (POCS). With the integration of drug structure and disease phenotype information, DrPOCS predicts potential associations between drugs and diseases with matrix completion. Benchmarking results demonstrate that our proposed approach outperforms popular existing approaches with high accuracy. In addition, a number of novel predicted indications are validated with various types of evidences, indicating the predictive power of our proposed approach.
Collapse
|
12
|
Olayan RS, Ashoor H, Bajic VB. DDR: efficient computational method to predict drug-target interactions using graph mining and machine learning approaches. Bioinformatics 2018; 34:1164-1173. [PMID: 29186331 PMCID: PMC5998943 DOI: 10.1093/bioinformatics/btx731] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 11/23/2017] [Indexed: 02/06/2023] Open
Abstract
Motivation Finding computationally drug–target interactions (DTIs) is a convenient strategy to identify new DTIs at low cost with reasonable accuracy. However, the current DTI prediction methods suffer the high false positive prediction rate. Results We developed DDR, a novel method that improves the DTI prediction accuracy. DDR is based on the use of a heterogeneous graph that contains known DTIs with multiple similarities between drugs and multiple similarities between target proteins. DDR applies non-linear similarity fusion method to combine different similarities. Before fusion, DDR performs a pre-processing step where a subset of similarities is selected in a heuristic process to obtain an optimized combination of similarities. Then, DDR applies a random forest model using different graph-based features extracted from the DTI heterogeneous graph. Using 5-repeats of 10-fold cross-validation, three testing setups, and the weighted average of area under the precision-recall curve (AUPR) scores, we show that DDR significantly reduces the AUPR score error relative to the next best start-of-the-art method for predicting DTIs by 31% when the drugs are new, by 23% when targets are new and by 34% when the drugs and the targets are known but not all DTIs between them are not known. Using independent sources of evidence, we verify as correct 22 out of the top 25 DDR novel predictions. This suggests that DDR can be used as an efficient method to identify correct DTIs. Availability and implementation The data and code are provided at https://bitbucket.org/RSO24/ddr/. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Rawan S Olayan
- King Abdullah University of Science and Technology (KAUST), Computational Bioscience Research Center (CBRC), Computer, Electrical and Mathematical Sciences and Engineering (CEMSE) Division, Thuwal, Saudi Arabia
| | - Haitham Ashoor
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut 06032, USA
| | - Vladimir B Bajic
- King Abdullah University of Science and Technology (KAUST), Computational Bioscience Research Center (CBRC), Computer, Electrical and Mathematical Sciences and Engineering (CEMSE) Division, Thuwal, Saudi Arabia
| |
Collapse
|
13
|
Iwata M, Hirose L, Kohara H, Liao J, Sawada R, Akiyoshi S, Tani K, Yamanishi Y. Pathway-Based Drug Repositioning for Cancers: Computational Prediction and Experimental Validation. J Med Chem 2018; 61:9583-9595. [PMID: 30371064 DOI: 10.1021/acs.jmedchem.8b01044] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Developing drugs with anticancer activity and low toxic side-effects at low costs is a challenging issue for cancer chemotherapy. In this work, we propose to use molecular pathways as the therapeutic targets and develop a novel computational approach for drug repositioning for cancer treatment. We analyzed chemically induced gene expression data of 1112 drugs on 66 human cell lines and searched for drugs that inactivate pathways involved in the growth of cancer cells (cell cycle) and activate pathways that contribute to the death of cancer cells (e.g., apoptosis and p53 signaling). Finally, we performed a large-scale prediction of potential anticancer effects for all the drugs and experimentally validated the prediction results via three in vitro cellular assays that evaluate cell viability, cytotoxicity, and apoptosis induction. Using this strategy, we successfully identified several potential anticancer drugs. The proposed pathway-based method has great potential to improve drug repositioning research for cancer treatment.
