Zhou Qiong, Wang Rui
Laboratory of Medical Oncology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China.
Front Cell Dev Biol. 2025 Aug 13;13:1637767. doi: 10.3389/fcell.2025.1637767. eCollection 2025.
BACKGROUND: Hepatocellular carcinoma (HCC) is a major cause of cancer-related mortality worldwide. It is often diagnosed at advanced stages, which limits treatment options. Although Donafenib is a standard therapy for advanced HCC, its effectiveness is often reduced by treatment failures. Alisertib, an Aurora-A kinase inhibitor, shows promise in enhancing the cytotoxic effects of Donafenib. This study investigates the combined therapeutic effects of these two agents. METHODS: Synergistic cytotoxicity was assessed via CCK-8 and colony formation assays. Ferroptosis activation was quantified through flow cytometry, lipid peroxidation, and measurements of reactive oxygen species (ROS), intracellular Fe, and GSH/GSSG. Mechanistic studies involved immunofluorescence for NF-κB/p65 localization, along with Western blotting, qPCR, and dual-luciferase reporter assays to evaluate protein and gene expression. Chromatin immunoprecipitation (ChIP) experiments were performed to analyze the binding of NF-κB/p65 to its endogenous promoters. xenografts were established to evaluate the antitumor efficacy and potential side effects of the combination treatment, supported by histological and immunohistochemical analyses. RESULTS: Optimal synergistic concentrations (Alisertib 2.5 µM + Donafenib 10 µM for HCCLM3; 5 µM for Huh7) induced profound ferroptotic cascades, evidenced by elevated ROS, lipid peroxides, and Fe accumulation concurrent with GSH depletion. The co-treatment potently inhibited p65 nuclear translocation while stabilizing IκBα, thereby suppressing NRF2-mediated antioxidant transcription. Xenograft models demonstrated marked tumor volume reduction with preserved organ architecture and hematological parameters, confirming clinical translatability. CONCLUSION: Alisertib is identified as a potent enhancer of Donafenib-induced ferroptosis through inhibition of the NF-κB/NRF2 pathway. This suggests a novel combinatorial strategy that targets ferroptosis through NF-κB inhibition. Further research is needed to translate these promising results into clinical practice.
背景:肝细胞癌(HCC)是全球癌症相关死亡的主要原因。它通常在晚期被诊断出来,这限制了治疗选择。尽管多纳非尼是晚期HCC的标准治疗方法,但其有效性常常因治疗失败而降低。阿利塞替尼是一种极光激酶A抑制剂,在增强多纳非尼的细胞毒性作用方面显示出前景。本研究调查了这两种药物的联合治疗效果。 方法:通过CCK-8和集落形成试验评估协同细胞毒性。通过流式细胞术、脂质过氧化以及活性氧(ROS)、细胞内铁和谷胱甘肽/氧化型谷胱甘肽(GSH/GSSG)的测量来量化铁死亡激活。机制研究包括用于NF-κB/p65定位的免疫荧光,以及用于评估蛋白质和基因表达的蛋白质印迹、qPCR和双荧光素酶报告基因试验。进行染色质免疫沉淀(ChIP)实验以分析NF-κB/p65与其内源性启动子的结合。建立异种移植模型以评估联合治疗的抗肿瘤疗效和潜在副作用,并辅以组织学和免疫组织化学分析。 结果:最佳协同浓度(HCCLM3为阿利塞替尼2.5 μM + 多纳非尼10 μM;Huh7为5 μM)诱导了深刻的铁死亡级联反应,表现为ROS、脂质过氧化物升高以及铁积累,同时谷胱甘肽耗竭。联合治疗有效抑制了p65核转位,同时稳定了IκBα,从而抑制了NRF2介导的抗氧化转录。异种移植模型显示肿瘤体积显著减小,器官结构和血液学参数得以保留,证实了临床可转化性。 结论:阿利塞替尼被确定为通过抑制NF-κB/NRF2途径增强多纳非尼诱导的铁死亡的有效增强剂。这提示了一种通过抑制NF-κB靶向铁死亡的新型联合策略。需要进一步研究将这些有前景的结果转化为临床实践。
Front Cell Dev Biol. 2025-8-13
JAMA Netw Open. 2024-10-1
Signal Transduct Target Ther. 2024-10-14
Cell Death Differ. 2024-4
CA Cancer J Clin. 2024
Biomed Pharmacother. 2023-9
Acta Pharm Sin B. 2023-7
Colloids Surf B Biointerfaces. 2023-7
Curr Treat Options Oncol. 2023-7