Suppr超能文献

新辅助奈拉替尼促进铁死亡并抑制自发 HER2 乳腺癌转移的新型同源模型中的脑转移。

Neoadjuvant neratinib promotes ferroptosis and inhibits brain metastasis in a novel syngeneic model of spontaneous HER2 breast cancer metastasis.

机构信息

Matrix Microenvironment & Metastasis Laboratory, Olivia Newton-John Cancer Research Institute, 145 Studley Road, Heidelberg, VIC, 3084, Australia.

School of Cancer Medicine, La Trobe University, Bundoora, VIC, 3086, Australia.

出版信息

Breast Cancer Res. 2019 Aug 13;21(1):94. doi: 10.1186/s13058-019-1177-1.

Abstract

BACKGROUND

Human epidermal growth factor receptor-2 (HER2)-targeted therapies prolong survival in HER2-positive breast cancer patients. Benefit stems primarily from improved control of systemic disease, but up to 50% of patients progress to incurable brain metastases due to acquired resistance and/or limited permeability of inhibitors across the blood-brain barrier. Neratinib, a potent irreversible pan-tyrosine kinase inhibitor, prolongs disease-free survival in the extended adjuvant setting, and several trials evaluating its efficacy alone or combination with other inhibitors in early and advanced HER2-positive breast cancer patients are ongoing. However, its efficacy as a first-line therapy against HER2-positive breast cancer brain metastasis has not been fully explored, in part due to the lack of relevant pre-clinical models that faithfully recapitulate this disease. Here, we describe the development and characterisation of a novel syngeneic model of spontaneous HER2-positive breast cancer brain metastasis (TBCP-1) and its use to evaluate the efficacy and mechanism of action of neratinib.

METHODS

TBCP-1 cells were derived from a spontaneous BALB/C mouse mammary tumour and characterised for hormone receptors and HER2 expression by flow cytometry, immunoblotting and immunohistochemistry. Neratinib was evaluated in vitro and in vivo in the metastatic and neoadjuvant setting. Its mechanism of action was examined by transcriptomic profiling, function inhibition assays and immunoblotting.

RESULTS

TBCP-1 cells naturally express high levels of HER2 but lack expression of hormone receptors. TBCP-1 tumours maintain a HER2-positive phenotype in vivo and give rise to a high incidence of spontaneous and experimental metastases in the brain and other organs. Cell proliferation/viability in vitro is inhibited by neratinib and by other HER2 inhibitors, but not by anti-oestrogens, indicating phenotypic and functional similarities to human HER2-positive breast cancer. Mechanistically, neratinib promotes a non-apoptotic form of cell death termed ferroptosis. Importantly, metastasis assays demonstrate that neratinib potently inhibits tumour growth and metastasis, including to the brain, and prolongs survival, particularly when used as a neoadjuvant therapy.

CONCLUSIONS

The TBCP-1 model recapitulates the spontaneous spread of HER2-positive breast cancer to the brain seen in patients and provides a unique tool to identify novel therapeutics and biomarkers. Neratinib-induced ferroptosis provides new opportunities for therapeutic intervention. Further evaluation of neratinib neoadjuvant therapy is warranted.

摘要

背景

人表皮生长因子受体 2(HER2)靶向治疗可延长 HER2 阳性乳腺癌患者的生存时间。获益主要源于系统疾病控制的改善,但多达 50%的患者因获得性耐药和/或抑制剂通过血脑屏障的通透性有限而进展为不可治愈的脑转移。奈拉替尼是一种有效的不可逆泛酪氨酸激酶抑制剂,可延长辅助治疗中的无病生存期,目前正在进行多项评估其在早期和晚期 HER2 阳性乳腺癌患者中单独使用或与其他抑制剂联合使用的疗效的试验。然而,由于缺乏能够忠实地重现这种疾病的相关临床前模型,其作为 HER2 阳性乳腺癌脑转移一线治疗的疗效尚未得到充分探索。在这里,我们描述了一种新型自发 HER2 阳性乳腺癌脑转移(TBCP-1)的同源模型的开发和特征,并利用该模型来评估奈拉替尼的疗效和作用机制。

方法

TBCP-1 细胞源自自发的 BALB/C 鼠乳腺肿瘤,通过流式细胞术、免疫印迹和免疫组化分析对激素受体和 HER2 表达进行了鉴定。在转移和新辅助治疗环境中评估了奈拉替尼的疗效。通过转录组谱分析、功能抑制测定和免疫印迹研究了其作用机制。

结果

TBCP-1 细胞天然表达高水平的 HER2,但缺乏激素受体的表达。TBCP-1 肿瘤在体内保持 HER2 阳性表型,并在大脑和其他器官中自发和实验性转移的发生率很高。细胞增殖/活力在体外被奈拉替尼和其他 HER2 抑制剂抑制,但不受抗雌激素的抑制,表明与人类 HER2 阳性乳腺癌具有表型和功能上的相似性。从机制上讲,奈拉替尼促进了一种称为铁死亡的非凋亡细胞死亡形式。重要的是,转移测定表明,奈拉替尼能够强烈抑制肿瘤生长和转移,包括脑转移,并延长了生存时间,尤其是在作为新辅助治疗时。

结论

TBCP-1 模型再现了患者中所见的 HER2 阳性乳腺癌自发扩散到大脑的情况,为鉴定新的治疗方法和生物标志物提供了独特的工具。奈拉替尼诱导的铁死亡为治疗干预提供了新的机会。需要进一步评估奈拉替尼新辅助治疗。

https://cdn.ncbi.nlm.nih.gov/pmc/blobs/286c/6693253/1403708a80c7/13058_2019_1177_Fig1_HTML.jpg

文献AI研究员

20分钟写一篇综述,助力文献阅读效率提升50倍。

立即体验

用中文搜PubMed

大模型驱动的PubMed中文搜索引擎

马上搜索

文档翻译

学术文献翻译模型,支持多种主流文档格式。

立即体验