Drug Repositioning

Drug Repurposing in Pancreatic Cancer: A Multi-Stakeholder Perspective to Improve Treatment Options for Pancreatic Cancer Patients

Thu, 2025-03-06 06:00

Cancer Manag Res. 2025 Mar 1;17:429-440. doi: 10.2147/CMAR.S483151. eCollection 2025.

ABSTRACT

Pancreatic cancer (PC) remains one of the most challenging malignancies to treat. Current therapeutic options are unsatisfactory, and there is an urgent need for more effective and less toxic drugs to improve the dismal prognosis of PC. In recent years, drug repurposing (DR) has emerged as an attractive strategy to identify novel treatments for PC by leveraging existing drugs approved for other indications. Through the use of electronic medical records, Artificial Intelligence, study of metabolic pathways, signalling pathways, and many other approaches, it has become much easier in recent years to identify potential novel uses for old drugs. Although policy, funding and research attention in this area are steadily growing, major challenges to efficient and effective patient-centric DR in PC need to be addressed. These include but are not limited to regulatory, financial and funding barriers and the lack of coordination and collaboration among several sectors and stakeholders. To explore the opportunities and challenges associated with DR in PC, a one-day multi-stakeholder meeting was held on 14th of November 2024 in Brussels, Belgium as part of the REMEDi4ALL project. This meeting provided a platform for researchers, clinicians, industry representatives, funders, regulatory experts, and patient advocates to discuss and propose actions to optimize and accelerate DR in PC. Insights from this meeting support the potential of DR to enhance PC treatment options while highlighting the importance of systemic and supportive changes in the regulatory, policy and funding landscapes, interdisciplinary collaboration, data sharing, and patient involvement in driving therapeutic innovation. This summary highlights key outcomes and recommendations from the meeting in informing future efforts to advance DR initiatives in the context of PC.

PMID:40046652 | PMC:PMC11881603 | DOI:10.2147/CMAR.S483151

Categories: Literature Watch

An Attention-Aware Multi-Task Learning Framework Identifies Candidate Targets for Drug Repurposing in Sarcopenia

Thu, 2025-03-06 06:00

J Cachexia Sarcopenia Muscle. 2025 Apr;16(2):e13661. doi: 10.1002/jcsm.13661.

ABSTRACT

BACKGROUND: Sarcopenia presents a pressing public health concern due to its association with age-related muscle mass decline, strength loss and reduced physical performance, particularly in the growing older population. Given the absence of approved pharmacological therapies for sarcopenia, the need to discover effective pharmacological interventions has become critical.

METHODS: To address this challenge and discover new therapies, we developed a novel Multi-Task Attention-aware method for Multi-Omics data (MTA-MO) to extract complex biological insights from various biomedical data sources, including transcriptome, methylome and genome data to identify drug targets and discover new therapies. Additionally, MTA-MO integrates human protein-protein interaction (PPI) networks and drug-target networks to improve target identification. The novel method is applied to a multi-omics dataset that included 1055 participants aged 20-50 (mean (± SD) age 36.88 (± 8.64)), comprising 37.82% African-American and 62.18% Caucasian/White individuals. Physical activity levels were self-reported and categorized into three groups: ≥ 3 times/week, < 3 times/week and no regular exercise. Mean (± SD) measures for grip strength, appendicular lean mass (ALM), exercise frequency and smoking status (no/yes, n (%)) were 38.72 (± 8.93) kg, 28.65 (± 4.63) kg, 4.31 (± 1.79) and 30.81%/69.19%, respectively. Significant differences (p < 0.05) were found between groups in age, ALM, smoking, and consumption of milk, alcohol, beer and wine.

