Hostname: page-component-848d4c4894-2pzkn Total loading time: 0 Render date: 2024-05-14T09:04:25.893Z Has data issue: false hasContentIssue false

Drug repurposing: databases and pipelines

Published online by Cambridge University Press:  25 July 2023

Arjun V. Kowshik
Affiliation:
Department of Biotechnology, PES University, Bengaluru, India
Megha Manoj
Affiliation:
Department of Biotechnology, PES University, Bengaluru, India
Siddarth Sowmyanarayan
Affiliation:
Department of Biotechnology, PES University, Bengaluru, India
Jhinuk Chatterjee*
Affiliation:
Department of Biotechnology, PES University, Bengaluru, India
*
Corresponding author: Jhinuk Chatterjee; Email: jhinukchatterjee@pes.edu
Rights & Permissions [Opens in a new window]

Abstract

The concept of drug repurposing is focused on the repositioning of drug molecules that have already undergone safety trials. There are different strategies for drug repurposing. Network-based strategy focuses on the evaluation of drug combinations in a molecular environment with multi-target hits and analysis of drug interactions. Implementation of any in silico strategy requires several databases and pipelines for executing the process of shortlisting appropriate drugs.

Type
Editorial
Copyright
© The Author(s), 2023. Published by Cambridge University Press

Adherence to the notion of one drug used for one protein as treatment has failed to provide productive solutions for several disorders caused by multiple diverse factors. Currently, pharmaceutical companies are aiming to reposition approved drugs as treatment for various diseases as de novo drug designing is not only slow, but also cumbersome.

The concept of drug repurposing is focused on the sensible repositioning of a plethora of drug molecules that have already undergone safety trials thereby avoiding extensive investment. Drug repurposing is evaluated as a low-risk and highly efficient strategy due to the investment of $1.6 billion, which is nearly one-tenth of the monetary commitment required for de novo designing of a drug,Reference Xue, Li, Xie and Wang 1 antibiotic resistance,Reference Younis, Thangamani and Seleem 2 rare diseases,Reference Roessler, Knoers, van Haelst and van Haaften 3 oncological studies,Reference Schein 4 inflammatory disorders,Reference Kingsmore, Grammer and Lipsky 5 and neurological diseasesReference Massey and Robertson 6 highlights its importance.

The scope of drug repurposing broadened massively since the COVID-19 pandemic and most of the drugs used to treat moderate to severe COVID-19 infections were initially screened using repurposing pipelines.Reference Galindez, Matschinske and Rose 7

Different approaches like signature matching, genetic association, and pathway mapping can be used for drug repurposing.Reference Pushpakom, Iorio and Eyers 8 Two main strategies are namely drug-based, wherein a known FDA-approved drug is chosen as the starting point and either on-target or off-target drug repositioning is performed, and target-based, wherein a target molecule or biomarker is chosen as the starting and all FDA-approved drugs are screened against this target molecule using computational approaches.Reference Spitschak, Gupta, Singh, Logotheti and Pützer 9

Network-based approach facilitates the evaluation of drug combinations in a molecular environment with multi-target hits and analysis of drug interactions.Reference Wu, Wang and Chen 10 One of the key strategies is interactome construction using different genes and related gene products.Reference Barabási, Gulbahce and Loscalzo 11 Networks called diseasomes link multiple disorders and disease-related genes to provide a better insight into how different diseases are linked to each other and this helps pinpoint those genes which form a major link between seemingly unrelated diseases and behave as targets of drug repositioning.Reference Barabási, Gulbahce and Loscalzo 11 Network medicine has proven to play a vital role in drug repurposing in identifying the drug targets by calculating the proximity scores between drug targets and disease genes in the human protein–protein interactome.Reference Cheng, Desai and Handy 12

Implementation of any in silico strategy requires extensive research using databases, and pipelines for executing the process of shortlisting appropriate drugs. Every drug repositioning pipeline requires a collection of all the FDA-approved drugs and their pharmacological properties, structural information of drugs and proteins, gene-level analytical resources, knowledge of the disease, and the pathways related to the disease.

