Int J App Pharm, Vol 16, Issue 3, 2024, 56-62Reviewl Article

REVITALIZING THERAPEUTICS: DRUG REPURPOSING AS A COST-EFFECTIVE STRATEGY FOR DRUG DEVELOPMENT

SHIVANI MAKHIJANI*

Department of Chemical Engineering, Rajabajar Science College, University of Calcutta, 92, Acharyya Prafulla Chandra Road, Kolkata-700009, West Bengal, India
*Corresponding author: Shivani Makhijani; *Email: shivanimakhijani.pharmacy@dmiher.edu.in

Received: 09 Oct 2023, Revised and Accepted: 01 Feb 2024


ABSTRACT

The process of developing new drugs is known for being drawn-out, expensive, risky, and having a high attrition rate. Drug repurposing has grown in favor recently as a practical way to speed up the development of new medicines while reducing the costs and time constraints associated with traditional drug research. The description of this study's pharmacological repurposing highlights its promise as a practical method to fill gaps in the market and revitalize treatment options. This review provides a full analysis of the ground-breaking tactic of repurposing medications, supported by numerous cases that demonstrate its revolutionary potential. We examine instances of repurposed drugs, such as thalidomide, sildenafil, and metformin, that have performed astoundingly well in a range of therapeutic settings despite being used outside of their original scope.

Overall, the paper's main goal-to study pharmacological repurposing as a potentially successful strategy for revitalizing treatments-is, succinctly summarized in this abstract. It highlights the potential benefits of this approach and how it might be used in the pharmaceutical industry's ongoing quest for more inexpensive and effective medicine development.

Keywords: Drug repurposing, Oncology, Anthelmintics, COVID-19, M. Tuberculosis and Sildenafil


INTRODUCTION

Despite improvements in technology and our understanding of human disease, we have seen far slower than anticipated progress in developing new therapeutics [1, 2]. It has been calculated that less than a dollar of value is generated for every dollar spent on research and development (R and D) on average [3]. Due to the increasing cost and length of time needed for new medication development, this may make the pharmaceutical business a less desirable alternative for investors.

Finding new medical uses for already approved, abandoned, shelved, and experimental drugs is a process known as drug repurposing, also known as drug repositioning, drug reprofiling, indication expansion, or indication shift. Although this tactic is not new, it has gained significant traction in the last ten years: around one-third of recent approvals are related to drug repurposing, and repurposed medications today account for about 25% of the pharmaceutical industry's yearly income [4]. The earliest stages of clinical trials can be omitted because the drug's efficacy, safety, and toxicity are already known, which reduces their cost and duration. A new drug must be developed for around 15 y before it can be sold, although repurposed drugs can be developed faster and for less money [5].

Existing compounds are those that have undergone successful Phase I or Phase II clinical studies and have a demonstrated safety and tolerability profile. As a result, a prospective repurposing medicine will have a well-established safety and toxicity profile and will have gathered data in preparation for regulatory approval [6]. The current review provides an overview of methods currently employed for repurposing and discusses case studies that demonstrate the effectiveness and utility of drug repurposing as evidenced by the significantly shorter development time for new drugs as a result of the availability of all pertinent clinical and toxicological data. This article discusses various repurposing strategies and the accompanying difficulties [7].

As we delve further into this review, we will cover successful case studies, various tactics employed in drug repurposing, as well as the difficulties that come with this method. By shedding light on the drug repurposing industry, we hope to shed light on its enormous potential to treat a variety of ailments, ultimately enhancing patient outcomes and altering the pharmaceutical industry.

Fig. 1: Illustration of traditional method and drug repurposing

Significance of drug repurposing in healthcare

A new medicine must adhere to strict guidelines in order to be sold. Due to the various physicochemical features of the chemical entities and the difficulty of scaling up the manufacturing, it takes a considerable investment to identify a medicine and subsequently develop it [8–10]. A key strategy in drug development is drug repurposing. According to some statistics, between 2007 and 2009, the US Food and Drug Administration (FDA) authorized around 30–40% of novel pharmaceuticals and biologics that can be classified as repurposed or repositioned products [11].

Many experts believe that repurposing medications can be more efficient than standard drug development techniques in terms of speed, cost, safety, and effectiveness. This is mostly due to the fact that since the initial stages of development that establish medicine safety have already been completed, researchers can skip them [12]. However, it could be challenging to pinpoint exactly how much time, risk, and money are saved due to some contradicting information [13].

