Medical OncologyPub Date : 2025-06-30DOI: 10.1007/s12032-025-02844-9
Md Sakib Al Hasan, Yasin Emon, Mohammad Y Alshahrani, Md Mizan, Mohammed Burhan Uddin, Ali Mohamod Wasaf Hasan, Md Abu Sayeed, Emon Mia, Noshin Tasnim Yana, Rakib Hossan, Muhammad Torequl Islam
{"title":"Coumarin derivatives as anticancer agents targeting PI3K-AKT-mTOR pathway: a comprehensive literature review.","authors":"Md Sakib Al Hasan, Yasin Emon, Mohammad Y Alshahrani, Md Mizan, Mohammed Burhan Uddin, Ali Mohamod Wasaf Hasan, Md Abu Sayeed, Emon Mia, Noshin Tasnim Yana, Rakib Hossan, Muhammad Torequl Islam","doi":"10.1007/s12032-025-02844-9","DOIUrl":"10.1007/s12032-025-02844-9","url":null,"abstract":"<p><p>Coumarin is a natural benzopyrone compound known for its diverse pharmacological activities, including anticancer, anti-inflammatory, and antioxidant properties. But its derivatives' exploration in cancer, specially PI3K-AKT-mTOR pathways-based cancer activity not explored yet. This review aims to evaluate the anticancer potential of coumarin derivatives' by exploring their modulatory effects on the PI3K-AKT-mTOR signaling pathway across diverse cancer types. This review adopted a systematic literature approach, emphasizing studies across various cancer types and focusing on coumarin's mechanisms and therapeutic potential. Results reveal that coumarin derivatives suppress PI3K-AKT-mTOR pathway activity across cancers, such as liver, breast, and colorectal, with IC<sub>50</sub> values ranging from 4 µM to > 200 µM in vitro and confirmed effects in vivo. The modulation of other pathways, including NF-κB and MAPK, underscores their multi-targeted anticancer action. Despite promising preclinical efficacy, challenges like low bioavailability, potential hepatotoxicity, and systemic toxicity persist. Structure-activity relationship (SAR) studies suggest that introducing specific functional groups can enhance selectivity, reduce toxicity, and improve therapeutic outcomes. The conclusion reinforces the potential of coumarin derivatives as novel anticancer agents, advocating for structural optimizations and clinical investigations to overcome pharmacokinetic barriers and maximize therapeutic benefits. This exploration offers a strategic perspective on utilizing coumarin-based molecules in advancing cancer therapeutics.</p>","PeriodicalId":18433,"journal":{"name":"Medical Oncology","volume":"42 8","pages":"301"},"PeriodicalIF":2.8,"publicationDate":"2025-06-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144528661","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Ovarian cancer, neutrophil hitchhiking, and NETs: unraveling their role in pathogenesis and management.","authors":"Shahrzad Amirlatifi, Shabnam Forouzin, Elahe Sadati, Somayeh Moradpanah, Ramina Fazeli, Paria Panahinia, Nasim Zarifi, Sania Rahimi","doi":"10.1007/s12032-025-02860-9","DOIUrl":"https://doi.org/10.1007/s12032-025-02860-9","url":null,"abstract":"<p><p>Ovarian cancer is the most lethal gynecologic malignancy, driven by extensive genetic heterogeneity, clonal evolution, and resistance to therapy. Despite advances in treatment, recurrence remains common, highlighting the urgent need for novel therapeutic strategies. Neutrophil extracellular traps (NETs)-web-like DNA-protein complexes released during NETosis-have recently emerged as key facilitators of tumor progression across multiple cancers. However, their role in ovarian cancer remains underexplored. NETs contribute to metastasis, angiogenesis, immune evasion, and chemoresistance, creating a pro-tumorigenic microenvironment. This review identifies critical knowledge gaps in NET-mediated mechanisms in ovarian cancer and introduces, for the first time in this context, the concept of neutrophil hitchhiking, whereby circulating tumor cells exploit neutrophils to enhance metastatic dissemination. We examine molecular pathways driving NETosis and their influence on the tumor microenvironment, highlighting how NET-targeted therapies-including NETosis inhibitors and immune modulators-offer promising avenues to suppress metastasis, restore immune surveillance, and improve treatment outcomes. By illuminating this underexplored axis of neutrophil-tumor interaction, we aim to stimulate research and therapeutic development that could transform ovarian cancer management.</p>","PeriodicalId":18433,"journal":{"name":"Medical Oncology","volume":"42 8","pages":"302"},"PeriodicalIF":2.8,"publicationDate":"2025-06-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144528664","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Medical OncologyPub Date : 2025-06-30DOI: 10.1007/s12032-025-02862-7
Kiramat Ali Shah, Anam Razzaq, Tariq Ali, Saeed Ullah, Bengang You, Haroon Iqbal, Jing-Hao Cui