RESULTS: Using the MTA-MO method, we identified 639 gene targets, and by analysing PPIs and querying public databases, we narrowed this list down to seven potential hub genes associated with sarcopenia (ESR1, ATM, CDC42, EP300, PIK3CA, EGF and PTK2B). These findings were further validated through diverse levels of pathobiological evidence associated with sarcopenia. Gene Ontology and KEGG pathways analysis highlighted five key functions and signalling pathways relevant to skeletal muscle. The interaction network analysis identified three transcriptional factors (GATA2, JUN and FOXC1) as the key transcriptional regulators of the seven potential genes. In silico analysis of 1940 drug candidates identified canagliflozin as a promising candidate for repurposing in sarcopenia, demonstrating the strongest binding affinity to the PTK2B protein (inhibition constant 6.97 μM). This binding is stabilized by hydrophobic bonds, Van der Waals forces, pi-alkyl interactions and pi-anion interactions around PTK2B's active residues, suggesting its potential as a therapeutic option.

CONCLUSIONS: Our novel approach effectively integrates multi-omics data to identify potential treatments for sarcopenia. The findings suggest that canagliflozin could be a promising therapeutic candidate for sarcopenia.

PMID:40045692 | DOI:10.1002/jcsm.13661

Categories: Literature Watch

Leishmaniases: Strategies in treatment development

Wed, 2025-03-05 06:00

Parasite. 2025;32:18. doi: 10.1051/parasite/2025009. Epub 2025 Mar 5.

ABSTRACT

Leishmaniases are vector-borne parasitic diseases that pose a threat to over 1 billion people worldwide. The parasites target cells of the reticulohistiocytic system, such as macrophages, where they replicate. The disease manifests in various forms, ranging from localized cutaneous leishmaniasis to life-threatening visceral forms, which are fatal in 95% of cases without treatment. Current treatments rely on the invasive administration of toxic and expensive drugs that are increasingly encountering resistance. Therefore, finding alternative treatments for this disease is imperative. This literature review focuses on recent advancements in alternative treatments and aims to present the various strategies designed to address current limitations, including cost, toxicity, off-target effects, administration routes, and the emergence of drug resistance. Starting with an overview of the existing approved treatments and their specific limitations, we categorize treatment development strategies into five key sections: (i) combination therapies using existing approved treatments to enhance efficacy and reduce resistance; (ii) nanoparticle formulations, which enable targeted delivery to infected organs and improved therapeutic efficiency; (iii) drug repositioning, a strategy that has already contributed to the approval of over half of current therapeutic compounds; (iv) immunomodulation, used in conjunction with standard chemotherapies to enhance treatment efficacy and lower relapse rates; and (v) ethnobotanicals, which have demonstrated promising in vitro results by combining low toxicity, immunomodulatory properties, and potent anti-parasitic effects. In summary, this review outlines current strategies in treatment development, emphasizing their advantages over conventional therapies while acknowledging their limitations.

PMID:40043198 | DOI:10.1051/parasite/2025009

Categories: Literature Watch

Comparative in vitro and in silico evaluation of the toxic effects of metformin and/or ascorbic acid, new treatment options in the treatment of Melasma

Wed, 2025-03-05 06:00

Toxicol Res (Camb). 2025 Feb 27;14(1):tfaf025. doi: 10.1093/toxres/tfaf025. eCollection 2025 Feb.

ABSTRACT

Melasma is a chronic condition that leads to the buildup of melanin pigment in the epidermis and dermis due to active melanocytes. Even though it is considered a non-life-threatening condition, pigment disorders have a negative impact on quality of life. Since melasma treatment is not sufficient and complicated, new treatment options are sought. Research on metformin and ascorbic acid suggested that they might be used against melasma in the scope of "drug repositioning."The MNT-1 human melanoma cell line was used to assess the effects of metformin, ascorbic acid, and metformin+ascorbic acid combination on cytotoxicity and oxidative stress. Melanin, cAMP, L-3,4-dihydroxyphenylalanine (L-DOPA) and tyrosinase levels were determined by commercial ELISA kits and tyrosinase gene expression was analyzed with RT-qPCR. Cytopathological evaluations were performed by phase contrast microscopy. Tyrosinase expression was determined by immunofluorescence (IF) staining of MNT-1 cells. The online service TargetNet was used for biological target screening. The parameters were not significantly altered by ascorbic acid applied at non-cytotoxic concentrations. On the contrary, metformin dramatically raised tyrosinase and intracellular ROS levels. Moreover, intracellular ROS levels and tyrosinase levels were found to be considerably elevated with the combined treatment. Also, potential metformin and ascorbic acid interactions were determined. According to the results, it can be said that these parameters were not significantly altered by ascorbic acid. On the contrary, metformin dramatically raised tyrosinase and intracellular oxidative stress levels. Moreover, intracellular oxidative stress and tyrosinase levels were elevated with the combined treatment. In conclusion, individual treatments of ascorbic acid or metformin may only provide a limited effect when treating melasma and extensive in vitro and in vivo research are required.