Collapse
Affiliation(s)
- Michio Iwata
- Department of Bioscience and Bioinformatics, Faculty of Computer Science and Systems Engineering , Kyushu Institute of Technology , 680-4 Kawazu , Iizuka , Fukuoka 820-8502 , Japan
| | - Lisa Hirose
- Project Division of ALA Advanced Medical Research, The Institute of Medical Science , The University of Tokyo , 4-6-1 Shirokanedai , Minato-ku , Tokyo 108-8639 , Japan
| | - Hiroshi Kohara
- Project Division of ALA Advanced Medical Research, The Institute of Medical Science , The University of Tokyo , 4-6-1 Shirokanedai , Minato-ku , Tokyo 108-8639 , Japan.,Division of Molecular and Clinical Genetics, Department of Molecular Genetics, Medical Institute of Bioregulation , Kyushu University , 3-1-1 Maidashi , Higashi-ku , Fukuoka, Fukuoka 812-8582 , Japan
| | - Jiyuan Liao
- Project Division of ALA Advanced Medical Research, The Institute of Medical Science , The University of Tokyo , 4-6-1 Shirokanedai , Minato-ku , Tokyo 108-8639 , Japan.,Division of Molecular and Clinical Genetics, Department of Molecular Genetics, Medical Institute of Bioregulation , Kyushu University , 3-1-1 Maidashi , Higashi-ku , Fukuoka, Fukuoka 812-8582 , Japan
| | - Ryusuke Sawada
- Medical Institute of Bioregulation , Kyushu University , 3-1-1 Maidashi , Higashi-ku , Fukuoka, Fukuoka 812-8582 , Japan
| | - Sayaka Akiyoshi
- Medical Institute of Bioregulation , Kyushu University , 3-1-1 Maidashi , Higashi-ku , Fukuoka, Fukuoka 812-8582 , Japan
| | - Kenzaburo Tani
- Project Division of ALA Advanced Medical Research, The Institute of Medical Science , The University of Tokyo , 4-6-1 Shirokanedai , Minato-ku , Tokyo 108-8639 , Japan.,Division of Molecular Design, Research Center for Systems Immunology, Medical Institute of Bioregulation , Kyushu University , 3-1-1 Maidashi , Higashi-ku , Fukuoka, Fukuoka 812-8582 , Japan
| | - Yoshihiro Yamanishi
- Department of Bioscience and Bioinformatics, Faculty of Computer Science and Systems Engineering , Kyushu Institute of Technology , 680-4 Kawazu , Iizuka , Fukuoka 820-8502 , Japan.,PRESTO , Japan Science and Technology Agency , Kawaguchi , Saitama 332-0012 , Japan
| |
Collapse
|
14
|
Khalid Z, Sezerman OU. Computational drug repurposing to predict approved and novel drug-disease associations. J Mol Graph Model 2018; 85:91-96. [PMID: 30130693 DOI: 10.1016/j.jmgm.2018.08.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 08/07/2018] [Accepted: 08/10/2018] [Indexed: 11/24/2022]
Abstract
The Drug often binds to more than one targets defined as polypharmacology, one application of which is drug repurposing also referred as drug repositioning or therapeutic switching. The traditional drug discovery and development is a high-priced and tedious process, thus making drug repurposing a popular alternate strategy. We proposed an integrative method based on similarity scheme that predicts approved and novel Drug targets with new disease associations. We combined PPI, biological pathways, binding site structural similarities and disease-disease similarity measures. The results showed 94% Accuracy with 0.93 Recall and 0.94 Precision measure in predicting the approved and novel targets surpassing the existing methods. All these parameters help in elucidating the unknown associations between drug and diseases for finding the new uses for old drugs.
