View Article

Abstract

Drug reprofiling, also referred to as drug repurposing or repositioning, finds new therapeutic applications for drugs that are already on the market or have already failed. It provides quicker, less expensive, and less dangerous alternatives to traditional drug discovery. In in addition to addressing unmet medical needs in a variety of fields, such as infectious, neurodegenerative, oncologic, cardiovascular, metabolic, autoimmune, parasitic, and rare diseases, this strategy makes use of established safety and pharmacokinetic profiles to shorten development timelines and costs. Phenotypic and target-based approaches as well as computational methods like molecular docking are examples of strategies. Its effects during the COVID-19 pandemic and in chronic conditions where innovation is desperately needed are demonstrated by case studies. Drug reprofiling enhances drug lifecycles, speeds up personalised medicine, and supports global health initiatives by integrating into research pipelines and regulatory frameworks.

Keywords

Drug Reprofiling, drug repurposing, therapeutic innovation, repositioning, computational methods, phenotypic screening, molecular docking, COVID-19, rare diseases, personalized medicine

Introduction

Drug reprofiling, also known as drug repurposing or drug repositioning, is an emerging strategy in pharmaceutical research that focuses on identifying new therapeutic uses for existing or previously failed drugs(1). Unlike traditional drug discovery—which involves de novo synthesis, extensive preclinical studies, and time-consuming clinical trials—drug reprofiling offers a faster, cost-effective, and lower-risk approach by utilizing the known pharmacological and safety profiles of approved or investigational drugs(2). The process of developing a new drug from scratch can take over 10–15 years and cost billions of dollars, with a high rate of failure, especially in the later stages of clinical trials(3). Drug reprofiling circumvents many of these challenges by capitalizing on existing data, thereby significantly reducing development time and financial burden(4). It is particularly valuable in addressing urgent healthcare needs, such as emerging infectious diseases, rare diseases, and cancers, where treatment options are limited or non-existent(5). Numerous medications that have been repurposed have shown impressive results(6). For example, thalidomide, which was once discontinued because of its teratogenic effects, has been successfully repurposed for the treatment of leprosy sequelae and multiple myeloma(7). Another prominent example is sildenafil, which was first created to treat angina but was later used to treat pulmonary hypertension and erectile dysfunction(8). Advancements in computational biology, bioinformatics, and artificial intelligence have revolutionized drug reprofiling(9). In silico methods, molecular docking, network pharmacology, and machine learning now enable the rapid screening of large drug libraries against diverse disease targets(10). These technologies facilitate the identification of novel drug-disease relationships and support the rationale for clinical repurposing studies(11). In the current pharmaceutical landscape, drug reprofiling not only enhances the lifecycle of existing drugs but also contributes to personalized medicine and global health initiatives(12). Its integration into regulatory frameworks and drug development pipelines signifies a paradigm shift towards more efficient and sustainable therapeutic innovation(13).

1.2 Stages of Drug Reprofiling Without AI Involvement

Drug reprofiling, also known as drug repositioning, refers to the strategy of identifying new therapeutic applications for existing or previously studied drugs(14). While modern approaches often involve artificial intelligence and machine learning, traditional reprofiling techniques rely on well-established experimental methods, clinical observations, literature surveys, and conventional data analysis(15). These methods are still widely used and continue to contribute significantly to drug discovery(16). The following are the major stages of drug reprofiling without the involvement of AI:

1.2.1 Selection and literature review

Finding appropriate medication candidates for reprofiling is the initial step(17). Frequently taken into consideration are medications that have already received approval, been withdrawn for non-safety reasons, or been put on hold because of ineffectiveness(18). The pharmacological characteristics, safety profiles, and past clinical results of these medications are evaluated using literature reviews, regulatory documents, and clinical trial databases(19). Drugs with off-target effects or unexpected therapeutic responses documented in previous trials or case studies receive special attention(20).

1.2.2 Target identification through experimental and literature-based approaches

Target identification is done manually by looking through the available pharmacological data and experimental trials when AI technologies are not available. Researchers look into potential secondary targets based on published data on the drug's mechanisms of action, known biochemical interactions, and structural similarities with other medications. Physiological effects noted, Scientific journals, pharmacology textbooks, biochemical assays, and route analysis from experimental research are typically the sources of these insights (21).

1.2.3 Disease Association Based on Clinical and Experimental Evidence

Linking the medication to illnesses where these targets are implicated comes next, after potential new targets have been identified. This procedure depends on a thorough comprehension of clinical pathology, illness causes, and the body of available literature. A medication with anti-inflammatory properties, for example, may be taken into consideration for disorders like arthritis, inflammatory bowel disease, or neuroinflammatory diseases. At this point, observational research, clinical case reports, and experimental illness models are crucial resources(22,23).

1.2.4 Preclinical evaluation

Through laboratory based, the preclinical stage confirms the drug's potential in the novel indication. These consist of:

  • In vitro experiments: Drug testing on specific disease-relevant cell lines to evaluate cellular effects, cytotoxicity, and biomarker expression(24).
  • In vivo studies: Animal models are used to assess pharmacological activity, dosing, and disease outcomes(25).

These studies help determine whether the drug demonstrates sufficient efficacy in the new therapeutic context before progressing to human trials.

1.2.5 Clinical trials for the new indication

The medication moves on to clinical investigation if preclinical findings show promise. Phase I trials may be skipped in favor of Phase II or III trials, which concentrate on efficacy and the best dosage in the novel therapeutic area, because the safety of the medicine has already been demonstrated. These experiments could be carried out as

Investigator-initiated trials

  • Randomized controlled studies
  • Off-label use assessments (in some regulatory environments)

Trial design and ethical approvals are crucial in this stage to ensure robust and reproducible results(26).

1.2.6 Regulatory approval

Following successful clinical trials, a submission is made to the respective drug regulatory authority for approval of the drug’s new indication (27). Depending on the jurisdiction and nature of the change, a Supplemental New Drug Application (sNDA) or a formal request for label expansion is submitted(28). Regulatory agencies like the FDA, EMA, or CDSCO evaluate the submitted data to determine the drug's benefit-risk ratio in the new context (29).

1.2.7 Post-marketing surveillance

Once approved, the repurposed drug is monitored for real-world safety and effectiveness. Post-marketing surveillance involves:

  • Reporting and evaluation of adverse drug reactions (ADRs)
  • Collection of long-term safety data
  • Ongoing assessment of therapeutic outcomes

This stage ensures the continued safety and efficacy of the drug in the general population and may lead to further label updates or risk management plans (30,31).

Fig:01

1.3 Importance of Drug Reprofiling

Drug reprofiling plays an increasingly vital role in pharmaceutical innovation and global health strategy (32). Its importance stems from a combination of scientific, economic, and humanitarian benefits (33). Below are the key reasons why drug reprofiling has become a core component of modern drug development:

1.3.1 Time and cost efficiency

Traditional drug development is time-consuming and expensive (34). Drug reprofiling bypasses many early-stage steps—such as toxicology studies and Phase I clinical trials—because thesafety and pharmacokinetics of the drug are already known (35). This dramatically reduces development time and costs (36).

Fig:02

1.3.2 Lower risk of failure

Many drugs fail in clinical trials due to unforeseen toxicity or poor bioavailability. Reprofiled drugs, having already passed safety evaluations, present a lower risk during development and clinical testing for new indications (37).

1.3.3 Fast response to public health emergencies

In emergency situations, such as infectious disease outbreaks, reprofiling enables a rapid therapeutic response (38). This was evident during the COVID-19 pandemic, where several existing drugs were quickly tested and used under emergency authorizations (39).

1.3.4 Maximizes existing drug libraries

Pharmaceutical companies often have a portfolio of previously developed compounds that were never commercialized(40). Reprofiling helps revive these assets by identifying new therapeutic areas, thus improving return on investment and resource utilization(41).

1.2.5 Expands treatment for rare and neglected diseases

Reprofiling is particularly valuable for rare diseases and neglected tropical conditions where new drug development is not financially viable(42). It offers an accessible route to new therapies, improving global equity in healthcare(43).

1.2.6 Encourages interdisciplinary research and collaboration

The reprofiling process encourages collaboration between pharmacologists, clinicians, biochemists, and regulatory authorities (44). This multidisciplinary effort promotes innovation and knowledge sharing across different sectors of healthcare and research (45).

1.4 An Ideal Candidate for Drug Reprofiling

Choosing the best candidate is a crucial first stage in the drug reprofiling process that has a big impact on the overall outcome (46). A therapeutic candidate with a thorough safety and pharmacokinetic profile is the best candidate for reprofiling since it enables researchers to avoid numerous expensive and time-consuming early phases of drug development (47). Usually, a pool of previously approved medications, compounds that have been shelved, or medications that failed studies for reasons other than toxicity produces these candidates (48).

Key Characteristics of an Ideal Candidate

1.4.1 Established safety and tolerability

Drugs that have already been tested in humans—either in completed clinical trials or approved for other indications—are prime candidates. Their safety profile is well-documented, reducing the risk of adverse outcomes and regulatory hurdles in the new indication (49).

1.4.2 Known pharmacokinetic and pharmacodynamic properties

A deep understanding of the drug’s absorption, distribution, metabolism, excretion (ADME), and mechanism of action (MOA) makes it easier to predict its behavior in different disease conditions. This data also aids in determining dosage and treatment regimens for the new use(50).

1.4.3 Evidence of off-target or pleiotropic effects

Drugs that exhibit multiple mechanisms of action or off-target effects may offer therapeutic benefits beyond their original indication. These secondary effects, once considered side effects, can often serve as clues to potential new applications (51).

1.4.4 History of off-label use or clinical anecdotes

Drugs that have been used off-label or have anecdotal evidence suggesting benefits in other conditions are often strong reprofiling candidates. Such observations can serve as real-world indicators of therapeutic potential (52).

1.4.5 Previously shelved drugs (non-safety reasons)

Many compounds are discontinued during development due to strategic decisions, commercial limitations, or failure to meet efficacy endpoints for a specific disease. If safety is not a concern, these shelved drugs may be re-evaluated for other indications (53).

1.4.6 Chemical and formulation stability

Candidates with stable formulations, acceptable shelf-life, and scalable manufacturing processes are favourable, as these factors ease the transition into clinical testing for a new indication (54).

Examples of reprofiled drugs:Several well-known drugs began their journey with one intended use but were successfully repurposed for entirely different therapeutic areas:

  • Thalidomide: Initially used as a sedative, later reprofiled for multiple myeloma and erythema nodosum leprosum (55).
  • Sildenafil: Originally developed for angina, reprofiled for erectile dysfunction and pulmonary arterial hypertension (56).
  • Minoxidil: First approved as an antihypertensive, later repositioned for androgenic alopecia (57).

These success stories highlight how a deep understanding of drug biology, combined with clinical insight, can lead to effective reprofiling strategies.

1.5 Strategies for Drug Reprofiling

Drug reprofiling employs a diverse set of strategies to identify new therapeutic applications for existing drugs. These strategies range from observational and experimental approaches to systematic, molecular, and mechanism-based techniques. Choosing the right strategy depends on the type of drug, disease target, and available data (58).