PMID:40040652 | PMC:PMC11878769 | DOI:10.1093/toxres/tfaf025

Categories: Literature Watch

Drug-Target Interaction Prediction via Deep Multimodal Graph and Structural Learning

Wed, 2025-03-05 06:00

Annu Int Conf IEEE Eng Med Biol Soc. 2024 Jul;2024:1-7. doi: 10.1109/EMBC53108.2024.10782657.

ABSTRACT

Drug-target interaction (DTI) prediction speeds up drug repurposing, accelerates drug screening, and reduces drug design timeframe. Previous DTI prediction frameworks lack consideration for the multimodal nature of DTI, advanced feature representation techniques, and generalizability on unseen drugs and proteins. Therefore, we propose a novel framework that combines a multimodal graph neural network with direct, molecular-level structural learning via model ensembling. We use a multimodal biomedical that contains drugs, proteins, diseases, and pathways, all of which have meaningful feature embeddings generated via language models or knowledge graphs. We also employ a structural learning module that exploits molecular-level information and runs independently from the graph. Lastly, the graph and structural modules are combined, forming the optimal prediction. Our proposed framework outperformed multiple benchmark DTI frameworks on real-world datasets. After testing on an independent dataset, we conclude our framework is generalizable to unseen drugs and proteins. Our model can be easily extended to other biomedical link prediction problems, such as drug-drug interaction.

PMID:40040060 | DOI:10.1109/EMBC53108.2024.10782657

Categories: Literature Watch

Old drugs, new challenges: reassigning drugs for cancer therapies

Tue, 2025-03-04 06:00

Cell Mol Biol Lett. 2025 Mar 5;30(1):27. doi: 10.1186/s11658-025-00710-0.

ABSTRACT

The "War on Cancer" began with the National Cancer Act of 1971 and despite more than 50 years of effort and numerous successes, there still remains much more work to be done. The major challenge remains the complexity and intrinsic polygenicity of neoplastic diseases. Furthermore, the safety of the antitumor therapies still remains a concern given their often off-target effects. Although the amount of money invested in research and development required to introduce a novel FDA-approved drug has continuously increased, the likelihood for a new cancer drug's approval remains limited. One interesting alternative approach, however, is the idea of repurposing of old drugs, which is both faster and less costly than developing new drugs. Repurposed drugs have the potential to address the shortage of new drugs with the added benefit that the safety concerns are already established. That being said, their interactions with other new drugs in combination therapies, however, should be tested. In this review, we discuss the history of repurposed drugs, some successes and failures, as well as the multiple challenges and obstacles that need to be addressed in order to enhance repurposed drugs' potential for new cancer therapies.

PMID:40038587 | DOI:10.1186/s11658-025-00710-0

Categories: Literature Watch

Inhibition of the STAT3/Fanconi anemia axis is synthetic lethal with PARP inhibition in breast cancer

Tue, 2025-03-04 06:00

Nat Commun. 2025 Mar 4;16(1):2159. doi: 10.1038/s41467-025-57476-4.

ABSTRACT

The targeting of cancer stem cells (CSCs) has proven to be an effective approach for limiting tumor progression, thus necessitating the identification of new drugs with anti-CSC activity. Through a high-throughput drug repositioning screen, we identify the antibiotic Nifuroxazide (NIF) as a potent anti-CSC compound. Utilizing a click chemistry strategy, we demonstrate that NIF is a prodrug that is specifically bioactivated in breast CSCs. Mechanistically, NIF-induced CSC death is a result of a synergistic action that combines the generation of DNA interstrand crosslinks with the inhibition of the Fanconi anemia (FA) pathway activity. NIF treatment mimics FA-deficiency through the inhibition of STAT3, which we identify as a non-canonical transcription factor of FA-related genes. NIF induces a chemical HRDness (Homologous Recombination Deficiency) in CSCs that (re)sensitizes breast cancers with innate or acquired resistance to PARP inhibitor (PARPi) in patient-derived xenograft models. Our results suggest that NIF may be useful in combination with PARPi for the treatment of breast tumors, regardless of their HRD status.

