Open access peer-reviewed chapter

In Silico Drug Repurposing: An Effective Tool to Accelerate the Drug Discovery Process

Written By

Kareti Srinivasa Rao and P. Subash

Submitted: 19 November 2022 Reviewed: 02 December 2022 Published: 23 December 2022

DOI: 10.5772/intechopen.109312

From the Edited Volume

Drug Repurposing - Advances, Scopes and Opportunities in Drug Discovery

Edited by Mithun Rudrapal

Chapter metrics overview

112 Chapter Downloads

View Full Metrics

Abstract

Repurposing “old” drugs to treat both common and rare diseases is increasingly emerging as an attractive proposition due to the use of de-risked compounds, with potential for lower overall development costs and shorter development timelines. This is due to the high attrition rates, significant costs, and slow pace of new drug discovery and development. Drug repurposing is the process of finding new, more efficient uses for already-available medications. Numerous computational drug repurposing techniques exist, there are three main types of computational drug-repositioning methods used on COVID-19 are network-based models, structure-based methods and artificial intelligence (AI) methods used to discover novel drug–target relationships useful for new therapies. In order to assess how a chemical molecule can interact with its biological counterpart and try to find new uses for medicines already on the market, structure-based techniques made it possible to identify small chemical compounds capable of binding macromolecular targets. In this chapter, we explain strategies for drug repurposing, discuss about difficulties encountered by the repurposing community, and suggest reported drugs through the drug repurposing. Moreover, metabolic and drug discovery network resources, tools for network construction, analysis and protein–protein interaction analysis to enable drug repurposing to reach its full potential.

Keywords

  • drug repurposing
  • protein–protein interaction
  • drug discovery
  • COVID-19
  • pharmacological repositioning

1. Introduction

Drug repurposing, also known as drug repositioning, is a strategy for speeding up the medicine discovery process by identifying a new therapeutic usage for an already-approved drug for a different indication. One of the outcomes of polypharmacology is the increased success and applicability of drug repurposing, which is a manifestation of the transition from a single to multitarget paradigm in drug discovery [1]. COVID-19 has now been labelled a pandemic, necessitating the development of novel medicines as we move beyond containment. It is unrealistic to meet the current global crisis by developing new pharmaceuticals from the ground up because it is a lengthy procedure. Drug repurposing is a new method in which current drugs that have been proven safe in humans are repurposed to treat diseases that are difficult to cure. While taking these repurposed medications alone may not provide a meaningful clinical advantage, strategically combining them into a cocktail could be quite useful [2].

Repositioning previously approved medications is a promising practice since it lowers the cost and length of the drug development pipeline while also lowering the risk of unexpected side effects. The ability to quickly screen candidates in silico and limit the number of prospective repositioning candidates makes computational repositioning particularly interesting. What is not obvious is how effective such strategies are at generating clinically useful repositioning hypotheses is represented in Figure 1 [3]. The SARS-CoV-2 virus causes a respiratory infection that can lead to pneumonia. COVID-19 has a mortality rate of 2–3.5%, which rises with age and the presence of comorbidities (e.g., hypertension, cardiac insufficiency, diabetes, and asthma). By April 15, 2020, the new coronavirus has infected 2,033,406 people worldwide and killed over 130,000 people [4]. COVID-19 has depleted health systems around the world, leading countries to take drastic measures such as closing land borders and instituting social distancing regulations to halt the disease’s spread [5].

Figure 1.

In silico-based drug repurposing.

The new coronavirus (SARS-CoV-2), which causes COVID-19, has swiftly become a global danger to public health and the economy [5, 6]. SARS-CoV-2, according to recent clinical reports, produces both mild, self-limiting respiratory tract infection and severe progressive pneumonia, which can lead to multiorgan failure and death. Despite the severity of some cases, no pathogen-specific antivirals are currently available to treat this infection. As a result, several studies have looked at the anti-SARS-CoV-2 activity of currently available medicines [7].