The following are major strategies widely used in drug reprofiling:

      1. Phenotypic modelling

Using this method, medications that produce phenotypic alterations akin to those seen in illness states are found.

Thalidomide, for instance, was first sold as a sedative but was later used to treat multiple myeloma after researchers noticed that it had anti-angiogenic effects in phenotypic screening(59).

1.5.2. Target based methods

 Drugs that interact with particular molecular targets known to be involved in disease processes are identified using these techniques.

For instance, sildenafil (Viagra) was once created to treat hypertension and angina pectoris, but when researchers found that it specifically inhibited phosphodiesterase type 5 (PDE5), it was repurposed to treat erectile dysfunction (60).

1.5.3. Knowledge-based methods

 These make use of databases, literature, and current scientific knowledge to find possible new applications for already-approved medications. An illustration of this would be the repurposing of celecoxib, which was first used as an  nonsteroidal anti-inflammatory drug (NSAID) used to treat mild to moderate pain and help relieve symptoms of arthritis. Celecoxib is a selective cyclooxygenase-2 (COX-2) inhibitor. showed that combining celecoxib with paclitaxel might be an effective treatment for ovarian cancer (61).

1.5.4. Signature-based methods

These techniques look for possible therapeutic matches by comparing drug induced gene expression profiles with illness signatures. For instance, researchers utilized the Connectivity Map to determine that the anticonvulsant   topiramate's gene expression signature counteracted alterations in gene expression linked to alcohol addiction, leading to its repurposing for the treatment of alcohol dependency (62).

1.5.6. Pathway-based and network-based reprofiling

Focuses on medications that alter biological networks or cellular signalling pathways that are prevalent in many disorders.

  • Beneficial when illnesses have common pathway-level mechanisms as opposed to distinct molecular targets.
  • Network-based techniques investigate drug interactions in gene-regulatory or protein-protein interaction (PPI) networks.

Rapamycin (sirolimus), originally an immunosuppressant that targets mTOR, was repurposed for treating tuberous sclerosis complex (TSC) after researchers discovered that TSC is characterized by hyperactivation of the mTOR pathway (63).

1.5.7.Targeted mechanism-based reprofiling
This method compares the drug's pharmacological mechanism to the pathophysiology of an alternative illness.For instance, because neurodegenerative illnesses overlap inflammatory mechanisms, anti-inflammatory medications have been repurposed. An in-depth knowledge of pharmacology and pathophysiology is necessary for mechanism-based matching.

Example: Rapamycin (sirolimus), originally an immunosuppressant that targets mTOR, was repurposed for treating tuberous sclerosis complex (64).

1.5.8. Molecular docking and structural modelling

This computational approach predicts how drugs bind to target proteins, enabling identification of new drug-target interactions i.e it helps predict binding affinity and interaction mode between a drug and a protein involved in another disease.

Example: Raloxifene, originally approved for osteoporosis, was repurposed for breast cancer prevention after molecular docking studies revealed its binding affinity to oestrogen receptors in breast tissue (65).

2. Case Study And Applications

2.1 COVID-19

The COVID-19 pandemic triggered an urgent need for effective treatments (66). Developing new drugs from scratch would take years, so researchers turned to drug repurposing—identifying existing drugs with potential to treat SARS-CoV-2(67). This strategy allowed for quicker deployment due to pre-existing safety data. Several drugs, originally designed for other diseases, were evaluated globally, with mixed outcomes(68).

Table 1: Comparative Table of Repurposed Drugs for COVID-19

Drug

Original Use

Mechanism of Action

COVID-19 Use

Efficacy Summary

Approval Status

Remdesivir(69,70)

Ebola

Inhibits viral RNA polymerase

Moderate to severe cases

Shortens recovery time

FDA approved

Dexamethasone(71)

Inflammatory diseases

Corticosteroid; reduces inflammation

Severe cases with oxygen support

Reduces mortality

Globally recommended

Favipiravir(72)

Influenza

Inhibits RNA polymerase

Mild to moderate cases

Mixed results

Emergency use in some countries

Hydroxychloroquine(73)

Malaria, Lupus

Alters endosomal pH

Early-stage use

No proven benefit

Not recommended

Ivermectin(74)

Parasitic infections

Inhibits importin protein pathway

Mild cases (off-label)

Conflicting evidence

Not recommended

Tocilizumab(75)

Rheumatoid arthritis

IL-6 receptor antagonist

Severe cases with inflammation

Improves survival with steroids

Conditional approval

Baricitinib(76)

Rheumatoid arthritis

JAK inhibitor

Moderate to severe cases

Improves outcomes with Remdesivir

Emergency use authorized

2.2. Neurodegenerative Disorders

Neurodegenerative disorders such as Alzheimer's disease (AD), Parkinson’s disease (PD), amyotrophic lateral sclerosis (ALS), and Huntington’s disease (HD) are characterized by progressive neuronal dysfunction and lack effective curative treatments(77,78). Traditional drug development for these diseases faces high failure rates and long timelines. Repurposing existing drugs with known safety profiles can significantly reduce the cost and time of developing new therapies, especially for rare or complex CNS disorders(79,80,81).

Flowchart: Repurposing Pipeline for Neurodegenerative Diseases

FDA-Approved Drug Mechanistic Screening Disease Model Testing Phase I/II Trials in ND Patients Regulatory Repositioning(87)

Table 2: Comparative Table of Repurposed Drugs For ND

Drug

Original Use

ND Target

Mechanism

Memantine(82)

Alzheimer’s

PD, ALS

NMDA receptor antagonist

Riluzole(83)

ALS

AD, HD

Glutamate inhibition

Rasagiline(84)

Parkinson’s

AD

MAO-B inhibition

Metformin(85)

Type 2 Diabetes

AD, PD

AMPK activation

Nilotinib(86)

Leukemia

PD, AD

Tyrosine kinase inhibition

2.3cancer

Cancer remains a major global health challenge. The high cost and time-consuming nature of novel drug development make drug repurposing an attractive alternative. The traditional path of cancer drug development is lengthy, high-risk, and expensive(88).

2.3.1. Colon and Breast Cancer

Table 3 : Comparative Table of Repurposed Drugs in Colon and Breast

Drug

Original Use

Cancer Target

Mechanism

Metformin(89)

Type 2 Diabetes

Breast, Colon

AMPK activation, mTOR inhibition

Aspirin(90)

Analgesic/NSAID

Colon

COX inhibition, anti-inflammatory

Disulfiram(91)

Alcohol dependence

Breast, Colon

ROS generation, proteasome inhibition

Itraconazole(92)

Antifungal

Breast, Colon

Hedgehog pathway, angiogenesis inhibition

Propranolol(93)

Hypertension

Breast

Beta-adrenergic blockade, anti-angiogenesis

Drug Repurposing Strategy Flowchart

Drug with Known Safety → Preclinical Testing in Cancer Models → Mechanistic Studies → Clinical Trials (Phase I/II) → Potential New Indication(94)

2.4. Parasitic Infection

Parasitic infections such as malaria, leishmaniasis, trypanosomiasis, and schistosomiasis affect millions globally, particularly in tropical and subtropical regions (95). The traditional drug discovery pipeline is time-consuming and expensive, prompting interest in drug repurposing — using existing approved or investigational drugs for new therapeutic indications(96,97).

Table 4: Comparative Table of Repurposed Drugs For Parasitic Infection

Drug

Original Use

Repurposed For

Mechanism in Parasitic Infection

Status

Miltefosine(98)

Breast cancer

Leishmaniasis

Disrupts parasite cell membrane

Approved

Nitazoxanide(99)

Diarrhea (Cryptosporidium)

Giardiasis, Amebiasis

Inhibits PFOR enzyme-dependent electron transfer

Approved

Metronidazole(100)

Anaerobic bacterial infections

Amoebiasis, Giardiasis

DNA synthesis inhibition in anaerobic organisms

Approved

Eflornithine(101)

Cancer (ornithine decarboxylase inhibitor)

Trypanosomiasis

Inhibits polyamine biosynthesis

Approved (WHO)

Chloroquine(102)

Malaria

Amebiasis, Leishmaniasis

Interferes with heme detoxification in parasites

Repurposing trials

    1. . Auto Immune Diseases

Autoimmune diseases result from immune system dysregulation(103). Drug repurposing in this domain focuses on utilizing existing immunomodulatory or anti-inflammatory drugs developed for other indications(104).

Table 5: Comparative Table Of Repurposed Drugs For Autoimmune Diseases

Drug

Original use

Repurposed Autoimmune Use

Mechanism

Hydroxychloroquine(105)

Antimalarial

SLE, Rheumatoid Arthritis

TLR inhibition, reduces antigen presentation

Methotrexate(106)

Chemotherapy (cancer)

Psoriasis, Crohn’s Disease

Inhibits folate metabolism; immunosuppressive

Azathioprine(107)

Anticancer(leukemia)

Systemic sclerosis, Myositis

Purine synthesis inhibition, reduces T/B cells

Infliximab, Adalimumab(108)

Anti-TNF (Crohn’s)

RA, Psoriasis, Ankylosing Spondylitis

Neutralizes TNF-α

Tofacitinib, Baricitinib(109)

JAK inhibitors

RA, Psoriasis

Inhibits JAK/STAT pathway

2.6. Rare Diseases

Rare diseases, also known as orphan diseases, are conditions that affect a small percentage of the population(110). These diseases are often genetic in origin, chronic, and life-threatening, with limited treatment options available (111). Due to their low prevalence, rare diseases often face challenges in diagnosis, research, and drug development, making awareness and innovation in this field critically important(112).

Table 6: Comparative Table Of Repurposed Drugs For Rare Diseases

Drug

Original Use

Repurposed Rare Disease Use

Mechanism

Penicillamine(113)

Anti-leukemic

Wilson’s disease

Copper chelator

Valproic Acid(114)

Antiepileptic

Niemann–Pick Type C

HDAC inhibition, improves lysosomal function

Lisinopril(115)

Hypertension (ACE inhibitor)

Alport syndrome (renal disease)

Reduces glomerular pressure

Tacrolimus(116)

Immunosuppressant (transplants)

Myasthenia Gravis ,DMD

T-cell suppression

Azithromycin(117)

Antibiotic (macrolide)

Cystic fibrosis

Anti-inflammatory in airway

Desipramine(118)

Antidepressant

Rett syndrome

Enhances norepinephrine, possible neuroprotective effect

2.7. Infectious Diseases

Infectious diseases are disorders caused by microorganisms such as bacteria, viruses, fungi, or parasites. These diseases can spread directly or indirectly from one person to another, through contaminated food, water, air, or vectors like mosquitoes(1

Table 7: Comparative Table Of Repurposed Drugs For Infectious Diseases

Drug

Original Use

Repurposed Infectious Use

Mechanism

Chloroquine, Hydroxychloroquine(120)

Antimalarial

COVID-19 (initially), Viral infections

Endosomal pH modulation, antiviral entry blocker

Lopinavir-Ritonavir(121)

Antiviral (HIV protease inhibitor)

COVID-19, SARS

Viral protease inhibition

Ivermectin(122)

Antiparasitic (helminths)

COVID-19 (investigational)

Blocks viral nuclear transport proteins

Fluvoxamine(123)

Antidepressant (SSRI)

COVID-19

S1R agonist, reduces cytokine storm

Azithromycin(124)

Antibiotic (macrolide)

COVID-19 (with hydroxychloroquine)

Anti-inflammatory and possible antiviral

Indomethacin(125)

NSAID

Dengue, Zika (supportive)

Antiviral effect in vitro via prostaglandin inhibition

2.8. Cardiovascular Diseases

Cardiovascular diseases remain the leading cause of mortality globally(126). Drug reprofiling in cardiology aims to find new therapeutic benefits of existing drugs, particularly for heart failure, hypertension, arrhythmias, atherosclerosis, and pulmonary hypertension.It saves time and cost compared to de novo drug development(127,128).

