Skip to main content
Springer Nature - PMC COVID-19 Collection logoLink to Springer Nature - PMC COVID-19 Collection
. 2023 Feb 21;49(2):157–166. doi: 10.1134/S1068162023020139

Drug Repurposing: An Effective Tool in Modern Drug Discovery

V S Kulkarni 1, V Alagarsamy 1,, V R Solomon 1, P A Jose 1, S Murugesan 2
PMCID: PMC9945820  PMID: 36852389

Abstract

Drug repurposing is using an existing drug for a new treatment that was not indicated before. It has received immense attention during the COVID-19 pandemic emergency. Drug repurposing has become the need of time to fasten the drug discovery process and find quicker solutions to the over-exerted healthcare scenario and drug needs. Drug repurposing involves identifying the drug, evaluating its efficiency using preclinical models, and proceeding to phase II clinical trials. Identification of the drug candidate can be made through computational and experimental approaches. This approach usually utilizes public databases for drugs. Data from primary and translational research, clinical trials, anecdotal reports regarding off-label uses, and other published human data information available are included. Using artificial intelligence algorithms and other bioinformatics tools, investigators systematically try to identify the interaction between drugs and protein targets. It can be combined with genetic data, clinical analysis, structure (molecular docking), pathways, signatures, targets, phenotypes, binding assays, and artificial intelligence to get an optimum outcome in repurposing. This article describes the strategies involved in drug repurposing and enlists a series of repurposed drugs and their indications.

Keywords: drug repurposing, clinical trials, molecular docking, drug discovery, post-market safety


INTRODUCTION

STAGES OF DRUG REPURPOSING

IMPORTANCE OF DRUG REPURPOSING

AN IDEAL CANDIDATE FOR REPURPOSING

STRATEGIES FOR DRUG REPURPOSING

Phenotypic Screening

Target-Based Methods

Knowledge-Based Methods

Signature-Based Methods

Pathway- or Network-Based Methods

Targeted Mechanism-Based Methods

Molecular Docking

Application of Drug repurposing in Drug Discovery

Challenges

CONCLUSIONS

REFERENCES

INTRODUCTION

Drug repurposing is the technique of using an existing drug or drug candidate for a new treatment or medical condition for which it was not indicated before [1]. It was initially developed to treat a different medical condition. It has been described as a serendipitous process that happens unexpectedly. In this process, the undesired side effects of drug molecules can also be a pointer to exploring the possibility of its effectiveness in an entirely different medical condition [2]. Usually, drugs with established safety in humans and tested and developed for efficacy in a particular disease other than the one for which they were developed [3]. This process brings the drugs directly to preclinical and clinical trials, skipping the drug development process, and thus reducing risk and costs [4].

STAGES OF DRUG REPURPOSING

The stages of repurposing are elucidated in Fig. 1 [17].

Fig. 1.

Fig. 1.

Stages of Repurposing.

IMPORTANCE OF DRUG REPURPOSING

Repurposing can identify new compounds based on phenotypic benefits without explicitly defining the mechanism of action. This can be directly tested in preclinical animal models, and these results are more applicable to clinical applications and research. It may progress directly straight to Phase II clinical trials [5]. There is a minimum risk of failure with repurposed drugs [6]. The difference between traditional drug discovery and drug repurposing is described in Table 1.

Table 1.  .

The main difference between Traditional Drug Discovery System and Drug Repurposing [613]

TRADITIONAL DRUG DISCOVERY DRUG REPURPOSING

Include 5 stages:

  • Discovery and preclinical

  • Safety review

  • Clinical research

  • FDA review

  • FDA post-market safety monitoring

Include 4 stages:

    • Compound identification

    • Compound acquisition

    • Development

    • FDA post-market safety monitoring

Generally, more time consuming Less time consuming
High investment or cost Lesser investment compared to traditional drug discovery
More risk of failure Less risk of failure
Clinical efficacy and safety profile should be evaluated Clinical efficacy and safety profiles already exist

Repurposing drugs have a significant advantage in decreasing the development cost and time to market over standard discovery. Data like pharmacokinetics, toxicology, and safety data from the standard discovery process [1416].