Advertisement

2. Faster development times and reduced risks

Attempts to speed up the development of a medication are typically accompanied by an increase in risk. Drug repositioning, on the other hand, offers a solution to the problem. Repositioning candidates have generally gone through numerous phases of clinical development and hence have well-known safety and pharmacokinetic properties, which reduces development risk. Shorter paths to the clinic are also possible because in vitro and in vivo screening, chemical optimization, toxicity, bulk production, formulation development, and even early clinical development have all been achieved in many situations. In summary, these considerations allow for the reduction of many years from the path to market, as well as major risks and costs (Figure 2). As a result, repositioning may provide a superior risk-to-reward ratio than other medication development tactics. These benefits have not gone unnoticed by venture capital firms looking for high value exits for their companies in the near future. Because of the strong response such firms have had from the public equity markets, it is nearly impossible for venture capitalists to invest in a therapeutics company without drug prospects in or approaching clinical trials in 2004. Indeed, repositioning allows for the rapid creation of such a pipeline, and repositioning firms are having little issue with getting venture capital.

Figure 2.

A comparison of traditional de novo drug discovery and development versus drug repositioning. (a) It is well known that from concept to marketable medicine, de novo drug research and development takes 10–17 years [8]. The probability of success is lower than 10% [9]. (b) because repositioning candidates have frequently gone through numerous phases of development for their initial indication, several phases common to de novo drug discovery and development can be avoided, allowing for a reduction in time and risk. ADMET is an abbreviation for absorption, distribution, metabolism, excretion, and toxicity; EMEA is an abbreviation for European medicines agency; FDA is an abbreviation for Food and Drug Administration; IP is an abbreviation for intellectual property; and MHLW is an abbreviation for Ministry of Health, labour, and welfare.

Advertisement

3. Drug repositioning opportunities

Drug repositioning is a potential approach that is gaining traction among governments and pharmaceutical corporations due to its critical role in decreasing time, cost, and risk in the development of treatments for cancer and other terminal diseases. As this technique became more widely known, multidisciplinary teams of researchers and scientists attempted, with varying degrees of efficiency and success, to computationally study the potential of repositioning drugs to treat other diseases and identify alternative indications, regardless of whether the drug in question was approved, withdrawn, in clinical trials, or failed. Despite the fact that drug repositioning is a relatively new technique, the traditional, costly, and risky de novo drug development process is still necessary for discovering and testing new drugs; however, incorporating some computational drug repositioning models into this process can help to move drugs forward in the development pipeline and ultimately improve drug efficiencies in clinical trials. The potential for drug repositioning to help create the critical medications needed to combat the present coronavirus outbreak cannot be overstated [10].

Advertisement

4. Challenges and opportunities

Traditional drug development strategies are risky, expensive, and prone to failure. As a result, drug repositioning has recently gained attention, and it expedites the release of medications for clinical usage. Drug repositioning, on the other hand, is a complicated process involving a variety of aspects, including technology, business models, patents, investment, and market demands. Despite the fact that many medical databases have been built, determining the best strategy to fully utilise huge volumes of medical data remains a challenge. New techniques for drug repositioning are urgently needed. Another problem that needs to be addressed is intellectual property (IP). IP protection for repositioning medications is minimal [11]. Some novel drug-targeted-disease connections discovered by repositioning researchers, for example, were corroborated by papers or online databases; yet, according to the law, it is difficult to seek IP protection for such associations. Some repositioned medications are unable to enter the market due to intellectual property issues. Furthermore, some repositioning attempts have to be abandoned, wasting both time and money [12]. Because the existing commercial model is serial and produces overlapping investment concerns, it is required to develop a new commercial model. Challenges accompany opportunities. An unintentional finding in the 1920s was the first example of medication repositioning. More ways of speeding up the process of drug repositioning have been proposed after nearly a century of development. As a result, medication repositioning has made significant progress. Table 1 contains 75 examples of pharmacological repositioning culled from the extensive literature. To increase the performance of drug repositioning in this circumstance, massive machine learning techniques were applied. Experimental procedures, such as target screening approaches, have been developed in addition to computational approaches to provide direct proof of correlations between medications and diseases [11, 12, 24].