Table 8 : Comparative Table Of Repurposed Drugs For CVD

Drug

Original indication

Repurposed for

Mechanism in CVD Context

Empagliflozin, Dapagliflozin(129)

Type 2 Diabetes

Heart Failure (HFrEF, HFpEF)

SGLT2 inhibitors; improve cardiac metabolism and reduce preload

Sildenafil, Tadalafil(130)

Erectile Dysfunction

Pulmonary Arterial Hypertension (PAH)

PDE5 inhibition → vasodilation of pulmonary vasculature

Methotrexate (low dose)(131)

Cancer (chemotherapy)

Atherosclerosis, Post-MI remodeling

Anti-inflammatory (IL-1β, CRP reduction)

Colchicine(132)

Immunosuppressant (RA)

Pericarditis, Post -MI inflammation

Inhibits neutrophil migration and inflammasome activation

Statins (e.g. Atorvastatin)(133)

Lipid Disorders

Heart Failure, Stroke

Beyond lipid lowering: plaque stabilization, anti-inflammatory

Fluoxetine, Sertraline(134)

Antidepressants(SSRIs)

Ischemic heart disease, stroke prevention

Platelet inhibition and mood stabilization in cardiac patients

Liraglutide, Semaglutide(135)

Antidiabetic (GLP-1 RA)

CVD risk reduction in diabetes

Improves endothelial function and weight reduction

Ranolazine(136)

Antianginal

Heart failure with preserved ejection fraction (HFpEE)

Late sodium current inhibition → improves myocardial relaxation

Clinical Benefits of Cardiovascular Drug Repurposing:

  • Cost-effectiveness: Avoids early-phase trials.
  • Reduced time to market.
  • Multifactorial benefit: Many CVDs have metabolic, inflammatory, and vascular components that benefit from multitarget drugs.

Enhanced compliance: Patients with comorbidities (e.g., diabetes + CVD) benefit from dual-action drugs like SGLT2 inhibitors(137).

Fig:03

2.9.Metabolic Disorders

Metabolic disorders are conditions that disrupt normal metabolism — the process your body uses to convert food into energy and building blocks for cells. These disorders can be inherited (genetic) or acquired due to lifestyle, disease, or medication(138).

Table 9: Comparative Table Of Repurposed Drugs For MD

Drug

Original Use

Repurposed Use

Mechanism

Metformin(139)

Type 2 Diabetes

PCOS, cancer, anti-aging

Activates AMPK, reduces hepatic glucose production

Thiazolidinediones (e.g., Pioglitazone)(140)

Type 2 Diabetes

NAFLD, PCOS

PPARγ agonism, improves insulin sensitivity

SGLT2 Inhibitors (e.g., Dapagliflozin)(141)

Diabetes

Heart failure, CKD

Promotes glucosuria, reduces cardiac stress

Statins(142)

Hyperlipidemia

Alzheimer's, NAFLD

Inhibits HMG-CoA reductase, anti-inflammatory

Colesevelam(143)

Hyperlipidemia

Type 2 Diabetes

Bile acid sequestration improves glycemic control

Topiramate(144)

Epilepsy

Obesity, binge eating disorder

Inhibits carbonic anhydrase, suppresses appetite

Hydroxychloroquine(145)

Malaria

Type 2 Diabetes, RA

Reduces inflammation, improves insulin sensitivity

Bromocriptine(146)

Parkinson’s Disease

Type 2 Diabetes

Dopamine D2 receptor agonist; resets circadian rhythm

Orlistat(147)

Obesity

Type 2 Diabetes

Inhibits lipase, reduces fat absorption

Fig:04

2.10.Drug Repurposing For Kidney Ailments

Drug reprofiling (or repurposing) for kidney ailments is a powerful strategy for finding new therapeutic uses for existing drugs in the treatment of kidney diseases. This approach is particularly valuable given the high unmet medical needs in chronic kidney disease (CKD), acute kidney injury (AKI), and other renal disorders. By leveraging drugs with established safety profiles, researchers can accelerate the treatment options for kidney patients(148,149,150).

Table 10: Comparative Table Of Repurposed Drugs For Kidney Ailments

Drug

Original Use

Repurposed Use in Kidney Disease

Mechanism of Action

Kidney Benefit

SGLT2 Inhibitors (e.g., Dapagliflozin, Empagliflozin)(151)

Type 2 Diabetes

CKD, Heart Failure

Inhibit SGLT2 → glucosuria, ↓ glomerular pressure

Slows CKD progression, cardio-renal protection

Epoetin, Darbepoetin(152)

Anemia in cancer

Anemia in CKD

Stimulates erythropoiesis (RBC production)

Treats CKD-associated anemia

ACE Inhibitors (e.g., Ramipril)(153)

Hypertension, Heart Failure

CKD, Proteinuria

Inhibits RAAS → ↓ BP and glomerular pressure

Reduces proteinuria, protects renal function

Statins (e.g., Atorvastatin)(154)

Hyperlipidemia

CKD, Proteinuria

Inhibits HMG-CoA reductase → ↓ cholesterol, anti-inflammatory

↓ Cardiovascular risk, antiproteinuric effect

Spironolactone, Eplerenone(155)

Hypertension, Heart Failure

CKD, Proteinuria

Aldosterone antagonism → ↓ sodium retention, fibrosis

Reduces proteinuria, glomerular fibrosis

Colchicine(156)

Gout, FMF

CKD (fibrosis)

Inhibits inflammasome → anti-fibrotic

↓ Kidney fibrosis and inflammation

N-acetylcysteine (NAC)(157)

Acetaminophen overdose

AKI, Contrast-Induced Nephropathy

Antioxidant, scavenges ROS

Prevents/treats AKI, especially post-contrast

Sirolimus (Rapamycin)(158)

Organ transplant immunosuppressant

Alport Syndrome, FSGS

mTOR inhibition → anti-proliferative, anti-fibrotic

↓ Proteinuria, slows glomerular scarring

Pentoxifylline(159)

Peripheral vascular disease

Diabetic Nephropathy

Improves blood flow, anti-inflammatory

↓ Proteinuria, improves kidney function

Lithium(160)

Bipolar disorder

Renal regeneration (experimental)

Inhibits GSK-3 → cell regeneration

Promotes renal repair (preclinical stage)

2.11.HMG CoA – Reductase Inhibitor

Table 11: Comparative Table Of Repurposed HMG CoA – Reductase Inhibitors

Drug Name

Original Indication

Repurposed Use

Mechanism Behind Reprofiling

Status

Atorvastatin(161)

Hyperlipidemia

Alzheimer's disease, Multiple sclerosis

Anti-inflammatory, improves cerebral blood flow

Clinical trials, observational

Simvastatin(162)

Hyperlipidemia

Rheumatoid arthritis, Osteoporosis

Reduces inflammation, affects bone metabolism

Investigational

Lovastatin(163)

Hyperlipidemia

Cancer (breast, prostate, glioma)

Inhibits tumor cell proliferation via mevalonate pathway suppression

Preclinical to early trials

Rosuvastatin(164)

Hyperlipidemia

Pulmonary hypertension

Improves endothelial function, reduces vascular remodeling

Investigational

CONCLUSION

Drug reprofiling has emerged as a key tactic in modern medicine, transforming the way existing drugs are used for new purposes.   By avoiding early-stage safety evaluations, it significantly reduces time, cost, and risk when compared to traditional drug development.   Its use spans a wide range of illnesses, from cancer and neurodegenerative conditions to rare and infectious diseases, indicating its adaptability and impact on global health.   The integration of computational, observational, and experimental approaches has expanded its scope, improving treatment accessibility for underserved populations and enabling faster responses to public health emergencies. Drug reprofiling is expected to remain a crucial element of pharmaceutical innovation as interdisciplinary collaboration increases and regulatory pathways shift, maximising available resources and driving worldwide advances in precision and personalised medicine.

ACKNOWLEGEMENT

We, students of 7th semester B. Pharm, take the privilege to acknowledge to all those who helped me in the project work entitled as “Drug Reprofiling in the Age of Necessity: A Strategic-Clinical Synthesis for Therapeutic Clinical Outcome”. At first, we express our deep sense of gratitude indebtedness to the Department of Pharmacology of Mar Dioscorus College of Pharmacy, for helping us in the completion of our practice school. We are extremely thankful to DR. PREEJA. G. PILLAI, our principal, for her guidance and support throughout the work. We owe our profound recognition to DR. SINCHU YESUDANAM, our guide for enlightening us with knowledge, valuable suggestions and for her remarkable encouragement which made us complete our work. We are also grateful to DR. JOYAMMA VARKEY for providing us with valuable education throughout our practice school. We express our sincere thanks to one and all that accompanied and helped throughout our educational career.

