首页> 美国卫生研究院文献>Neuro-Oncology >Cyclopamine cooperates with EGFR inhibition to deplete stem-like cancer cells in glioblastoma-derived spheroid cultures
【2h】

Cyclopamine cooperates with EGFR inhibition to deplete stem-like cancer cells in glioblastoma-derived spheroid cultures

机译:环巴胺与EGFR抑制协同作用以耗尽胶质母细胞瘤来源的球体培养物中的干样癌细胞

代理获取
本网站仅为用户提供外文OA文献查询和代理获取服务,本网站没有原文。下单后我们将采用程序或人工为您竭诚获取高质量的原文,但由于OA文献来源多样且变更频繁,仍可能出现获取不到、文献不完整或与标题不符等情况,如果获取不到我们将提供退款服务。请知悉。

摘要

Putative cancer stem cells have been identified in glioblastoma (GBM), associated with resistance to conventional therapies. Overcoming this resistance is a major challenge to manage this deadly brain tumor. Epidermal growth factor receptor (EGFR) is commonly amplified, over-expressed, and/or mutated in GBM, making it a compelling target for therapy. This study investigates the behavior of 3 primary neurosphere (NS) cell lines and their adherent counterparts originated from human GBM resections, when treated with EGFR–tyrosine kinase inhibitor erlotinib, associated or not with cyclopamine, a hedgehog pathway inhibitor. Adherent cells cultured in the presence of serum expressed the glial fibrillary acidic protein, whereas NS-forming cells cultured in serum-free medium expressed CD133, nestin, and Oct-4, markers of neural stem and progenitor cells. For the 3 adherent cell lines, erlotinib has a moderate effect (50% inhibitory concentration [IC50], >10 µM). Conversely, erlotinib induced a strong cell growth inhibition (IC50, <1 µM) on NS-forming cells, related to the EGFR gene amplification and EGFR protein expression. A short exposure to erlotinib reduced nestin-positive cell proliferation, but NS-initiating activity and self-renewal were not altered. EGFR pathway seems essential for GBM progenitor cell proliferation but dispensable for cancer stem-like cell self-renewal. Inhibition of hedgehog pathway with cyclopamine was evaluated in association with erlotinib on NS growth. Although each drug separately had no effect on sphere initiation, their combination significantly decreased the sphere number (P < .001). Our findings show synergic efficiency for erlotinib-cyclopamine association and provide a suitable in vitro model to explore drug combinations on GBM cells.
机译:在胶质母细胞瘤(GBM)中已鉴定出推定的癌症干细胞,与传统疗法的耐药性有关。克服这种抵抗力是应对这种致命脑瘤的主要挑战。表皮生长因子受体(EGFR)通常在GBM中扩增,过度表达和/或突变,使其成为引人注目的治疗靶标。这项研究调查了当使用EGFR-酪氨酸激酶抑制剂厄洛替尼治疗或与环巴胺(一种刺猬蛋白途径抑制剂)相关或不相关时,三种主要神经球(NS)细胞系及其粘附对应物源自人GBM切除的行为。在血清存在下培养的贴壁细胞表达神经胶质纤维酸性蛋白,而在无血清培养基中培养的NS形成细胞表达CD133,巢蛋白和Oct-4(神经干细胞和祖细胞的标志物)。对于3种贴壁细胞系,厄洛替尼具有中等作用(50%抑制浓度[IC50],> 10 µM)。相反,厄洛替尼对NS形成细胞具有强烈的细胞生长抑制作用(IC50,<1 µM),这与EGFR基因扩增和EGFR蛋白表达有关。短期暴露于厄洛替尼会降低巢蛋白阳性细胞的增殖,但不会改变NS起始活性和自我更新。 EGFR途径对于GBM祖细胞增殖似乎必不可少,但对于癌症干样细胞自我更新是必不可少的。与厄洛替尼对NS生长的抑制作用一起评估了环巴胺对刺猬通路的抑制作用。尽管每种药物分别对球的起始没有影响,但它们的组合显着降低了球的数量(P <.001)。我们的发现显示了厄洛替尼-环巴胺缔合的协同效率,并提供了合适的体外模型来探索GBM细胞上的药物组合。

著录项

相似文献

  • 外文文献
  • 中文文献
  • 专利
代理获取

客服邮箱:kefu@zhangqiaokeyan.com

京公网安备:11010802029741号 ICP备案号:京ICP备15016152号-6 六维联合信息科技 (北京) 有限公司©版权所有
  • 客服微信

  • 服务号