AN IDEAL CANDIDATE FOR REPURPOSING

A drug has undergone clinical drug development and has been marketed as an ideal candidate for repurposing. A drug with well-established safety and toxicity studies in previous clinical trials, approved by the regulatory authorities, can skip clinical trials with sufficient data support and justification. The mechanism of action of the selected drug shall be established [3].

Those drugs that have gone through several stages of clinical development and have been unsuccessful for reasons other than safety are ideal candidates for repurposing. Some drug repurposing has occurred during clinical trials, like the well-known Viagra (sildenafil) indicated initially for hypertension and angina. The new role of treating erectile dysfunction was unravelled during the clinical trials.

There are examples of abandoned drugs with their toxicity resurfaced with different indications. Thalidomide, indicated for vomiting, was used to treat nausea in pregnant women and resulted in several congenital disabilities. Due to this tragic effect on fetal development, its use was banned or restricted in several countries. Later, the drug was repurposed for leprosy and multiple myeloma [18]. However, this type of repurposing has met objections from the scientific community [19]. The benefit-to–to-risk ratio can be considered during repurposing cases. A rational decision from the regulatory authorities is essential to address the concern “Is it worth the risk? Or can an existing therapy perform better than the repurposed drug?”.

STRATEGIES FOR DRUG REPURPOSING

This approach usually utilizes public databases for drugs. Data from primary and translational research, clinical trials, anecdotal reports regarding off-label uses, and other published human data information available are included. Using artificial intelligence algorithms and other bioinformatics tools, investigators systematically try to identify the interaction between drugs and protein targets. In-silico drug repositioning is a powerful technology with significant advantages, including speed and reduced costs [20]. There are usually three kinds of approaches; computational approach, biological experimental approach, and mixed approach, which are described in Table 2 [17].

Table 2.  .

Drug repurposing can be drug-oriented, disease-oriented, and treatment-oriented

Drug-oriented Information of drugs Disease-oriented Treatment oriented
Off-label use of drugs Information on disease pathway Disease omics data
Phenotypic screening Disease omics data All information related to treatment strategies
Target 3D structure of the drug Genetics data of disease Genetics genomics
Chemical structure of drugs and ligands Protein interaction network Proteomics metabolics
An adverse effect of drugs

Traditional phenotype-based screening methods do not need prior knowledge, and the repositioned drugs are just serendipitously tested. The integrated knowledge and elucidated drug action mechanisms increase with the complexity of modelling methods [4]. These methods are listed in Table 3.

Table 3.  .

Methods of Repurposing [4]

Method Required knowledge#
Blinded search or screening method

Off-label use

Phenotypic screening

Target-based methods Phenotypic screening; Target 3D structure, chemical structure information of drugs and ligands
Knowledge-based methods

Drug–target information, chemical structure information of targets and adverse effects (clinical trial information)

Regulatory approval labels and adverse effects

Available pathway information of disease

Signature-based methods

Disease omics data; Genetics data; Drug omics data

Disease omics and drug omics data

Pathway- or network-based methods Disease omics data, available pathway information, and protein interaction network; Drug omics data
Targeted mechanism-based methods Drug omics data, disease pathway and protein interaction network

# Repurposing methods require various skills and knowledge of various factors, that is, it may be drug-oriented or disease-oriented, or    treatment-oriented [4]

Phenotypic Screening

The phenotypic screening method was used to discover molecules and biologics approved by the regulatory bodies. These methods do not include pharmaceutical or biological information and therefore are less likely to help elucidate any mechanisms of action of drugs. Most depend on serendipitous identification from tests aimed at specific diseases and medicines. The advantage of these methods (off-label use and phenotypic screening) is that they have a high chance of application to many drugs or conditions [4, 21].

Target-Based Methods

This method requires specific knowledge about the targets, such as 3D protein structures. Knowledge-based methods require knowledge about drugs or diseases, such as adverse effects, regulatory approval labels, records of clinical trials, and published disease biomarkers (potential targets) or disease pathways). These methods enable researchers to quickly screen any number of drug molecules with a known chemical structure (e.g., Simplified Molecular Input Line-Entry System SMILES). Target-based drug repositioning methods include.

• In-vitro and in vivo high-throughput and/or high-content screening (HTS/HCS) of drugs for a protein or biomarker of interest and

• In-silico screening of drugs or compounds from drug libraries, such as ligand-based screening or docking.