REFERENCES

  1. Ashburn TT, Thor KB. Drug repositioning: identifying and developing new uses for existing drugs. Nat Rev Drug Discov. 2004;3(8):673-83.
  2. Pushpakom S, Iorio F, Eyers PA, et al. Drug repurposing: progress, challenges and recommendations. Nat Rev Drug Discov. 2019;18(1):41-58.
  3. Nosengo N. Can you teach old drugs new tricks? Nature. 2016;534(7607):314-6.
  4. Li J, Zheng S, Chen B, et al. A survey of current trends in computational drug repositioning. Brief Bioinform. 2016;17(1):2-12.
  5. Sleigh SH, Barton CL. Repurposing strategies for therapeutics. Pharm Med. 2010;24(3):151-9.
  6. Chong CR, Sullivan DJ Jr. New uses for old drugs. Nature. 2007;448(7154):645-6.
  7. Langedijk J, Mantel-Teeuwisse AK, Slijkerman DS, Schutjens MH. Drug repositioning and repurposing: terminology and definitions in literature. Drug Discov Today. 2015;20(8):1027-34.
  8. Oprea TI, Bauman JE, Bologa CG, et al. Drug repurposing from an academic perspective. Drug Discov Today Ther Strateg. 2011;8(3-4):61-9.
  9. Vamathevan J, Clark D, Czodrowski P, et al. Applications of machine learning in drug discovery and development. Nat Rev Drug Discov. 2019;18(6):463-77.
  10. Sahu KK, Mishra AK. Repositioning of drugs in COVID-19: a review. J Biomol Struct Dyn. 2021;39(7):2679-92.
  11. Breckenridge A, Jacob R. Overcoming the legal and regulatory barriers to drug repurposing. Nat Rev Drug Discov. 2019;18(1):1-2.
  12. Mak K-K, Pichika MR. Artificial intelligence in drug development: present status and future prospects. Drug Discov Today. 2019;24(3):773-80.
  13. Cavalla D. Predictive methods in drug repositioning. In: Barratt MJ, Frail DE, editors. Drug Repositioning: Bringing New Life to Shelved Assets and Existing Drugs. Hoboken (NJ): Wiley; 2012. p. 153–76.
  14. Ashburn TT, Thor KB. Nat Rev Drug Discov. 2004;3(8):673-683.
  15. Li J, Zheng S, Chen B, et al. Brief Bioinform. 2016;17(1):2-12.
  16. Pushpakom S, Iorio F, Eyers PA, et al. Nat Rev Drug Discov. 2019;18(1):41-58.
  17. Sleigh SH, Barton CL. Pharm Med. 2010;24(3):151-159.
  18. Chong CR, Sullivan DJ Jr. Nature. 2007;448(7154):645-646.
  19. Langedijk J, Mantel-Teeuwisse AK, Slijkerman DS, Schutjens MH. Drug Discov Today. 2015;20(8):1027-1034.
  20. Nosengo N. Nature. 2016;534(7607):314-316.
  21. Vohora D, Singh G. Pharmaceutical Medicine and Translational Clinical Research. Academic Press; 2018.
  22. Oprea TI, Bauman JE, Bologa CG, et al. Drug Discov Today Ther Strateg. 2011;8(3-4):61-69.
  23. Sahoo BM, Ravi Kumar BVV, Mahapatra MK, et al. Comput Biol Med. 2021;137:104851.
  24. Eder J, Sedrani R, Wiesmann C. Nat Rev Drug Discov. 2014;13(8):577-587.
  25. Garcia-Serna R, Vidal D, Remez N, Mestres J. Trends Pharmacol Sci. 2015;36(10):637-645.
  26. FDA. Guidance for Industry: Drug Development and Review Definitions. 2020.
  27. EMA. Guideline on Clinical Trials in Small Populations. 2006.
  28. CDSCO. Regulatory Pathways for Drug Repurposing in India. 2021.
  29. Breckenridge A, Jacob R. Nat Rev Drug Discov. 2019;18(1):1-2.
  30. FDA. FDA Adverse Event Reporting System (FAERS). 2023.
  31. EMA. Good Pharmacovigilance Practices (GVP). 2022.
  32. Ashburn TT, Thor KB. Nat Rev Drug Discov. 2004;3(8):673-83.
  33. Pushpakom S, Iorio F, Eyers PA, et al. Nat Rev Drug Discov. 2019;18(1):41-58.
  34. DiMasi JA, Grabowski HG, Hansen RW. J Health Econ. 2016;47:20-33.
  35. Kola I, Landis J. Nat Rev Drug Discov. 2004;3(8):711-6.
  36. Langedijk J, Mantel-Teeuwisse AK, Slijkerman DS, Schutjens MH. Drug Discov Today. 2015;20(8):1027-34.
  37. Nosengo N. Nature. 2016;534(7607):314-6.
  38. Zhou Y, Hou Y, Shen J, et al. Cell Discov. 2020;6:14.
  39. Beigel JH, Tomashek KM, Dodd LE, et al. N Engl J Med. 2020;383(19):1813-26.
  40. Breckenridge A, Jacob R. Nat Rev Drug Discov. 2019;18(1):1-2.
  41. Jin G, Wong ST. Drug Discov Today. 2014;19(5):637-44.
  42. Sahoo BM, Ravi Kumar BVV, Mahapatra MK, et al. Comput Biol Med. 2021;137:104851.
  43. Sleigh SH, Barton CL. Pharm Med. 2010;24(3):151-9.
  44. Vamathevan J, Clark D, Czodrowski P, et al. Nat Rev Drug Discov. 2019;18(6):463-77.
  45. Oprea TI, Bauman JE, Bologa CG, et al. Drug Discov Today Ther Strateg. 2011;8(3-4):61-9.
  46. Ashburn TT, Thor KB. Nat Rev Drug Discov. 2004;3(8):673-83.
  47. Nosengo N. Nature. 2016;534(7607):314-6.
  48. Li J, Zheng S, Chen B, et al. Brief Bioinform. 2016;17(1):2-12.
  49. Pushpakom S, Iorio F, Eyers PA, et al. Nat Rev Drug Discov. 2019;18(1):41-58.
  50. Jin G, Wong ST. Drug Discov Today. 2014;19(5):637-44.
  51. Keiser MJ, Setola V, Irwin JJ, et al. Nature. 2009;462(7270):175-81.
  52. Langedijk J, Mantel-Teeuwisse AK, Slijkerman DS, Schutjens MH. Drug Discov Today. 2015;20(8):1027-34.
  53. Chong CR, Sullivan DJ Jr. Nature. 2007;448(7154):645-6.
  54. Cavalla D. Drug Repositioning: Bringing New Life to Shelved Assets and Existing Drugs. Wiley; 2012.
  55. Franks ME, Macpherson GR, Figg WD. Lancet Oncol. 2004;5(7):375-82.
  56. Boolell M, Allen MJ, Ballard SA, et al. Int J Impot Res. 1996;8(2):47-52.
  57. Messenger AG, Rundegren J. Dermatol Clin. 2007;25(4):587-98.
  58. Li J, Zheng S, Chen B, et al. Brief Bioinform. 2016;17(1):2-12.
  59. Franks ME, Macpherson GR, Figg WD. Lancet Oncol. 2004;5(7):375-82.
  60. Boolell M, Allen MJ, Ballard SA, et al. Int J Impot Res. 1996;8(2):47-52.
  61. Ali K, Middleton MR, Garcia M, et al. Celecoxib as an adjuvant in paclitaxel chemotherapy. J Clin Oncol. 2005;23(16_suppl):9614.
  62. Jia P, Wang L, Fan Y, et al. BMC Bioinformatics. 2013;14(Suppl 16):S3.
  63. Franz DN, Leonard J, Tudor C, et al. N Engl J Med. 2006;354(1):18-28.
  64. Johnson SC, Rabinovitch PS, Kaeberlein M. Nat Rev Drug Discov. 2013;12(5):377-88.
  65. Martinkovich S, Shah D, Planey SL, Arnott JA. Steroids. 2014;90:13-25.
  66.  Li G, De Clercq E. Therapeutic options for the 2019 novel coronavirus (2019-nCoV). Nat Rev Drug Discov. 2020 Mar;19(3):149–150.
  67.  Pushpakom S, Iorio F, Eyers PA, et al. Drug repurposing: progress, challenges and recommendations. Nat Rev Drug Discov. 2019 Jan;18(1):41–58.
  68.  Zhou Y, Wang F, Tang J, Nussinov R, Cheng F. Artificial intelligence in COVID-19 drug repurposing. The Lancet Digital Health. 2020 Aug;2(8):e395–e404.
  69.  Harrison C. Coronavirus puts drug repurposing on the fast track. Nat Biotechnol. 2020 Apr;38(4):379–381.
  70.  Singh TU, Parida S, Lingaraju MC, Kesavan M, Kumar D, Singh RK. Drug repurposing approach to fight COVID-19. Pharmacol Rep. 2020 Aug;72(6):1479–1508.
  71.  Beigel JH, Tomashek KM, Dodd LE, et al. Remdesivir for the Treatment of Covid-19 — Final Report. N Engl J Med. 2020 Nov;383(19):1813–1826.
  72.  FDA. Remdesivir EUA Letter of Authorization. [Internet]. 2020 [cited 2025 Aug 7]. Available from: https://www.fda.gov/media/137564/download
  73.  RECOVERY Collaborative Group. Dexamethasone in Hospitalized Patients with Covid-19. N Engl J Med. 2021 Feb;384(8):693–704.
  74.  Chen C, Zhang Y, Huang J, et al. Favipiravir versus Arbidol for COVID-19: a randomized clinical trial. medRxiv. 2020; doi:10.1101/2020.03.17.20037432.
  75.  WHO. “Solidarity” clinical trial for COVID-19 treatments. [Internet]. 2021 [cited 2025 Aug 7]. Available from: https://www.who.int/emergencies/diseases/novel-coronavirus-2019/global-research-on-novel-coronavirus-2019-ncov/solidarity-clinical-trial-for-covid-19-treatments
  76.  Bryant A, Lawrie TA, Dowswell T, et al. Ivermectin for prevention and treatment of COVID?19 infection: a systematic review, meta?analysis, and trial sequential analysis to inform clinical guidelines. Am J Ther. 2021;28(4):e434e460.
  77.  Dugger BN, Dickson DW. Pathology of neurodegenerative diseases. Cold Spring Harb Perspect Biol. 2017;9(7):a028035.
  78.  Cummings J, Lee G, Zhong K, Fonseca J, Taghva K. Alzheimer's disease drug development pipeline: 2021. Alzheimers Dement (N Y). 2021;7(1):e12179.
  79.  Mullard A. Failures overshadowing CNS drug pipeline. Nat Rev Drug Discov. 2010;9(10):693–694.
  80. Pushpakom S, Iorio F, Eyers PA, et al. Drug repurposing: progress, challenges and recommendations. Nat Rev Drug Discov. 2019;18(1):41–58.
  81. Ashburn TT, Thor KB. Drug repositioning: identifying and developing new uses for existing drugs. Nat Rev Drug Discov. 2004;3(8):673–683.
  82. Lipton SA. Paradigm shift in neuroprotection by NMDA receptor blockade: memantine and beyond. Nat Rev Drug Discov. 2006;5(2):160–170.
  83. Cheah BC, Vucic S, Krishnan AV, et al. Riluzole, neuroprotection and amyotrophic lateral sclerosis. Curr Med Chem. 2010;17(18):1942–1199.
  84. Weinreb O, Amit T, Bar-Am O, Youdim MBH. Rasagiline: a novel anti-Parkinsonian monoamine oxidase-B inhibitor with neuroprotective activity. Prog Neurobiol. 2010;92(3):330–344.
  85. Rotermund C, Machetanz G, Fitzgerald JC. The therapeutic potential of metformin in neurodegenerative diseases. Front Endocrinol (Lausanne). 2018;9:400.
  86. Pagan FL, Hebron ML, Wilmarth B, et al. Nilotinib effects in Parkinson’s disease and dementia with Lewy bodies. J Parkinsons Dis. 2016;6(3):503–517.
  87. Cavalla D. Predictive value of preclinical models in CNS drug development. Drug Discov Today. 2019;24(1):30–36.
  88. DiMasi JA, Grabowski HG, Hansen RW. Innovation in the pharmaceutical industry: new estimates of R&D costs. J Health Econ. 2016;47:20–33.
  89. Dowling RJ, Goodwin PJ, Stambolic V. Understanding the benefit of metformin use in cancer treatment. BMC Med. 2011;9:33.
  90. Chan AT, Giovannucci EL, Meyerhardt JA, et al. Long-term use of aspirin and nonsteroidal anti-inflammatory drugs and risk of colorectal cancer. JAMA. 2005;294(8):914–923.
  91. Skrott Z, Mistrik M, Andersen KK, et al. Alcohol-abuse drug disulfiram targets cancer via p97 segregase adaptor NPL4. Nature. 2017;552(7684):194–199.
  92. Kim J, Tang JY, Gong R, et al. Itraconazole, a commonly used antifungal that inhibits Hedgehog pathway activity and cancer growth. Cancer Cell. 2010;17(4):388–399.
  93. Montoya A, Amaya CN, Belmont A, et al. Use of non-selective β-blockers is associated with decreased metastasis and improved recurrence-free survival in breast cancer. Oncotarget. 2017;8(41):70684–70693.
  94. Pantziarka P, Bouche G, Meheus L, Sukhatme V, Sukhatme VP, Vikas P. The repurposing drugs in oncology (ReDO) project. Ecancermedicalscience. 2014;8:442.
  95. Hotez PJ, Kamath A. Neglected tropical diseases in sub-Saharan Africa: review of their prevalence, distribution, and disease burden. PLoS Negl Trop Dis. 2009;3(8):e412.
  96. Chatelain E. Chagas disease drug discovery: toward a new era. J Biomol Screen. 2015;20(1):22–35.
  97. Ashburn TT, Thor KB. Drug repositioning: identifying and developing new uses for existing drugs. Nat Rev Drug Discov. 2004;3(8):673–683.
  98. Sundar S, Jha TK, Thakur CP, et al. Oral miltefosine for Indian visceral leishmaniasis. N Engl J Med. 2002;347(22):1739–1746.
  99. Müller J, Hemphill A. Effects of nitazoxanide on Giardia lamblia: a mini review. Parasitol Res. 2013;112(10):3537–3542.