There are different categories of databases commonly used in drug repurposing.

Molecular structure of drugs: ChEMBLReference Davies, Nowotka and Papadatos 13 (https://www.ebi.ac.uk/chembl/) is a manually curated database containing drugs and molecules with high bioactivity. PubChemReference Kim, Thiessen and Bolton 14 (https://pubchem.ncbi.nlm.nih.gov/) is a database of chemical molecules by name, structure, molecular formula, biological activities, physical properties, safety, and other identifiers. DrugBankReference Wishart, Knox and Guo 15 (https://go.drugbank.com/) is a manually curated comprehensive database of drugs and their targets, groups, pharmacological parameters, interactions, transporters, mechanism of action, and useful properties. ChemSpiderReference Pence and Williams 16 (http://www.chemspider.com/) is a chemical structure database providing all the properties, literature review, vendors, and similar molecules to a particular ligand.

Drug targets: BindingDBReference Liu, Lin, Wen, Jorissen and Gilson 17 (https://www.bindingdb.org/rwd/bind/index.jsp) provides information on binding affinities and interactions of all ligand molecules with the target and off-target proteins. Drug Target CommonsReference Tang, Tanoli and Ravikumar 18 (https://drugtargetcommons.fimm.fi/) provides annotations regarding bioactivities of drug–target interactions and relation with multiple proteins in the body accompanied by bioassay data. STITCHReference Kuhn, von Mering, Campillos, Jensen and Bork 19 (http://stitch.embl.de) produces a detailed map of the drug–protein interactions and further protein–protein relations to identify all possible drug targets. TTDReference Zhou, Zhang and Lian 20 (https://db.idrblab.net/ttd/) is an organized platform for identifying drugs for a specific target protein along with patient data and literature. DrugCentralReference Avram, Bologa and Holmes 21 (https://drugcentral.org/) has up-to-date drug information based on protein targets for repositioning.

Drug side effects and toxicity: SIDERReference Kuhn, Letunic, Jensen and Bork 22 (http://sideeffects.embl.de/) is a collection of the drug indications and recorded adverse drug reactions of all marketed drugs. TOXRICReference Wu, Yan and Han 23 (https://toxric.bioinforai.tech/home) is a novel repository with metabolic reactions and categorized toxicological data for all drugs.

Protein and Gene related: UniProt 24 (https://www.uniprot.org/) has the highest quality information regarding the structural and functional annotation of discovered proteins. GproteinDBReference Pándy-Szekeres, Esguerra and Hauser 25 (https://gproteindb.org/?) stores structural, functional, mutational, and endogenous ligand information of all G-coupled proteins in the body. PDBReference Berman, Westbrook and Feng 26 (https://www.rcsb.org/) has 3-D X-ray crystallographic and NMR structures of proteins along with important structural annotations. SCOPeReference Chandonia, Guan, Lin, Yu, Fox and Brenner 27 (https://scop.berkeley.edu/) is a highly curated structural classification of proteins focusing on protein similarity and interactions. HuRIReference Luck, Kim and Lambourne 28 (http://interactome-atlas.org/) has mapping of the interaction of the target protein with all other proteins in the body along with their relationships. HINTReference Das and Hint 29 (http://hint.yulab.org/) stores binary and co-complex protein interactions mapped to generate interactome. STRINGReference Szklarczyk, Gable and Nastou 30 (https://string-db.org/) provides all the functional associations of a protein in the form of an interactome along with sequence similarity, disease pathway, and latest annotations. OMIMReference Amberger, Bocchini, Schiettecatte, Scott and Hamosh 31 (https://www.omim.org/) is a thorough assembly of all human genes with reference to phenotypic expression in disorders, structure, location, mapping, and functioning. GenBankReference Benson, Karsch-Mizrachi, Lipman, Ostell and Wheeler 32 (https://www.ncbi.nlm.nih.gov/genbank/) is genetic sequence collection with protein translation information and gene similarities. BioGRIDReference Stark, Breitkreutz, Reguly, Boucher, Breitkreutz and Tyers 33 (https://thebiogrid.org/) stores detailed description of the gene–gene and gene-chemical interactions. DGIdbReference Cotto, Wagner and Feng 34 (https://www.dgidb.org/) has curated details on all the gene interactions for a drug and vice versa. IntActReference Hermjakob, Montecchi-Palazzi and Lewington 35 (https://www.ebi.ac.uk/intact/home) provides molecular interactions specifically protein–protein and RNA-protein interactions. GEOReference Edgar, Domrachev and Lash 36 (https://www.ncbi.nlm.nih.gov/geo/) accepts array and sequence-based data and provides tools to query and download experiments and curated gene expression profiles.