{"title":"Role of nanomedicines in lung cancer treatment and diagnosis: opportunities and challenges.","authors":"Kiramat Ali Shah, Anam Razzaq, Tariq Ali, Saeed Ullah, Bengang You, Haroon Iqbal, Jing-Hao Cui","doi":"10.1007/s12032-025-02862-7","DOIUrl":"https://doi.org/10.1007/s12032-025-02862-7","url":null,"abstract":"<p><p>Lung cancer (LC) is one of the leading causes of cancer-related mortality globally, and despite numerous available treatment modalities, the 5-year survival rate for LC patients remains approximately 20%. A large proportion of LC patients are diagnosed at advanced stages, and conventional treatments frequently exhibit poor efficacy and are often associated with numerous side effects. Therefore, there is an urgent need for innovative therapeutic strategies to meet the needs of LC patients. Recent advances in nanotechnology have opened new avenues for the diagnosis and treatment of LC, particularly through the use of nanoparticles (NPs) as a drug delivery system, which offer enhanced therapeutic potential. Due to their unique physicochemical properties, NPs are widely employed in LC drug delivery systems. They can efficiently load pharmaceuticals and specifically target cancer cells to enhance diagnostics and treatment. Additionally, NPs can encapsulate multiple therapeutic agents, including targeted molecules and photosensitive reagents. Despite the substantial promise of nanotechnology in LC treatment, several challenges remain in its clinical application. This review represents a comprehensive overview of LC, encompassing risk factors, types, stages, mechanisms, and traditional treatment strategies. Furthermore, we thoroughly emphasized the current nanotechnology-based diagnostics and therapies for LC. In summary, the ongoing advancement of drug delivery systems highlights the potential of nanotechnology to enhance traditional LC treatments and facilitate the development of new therapeutic strategies. Future research will focus on optimizing the unique properties of various NPs to enhance treatment outcomes and offer personalized therapeutic strategies, ultimately improving the prognosis and quality of life for LC patients.</p>","PeriodicalId":18433,"journal":{"name":"Medical Oncology","volume":"42 8","pages":"305"},"PeriodicalIF":2.8,"publicationDate":"2025-06-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144528679","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Medical OncologyPub Date : 2025-06-30DOI: 10.1007/s12032-025-02873-4
Anita Choudhary, Anil Kumar, Anjana Munshi
{"title":"Genetic variants in oncogenic miRNA and 3' untranslated region of tumor suppressor genes: emerging insight into cancer genetics.","authors":"Anita Choudhary, Anil Kumar, Anjana Munshi","doi":"10.1007/s12032-025-02873-4","DOIUrl":"https://doi.org/10.1007/s12032-025-02873-4","url":null,"abstract":"<p><p>The miRNAs are key regulators of post-transcriptional gene expression. These are associated with the different molecular mechanisms which are engaged in the pathogenesis of various cancers, including breast cancer. The tumor suppressor and oncogenic miRNAs have a significant impact on cell proliferation, metastasis, angiogenesis, and apoptotic pathways. Variation in oncogenic miRNA encoding and the 3' UTR of the tumor suppressor genes associated with the development and prognosis of the cancer is being explored. These genetic variants alter oncogenic miRNAs' stability, target recognition, and binding ability, thereby resulting in the dysregulation of their target tumor suppressor gene, leading to uncontrolled cell division, a significant hallmark of cancer. The current review has been compiled to explore the genetic variation reported in the oncogenic miRNA encoding and 3' UTR of their target tumor suppressor genes associated with the development of breast cancer and its progression, focusing on the associated molecular mechanisms. Further, an effort has been made to discuss the possible therapeutic strategies, especially anti-miR, RNA interference, CRISPR/Cas, and ASOs, that have the potential to restore the function of the dysregulated tumor suppressor as well as oncogenic miRNA encoding genes.