PMID:40038300 | DOI:10.1038/s41467-025-57476-4

Categories: Literature Watch

DD-HGNN: Drug-Disease Association Prediction Via General Hypergraph Neural Network With Hierarchical Contrastive Learning and Cross Attention Learning

Tue, 2025-03-04 06:00

IEEE J Biomed Health Inform. 2025 Feb 21;PP. doi: 10.1109/JBHI.2025.3542784. Online ahead of print.

ABSTRACT

The research on identifying drug-disease associations (DDAs) is widely used in scenarios such as drug development, clinical decision-making, and drug repurposing, holding significant biological and medical significance. Existing methods for drug-disease association prediction have achieved decent performance, they primarily rely on simplistic drug-disease association graphs or similarity graphs. These methods often struggle to capture the high-order correlations of complex multimodal data, limiting their ability to handle the complexity of data associations effectively. In addition, real drug-disease associations are highly sparse, posing a significant challenge to prediction accuracy. To tackle these issues, we propose a general hypergraph neural network framework for drug-disease association prediction based on hierarchical contrastive learning and cross-attention learning. It leverages hypergraph neural networks to learn representations of drugs and diseases carrying high-order correlations and strengthens representation quality using interactive attention learning and hierarchical contrastive learning. Meanwhile, the -weighted loss function is utilized to adapt to the high sparsity property of real drug-disease associations during model training and improve prediction performance. Extensive experiments demonstrate that DD-HGNN surpasses other state-of-the-art methods in predicting drug-disease associations and further validation through case studies on Leukemia and Colorectal Neoplasms underscores its reliability.

PMID:40036538 | DOI:10.1109/JBHI.2025.3542784

Categories: Literature Watch

Identification of a group of 9-amino-acridines that selectively downregulate regulatory T cell functions through FoxP3

Tue, 2025-03-04 06:00

iScience. 2025 Jan 31;28(3):111931. doi: 10.1016/j.isci.2025.111931. eCollection 2025 Mar 21.

ABSTRACT

FoxP3+ regulatory T cells (Tregs) are responsible for immune homeostasis by suppressing excessive anti-self-immunity. Tregs facilitate tumor growth by inhibiting anti-tumor immunity. Here, we explored the targeting of FoxP3 as a basis for new immunotherapies. In a high-throughput phenotypic screening of a drug repurposing library using human primary T cells, we identified quinacrine as a FoxP3 downregulator. In silico searches based on the structure of quinacrine, testing of sub-libraries of analogs in vitro, and validation identified a subset of 9-amino-acridines that selectively abrogated Treg suppressive functions. Mechanistically, these acridines interfered with the DNA-binding activity of FoxP3 and inhibited FoxP3-regulated downstream gene regulation. Release from Treg suppression by 9-amino-acridines increased anti-tumor immune responses both in cancer patient samples and in mice in a syngeneic tumor model. Our study highlights the feasibility of screening for small molecular inhibitors of FoxP3 as an approach to pursuing Treg-based immunotherapy.

PMID:40034859 | PMC:PMC11872463 | DOI:10.1016/j.isci.2025.111931

Categories: Literature Watch

Phytochemical synergies in BK002: advanced molecular docking insights for targeted prostate cancer therapy

Tue, 2025-03-04 06:00

Front Pharmacol. 2025 Feb 17;16:1504618. doi: 10.3389/fphar.2025.1504618. eCollection 2025.