Pathways: KEGGReference Kanehisa and Goto 37 (https://www.genome.jp/kegg/) stores detailed map of the molecular reactions, relations, and interactions along with the biochemistry of the disease. ReactomeReference Fabregat, Jupe and Matthews 38 (https://reactome.org/) has representation of entire pathways, proteins, reactions, and interactions specific to diseases and protein complexes in a hierarchical manner. Pathway CommonsReference Cerami, Gross and Demir 39 (https://www.pathwaycommons.org/) is used for illustrious pathway visualization for various biochemical processes and diseased states.

Disease-associated: PhenopediaReference Yu, Clyne, Khoury and Gwinn 40 (https://phgkb.cdc.gov/PHGKB/startPagePhenoPedia.action) offers summarized information in a disease-centric view on human genetic associations. DisGeNETReference Piñero, Ramírez-Anguita and Saüch-Pitarch 41 (https://www.disgenet.org/) is a large collection of genes and variants associated with human diseases and integrates data from various repositories to provide various gene-disease and variant-disease associations. DISEASES—Database CommonsReference Grissa, Junge, Oprea and Jensen 42 (https://diseases.jensenlab.org/) provides records of the major genes, proteins, and experiments relating to a disease.

Repurposed Drugs: ReDO_DBReference Pantziarka, Verbaanderd and Sukhatme 43 (https://www.anticancerfund.org/en/redo-db) stores information on compounds that are noncancer drugs showing anticancer activity. DrugRepVReference Rajput, Kumar, Megha, Thakur and Kumar 44 (https://bioinfo.imtech.res.in/manojk/drugrepv/) stores manually curated drugs and chemicals that display antiviral activity against epidemic and pandemic viruses. ReFRAMEReference Janes, Young and Chen 45 (https://reframedb.org/) contains nearly all small molecules that have reached clinical development or undergone significant preclinical profiling.

Drug repurposing pipelines focus on obtaining the best set of candidate drug molecules for the selected disease through rigorous computational techniques and intensified analytical measures.

Computational Analysis of Novel Drug Opportunities (CANDO)Reference Mangione, Falls, Chopra and Samudrala 46 (https://github.com/ram-compbio/CANDO) is an open-source platform for the analysis of drug interactions on a proteomic scale. In CANDO drug–protein interaction signatures are generated from an extensive library of known interaction mappings and compared and screened based on their similarities to the drugs used for the same disease. In total, 51 of the 276 molecules predicted by this platform against SARS-CoV-2 are explored in clinical studies and have demonstrated promising activity.Reference Mangione, Falls and Samudrala 47

KsRepoReference Brown, Kong and Kohane 48 (https://github.com/adam-sam-brown/ksRepo) is an R-based open-source expression-level platform for drug repurposing that identifies potential candidates enrichment scores. KsRepo analyses and compares RNA-seq data to known gene–drug interactions and also exhibits flexibility in data set types and can also predict drug candidates with limited information on drug–gene interactions.Reference Traylor, Sheppard and Ravikumar 49

SperoPredictorReference Ahmed, Lee and Samantasinghar 50 is a generic repurposing framework consisting of multiple machine learning algorithms like Random Forest, Tree Ensemble, and Gradient Boosted Trees to predict repurposable drug candidates. SperoPredictor pipeline involves data collection, training, and subsequent deployment of machine learning models followed by literature and molecular docking-based validation.