The preclinical, pharmacokinetic, pharmacodynamic, and toxicity characteristics of the drug are all known, which lowers the risk of compound development and is one of the key benefits of a drug repurposing procedure. As a result, the medicine can be used in Phase II and III clinical studies quickly, resulting in lower development costs [14], a higher return on investment, and a shorter development period [15]. From the standpoint of intellectual property (IP) and patent protection, drug repurposing is also intriguing because, assuming the new use is not specifically stated and supported by the original patents, patent protection for a new use of an existing drug whose composition of matter patents are still in force may be obtained [16].

One effective example of repurposing is the phosphodiesterase type 5 (PDE5) inhibitor sildenafil. The FDA granted sildenafil approval for its use in treating erectile dysfunction despite the fact that it was initially created to treat hypertension. Later, it was modified to treat the uncommon condition pulmonary hypertension [17].

Challenges in drug repurposing

Despite recent growth in popularity, there are fewer applications than anticipated due to several challenges to effective deployment.

Strategies for accelerating drug repurposing

Infectious diseases: repurposing for rapid response

Anthelmintics for drug repurposing

A class of substances known as anthelmintics exhibit anti-infective activity against helminths that colonize the human intestine [25]. Anthelmintics come in a number of chemical forms and work by altering the metabolism of the parasite (worm) or paralyzing it so that the parasite can be killed by the immune system of the host. It has been shown that several anthelmintics have the ability to block important oncogenic pathways, including Wnt/b-catenin and STAT3 [25]; therefore, their application for cancer treatment has been considered.

The updated knowledge regarding anthelmintics with anti-cancer activity and their potential use as anti-cancer medicines (i.e., repositioned pharmaceuticals) is summarized in this paragraph.

The anti-tumor properties of albendazole have been proven in vitro against hepatocellular carcinoma (HCC) and colorectal carcinoma (CRC), as well as in vivo against a xenograft model of peritoneal carcinomatosis. Additionally, the drug has demonstrated antiproliferative effects on tumor cells, including those resistant to other microtubule-targeting medications (such as leukemia and ovarian cancer cells) [26].

Table 1: list of anthelmintic that have been repositioned in oncology and have shown to have anti-cancer effects

Drug name New indication Comments Reference
Ivermectin Colon and Lung Cancer Block the T-cell factor (TCF) family transcriptional factor's main signaling pathway, which is influenced by the wingless-related integration site (wnt) gene. [26]
Levamisole Hepatocellular carcinoma Boost the tumor necrosis factor levelDeath receptor 4 (DR4)-independent apoptosis rate by limiting the phosphorylation of c-Jun N-terminal kinase (JNK)-related apoptosis-inducing ligand (TRAIL) [25]
Mebendazole Colorectal cancer Mebendazole inhibits a number of processes that help tumors grow, including angiogenesis, the polymerization of tubulin, pro-survival pathways, and matrix metalloproteinases. [27]
Praziquantel Anticancer When combined with paclitaxel, praziquantel inhibits cell growth and induces death in a variety of cancer cells, including CRC DLD-1. [25]

Drug repurposing for mycobactrium tuberculosis

The bacterium mycobacterium tuberculosis, which causes tuberculosis (TB), is now regarded as the world's most common cause of bacterial infection-related death [28]. According to recent estimates, 10 million individuals worldwide contract M. TB each year, infecting more than 30% of the world's population [28]. Fortunately, the finding of novel therapeutic options against extensively drug-resistant (XDR-TB) strains of tuberculosis, which are causing infections that are incurable, is currently seen as a very promising strategy through pharmacological repurposing of antibiotics [29]. Although there are a number of experimental and in silico methods to uncover drugs with repurposing potential, knowledge-based strategies, molecular docking, and phenotypic screening are the most frequently employed [30].

Metformin, a medication for type 2 diabetes, has been scientifically shown to have anti-tuberculosis effects (table 2). Particularly, metformin enhances phagosome-lysosome fusion and increases ROS concentration during the oxidative burst, preventing bacterial colonization, reducing lung damage and chronic inflammation, enhancing the immune response against tuberculosis, and enhancing the activity of conventional anti-tuberculosis medication [31-33].

Table 2: List of drugs repurposed against M. tuberculosis

Repurposed drugs Primary mechanism of action Reference
Transition metals (Cu2+and Co2+) Interfering with urease [36]
Eltrombopag Inhibition of Zmp1 and PDF [37]
Metformin phagosome–lysosome fusion [31-33]
Simvastatin HMG-CoA inhibition [38]
Doxycycline Matrix metalloprotease inhibition [39, 40]
Gefitinib EGFR inhibition [41]

Similar to the way statins are used to treat hypercholesterolemia and atherosclerotic cardiovascular disease, they are also a very promising source of antimicrobial substances that are effective against M. tuberculosis [34]. Simvastatin, for instance, can lessen the amount of germs present inside cells when taken with other antitubercular drugs [35, 34]. This drug's mechanism of action appears to include inhibiting the 3-hydroxy-3-methylglutaryl-coenzyme enzyme.