  100. Upcroft JA, Upcroft P. Drug targets and mechanisms of resistance in the anaerobic protozoa. Clin Microbiol Rev. 2001;14(1):150–164.
  101. Barrett MP, Vincent IM, Burchmore RJ, et al. Drug resistance in kinetoplastid protozoa: mechanisms, mutations and diagnosis. Nat Rev Microbiol. 2011;9(6):430–440.
  102. Vale N, Moreira R, Gomes P. Primaquine revisited six decades after its discovery. Eur J Med Chem. 2009;44(3):937–953.
  103. Davidson A, Diamond B. Autoimmune diseases. N Engl J Med. 2001;345(5):340–350.
  104. Zhang Y, He D, Li M, et al. Drug repurposing for autoimmune diseases: a literature-based review. Front Pharmacol. 2021;12:654428.
  105. Schrezenmeier E, Dörner T. Mechanisms of action of hydroxychloroquine and chloroquine: implications for rheumatology. Nat Rev Rheumatol. 2020;16(3):155–166.
  106. Visser K, van der Heijde D. Risk and management of liver toxicity during methotrexate treatment in rheumatoid and psoriatic arthritis. Clin Exp Rheumatol. 2009;27(6):1017–1025.
  107. Rosenblum H, Amital H. Anti-TNF therapy: safety aspects of taking the risk. Autoimmun Rev. 2011;10(9):563–568.
  108. Feldmann M, Maini RN. Anti-TNF alpha therapy of rheumatoid arthritis: what have we learned? Annu Rev Immunol. 2001;19:163–196.
  109. Schwartz DM, Kanno Y, Villarino A, et al. JAK inhibition as a therapeutic strategy for immune and inflammatory diseases. Nat Rev Drug Discov. 2017;16(12):843–862.
  110. Institute of Medicine (US) Committee on Accelerating Rare Diseases Research and Orphan Product Development. Rare Diseases and Orphan Products: Accelerating Research and Development. Washington (DC): National Academies Press (US); 2010.
  111. Ferreira CR. The burden of rare diseases. Am J Med Genet A. 2019;179(6):885–892.
  112. Austin CP, Cutillo CM, Lau LPL, et al. Future of rare diseases research 2017–2027: an IRDiRC perspective. Clin Transl Sci. 2018;11(1):21–27.
  113. Walshe JM. Penicillamine, a new oral therapy for Wilson’s disease. Am J Med. 1956;21(4):487–495.
  114. Pipalia NH, Cosner CC, Huang A, et al. Histone deacetylase inhibitor treatment dramatically reduces cholesterol accumulation in Niemann–Pick type C1 mutant human fibroblasts. Proc Natl Acad Sci USA. 2011;108(14):5620–5625.
  115. Gross O, Licht C, Anders HJ, et al. Early angiotensin-converting enzyme inhibition in Alport syndrome delays renal failure and improves life expectancy. Kidney Int. 2012;81(5):494–501.
  116. Mantegazza R, Antozzi C. When myasthenia gravis is deemed refractory: clinical signposts and treatment strategies. Ther Adv Neurol Disord. 2018;11:1756285617749134.
  117. Saiman L, Anstead M, Mayer-Hamblett N, et al. Effect of azithromycin on pulmonary function in patients with cystic fibrosis uninfected with Pseudomonas aeruginosa: a randomized controlled trial. JAMA. 2010;303(17):1707–1715.
  118. Kishi N, Macklis JD. MECP2 is progressively expressed in post-migratory neurons and is involved in neuronal maturation rather than cell fate decisions. Mol Cell Neurosci. 2004;27(3):306–321.
  119. Morens DM, Fauci AS. Emerging pandemic diseases: how we got to COVID-19. Cell. 2020;182(5):1077–1092.
  120. WHO Solidarity Trial Consortium. Repurposed antiviral drugs for COVID-19 — interim WHO Solidarity trial results. N Engl J Med. 2021;384(6):497–511.
  121. Cao B, Wang Y, Wen D, et al. A trial of lopinavir–ritonavir in adults hospitalized with severe Covid-19. N Engl J Med. 2020;382(19):1787–1799.
  122. Caly L, Druce JD, Catton MG, et al. The FDA-approved drug ivermectin inhibits the replication of SARS-CoV-2 in vitro. Antiviral Res. 2020;178:104787.
  123. Lenze EJ, Mattar C, Zorumski CF, et al. Fluvoxamine vs placebo and clinical deterioration in outpatients with symptomatic COVID-19: a randomized clinical trial. JAMA. 2020;324(22):2292–2300.
  124. Gautret P, Lagier JC, Parola P, et al. Hydroxychloroquine and azithromycin as a treatment of COVID-19: results of an open-label non-randomized clinical trial. Int J Antimicrob Agents. 2020;56(1):105949.
  125. Amici C, Di Caro A, Ciucci A, et al. Indomethacin has a potent antiviral activity against SARS coronavirus. Antivir Ther. 2006;11(8):1021–1030.
  126. Roth GA, Mensah GA, Johnson CO, et al. Global burden of cardiovascular diseases and risk factors, 1990–2019: update from the GBD 2019 study. J Am Coll Cardiol. 2020;76(25):2982–3021.
  127. Benjamin EJ, Muntner P, Alonso A, et al. Heart disease and stroke statistics—2019 update: a report from the American Heart Association. Circulation. 2019;139(10):e56–e528.
  128. Pushpakom S, Iorio F, Eyers PA, et al. Drug repurposing: progress, challenges and recommendations. Nat Rev Drug Discov. 2019;18(1):41–58.
  129. McMurray JJV, Solomon SD, Inzucchi SE, et al. Dapagliflozin in patients with heart failure and reduced ejection fraction. N Engl J Med. 2019;381(21):1995–2008.
  130. Galie N, Ghofrani HA, Torbicki A, et al. Sildenafil citrate therapy for pulmonary arterial hypertension. N Engl J Med. 2005;353(20):2148–2157.
  131. Ridker PM, Everett BM, Thuren T, et al. Antiinflammatory therapy with canakinumab for atherosclerotic disease. N Engl J Med. 2017;377(12):1119–1131.
  132. Tardif JC, Kouz S, Waters DD, et al. Efficacy and safety of low-dose colchicine after myocardial infarction. N Engl J Med. 2019;381(26):2497–2505.
  133. Davignon J. Beneficial cardiovascular pleiotropic effects of statins. Circulation. 2004;109(23 Suppl 1):III39–III43.
  134. Celano CM, Huffman JC. Depression and cardiac disease: a review. Cardiol Rev. 2011;19(3):130–142.
  135. Marso SP, Daniels GH, Brown-Frandsen K, et al. Liraglutide and cardiovascular outcomes in type 2 diabetes. N Engl J Med. 2016;375(4):311–322.
  136. Gheorghiade M, Konstam MA, Burnett JC, et al. Short-term clinical effects of ranolazine in patients with acute heart failure syndromes: the RALI-DHF trial. J Am Coll Cardiol. 2011;58(7):747–755.
  137. Feher M. The value of drug repurposing in the cardiovascular therapeutic area. Expert Rev Cardiovasc Ther. 2021;19(2):81–92.
  138. Saudubray JM, Baumgartner MR, Walter JH. Inborn Metabolic Diseases: Diagnosis and Treatment. 6th ed. Springer; 2016.
  139. Rena G, Hardie DG, Pearson ER. The mechanisms of action of metformin. Diabetologia. 2017;60(9):1577–1585.
  140. Heerspink HJL, Stefánsson BV, Correa-Rotter R, et al. Dapagliflozin in patients with chronic kidney disease. N Engl J Med. 2020;383(15):1436–1446.
  141. Sahebkar A, Serban C, Mikhailidis DP, et al. Statin therapy reduces liver aminotransferases in patients with non-alcoholic fatty liver disease: a systematic review and meta-analysis of controlled clinical trials. J Clin Lipidol. 2016;10(2):519–533.
  142. Fonseca VA, Handelsman Y, Staels B. Colesevelam hydrochloride: a non-systemic lipid-altering drug. Cardiovasc Drugs Ther. 2010;24(1):5–11.
  143. Bray GA, Ryan DH. Update on obesity pharmacotherapy. Ann N Y Acad Sci. 2014;1311:1–13.
  144. Rekedal LR, Massarotti E, Garg R, et al. Changes in glycosylated hemoglobin after initiation of hydroxychloroquine or methotrexate treatment in diabetes patients with rheumatic diseases. Arthritis Rheum. 2010;62(12):3569–3573.
  145. Pijl H, Ohashi S, Matsuda M, et al. Bromocriptine: a novel approach to the treatment of type 2 diabetes. Diabetes Care. 2000;23(8):1154–1161.
  146. Torgerson JS, Hauptman J, Boldrin MN, et al. XENical in the prevention of diabetes in obese subjects (XENDOS) study: a randomized study. Diabetes Care. 2004;27(1):155–161.
  147. Henry RR, Smith SR, Schwartz SL, et al. Effects of weight loss with orlistat on glucose tolerance and progression to type 2 diabetes in obese patients with impaired glucose tolerance. Diabetes Care. 2001;24(1):133–140.
  148. Jha V, Garcia-Garcia G, Iseki K, et al. Chronic kidney disease: global dimension and perspectives. Lancet. 2013;382(9888):260–272.
  149. Himmelfarb J, Tuttle K. Challenges in the design of clinical trials for chronic kidney disease: lessons learned and future directions. Kidney Int. 2014;86(5):894–903.
  150. Pushpakom S, Iorio F, Eyers PA, et al. Drug repurposing: progress, challenges and recommendations. Nat Rev Drug Discov. 2019;18(1):41–58.
  151. Wheeler DC, Stefánsson BV, Jongs N, et al. Effects of dapagliflozin on major adverse kidney and cardiovascular events in patients with diabetic and non-diabetic chronic kidney disease: a prespecified analysis from the DAPA-CKD trial. Lancet Diabetes Endocrinol. 2021;9(1):22–31.
  152. Locatelli F, Bárány P, Covic A, et al. Kidney Disease: Improving Global Outcomes guidelines on anaemia management in chronic kidney disease: a European Renal Best Practice position statement. Nephrol Dial Transplant. 2013;28(6):1346–1359.
  153. Lewis EJ, Hunsicker LG, Clarke WR, et al. Renoprotective effect of the angiotensin-receptor antagonist irbesartan in patients with nephropathy due to type 2 diabetes. N Engl J Med. 2001;345(12):851–860.
  154. Tonelli M, Isles C, Curhan GC, et al. Effect of statins on kidney function, proteinuria, and progression of chronic kidney disease: a meta-analysis of randomized trials. BMJ. 2011;343:d1895.
  155. Bomback AS, Klemmer PJ. The incidence and implications of aldosterone breakthrough. Nat Clin Pract Nephrol. 2007;3(9):486–492.
  156. Yilmaz MI, Romano M, Basarali MK, et al. The potential role of colchicine in reducing inflammation and proteinuria in chronic kidney disease. Kidney Blood Press Res. 2021;46(2):149–158.
  157. Tepel M, van der Giet M, Schwarzfeld C, et al. Prevention of radiographic-contrast-agent-induced reductions in renal function by acetylcysteine. N Engl J Med. 2000;343(3):180–184.
  158. Song X, Xue X, Wang Y, et al. Sirolimus as a therapeutic option in focal segmental glomerulosclerosis and Alport syndrome: an experimental review. Nephrol Dial Transplant. 2021;36(2):301–308.
  159. Navarro-González JF, Mora-Fernández C, Muros M, et al. Pentoxifylline for renal protection in diabetic nephropathy: a systematic review and meta-analysis. PLoS One. 2013;8(9):e74431.
  160. Hoorn EJ, Walsh SB, McCormick JA, et al. The calcineurin inhibitor tacrolimus activates the renal sodium chloride cotransporter to cause hypertension. Nat Med. 2011;17(10):1304–1309.
  161. Sparks DL, Sabbagh MN, Connor DJ, et al. Atorvastatin for the treatment of mild to moderate Alzheimer disease: preliminary results. Arch Neurol. 2005;62(5):753–757.
  162. Panonnummal R,Varkey J. Anti-inflammatory activity of HMG CoA reductase inhibitors: a comparative study. Int J Pharm Pharm Sci. 2015;7(3):468-9.
  163. Jiang P, Mukthavaram R, Chao Y, et al. In vitro and in vivo anticancer effects of mevalonate pathway inhibition by lovastatin in glioblastoma. Cancer Lett. 2014;349(2):161–170.
  164. Wilkins MR, Ali O, Bradlow W, et al. Simvastatin as a treatment for pulmonary hypertension trial. Am J Respir Crit Care Med. 2010;181(10):1106–1113.

