Ivana Nikolic , Aleksandar Lazovic , Isidora Stanisavljevic , Marija Andjelkovic , Suzana Popovic , Sladjana Pavlovic , Milena Jurisevic , Marina Mitrovic
{"title":"Duloxetine's potential dual antitumor and immunomodulatory role in apoptosis and autophagy signaling pathways in cancer: In Vitro and In Vivo evidence","authors":"Ivana Nikolic , Aleksandar Lazovic , Isidora Stanisavljevic , Marija Andjelkovic , Suzana Popovic , Sladjana Pavlovic , Milena Jurisevic , Marina Mitrovic","doi":"10.1016/j.ejps.2025.107165","DOIUrl":null,"url":null,"abstract":"<div><h3>Background</h3><div>Cancer remains the second leading cause of mortality worldwide, underscoring the urgent need for novel therapeutic strategies. Drug repurposing is an effective strategy to address current cancer challenges, such as the resistance and toxicity associated with traditional chemotherapy. Among the various psychotropic drugs, antidepressants are emerging as promising candidates due to their demonstrated anticancer activity.</div></div><div><h3>Methods</h3><div>We evaluated duloxetine (DUL), a serotonin-norepinephrine reuptake inhibitor, for anticancer and immunomodulatory activity by performing comprehensive <em>in vitro</em> and <em>in vivo</em> experiments. In HCT116 (colon), HeLa (cervical), and MDA-MB-231 (triple-negative breast cancer) human cancer cells, and 4T1 mouse breast cancer cells, we investigated DUL’s cytotoxicity, apoptosis, autophagy, cell cycle arrest, and migration inhibition through MTT assay, flow cytometry, and immunofluorescence methods. An orthotopic mouse model of breast cancer was utilized to investigate the <em>in vivo</em> tumor inhibitory effects of DUL, along with its systemic toxicity and immunomodulatory properties. Gene expression (p53, Beclin-1), cytokine profile, and CD3<sup>+</sup> <em>T</em> cell activation were examined by mRNA sequencing and ELISA kits to explore underlying mechanisms.</div></div><div><h3>Results</h3><div>For the first time, our <em>in vitro</em> results indicated that DUL caused dose-dependent cytotoxicity, apoptosis, autophagy, and cell cycle arrest in all cancer lines tested, which was more selective than 5-fluorouracil (5-FU). Mechanistically, DUL modulated apoptotic (Bcl-2, Bax, caspase-3), autophagic (p62), and survival (pAkt) signaling, disrupted mitochondrial membrane potential, and inhibited cell migration. <em>In vivo</em>, DUL inhibited tumor growth without inducing hepatic or renal toxicity. Notably, DUL stimulated the production of pro-inflammatory cytokines (IFN-γ, IL-1β, TNF-α), enhanced the immunity of CD3<sup>+</sup> <em>T</em> cells, and increased the expression of pro-apoptotic p53 and autophagic Beclin-1 genes, reflecting its dual antitumor and immunomodulatory actions.</div></div><div><h3>Conclusion</h3><div>Our findings revealed for the first time that DUL is a promising repurposed drug for cancer treatment, as it has demonstrated proven antitumor efficacy and immune-stimulating properties. This novel dual role of DUL warrants further investigation in clinical oncology.</div></div>","PeriodicalId":12018,"journal":{"name":"European Journal of Pharmaceutical Sciences","volume":"212 ","pages":"Article 107165"},"PeriodicalIF":4.7000,"publicationDate":"2025-06-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"European Journal of Pharmaceutical Sciences","FirstCategoryId":"3","ListUrlMain":"https://www.sciencedirect.com/science/article/pii/S0928098725001642","RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"PHARMACOLOGY & PHARMACY","Score":null,"Total":0}
引用次数: 0
Abstract
Background
Cancer remains the second leading cause of mortality worldwide, underscoring the urgent need for novel therapeutic strategies. Drug repurposing is an effective strategy to address current cancer challenges, such as the resistance and toxicity associated with traditional chemotherapy. Among the various psychotropic drugs, antidepressants are emerging as promising candidates due to their demonstrated anticancer activity.
Methods
We evaluated duloxetine (DUL), a serotonin-norepinephrine reuptake inhibitor, for anticancer and immunomodulatory activity by performing comprehensive in vitro and in vivo experiments. In HCT116 (colon), HeLa (cervical), and MDA-MB-231 (triple-negative breast cancer) human cancer cells, and 4T1 mouse breast cancer cells, we investigated DUL’s cytotoxicity, apoptosis, autophagy, cell cycle arrest, and migration inhibition through MTT assay, flow cytometry, and immunofluorescence methods. An orthotopic mouse model of breast cancer was utilized to investigate the in vivo tumor inhibitory effects of DUL, along with its systemic toxicity and immunomodulatory properties. Gene expression (p53, Beclin-1), cytokine profile, and CD3+T cell activation were examined by mRNA sequencing and ELISA kits to explore underlying mechanisms.
Results
For the first time, our in vitro results indicated that DUL caused dose-dependent cytotoxicity, apoptosis, autophagy, and cell cycle arrest in all cancer lines tested, which was more selective than 5-fluorouracil (5-FU). Mechanistically, DUL modulated apoptotic (Bcl-2, Bax, caspase-3), autophagic (p62), and survival (pAkt) signaling, disrupted mitochondrial membrane potential, and inhibited cell migration. In vivo, DUL inhibited tumor growth without inducing hepatic or renal toxicity. Notably, DUL stimulated the production of pro-inflammatory cytokines (IFN-γ, IL-1β, TNF-α), enhanced the immunity of CD3+T cells, and increased the expression of pro-apoptotic p53 and autophagic Beclin-1 genes, reflecting its dual antitumor and immunomodulatory actions.
Conclusion
Our findings revealed for the first time that DUL is a promising repurposed drug for cancer treatment, as it has demonstrated proven antitumor efficacy and immune-stimulating properties. This novel dual role of DUL warrants further investigation in clinical oncology.
期刊介绍:
The journal publishes research articles, review articles and scientific commentaries on all aspects of the pharmaceutical sciences with emphasis on conceptual novelty and scientific quality. The Editors welcome articles in this multidisciplinary field, with a focus on topics relevant for drug discovery and development.
More specifically, the Journal publishes reports on medicinal chemistry, pharmacology, drug absorption and metabolism, pharmacokinetics and pharmacodynamics, pharmaceutical and biomedical analysis, drug delivery (including gene delivery), drug targeting, pharmaceutical technology, pharmaceutical biotechnology and clinical drug evaluation. The journal will typically not give priority to manuscripts focusing primarily on organic synthesis, natural products, adaptation of analytical approaches, or discussions pertaining to drug policy making.
Scientific commentaries and review articles are generally by invitation only or by consent of the Editors. Proceedings of scientific meetings may be published as special issues or supplements to the Journal.