Drug repurposing for dengue

The most common virus spread by mosquitoes in the world today is dengue. Four antigenically different serotypes of the dengue virus (DENV), notably DENV1-4, are the cause of this viral illness that is spread by mosquitoes. There is still a need for an efficient antiviral treatment for Dengue Virus infection, and several replicative cycle inhibitors are now being investigated. The repurposing of licensed medications used for other diseases to find novel inhibitors of this pathogen represents an appealing approach for a swift therapeutic intervention [42], given the rapid spread of DENV and the typical timescale necessary for bringing a new drug to market. The repositioning of well-known medications for the prevention of DENV replication will be described in detail in the following paragraphs through a number of exemplary examples.

Fig. 2: Life cycle of dengue and steps inhibited by repurposed drug

A powerful protease inhibitor, nelfinavir (AG1343) is effective against human immunodeficiency virus type 1 (HIV-1) (24). An effort to reposition peptidomimetics against Nelfinavir and other viral protease inhibitors, such as Lopinavir and Ritonavir, were chosen as treatments for dengue virus infection using computer-aided drug design and molecular modeling [43]. Positive interactions between Nelfinavir and DENV NS2B-NS3 protease were found using molecular docking calculations and molecular dynamics simulations [42].

Similar to this, the antimalarial drug chloroquine has also been used to treat rheumatoid arthritis, systemic lupus erythematosus, and amebic liver abscesses by systemic therapy [44]. In the DENV scenario, chloroquine was utilized in a number of medication repositioning trials. Plaque test and qRT-PCR results demonstrated that it can prevent Dengue Virus Type 2 multiplication in Vero cells at a dose of 5 g/ml (the cytotoxic level is 500 g/ml) [45].

Table 3: List of repurposed drugs against dengue virus

Drug Original indication Activity against dengue virus Reference
Nelfinavir Antivirals NS2B-NS3 protease inhibition [42, 43]
Balapiravir Antivirals RNA-dependent RNA polymerase inhibition [46, 47]
Chloroquine Antimalarics Inhibits low-pH dependent entry steps [45]
Amodiaquine Antimalarics Inhibits low-pH dependent entry steps [48]
Celgosivir Antidiabetics Accumulation of NS1 in ER [49, 50]
Montelukast Antihistamines Reduction of vascular leakage DENV-induced [51]
Dasatinib Anticancers Src Fyn kinases inhibition [52]

Drug repurposing for breast cancer

Globally, physical examinations, breast scans, and tissue biopsies are used to diagnose breast cancer, one of the most prevalent cancers in women. 2018 saw a total of 2.089 million new instances of breast cancer, of which 627,000 cases resulted in death [53]. About 15% of all female cancer deaths worldwide were caused by this [53].

In India, there were 87,090 reported fatalities and 162,468 new cases of breast cancer in 2018, according to NICPR. According to one of the most current studies on the risk of breast cancer in India, 1 in 28 women will have breast cancer throughout their lifetime. In India, the incidence/mortality ratio is 0.48, which is more than in other nations [68].

The therapy options are quite expensive and have negative side effects. Repositioning older, off-patent, non-cancer medications with clinical approval and known targets into newer indications is analogous to employing older weaponry in a more recent conflict. Over the past ten years, various drugs have been repositioned for the treatment of breast cancer, including alkylating substances, anthracyclins, antimetabolites, CDK4/6 inhibitors, aromatase inhibitors, mTOR inhibitors, and mitotic inhibitors [54].

The PI3K/AKT/mTOR signaling pathway is inhibited by the mTOR kinase inhibitor everolimus. Everolimus was initially licensed for the treatment of pancreatic cancer in 2011, renal transplant immunosuppression, and renal cancer in 2009. Following the successful conclusion of a phase III clinical trial known as "Breast Cancer Trial of Oral Everolimus-2 (BOLERO-2)" that included everolimus in combination with exemestane, everolimus was approved by the US FDA in 2012 for the treatment of HR+, HER2-advanced metastatic cancers that are resistant to letrozole or anastrazole [55, 56].

A N,N',N''-triethylenephosphoramide (TEPA) derivative called thiotepa was introduced in 1953 as an immunosuppressive medication for transplantation in hematological illnesses [57]. The medication was subsequently advised for solid tumors in 1959 [58] and breast cancer (0.3 to 0.4 mg/kg IV repeated every 1 to 4 w) in 1963.