Reference

  1. Ashburn TT, Thor KB. Drug repositioning: identifying and developing new uses for existing drugs. Nat Rev Drug Discov. 2004;3(8):673-83.
  2. Pushpakom S, Iorio F, Eyers PA, et al. Drug repurposing: progress, challenges and recommendations. Nat Rev Drug Discov. 2019;18(1):41-58.
  3. Nosengo N. Can you teach old drugs new tricks? Nature. 2016;534(7607):314-6.
  4. Li J, Zheng S, Chen B, et al. A survey of current trends in computational drug repositioning. Brief Bioinform. 2016;17(1):2-12.
  5. Sleigh SH, Barton CL. Repurposing strategies for therapeutics. Pharm Med. 2010;24(3):151-9.
  6. Chong CR, Sullivan DJ Jr. New uses for old drugs. Nature. 2007;448(7154):645-6.
  7. Langedijk J, Mantel-Teeuwisse AK, Slijkerman DS, Schutjens MH. Drug repositioning and repurposing: terminology and definitions in literature. Drug Discov Today. 2015;20(8):1027-34.
  8. Oprea TI, Bauman JE, Bologa CG, et al. Drug repurposing from an academic perspective. Drug Discov Today Ther Strateg. 2011;8(3-4):61-9.
  9. Vamathevan J, Clark D, Czodrowski P, et al. Applications of machine learning in drug discovery and development. Nat Rev Drug Discov. 2019;18(6):463-77.
  10. Sahu KK, Mishra AK. Repositioning of drugs in COVID-19: a review. J Biomol Struct Dyn. 2021;39(7):2679-92.
  11. Breckenridge A, Jacob R. Overcoming the legal and regulatory barriers to drug repurposing. Nat Rev Drug Discov. 2019;18(1):1-2.
  12. Mak K-K, Pichika MR. Artificial intelligence in drug development: present status and future prospects. Drug Discov Today. 2019;24(3):773-80.
  13. Cavalla D. Predictive methods in drug repositioning. In: Barratt MJ, Frail DE, editors. Drug Repositioning: Bringing New Life to Shelved Assets and Existing Drugs. Hoboken (NJ): Wiley; 2012. p. 153–76.
  14. Ashburn TT, Thor KB. Nat Rev Drug Discov. 2004;3(8):673-683.
  15. Li J, Zheng S, Chen B, et al. Brief Bioinform. 2016;17(1):2-12.
  16. Pushpakom S, Iorio F, Eyers PA, et al. Nat Rev Drug Discov. 2019;18(1):41-58.
  17. Sleigh SH, Barton CL. Pharm Med. 2010;24(3):151-159.
  18. Chong CR, Sullivan DJ Jr. Nature. 2007;448(7154):645-646.
  19. Langedijk J, Mantel-Teeuwisse AK, Slijkerman DS, Schutjens MH. Drug Discov Today. 2015;20(8):1027-1034.
  20. Nosengo N. Nature. 2016;534(7607):314-316.
  21. Vohora D, Singh G. Pharmaceutical Medicine and Translational Clinical Research. Academic Press; 2018.
  22. Oprea TI, Bauman JE, Bologa CG, et al. Drug Discov Today Ther Strateg. 2011;8(3-4):61-69.
  23. Sahoo BM, Ravi Kumar BVV, Mahapatra MK, et al. Comput Biol Med. 2021;137:104851.
  24. Eder J, Sedrani R, Wiesmann C. Nat Rev Drug Discov. 2014;13(8):577-587.
  25. Garcia-Serna R, Vidal D, Remez N, Mestres J. Trends Pharmacol Sci. 2015;36(10):637-645.
  26. FDA. Guidance for Industry: Drug Development and Review Definitions. 2020.
  27. EMA. Guideline on Clinical Trials in Small Populations. 2006.
  28. CDSCO. Regulatory Pathways for Drug Repurposing in India. 2021.
  29. Breckenridge A, Jacob R. Nat Rev Drug Discov. 2019;18(1):1-2.
  30. FDA. FDA Adverse Event Reporting System (FAERS). 2023.
  31. EMA. Good Pharmacovigilance Practices (GVP). 2022.
  32. Ashburn TT, Thor KB. Nat Rev Drug Discov. 2004;3(8):673-83.
  33. Pushpakom S, Iorio F, Eyers PA, et al. Nat Rev Drug Discov. 2019;18(1):41-58.
  34. DiMasi JA, Grabowski HG, Hansen RW. J Health Econ. 2016;47:20-33.
  35. Kola I, Landis J. Nat Rev Drug Discov. 2004;3(8):711-6.
  36. Langedijk J, Mantel-Teeuwisse AK, Slijkerman DS, Schutjens MH. Drug Discov Today. 2015;20(8):1027-34.
  37. Nosengo N. Nature. 2016;534(7607):314-6.
  38. Zhou Y, Hou Y, Shen J, et al. Cell Discov. 2020;6:14.
  39. Beigel JH, Tomashek KM, Dodd LE, et al. N Engl J Med. 2020;383(19):1813-26.
  40. Breckenridge A, Jacob R. Nat Rev Drug Discov. 2019;18(1):1-2.
  41. Jin G, Wong ST. Drug Discov Today. 2014;19(5):637-44.
  42. Sahoo BM, Ravi Kumar BVV, Mahapatra MK, et al. Comput Biol Med. 2021;137:104851.
  43. Sleigh SH, Barton CL. Pharm Med. 2010;24(3):151-9.
  44. Vamathevan J, Clark D, Czodrowski P, et al. Nat Rev Drug Discov. 2019;18(6):463-77.
  45. Oprea TI, Bauman JE, Bologa CG, et al. Drug Discov Today Ther Strateg. 2011;8(3-4):61-9.
  46. Ashburn TT, Thor KB. Nat Rev Drug Discov. 2004;3(8):673-83.
  47. Nosengo N. Nature. 2016;534(7607):314-6.
  48. Li J, Zheng S, Chen B, et al. Brief Bioinform. 2016;17(1):2-12.
  49. Pushpakom S, Iorio F, Eyers PA, et al. Nat Rev Drug Discov. 2019;18(1):41-58.
  50. Jin G, Wong ST. Drug Discov Today. 2014;19(5):637-44.
  51. Keiser MJ, Setola V, Irwin JJ, et al. Nature. 2009;462(7270):175-81.
  52. Langedijk J, Mantel-Teeuwisse AK, Slijkerman DS, Schutjens MH. Drug Discov Today. 2015;20(8):1027-34.
  53. Chong CR, Sullivan DJ Jr. Nature. 2007;448(7154):645-6.
  54. Cavalla D. Drug Repositioning: Bringing New Life to Shelved Assets and Existing Drugs. Wiley; 2012.
  55. Franks ME, Macpherson GR, Figg WD. Lancet Oncol. 2004;5(7):375-82.
  56. Boolell M, Allen MJ, Ballard SA, et al. Int J Impot Res. 1996;8(2):47-52.
  57. Messenger AG, Rundegren J. Dermatol Clin. 2007;25(4):587-98.
  58. Li J, Zheng S, Chen B, et al. Brief Bioinform. 2016;17(1):2-12.
  59. Franks ME, Macpherson GR, Figg WD. Lancet Oncol. 2004;5(7):375-82.
  60. Boolell M, Allen MJ, Ballard SA, et al. Int J Impot Res. 1996;8(2):47-52.
  61. Ali K, Middleton MR, Garcia M, et al. Celecoxib as an adjuvant in paclitaxel chemotherapy. J Clin Oncol. 2005;23(16_suppl):9614.
  62. Jia P, Wang L, Fan Y, et al. BMC Bioinformatics. 2013;14(Suppl 16):S3.
  63. Franz DN, Leonard J, Tudor C, et al. N Engl J Med. 2006;354(1):18-28.
  64. Johnson SC, Rabinovitch PS, Kaeberlein M. Nat Rev Drug Discov. 2013;12(5):377-88.
  65. Martinkovich S, Shah D, Planey SL, Arnott JA. Steroids. 2014;90:13-25.
  66.  Li G, De Clercq E. Therapeutic options for the 2019 novel coronavirus (2019-nCoV). Nat Rev Drug Discov. 2020 Mar;19(3):149–150.
  67.  Pushpakom S, Iorio F, Eyers PA, et al. Drug repurposing: progress, challenges and recommendations. Nat Rev Drug Discov. 2019 Jan;18(1):41–58.
  68.  Zhou Y, Wang F, Tang J, Nussinov R, Cheng F. Artificial intelligence in COVID-19 drug repurposing. The Lancet Digital Health. 2020 Aug;2(8):e395–e404.
  69.  Harrison C. Coronavirus puts drug repurposing on the fast track. Nat Biotechnol. 2020 Apr;38(4):379–381.
  70.  Singh TU, Parida S, Lingaraju MC, Kesavan M, Kumar D, Singh RK. Drug repurposing approach to fight COVID-19. Pharmacol Rep. 2020 Aug;72(6):1479–1508.
  71.  Beigel JH, Tomashek KM, Dodd LE, et al. Remdesivir for the Treatment of Covid-19 — Final Report. N Engl J Med. 2020 Nov;383(19):1813–1826.
  72.  FDA. Remdesivir EUA Letter of Authorization. [Internet]. 2020 [cited 2025 Aug 7]. Available from: https://www.fda.gov/media/137564/download
  73.  RECOVERY Collaborative Group. Dexamethasone in Hospitalized Patients with Covid-19. N Engl J Med. 2021 Feb;384(8):693–704.
  74.  Chen C, Zhang Y, Huang J, et al. Favipiravir versus Arbidol for COVID-19: a randomized clinical trial. medRxiv. 2020; doi:10.1101/2020.03.17.20037432.
  75.  WHO. “Solidarity” clinical trial for COVID-19 treatments. [Internet]. 2021 [cited 2025 Aug 7]. Available from: https://www.who.int/emergencies/diseases/novel-coronavirus-2019/global-research-on-novel-coronavirus-2019-ncov/solidarity-clinical-trial-for-covid-19-treatments
  76.  Bryant A, Lawrie TA, Dowswell T, et al. Ivermectin for prevention and treatment of COVID?19 infection: a systematic review, meta?analysis, and trial sequential analysis to inform clinical guidelines. Am J Ther. 2021;28(4):e434e460.
  77.  Dugger BN, Dickson DW. Pathology of neurodegenerative diseases. Cold Spring Harb Perspect Biol. 2017;9(7):a028035.
  78.  Cummings J, Lee G, Zhong K, Fonseca J, Taghva K. Alzheimer's disease drug development pipeline: 2021. Alzheimers Dement (N Y). 2021;7(1):e12179.
  79.  Mullard A. Failures overshadowing CNS drug pipeline. Nat Rev Drug Discov. 2010;9(10):693–694.
  80. Pushpakom S, Iorio F, Eyers PA, et al. Drug repurposing: progress, challenges and recommendations. Nat Rev Drug Discov. 2019;18(1):41–58.
  81. Ashburn TT, Thor KB. Drug repositioning: identifying and developing new uses for existing drugs. Nat Rev Drug Discov. 2004;3(8):673–683.
  82. Lipton SA. Paradigm shift in neuroprotection by NMDA receptor blockade: memantine and beyond. Nat Rev Drug Discov. 2006;5(2):160–170.
  83. Cheah BC, Vucic S, Krishnan AV, et al. Riluzole, neuroprotection and amyotrophic lateral sclerosis. Curr Med Chem. 2010;17(18):1942–1199.
  84. Weinreb O, Amit T, Bar-Am O, Youdim MBH. Rasagiline: a novel anti-Parkinsonian monoamine oxidase-B inhibitor with neuroprotective activity. Prog Neurobiol. 2010;92(3):330–344.
  85. Rotermund C, Machetanz G, Fitzgerald JC. The therapeutic potential of metformin in neurodegenerative diseases. Front Endocrinol (Lausanne). 2018;9:400.
  86. Pagan FL, Hebron ML, Wilmarth B, et al. Nilotinib effects in Parkinson’s disease and dementia with Lewy bodies. J Parkinsons Dis. 2016;6(3):503–517.
  87. Cavalla D. Predictive value of preclinical models in CNS drug development. Drug Discov Today. 2019;24(1):30–36.
  88. DiMasi JA, Grabowski HG, Hansen RW. Innovation in the pharmaceutical industry: new estimates of R&D costs. J Health Econ. 2016;47:20–33.
  89. Dowling RJ, Goodwin PJ, Stambolic V. Understanding the benefit of metformin use in cancer treatment. BMC Med. 2011;9:33.
  90. Chan AT, Giovannucci EL, Meyerhardt JA, et al. Long-term use of aspirin and nonsteroidal anti-inflammatory drugs and risk of colorectal cancer. JAMA. 2005;294(8):914–923.
  91. Skrott Z, Mistrik M, Andersen KK, et al. Alcohol-abuse drug disulfiram targets cancer via p97 segregase adaptor NPL4. Nature. 2017;552(7684):194–199.
  92. Kim J, Tang JY, Gong R, et al. Itraconazole, a commonly used antifungal that inhibits Hedgehog pathway activity and cancer growth. Cancer Cell. 2010;17(4):388–399.
  93. Montoya A, Amaya CN, Belmont A, et al. Use of non-selective β-blockers is associated with decreased metastasis and improved recurrence-free survival in breast cancer. Oncotarget. 2017;8(41):70684–70693.
  94. Pantziarka P, Bouche G, Meheus L, Sukhatme V, Sukhatme VP, Vikas P. The repurposing drugs in oncology (ReDO) project. Ecancermedicalscience. 2014;8:442.
  95. Hotez PJ, Kamath A. Neglected tropical diseases in sub-Saharan Africa: review of their prevalence, distribution, and disease burden. PLoS Negl Trop Dis. 2009;3(8):e412.
  96. Chatelain E. Chagas disease drug discovery: toward a new era. J Biomol Screen. 2015;20(1):22–35.
  97. Ashburn TT, Thor KB. Drug repositioning: identifying and developing new uses for existing drugs. Nat Rev Drug Discov. 2004;3(8):673–683.
  98. Sundar S, Jha TK, Thakur CP, et al. Oral miltefosine for Indian visceral leishmaniasis. N Engl J Med. 2002;347(22):1739–1746.
  99. Müller J, Hemphill A. Effects of nitazoxanide on Giardia lamblia: a mini review. Parasitol Res. 2013;112(10):3537–3542.