Compared to blinded methods, targeted-based methods improve the chances of drug discovery as targets are directly linked to the disease mechanism. Integrating target information and drug repurposing increases the possibility of finding therapeutically beneficial compounds [4, 21, 22].

Knowledge-Based Methods

These methods apply bioinformatics or cheminformatics approaches to include the available information of drugs, drug-target networks, chemical structures of targets and drugs, clinical trial information, FDA approval labels, signalling or metabolic pathways, and so on, into drug-repositioning studies. The information content of blinded and target-based methods may not be sufficient to identify new mechanisms beyond the known targets. Knowledge-based methods incorporate known information into predicting unknown mechanisms, such as novel drug targets, obscure drug-drug similarities, and new disease biomarkers. Knowledge-based methods include much-known information into the drug repositioning process to improve prediction accuracy [4, 21].

Signature-Based Methods

Signature-based drug-repurposing methods use gene signatures derived from disease omics data with or without treatment to discover unknown off-target or disease mechanisms. Such genomics data can be assessed through publicly available databases, such as NCBI-GEO (http://www.ncbi.nlm.nih.gov/geo/), SRA Sequence Read Archive (http://www.ncbi. nlm.nih.gov/Traces/sra/), CMAP Connectivity Map and CCLE Cancer Cell Line Encyclopedia. Signature-based methods help uncover unknown mechanisms of action of molecules and drugs. Using computational approaches includes molecular-level mechanisms, such as significantly changed genes [4, 21].

Pathway- or Network-Based Methods

This method utilizes genetic disease data, available signalling or metabolic pathways, and protein interaction networks to reconstruct disease-specific pathways, thereby identifying the key target for repurposing drugs. They help narrow general signalling networks from a large number of proteins down to a specific network with a few proteins (or targets). These methods use pathway analysis or network biology methods to discover essential pathways from diseases' genetic, genomic, proteomic, and metabolic data to find new targets for repositioned drugs. Example: Signalling mechanisms of metastatic subtypes of breast cancer because the subtype signalling mechanisms are hard to elucidate from existing breast cancer pathways or the gene signatures [4, 21].

Targeted Mechanism-Based Methods

These methods integrate treatment omics (genetic) data, available signalling pathway information, and protein interaction networks to delineate the unknown mechanisms of the action of drugs. It aims to discover drug action mechanisms by identifying off-target or targeted pathways of treated drugs using drug omics data (before and after drug treatment). For example, drug resistance is an issue in cancer therapy. Although patients initially respond well to a drug, they often acquire resistance to that drug after a few months of treatment. Hence, successful drug treatment needs additional information about the mechanisms of action of drugs to find better drug targets. However, there are fewer studies on targeted mechanism-based methods that developed elegant computational models to predict the drug effects and related targeted pathways. This is because of the difficulties in deriving effective computational models [4, 21].

Molecular Docking

Molecular docking is a versatile tool used to predict the geometry and to score the interaction of a protein in a complex with a small-molecule ligand. Docking can be performed by docking a known drug into a large set of different target structures or a database of approved medications into one intended target. Molecular docking is a convenient and fast method to screen large libraries of ligands and targets, with a full range of sampling options [23]. 3D structures of the target shall be available through crystallography, nuclear magnetic resonance (NMR), or comparative models to carry out docking. Drawbacks and limitations include approximate scoring function and imperfect binding mode placement algorithms. However, these problems can be overcome by postprocessing docking results with more accurate scoring functions and other criteria [24].

Application of Drug Repurposing in Drug Discovery

Besides developing new treatment options (such as immunotherapy and host-directed therapies), scientists worldwide are working to repurpose existing drugs against SARS-CoV-2 [27]. Table 2 gives a detailed list of repurposed drugs, and a few are repurposed for the treatment of COVID-19. Another example is metformin, an antidiabetic drug that shows anticancer effects by decreasing the incidence of different cancers and inhibiting the proliferation and migration of cancer cells, activating apoptosis, and reducing EMT (epithelial-mesenchymal transition) and metastasis. Repurposing helps overcome antibiotic resistance. For example, TB strains resistant to currently used drug combinations are found in all parts of the world. The product antibiotic, pyridomycin, discovered in the 1950s, is repurposed to treat TB, which takes the place of isoniazid [25, 26]. Table 4 contains a detailed list of repurposed drugs with their initial indication and repurposed use.