Drug repurposing for COVID-19

The use of repurposed medications has been used to treat a number of epidemic diseases, and the coronavirus disease 2019 (COVID-19) pandemic is one such instance [59]. A new coronavirus known as severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is the source of COVID-19 [59]. A key phase in the infection process is the interaction of the viral spike protein with the human ACE2 and TMPRSS2 enzymes, according to the evidence for the mechanism of infection, which was also derived from earlier investigations on coronaviruses. The peptidase domain of the human ACE2 enzyme interacts to the receptor-binding domain of the spike protein [60].

Remdesivir is thought to be a promising drug candidate for repurposing against COVID-19 based on the existing understanding of its use in SARS-CoV2 infection. A nucleoside analog called Remdesivir (GS-5734) was initially created by Gilead Sciences Inc, a biopharmaceutical company with headquarters in the United States, to combat Ebola viruses. Despite the medication's ineffectiveness against the Ebola virus, preliminary findings from non vitro and in vivo preclinical research, as well as case reports, point to its effectiveness against the SARS-CoV2 virus [61-63].

Favipiravir is a prodrug that is produced by T-1105's pyrazine moiety being chemically modified. Because there is currently no proven cure for COVID-19 and there is a strong focus on repurposing existing medications because to the lengthy time it would take to develop new ones, favipiravir is swiftly becoming the medicine of choice. One new repurposing medication for the treatment of novel viruses is favipiravir [67].

The primary proteases of the coronavirus and HIV are aspartic and cysteine, respectively, in the lopinavir-ritonavir combination. It has been found that the non-specific protease inhibition of protease inhibitors used in HIV therapy makes them effective against SARS-CoV. The SARS-CoV2 and HIV-1 proteases have comparable binding energies to lopinavir [64]. Significant virus clearance has been accomplished in SARS-CoV2 patients with lopinavir-ritonavir treatment [65,66]. A 47-year-old patient quickly improved with extra lopinavir and ritonavir tablet therapy after failing to respond to methylprednisolone and interferon therapy.

CONCLUSION

The identification of new indications for currently existing pharmaceuticals (drug repositioning) could improve and boost the actual number of new medicines that reach the market because the traditional drug-discovery technique is frequently a lengthy, difficult, and expensive procedure. Thanks to the faster drug discovery route on which this technique is based, drug repositioning strives to meet the need for new medications for a given disease, notably for developing diseases or for those yet without treatment. For usage as antitubercular medicines, certain substances have previously received approval or are in the final phases of clinical testing. The most effective medications target the activation or inhibition of host genes that enable bacterial colonization, which may also prevent the selection of new strains that are resistant to antibiotics.

FUNDING

Nil

AUTHORS CONTRIBUTIONS

All authors contributed equally

CONFLICT OF INTERESTS

The authors declare that there are no conflicts of interest in this review article.

REFERENCES

  1. Ashburn TT, Thor KB. Drug repositioning: identifying and developing new uses for existing drugs. Nat Rev Drug Discov. 2004 Nov 22;3(8):673-83. doi: 10.1038/nrd1468, PMID 15286734.

  2. Scannell JW, Blanckley A, Boldon H, Warrington B. Diagnosing the decline in pharmaceutical R&D efficiency. Nat Rev Drug Discov. 2012 Mar 1;11(3):191-200. doi: 10.1038/nrd3681, PMID 22378269.

  3. Naylor S, Kauppi DM, Schonfeld JP. Therapeutic drug repurposing, repositioning and rescue part II: business review. Roundtable on translating genomic-based research for health; board on health sciences policy; Institute of Medicine. Drug repurposing and repositioning: workshop summary. Drug Discov World. Washington (DC): National Academies Press (US). 2014 Aug 8:16(2):57-72, PMID 24872991.

  4. Chong CR, Sullivan DJ Jr. New uses for old drugs. Nature. 2007 Aug 9;448(7154):645-6. doi: 10.1038/448645a, PMID 17687303.

  5. Cha Y, Erez T, Reynolds IJ, Kumar D, Ross J, Koytiger G. Drug repurposing from the perspective of pharmaceutical companies. Br J Pharmacol. 2018 Jan;175(2):168-80. doi: 10.1111/bph.13798, PMID 28369768, PMCID PMC5758385.

  6. Parveen S, Alnoman RB, Bayazeed AA, Alqahtani AM. Computational insights into the drug repurposing and synergism of FDA-approved influenza drugs binding with SARS-CoV-2 protease against COVID-19. Am J Microbiol Res. 2020;8(3):93-102.

  7. Vaidya B, Parvathaneni V, Kulkarni NS, Shukla SK, Damon JK, Sarode A. Cyclodextrin modified erlotinib loaded PLGA nanoparticles for improved therapeutic efficacy against non-small cell lung cancer. Int J Biol Macromol. 2019 Feb 1;122:338-47. doi: 10.1016/j.ijbiomac.2018.10.181, PMID 30401652.