ABSTRACT

Achyranthes japonica (Miq.) Nakai (AJN) and Melandrium firmum (Siebold and Zucc.) Rohrb. (MFR) are medicinal plants recognized for their bioactive phytochemicals, including ecdysteroids, anthraquinones, and flavonoids. This study investigates the anticancer properties of key constituents of these plants, focusing on the BK002 formulation, a novel combination of AJN and MFR. Specifically, the research employs advanced molecular docking and in silico analyses to assess the interactions of bioactive compounds ecdysterone, inokosterone, and 20-hydroxyecdysone (20-HE) with key prostate cancer-related network proteins, including 5α-reductase, CYP17, DNMT1, Dicer, PD-1, and PD-L1. Molecular docking techniques were applied to evaluate the binding affinities contributions of the bioactive compounds in BK002 against prostate cancer-hub network targets. The primary focus was on enzymes like 5α-reductase and CYP17, which are central to androgen biosynthesis, as well as on cancer-related proteins such as DNA methyltransferase 1 (DNMT1), Dicer, programmed death-1 (PD-1), and programmed death ligand-1 (PD-L1). Based on data from prostate cancer patients, key target networks were identified, followed by in silico analysis of the primary bioactive components of BK002.In silico assessments were conducted to evaluate the safety profiles of these compounds, providing insights into their therapeutic potential. The docking studies revealed that ecdysterone, inokosterone, and 20-hydroxyecdysonec demonstrated strong binding affinities to the critical prostate cancer-related enzymes 5α-reductase and CYP17, contributing to a potential reduction in androgenic activity. These compounds also exhibited significant inhibitory interactions with DNMT1, Dicer, PD-1, and PD-L1, suggesting a capacity to interfere with key oncogenic and immune evasion pathways. Ecdysterone, inokosterone, and 20-hydroxyecdysone have demonstrated the ability to target key oncogenic pathways, and their favorable binding affinity profiles further underscore their potential as novel therapeutic agents for prostate cancer. These findings provide a strong rationale for further preclinical and clinical investigations, supporting the integration of BK002 into therapeutic regimens aimed at modulating tumor progression and immune responses.

PMID:40034825 | PMC:PMC11872924 | DOI:10.3389/fphar.2025.1504618

Categories: Literature Watch

Genomics-Informed Drug Repurposing Strategy Identifies Novel Therapeutic Targets for Metabolic Dysfunction-Associated Steatotic Liver Disease

Tue, 2025-03-04 06:00

medRxiv [Preprint]. 2025 Feb 21:2025.02.18.25321035. doi: 10.1101/2025.02.18.25321035.

ABSTRACT

Identification of drug-repurposing targets with genetic and biological support is an economically and temporally efficient strategy for improving treatment of diseases. We employed a cross-disciplinary approach to identify potential treatments for metabolic dysfunction associated steatotic liver disease (MASLD) using humans as a model organism. We identified 212 putative causal genes associated with MASLD using data from a large multi-ancestry genetic association study, of which 158 (74.5%) are novel. From this set we identified 57 genes that encode for druggable protein targets, and where the effects of increasing genetically predicted gene expression on MASLD risk align with the function of that drug on the protein target. These potential targets were then evaluated for evidence of efficacy using Mendelian randomization, pathway analysis, and protein structural modeling. Using these approaches, we present compelling evidence to suggest activation of FADS1 by icosopent ethyl as well as S1PR2 by fingolimod could be promising therapeutic strategies for MASLD.

PMID:40034783 | PMC:PMC11875238 | DOI:10.1101/2025.02.18.25321035

Categories: Literature Watch

A Large-Scale Genome-wide Association Study of Blood Pressure Accounting for Gene-Depressive Symptomatology Interactions in 564,680 Individuals from Diverse Populations

Tue, 2025-03-04 06:00

Res Sq [Preprint]. 2025 Feb 17:rs.3.rs-6025759. doi: 10.21203/rs.3.rs-6025759/v1.