Drug nameNew indicationOrigin indicationMining ApproachesReferences
AtomoxetineADHDParkinson’s diseaseNetwork Approach[13, 14]
AllopurinolGoutTumour lysis syndromeExperiment[13]
AmphetamineHyperkinesis in children (attention deficit hyperactivity disorder, ADHD)StimulantSemantic Approach[13, 15]
ApomorphineErectile dysfunctionParkinson’s diseaseExperiment[16]
AspirinColorectal cancerAnalgesic, antipyreticExperiment[16]
BudesonideColitis, UlcerativeAsthmaComputational Approach[17, 18]
BupropionSmoking cessationDepressionExperiment[13, 19]
CelecoxibColon, colorectal, lung, and breast cancer are all linked to familial adenomatous polyposis.Osteoarthritis and adult rheumatoid, arthritisComputational Approach[17, 20]
ChlorpromazineNon-sedating tranquillizerAntiemetic/antihistamineExperiment[17]
CrizotinibNSCLCClinical trials for anaplastic large-cell lymphomaExperiment[21]
CymbaltaDiabetic peripheral neuropathyDepressionExperiment[13]
DapoxetinePremature ejaculationAnalgesia and depressionExperiment[13]
DoxepinInsomnia antipruriticAntidepressantExperiment[19]
DrospirenoneHypertensionOral contraceptiveExperiment[22]
DuloxetineStress urinary incontinence, fibromyalgia,DepressionComputational Approach[19]
Duloxetinechronic, musculoskeletal pain, shoulder pain, back pain, osteoa-arthritis kneeDiabetic NeuropathiesExperiment[17]
EflornithineReduction of unwanted facial hair in womenAnti-infectiveExperiment[17]
EtanerceptAsthmaRheumatoid arthritisNetwork Approach[22]
EverolimusPancreatic neuroendocrine tumoursImmunosuppressantText-mining Approach[21]
FinasterideHair lossBenign prostatic hyperplasiaExperiment[13, 17]
FludrocortisoneHypertensionCerebral salt wasting syndromeExperiment[22]
FluoxetinePremenstrual dysphoric disorderDepressionNetwork Propagation[13]
FurosemideBartter syndromeEdema associated with congestive heart failureExperiment[22]
GalantamineAlzheimer’s diseasePolio, paralysis and anaesthesiaNetwork Approach[17]
GemcitabineAnticancer agentAntiviralExperiment[16]
HydroxychloroquineAnti-arthritic systemic lupus erythematosusAntiparasiticExperiment[19]
ImatinibGISTBCR-ABLExperiment[21]
ImidaprilCancer cachexiaHypertensionExperiment[19]
InfliximabDifferent arthritis forms; Alzheimer’s diseaseCrohn’s diseaseExperiment[19]
LeflunomideProstate cancerRheumatoid arthritisNetwork Approach[16]
LidocaineOral corticosteroid dependent asthma, arrhythmiaLocal anaesthesiaExperiment[13]
LumiganHypotrichosis simplexGlaucomaExperiment[13]
MecamylamineADHDModerately severe to severe essential hypertension and uncomplicated cases of malignant hypertensionExperiment[17]
MetforminBreast, adenocarcinoma, prostate, colorectal cancerDiabetes mellitusExperiment[16]
MethotrexateOsteosarcoma, breast cancer, Hodgkin lymphomaAcute leukaemiaNetwork Approach[13]
MethotrexateRheumatoid arthritisCancerExperiment[16]
MifepristonePsychotic major depression, Cushing’s syndromePregnancy terminationExperiment[13]
MilnacipranFibromyalgiaDepressionExperiment[13]
MiltefosineVisceral and cutaneous leishmaniosisBreast cancerExperiment[19]
MinocyclineOvarian cancer, gliomaAcneExperiment[16]
MonoxideHair lossHypertensionExperiment[13]
Mycophenolate mofetilRenal symptoms of systemic lupus erythematosusTransplanted organ rejectionExperiment[19]
NaltrexoneAlcohol withdrawalOpioid addictionExperiment[19]
NelfinavirIn clinical trials for multiple cancerAIDSNetwork Approach[21]
NitroxolineBladder, breast cancerAntibioticExperiment[16]
NoscapineMultiple cancer typesAntitussive,antimalarial,analgesicExperiment[16]
PaclitaxelRestenosisCancerNetwork Approach[13]
PegvisomantHypercholesterolemiaAcromegalyExperiment[22]
PerindoprilAlzheimer’s diseaseHypertensionNetwork Approach[22]
PhentolamineImpaired night vision, dental anaesthesia reversal agentHypertensionNetwork Approach[13]
PioglitazoneNonalcoholic steatohepatitisType 2 diabetes mellitusExperiment[23]
RaloxifeneOsteoporosisBreast and prostate cancerExperiment[19]
RapamycinColorectal cancer, lymphoma, leukaemiaImmunosuppressantComputational Approach[16]
RequipRestless legsParkinson’s diseaseExperiment[13]
Retinoic acidAcute promyelocytic leukaemiaAcneExperiment[19]
RopineroleParkinson’s, restless legs syndromeHypertensionExperiment[17]
SibutramineObesityDepressionExperiment[13, 16]
Sildenafil citrateErectile dysfunction (approved)Hypertension, anginaExperiment[13, 19]
StatinsCancer, leukaemiaMyocardial infarctionNetwork Approach[16]
SunitinibPancreatic tumours/Gastrointestinal TumourGIST, renal cell carcinomaNetwork Approach[21]
TadalafilMale erectile dysfunctionInflammation and cardiovascular disease,Experiment[17]
TadalafilProstate cancer, hypertension, pulmonary hyperplasia and prostatic hyperplasiaImpotenceNetwork Approach[17]
ThalidomideModerate to severe erythema nodosum leprosum cutaneous symptoms in leprosy and multiple myelomaSedation, nausea and insomniaExperiment[17]
ThalidomideLeprosy, multiple myelomaMorning sicknessNetwork Approach[13]
ThalidomideErythema nodosum leprosumAnti-emeticExperiment[19]
ThiocolchicosideLeukaemia, multiple myelomaMuscle relaxantNetwork Approach[16]
TofisopamIrritable bowel syndromeAnxiety-related conditionsExperiment[17]
TopiramateMigraine,bu1imiaEpilepsyExperiment[13]
TrastuzumabHER2-positive metastatic gastric cancerHER2-positive breast cancerNetwork Approach[21]
Valproic acidSolid tumours, LeukaemiaAntiepilepticExperiment[16]
VesnarinoneOral cancer, leukaemia, lymphomaCardioprotectiveExperiment[16]
WortmanninLeukaemiaAntifungalExperiment[16]
ZidovudineHIV/AIDSCancerExperiment[13]
Zoledronic acidMultiple myeloma, prostate cancer, breast cancerAnti-bone resorptionExperiment[16]