  100. Upcroft JA, Upcroft P. Drug targets and mechanisms of resistance in the anaerobic protozoa. Clin Microbiol Rev. 2001;14(1):150–164.
  101. Barrett MP, Vincent IM, Burchmore RJ, et al. Drug resistance in kinetoplastid protozoa: mechanisms, mutations and diagnosis. Nat Rev Microbiol. 2011;9(6):430–440.
  102. Vale N, Moreira R, Gomes P. Primaquine revisited six decades after its discovery. Eur J Med Chem. 2009;44(3):937–953.
  103. Davidson A, Diamond B. Autoimmune diseases. N Engl J Med. 2001;345(5):340–350.
  104. Zhang Y, He D, Li M, et al. Drug repurposing for autoimmune diseases: a literature-based review. Front Pharmacol. 2021;12:654428.
  105. Schrezenmeier E, Dörner T. Mechanisms of action of hydroxychloroquine and chloroquine: implications for rheumatology. Nat Rev Rheumatol. 2020;16(3):155–166.
  106. Visser K, van der Heijde D. Risk and management of liver toxicity during methotrexate treatment in rheumatoid and psoriatic arthritis. Clin Exp Rheumatol. 2009;27(6):1017–1025.
  107. Rosenblum H, Amital H. Anti-TNF therapy: safety aspects of taking the risk. Autoimmun Rev. 2011;10(9):563–568.
  108. Feldmann M, Maini RN. Anti-TNF alpha therapy of rheumatoid arthritis: what have we learned? Annu Rev Immunol. 2001;19:163–196.
  109. Schwartz DM, Kanno Y, Villarino A, et al. JAK inhibition as a therapeutic strategy for immune and inflammatory diseases. Nat Rev Drug Discov. 2017;16(12):843–862.
  110. Institute of Medicine (US) Committee on Accelerating Rare Diseases Research and Orphan Product Development. Rare Diseases and Orphan Products: Accelerating Research and Development. Washington (DC): National Academies Press (US); 2010.
  111. Ferreira CR. The burden of rare diseases. Am J Med Genet A. 2019;179(6):885–892.
  112. Austin CP, Cutillo CM, Lau LPL, et al. Future of rare diseases research 2017–2027: an IRDiRC perspective. Clin Transl Sci. 2018;11(1):21–27.
  113. Walshe JM. Penicillamine, a new oral therapy for Wilson’s disease. Am J Med. 1956;21(4):487–495.
  114. Pipalia NH, Cosner CC, Huang A, et al. Histone deacetylase inhibitor treatment dramatically reduces cholesterol accumulation in Niemann–Pick type C1 mutant human fibroblasts. Proc Natl Acad Sci USA. 2011;108(14):5620–5625.
  115. Gross O, Licht C, Anders HJ, et al. Early angiotensin-converting enzyme inhibition in Alport syndrome delays renal failure and improves life expectancy. Kidney Int. 2012;81(5):494–501.
  116. Mantegazza R, Antozzi C. When myasthenia gravis is deemed refractory: clinical signposts and treatment strategies. Ther Adv Neurol Disord. 2018;11:1756285617749134.
  117. Saiman L, Anstead M, Mayer-Hamblett N, et al. Effect of azithromycin on pulmonary function in patients with cystic fibrosis uninfected with Pseudomonas aeruginosa: a randomized controlled trial. JAMA. 2010;303(17):1707–1715.
  118. Kishi N, Macklis JD. MECP2 is progressively expressed in post-migratory neurons and is involved in neuronal maturation rather than cell fate decisions. Mol Cell Neurosci. 2004;27(3):306–321.
  119. Morens DM, Fauci AS. Emerging pandemic diseases: how we got to COVID-19. Cell. 2020;182(5):1077–1092.
  120. WHO Solidarity Trial Consortium. Repurposed antiviral drugs for COVID-19 — interim WHO Solidarity trial results. N Engl J Med. 2021;384(6):497–511.
  121. Cao B, Wang Y, Wen D, et al. A trial of lopinavir–ritonavir in adults hospitalized with severe Covid-19. N Engl J Med. 2020;382(19):1787–1799.
  122. Caly L, Druce JD, Catton MG, et al. The FDA-approved drug ivermectin inhibits the replication of SARS-CoV-2 in vitro. Antiviral Res. 2020;178:104787.
  123. Lenze EJ, Mattar C, Zorumski CF, et al. Fluvoxamine vs placebo and clinical deterioration in outpatients with symptomatic COVID-19: a randomized clinical trial. JAMA. 2020;324(22):2292–2300.
  124. Gautret P, Lagier JC, Parola P, et al. Hydroxychloroquine and azithromycin as a treatment of COVID-19: results of an open-label non-randomized clinical trial. Int J Antimicrob Agents. 2020;56(1):105949.
  125. Amici C, Di Caro A, Ciucci A, et al. Indomethacin has a potent antiviral activity against SARS coronavirus. Antivir Ther. 2006;11(8):1021–1030.
  126. Roth GA, Mensah GA, Johnson CO, et al. Global burden of cardiovascular diseases and risk factors, 1990–2019: update from the GBD 2019 study. J Am Coll Cardiol. 2020;76(25):2982–3021.
  127. Benjamin EJ, Muntner P, Alonso A, et al. Heart disease and stroke statistics—2019 update: a report from the American Heart Association. Circulation. 2019;139(10):e56–e528.
  128. Pushpakom S, Iorio F, Eyers PA, et al. Drug repurposing: progress, challenges and recommendations. Nat Rev Drug Discov. 2019;18(1):41–58.
  129. McMurray JJV, Solomon SD, Inzucchi SE, et al. Dapagliflozin in patients with heart failure and reduced ejection fraction. N Engl J Med. 2019;381(21):1995–2008.
  130. Galie N, Ghofrani HA, Torbicki A, et al. Sildenafil citrate therapy for pulmonary arterial hypertension. N Engl J Med. 2005;353(20):2148–2157.
  131. Ridker PM, Everett BM, Thuren T, et al. Antiinflammatory therapy with canakinumab for atherosclerotic disease. N Engl J Med. 2017;377(12):1119–1131.
  132. Tardif JC, Kouz S, Waters DD, et al. Efficacy and safety of low-dose colchicine after myocardial infarction. N Engl J Med. 2019;381(26):2497–2505.
  133. Davignon J. Beneficial cardiovascular pleiotropic effects of statins. Circulation. 2004;109(23 Suppl 1):III39–III43.
  134. Celano CM, Huffman JC. Depression and cardiac disease: a review. Cardiol Rev. 2011;19(3):130–142.
  135. Marso SP, Daniels GH, Brown-Frandsen K, et al. Liraglutide and cardiovascular outcomes in type 2 diabetes. N Engl J Med. 2016;375(4):311–322.
  136. Gheorghiade M, Konstam MA, Burnett JC, et al. Short-term clinical effects of ranolazine in patients with acute heart failure syndromes: the RALI-DHF trial. J Am Coll Cardiol. 2011;58(7):747–755.
  137. Feher M. The value of drug repurposing in the cardiovascular therapeutic area. Expert Rev Cardiovasc Ther. 2021;19(2):81–92.
  138. Saudubray JM, Baumgartner MR, Walter JH. Inborn Metabolic Diseases: Diagnosis and Treatment. 6th ed. Springer; 2016.
  139. Rena G, Hardie DG, Pearson ER. The mechanisms of action of metformin. Diabetologia. 2017;60(9):1577–1585.
  140. Heerspink HJL, Stefánsson BV, Correa-Rotter R, et al. Dapagliflozin in patients with chronic kidney disease. N Engl J Med. 2020;383(15):1436–1446.
  141. Sahebkar A, Serban C, Mikhailidis DP, et al. Statin therapy reduces liver aminotransferases in patients with non-alcoholic fatty liver disease: a systematic review and meta-analysis of controlled clinical trials. J Clin Lipidol. 2016;10(2):519–533.
  142. Fonseca VA, Handelsman Y, Staels B. Colesevelam hydrochloride: a non-systemic lipid-altering drug. Cardiovasc Drugs Ther. 2010;24(1):5–11.
  143. Bray GA, Ryan DH. Update on obesity pharmacotherapy. Ann N Y Acad Sci. 2014;1311:1–13.
  144. Rekedal LR, Massarotti E, Garg R, et al. Changes in glycosylated hemoglobin after initiation of hydroxychloroquine or methotrexate treatment in diabetes patients with rheumatic diseases. Arthritis Rheum. 2010;62(12):3569–3573.
  145. Pijl H, Ohashi S, Matsuda M, et al. Bromocriptine: a novel approach to the treatment of type 2 diabetes. Diabetes Care. 2000;23(8):1154–1161.
  146. Torgerson JS, Hauptman J, Boldrin MN, et al. XENical in the prevention of diabetes in obese subjects (XENDOS) study: a randomized study. Diabetes Care. 2004;27(1):155–161.
  147. Henry RR, Smith SR, Schwartz SL, et al. Effects of weight loss with orlistat on glucose tolerance and progression to type 2 diabetes in obese patients with impaired glucose tolerance. Diabetes Care. 2001;24(1):133–140.
  148. Jha V, Garcia-Garcia G, Iseki K, et al. Chronic kidney disease: global dimension and perspectives. Lancet. 2013;382(9888):260–272.
  149. Himmelfarb J, Tuttle K. Challenges in the design of clinical trials for chronic kidney disease: lessons learned and future directions. Kidney Int. 2014;86(5):894–903.
  150. Pushpakom S, Iorio F, Eyers PA, et al. Drug repurposing: progress, challenges and recommendations. Nat Rev Drug Discov. 2019;18(1):41–58.
  151. Wheeler DC, Stefánsson BV, Jongs N, et al. Effects of dapagliflozin on major adverse kidney and cardiovascular events in patients with diabetic and non-diabetic chronic kidney disease: a prespecified analysis from the DAPA-CKD trial. Lancet Diabetes Endocrinol. 2021;9(1):22–31.
  152. Locatelli F, Bárány P, Covic A, et al. Kidney Disease: Improving Global Outcomes guidelines on anaemia management in chronic kidney disease: a European Renal Best Practice position statement. Nephrol Dial Transplant. 2013;28(6):1346–1359.
  153. Lewis EJ, Hunsicker LG, Clarke WR, et al. Renoprotective effect of the angiotensin-receptor antagonist irbesartan in patients with nephropathy due to type 2 diabetes. N Engl J Med. 2001;345(12):851–860.
  154. Tonelli M, Isles C, Curhan GC, et al. Effect of statins on kidney function, proteinuria, and progression of chronic kidney disease: a meta-analysis of randomized trials. BMJ. 2011;343:d1895.
  155. Bomback AS, Klemmer PJ. The incidence and implications of aldosterone breakthrough. Nat Clin Pract Nephrol. 2007;3(9):486–492.
  156. Yilmaz MI, Romano M, Basarali MK, et al. The potential role of colchicine in reducing inflammation and proteinuria in chronic kidney disease. Kidney Blood Press Res. 2021;46(2):149–158.
  157. Tepel M, van der Giet M, Schwarzfeld C, et al. Prevention of radiographic-contrast-agent-induced reductions in renal function by acetylcysteine. N Engl J Med. 2000;343(3):180–184.
  158. Song X, Xue X, Wang Y, et al. Sirolimus as a therapeutic option in focal segmental glomerulosclerosis and Alport syndrome: an experimental review. Nephrol Dial Transplant. 2021;36(2):301–308.
  159. Navarro-González JF, Mora-Fernández C, Muros M, et al. Pentoxifylline for renal protection in diabetic nephropathy: a systematic review and meta-analysis. PLoS One. 2013;8(9):e74431.
  160. Hoorn EJ, Walsh SB, McCormick JA, et al. The calcineurin inhibitor tacrolimus activates the renal sodium chloride cotransporter to cause hypertension. Nat Med. 2011;17(10):1304–1309.
  161. Sparks DL, Sabbagh MN, Connor DJ, et al. Atorvastatin for the treatment of mild to moderate Alzheimer disease: preliminary results. Arch Neurol. 2005;62(5):753–757.
  162. Panonnummal R,Varkey J. Anti-inflammatory activity of HMG CoA reductase inhibitors: a comparative study. Int J Pharm Pharm Sci. 2015;7(3):468-9.
  163. Jiang P, Mukthavaram R, Chao Y, et al. In vitro and in vivo anticancer effects of mevalonate pathway inhibition by lovastatin in glioblastoma. Cancer Lett. 2014;349(2):161–170.
  164. Wilkins MR, Ali O, Bradlow W, et al. Simvastatin as a treatment for pulmonary hypertension trial. Am J Respir Crit Care Med. 2010;181(10):1106–1113.