ABSTRACT

Background Gene-environment interactions may enhance our understanding of hypertension. Our previous study highlighted the importance of considering psychosocial factors in gene discovery for blood pressure (BP) but was limited in statistical power and population diversity. To address these challenges, we conducted a multi-population genome-wide association study (GWAS) of BP accounting for gene-depressive symptomatology (DEPR) interactions in a larger and more diverse sample. Results Our study included 564,680 adults aged 18 years or older from 67 cohorts and 4 population backgrounds (African (5%), Asian (7%), European (85%), and Hispanic (3%)). We discovered seven novel gene-DEPR interaction loci for BP traits. These loci mapped to genes implicated in neurogenesis ( TGFA , CASP3 ), lipid metabolism ( ACSL1 ), neuronal apoptosis ( CASP3 ), and synaptic activity ( CNTN6 , DBI ). We also identified evidence for gene-DEPR interaction at nine known BP loci, further suggesting links between mood disturbance and BP regulation. Of the 16 identified loci, 11 loci were derived from African, Asian, or Hispanic populations. Post-GWAS analyses prioritized 36 genes, including genes involved in synaptic functions ( DOCK4 , MAGI2 ) and neuronal signaling ( CCK , UGDH , SLC01A2 ). Integrative druggability analyses identified 11 druggable candidate gene targets, including genes implicated in pathways linked to mood disorders as well as gene products targeted by known antihypertensive drugs. Conclusions Our findings emphasize the importance of considering gene-DEPR interactions on BP, particularly in non-European populations. Our prioritized genes and druggable targets highlight biological pathways connecting mood disorders and hypertension and suggest opportunities for BP drug repurposing and risk factor prevention, especially in individuals with DEPR.

PMID:40034430 | PMC:PMC11875294 | DOI:10.21203/rs.3.rs-6025759/v1

Categories: Literature Watch

Repurposing the anti-parasitic agent pentamidine for cancer therapy; a novel approach with promising anti-tumor properties

Mon, 2025-03-03 06:00

J Transl Med. 2025 Mar 3;23(1):258. doi: 10.1186/s12967-025-06293-w.

ABSTRACT

Pentamidine (PTM) is an aromatic diamidine administered for infectious diseases, e.g. sleeping sickness, malaria, and Pneumocystis jirovecii pneumonia. Due to similarities of cellular mechanisms between human cells and such infections, PTM has also been proposed for repurposing in non-infectious diseases such as cancer. Indeed, by modulating different signaling pathways such as PI3K/AKT, MAPK/ERK, p53, PD-1/PD-L1, etc., PTM has been shown to inhibit different properties of cancer, including proliferation, invasion, migration, hypoxia, and angiogenesis, while inducing anti-tumor immune responses and apoptosis. Given the promising implications of PTM for cancer treatment, however, the clinical translation of PTM in cancer is not without certain challenges. In fact, clinical trials have shown that systemic administration of PTM can be concurrent with serious adverse effects, e.g. hypoglycemia. Therefore, to reduce the administered doses of PTM, lower the risk of adverse effects, and prevent any potential drug resistance, while maintaining the anti-tumor efficacy, two main strategies have been suggested. One is combination therapy that employs PTM in conjunction with other anti-cancer modalities, such as chemotherapy and radiotherapy, and attacks tumor cells with significant additive or synergistic anti-tumor effects. The other is developing PTM-loaded nanocarrier drug delivery systems e.g. pegylated liposomes, chitosan-coated niosomes, squalene-based nanoparticles, hyaluronated lipid-polymer hybrid nanoparticles, etc., that offer enhanced pharmacokinetic characteristics, including increased bioavailability, sit-targeting, and controlled/sustained drug release. This review highlights the anti-tumor properties of PTM that favor its repurposing for cancer treatment, as well as, PTM-based combination therapies and nanocarrier delivery systems which can enhance therapeutic efficacy and simultaneously reduce toxicity.

PMID:40033361 | DOI:10.1186/s12967-025-06293-w

Categories: Literature Watch

Drug Repurposing Tactics in the USA: Known Active Pharmaceutical Ingredients in New Indications

Mon, 2025-03-03 06:00

Pulm Pharmacol Ther. 2025 Mar 1:102348. doi: 10.1016/j.pupt.2025.102348. Online ahead of print.

NO ABSTRACT

PMID:40032240 | DOI:10.1016/j.pupt.2025.102348

Categories: Literature Watch

DSANIB: drug-target interaction predictions with dual-view synergistic attention network and information bottleneck strategy

Mon, 2025-03-03 06:00

IEEE J Biomed Health Inform. 2024 Nov 13;PP. doi: 10.1109/JBHI.2024.3497591. Online ahead of print.