  8. Kulkarni NS, Guererro Y, Gupta N, Muth A, Gupta V. Exploring potential of quantum dots as dual modality for cancer therapy and diagnosis. J Drug Deliv Sci Technol. 2019;49:352-64. doi: 10.1016/j.jddst.2018.12.010.

  9. Kulkarni NS, Parvathaneni V, Shukla SK, Barasa L, Perron JC, Yoganathan S. Tyrosine kinase inhibitor conjugated quantum dots for non-small cell lung cancer (NSCLC) treatment. Eur J Pharm Sci. 2019 May 15;133:145-59. doi: 10.1016/j.ejps.2019.03.026, PMID 30946965.

  10. Graul AI, Sorbera L, Pina P, Tell M, Cruces E, Rosa E. The year’s new drugs & biologics–2009. Drug News Perspect. 2010 Jan-Feb;23(1):7-36. doi: 10.1358/dnp.2010.23.1.1440373, PMID 20155217.

  11. Ashburn TT, Thor KB. Drug repositioning: identifying and developing new uses for existing drugs. Nat Rev Drug Discov. 2004 Nov 22;3(8):673-83. doi: 10.1038/nrd1468, PMID 15286734–683.

  12. Krishnamurthy. Grimshaw N, AA, Axson SA, Choe SH, Miller JE. Drug repurposing: a systematic review on root causes, barriers and facilitators. BMC Health Serv Res. 2022;22(1):1-17.

  13. Mignani S, Huber S, Tomás H, Rodrigues J, Majoral JP. Why and how have drug discovery strategies in pharma changed? What are the new mindsets? Drug Discov Today. 2016 Feb;21(2):239-49. doi: 10.1016/j.drudis.2015.09.007, PMID 26376356.

  14. Novac N. Challenges and opportunities of drug repositioning. Trends Pharmacol Sci. 2013 May;34(5):267-72. doi: 10.1016/j.tips.2013.03.004, PMID 23582281.

  15. Vanhaelen Q, Mamoshina P, Aliper AM, Artemov A, Lezhnina K, Ozerov I. Design of efficient computational workflows for in silico drug repurposing. Drug Discov Today. 2017 Feb;22(2):210-22. doi: 10.1016/j.drudis.2016.09.019, PMID 27693712.

  16. Hatzimouratidis K. Sildenafil in the treatment of erectile dysfunction: an overview of the clinical evidence. Clin Interv Aging. 2006;1(4):403-14. doi: 10.2147/ciia.2006.1.4.403, PMID 18046917, PMCID PMC2699643.

  17. Hurle MR, Yang L, Xie Q, Rajpal DK, Sanseau P, Agarwal P. Computational drug repositioning: from data to therapeutics. Clin Pharmacol Ther. 2013 Apr;93(4):335-41. doi: 10.1038/clpt.2013.1, PMID 23443757.

  18. Knight JM, Kerswill SA, Hari P, Cole SW, Logan BR, D’Souza A. Repurposing existing medications as cancer therapy: design and feasibility of a randomized pilot investigating propranolol administration in patients receiving hematopoietic cell transplantation. BMC Cancer. 2018;18(1):593. doi: 10.1186/s12885-018-4509-0, PMID 29793446.

  19. Ashburn TT, Thor KB. Drug repositioning: identifying and developing new uses for existing drugs. Nat Rev Drug Discov. 2004 Nov 22;3(8):673-83. doi: 10.1038/nrd1468, PMID 15286734.

  20. S. Pushpakom S, Iorio F, Eyers PA, Escott KJ, Hopper S, Wells A. Drug repurposing: progress, challenges and recommendations. Nat Rev Drug Discov. 2019 Jan;18(1):41-58. doi: 10.1038/nrd.2018.168, PMID 30310233.

  21. Liu Z, Fang H, Reagan K, Xu X, Mendrick DL, Slikker W Jr. In silico drug repositioning: what we need to know. Drug Discov Today. 2013 Feb;18(3-4):110-5. doi: 10.1016/j.drudis.2012.08.005, PMID 22935104.

  22. Scotti L, Mendonca Junior FJ, Ishiki HM, Ribeiro FF, Singla RK, Barbosa Filho JM. Docking studies for multi-target drugs. Curr Drug Targets. 2017;18(5):592-604. doi: 10.2174/1389450116666150825111818, PMID 26302806.

  23. Challener Cynthia A. Can artificial intelligence take the next step for drug repositioning? PharmTech. 2018;42(9):22-6.