Collapse
|
15
|
Shameer K, Glicksberg BS, Hodos R, Johnson KW, Badgeley MA, Readhead B, Tomlinson MS, O’Connor T, Miotto R, Kidd BA, Chen R, Ma’ayan A, Dudley JT. Systematic analyses of drugs and disease indications in RepurposeDB reveal pharmacological, biological and epidemiological factors influencing drug repositioning. Brief Bioinform 2018; 19:656-678. [PMID: 28200013 PMCID: PMC6192146 DOI: 10.1093/bib/bbw136] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 11/29/2016] [Indexed: 12/22/2022] Open
Abstract
Increase in global population and growing disease burden due to the emergence of infectious diseases (Zika virus), multidrug-resistant pathogens, drug-resistant cancers (cisplatin-resistant ovarian cancer) and chronic diseases (arterial hypertension) necessitate effective therapies to improve health outcomes. However, the rapid increase in drug development cost demands innovative and sustainable drug discovery approaches. Drug repositioning, the discovery of new or improved therapies by reevaluation of approved or investigational compounds, solves a significant gap in the public health setting and improves the productivity of drug development. As the number of drug repurposing investigations increases, a new opportunity has emerged to understand factors driving drug repositioning through systematic analyses of drugs, drug targets and associated disease indications. However, such analyses have so far been hampered by the lack of a centralized knowledgebase, benchmarking data sets and reporting standards. To address these knowledge and clinical needs, here, we present RepurposeDB, a collection of repurposed drugs, drug targets and diseases, which was assembled, indexed and annotated from public data. RepurposeDB combines information on 253 drugs [small molecules (74.30%) and protein drugs (25.29%)] and 1125 diseases. Using RepurposeDB data, we identified pharmacological (chemical descriptors, physicochemical features and absorption, distribution, metabolism, excretion and toxicity properties), biological (protein domains, functional process, molecular mechanisms and pathway cross talks) and epidemiological (shared genetic architectures, disease comorbidities and clinical phenotype similarities) factors mediating drug repositioning. Collectively, RepurposeDB is developed as the reference database for drug repositioning investigations. The pharmacological, biological and epidemiological principles of drug repositioning identified from the meta-analyses could augment therapeutic development.
Collapse
Affiliation(s)
- Khader Shameer
- Institute of Next Generation Healthcare, Mount Sinai Health System, New York,
NY, USA
| | - Benjamin S Glicksberg
- Icahn School of Medicine at Mount Sinai, Mount Sinai Health System, New York,
NY, USA
| | - Rachel Hodos
- Icahn School of Medicine at Mount Sinai, Mount Sinai Health System, New York,
NY, USA
- New York University, New York, NY, USA
| | - Kipp W Johnson
- Icahn School of Medicine at Mount Sinai, Mount Sinai Health System, New York,
NY, USA
| | - Marcus A Badgeley
- Icahn School of Medicine at Mount Sinai, Mount Sinai Health System, New York,
NY, USA
| | - Ben Readhead
- Institute of Next Generation Healthcare, Mount Sinai Health System, New York,
NY, USA
| | - Max S Tomlinson
- Institute of Next Generation Healthcare, Mount Sinai Health System, New York,
NY, USA
| | | | - Riccardo Miotto
- Institute of Next Generation Healthcare, Mount Sinai Health System, New York,
NY, USA
| | - Brian A Kidd
- Institute of Next Generation Healthcare, Mount Sinai Health System, New York,
NY, USA
| | - Rong Chen
- Clinical Genome Informatics, Icahn Institute of Genetics and Multiscale
Biology, Mount Sinai Health System, New York, NY
| | - Avi Ma’ayan
- Mount Sinai Center for Bioinformatics, Mount Sinai Health System, New York,
NY
| | - Joel T Dudley
- Institute of Next Generation Healthcare, Mount Sinai Health System, New York,
NY, USA
- Department of Genetics and Genomic Sciences, Mount Sinai Health System, New
York, NY, USA
- Department of Population Health Science and Policy, Mount Sinai Health System,
New York, NY, USA
- Director of Biomedical Informatics, Icahn School of Medicine at Mount Sinai,
Mount Sinai Health System, New York, NY
| |
Collapse
|
16
|
Fukunishi Y, Yamashita Y, Mashimo T, Nakamura H. Prediction of Protein-compound Binding Energies from Known Activity Data: Docking-score-based Method and its Applications. Mol Inform 2018; 37:e1700120. [PMID: 29442436 PMCID: PMC6055825 DOI: 10.1002/minf.201700120] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 01/22/2018] [Indexed: 12/18/2022]
Abstract
We used protein-compound docking simulations to develop a structure-based quantitative structure-activity relationship (QSAR) model. The prediction model used docking scores as descriptors. The binding free energy was approximated by a weighted average of docking scores for multiple proteins. This approximation was based on a pharmacophore model of receptor pockets and compounds. The weights of the docking scores were restricted to small values to avoid unrealistic weights by a regularization term. Additional outlier elimination improved the results. We applied this method to two groups of targets. The first target was the kinase family. The cross-validation results of 107 kinase proteins showed that the RMSE of predicted binding free energies was 1.1 kcal/mol. The second target was the matrix metalloproteinase (MMP) family, which has been difficult for docking programs. MMPs require metal-binding groups in their inhibitor structures in many cases. A quantum effect contributes to the metal-ligand interaction. Despite this difficulty, the present method worked well for the MMPs. This method showed that the RMSE of predicted binding free energies was 1.1 kcal/mol. In comparison, with the original docking method the RMSE was 1.7 kcal/mol. The results suggest that the present QSAR model should be applied to general target proteins.