Table 1.

Pharmacological repositioning culled from the extensive literature.

Advertisement

5. Drug-based computational approaches

The structure and chemical characteristics of a medicinal compound are clearly linked to its final therapeutic effectiveness. As a result, repositioning options for medicinal molecules can be investigated based on chemical similarities. The rationale for this method is based on quantitative connections between chemical structures and biological activity that are well-known (QSAR). Although identical structures in biological systems do not always act the same, computational techniques for drug repositioning can take use of the degrees of resemblance that exist. Chemical similarity techniques work by extracting a set of chemical properties for each drug in a group of medications, then clustering or creating networks based on the recovered features to relate the drugs directly to one another [25]. Simple chemical associations or looking for specific biological traits, such as known drug targets, enriched in the resulting correlations can subsequently be used to infer therapeutic repositioning prospects.

Chemical systems biology is being used to identify new drugs in a network. A unique method of modelling and predicting drug–target interactions is statistical modelling of similarities in chemical structure between medicines and possible ligands [26, 27]. Before and after modelling with chemical drug – ligand interactions, network mapping of a wide range of drugs to protein targets enabled the prediction of new targets, including primary sites of action and off-target proteins as explanations for well-known side effects, with new and unexpected drug binding revealed across major categories of proteins unrelated by sequence or structure. A number of modelling predictions were validated using binding assays, proving the method’s efficacy [28]. A generated network of chemogenomic space exhibited a high level of interaction between gene families, giving tractable drug combinations the ability to act on projected targets, by integrating structure – activity data for predicted multiple target binding compounds [29]. This demonstrates how networks can be used as templates for statistical and computational modelling predictions of drug–ligand interactions, and it adds to our understanding of polypharmacology, or the particular binding of a molecule to two or more biological targets [27]. Computational tools for drug discovery are represented in Table 2.