  24. Laudisi F, Maronek M, Di Grazia A, Monteleone G, Stolfi C. Repositioning of anthelmintic drugs for the treatment of cancers of the digestive system. Int J Mol Sci. 2020 Jul 13;21(14):4957. doi: 10.3390/ijms21144957, PMID 32668817, PMCID PMC7404055.

  25. Armando RG, Mengual Gomez DL, Gomez DE. New drugs are not enough‑drug repositioning in oncology: an update. Int J Oncol. 2020 Mar;56(3):651-84. doi: 10.3892/ijo.2020.4966. PMID 32124955, PMCID PMC7010222.

  26. Guerini AE, Triggiani L, Maddalo M, Bonù ML, Frassine F, Baiguini A. Mebendazole as a candidate for drug repurposing in oncology: an extensive review of current literature. Cancers (Basel). 2019 Aug 31;11(9):1284. doi: 10.3390/cancers11091284, PMID 31480477, PMCID PMC6769799.

  27. Russell DG. Mycobacterium tuberculosis and the intimate discourse of a chronic infection. Immunol Rev. 2011 Mar;240(1):252-68. doi: 10.1111/j.1600-065X.2010.00984.x, PMID 21349098, PMCID PMC3174472.

  28. Kaur D, Mathew S, Nair CGS, Begum A, Jainanarayan AK, Sharma M. Structure-based drug discovery for designing leads for the non-toxic metabolic targets in multi-drug resistant Mycobacterium tuberculosis. J Transl Med. 2017 Dec 21;15(1):261. doi: 10.1186/s12967-017-1363-9, PMID 29268770, PMCID PMC5740895.

  29. Konreddy AK, Rani GU, Lee K, Choi Y. Recent drug-repurposing-driven advances in the discovery of novel antibiotics. Curr Med Chem. 2019;26(28):5363-88. doi: 10.2174/0929867325666180706101404, PMID 29984648.

  30. Naicker N, Sigal A, Naidoo K. Metformin as host-directed therapy for TB treatment: scoping review. Front Microbiol. 2020 Apr 29;11:435. doi: 10.3389/fmicb.2020.00435, PMID 32411100, PMCID PMC7201016.

  31. Tiberi S, du Plessis N, Walzl G, Vjecha MJ, Rao M, Ntoumi F. Tuberculosis: progress and advances in development of new drugs, treatment regimens, and host-directed therapies. Lancet Infect Dis. 2018 Jul;18(7):e183-98. doi: 10.1016/S1473-3099(18)30110-5. Erratum in: Lancet infect, Dis. 2018 Apr 27. PMID 29580819.

  32. Mishra R, Krishan S, Siddiqui AN, Kapur P, Khayyam KU, Sharma M. Potential role of adjuvant drugs on efficacy of first-line oral antitubercular therapy: drug repurposing. Tuberculosis (Edinb). 2020 Jan;120:101902. doi: 10.1016/j.tube.2020.101902, PMID 32090863.

  33. Miro Canturri A, Ayerbe Algaba R, Smani Y. Corrigendum: drug repurposing for the treatment of bacterial and fungal infections. Front Microbiol. 2022;13:844615. doi: 10.3389/fmicb.2022.844615, PMID 35283839.

  34. Pacios O, Blasco L, Bleriot I, Fernandez Garcia L, Gonzalez Bardanca M, Ambroa A. Strategies to combat multidrug-resistant and persistent infectious diseases. Antibiotics (Basel). 2020 Feb 6;9(2):65. doi: 10.3390/antibiotics9020065, PMID 32041137, PMCID PMC7168131.

  35. Coelho TS, Halicki PCB, Silva L Jr, de Menezes Vicenti JR, Gonçalves BL, Almeida da Silva PE. Metal-based antimicrobial strategies against intramacrophage mycobacterium tuberculosis. Lett Appl Microbiol. 2020 Aug;71(2):146-53. doi: 10.1111/lam.13298, PMID 32286695.

  36. Battah B, Chemi G, Butini S, Campiani G, Brogi S, Delogu G. A repurposing approach for uncovering the anti-tubercular activity of FDA-approved drugs with potential multi-targeting profiles. Molecules. 2019 Nov 29;24(23):4373. doi: 10.3390/molecules24234373, PMID 31795400, PMCID PMC6930672.

  37. Guerra-De-Blas PDC, Bobadilla-Del-Valle M, Sada Ovalle I, Estrada Garcia I, Torres Gonzalez P, Lopez Saavedra A. Simvastatin enhances the immune response against Mycobacterium tuberculosis. Front Microbiol. 2019 Sep 20;10:2097. doi: 10.3389/fmicb.2019.02097, PMID 31616387, PMCID PMC6764081.