Photo
Dr. Sinchu Yesudanam
Corresponding author

Mar Dioscorus College of Pharmacy.

Photo
Anshu Shyjin
Co-author

Mar Dioscorus College of Pharmacy.

Photo
M. Muhammed Aslam
Co-author

Mar Dioscorus College of Pharmacy.

Dr. Sinchu Yesudanam*, Anshu Shyjin, M. Muhammed Aslam, Drug Reprofiling in the Age of Necessity: A Strategic-Clinical Synthesis for Therapeutic Clinical Outcome, Int. J. of Pharm. Sci., 2025, Vol 3, Issue 8, 1667-1688. https://doi.org/10.5281/zenodo.16881340

More related articles
Molecular Docking: A Technique For Discovering Tel...
Vidya Magar , Karna Khavane, Shailesh Patwekar, Santosh Shelke, ...
A Review On Millets And Their Types ...
Pragya Nigam, Dharmesh Sisodiya, Kamlesh Dashora, ...
A Review On : Fast Dissolving Oral Thin Film...
Pote Ashwini.B, Zarad Snehal, Ashok Dalimbe , ...
Anti-Acne Gel Comprehensive Review of Formulation, Efficacy, Safety and Clinical...
Sachin Bhalekar , Sanika Bankhele , Monika Ranpise , Bhakti Ingale , ...
Liposome-Novel Drug Delivery Systems...
Utkarsha Gadekar , Ulka Mote, ...
Solid Lipid Nanoparticle for Topical Drug Delivery...
Arya Ashok, Praveen Raj R., Riya Marium Philip, ...
Related Articles
Comparative Study Of Vildagliptin Branded And Generic Marketed Antidiabetics Tab...
Shivani S. Pangarekar, Tanaya S. Pawar, Sakshi S. Rashinkar, Ganesh V. Taware, ...
A Comprehensive Review of Insights into Rheumatoid Arthritis: From Pathophysiolo...
Sanika Gurav, Shruti Choudhari, Shweta Patil, Tejasvini Kamble, Shradha Lokare, Dr. Jadge D. R., ...
In Silico Molecular Docking Investigation to Develop Novel N-[4-nitro-2-(trifluo...
Soumallya Chakraborty, Somenath Bhattacharya, Santu Mandal, ...
A Review On Orodispersible Tablets...
A. Manoj Kumar PATRO, Ali Osman SARIKAYA, Emre Erol ALDEN?Z, Udaya Kumar DUDE, ...
Molecular Docking: A Technique For Discovering Telomerase Inhibitors For Cancer ...
Vidya Magar , Karna Khavane, Shailesh Patwekar, Santosh Shelke, ...
More related articles
Molecular Docking: A Technique For Discovering Telomerase Inhibitors For Cancer ...
Vidya Magar , Karna Khavane, Shailesh Patwekar, Santosh Shelke, ...
A Review On Millets And Their Types ...
Pragya Nigam, Dharmesh Sisodiya, Kamlesh Dashora, ...
A Review On : Fast Dissolving Oral Thin Film...
Pote Ashwini.B, Zarad Snehal, Ashok Dalimbe , ...
Molecular Docking: A Technique For Discovering Telomerase Inhibitors For Cancer ...
Vidya Magar , Karna Khavane, Shailesh Patwekar, Santosh Shelke, ...
A Review On Millets And Their Types ...
Pragya Nigam, Dharmesh Sisodiya, Kamlesh Dashora, ...
A Review On : Fast Dissolving Oral Thin Film...
Pote Ashwini.B, Zarad Snehal, Ashok Dalimbe , ...