Table 4.  .

Examples of repurposed drugs

S. No. Drug Discovered Repurposed Ref.
1 Amiloride Acid-sensing ion channel antagonist Secondary progressive multiple sclerosis (SPMS) [26]
2 Anastrazole Ovulation induction Breast cancer [9]
3 Angiotensin-converting enzyme 2 (ACE2) inhibitor, angiotensin receptor blocker (ARB) and statins Antihypertensives Effective against SARS-CoV-2 (COVID-19) [Few controversies are seen apart from promising results] [27]
4 Aripiprazole Antipsychotic/Antidepressant Active against fungal biofilms [28]
5 Artesunate Anti-infective Active against fungal biofilms [28]
6 Aspirin and ibuprofen Inflammation Antibacterial and antifungal [25]
7 Atorvastatin (generic Lipitor) Hyper-cholesterolaemia Cavernous angioma [26]
8 Auranofin Rheumatoid arthritis Antibacterial and antifungal [25]
9 Avermectin B1a Anti-infective Active against fungal biofilms [28]
10 Azathioprine Crohn’s disease Antibacterial and antifungal [25]
11 Bacitracin Anti-infective Active against fungal biofilms [28]
12 Benzbromarone Vasodilator Active against fungal biofilms [28]
13 Bithionate disodium Anti-infective Active against fungal biofilms [28]
14 Bleomycin Antitumor Active against fungal biofilms [28]
15 Bromperidol Antipsychotic/Antidepressant Active against fungal biofilms [28]
16 Broxyquinoline Anti-infective Active against fungal biofilms [28]
17 Capecitabine Colon cancer Breast cancer [9]
18 Carboplatin Antitumor Active against fungal biofilms [28]
19 Celecoxib Anti-inflammatory/Immunomodulatory Active against fungal biofilms [28]
20 Chloroquine Anti-malarial Active against fungal biofilms [28]
21 Cisplatin Antitumor Active against fungal biofilms [28]
22 Clarithromycin Anti-infective Active against fungal biofilms [28]
23 Clarithromycin, pioglitazone, and treosulfan

Antibiotic

Antidiabetic

Non-small cell lung cancer [29]
24 Clomiphene Fertility Antibacterial and antifungal [25]
25 Cyclophosphamide As immuno-modulator in autoimmune diseases Breast cancer [9]
26 Cyclosporine Anti-inflammatory/Immunomodulatory Active against fungal biofilms [28]
27 Dacarbazine Antitumor Active against fungal biofilms [28]
28 Daunorubicin Acute myeloid leukemia, acute lymphocytic leukemia, chronic myelogenous leukemia, and Kaposi’s sarcoma Antibacterial and antifungal [25]
29 Dexpramipexole ALS and other neurological diseases: phase 3 trials did not meet the endpoint Hypereosinophilic syndromes [26]
30 Diazepam Antipsychotic/Antidepressant Active against fungal biofilms [28]
31 Digoxin Treatment for cardiac diseases Anticancer [30]
32 Dihydroartemisinin Anti-infective Active against fungal biofilms [28]
33 Disulfiram (Antabuse) Reduces ethanol tolerance in alcoholism Metastatic breast cancer & Alzheimer’s disease [26]
34 Docetaxel Hormone-refractory prostate cancer Breast cancer and active against fungal biofilms [9, 28]
35 Doxepin Antipsychotic/Antidepressant Active against fungal biofilms 28]
36 Doxorubicin Antibiotic from Streptomyces peucetiusbacterium, Bladder, breast, stomach, lung, ovarian, and thyroid cancers [9, 25]
37 Ebastine Anti-inflammatory/Immunomodulatory Active against fungal biofilms [28]
38 Ebselen Bipolar disorder and ischemic stroke Antibacterial and antifungal [25]
39 Edaravone Neuroprotective agent in acute ischemic stroke and ALS Multiple sclerosis [26]
40 Eltrombopag Anti-inflammatory/Immunomodulatory Active against fungal biofilms [28]
41 Esketamine (S enantiomer of ketamine) Intravenous anesthetic Treatment-resistant major depressive disorder (TRD) [31]
42 Etodolac Anti-inflammatory/Immunomodulatory Active against fungal biofilms [28]
43 Everolimus (Votubia, Evertor) Immunosuppressants during organ transplants, wound healing Breast cancer [9]
44 Exemestane Ovulation induction Breast cancer [9]
45 Favipiravir Inhibitors of RNA-dependent RNA polymerase of virus (Antiviral drug) Effective against SARS-CoV-2 (COVID-19) [More studies ae required] [27, 34]
46 Fenofibrate Reduces, triglyceride-rich particles (LDL) in plasma Reduces macrophage recruitment in abdominal aortic aneurysm [26]
47 Finasteride Benign prostatic hyperplasia Antibacterial and antifungal [25]
48 Floxuridine Colorectal cancer Antibacterial and antifungal [25]
49 Fluorouracil Keratoacanthomas, actinic keratosis, and skin warts Breast cancer [9]
50 Fluorouracil Solid tumors Antibacterial and antifungal [25]
51 Fluoxetine Antipsychotic/Antidepressant, Serotonin selective reuptake inhibitor (SSRI) Active against fungal biofilms, secondary progressive multiple sclerosis (SPMS)