  38. Kim JH, O’Brien KM, Sharma R, Boshoff HIM, Rehren G, Chakraborty S. A genetic strategy to identify targets for the development of drugs that prevent bacterial persistence. Proc Natl Acad Sci USA. 2013;110(47):19095-100. doi: 10.1073/pnas.1315860110, PMID 24191058.

  39. Ranjbar S, Haridas V, Nambu A, Jasenosky LD, Sadhukhan S, Ebert TS. Cytoplasmic RNA sensor pathways and nitazoxanide broadly inhibit intracellular mycobacterium tuberculosis growth. iScience. 2019 Dec 20;22:299-313. doi: 10.1016/j.isci.2019.11.001, PMID 31805434, PMCID PMC6909047.

  40. Torfs E, Piller T, Cos P, Cappoen D. Opportunities for overcoming Mycobacterium tuberculosis Drug resistance: emerging mycobacterial targets and host-directed therapy. Int J Mol Sci. 2019 Jun 12;20(12):2868. doi: 10.3390/ijms20122868, PMID 31212777, PMCID PMC6627145.

  41. Botta L, Rivara M, Zuliani V, Radi M. Drug repurposing approaches to fight dengue virus infection and related diseases. Front Biosci (Landmark Ed). 2018 Jan 1;23(6):997-1019. doi: 10.2741/4630, PMID 28930586.

  42. Bhakat S, Delang L, Kaptein S, Neyts J, Leyssen P, Jayaprakash V. Reaching beyond HIV/HCV: nelfinavir as a potential starting point for broad-spectrum protease inhibitors against dengue and Chikungunya virus. RSC Adv. 2015;5(104):85938-49. doi: 10.1039/C5RA14469H.

  43. Eyer L, Valdes JJ, Gil VA, Nencka R, Hrebabecky H, Sala M. Nucleoside inhibitors of tick-borne encephalitis virus. Antimicrob Agents Chemother. 2015 Sep;59(9):5483-93. doi: 10.1128/AAC.00807-15, PMID 26124166, PMCID PMC4538560.

  44. Rolain JM, Colson P, Raoult D. Recycling of chloroquine and its hydroxyl analogue to face bacterial, fungal and viral infections in the 21st century. Int J Antimicrob Agents. 2007 Oct;30(4):297-308. doi: 10.1016/j.ijantimicag.2007.05.015, PMID 17629679, PMCID PMC7126847.

  45. Klumpp K, Leveque V, Le Pogam S, Ma H, Jiang WR, Kang H. The novel nucleoside analog R1479 (4’-azidocytidine) is a potent inhibitor of NS5B-dependent RNA synthesis and hepatitis C virus replication in cell culture. J Biol Chem. 2006;281(7):3793-9. doi: 10.1074/jbc.M510195200, PMID 16316989.

  46. Nguyen NM, Tran CN, Phung LK, Duong KT, Huynh A, Farrar J. A randomized, double-blind placebo-controlled trial of Balapiravir, a polymerase inhibitor, in adult dengue patients. J Infect Dis. 2013 May 1;207(9):1442-50. doi: 10.1093/infdis/jis470, PMID 22807519, PMCID PMC3610419.

  47. Boonyasuppayakorn S, Reichert ED, Manzano M, Nagarajan K, Padmanabhan R. Amodiaquine, an antimalarial drug, inhibits dengue virus type 2 replication and infectivity. Antiviral Res. 2014 Jun;106:125-34. doi: 10.1016/j.antiviral.2014.03.014, PMID 24680954, PMCID PMC4523242.

  48. Durantel D. Celgosivir, an alpha-glucosidase I inhibitor for the potential treatment of HCV infection. Curr Opin Investig Drugs. 2009 Aug;10(8):860-70, PMID 19649930.

  49. Whitby K, Taylor D, Patel D, Ahmed P, Tyms AS. Action of celgosivir (6 O-butanoyl castanospermine) against the pestivirus BVDV: implications for the treatment of hepatitis C. Antivir Chem Chemother. 2004 May;15(3):141-51. doi: 10.1177/095632020401500304, PMID 15266896.

  50. St John AL. Influence of mast cells on dengue protective immunity and immune pathology. PLOS Pathog. 2013;9(12):e1003783. doi: 10.1371/journal.ppat.1003783, PMID 24367254, PMCID PMC3868513.

  51. Talpaz M, Shah NP, Kantarjian H, Donato N, Nicoll J, Paquette R. Dasatinib in imatinib-resistant philadelphia chromosome-positive leukemias. N Engl J Med. 2006 Jun 15;354(24):2531-41. doi: 10.1056/NEJMoa055229, PMID 16775234.