</p>","PeriodicalId":18433,"journal":{"name":"Medical Oncology","volume":"42 8","pages":"303"},"PeriodicalIF":2.8,"publicationDate":"2025-06-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144528662","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"pH-responsive nanocapsules loaded with 5-fluorouracil-coated green-synthesized CuO-ZnO NPs for enhanced anticancer activity against HeLa cells.","authors":"Gouranga Dutta, Abimanyu Sugumaran, Damodharan Narayanasamy","doi":"10.1007/s12032-025-02847-6","DOIUrl":"https://doi.org/10.1007/s12032-025-02847-6","url":null,"abstract":"<p><p>Cancer represents a form of unregulated cellular proliferation, persisting as a significant challenge to global health. Despite the progress made in therapeutic interventions, challenges such as inadequate drug selectivity, systemic toxicity, and the development of drug resistance continue to pose considerable obstacles. In response to these barriers, we prepared pH (Stimuli)-responsive gelatin (GA) nanocapsules (NCs) that encapsulate 5-fluorouracil (5-FU)-coated CuO-ZnO nanoparticles (NPs) to achieve a combined effect in anticancer activity. The CuO-ZnO nanoparticles have been chosen for their combined antitumor efficacy of CuO and ZnO NPs. CuO-ZnO NPs synthesized through an environmentally friendly approach utilizing Trichosanthes dioica fruit extract demonstrated a size of 35.79 ± 6.04 nm. The drug-coated NPs were encapsulated within a gelatin matrix, which was stabilized with poloxamer 188 (Po) through modified emulsion techniques. Various characterizations were conducted using FTIR, XRD, TEM, and XPS, yielding valuable insights into the structural integrity and morphology of CuO-ZnO NPs and NCs. The NCs' average size was found to be 331.4 ± 38.7 nm with an average zeta potential of -15.6 ± 4.98 mV. Studies found drug release is sensitive to acidic pH 5.6, with a more rapid release than pH 7.4 (~25% 48 h), increasing tumor delivery selectivity. The assessment of cytotoxicity (MTT assay) on HeLa cells revealed a markedly reduced IC<sub>50</sub> (13.71 ± 2.6 µg/mL) for nanocapsules in comparison with CuO-ZnO NPs and 5-Fu, suggesting a combined interaction. The confirmation of the apoptosis feature was achieved via AO/EtBr staining. The results underscore the promise of PoGA-5Fu(CuO-ZnO) nanocapsules as a targeted approach to cancer therapy, demonstrating improved efficacy while minimizing systemic toxicity.</p>","PeriodicalId":18433,"journal":{"name":"Medical Oncology","volume":"42 8","pages":"300"},"PeriodicalIF":2.8,"publicationDate":"2025-06-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144528667","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Medical OncologyPub Date : 2025-06-29DOI: 10.1007/s12032-025-02851-w
Benjo Chalissery, Sahana Nagappa Moger, K P Shreyas, Jismon Jose, V K Ashith
{"title":"Recent developments on copper incorporation of polymer thin films for cancer cell treatment: review.","authors":"Benjo Chalissery, Sahana Nagappa Moger, K P Shreyas, Jismon Jose, V K Ashith","doi":"10.1007/s12032-025-02851-w","DOIUrl":"https://doi.org/10.1007/s12032-025-02851-w","url":null,"abstract":"<p><p>Cancer remains a critical global health challenge, with conventional treatments such as chemotherapy, radiation, and immunotherapy often hindered by issues like poor selectivity, systemic toxicity, and drug resistance. Recent advancements in materials science, particularly in the field of nanotechnology and drug delivery systems, have opened new avenues for more effective cancer therapies. This review focuses on copper-doped polymer thin films-an emerging class of therapeutic materials with promising anti-cancer properties. Copper, a biologically essential trace element, contributes to cancer cell apoptosis by inducing oxidative stress and has also been shown to enhance the efficacy of chemotherapeutic agents, helping to overcome drug resistance in various cancers. The paper reviews a broad spectrum of recent literature, encompassing both experimental and theoretical studies related to the synthesis, structural characterization, and biomedical performance of these materials. Methodologies such as in vitro cytotoxicity assays, in vivo treatment models, and physicochemical analyses are discussed to provide a comprehensive understanding of their therapeutic potential. The review concludes by highlighting the advantages, challenges, and future prospects of integrating copper-doped polymer thin films into advanced cancer treatment strategies.</p>","PeriodicalId":18433,"journal":{"name":"Medical Oncology","volume":"42 8","pages":"298"},"PeriodicalIF":2.8,"publicationDate":"2025-06-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144528668","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Medical OncologyPub Date : 2025-06-28DOI: 10.1007/s12032-025-02863-6
Allah Rakha Yaseen, Muhammad Suleman, Khalid J Alzahrani, Khalaf F Alsharif, Fuad M Alzahrani