DatabasesFunctionsURL
BINDPortal for biomolecular interaction networkshttp://bond.unleashedinformatics.com/
BioGRIDA database of physical and genetic interactions of many organismshttp://thebiogrid.org
DIPA database for experimentally determined protein interactionshttp://dip.doe-mbi.ucla.edu/dip/Main.cgi
GWASResource of genome-wide association studieshttp://gwas.nih.gov
HPRDHuman proteome database HPIDwww.hprd.org/
HPIDA human-protein interaction databasehttp://wilab.inha.ac.kr/hpid/
IntActOpen source analysis tools for molecular interaction datahttp://www.ebi.ac.uk/intact/
MINTDatabase of curated molecular interactionshttp://160.80.34.4/mint/
MIPSDatabase of mammalian protein–protein interactions that has been manually curated.http://mips.helmholtz-muenchen.de/proj/ppi/
OMIMA repository of human genes and genetic diseases.http://www.ncbi.nlm.nih.gov/omim
STRINGKnown and predicted protein–protein interactions databasehttp://string-db.org/
Metabolic network resources
BRENDAA comprehensive enzyme databasewww.brenda-enzymes.info/
KEGGA comprehensive database on metabolic pathways, diseases, drugs, and other topics.http://www.genome.jp/kegg/
REACTOMEOpen access curated pathway databasehttp://www.reactome.org/ReactomeGWT/entrypoint.html
Drug Discovery Network resources
CPNMContext-specific Protein Network Minerhttp://www.biotextminer.com/CPNM/index.html
Drug bankInformation on drugs and their targetshttp://www.drugbank.ca/
PROMISCUOUSResource of drugs, proteins and side effectshttp://bioinformatics.charite.de/promiscuous/
STITCHA database of known and projected drug-protein interactionshttp://stitch.embl.de/
ZINCChemical compounds that are commercially available are stored in a databasehttp://zinc.docking.org/
Tools for network construction
CobwebA tool for visualising and exploring networks.http://bioinformatics.charite.de/cobweb/
CytoscapeTool for network visualisation and data integration. Many plugins are available for various types of analysis.http://www.cytoscape.org/
NAViGaTORNetwork visualisation toolhttp://ophid.utoronto.ca/navigator/
Tools for network analysis
PajekA large network visualisation and analysis toolhttp://vlado.fmf.uni-lj.si/pub/networks/pajek/
GephiA dynamic network visualisation tool that also allows for specialising, filtering, navigating, altering, and clustering of network data.https://gephi.org/
BIANAAutomated network data integration and analysis using other tools such as Cytoscapehttp://sbi.imim.es/web/index.php/research/servers/biana?
POINeTPPI searching, analysis and visualisation toolhttp://poinet.bioinformatics.tw/
Network analyserPlugin for analysing and visualising molecular interaction networks, as well as computing specific network topological metrics.http://med.bioinf.mpi-inf.mpg.de/netanalyzer/

Table 2.

Tools for protein–protein interaction analysis.

Advertisement

6. Applications of personalised medicine and drug repositioning

The utilisation of personalised medicine methodologies to investigate particular diseases and reposition medications for these diseases has far-reaching diagnostic and therapy implications. Both of these approaches are particularly useful for rare diseases or disease subtypes that are difficult to investigate and conduct clinical trials for due to their rarity [30]. They’re also important for patients who are resistant to or have developed resistance to medicines and do not have any other options for treatment. We’ll look at how customised medicine and drug repositioning methods can help in these two cases in this section.

Advertisement

7. Orphan or rare diseases

Any disease that affects a small percentage of the population is classified as an orphan or uncommon disease. The majority of known uncommon diseases are genetic in nature, and so they affect people for the rest of their lives. Many manifest early in life, and approximately 30% of children with rare diseases die before reaching the age of five. There is no commonly agreed-upon cut-off figure for determining whether or not a disease is rare. The Rare Disease Act of 2002, for example, defines a rare sickness as any disease or condition that affects fewer than 200,000 people in the United States, whereas in Japan, a rare disease is defined as one that affects fewer than 50,000 people. Rare diseases, on the other hand, are defined by the European Commission on Public Health as those that are life-threatening or chronically debilitating and have such a low prevalence (1 in 2000 individuals) that they require special coordinated efforts to combat. Furthermore, a sickness that is rare in one part of the world or among a specific group of people may be widespread in another. An individual uncommon disease may have a low incidence. However, the 6000 identified rare diseases collectively impact around 25 million Americans, or about 10% of the total [31]. Because rare diseases are defined by therapy availability, resource scarcity, and disease severity, they are now referred to as orphan diseases (ODs), especially since the orphan drug movement began in the United States in 1983. As a result, the United States Orphan Medication Act (1983) covers both rare and non-rare diseases for which there is no reasonable expectation that the cost of developing and commercialising a drug for such a disease in the United States will be recouped via drug sales in the United States. About 6000 rare or OD diseases have been recognised, and the National Institutes of Health’s Office of Rare Diseases (ORD) keeps track of them (NIH). While some of the mentioned ODs are well-known (e.g., cystic fibrosis, Huntington’s disease), the majority of people are unaware of numerous ODs with patient numbers of less than a hundred. Each year, about 250 new ODs and diseases are characterised [32]. The ODA was created to support the research and marketing of medications (orphan pharmaceuticals) for the treatment of ODs and other disorders. The ODA arose in response to the modest number of orphan medications approved in the United States in the years leading up to the ODA’s approval [33]. Unfortunately, the drug research process for ODs is the same as it is for any other disease: it is extremely costly and time-consuming.