Collapse
Affiliation(s)
- Yoshifumi Fukunishi
- Molecular Profiling Research Center for Drug Discovery (molprof)National Institute of Advanced Industrial Science and Technology (AIST)2-3-26Aomi, Koto-ku, Tokyo135-0064Japan
| | - Yasunobu Yamashita
- Technology Research Association for Next-Generation Natural Products Chemistry2-3-26, Aomi, Koto-kuTokyo135-0064Japan
| | - Tadaaki Mashimo
- Technology Research Association for Next-Generation Natural Products Chemistry2-3-26, Aomi, Koto-kuTokyo135-0064Japan
- IMSBIO Co., Ltd.Owl Tower, 4-21-1Higashi-Ikebukuro, Toshima-kuTokyo170-0013Japan
| | - Haruki Nakamura
- Institute for Protein ResearchOsaka University3-2 YamadaokaSuita, Osaka565-0871Japan
| |
Collapse
|
17
|
Repurposing drugs for glioblastoma: From bench to bedside. Cancer Lett 2018; 428:173-183. [PMID: 29729291 DOI: 10.1016/j.canlet.2018.04.039] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 04/25/2018] [Accepted: 04/26/2018] [Indexed: 02/07/2023]
Abstract
Glioblastoma multiforme is the most common, aggressive and lethal type of brain tumor. It is a stage IV cancer disease with a poor prognosis, as the current therapeutic options (surgery, radiotherapy and chemotherapy) are not able to eradicate tumor cells. The approach to treat glioblastoma has not suffered major changes over the last decade and temozolomide (TMZ) remains the mainstay for chemotherapy. However, resistance mechanisms to TMZ and other chemotherapeutic agents are becoming more frequent. The lack of effective options is a reality that may be counterbalanced by repositioning known and commonly used drugs for other diseases. This approach takes into consideration the available pharmacokinetic, pharmacodynamic, toxicity and safety data, and allows a much faster and less expensive drug and product development process. In this review, an extensive literature search is conducted aiming to list drugs with repurposing usage, based on their preferential damage in glioblastoma cells through various mechanisms. Some of these drugs have already entered clinical trials, exhibiting favorable outcomes, which sparks their potential application in glioblastoma treatment.
Collapse
|
18
|
Murtazalieva KA, Druzhilovskiy DS, Goel RK, Sastry GN, Poroikov VV. How good are publicly available web services that predict bioactivity profiles for drug repurposing? SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2017; 28:843-862. [PMID: 29183230 DOI: 10.1080/1062936x.2017.1399448] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 10/29/2017] [Indexed: 06/07/2023]
Abstract
Drug repurposing provides a non-laborious and less expensive way for finding new human medicines. Computational assessment of bioactivity profiles shed light on the hidden pharmacological potential of the launched drugs. Currently, several freely available computational tools are available via the Internet, which predict multitarget profiles of drug-like compounds. They are based on chemical similarity assessment (ChemProt, SuperPred, SEA, SwissTargetPrediction and TargetHunter) or machine learning methods (ChemProt and PASS). To compare their performance, this study has created two evaluation sets, consisting of (1) 50 well-known repositioned drugs and (2) 12 drugs recently patented for new indications. In the first set, sensitivity values varied from 0.64 (TarPred) to 1.00 (PASS Online) for the initial indications and from 0.64 (TarPred) to 0.98 (PASS Online) for the repurposed indications. In the second set, sensitivity values varied from 0.08 (SuperPred) to 1.00 (PASS Online) for the initial indications and from 0.00 (SuperPred) to 1.00 (PASS Online) for the repurposed indications. Thus, this analysis demonstrated that the performance of machine learning methods surpassed those of chemical similarity assessments, particularly in the case of novel repurposed indications.
