Revision Article
Reposicionamiento de Fármacos: Nuevos Caminos para Medicamentos Conocidos
1 Laboratorio de Biotecnología Farmacéutica, Centro de Biotecnología Genómica, Instituto Politécnico Nacional, Reynosa, Tamaulipas, México.
2 Laboratorio de Investigación en Farmacia, Farmacobiología y Toxicobiología, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Sinaloa, Ciudad Universitaria, Culiacán, Sinaloa, México.
Drug repositioning is a strategy that leverages approved medications to treat diseases for which they were not ori- ginally designed. This approach reduces the time and costs associated with developing new drugs, facilitating the disco- very of new therapeutic options. This review aims to explore the methodologies used, the current challenges, and the potential impact of this approach in medicine. A search was conducted in the PubMed and ScienceDirect databases using keywords related to drug repositioning, combining terms such as “repurposing” with different types of medications. The most recent information was prioritized, specifically for the last 15 years (with 2010 used as a threshold), and articles deemed irrelevant or outdated were excluded. Successful repositioning cases are highlighted, such as ibuprofen, a Non-Steroidal Anti-Inflammatory Drug (NSAID), which may offer benefits in neurodegenerative diseases. Acetylsalicylic acid, initially designed for hypertension and angina, has presented activity against cancer and Alzheimer’s disease. Omeprazole, used for acid-related secretory disorders, has shown antiparasitic activity. These examples emphasize how approved drugs, with established safety profiles, can have new uses. Drug repositioning, supported by bioinformatics tools, represents a promising avenue for treating neglected diseases and improving existing therapies through drug combinations that optimize efficacy and reduce side effects. Keywords: Repurposing, non-steroidal anti-inflammatory, sildenafil, omeprazole, bioinformatics.
El reposicionamiento de fármacos es una estrategia que aprovecha medicamentos aprobados para tratar enferme- dades distintas a las originalmente previstas. Este enfoque reduce tiempo y costos asociados al desarrollo de nuevos fármacos, facilitando el descubrimiento de nuevas opciones terapéuticas. Esta revisión tiene como objetivo explorar las metodologías utilizadas, los desafíos actuales y el impacto potencial de este enfoque en la medicina. Se realizó una búsqueda en las bases de datos PubMed y ScienceDirect utilizando palabras clave relacionadas con el reposiciona- miento de fármacos, combinando términos como “repurpo- sing” con diferentes tipos de medicamentos. Se priorizó la
*Author for correspondence: Gildardo Rivera e-mail: giriveras@ipn.mx
Received: February 21, 2025
Accepted: April 25, 2025
Published: May 30, 2025
información más reciente, específicamente de los últimos 15 años (tomando 2010 como límite), y se excluyeron artículos irrelevantes o desactualizados. Se destacan casos exitosos de reposicionamiento, como el ibuprofeno, un antiinflamatorio no esteroideo (AINE) estudiado contra enfermedades neuro- degenerativas. El ácido acetilsalicílico, diseñado inicialmente para la hipertensión y angina, que ha mostrado actividad contra el cáncer y Alzheimer; y el omeprazol, utilizado para la gastritis, ha demostrado actividad antiparasitaria. Estos ejemplos subrayan cómo fármacos aprobados, con perfiles de seguridad establecidos, pueden tener nuevos usos. El reposicionamiento de fármacos, apoyado por herramientas bioinformáticas, representa una vía prometedora para tratar enfermedades desatendidas y mejorar terapias mediante combinaciones que optimizan la eficacia y reducen efectos secundarios.
The strategy of identifying new therapeutic uses for exis- ting drugs has long been a well-established approach in pharmaceutical research. However, recent advances in computational tools and omics technologies have brought renewed innovation to this field, enabling more systematic, large-scale, and data-driven repositioning efforts. These mo- dern methodologies offer a cost-effective and time-efficient alternative to traditional drug development, by leveraging existing pharmacological and safety profiles to accelerate the discovery of new indications. This approach has proven par- ticularly valuable in fields such as oncology (Xia et al., 2024), infectious diseases (Devillers, 2018; Pires et al., 2021; Pizzorno et al., 2019), and neurodegenerative disorders (Chaffey et al., 2022; Onódi et al., 2023). Despite its advantages, drug repo- sitioning still faces challenges, including the identification of novel therapeutic targets and the experimental validation of computational predictions (Deshpande et al., 2020; Juá- rez‐Saldivar et al., 2021; Rashid et al., 2023). The objective of this review is to provide a comprehensive overview of drug repositioning as a strategy for accelerating drug discovery, with a particular focus on distinct family drugs. By integra- ting computational approaches, omics technologies, and
Volume XXVII
DOI: 10.18633/biotecnia.v27.2591
Journal of biological and health sciences http://biotecnia.unison.mx
Universidad de Sonora
ISSN: 1665-1456
experimental validation, this review aims to highlight current advances and in silico strategies to improve the identification and development of repositioned drug candidates.
A systematic literature search was conducted using the Pub- Med and ScienceDirect databases to identify relevant studies on drug repositioning. The search strategy involved the use of specific keywords. The primary search terms included “drug repositioning,” “drug repurposing,” and “therapeutic repositioning,” which were combined with different classes of drugs, such as “NSAIDs” and diseases, to ensure a compre- hensive retrieval of relevant publications. To maintain the relevance and accuracy of the review, only articles published within the last fifteen years were considered, setting 2010 as the lower limit, prioritizing the most recent ones. This time- frame was chosen to reflect the most recent advancements in the field while ensuring that foundational studies were included. Studies were filtered based on relevance, prioriti- zing peer-reviewed articles, systematic reviews, and original research papers. Articles deemed outdated, redundant, or lacking methodological rigor were excluded.
Repurposing of non-steroidal anti-inflammatory drugs Non-steroidal anti-inflammatory drugs (NSAIDs) are a class of medications approved by the Food and Drug Administration (FDA) for their antipyretic, anti-inflammatory, and analgesic properties. They are commonly used to manage muscle pain, menstrual cramps, arthritis, fever, gout, and migraines, and as alternatives to opioids in some acute trauma situations (Sobhani et al., 2023). However, NSAIDs have demonstrated several activities than those originally reported, for exam- ple, acetylsalicylic acid (ASA), marketed and better known as Aspirin®, first was designed as an NSAID and then as an antiplatelet drug (Patrono and Rocca, 2021). The inhibitory action of this compound on blood platelets was not disco- vered until the late 1960’s, and it has been little more than a decade since this property was linked to the inhibition of the cyclooxygenase enzymes, mostly COX-1 through Ser530 prevents the production of prostaglandin and thromboxane A2, responsible for inducing platelet aggregation and vaso- constriction in endothelial cells. This mechanism of action explains the effect of ASA in preventing thrombosis of the ce- rebral vessels and coronary arteries (Fijałkowski et al., 2022).
In the last decade, new potential activities have been re- ported for ASA, including its antiviral properties. Its effect has been evaluated against a broad range of human pathogenic viruses, including adenovirus (Adeno 5), herpes simplex virus (HSV-1), human influenza A virus (FluA H1N1), respiratory syncytial virus (RSV), Coxsackie virus (CA9), and several human rhinoviruses (HRV). Specifically, ASA’s influence was analyzed in two HRV subtypes from both the “minor group” (HRV1A, HRV2) and the “major group” (HRV14, HRV39). In vitro studies were conducted using plaque reduction assays, demonstrating a dose-dependent effect of ASA against FluA H1N1, with a half-maximal effective concentration (EC50)
value of 0.66 mmol/L. For CA9, HRV2, HRV14, and HRV39, the EC50 values were 0.98, 0.69, 0.21, and 0.1 mmol/L, respecti- vely, whereas for the remaining viruses, the values exceeded 1 mmol/L. Interestingly, only RNA viruses were susceptible to the antiviral activity of ASA, suggesting a certain degree of specificity (Glatthaar-Saalmüller et al., 2017).