Advertisement

8. Discussion and conclusion

After looking at the various ways that computational drug repositioning strategies and models have been used to identify novel therapeutic interactions, we can conclude that each strategy and approach has its own set of benefits and drawbacks, and that combining different strategies and approaches often results in a higher success rate. Despite the fact that we have some excellent computational drug repositioning models, establishing robust models is still a difficult endeavour. Because of the intricacy of mapping such theoretical approaches to imitate actual organisms behaviour, as well as other difficulties such as missing, skewed, and erroneous data, one of the key challenges is bringing theoretical computing ideas into action. For example, reliable gene expression signature profiles may be difficult to define due to a variety of factors, including differences in experimental conditions (e.g., environment variables and patient age) between experiments, which can lead to data discrepancies in gene expression signatures, contributing to biased data. Furthermore, when these genes are employed as medication targets, there may not always be large changes in gene expression, which might lead to erroneous results. Furthermore, when using the chemical structure and molecular information technique, the dearth of high-resolution structural data for drug targets makes it difficult to detect potential drug-target interactions. Another issue that computational drug repositioning models face is the absence of reliable gold-standard datasets with which to evaluate their efficacy.

We offer a brief overview of the subject of computational drug repositioning, with a focus on analytically validating such methods. We cover the three methods of validation that are currently in use, as well as the challenges with consistency and essential assumptions that each of them makes. Finally, we offer an approach for increasing the validity of computational repositioning validation.