Single-cell Guided Pipeline to Aid Repurposing of Drugs (ASGARD)Reference He, Xiao and Liang 51 (https://github.com/lanagarmire/ASGARD) makes use of scRNA-seq data of single cell and cell clusters in the disease to predict potentially repurposable drugs. ASGARD processes scRNA-seq data for differential gene analysis and identifies drug candidates which reverse gene expression.

One of the most recent approaches to drug repurposing is using network medicine to identify biomarkers and target molecules through interactomes.Reference Caldera, Buphamalai, Müller and Menche 52 Searching off-lAbel dRUg aNd NEtwoRk (SAveRUNNER)Reference Fiscon and Paci 53 (https://github.com/sportingCode/SAveRUNNER) is a R-based tool that predicts drugs that can be repositioned by preparing a drug-disease association with the help of human interactome. SAveRUNNER prepares a drug-disease association by calculating the network-based proximity scores which show how close the drug molecule is to the disease in the human interactome. It has been successfully used to repurpose drugs for diseases such as COVID-19,Reference Fiscon, Conte, Farina and Paci 54 Amyotrophic Lateral Sclerosis,Reference Fiscon, Conte, Amadio, Volonté and Paci 55 Breast cancer,Reference Conte, Sibilio, Fiscon and Paci 56 and cardiovascular disease.Reference Paci, Fiscon and Conte 57

The Konstanz-Integration Miner (KNIME)Reference Tuerkova and Zdrazil 58 (www.knime.com) is an open-source platform used to generate semi-automated workflows which are intensely used for drug repurposing. The approach used in KNIME is based on mining of data from various exhaustive databases and processing, analyzing, and visualizing this data in order to shortlist the drugs that can be repurposed.

Conclusion

With an increasing demand to find cures for diseases using faster and more cost-effective methods, drug repurposing is proving to be the future of medicine and the more sensible approach as compared to de novo drug design by reducing the number of clinical and experimental trials and making strong predictions about which drugs can be repositioned, thus, paving the way for successful treatments.

Financial support

There was no funding source pertaining to this research work.

Author contribution

Conceptualization: J.C.; Formal analysis: A.V.K., M.M., S.S.; Methodology: A.V.K., M.M., S.S.; Project administration: J.C.; Resources: A.V.K., M.M., S.S.; Software: A.V.K., M.M., S.S.; Supervision: J.C.; Writing—original draft: A.V.K., M.M., S.S.; Writing—review and editing: J.C.

Disclosure

The authors do not have any competing interests to disclose.