More recently, related to the specificity of previous work, ASA activity was tested against Bunyamwera virus (BUNV), a human pathogen RNA virus (Fernández-Sánchez et al., 2023). Authors use viral RNA polymerases as targets, proteins essential for viral genome replication and share sequence as well as structural similarities across different families of RNA viruses (Horst et al., 2019). Their approach was combining both ligand-based and structure-based molecular models, using a known influenza endonuclease inhibitor as reference 2,4-dioxo-4-phenylbutanoic acid (DPBA), obtaining FDA- approved drugs from DrugBank Database, and using crystal structure of the Hantaan virus endonuclease as target to obtain hit compounds through molecular docking. ASA was one of the hit compounds and was selected because of its si- milarity to DPBA. From the top five hit compounds, only ASA showed antiviral activity against BUNV, with an IC50 of 2.02 mM. ASA significantly reduced viral titters in BUNV-infected Vero cells, decreasing the expression of viral proteins Gc and N in a dose-dependent manner. ASA also inhibited the formation of Golgi-associated viral replication structures and protected the Golgi complex from BUNV-induced fragmen- tation, suggesting an interaction with vRNA polymerase that disrupts viral replication. Similar effects were observed with the vRNA polymerase inhibitor Ribavirin. Notably, the remo- val of ASA from the culture medium restored normal Golgi morphology and viral structures, indicating that its antiviral effects are reversible. While ASA effectively inhibits BUNV replication at 2 mM, its anticoagulant properties limit its use in hemorrhagic fever cases. However, ASA remains a cost- effective and safe candidate for broad-spectrum antiviral therapy, with potential applications against select bunyavi- rus infections (Fernández-Sánchez et al., 2023). The structural and functional similarities between influenza virus and bun- yavirus endonucleases suggest that inhibitors designed for influenza could be repurposed to target bunyaviruses. This rationale guided an in silico screening of the DrugBank data- base, which includes approximately 10,000 compounds with prior human use. Using computational methods rather than manual selection allowed for a systematic evaluation of che- mical similarities to DPBA, a known influenza endonuclease inhibitor. Virtual screening not only identified structurally related candidates, but also confirmed their compatibility with the catalytic site, a critical factor for effective inhibition. These findings underscore the importance of computational approaches in drug repurposing efforts, facilitating the iden- tification of potential antiviral agents against bunyaviruses.
Several studies have explored the potential ASA benefits in COVID-19 patients, particularly regarding its antithrom- botic and anti-inflammatory properties (Tantry et al., 2021). Observational cohort studies have reported that ASA use
before or shortly after hospitalization was associated with reduced rates of mechanical ventilation, intensive care unit (ICU) admission, and in-hospital mortality, without signifi- cant differences in thrombosis or major bleeding (Chow et al., 2021). In American veterans with COVID-19, preexisting ASA prescriptions correlated with a lower mortality risk at both 14 and 30 days (Osborne et al., 2021). Similarly, in-hospital ASA use was linked to a decreased incidence of mortality in a propensity score-matched study (Meizlish et al., 2021). Another retrospective analysis suggested that ASA users had a lower risk of COVID-19 infection and a shorter disease du- ration (Merzon et al., 2021). Overall, prior and in-hospital ASA use at doses ranging from 75 to 325 mg per day has been associated with improved survival outcomes in COVID-19 patients (Liu et al., 2021).
Additionally, ASA has been studied to prevent cardio- vascular diseases (CVD) linked to a first heart attack or stroke (Cofer et al., 2022). COX-1 and P2Y12 inhibitors are two classes of antiplatelet drugs approved to treat acute coronary syn- drome and secondary prevention. Therefore, the combina- tion of ASA (COX-1 inhibitor) and clopidogrel (P2Y12 inhibitor) is the standard treatment for atherosclerotic cardiovascular disease as per the 2015 European Society of Cardiology gui- delines (Qaderdan et al., 2015; Saqallah et al., 2024).
ASA has presented activity on distinct types of cancer, like colorectal, where studies suggest that the reduction in risk of colorectal cancer is present in older people who have been using ASA since a younger age (Shureiqi, 2022). Re- cently, in silico and in vitro approaches showed that ASA also has good affinity to estrogen receptor alpha (ERα) and cause a decrease of mRNA levels of ERα followed by a decrease in protein expression, suggesting that ASA could be used as a potential anti-breast cancer agent (Guo et al., 2021; Kaur et al., 2024). In one study, ASA was docked onto three ERα crystals, each bound to a different type of ligand: agonist (1GWR), antagonist (5UFX), and selective estrogen receptor modulator (SERM) (3ERT) (Figure 1). The aim was to evaluate whether agonists (such as 17-β estradiol), antagonists (such as OP1074), or SERMs (such as tamoxifen) have unique bin- ding patterns and to compare ASA’s behavior with them. The interactions between ASA and 3ERT showed HBs with residues Gly-521, Thr-347, and His-524, with His-524 being common in both antagonist-bound and SERM-bound ERα complexes. The Thr-347 interaction was exclusive to anta- gonist-bound ERα complexes. To assess the stability of ASA on different crystals, Molecular Dynamics (MD) studies were carried out. ASA was unstable on the 5UFX crystal, whereas ASA remained stable on the 1GWR and 3ERT crystals during
Figure 1. Crystallographic structures of estrogen receptor alpha (ERα) in complex with different types of inhibitors. A) ERα in complex with the agonist inhibitor 17-β estradiol. B) ERα in complex with the antagonist inhibitor OP1074. C) ERα in complex with the selective modulator inhibitor tamoxifen. Each ERα chain is represented in a different color (yellow, blue, green, and purple), and the inhibitor is shown in red. The structure of the inhibitor is displayed below each crystal. Figura 1. Estructuras cristalográficas del receptor de estrógenos alfa (ERα) en complejo con diferentes tipos de inhibidores. A) ERα en complejo con el inhibidor agonista 17-β estradiol. B) ERα en complejo con el inhibidor antagonista OP1074. C) ERα en complejo con el inhibidor modulador selectivo tamoxifen. Cada cadena del ERα se representa en un color diferente (amarillo, azul, verde y púrpura) y el inhibidor se muestra en color rojo. La estructura del inhibidor se muestra en la parte inferior de cada cristal.
a 500 ns simulation. The 3ERT + ASA complex showed a lower ligand RMSD (~1 Å), greater receptor stability, and higher average interaction energy (-26.86 kcal/mol) compared to the 1GWR complex (-22.73 kcal/mol), indicating stronger and more stable binding in the SERM-bound ERα conformation. Finally, following MD trajectories, authors found in 1GWR
+ ASA complex, ASA maintained HB with residues Arg-394, Glu-323, and Lys-449, and showed hydrophobic contacts with Pro-324. In 3ERT + ASA, it retained interactions with re- sidues Gly-521, Thr-347, and His-524, as well as water bridge interactions with Ala-350 and Leu-525. These interactions were similar to those observed in antagonist- and SERM- bound ERα complexes, suggesting a potential antagonistic behavior of ASA. Secondary structure content, radius of gyra- tion, and solvent-accessible surface area were comparable in both stable systems. Overall, 3ERT + ASA demonstrated more favorable dynamic and energetic properties, supporting its potential relevance in drug repositioning efforts, and his stability and interaction pattern suggests that ASA might behave like an antagonist (Kaur et al., 2024).