Collapse
Affiliation(s)
- K A Murtazalieva
- a Institute of Biomedical Chemistry , Moscow , Russia
- b Pirogov Russian National Research Medical University , Moscow , Russia
| | | | - R K Goel
- c Punjabi University , Patiala , Punjab , India
| | - G N Sastry
- d CSIR-Indian Institute of Chemical Technology , Hyderabad , India
| | - V V Poroikov
- a Institute of Biomedical Chemistry , Moscow , Russia
| |
Collapse
|
19
|
Domínguez Á, Muñoz E, López MC, Cordero M, Martínez JP, Viñas M. Transcriptomics as a tool to discover new antibacterial targets. Biotechnol Lett 2017; 39:819-828. [PMID: 28289911 DOI: 10.1007/s10529-017-2319-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 03/07/2017] [Indexed: 12/20/2022]
Abstract
The emergence of antibiotic-resistant pathogens, multiple drug-resistance, and extremely drug-resistant strains demonstrates the need for improved strategies to discover new drug-based compounds. The development of transcriptomics, proteomics, and metabolomics has provided new tools for global studies of living organisms. However, the compendium of expression profiles produced by these methods has introduced new scientific challenges into antimicrobial research. In this review, we discuss the practical value of transcriptomic techniques as well as their difficulties and pitfalls. We advocate the construction of new databases of transcriptomic data, using standardized formats in addition to standardized models of bacterial and yeast similar to those used in systems biology. The inclusion of proteomic and metabolomic data is also essential, as the resulting networks can provide a landscape to rationally predict and exploit new drug targets and to understand drug synergies.
Collapse
Affiliation(s)
- Ángel Domínguez
- Department of Microbiology and Genetics, Universidad de Salamanca, Plaza de los Drs. de la Reina s/n, 37007, Salamanca, Spain.
| | - Elisa Muñoz
- Department of Cell Biology & Pathology, Universidad de Salamanca, Salamanca, Spain
| | - M Carmen López
- Department of Microbiology and Genetics, Universidad de Salamanca, Plaza de los Drs. de la Reina s/n, 37007, Salamanca, Spain
| | - Miguel Cordero
- Department of Medicine, Universidad de Salamanca, Salamanca, Spain
| | - José Pedro Martínez
- Department of Microbiology & Ecology, Universitat de Valencia/Estudi General (UVEG), Valencia, Spain
| | - Miguel Viñas
- Department of Pathology and Experimental Therapeutics, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
20
|
Fukunishi Y, Yamasaki S, Yasumatsu I, Takeuchi K, Kurosawa T, Nakamura H. Quantitative Structure-activity Relationship (QSAR) Models for Docking Score Correction. Mol Inform 2017; 36:1600013. [PMID: 28001004 PMCID: PMC5297997 DOI: 10.1002/minf.201600013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 04/01/2016] [Indexed: 01/26/2023]
Abstract
In order to improve docking score correction, we developed several structure-based quantitative structure activity relationship (QSAR) models by protein-drug docking simulations and applied these models to public affinity data. The prediction models used descriptor-based regression, and the compound descriptor was a set of docking scores against multiple (∼600) proteins including nontargets. The binding free energy that corresponded to the docking score was approximated by a weighted average of docking scores for multiple proteins, and we tried linear, weighted linear and polynomial regression models considering the compound similarities. In addition, we tried a combination of these regression models for individual data sets such as IC50 , Ki , and %inhibition values. The cross-validation results showed that the weighted linear model was more accurate than the simple linear regression model. Thus, the QSAR approaches based on the affinity data of public databases should improve docking scores.