[28, 26

]

52 Fluvastatin Lipid-lowering Active against fungal biofilms [28]
53 Fulvestrant Antiestrogen Breast cancer [9]
54 Gallium nitrate Lymphoma and bladder cancer Antibacterial and antifungal [25]
55 Gemcitabine Anti-viral drug Breast cancer [9]
56 Goserelin Prostate cancer, uterine fibroids, assisted reproduction Breast cancer [9]
57 γ-Secretase inhibitors (GSI) Alzheimer disease: prevent amyloid precursor cleavage Several inhibitors are being tested against a variety of cancers [26]
58 Human Albumin Blood additive Immuno-restoration [26]
59 Hydroxychloroquine Antimalarial Antiviral drug (HIV, Chicken guinea, dengue, SARS-CoV-2) [34]
60 Imipramine Antipsychotic/Antidepressant Active against fungal biofilms [28]
61 Iodoquinol Anti-infective Active against fungal biofilms [28]
62 Itraconazole Antifungal Anticancer [30]
63 Ivermectin Anti-parasitic Effective against SARS-CoV-2 (COVID-19) [safe in conventional doses] [27]
64 Ketoprofen Anti-inflammatory/Immunomodulatory Active against fungal biofilms [28]
65 Ketorolac Anti-inflammatory/Immunomodulatory Active against fungal biofilms [28]
66 Letrozole Ovulation induction Breast cancer [9]
67 Lopinavir/ritonavir Antiviral drug Effective against SARS-CoV-2 (COVID-19) Drug needs to be further investigated] [27, 34]
68 Lorazepam Antipsychotic/Antidepressant Active against fungal biofilms [28]
69 Losartan Blood pressure reduction Alzheimer disease [26]
70 Lovastatin Lipid-lowering Active against fungal biofilms [28]
71 Loxapine Antipsychotic and antischizophrenia Irritability associated with autism [26]
72 Mebendazole Antiparasitic/Helminthiasis/Anti-infective Brain cancer (i.e., medulloblastoma and glioblastoma)/Antibacterial and antifungal/Active against fungal biofilms [25, 28, 29]
73 Meloxicam Anti-inflammatory/Immunomodulatory Active against fungal biofilms [28]
74 Metformin Diabetes Anti-nonsmall cell lung cancer, and augmented resistance in aging, Colo rectal cancer [26, 32]
75 Methotrexate Leukemia Breast cancer [9]
76 Mibefradil (Posicor) Antihypertensive, calcium channel blocker Short term use as an adjuvant in cancer therapy [26]
77 Midazolam Antipsychotic/Antidepressant Active against fungal biofilms [28]
78 Mifepristone Emergency contraceptive Cushing’s syndrome [29]
79 Mycophenolic acid Immunosuppressant Anticancer [30]
80 Nelfinavir HIV protease inhibitor Solid tumors [26]
81 Niclosamide Helminthiasis/Anti-infective