ABSTRACT

Prediction of drug-target interactions (DTIs) is one of the crucial steps for drug repositioning. Identifying DTIs through bio-experimental manners is always expensive and time-consuming. Recently, deep learning-based approaches have shown promising advancements in DTI prediction, but they face two notable challenges: (i) how to explicitly capture local interactions between drug-target pairs and learn their higher-order substructure embeddings; (ii) How to filter out redundant information to obtain effective embeddings for drugs and targets. Results: In this study, we propose a novel approach, termed DSANIB, to infer potential interactions between drugs and targets. DSANIB comprises two primary components: (1) DSAN component: The Inter-view Attention Network Module explicitly learns the local interactions between drugs and targets, while the Intra-view Attention Network Module aggregates information from local interaction features to obtain their higher-order substructure embeddings. (2) Information Bottleneck (IB) component: DSANIB adopts the IB strategy, which could retain relevant information while minimizing the redundant features to obtain their discriminative representations. Extensive experimental results demonstrate that DSANIB outperforms other SOTA prediction models. In addition, visualization of drug and target embeddings learned through DSANIB could provide interpretable insights for the prediction results. Availability: The source code has been made publicly available on GitHub https://github.com/Zzz-Soar/DSANIB.

PMID:40030194 | DOI:10.1109/JBHI.2024.3497591

Categories: Literature Watch

Drug repurposing in amyotrophic lateral sclerosis (ALS)

Mon, 2025-03-03 06:00

Expert Opin Drug Discov. 2025 Mar 3. doi: 10.1080/17460441.2025.2474661. Online ahead of print.

ABSTRACT

INTRODUCTION: Identifying treatments that can alter the natural history of amyotrophic lateral sclerosis (ALS) is challenging. For years, drug discovery in ALS has relied upon traditional approaches with limited success. Drug repurposing, where clinically approved drugs are reevaluated for other indications, offers an alternative strategy that overcomes some of the challenges associated with de novo drug discovery. Whilst not a new concept, the potential of drug repurposing in ALS is yet to be fully realized.

AREAS COVERED: In this review, the authors discuss the challenge of drug discovery in ALS and specifically examine the potential of drug repurposing for the identification of new effective treatments. The authors consider a broad range of approaches, from screening in experimental models to computational approaches, and outline some general principles for pre-clinical and clinical research to help bridge the translational gap. Literature was reviewed from original publications, press releases and clinical trials.

EXPERT OPINION: Despite the remaining challenges, drug repurposing offers the opportunity to improve therapeutic options for ALS patients. Nevertheless, stringent pre-clinical research will be necessary to identify the most promising compounds while innovative experimental medicine studies will also be paramount to bridge the aforementioned translational gap. The authors further highlight the importance of combining expertise across academia, industry and wider stakeholders, which will be key in the successful delivery of repurposed therapies to the clinic.

PMID:40029669 | DOI:10.1080/17460441.2025.2474661

Categories: Literature Watch

Darunavir inhibits dengue virus replication by targeting the hydrophobic pocket of the envelope protein

Sun, 2025-03-02 06:00

Biochem Pharmacol. 2025 Feb 28:116839. doi: 10.1016/j.bcp.2025.116839. Online ahead of print.

ABSTRACT

Dengue viruses (DENV) pose significant health threats, with no approved antiviral drugs currently available, creating an urgent need for new therapies. This study screened FDA-approved drugs for their antiviral ability against DENV and identified three promising candidates: darunavir (DRV), domperidone, and tetracycline. DRV demonstrated the highest efficacy against three DENV serotypes, with half-maximal effective concentrations (EC50) below 1 µM, surpassing the performance of tetracycline and domperidone. It effectively blocked DENV envelope (E) protein attachment to two type cells with EC50 values less than 0.2 μM. Domperidone reduced DENV-2 attachment to TE671 cells (EC50 = 3.08 μM) but was less effective in BHK-21 cells, while tetracycline inhibited NS3 protease (IC50 = 1.12 μM). Among DRV's structurally related drugs, fosamprenavir (FPV) significantly reduced DENV infectivity and virus yield, with EC50 values below 0.5 µM. In vivo, DRV at 1, 2, and 5 mg/kg achieved 100 % survival in suckling mice, compared to 83.5 % with FPV. Real-time RT-PCR showed DRV more effectively reduced DENV-2 RNA in mouse brains than FPV. Molecular docking showed DRV and FPV bind tightly to the DENV-2 E protein's N-octyl-β-D-glucoside (βOG) hydrophobic pocket, with DRV forming stronger interactions than FPV. Chimeric DENV-2 single-round infectious particle tests confirmed DRV's effective targeting of this pocket, though mutations at K128, L198, Q200, I270, and T280 reduced its efficacy. These findings highlight DRV as a potent antiviral agent against DENV, targeting the E protein's βOG hydrophobic pocket, with the potential for rapid deployment in treating and preventing infections.