  52. Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018 Nov;68(6):394-424. doi: 10.3322/caac.21492. Erratum in: CA Cancer J Clin. 2020 Jul;70(4):313, PMID 30207593.

  53. Aggarwal S, Verma SS, Aggarwal S, Gupta SC. Drug repurposing for breast cancer therapy: old weapon for new battle. Semin Cancer Biol. 2021 Jan;68:8-20. doi: 10.1016/j.semcancer.2019.09.012, PMID 31550502, PMCID PMC7128772.

  54. Royce ME, Osman D. Everolimus in the treatment of metastatic breast cancer. Breast Cancer (Auckl). 2015 Sep 6;9:73-9. doi: 10.4137/BCBCR.S29268, PMID 26417203, PMCID PMC4571987.

  55. Baselga J, Campone M, Piccart M, Burris HA, Rugo HS, Sahmoud T. Everolimus in postmenopausal hormone-receptor-positive advanced breast cancer. N Engl J Med. 2012 Feb 9;366(6):520-9. doi: 10.1056/NEJMoa1109653, PMID 22149876, PMCID PMC5705195.

  56. Sykes MP, Karnofsky DA, Philips FS, Burchenal JH. Clinical studies on triethylenephosphoramide and diethylenephosphoramide, compounds with nitrogen-mustard‐like activity. Cancer. 1953;6(1):142-8. doi: 10.1002/1097-0142(195301)6:1<142::AID-CNCR2820060114>3.0.CO;2-W.

  57. Kim KW, Roh JK, Wee HJ, Kim C. Cancer drug discovery, Springer; 2016.

  58. Pawar AY. Combating devastating COVID-19 by drug repurposing. Int J Antimicrob Agents. 2020 Aug;56(2):105984. doi: 10.1016/j.ijantimicag.2020.105984, PMID 32305589, PMCID PMC7162749.

  59. Dotolo S, Marabotti A, Facchiano A, Tagliaferri R. A review on drug repurposing applicable to COVID-19. Brief Bioinform. 2021 Mar 22;22(2):726-41. doi: 10.1093/bib/bbaa288, PMID 33147623, PMCID PMC7665348.

  60. Zhou P, Yang XL, Wang XG, Hu B, Zhang L, Zhang W. Addendum: a pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature. 2020;588(7836):E6. doi: 10.1038/s41586-020-2951-z, PMID 33199918.

  61. Holshue ML, DeBolt C, Lindquist S, Lofy KH, Wiesman J, Bruce H. First case of 2019 novel coronavirus in the United States. N Engl J Med. 2020;382(10):929-36. doi: 10.1056/NEJMoa2001191, PMID 32004427.

  62. Nicastri E, Petrosillo N, Ascoli Bartoli T, Lepore L, Mondi A, Palmieri F. National institute for the infectious diseases ”L. Spallanzani”, IRCCS. Recommendations for COVID-19 clinical management. Infect Dis Rep. 2020 Mar 16;12(1):8543. doi: 10.4081/idr.2020.8543, PMID 32218915, PMCID PMC7097833.

  63. Ortega JT, Serrano ML, Pujol FH, Rangel HR. Unrevealing sequence and structural features of novel coronavirus using in silico approaches: the main protease as molecular target. Excli J. 2020 Mar 17;19:400-9. doi: 10.17179/excli2020-1189, PMID 32210741, PMCID PMC7081067.

  64. Lim J, Jeon S, Shin HY, Kim MJ, Seong YM, Lee WJ. Case of the index patient who caused tertiary transmission of COVID-19 infection in Korea: the application of lopinavir/ritonavir for the treatment of COVID-19 infected pneumonia monitored by quantitative RT-PCR. J Korean Med Sci. 2020 Feb 17;35(6):e79. doi: 10.3346/jkms.2020.35.e79, PMID 32056407, PMCID PMC7025910.

  65. Xu K, Cai H, Shen Y, Ni Q, Chen Y, Hu S. [Management of COVID-19: the Zhejiang experience. Zhejiang Da Xue Xue Bao Yi Xue Ban. 2020 Feb 21;49(2):147-57. doi: 10.3785/j.issn.1008-9292.2020.02.02, PMID 32391658, PMCID PMC8800711.

  66. Jyothi BJ, Kavya VR. “Ultraviolet spectrophotometric method development for estimation of new antiviral repurposing drug favipiravir. Asian J Pharm Clin Res. 2021;14(7 Jul):67-9. doi: 10.22159/ajpcr.2021.

  67. Choudhary DG, K Kumar J. A study to assess knowledge, attitude and practice on breast cancer among women in government general hospital. Asian Journal of Pharmaceutical and Clinical Research. 2021;14(2):60-5. doi: 10.22159/ajpcr.2021.v14i2.40244.