Another example of NSAID repurposing is ibuprofen, a drug developed as an anti-rheumatic in the 1960’s, that now is a drug with anti-inflammation and antipyretic properties, and it’s one of the analgesics mostly used worldwide. A few studies have also explored NSAID-based treatments targe- ting key features of cystic fibrosis (CF) lung disease, particu- larly chronic infections and inflammation, which are major contributors to morbidity in CF patients (Lands and Stano- jevic, 2016). Ibuprofen not only apports to the treatment of CF because of its anti-inflammatory activity but also has antibacterial and antifungal activities that could combat the causes of the inflammation. For example, Shah et al. (2018) tested the activity of ibuprofen against CF-associated Gram-negative pathogens. Their results showed that ibu- profen reduces in vitro the growth of different Pseudomonas aeruginosa strains (PAO1, M57-15, T63547, and H25815) and Burkholderia spp. in a dose-dependent manner. P. aeruginosa had a significant reduction in ATP after 100 μg/mL, 30-min exposure to ibuprofen. In vivo studies achieved lower CFU counts of P. aeruginosa PAO1 in the lung and the spleen, and a significant survival advantage (92%) was observed in mice after ibuprofen oral treatment (Shah et al., 2018).
Antiviral activity has also been studied for ibuprofen. From a dataset that contained 382 experimental and 267 approved drugs, Veljkovic et al. (2015) carried out a blind molecular docking on glycoprotein GP1 from the Ebola vi- rus. This protein is not crystalized, but they previously had obtained a model (Veljkovic et al., 2015). Results suggested that ibuprofen binds to Elastin Microfibril Interface Located Proteins (EMILINs) binding domain in GP1, involved in inte- raction between GP1 and endothelial extracellular matrix (ECM) (Veljkovic et al., 2015). This is just an in silico approach but it was a good start for further investigation of ibuprofen against the Ebola virus.
Recently, Borba-Santos et al. (2021) found that ibupro- fen had low in vitro antifungal activity against Sporothrix
brasiliensis and S. schenckii (MIC median of 256 μg/mL), but when is co-administrated with antifungal drugs (amphote- ricin B, itraconazole, and terbinafine), it was able to reduce the concentrations required to inhibit fungal growth in vitro. In this study, the best combination obtained was ibuprofen plus amphotericin B against S. schenckii. This combination achieved a great decrease in the dose of amphotericin B (up to 125-fold) (Borba-Santos et al., 2021). Given the high toxicity of amphotericin B and the associated side effects experienced by patients, as well as the possibility that these effects are replicated in vivo, reducing the dose of this drug could represent a significant benefit for the treatment of the patient due to their joint activity with ibuprofen.
Drugs repurposed through serendipity are also an example of success. Sildenafil, a phosphodiesterase-5 (PDE5) inhibitor (Figure 2) also known as Viagra®, started clinical development as an agent for the treatment of hypertension and angina, a disease that is characterized by present chest pain caused by the less quantity of blood that arrives at the heart, and sub- sequently evolved into a revolutionary new oral treatment for erectile dysfunction and pulmonary hypertension during clinical trials (Ghofrani et al., 2006).
Nitric oxide (NO) functions as an inhibitory neuro- transmitter in the gastrointestinal system, promoting the production of guanosine 3′,5′-cyclic monophosphate (cGMP) following swallowing. This process reduces lower esophageal sphincter (LES) pressure by inducing smooth muscle relaxa- tion. The cGMP that accumulates is eventually broken down by PDE-5. Sildenafil, a powerful PDE-5 inhibitor, helps main- tain cGMP levels by preventing its degradation, which leads to continued smooth muscle relaxation. Although, sildenafil is primarily used to treat erectile dysfunction by relaxing smooth muscle in the arterioles of the corpus cavernosum, it also reduces LES pressure, decreases esophageal propulsion, and slows gastric emptying (Wong et al., 2021).
In the last decade, sildenafil has also demonstrated to improve the antineoplastic drugs effects, for example, Das et al. (2010) obtained an increased antineoplastic activity when they combined sildenafil with doxorubicin (a broad-spectrum antitumor-antibiotic that has been widely used for treatment of several cancers, including breast, ovarian, and prostate cancers) against bone metastasis PC-3 and human prostate cancer DU145 cells. Doxorubicin alone induces apoptosis of 7.52% and 45.02% of PC-3 and DU145 cells, respectively. Co-treatment with sildenafil and doxorubicin increases apoptosis in both cell lines (18.71% in PC-3 and 56.82% in DU145 cells). Interestingly, sildenafil doesn’t have an effect alone against these cell lines (Das et al., 2010). In 2020, another study showed that exposure of the breast cancer cell line MDA-MB-231 to low concentrations of sildenafil (25 µM) inhibited cancer cell proliferation, promoted apoptosis, and reduced tumor growth. This antitumor effect was linked to sildenafil’s impact on the expression of HSP90, a molecular chaperone that facilitates the degradation of protein kinase
Figure 2. Crystallographic structure of phosphodiesterase 4 (PDE4) with sildenafil bounded to the catalytic pocket. The three-dimensional structure of PDE4 is shown in grey. A sulfate ion is represented in red-yellow, magnesium in green, zinc in black, and sildenafil in blue. The chemical structure of sildenafil is shown on the right.
Figura 2. Estructura cristalográfica de la fosfodiesterasa 4 (PDE4) con sildenafil unido a la cavidad catalítica. En color gris, la estructura tridimensional de PDE. En rojo-amarillo, un sulfato. En verde, magnesio. En negro, zinc. En azul, sildenafil. A la derecha se muestra la estructura del sildenafil.
D2 (PKD2), a serine/threonine kinase crucial for maintaining cancer cell proliferation and viability (Chen et al., 2020).
Many cancers are characterized by PDE5 overexpres- sion, which suggests their involvement in carcinogenesis, and may be linked to the variation in the ratio of cGMP to cAMP and PDE levels among healthy and malignant tissues, notably in those with higher cGMP PDE activity (Savai et al., 2010). Therefore, sildenafil was evaluated in vivo and in silico as antineoplastic potential against Ehrlich Ascites Carcinoma. Sildenafil, both as a mono-treatment and in combination with cisplatin, significantly reduced tumor cell count, viabili- ty, growth rate, and proliferation while promoting apoptosis and causing cell cycle arrest at G0/G1 and sub-G1 phases. It also enhanced the cellular immune response by increasing plasma levels of granzyme B and IFN-γ, and improved the proportion of cytotoxic (CD3+) and helper T cells (CD3+CD4+) in the spleen while decreasing regulatory T cells. To evaluate the potential mechanism of action of sildenafil, some crystals as Major Histocompatibility (MHC), Lymphocyte-Specific Protein Tyrosine Kinase (Lck), Mitogen-Activated Protein Ki- nases (MAPKs) and PDE, were retrieved to molecular docking. Sildenafil achieved excellent docking scores -17.5 and -18.1 Kcal/mol with Lck and MAPKs, respectively, very close to the docking score achieved by sildenafil with PDE (-18.3 Kcal/ Mol). Docking of sildenafil with MHC achieved only a doc- king score of – 6.4 Kcal/mol. To this end, LcK and MAPKs were identified as potential targets for sildenafil, and their docking results were further analyzed. sildenafil was found to bind in the ATP binding site of Lck, with a docking score of -17.5 kcal/mol, forming a HB with the crucial gatekeeper residue Thr316. Additionally, sildenafil interacts with Asp382, Gly252,
and Gly254 through a HB and with Lys273 via an arene inte- raction. In contrast, the co-crystal ligand binds with Val259 through an HB and with Thr316 and Met319 through HBs. On the other hand, sildenafil exhibited a similar binding pattern to the co-crystallized ligand in the p38α MAPK binding site, with docking scores of -18.3 and -17.9 kcal/mol, respectively. The nitrogen atom of sildenafil’s piperidine ring forms a HB with Lys53 in the active site, while the co-crystal ligand interacts with Lys53 via one HB and two arene interactions. sildenafil also forms hydrophobic bonds with Met109 in the hinge region, while the co-crystal ligand accepts HB. Addi- tionally, sildenafil demonstrated enhanced binding through three hydrophobic interactions with Gly31 and Val30. In conclusion, in silico data suggest that Lck and MAPKs are potential targets of immunomodulatory, apoptotic, and anti- proliferative sildenafil’s activity (Morsi et al., 2023).