Antibacterial and antifungal and active against fungal biofilms

Treats multidrug-resistant leukemia

[25, 28, 33]
82 Nitazoxanide Antiprotozoal agent Influenza [26]
83 Nitroxoline Anti-infective Active against fungal biofilms [28]
84 Nortriptyline Antipsychotic/Antidepressant Active against fungal biofilms [28]
85 Oxyclozanide Helminthiasis Antibacterial and antifungal [25]
86 Paclitaxel Ovarian cancer, atrial restenosis Breast cancer [9]
87 Pentetic acid Hypocalcaemia Antibacterial and antifungal [25]
88 Perhexiline maleate Anti-anginal Active against fungal biofilms [28]
89 Phenobarbitone Anticonvulsant Active against fungal biofilms [28]
90 Pimozide Severe Tourette’s syndrome and schizophrenia Antibacterial and antifungal [25]
91 Promethazine Anti-inflammatory/Immunomodulatory Active against fungal biofilms [28]
92 Propanolol Antiarrhythmic Active against fungal biofilms [28]
93 Pyrvinium pamoate Anti-infective Active against fungal biofilms [28]
94 Quinacrine Helminthiasis Antibacterial and antifungal [25]
95 Raloxifene Osteoporosis in postmenopausal women Breast cancer [9]
96 Rapamune Anti-inflammatory/Immunomodulatory Active against fungal biofilms [28]
97 Remdesvir Inhibitors of RNA-dependent RNA polymerase in virus (Antiviral drug) Effective against SARS-CoV-2

[27,

34]

98 Ribavirin Antiviral drug Effective against SARS-CoV-2(COVID-19) [27, 34]
99 Rifampicin Anti-infective Active against fungal biofilms [28]
100 Riluzol Glutamate antagonist Secondary progressive multiple sclerosis (SPMS) [26]
101 Saracatinib Cancer therapy Mild to moderate Alzheimer disease [26]
102 Sildenafil (Viagra) Angina Erectile dysfunction, [29]
103 Silymarin Anti-hepatotoxic Active against fungal biofilms [28]
104 Simvastatin Hyper-cholesterolemia (Lipid-lowering) Antibacterial and antifungal [25, 28]
105 Sirolimus and Zoledronic acid Prophylaxis of organ rejection, Osteoporosis respectively Osteosarcoma (combined with Metzolimos, metronomic cyclophosphamide, methotrexate) [29]
106 Statins Hyper-cholesterolaemia Oncology [26]
107 Streptozotocin Pancreatic islet cell cancer Antibacterial and antifungal [25]
108 Sulfadiazine Anti-infective Active against fungal biofilms [28]
109 Sulfadimethoxine Anti-infective Active against fungal biofilms [28]
110 Sulfamethoxazole Anti-infective Active against fungal biofilms [28]
111 Sulfamethoxy-pyridazine Anti-infective Active against fungal biofilms [28]
112 Tacrolimus Anti-inflammatory/Immunomodulatory Active against fungal biofilms [28]
113 Tamoxifen Breast cancer, Albright syndrome, ovulation induction Antibacterial and antifungal [9, 25]
114 Teicoplanin Antibacterial Antiviral, potentially repurposable for COVID-19 treatment [27]
115 Telmisartan Blood pressure reduction Abdominal aortic aneurysm [26]
116 Thalidomide Antiemetic Hanson’s disease, Leprosy [26]
117 Thiotepa Immunosuppressant Breast cancer [9]
118 Tigecycline Anti-infective Active against fungal biofilms [28]
119 Tocilizumab Immunosuppressive drug for cytokine release syndrome Severe COVID-19 infection [27, 34]
120 Toremifene Infertility with an ovulatory disorder Breast cancer [9]
121 Valproic acid Anticonvulsant Active against fungal biofilms [28]
122. Verapamil Antiarrhythmic Active against fungal biofilms [28]
123. Vinblastine Hodgkin lymphoma, non-Hodgkin’s lymphoma, histiocytosis Breast cancer [9]
124. Yohimbine hydrochloride Vasodilator Active against fungal Biofilms [28]

Challenges

Although screening efforts are relatively inexpensive, approved drug clinical trials are expensive. The advantage of drug repurposing is that the early stages of clinical development are complete; hence, the drugs can proceed to clinical studies. However, doing clinical studies directly without preclinical studies is a risk. Inaccurate identification of drugs may prove to have no significant impact on therapy or mortality rates, and it may result in a loss in terms of treatment and expense.