Collapse
Affiliation(s)
- Yoshifumi Fukunishi
- Molecular Profiling Research Center for Drug Discovery (molprof), National Institute of Advanced Industrial Science and Technology (AIST), 2-3-26, Aomi, Koto-ku, Tokyo, 135-0064, Japan
| | - Satoshi Yamasaki
- Technology Research Association for Next-Generation Natural Products Chemistry, 2-3-26, Aomi, Koto-ku, Tokyo, 135-0064, Japan
| | - Isao Yasumatsu
- Technology Research Association for Next-Generation Natural Products Chemistry, 2-3-26, Aomi, Koto-ku, Tokyo, 135-0064, Japan
- Daiichi Sankyo RD Novare Co., Ltd., 1-16-13, Kita-Kasai, Edogawa-ku, Tokyo, 134-8630, Japan
| | - Koh Takeuchi
- Molecular Profiling Research Center for Drug Discovery (molprof), National Institute of Advanced Industrial Science and Technology (AIST), 2-3-26, Aomi, Koto-ku, Tokyo, 135-0064, Japan
| | - Takashi Kurosawa
- Technology Research Association for Next-Generation Natural Products Chemistry, 2-3-26, Aomi, Koto-ku, Tokyo, 135-0064, Japan
- Hitachi Solutions East Japan, 12-1 Ekimaehoncho, Kawasaki-ku, Kanagawa, 210-0007, Japan
| | - Haruki Nakamura
- Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
21
|
Liu Z, Fang H, Slikker W, Tong W. Potential Reuse of Oncology Drugs in the Treatment of Rare Diseases. Trends Pharmacol Sci 2016; 37:843-857. [DOI: 10.1016/j.tips.2016.06.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 06/27/2016] [Accepted: 06/30/2016] [Indexed: 12/23/2022]
|
22
|
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
|
23
|
Jia Z, Liu Y, Guan N, Bo X, Luo Z, Barnes MR. Cogena, a novel tool for co-expressed gene-set enrichment analysis, applied to drug repositioning and drug mode of action discovery. BMC Genomics 2016; 17:414. [PMID: 27234029 PMCID: PMC4884357 DOI: 10.1186/s12864-016-2737-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 05/11/2016] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Drug repositioning, finding new indications for existing drugs, has gained much recent attention as a potentially efficient and economical strategy for accelerating new therapies into the clinic. Although improvement in the sensitivity of computational drug repositioning methods has identified numerous credible repositioning opportunities, few have been progressed. Arguably the "black box" nature of drug action in a new indication is one of the main blocks to progression, highlighting the need for methods that inform on the broader target mechanism in the disease context. RESULTS We demonstrate that the analysis of co-expressed genes may be a critical first step towards illumination of both disease pathology and mode of drug action. We achieve this using a novel framework, co-expressed gene-set enrichment analysis (cogena) for co-expression analysis of gene expression signatures and gene set enrichment analysis of co-expressed genes. The cogena framework enables simultaneous, pathway driven, disease and drug repositioning analysis. Cogena can be used to illuminate coordinated changes within disease transcriptomes and identify drugs acting mechanistically within this framework. We illustrate this using a psoriatic skin transcriptome, as an exemplar, and recover two widely used Psoriasis drugs (Methotrexate and Ciclosporin) with distinct modes of action. Cogena out-performs the results of Connectivity Map and NFFinder webservers in similar disease transcriptome analyses. Furthermore, we investigated the literature support for the other top-ranked compounds to treat psoriasis and showed how the outputs of cogena analysis can contribute new insight to support the progression of drugs into the clinic. We have made cogena freely available within Bioconductor or https://github.com/zhilongjia/cogena . CONCLUSIONS In conclusion, by targeting co-expressed genes within disease transcriptomes, cogena offers novel biological insight, which can be effectively harnessed for drug discovery and repositioning, allowing the grouping and prioritisation of drug repositioning candidates on the basis of putative mode of action.