In a recent study, sildenafil showed a reduction in the likelihood of Alzheimer’s Disease (AD), with a 54% reduced incidence, finding that sildenafil treatment reduced taurine hyperphosphorylation (pTau181 and pTau205) in a dose- dependent manner in both familial and sporadic AD patient isogenic pairs of wild-type and mutant induced pluripotent cell (iPSC) derived neurons. Moreover, the same study showed that sildenafil targets AD-related genes and patho- biological pathways, supporting the potential of this erectile dysfunction drug as a candidate drug for the treatment of AD (Gohel et al., 2024).
The expanding therapeutic potential of sildenafil opens new avenues for both neurological and oncological treatments. Its capacity to reduce tau hyperphosphorylation and modulate Alzheimer’s disease-related pathways suggests
that it could become a viable option in the development of disease-modifying therapies for neurodegenerative disor- ders not only tested alone but also tested as a co-treatment with other reported compounds of these diseases (Gohel et al., 2024).
Fang et al. (2021) explored the potential of repurposing sildenafil as a treatment for AD using an endophenotype- based in silico network. Sildenafil was significantly associa- ted with a 69% reduced risk of AD, based on retrospective pharmaco-epidemiological analyses of insurance claims data from 7.23 million individuals. This association remained consistent across multiple drug cohorts, even after adjusting for factors such as age, sex, race, and comorbidities. Mecha- nistically, sildenafil was shown to increase neurite growth and decrease phospho-tau expression in neuron models derived from AD patient-induced pluripotent stem cells, suggesting a potential therapeutic effect for AD. Further- more, the study demonstrated a network-based approach for AD drug discovery, integrating multi-omics data from AD transgenic mouse models, which improved predictions for potential treatments. Comprehensive subgroup analyses also indicated that sildenafil was associated with a reduced risk of AD across various demographic and health condition
subgroups, strengthening its potential as a candidate for AD prevention or treatment. Overall, these findings highlight sildenafil’s promise as a repurposed drug for AD, supported by both epidemiological data and mechanistic evidence (Fang et al., 2021).
Proton-pump inhibitors (PPI) are a group of benzimidazole- substituted compounds that are employed to treat gastric and duodenal ulcers, gastroesophageal reflux disease, and other excessive acid-related secretory disorders. PPIs as ome- prazole and their analogues (esomeprazole, pantoprazole, lansoprazole and rabeprazole) since 2011 have been studied as new antiprotozoal agents.
Pérez-Villanueva et al. (2011) reported PPIs antiprotozoal activity against Trichomonas vaginalis, Giardia intestinalis, and Entamoeba histolytica. All PPIs had activity in the nanomolar range against the three parasites. PPIs were 1.9-3.1 times more active than metronidazole against T. vaginalis and 12.8-78.1 times more active against G. intestinalis. Besides, rabeprazole and pantoprazole were 14.8 and 134.6 times more active than metronidazole against E. histolytica. The authors highlight the activity of pantoprazole against the
Figure 3. Crystal structure of Giardia intestinalis triosephosphate isomerase in its homodimeric form (PDB: 4BI7) and proton pump inhibitors structure. Since only mutant crystals of this protein are available with altered cysteines, the structure was modeled to represent the wild-type form (only the Ala202Cys mutation was introduced). A) Cysteines 14, 127, 202, and 228 are shown in yellow in stick representation. Cysteine 222 is shown in pink; this residue is derivatized by proton pump inhibitors, leading to its giardicidal activity. B) Structure of proton pump inhibitors. From top to bottom: omeprazole, esomeprazole, pantoprazole, lansoprazole, and rabeprazole.
Figura 3. Estructura cristalográfica de la triosa fosfato isomerasa de Giardia intestinalis en su forma homodimérica (PDB:4BI7) y estructura de los inhibidores de la bomba de protones. Dado que existen solo cristales mutados en las cisteínas de esta proteína, se modeló para tenerlo en su forma Wild-type (únicamente se hizo el cambio Ala202Cys). En color amarillo, las cisteínas en representación stick (cisteínas 14, 127, 202 y 228). En color rosa, la císteina 222, la cual se derivatiza por los inhibidores de la bomba de protones para causar su actividad giardicida. B) Estructura de los inhibidores de la bomba de protones. De arriba abajo: omeprazol, esomeprazol, pantoprazol, lansoprazol y rabeprazol.
three anaerobic protozoa tested (with IC50 values of 0.0756, 0.0157, and 0.0026 µM against T. vaginalis, G. intestinalis, and
E. histolytica, respectively) (Pérez-Villanueva et al., 2011).
After, Reyes-Vivas et al. (2014) showed the mechanism of action of PPIs, specifically omeprazole, on triosephosphate isomerase of G. lamblia (GlTIM). This activity is linked to the inactivation of triose phosphate isomerase (TIM) of the pa- rasite in a specific manner through interaction with Cys 222 (Figure 3), because PPIs bind covalently to sulfhydryl groups of cysteine residues deriving in the cytotoxic effect caused by omeprazole on the trophozoites of G. lamblia, proposing GlTIM as a potential target for drug design (Reyes-Vivas et al., 2014).
García-Torres et al. (2016) tested the effects of PPIs on the activity of GlTIM wild-type. The exposure of this glycolytic enzyme to esomeprazole, pantoprazole, and lansoprazole induced the inactivation of GlTIM in a very similar manner to omeprazole, losing 100% of its activity when these PPIs were incubated at 1 mM. Prominently, the efficacy of rabeprazole is remarkable because the concentration required to com- pletely inactivate GlTIM was only 0.12 mM. The IC50 values of PPIs were approximately 0.30 mM, while the IC50 of rabepra- zole was close to 0.03 mM. Furthermore, human TIM (HsTIM) was resistant to all concentrations of PPIs tested in this study, highlighting that when 50% of GlTIM was lost, HsTIM remai- ned at almost 100% of its original activity (García-Torres et al., 2016). This study provides valuable insights into how struc- turally similar compounds, like omeprazole and its analogs, could potentially be repurposed for targeting conserved proteins such as TIM (target of interest for protozoan infec- tions). While evidence of the activity of PPIs against various protozoa has been observed, further research is necessary. Specifically, modifications to the formulation or pharmaceu- tical presentation of PPIs may be required to enhance their efficacy against protozoan parasites. The widespread use and approval of PPIs for treating acid-related disorders could streamline the development process for therapies targeting protozoan infections, saving both time and resources in the development of new treatments.
Antidiabetics are part of a large group of drugs that are di- rected to reduce polysaccharide degradation and intestinal glucose absorption, decrease hepatic glucose production, or increase insulin secretion directly and indirectly (T. A. Des- hpande et al., 2022). Interestingly, this family has been well studied on different biological activities such as antibacterial (Zhen et al., 2024), antiparasitic (Juárez-Saldivar et al., 2020; Rub et al., 2019; Vázquez et al., 2024), AD (Shi et al., 2023), among others.