References

Xue, H, Li, J, Xie, H, Wang, Y. Review of drug repositioning approaches and resources. Int J Biol Sci. 2018;14(10):12321244.CrossRefGoogle ScholarPubMed
Younis, W, Thangamani, S, Seleem, MN. Repurposing non-antimicrobial drugs and clinical molecules to treat bacterial infections. Curr Pharm Des. 2015;21(28):41064111.CrossRefGoogle ScholarPubMed
Roessler, HI, Knoers, NVAM, van Haelst, MM, van Haaften, G. Drug repurposing for rare diseases. Trends Pharmacol Sci. 2021;42(4):255267.CrossRefGoogle ScholarPubMed
Schein, CH. Repurposing approved drugs for cancer therapy. Br Med Bull. 2021;137(1):1327.CrossRefGoogle ScholarPubMed
Kingsmore, KM, Grammer, AC, Lipsky, PE. Drug repurposing to improve treatment of rheumatic autoimmune inflammatory diseases. Nat Rev Rheumatol. 2020;16(1):3252.CrossRefGoogle ScholarPubMed
Massey, TH, Robertson, NP. Repurposing drugs to treat neurological diseases. J Neurol. 2018;265(2):446448.CrossRefGoogle ScholarPubMed
Galindez, G, Matschinske, J, Rose, TD, et al. Lessons from the COVID-19 pandemic for advancing computational drug repurposing strategies. Nat Comput Sci. 2021;1(1):3341.CrossRefGoogle ScholarPubMed
Pushpakom, S, Iorio, F, Eyers, PA, et al. Drug repurposing: progress, challenges and recommendations. Nat Rev Drug Discov. 2019;18(1):4158.CrossRefGoogle ScholarPubMed
Spitschak, A, Gupta, S, Singh, KP, Logotheti, S, Pützer, BM. Drug repurposing at the interface of melanoma immunotherapy and autoimmune disease. Pharmaceutics. 2022;15(1):83.CrossRefGoogle ScholarPubMed
Wu, Z, Wang, Y, Chen, L. Network-based drug repositioning. Mol Biosyst. 2013;9(6):12681281.CrossRefGoogle ScholarPubMed
Barabási, AL, Gulbahce, N, Loscalzo, J. Network medicine: a network-based approach to human disease. Nat Rev Genet. 2011;12(1):5668.CrossRefGoogle ScholarPubMed
Cheng, F, Desai, RJ, Handy, DE, et al. Network-based approach to prediction and population-based validation of in silico drug repurposing. Nat Commun. 2018;9:2691.CrossRefGoogle ScholarPubMed
Davies, M, Nowotka, M, Papadatos, G, et al. ChEMBL web services: streamlining access to drug discovery data and utilities. Nucleic Acids Res. 2015;43(W1):W612W620.CrossRefGoogle ScholarPubMed
Kim, S, Thiessen, PA, Bolton, EE, et al. PubChem substance and compound databases. Nucleic Acids Res. 2016;44(D1):D1202D1213.CrossRefGoogle ScholarPubMed
Wishart, DS, Knox, C, Guo, AC, et al. DrugBank: a knowledgebase for drugs, drug actions and drug targets. Nucleic Acids Res. 2008;36(Database issue):D901D906.CrossRefGoogle ScholarPubMed
Pence, HE, Williams, A. ChemSpider: an online chemical information resource. J Chem Educ. 2010;87(11):112311242010.CrossRefGoogle Scholar
Liu, T, Lin, Y, Wen, X, Jorissen, RN, Gilson, MK. BindingDB: a web-accessible database of experimentally determined protein-ligand binding affinities. Nucleic Acids Res. 2007;35(Database issue):D198D201.CrossRefGoogle ScholarPubMed
Tang, J, Tanoli, ZU, Ravikumar, B, et al. Drug target commons: a community effort to build a consensus knowledge base for drug-target interactions. Cell Chem Biol. 2018;25(2):224229.e2.CrossRefGoogle Scholar
Kuhn, M, von Mering, C, Campillos, M, Jensen, LJ, Bork, P. STITCH: interaction networks of chemicals and proteins. Nucleic Acids Res. 2008;36(Database issue):D684D688.CrossRefGoogle ScholarPubMed
Zhou, Y, Zhang, Y, Lian, X, et al. Therapeutic target database update 2022: facilitating drug discovery with enriched comparative data of targeted agents. Nucleic Acids Res. 2022;50(D1):D1398D1407.CrossRefGoogle ScholarPubMed