Collapse
Affiliation(s)
- Zhilong Jia
- Department of Chemistry and Biology, College of Science, National University of Defense Technology, Changsha, Hunan, 410073, People's Republic of China
- William Harvey Research Institute, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - Ying Liu
- Hunan Key Laboratory of Medical Epigenomics, Department of Dermatology, Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People's Republic of China
| | - Naiyang Guan
- College of Computer, National University of Defense Technology, Changsha, 410073, People's Republic of China
- National Laboratory for Parallel and Distributed Processing, National University of Defense Technology, Changsha, 410073, People's Republic of China
| | - Xiaochen Bo
- Beijing Institute of Radiation Medicine, Beijing, 100850, People's Republic of China
| | - Zhigang Luo
- College of Computer, National University of Defense Technology, Changsha, 410073, People's Republic of China.
- National Laboratory for Parallel and Distributed Processing, National University of Defense Technology, Changsha, 410073, People's Republic of China.
| | - Michael R Barnes
- William Harvey Research Institute, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK.
| |
Collapse
|
24
|
Alaimo S, Bonnici V, Cancemi D, Ferro A, Giugno R, Pulvirenti A. DT-Web: a web-based application for drug-target interaction and drug combination prediction through domain-tuned network-based inference. BMC SYSTEMS BIOLOGY 2015; 9 Suppl 3:S4. [PMID: 26050742 PMCID: PMC4464606 DOI: 10.1186/1752-0509-9-s3-s4] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND The identification of drug-target interactions (DTI) is a costly and time-consuming step in drug discovery and design. Computational methods capable of predicting reliable DTI play an important role in the field. Algorithms may aim to design new therapies based on a single approved drug or a combination of them. Recently, recommendation methods relying on network-based inference in connection with knowledge coming from the specific domain have been proposed. DESCRIPTION Here we propose a web-based interface to the DT-Hybrid algorithm, which applies a recommendation technique based on bipartite network projection implementing resources transfer within the network. This technique combined with domain-specific knowledge expressing drugs and targets similarity is used to compute recommendations for each drug. Our web interface allows the users: (i) to browse all the predictions inferred by the algorithm; (ii) to upload their custom data on which they wish to obtain a prediction through a DT-Hybrid based pipeline; (iii) to help in the early stages of drug combinations, repositioning, substitution, or resistance studies by finding drugs that can act simultaneously on multiple targets in a multi-pathway environment. Our system is periodically synchronized with DrugBank and updated accordingly. The website is free, open to all users, and available at http://alpha.dmi.unict.it/dtweb/. CONCLUSIONS Our web interface allows users to search and visualize information on drugs and targets eventually providing their own data to compute a list of predictions. The user can visualize information about the characteristics of each drug, a list of predicted and validated targets, associated enzymes and transporters. A table containing key information and GO classification allows the users to perform their own analysis on our data. A special interface for data submission allows the execution of a pipeline, based on DT-Hybrid, predicting new targets with the corresponding p-values expressing the reliability of each group of predictions. Finally, It is also possible to specify a list of genes tracking down all the drugs that may have an indirect influence on them based on a multi-drug, multi-target, multi-pathway analysis, which aims to discover drugs for future follow-up studies.
Collapse
|
25
|
Abstract
Drug repositioning or repurposing has received much coverage in the scientific literature in recent years and has been responsible for the generation of both new intellectual property and investigational new drug submissions. The literature indicates a significant trend toward the use of computational- or informatics-based methods for generating initial repositioning hypotheses, followed by focused assessment of biological activity in phenotypic assays. Another viable method for drug repositioning is in vitro screening of known drugs or drug-like molecules, initially in disease-relevant phenotypic assays, to identify and validate candidates for repositioning. This approach can use large compound libraries or can focus on subsets of known drugs or drug-like molecules. In this short review, we focus on ways to generate and validate repositioning candidates in disease-related in vitro and phenotypic assays, and we discuss specific examples of this approach as applied to a variety of disease areas. We propose that in vitro screens offer several advantages over biochemical or in vivo methods as a starting point for drug repositioning.