Ipragliflozin is an oral antidiabetic drug that belongs to the class of S sodium-glucose co-transporter 2 (SGLT2) inhibitors. It works by inhibiting the SGLT2 protein in the proximal tubule of the kidney, which reduces glucose reab- sorption and increases glucose excretion through the urine. Its primary indication is the treatment of type 2 diabetes
mellitus (T2D) (Alkabbani and Gamble, 2021). Zhen et al. (2024) did in silico and in vitro approaches where ipragliflozin presented anti-Mycobacterium tuberculosis activity. In this study, they predicted Rv1151c (a bifunctional enzyme with both deacetylation and desuccinylation activities, which plays an important role in M. tuberculosis drug resistance and stress responses) crystal structure and identified binding sites based on the druggability score. Once they chose what they called binding site 1, a structure-based virtual screening against predicted Rv1151c structure was carried out in site
1. Ipragliflozin is predicted to bind deeply within binding pocket 1 of the Rv1151c protein. Several residues—including Tyr183, Pro184, Leu155, Phe151, and Trp150—contribute to hydrophobic interactions with the ligand. Additionally, ipra- gliflozin forms one HB with each of the residues Ser179 and Thr178, two HBs with Gln86, as well as two π-π interactions with Phe151 and one π-π interaction with Phe20. MD study showed ipragliflozin and Rv1151c are stable after 55 ns. Fi- nally, in vitro assay against M. tuberculosis H37Rv was made, demonstrating ipragliflozin MIC90 of 0.1 mg/mL in a dose- dependent manner, indicating that ipragliflozin is an anti-M. tuberculosis agent by targeting Rv1151c predicted structure (Zhen et al., 2024).
Glyburide, gliquidone, and glipizide, part of the sulfon- ylureas family (second-generation), were studied against the parasite Trypanosoma cruzi, the ethiological agent of Chagas disease (Gómez-Ochoa et al., 2022). Juárez-Saldivar et al. (2020) carried out a study with an in silico and in vitro approach of FDA-approved drugs against Chagas disease. Ten potential compounds where selected through molecular docking of a 1857 FDA-approved drugs library on the bifunctional enzy- me dihydrofolate reductase-thymidylate synthase (DHFR-TS) of T. cruzi, and tested on epimastigotes, the cultivable form of the parasite. Glypzide showed interactions with amino acids Ala28, Ser83, and Gly156 through HB and forms hydrophobic interactions with Ile41, Phe52, Phe88, and Leu91, Glyburide forms HB with Ala28, Ser83, Ile154, Gly156, and Tyr160, while also interacting hydrophobically with Ile35, Ile41, and Thr80, and Gliquidone interacts with Ala28, Gly156, and Tyr160 through HB, and forms hydrophobic interactions with Ile35 And Thr80, along with π-stacking with Phe52. They found that glipizide, glyburide, and gliquidone presented the lowest free energy of binding against DHFR-TS, and had growth inhibitory activity, with IC50 values of 6, 13.4, 12, and 66 µM, respectively. Authors reported that residues Ile-41 and Phe-52, corresponded to the interaction profile of known DHFR-TS inhibitors (Figure 4), which are present in the three antidiabetics drugs mentioned (Juárez-Saldivar et al., 2020)
More recently, these three sulfonylurea drugs have been tested in combination with benznidazole (Bzn) against T. cruzi. This study showed that in monotherapy, the antidia- betic drugs demonstrated antiparasitic effects by preventing the progression of infection (release of trypomastigotes), with an IC50 ranging from 8.4 to 14.3 µM, compared to Bzn, which had an IC50 of 0.26 µM. However, in combination therapy, the addition of just 0.5 or 1 µM of the antidiabetics reduced the
Figure 4. Three-dimensional structure of Trypanosoma cruzi dihydrofolate reductase-thymidylate synthase and antidiabetic compounds with trypanocidal activity. A) Three-dimensional structure of dihydrofolate reductase-thymidylate synthase. In red, isoleucine 41 and phenylalanine 52 amino acid residues, identified by the authors as important for determining whether a compound has inhibitory potential against the enzyme. B) Antidiabetic compounds that showed activity against the enzyme. From top to bottom: glyburide, gliquidone, and glipizide.
Figura 4. Estructura tridimensional del dihidrofolato reductasa-timidilato sintasa de Trypanosoma cruzi y los antidiabéticos con actividad tripanocida.
A) Estructura tridimensional de dihidrofolato reductasa-timidilato sintasa. En rojo, los residuos isoleucina 41 y fenilalanina 52, señalados por los autores como importantes para determinar si un compuesto presenta potencial de inhibición en la enzima. B) Antidiabéticos que presentaron actividad contra la enzima. De arriba abajo: gliburida, gliquidona y glipzida.
IC50 of Bnz by 5 to 10 times, bringing it down to 0.03–0.05 µM. Notably, when used alone, the antidiabetic drugs re- duced the infection in mice by 40–60%, similar to the 80% reduction observed with Bnz. Furthermore, the combination of Bnz and antidiabetics altered the antioxidant metabolites in epimastigotes. Glyburide and gliquidone alone decreased the levels of glutathione and trypanothione and increased those of cysteine, while glipizide alone increased the level of trypanothione and decreased the levels of cysteine and glutathione. The changes in these antioxidant metabolites suggested perturbation of the antioxidant system in the parasite (Vázquez et al., 2024). This research highlights the importance of continuing the investigation of previously re- ported drugs with activity, as well as the consideration that, as observed in the parasite’s antioxidant system, potential repurposed drugs may have multiple targets. Therefore, en- zymes of the T. cruzi antioxidant system, such as glutathione synthetase, trypanothione synthetase, and trypanothione reductase, could be evaluated in conjunction with DHFR-TS.
Also, glyburide showed activity against the parasite Leishmania donovani using an in vitro and in silico approach. Molecular docking was conducted against Leishmania donovani-Trypanothione synthetase (LdTrySyn), a critical enzyme for the parasite’s survival as it detoxifies nitric oxi- de species produced by the host’s immune system. Results showed that glyburide interacts effectively with key residues in the LdTrySyn’s catalytic site as Asp348, Ser349, and Arg553, and in vitro growth reversibility assay shows that growth of treated parasite was not reversed when transferred to fresh culture media after 7 days, highlighting LdTrySyn as a drug target against L. donovani (Rub et al., 2019).
Metformin, derived from a plant named Galena offi- cinalis, is currently widely used as a first-line drug for the treatment of T2D. Some studies have proved the effect of metformin on the prophylaxis and treatment of neurop- sychiatric degenerative diseases in addition to its significant hypoglycemic effects, especially in patients with T2D (Orkaby et al., 2017). Shi et al. proposed an in silico approach to esta- blish a relationship between T2D and AD. From 89 targets, catalase presented the best free binding energy with met- formin. Metformin was predicted to interact with Glu330 and Ser120 twice. The cat-metformin complex was selected for 50 ns MD simulation and system stability analysis. The CAT- metformin complex was relatively stable at around 30 ns, the RMSD fluctuates at 0.2 nm (2 Å), and the stability was given by Asp259 amino acid residue through three HBs (Shi et al., 2023). However, these results are just based on computer simulations and big data analysis, which need to be further verified by cell or animal experiments.