{"title":"Unleashing the future of cancer immunotherapy: in silico design of a multi-epitope and mRNA vaccine duo targeting EWSR1-ATF1, EWSR1-CREB1, and PRAME to conquer clear cell sarcoma using immunoinformatics approaches.","authors":"Allah Rakha Yaseen, Muhammad Suleman, Khalid J Alzahrani, Khalaf F Alsharif, Fuad M Alzahrani","doi":"10.1007/s12032-025-02863-6","DOIUrl":"https://doi.org/10.1007/s12032-025-02863-6","url":null,"abstract":"<p><p>Clear cell sarcoma (CCS) is a rare and aggressive soft tissue malignancy characterized by a high metastatic propensity and recurrence rate, with conventional non-specific treatment modalities often yielding limited efficacy and substantial adverse effects. This study aimed to design novel vaccine candidates: a multi-epitope vaccine (MEV) and an mRNA-based vaccine, both targeting the EWSR1-ATF1 and EWSR1-CREB1 fusion proteins, along with the PRAME antigen, to stimulate robust tumor-specific immune responses. Advanced immunoinformatics approaches were employed to identify highly antigenic B-cell and T-cell epitopes while minimizing potential allergenicity and toxicity, ensuring optimal safety and efficacy. The MEV construct was engineered with GM-CSF as an adjuvant to enhance dendritic cell activation and antigen presentation, with EAAK and AAY linkers incorporated to improve structural integrity and epitope processing. For the mRNA vaccine, the MEV was codon-optimized and incorporated into a stable mRNA construct with a 5' cap, Kozak sequence, and poly(A) tail to enhance the translation efficiency and prolong antigen expression. Structural and molecular dynamics simulations confirmed strong and stable interactions of both vaccine constructs with Toll-Like Receptor-3 (TLR-3), supporting their potential for effective immune activation. Furthermore, population coverage analysis demonstrated a global reach of 99.48%, ensuring broad immunogenicity across diverse genetic backgrounds. In silico immune response simulations predicted a sustained immune activation lasting over 417 days, characterized by robust cytokine secretion, strong memory cell formation, and high antibody titers following a three-dose regimen. These findings suggest that both the multi-epitope and mRNA-based vaccine candidates hold substantial promise as novel and precise immunotherapeutic interventions for CCS, potentially overcoming the limitations of existing treatment approaches and significantly enhancing long-term patient prognosis.</p>","PeriodicalId":18433,"journal":{"name":"Medical Oncology","volume":"42 8","pages":"295"},"PeriodicalIF":2.8,"publicationDate":"2025-06-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144528681","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Medical OncologyPub Date : 2025-06-28DOI: 10.1007/s12032-025-02846-7
Deepshikha Rathore, Sweny Jain, Shanaya Patel, Nandani Dharwal, Nirali Shukla, Chirag Desai, Jigna Shah, Heena V Dave
{"title":"MicroRNA-driven organ-specific metastasis in triple-negative breast cancer: biogenesis, mechanisms, and therapeutic approaches.","authors":"Deepshikha Rathore, Sweny Jain, Shanaya Patel, Nandani Dharwal, Nirali Shukla, Chirag Desai, Jigna Shah, Heena V Dave","doi":"10.1007/s12032-025-02846-7","DOIUrl":"https://doi.org/10.1007/s12032-025-02846-7","url":null,"abstract":"<p><p>MicroRNAs or miRNAs are characterized as non-coding RNAs, and these are the critical regulators of gene expression post-transcription. Emerging evidence highlights their important role in both the progression and repression of cancer. miRNAs, found in both primary tumor cells and circulation, not only regulate primary tumor development but also regulate metastatic progression by affecting the behavior of primary tumor cells and resident cells at distant organs. Metastasis remains a major clinical challenge in cancer treatment, which limits the therapeutic efficacy and reduces patient survival significantly. This challenge is particularly evident in triple-negative breast cancers (TNBC). This malignancy responds poorly to current chemotherapies and lacks reliable biomarkers, thus, it lacks targeted therapy options. Notably, TNBC exhibits a high propensity for metastasis to critical organs such as the brain, bone, liver, and lung. The process of metastasis of TNBC primary tumor remains unclear, which highlights the critical need to uncover its molecular insights and organ-specific biomarkers. As key regulators of metastasis, microRNAs play a vital role in TNBC progression by modulating gene expression in primary tumor cells and functioning as circulating messenger molecules that affect distant metastatic sites. Furthermore, their potential as therapeutic targets are currently being explored in preclinical models, including the development of anti-miRNA oligonucleotides and miRNA mimics. Overall, this review offers an in-depth mechanism of TNBC metastasis and the molecular mechanisms driving the brain, bone, liver, and lung metastasis. The key focus is on understanding the role of miRNAs in directing organ-specific metastasis and evaluating their potential as biomarker for diagnosis and targeted treatment.</p>","PeriodicalId":18433,"journal":{"name":"Medical Oncology","volume":"42 8","pages":"296"},"PeriodicalIF":2.8,"publicationDate":"2025-06-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144528663","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Medical OncologyPub Date : 2025-06-28DOI: 10.1007/s12032-025-02853-8