Identifying a new therapeutic indication for an existing drug is a significant challenge. Choosing the right therapeutic area for the drug under investigation, evaluating the clinical trials with respect to the new therapeutic use, and deciding which stage of the clinical study or preclinical study shall be restarted are a few challenges repurposing. New preclinical and clinical trials may be required to be carried out if the available data are not satisfactory and do not comply with the requirements of regulatory agencies.

Another critical issue is patent application and intellectual property rights (IPR). Patents or IPR can prevent some repurposed drugs from entering the market, and IP protection for drug repurposing is minimal. Hence, the regulatory constraints and the risk of abandoning repurposing projects due to unsatisfactory results can discourage the investment of money or resources towards drug repurposing. A spike in market demand can be an excellent motivator to shift researchers and investors into action.

CONCLUSIONS

There are numerous diseases for which good therapeutic options have not been developed. The concept of repurposing a drug enables exploring the hidden potential of many molecules and better utilization of therapeutic agents. For better drug repositioning, more in-depth understanding along with integrated approaches between computational and experimental methods may be required to ensure high success rates of repositioned drugs.

COMPLIANCE WITH ETHICAL STANDARDS

The authors declare that they have no conflicts of interest.

This article does not contain any studies involving human participants performed by any of the authors and does not contain any studies involving animals performed by any of the authors.