In this review, case studies of NSAIDs, such as ASA and ibu- profen, illustrate the potential of repositioning well-known drugs for treating diseases beyond their original indications. For example, ASA has demonstrated antiviral properties, whi- le ibuprofen has shown promise in treating chronic infections, and inflammation in cystic fibrosis. These findings emphasize the importance of combining computational, experimental, and clinical approaches in the repositioning process, offering valuable insights into the broader therapeutic potential of these drugs.
Similarly, sildenafil’s unexpected shift from a hyperten- sion treatment to a successful therapeutic option for erectile dysfunction and pulmonary hypertension exemplifies how serendipitous discoveries can significantly impact drug repurposing strategies. This further illustrates the vast untap- ped potential within the pharmacological profiles of already approved drugs.
The ongoing repurposing of PPIs and antidiabetic drugs highlights their promising potential as therapeutic agents against parasitic infections. PPIs, traditionally used to treat acid-related disorders, have shown significant antiprotozoal activity, particularly against Trichomonas vaginalis, Giardia intestinalis, and Entamoeba histolytica. The observed efficacy of these compounds, notably pantoprazole, against various anaerobic protozoa, along with their ability to specifically tar- get parasite enzymes like triose phosphate isomerase (TIM), positions them as valuable candidates for further investi- gation in parasitic drug development. However, optimizing their formulation and pharmaceutical presentation remains a crucial step in enhancing their therapeutic potential.
Similarly, antidiabetic drugs such as ipragliflozin, sul- fonylureas (glipizide, glyburide, and gliquidone), and met- formin have demonstrated significant antiparasitic effects. Ipragliflozin’s activity against Mycobacterium tuberculosis, along with sulfonylureas’ ability to inhibit key enzymes in
T. cruzi and L. donovani, underscores the broad-spectrum potential of these drugs. Notably, the combination of these drugs with established therapies like Bzn has been shown to improve treatment outcomes, indicating the value of re- purposing these agents. Furthermore, metformin’s potential in treating neurodegenerative diseases such as AD adds another layer to its therapeutic utility, particularly in the context of its established role in managing type 2 diabetes.
The concept of drug repositioning has evolved signi- ficantly with the integration of advanced computational tools and omics technologies, making it a valuable approach for discovering new therapeutic uses for existing drugs. By leveraging the extensive safety and pharmacological profiles of approved drugs, repositioning provides a faster and more cost-effective alternative to traditional drug development. This strategy has shown considerable promise in various the- rapeutic areas, including oncology, infectious diseases, and neurodegenerative disorders. However, challenges remain, particularly in identifying novel therapeutic targets and validating computational predictions through experimental methods.
Acknowledgments to Secretaría de Investigación y Posgrado del Instituto Politécnico Nacional (SIP-20250835) and SECIHTI for scholarship.
The authors declare that there are no conflicts of interest.
Alkabbani, W. ane Gamble, J.M. 2021. Profile of ipragliflozin, an oral SGLT-2 inhibitor for the treatment of type 2 diabetes: The evidence to date. Drug Design, Development and Therapy, 15, 3057–3069. https://doi.org/10.2147/DDDT.S281602
Borba-Santos, L.P., Nucci, M., Ferreira-Pereira, A. and Rozental, S. 2021. Anti-sporothrix activity of ibuprofen combined with antifungal. Brazilian Journal of Microbiology, 52(1), 101. https://doi.org/10.1007/S42770-020-00327-9
Chaffey, L., Roberti, A. and Greaves, D.R. 2022. Drug repurposing in cardiovascular inflammation: Successes, failures, and future opportunities. Frontiers in Pharmacology, 13. https:// doi.org/10.3389/FPHAR.2022.1046406
Chen, L., Liu, Y., Becher, A., DIepold, K., Schmid, E., Fehn, A., Brunner, C., Rouhi, A., Chiosis, G., Cronauer, M., Seufferlein,
T. and Azoitei, N. 2020. Sildenafil triggers tumor lethality through altered expression of HSP90 and degradation of PKD2. Carcinogenesis, 41(10), 1421–1431. https://doi. org/10.1093/CARCIN/BGAA001
Chow, J.H., Khanna, A.K., Kethireddy, S., Yamane, D., Levine, A.,
Jackson, A.M., McCurdy, M.T., Tabatabai, A., Kumar, G., Park, P., Benjenk, I., Menaker, J., Ahmed, N., Glidewell, E., Presutto, E., Cain, S., Haridasa, N., Field, W., Fowler, J.G., … Mazzeffi, M.A. 2021. Aspirin use is associated with decreased mechanical ventilation, intensive care unit admission, and in-hospital mortality in hospitalized patients with coronavirus disease 2019. Anesthesia and Analgesia, 132(4), 930–941. https:// doi.org/10.1213/ANE.0000000000005292
Cofer, L.B., Barrett, T.J. and Berger, J.S. 2022. Aspirin for the primary prevention of cardiovascular disease: Time for a platelet- guided approach. Arteriosclerosis, Thrombosis, and Vascular Biology, 42(10), 1207–1216. https://doi.org/10.1161/ ATVBAHA.122.318020/ASSET/98D940F5-9138-47EB-8383- 2B11D24F3914/ASSETS/GRAPHIC/ATVBAHA.122.318020. FIG05.JPG
Das, A., Durrant, D., Mitchell, C., Mayton, E., Hoke, N.N.,
Salloum, F.N., Park, M.A., Qureshi, I., Lee, R., Dent, P. and Kukreja, R.C. 2010. Sildenafil increases chemotherapeutic efficacy of doxorubicin in prostate cancer and ameliorates cardiac dysfunction. Proceedings of the National Academy of Sciences of the United States of America, 107(42), 18202–18207. https://doi.org/10.1073/PNAS.1006965107/-/ DCSUPPLEMENTAL/PNAS.201006965SI.PDF
Deshpande, R.R., Tiwari, A.P., Nyayanit, N. and Modak, M. 2020. In silico molecular docking analysis for repurposing therapeutics against multiple proteins from SARS-CoV-2. European Journal of Pharmacology, 886. https://doi. org/10.1016/J.EJPHAR.2020.173430
Deshpande, T.A., Martínez-Málaga, J. and Priefer, R. 2022. Dithiocarbamates as potential PTP1B inhibitors for diabetes management. Results in Chemistry, 4, 100372. https://doi. org/10.1016/J.RECHEM.2022.100372
Devillers, J. (2018). Repurposing drugs for use against Zika virus infection. SAR and QSAR in Environmental Research, 29(2), 103–115. https://doi.org/10.1080/1062936X.2017.1411642