Collapse
Affiliation(s)
- Graeme F. Wilkinson
- Emerging Innovations, Innovative Medicines, AstraZeneca, Macclesfield, Cheshire, UK
| | | |
Collapse
|
26
|
AbdulHameed MDM, Tawa GJ, Kumar K, Ippolito DL, Lewis JA, Stallings JD, Wallqvist A. Systems level analysis and identification of pathways and networks associated with liver fibrosis. PLoS One 2014; 9:e112193. [PMID: 25380136 PMCID: PMC4224449 DOI: 10.1371/journal.pone.0112193] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Accepted: 10/13/2014] [Indexed: 01/18/2023] Open
Abstract
Toxic liver injury causes necrosis and fibrosis, which may lead to cirrhosis and liver failure. Despite recent progress in understanding the mechanism of liver fibrosis, our knowledge of the molecular-level details of this disease is still incomplete. The elucidation of networks and pathways associated with liver fibrosis can provide insight into the underlying molecular mechanisms of the disease, as well as identify potential diagnostic or prognostic biomarkers. Towards this end, we analyzed rat gene expression data from a range of chemical exposures that produced observable periportal liver fibrosis as documented in DrugMatrix, a publicly available toxicogenomics database. We identified genes relevant to liver fibrosis using standard differential expression and co-expression analyses, and then used these genes in pathway enrichment and protein-protein interaction (PPI) network analyses. We identified a PPI network module associated with liver fibrosis that includes known liver fibrosis-relevant genes, such as tissue inhibitor of metalloproteinase-1, galectin-3, connective tissue growth factor, and lipocalin-2. We also identified several new genes, such as perilipin-3, legumain, and myocilin, which were associated with liver fibrosis. We further analyzed the expression pattern of the genes in the PPI network module across a wide range of 640 chemical exposure conditions in DrugMatrix and identified early indications of liver fibrosis for carbon tetrachloride and lipopolysaccharide exposures. Although it is well known that carbon tetrachloride and lipopolysaccharide can cause liver fibrosis, our network analysis was able to link these compounds to potential fibrotic damage before histopathological changes associated with liver fibrosis appeared. These results demonstrated that our approach is capable of identifying early-stage indicators of liver fibrosis and underscore its potential to aid in predictive toxicity, biomarker identification, and to generally identify disease-relevant pathways.
Collapse
Affiliation(s)
- Mohamed Diwan M. AbdulHameed
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Materiel Command, Fort Detrick, Maryland, United States of America
| | - Gregory J. Tawa
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Materiel Command, Fort Detrick, Maryland, United States of America
| | - Kamal Kumar
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Materiel Command, Fort Detrick, Maryland, United States of America
| | - Danielle L. Ippolito
- U.S. Army Center for Environmental Health Research, Fort Detrick, MD, United States of America
| | - John A. Lewis
- U.S. Army Center for Environmental Health Research, Fort Detrick, MD, United States of America
| | - Jonathan D. Stallings
- U.S. Army Center for Environmental Health Research, Fort Detrick, MD, United States of America
| | - Anders Wallqvist
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Materiel Command, Fort Detrick, Maryland, United States of America
- * E-mail:
| |
Collapse
|
27
|
Liggi S, Drakakis G, Koutsoukas A, Cortes-Ciriano I, Martínez-Alonso P, Malliavin TE, Velazquez-Campoy A, Brewerton SC, Bodkin MJ, Evans DA, Glen RC, Carrodeguas JA, Bender A. Extending in silico mechanism-of-action analysis by annotating targets with pathways: application to cellular cytotoxicity readouts. Future Med Chem 2014; 6:2029-2056. [PMID: 25531967 DOI: 10.4155/fmc.14.137] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025] Open
Abstract
BACKGROUND An in silico mechanism-of-action analysis protocol was developed, comprising molecule bioactivity profiling, annotation of predicted targets with pathways and calculation of enrichment factors to highlight targets and pathways more likely to be implicated in the studied phenotype. RESULTS The method was applied to a cytotoxicity phenotypic endpoint, with enriched targets/pathways found to be statistically significant when compared with 100 random datasets. Application on a smaller apoptotic set (10 molecules) did not allowed to obtain statistically relevant results, suggesting that the protocol requires modification such as analysis of the most frequently predicted targets/annotated pathways. CONCLUSION Pathway annotations improved the mechanism-of-action information gained by target prediction alone, allowing a better interpretation of the predictions and providing better mapping of targets onto pathways.
Collapse
Affiliation(s)
- Sonia Liggi
- Centre for Molecular Informatics, Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|