References

  1. 1. Medina-Franco JL, Giulianotti MA, Welmaker GS, Houghten RA. Shifting from the single to the multitarget paradigm in drug discovery. Drug Discovery Today. 2013;18(9-10):495-501
  2. 2. Senanayake SL. Drug repurposing strategies for COVID-19. Future Drug Discovery. 2020;2(2):689-706
  3. 3. Brown AS, Patel CJ. A review of validation strategies for computational drug repositioning. Briefings in Bioinformatics. 2018;19(1):174-177
  4. 4. Coronavirus E. 13,968 Cases and 223 Deaths: Available from: https://www.worldometers.info/coronavirus/country/ethiopia. Accessed: 2020;27.
  5. 5. Lima WG, Brito JC, Overhage J, Nizer WS. The potential of drug repositioning as a short-term strategy for the control and treatment of COVID-19 (SARS-CoV-2): A systematic review. Archives of Virology. 2020;165(8):1729-1737
  6. 6. Rodriguez-Morales AJ, Cardona-Ospina JA, Gutiérrez-Ocampo E, Villamizar-Peña R, Holguin-Rivera Y, Escalera-Antezana JP, et al. Clinical, laboratory and imaging features of COVID-19: A systematic review and meta-analysis. Travel Medicine and Infectious Disease. 2020;34:101623
  7. 7. Qu YM, Kang EM, Cong HY. Positive result of Sars-Cov-2 in sputum from a cured patient with COVID-19. Travel Medicine and Infectious Disease. 2020;34:101619
  8. 8. Reichert JM. Trends in development and approval times for new therapeutics in the United States. Nature Reviews. Drug Discovery. 2003;2(9):695-702
  9. 9. Gilbert J, Henske P, Singh A. Rebuilding big pharma's business model. In vivo-New York then Norwalk. 2003;21(10):73-80
  10. 10. Carvalho T, Krammer F, Iwasaki A. The first 12 months of COVID-19: A timeline of immunological insights. Nature Reviews Immunology. 2021;21(4):245-256
  11. 11. Oprea TI, Bauman JE, Bologa CG, Buranda T, Chigaev A, Edwards BS, et al. Drug repurposing from an academic perspective. Drug Discovery Today: Therapeutic Strategies. 2011;8(3-4):61-69
  12. 12. Roin BN. Unpatentable drugs and the standards of patentability. Texas Law Review. 2008;87:503
  13. 13. Novac N. Challenges and opportunities of drug repositioning. Trends in Pharmacological Sciences. 2013;34(5):267-272
  14. 14. Yeu Y, Yoon Y, Park S. Protein localization vector propagation: A method for improving the accuracy of drug repositioning. Molecular BioSystems. 2015;11(7):2096-2102
  15. 15. Mullen J, Cockell SJ, Tipney H, Woollard PM, Wipat A. Mining integrated semantic networks for drug repositioning opportunities. PeerJ. 2016;4:e1558
  16. 16. Gupta SC, Sung B, Prasad S, Webb LJ, Aggarwal BB. Cancer drug discovery by repurposing: Teaching new tricks to old dogs. Trends in Pharmacological Sciences. 2013;34(9):508-517
  17. 17. Ashburn TT, Thor KB. Drug repositioning: Identifying and developing new uses for existing drugs. Nature Reviews. Drug Discovery. 2004;3(8):673-683
  18. 18. Ross MK, Yoon J, van der Schaar A, van der Schaar M. Discovering pediatric asthma phenotypes on the basis of response to controller medication using machine learning. Annals of The American Thoracic Society. 2018;15(1):49-58
  19. 19. Henriksen K, Christiansen C, Karsdal MA. Serological biochemical markers of surrogate efficacy and safety as a novel approach to drug repositioning. Drug Discovery Today. 2011;16(21-22):967-975
  20. 20. Nagaraj AB, Wang QQ , Joseph P, Zheng C, Chen Y, Kovalenko O, et al. Using a novel computational drug-repositioning approach (DrugPredict) to rapidly identify potent drug candidates for cancer treatment. Oncogene. 2018;37(3):403-414
  21. 21. Deotarse PP, Jain AS, Baile MB, Kolhe NS, Kulkarni AA. Drug repositioning: A review. International Journal of Pharmacy Research and Review. 2015;4:51-58
  22. 22. Wang ZY, Zhang HY. Rational drug repositioning by medical genetics. Nature Biotechnology. 2013;31(12):1080-1082
  23. 23. Wang ZY, Quan Y, Zhang HY. Medical genetic inspirations for anticancer drug repurposing. Trends in Pharmacological Sciences. 2014;35(1):1-3
  24. 24. Swinney DC, Anthony J. How were new medicines discovered? Nature Reviews. Drug Discovery. 2011;10(7):507-519
  25. 25. Eckert H, Bajorath J. Molecular similarity analysis in virtual screening: Foundations, limitations and novel approaches. Drug Discovery Today. 2007;12(5-6):225-233
  26. 26. Keiser MJ, Roth BL, Armbruster BN, Ernsberger P, Irwin JJ, Shoichet BK. Relating protein pharmacology by ligand chemistry. Nature Biotechnology. 2007;25(2):197-206
  27. 27. Hopkins AL. Network pharmacology: The next paradigm in drug discovery. Nature Chemical Biology. 2008;4(11):682-690
  28. 28. Keiser MJ, Setola V, Irwin JJ, Laggner C, Abbas AI, Hufeisen SJ, et al. Predicting new molecular targets for known drugs. Nature. 2009;462(7270):175-181
  29. 29. Paolini GV, Shapland RH, van Hoorn WP, Mason JS, Hopkins AL. Global mapping of pharmacological space. Nature Biotechnology. 2006;24(7):805-815
  30. 30. Li YY, Jones SJ. Drug repositioning for personalized medicine. Genome Medicine. Mar 2012;4(3):1-4
  31. 31. Rados C. Orphan products: Hope for people with rare diseases. FDA Consumer. 2003;11:10-15
  32. 32. Wästfelt M, Fadeel B, Henter JI. A journey of hope: Lessons learned from studies on rare diseases and orphan drugs. Journal of Internal Medicine. Jul 2006;260(1):1-10
  33. 33. Levitt JA, Kelsey JV. The orphan drug regulations and related issues. Food & Drug LJ. 1993;48:525

Written By

Kareti Srinivasa Rao and P. Subash

Submitted: 19 November 2022 Reviewed: 02 December 2022 Published: 23 December 2022