Fang, J., Zhang, P., Zhou, Y., Chiang, C.W., Tan, J., Hou, Y., Stauffer, S., Li, L., Pieper, A.A., Cummings, J. and Cheng, F. 2021. Endophenotype-based in silico network medicine discovery combined with insurance record data mining identifies sildenafil as a candidate drug for Alzheimer’s disease. Nature Aging, 1(12), 1175–1188. https://doi.org/10.1038/S43587- 021-00138-Z
Fernández-Sánchez, S.Y., Cerón-Carrasco, J.P., Risco, C. and Fernández de Castro, I. 2023. Antiviral activity of acetylsalicylic acid against Bunyamwera virus in cell culture. Viruses, 15(4), 948. https://doi.org/10.3390/V15040948/S1
Fijałkowski, Ł., Skubiszewska, M., Grześk, G., Koech, F.K. and Nowaczyk, A. 2022. Acetylsalicylic acid–primus inter pares in pharmacology. Molecules, 27(23), 8412. https://doi. org/10.3390/MOLECULES27238412
García-Torres, I., De La Mora-De La Mora, I., Marcial-Quino, J., Gómez-Manzo, S., Vanoye-Carlo, A., Navarrete-Vázquez, G., Colín-Lozano, B., Gutiérrez-Castrellón, P., Sierra-Palacios, E., López-Velázquez, G. and Enríquez-Flores, S. 2016. Proton pump inhibitors drastically modify triosephosphate isomerase from Giardia lamblia at functional and structural levels, providing molecular leads in the design of new antigiardiasic drugs. Biochimica et Biophysica Acta, 1860(1 Pt A), 97–107. https://doi.org/10.1016/J.BBAGEN.2015.10.021
Ghofrani, H.A., Osterloh, I.H. and Grimminger, F. 2006. Sildenafil: from angina to erectile dysfunction to pulmonary hypertension and beyond. Nature Reviews. Drug Discovery, 5(8), 689. https://doi.org/10.1038/NRD2030
Glatthaar-Saalmüller, B., Mair, K.H. and Saalmüller, A. 2017. Antiviral activity of aspirin against RNA viruses of the respiratory tract—an in vitro study. Influenza and Other Respiratory Viruses, 11(1), 85–92. https://doi.org/10.1111/ IRV.12421
Gohel, D., Zhang, P., Gupta, A.K., Li, Y., Chiang, C.W., Li, L., Hou, Y., Pieper, A.A., Cummings, J. and Cheng, F. 2024. Sildenafil as a candidate drug for Alzheimer’s disease: Real-world patient data observation and mechanistic observations from patient-induced pluripotent stem cell-derived neurons. Journal of Alzheimer’s Disease: JAD, 98(2), 643–657. https:// doi.org/10.3233/JAD-231391
Gómez-Ochoa, S.A., Rojas, L.Z., Echeverría, L.E., Muka, T. and Franco, O.H. 2022. Global, regional, and national trends of Chagas disease from 1990 to 2019: Comprehensive analysis of the global burden of disease study. Global Heart, 17(1). https://doi.org/10.5334/GH.1150
Guo, C.G., Ma, W., Drew, D.A., Cao, Y., Nguyen, L.H., Joshi, A.D.,
Ng, K., Ogino, S., Meyerhardt, J.A., Song, M., Leung, W.K., Giovannucci, E.L. and Chan, A.T. 2021. Aspirin use and risk of colorectal cancer among older adults. JAMA Oncology, 7(3),
1. https://doi.org/10.1001/JAMAONCOL.2020.7338 Juárez‐Saldivar, A., Barbosa‐Cabrera, E., Lara‐Ramírez, E.E., Paz‐
González, A.D., Martínez‐Vázquez, A.V., Bocanegra‐García, V., Palos, I., Campillo, N.E. and Rivera, G. 2021. Virtual screening of FDA‐approved drugs against triose phosphate isomerase from Entamoeba histolytica and Giardia lamblia identifies inhibitors of their trophozoite growth phase. International Journal of Molecular Sciences, 22(11), 5943. https://doi. org/10.3390/IJMS22115943/S1
Juárez-Saldivar, A., Schroeder, M., Salentin, S., Joachim Haupt, V., Saavedra, E.,Vázquez, C., Reyes-Espinosa, F., Herrera-Mayorga, V., Villalobos-Rocha, J.C., García-Pérez, C.A., Campillo, N.E. and Rivera, G. 2020. Computational drug Repositioning for Chagas disease using protein-ligand interaction profiling. International Journal of Molecular Sciences, 21(12), 4270. https://doi.org/10.3390/IJMS21124270
Kaur, D., Choudhury, C., Yadav, R., Kumari, L. and Bhatia, A. 2024. Aspirin as a potential drug repurposing candidate targeting estrogen receptor alpha in breast cancer: a molecular
dynamics and in-vitro study. Journal of Biomolecular Structure and Dynamics, 1–12. Advance online publication. https://doi.org/10.1080/07391102.2024.2308780
Lands, L.C. and Stanojevic, S. 2016. Oral non-steroidal anti- inflammatory drug therapy for lung disease in cystic fibrosis. The Cochrane Database of Systematic Reviews, 4, CD001505. https://doi.org/10.1002/14651858.CD001505.PUB4
Liu, Q., Huang, N., Li, A., Zhou, Y., Liang, L., Song, X., Yang, Z. and Zhou, X. 2021. Effect of low-dose aspirin on mortality and viral duration of the hospitalized adults with COVID-19. Medicine, 100(6), E24544. https://doi.org/10.1097/ MD.0000000000024544
Meizlish, M.L., Goshua, G., Liu, Y., Fine, R., Amin, K., Chang, E., DeFilippo, N., Keating, C., Liu, Y., Mankbadi, M., McManus, D., Wang, S.Y., Price, C., Bona, R.D., Ochoa Chaar, C.I., Chun,
H.J., Pine, A.B., Rinder, H.M., Siner, J.M., … Lee, A.I. 2021.
Intermediate‐dose anticoagulation, aspirin, and in‐hospital mortality in COVID‐19: A propensity score‐matched analysis. American Journal of Hematology, 96(4), 471. https://doi. org/10.1002/AJH.26102
Merzon, E., Green, I., Vinker, S., Golan-Cohen, A., Gorohovski, A., Avramovich, E., Frenkel-Morgenstern, M. and Magen, E. 2021. The use of aspirin for primary prevention of cardiovascular disease is associated with a lower likelihood of COVID‐19 infection. The Febs Journal, 288(17), 5179. https://doi. org/10.1111/FEBS.15784
Morsi, D.S., Barnawi, I.O., Ibrahim, H.M., El-Morsy, A.M., El Hassab,
M.A. and Abd El Latif, H.M. 2023. Immunomodulatory, apoptotic and anti-proliferative potentials of sildenafil in Ehrlich ascites carcinoma murine model: In vivo and in silico insights. International Immunopharmacology, 119, 110135. https://doi.org/10.1016/J.INTIMP.2023.110135
Onódi, Z., Koch, S., Rubinstein, J., Ferdinandy, P. and Varga, Z.V. 2023. Drug repurposing for cardiovascular diseases: New targets and indications for probenecid. British Journal of Pharmacology, 180(6), 685–700. https://doi.org/10.1111/ BPH.16001
Orkaby, A.R., Cho, K., Cormack, J., Gagnon, D.R. and Driver,
J.A. 2017. Metformin vs sulfonylurea use and risk of dementia in US veterans aged ≥65 years with diabetes. Neurology, 89(18), 1877–1885. https://doi.org/10.1212/ WNL.0000000000004586
Osborne, T.F., Veigulis, Z.P., Arreola, D.M., Mahajan, S.M., Roosli,
E. and Curtin, C.M. 2021. Association of mortality and aspirin prescription for COVID-19 patients at the Veterans Health Administration. PLoS ONE, 16(2), e0246825. https://doi. org/10.1371/JOURNAL.PONE.0246825
Patrono, C. and Rocca, B. 2021. Aspirin at 120: Retiring, recombining, or repurposing? Research and Practice in Thrombosis and Haemostasis, 5(4). https://doi.org/10.1002/ RTH2.12516