REFERENCES

  • 1.Oprea T.I. AAPS J. 2012;14:759–763. doi: 10.1208/s12248-012-9390-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Strittmatter S.M. Nat Med. 2014;20:590–591. doi: 10.1038/nm.3595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Dinić J., Efferth T., García-Sosa A.T., Grahovac J., Padrón J.M., Pajeva I., Rizzolio F., Saponara S., Spengler G., Tsakovska I. Drug Resist. Updates. 2020;52:100713. doi: 10.1016/j.drup.2020.100713. [DOI] [PubMed] [Google Scholar]
  • 4.Jin G., Wong S.T.C. Drug Discovery Today. 2014;19:637–644. doi: 10.1016/j.drudis.2013.11.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Turner N., Zeng X.Y., Osborne B., Rogers S., Ye J.M. Trends Pharmacol. Sci. 2016;37:379–389. doi: 10.1016/j.tips.2016.01.007. [DOI] [PubMed] [Google Scholar]
  • 6.Rudrapal, M., Khairnar, S.J., and Jadhav, A.G., Drug Repurposing (DR): An Emerging Approach in Drug Discovery, 2020. 10.5772/intechopen.93193
  • 7.Hughes J.P., Rees S., Kalindjian S.B., Philpott K.L. Br. J. Pharmacol. 2011;162:1239–1249. doi: 10.1111/j.1476-5381.2010.01127.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Kalita J., Chetia D., Rudrapal M. Med. Chem. 2020;16:928–937. doi: 10.2174/1573406415666190806154722. [DOI] [PubMed] [Google Scholar]
  • 9.Aggarwal S., Verma S.S., Aggarwal S., Gupta S.C. Semin. Cancer Biol. 2020;68:8–20. doi: 10.1016/j.semcancer.2019.09.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Cha Y., Erez T., Reynolds I.J., Kumar D., Ross J., Koytiger G., Kusko R., Zeskind B., Risso S., Kagan E., Papapetropoulos S., Grossman I., Laifenfeld D. Br. J. Pharmacol. 2018;175:168–180. doi: 10.1111/bph.13798. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Agrawal P. J. Pharmacovigil. 2018;6:1–2. doi: 10.1016/j.drudis.2020.10.010. [DOI] [Google Scholar]
  • 12.Allarakhia M. Drug Des. Dev. Ther. 2013;7:753–766. doi: 10.2147/DDDT.S46289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Parvathaneni V., Kulkarni N.S., Muth A., Gupta V. Drug Discovery Today. 2019;24:2076–2085. doi: 10.1016/j.drudis.2019.06.014. [DOI] [PubMed] [Google Scholar]
  • 14.Padhy B.M., Gupta Y.K. J. Postgrad. Med. 2011;57:153. doi: 10.4103/0022-3859.81870. [DOI] [PubMed] [Google Scholar]
  • 15.Agrawal P. J. Pharmacovigil. 2015;2:1–2. doi: 10.4172/2329-6887.S2-e002. [DOI] [Google Scholar]
  • 16.Pantziarka P., Bouche G., Meheus L., Sukhatme V., Sukhatme V.P., Vikas P. Ecancermedicalscience. 2014;8:442. doi: 10.3332/ecancer.2014.442. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Xue H., Li J., Xie H., Wang Y. Int. J. Biol. Sci. 2018;14:1232–1244. doi: 10.7150/ijbs.24612. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Kim J.H., Scialli A.R. Toxicol. Sci. 2011;122:1–6. doi: 10.1093/toxsci/kfr088. [DOI] [PubMed] [Google Scholar]
  • 19.Pannikar V. Lepr. Rev. 2003;74:286–288. doi: 10.47276/lr.74.3.286. [DOI] [PubMed] [Google Scholar]
  • 20.Abbruzzese C., Matteoni S., Signore M., Cardone L, Nath K., Glickson J.D., Paggi M.G. J. Exp. Clin. Cancer Res. 2017;36:169. doi: 10.1186/s13046-017-0642-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.March-Vila E., Pinzi L., Sturm N., Tinivella A., Engkvist O., Chen H., Rastelli G. Front. Pharmacol. 2017;8:298. doi: 10.3389/fphar.2017.00298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Napolitano F., Zhao Y., Moreira V.M., Tagliaferri R., Kere J., D’mato M., Greco D. J. Cheminform. 2013;5:30. doi: 10.1186/1758-2946-5-30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Kitchen D.B., Decornez H., Furr J.R., Bajorath J. Nat. Rev. Drug Discovery. 2004;3:935–949. doi: 10.1038/nrd1549. [DOI] [PubMed] [Google Scholar]
  • 24.Sgobba M., Caporuscio F., Anighoro A., Portioli C., Rastelli G. Eur. J. Med. Chem. 2012;58:431–440. doi: 10.1016/j.ejmech.2012.10.024. [DOI] [PubMed] [Google Scholar]
  • 25.Miró-Canturri A., Ayerbe-Algaba R., Smani Y. Front. Microbiol. 2019;10:41. doi: 10.3389/fmicb.2019.00041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Schein C.H. Med. Res. Rev. 2020;40:586–605. doi: 10.1002/med.21627. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Jean S.S., Hsueh P.R. Expert Rev. Anti Infect. Ther. 2020;18:843–847. doi: 10.1080/14787210.2020.1771181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.de Mello T.P., Silva L.N., Ramos L.S., Frota H.F., Branquinha M.H., dos Santos A.L.S. Curr. Top. Med. Chem. 2020;20:509–516. doi: 10.2174/156802662007200316142626. [DOI] [Google Scholar]
  • 29.Hernandez J.J., Pryszlak M., Smith L., Yanchus C., Kurji N., Shahani V.M., Molinski S.V. Front. Oncol. 2017;7:273. doi: 10.3389/fonc.2017.00273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Shim J.S., Liu J.O. Int. J. Biol. Sci. 2014;10:654–663. doi: 10.7150/ijbs.9224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Das J. J. Med. Chem. 2020;63:13514–13525. doi: 10.1021/acs.jmedchem.0c01193. [DOI] [PubMed] [Google Scholar]
  • 32.Sang, J., Tang, R., Yang, M., and Sun, Q., Biomed. Res. Int., 2020, pp. 1–9. 10.1155/2020/9312149 [DOI] [PMC free article] [PubMed]
  • 33.Hamdoun S., Jung P., Efferth T. Front. Pharmacol. 2017;8:1–11. doi: 10.3389/fphar.2017.00110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Ng Y.L., Salim C.K., Chu J.J.H. Pharmacol. Ther. 2021;228:1–14. doi: 10.1016/j.pharmthera.2021.107930. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Russian Journal of Bioorganic Chemistry are provided here courtesy of Nature Publishing Group

RESOURCES