PMID:40024350 | DOI:10.1016/j.bcp.2025.116839

Categories: Literature Watch

Gefitinib as an antimalarial: unveiling its therapeutic potential

Fri, 2025-02-28 06:00

Inflammopharmacology. 2025 Feb 28. doi: 10.1007/s10787-025-01682-5. Online ahead of print.

ABSTRACT

Resistant strains of Plasmodium spp. pose a great threat to healthcare. Drug repurposing is a smart, and an effective way to look for new alternatives for different ailments including malaria. Protein tyrosine kinases (PTKs) play a crucial role in growth, maturation as well as differentiation of Plasmodium and this study explores antimalarial activity of PTKs inhibitor gefitinib using in silico and experimental approaches. The drug showed considerable inhibitory activity against P. falciparum 3D7 (IC50 0.49 µg/mL) and RKL-9 (IC50 0.83 µg/mL) strains. Isobologram analysis revealed substantial synergism between gefitinib and artesunate. Gefitinib illustrated highest negative D-score towards phosphoethanolamine methyltransferase followed by PfPK5 and CDPK1. Its acute toxicity was 4 g/kg. Gefitinib (100 mg/kg) exhibited a dose-dependent curative activity against P. berghei with 91.09% chemo-suppression and the combination of gefitinib 100 mg/kg and AS 50 mg/kg exhibited complete parasite clearance with no recrudescence which was also evidenced by cytokine analysis, biochemical as well as histopathological studies. At length, gefitinib illustrated considerable antiplasmodial action by targeting phosphoethanolamine methyltransferase, PfPK5 and CDPK1. The combination of gefitinib (100 mg/kg) and AS (50 mg/kg) holds promise for malaria treatment. Further, research is being done to evaluate its pharmacokinetic properties.

PMID:40019687 | DOI:10.1007/s10787-025-01682-5

Categories: Literature Watch

Challenges in international investigator-led rare disease clinical trials and the case for optimism in inclusion body myositis

Fri, 2025-02-28 06:00

Clin Exp Rheumatol. 2025 Feb;43(2):309-315. doi: 10.55563/clinexprheumatol/dyjcsn. Epub 2025 Feb 26.

ABSTRACT

OBJECTIVES: This paper aims to provide insight into the challenges and opportunities of conducting an investigator-led, international, multicentre clinical trial for Inclusion Body Myositis (IBM), a rare inflammatory myopathy.

METHODS: An international, multicentre, randomised, controlled trial of a repurposed drug (sirolimus) was initiated based on promising results from a mono-centric pilot study. The progress of the trial was analysed to identify key challenges encountered and solutions developed.

RESULTS: This large, collaborative study has presented a mosaic of challenges and opportunities, many ubiquitous with investigator-led trials. Key challenges have included securing adequate funding, coordinating manufacture of placebo, negotiating international contracts, managing limited study budgets and delays linked to the COVID-19 pandemic. Alongside these challenges, the study team have found opportunities for creative and effective solutions, including the flexibility of building study databases, optimising digital data capture and harnessing patient involvement.

CONCLUSIONS: Instrumental to the progress of the trial has been the collaboration between site teams, patient partnership and adaptability.

PMID:40018747 | DOI:10.55563/clinexprheumatol/dyjcsn

Categories: Literature Watch

Pages