Pérez-Villanueva, J., Romo-Mancillas, A., Hernández-Campos, A., Yépez-Mulia, L., Hernández-Luis, F. and Castillo, R. 2011. Antiprotozoal activity of proton-pump inhibitors. Bioorganic & Medicinal Chemistry Letters, 21(24), 7351–7354. https:// doi.org/10.1016/J.BMCL.2011.10.028
Pires, D., Valente, S., Calado, M., Mandal, M., Azevedo-Pereira, J.M. and Anes, E. 2021. Repurposing saquinavir for host-directed therapy to control Mycobacterium tuberculosis infection. Frontiers in Immunology, 12. https://doi.org/10.3389/ FIMMU.2021.647728
Pizzorno, A., Padey, B., Terrier, O. and Rosa-Calatrava, M. 2019. Drug repurposing approaches for the treatment of influenza
viral infection: Reviving old drugs to fight against a long- lived enemy. Frontiers in Immunology, 10(MAR). https://doi. org/10.3389/FIMMU.2019.00531
Qaderdan, K., Ishak, M., Heestermans, A.A.C.M., De Vrey, E., Jukema, J.W., Voskuil, M., De Boer, M.J., Van’T Hof, A.W.J., Groenemeijer, B.E., Vos, G.J.A., Janssen, P.W.A., Bergmeijer, T.O., Kelder, J.C., Deneer, V.H.M. and Ten Berg, J.M. 2015. Ticagrelor or prasugrel versus clopidogrel in elderly patients with an acute coronary syndrome: Optimization of antiplatelet treatment in patients 70 years and older— rationale and design of the POPular AGE study. American Heart Journal, 170(5), 981–985.e1. https://doi.org/10.1016/J. AHJ.2015.07.030
Rashid, R.S.M., Temurlu, S., Abourajab, A., Karsili, P., Dinleyici, M., Al-Khateeb, B. and Icil, H. 2023. Drug repurposing of FDA compounds against α-glucosidase for the treatment of type 2 diabetes: Insights from molecular docking and molecular dynamics simulations. Pharmaceuticals (Basel, Switzerland),
16(4). https://doi.org/10.3390/PH16040555
Reyes-Vivas, H., De La Mora-De La Mora, I., Castillo-Villanueva, A., Yépez-Mulia, L., Hernández-Alcántara, G., Figueroa-Salazar, R., García-Torres, I., Gómez-Manzo, S., Méndez, S.T., Vanoye- Carlo, A., Marcial-Quino, J., Torres-Arroyo, A., Oria-Hernández, J., Gutiérrez-Castrellón, P., Enríquez-Flores, S. and López- Velázquez, G. 2014. Giardial triosephosphate isomerase as possible target of the cytotoxic effect of omeprazole in Giardia lamblia. Antimicrobial Agents and Chemotherapy, 58(12), 7072. https://doi.org/10.1128/AAC.02900-14
Rub, A., Shaker, K., Kashif, M., Arish, M., Dukhyil, A.A.B., Alshehri, B.M., Alaidarous, M.A., Banawas, S. and Amir, K. 2019. Repurposing glyburide as antileishmanial agent to fight against leishmaniasis. Protein and Peptide Letters, 26(5), 371–376. https://doi.org/10.2174/09298665266661903011
14012
Saqallah, F.G., Al-Najjar, B.O., Al-Kabariti, A.Y. and Abbas, M.A. 2024. Novel acridone derivatives as potential P2Y12 receptor inhibitors: integrating computational modeling and experimental analysis. Journal of Biomolecular Structure and Dynamics, 1–10. Advance online publication. https:// doi.org/10.1080/07391102.2024.2444421
Savai, R., Pullamsetti, S.S., Banat, G.A., Weissmann, N., Ghofrani, H.A., Grimminger, F. and Schermuly, R.T. 2010. Targeting cancer with phosphodiesterase inhibitors. Expert Opinion on Investigational Drugs, 19(1), 117–131. https://doi. org/10.1517/13543780903485642
Shah, P.N., Marshall-Batty, K.R., Smolen, J.A., Tagaev, J.A., Chen,
Q., Rodesney, C.A., Le, H.H., Gordon, V.D., Greenberg, D.E. and Cannon, C.L. 2018. Antimicrobial activity of ibuprofen against cystic fibrosis-associated gram-negative pathogens. Antimicrobial Agents and Chemotherapy, 62(3). https://doi. org/10.1128/AAC.01574-17
Shi, X., Li, L., Liu, Z., Wang, F. and Huang, H. 2023. Exploring the mechanism of metformin action in Alzheimer’s disease and type 2 diabetes based on network pharmacology, molecular
docking, and molecular dynamic simulation. Therapeutic Advances in Endocrinology and Metabolism, 14. https://doi. org/10.1177/20420188231187493
Shureiqi, I. 2022. Aspirin for colorectal cancer prevention: Age matters. Cancer Prevention Research, 15(9), 565–567. https://doi.org/10.1158/1940-6207.CAPR-22-0176
Sobhani, K., Li, J. and Cortes, M. 2023. Nonsteroidal anti- inflammatory drugs (NSAIDs). First aid perioperative ultrasound: Acute pain manual for surgical procedures, 127–138. https://doi.org/10.1007/978-3-031-21291-8_8
Tantry, U.S., Schror, K., Navarese, E.P., Jeong, Y.H., Kubica, J., Bliden, K.P. and Gurbel, P.A. 2021. Aspirin as an adjunctive pharmacologic therapy option for COVID-19: Anti- inflammatory, antithrombotic, and antiviral effects all in one agent. Journal of Experimental Pharmacology, 13, 957. https://doi.org/10.2147/JEP.S330776
Ter Horst, S., Conceição-Neto, N., Neyts, J. and Rocha-Pereira, J. 2019. Structural and functional similarities in bunyaviruses: Perspectives for pan-bunya antivirals. Reviews in Medical Virology, 29(3), e2039. https://doi.org/10.1002/RMV.2039
Vázquez, C., Encalada, R., Jiménez-Galicia, I., Gómez-Escobedo, R., Rivera, G., Nogueda-Torres, B. and Saavedra, E. 2024. Repurposing the antidiabetic drugs glyburide, gliquidone, and glipizide in combination with benznidazole for Trypanosoma cruzi infection. Pharmaceuticals (Basel, Switzerland), 18(1). https://doi.org/10.3390/PH18010021
Veljkovic, V., Glisic, S., Muller, C.P., Scotch, M., Branch, D.R., Perovic, V.R., Sencanski, M., Veljkovic, N. and Colombatti, A. 2015. In silico analysis suggests interaction between Ebola virus and the extracellular matrix. Frontiers in Microbiology, 6(FEB). https://doi.org/10.3389/FMICB.2015.00135
Veljkovic, V., Goeijenbier, M., Glisic, S., Veljkovic, N., Perovic, V.R., Sencanski, M., Branch, D.R. and Paessler, S. 2015. In silico analysis suggests repurposing of ibuprofen for prevention and treatment of EBOLA virus disease. F1000Research, 4,
104. https://doi.org/10.12688/F1000RESEARCH.6436.1 Wong, M.W., Yi, C.H., Liu, T.T., Lei, W.Y., Hung, J.S., Liu, C.Z. and
Chen, C.L. 2021. Effects of phosphodiesterase-5 inhibitor sildenafil on esophageal secondary peristalsis: Studies with high-resolution manometry. Journal of Gastroenterology and Hepatology, 36(2), 430–435. https://doi.org/10.1111/ JGH.15170
Xia, Y., Sun, M., Huang, H. and Jin, W.L. 2024. Drug repurposing for cancer therapy. Signal Transduction and Targeted Therapy, 9(1). https://doi.org/10.1038/S41392-024-01808-1
Zhen, J., Zhang, C., Huang, T., Xie, L., Yan, Y., Yan, S., Zhang, J., Huang, H. and Xie, J. 2024. Drug repurposing: An antidiabetic drug Ipragliflozin as Mycobacterium tuberculosis sirtuin-like protein inhibitor that synergizes with anti-tuberculosis drug isoniazid. International Journal of Biological Macromolecules, 282, 137003. https://doi.org/10.1016/J. IJBIOMAC.2024.137003