Avram, S, Bologa, CG, Holmes, J, et al. DrugCentral 2021 supports drug discovery and repositioning. Nucleic Acids Res. 2021;49(D1):D1160D1169.CrossRefGoogle ScholarPubMed
Kuhn, M, Letunic, I, Jensen, LJ, Bork, P. The SIDER database of drugs and side effects. Nucleic Acids Res. 2016;44(D1):D1075D1079.CrossRefGoogle ScholarPubMed
Wu, L, Yan, B, Han, J, et al. TOXRIC: a comprehensive database of toxicological data and benchmarks. Nucleic Acids Res. 2023;51(D1):D1432D1445.CrossRefGoogle ScholarPubMed
UniProt Consortium. UniProt: a hub for protein information. Nucleic Acids Res. 2015;43(D1):D204D212.CrossRefGoogle Scholar
Pándy-Szekeres, G, Esguerra, M, Hauser, AS, et al. The G protein database, GproteinDb. Nucleic Acids Res. 2022;50(D1):D518D525.CrossRefGoogle Scholar
Berman, HM, Westbrook, J, Feng, Z, et al. The protein data bank. Nucleic Acids Res. 2000;28(1):235242.CrossRefGoogle ScholarPubMed
Chandonia, JM, Guan, L, Lin, S, Yu, C, Fox, NK, Brenner, SE. SCOPe: improvements to the structural classification of proteins – extended database to facilitate variant interpretation and machine learning. Nucleic Acids Res. 2022;50(D1):D553D559.CrossRefGoogle Scholar
Luck, K, Kim, DK, Lambourne, L, et al. A reference map of the human binary protein interactome. Nature. 2020;580(7803):402408.CrossRefGoogle ScholarPubMed
Das, J, Hint, YH. High-quality protein interactomes and their applications in understanding human disease. BMC Syst Biol. 2012;6(1):112.CrossRefGoogle ScholarPubMed
Szklarczyk, D, Gable, AL, Nastou, KC, et al. The STRING database in 2021: customizable protein-protein networks, and functional characterization of user-uploaded gene/measurement sets. Nucleic Acids Res. 2021;49(D1):D605D612.CrossRefGoogle ScholarPubMed
Amberger, JS, Bocchini, CA, Schiettecatte, F, Scott, AF, Hamosh, A. OMIM.org: Online Mendelian Inheritance in Man (OMIM®), an online catalog of human genes and genetic disorders. Nucleic Acids Res. 2015;43(Database issue):D789D798.CrossRefGoogle ScholarPubMed
Benson, DA, Karsch-Mizrachi, I, Lipman, DJ, Ostell, J, Wheeler, DL. GenBank. Nucleic Acids Res. 2005;33(Database issue):D34D38.CrossRefGoogle ScholarPubMed
Stark, C, Breitkreutz, BJ, Reguly, T, Boucher, L, Breitkreutz, A, Tyers, M. BioGRID: a general repository for interaction datasets. Nucleic Acids Res. 2006;34(Database issue):D535D539.CrossRefGoogle ScholarPubMed
Cotto, KC, Wagner, AH, Feng, YY, et al. DGIdb 3.0: a redesign and expansion of the drug-gene interaction database. Nucleic Acids Res. 2018;46(D1):D1068D1073.CrossRefGoogle ScholarPubMed
Hermjakob, H, Montecchi-Palazzi, L, Lewington, C, et al. IntAct: an open source molecular interaction database. Nucleic Acids Res. 2004;32(Database issue):D452D455.CrossRefGoogle ScholarPubMed
Edgar, R, Domrachev, M, Lash, AE. Gene Expression Omnibus: NCBI gene expression and hybridization array data repository. Nucleic Acids Res. 2002;30(1):207210.CrossRefGoogle ScholarPubMed
Kanehisa, M, Goto, S. KEGG: kyoto encyclopedia of genes and genomes. Nucleic Acids Res. 2000;28(1):2730.CrossRefGoogle ScholarPubMed
Fabregat, A, Jupe, S, Matthews, L, et al. The reactome pathway knowledgebase. Nucleic Acids Res. 2018;46(D1):D649D655.CrossRefGoogle ScholarPubMed
Cerami, EG, Gross, BE, Demir, E, et al. Pathway Commons, a web resource for biological pathway data. Nucleic Acids Res. 2011;39(Database issue):D685D690.CrossRefGoogle Scholar
Yu, W, Clyne, M, Khoury, MJ, Gwinn, M. Phenopedia and Genopedia: disease-centered and gene-centered views of the evolving knowledge of human genetic associations. Bioinformatics. 2010;26(1):145146.CrossRefGoogle ScholarPubMed