Jing-Yi Liu, Xuan-Hao Liu, Nian-Nian Zhong, Yao Xiao, Guang-Rui Wang, Bing Liu, Lin-Lin Bu
{"title":"Barriers in bone tumor treatment: the emerging role of drug delivery systems.","authors":"Jing-Yi Liu, Xuan-Hao Liu, Nian-Nian Zhong, Yao Xiao, Guang-Rui Wang, Bing Liu, Lin-Lin Bu","doi":"10.1007/s12032-025-02853-8","DOIUrl":"https://doi.org/10.1007/s12032-025-02853-8","url":null,"abstract":"<p><p>Bone tumors are rare and diverse neoplasms with local and systemic impacts. Current therapies like surgery, chemotherapy, radiotherapy, targeted therapy, and immunotherapy have had mixed success; significant hurdles persist. Surgery may cause a series of complications and has limited applicability. Systemic chemotherapy notably has a narrow therapeutic window. Besides, the bone microenvironment is extremely complex. These aspects fuel tumor growth and hinder drug delivery. Innovations in drug delivery systems enable spatiotemporal drug control, enhancing tumor accumulation while minimizing systemic toxicity. Examples include bone-targeted nanoparticles (e.g., bisphosphonate-modified carriers), stimuli-responsive systems (pH/redox-sensitive release), and hybrid platforms (e.g., nanocarriers co-loading chemotherapeutics and immunomodulators). These strategies address tumor heterogeneity and microenvironmental barriers, demonstrating improved efficacy in preclinical models. In this review, we comprehensively summarize the most recent advancements in drug delivery systems designed for bone tumor therapy. The key approaches discussed are as follows: drug combination strategies, metal-organic frameworks and inorganic nanomaterials, specificity of bone tissue, bone-targeting strategies, organic combination of response strategies, nanocarrier-based delivery systems, and emerging technologies. Despite progress, challenges like scalability, biocompatibility, and regulatory hurdles limit clinical translation. Future directions include integrating AI for optimized drug delivery system design, developing personalized/patient-specific delivery methods, and exploring combinatorial approaches. This review synthesizes cutting-edge DDS technologies and addresses translational challenges, providing actionable insights to bridge laboratory discoveries and clinical applications.</p>","PeriodicalId":18433,"journal":{"name":"Medical Oncology","volume":"42 8","pages":"294"},"PeriodicalIF":2.8,"publicationDate":"2025-06-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144528660","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Role of Tigecycline in radiation sensitivity in colorectal cancer cell line.","authors":"Sepideh Hassanpour Khodaei, Shahnaz Sabetkam, Zeinab Mazloumi, Khadijeh Dizaji Asl, Ali Rafat","doi":"10.1007/s12032-025-02869-0","DOIUrl":"https://doi.org/10.1007/s12032-025-02869-0","url":null,"abstract":"<p><p>Colorectal cancer (CRC) is the third most common cancer worldwide. Despite the availability of conventional treatment options, challenges such as limited therapeutic alternatives and poor prognosis persist. Tigecycline is an inhibitor drug that blocks mitochondria-related proliferation in cancer cells and may enhance disease-free survival. We examined how Tigecycline plus radiotherapy affects CRC cell growth. Additionally, the study evaluated the expression of cancer cell markers and related genes. HCT-116 colorectal cancer cell lines were treated with the IC50 dose of Tigecycline and subsequently exposed to radiation. To evaluate the proliferation rate after treatment with IC50 dose, Ki-67 expression was analyzed using flow cytometry. Finally, the expression levels of cancer cell markers CD44 and the related genes SOX2 and OCT4 were analyzed. The results showed that Tigecycline reduced HCT-116 cell viability in a dose-dependent manner. IC50 dose of determined 93 um. Although the expression level of CD44 decreased significantly in the combination therapy groups, no significant difference was observed in Ki-67 expression among the treatment groups. Data are representative of three independent experiments. *P < 0.05. The findings suggest that combining Tigecycline with radiotherapy may have potential as a complementary strategy for colorectal cancer treatment, warranting further investigation.</p>","PeriodicalId":18433,"journal":{"name":"Medical Oncology","volume":"42 8","pages":"292"},"PeriodicalIF":2.8,"publicationDate":"2025-06-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144528680","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}