Piñero, J, Ramírez-Anguita, JM, Saüch-Pitarch, J, et al. The DisGeNET knowledge platform for disease genomics: 2019 update. Nucleic Acids Res. 2020;48(D1):D845D855.Google ScholarPubMed
Grissa, D, Junge, A, Oprea, TI, Jensen, LJ. Diseases 2.0: a weekly updated database of disease-gene associations from text mining and data integration. Database (Oxford). 2022;2022:baac019.CrossRefGoogle ScholarPubMed
Pantziarka, P, Verbaanderd, C, Sukhatme, V, et al. ReDO_DB: the repurposing drugs in oncology database. Ecancermedicalscience. 2018;12:886.CrossRefGoogle ScholarPubMed
Rajput, A, Kumar, A, Megha, K, Thakur, A, Kumar, M. DrugRepV: a compendium of repurposed drugs and chemicals targeting epidemic and pandemic viruses. Brief Bioinform. 2021;22(2):10761084.CrossRefGoogle ScholarPubMed
Janes, J, Young, ME, Chen, E, et al. The ReFRAME library as a comprehensive drug repurposing library and its application to the treatment of cryptosporidiosis. Proc Natl Acad Sci U S A. 2018;115(42):1075010755.CrossRefGoogle Scholar
Mangione, W, Falls, Z, Chopra, G, Samudrala, R. cando.py: open source software for predictive bioanalytics of large scale drug-protein-disease data. J Chem Inf Model. 2020;60(9):41314136.CrossRefGoogle ScholarPubMed
Mangione, W, Falls, Z, Samudrala, R. Optimal COVID-19 therapeutic candidate discovery using the CANDO platform. Front Pharmacol. 2022;13:970494.CrossRefGoogle ScholarPubMed
Brown, AS, Kong, SW, Kohane, IS, et al. ksRepo: a generalized platform for computational drug repositioning. BMC Bioinform. 2016;17:78.CrossRefGoogle ScholarPubMed
Traylor, JI, Sheppard, HE, Ravikumar, V, et al. Computational drug repositioning identifies potentially active therapies for chordoma. Neurosurgery. 2021;88(2):428436.CrossRefGoogle ScholarPubMed
Ahmed, F, Lee, JW, Samantasinghar, A, et al. SperoPredictor: an integrated machine learning and molecular docking-based drug repurposing framework with use case of COVID-19. Front Public Health. 2022;10:902123.CrossRefGoogle ScholarPubMed
He, B, Xiao, Y, Liang, H, et al. ASGARD is a single-cell guided pipeline to aid repurposing of drugs. Nat Commun. 2023;14:993.CrossRefGoogle ScholarPubMed
Caldera, M, Buphamalai, P, Müller, F, Menche, J. Interactome-based approaches to human disease. Curr. Opin. Syst. Biol. 2017;3:8894.CrossRefGoogle Scholar
Fiscon, G, Paci, P. SAveRUNNER: an R-based tool for drug repurposing. BMC Bioinform. 2021;22(1):150.CrossRefGoogle ScholarPubMed
Fiscon, G, Conte, F, Farina, L, Paci, P. SAveRUNNER: a network-based algorithm for drug repurposing and its application to COVID-19. PLoS Comput Biol. 2021;17(2):e1008686.CrossRefGoogle ScholarPubMed
Fiscon, G, Conte, F, Amadio, S, Volonté, C, Paci, P. Drug repurposing: a network-based approach to amyotrophic lateral sclerosis. Neurotherapeutics. 2021;18(3):16781691.CrossRefGoogle ScholarPubMed
Conte, F, Sibilio, P, Fiscon, G, Paci, P. A transcriptome- and interactome-based analysis identifies repurposable drugs for human breast cancer subtypes. Symmetry. 2022;14(11):2230.CrossRefGoogle Scholar
Paci, P, Fiscon, G, Conte, F, et al. Comprehensive network medicine-based drug repositioning via integration of therapeutic efficacy and side effects. NPJ Syst Biol Appl. 2022;8(1):12.CrossRefGoogle ScholarPubMed
Tuerkova, A, Zdrazil, B. A ligand-based computational drug repurposing pipeline using KNIME and Programmatic Data Access: case studies for rare diseases and COVID-19. J Cheminform. 2020;12:71.CrossRefGoogle ScholarPubMed