首页> 美国卫生研究院文献>International Journal of Nanomedicine >Co-delivery nanocarriers targeting folate receptor and encapsulating 2-deoxyglucose and α-tocopheryl succinate enhance anti-tumor effect in vivo
【2h】

Co-delivery nanocarriers targeting folate receptor and encapsulating 2-deoxyglucose and α-tocopheryl succinate enhance anti-tumor effect in vivo

机译:靶向叶酸受体并包裹2-脱氧葡萄糖和琥珀酸α-生育酚酸酯的共递送纳米载体在体内增强了抗肿瘤作用

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

A combination administration of chemical agents was highlighted to treat tumors. Recently, tumor cell has been found to be different from normal cell in metabolic manner. Most of cancer cells prefer aerobic glycolysis to mitochondrial oxidative phosphorylation (OXPHOS) to satisfy energy and biomass synthesis requirement to survive, grow and proliferate, which provides novel and potential therapeutic targets for chemotherapy. Here, 2-deoxy-d-glucose (2-DG), a potent inhibitor of glucose metabolism, was used to inhibit glycolysis of tumor cells; α-tocopheryl succinate (α-TOS), a water-insoluble vitamin E derivative, was chosen to suppress OXPHOS. Our data demonstrated that the combination treatment of 2-DG and α-TOS could significantly promote the anti-tumor efficiency in vitro compared with administration of the single drug. In order to maximize therapeutic activity and minimize negative side effects, a co-delivery nanocarrier targeting folate receptor (FR) was developed to encapsulate 2-DG and α-TOS simultaneously based on our previous work. Transmission electron microscope, dynamic light scattering method and UV-visible spectrophotometers were used to investigate morphology, size distribution and loading efficiency of the α-TOS-2-DG-loaded and FR-targeted nanoparticles (TDF NPs). The TDF NPs were found to possess a layer-by-layer shape, and the dynamic size was <100 nm. The final encapsulation efficiencies of α-TOS and 2-DG in TDF NPs were 94.3%±1.3% and 61.7%±7.7% with respect to drug-loading capacities of 8.9%±0.8% and 13.2%±2.6%, respectively. Almost no α-TOS release was found within 80 h, and release of 2-DG was sustained and slow within 72 h. The results of FR binding assay and fluorescence biodistribution revealed that TDF NPs could target FR highly expressed on tumor cell in vitro and in vivo. Further, in vivo anti-tumor experiments showed that TDF NPs had an improved biological function with less toxicity. Thus, our work indicates that the co-delivery TDF NPs have a great potential in tumor therapy.
机译:强调了化学药剂的联合施用以治疗肿瘤。最近,已经发现肿瘤细胞在代谢方式上与正常细胞不同。大多数癌细胞更喜欢有氧糖酵解而不是线粒体氧化磷酸化(OXPHOS),以满足能量和生物质合成生存,生长和增殖的需求,这为化学疗法提供了新的潜在治疗靶点。在这里,使用2-脱氧-d-葡萄糖(2-DG)(一种有效的葡萄糖代谢抑制剂)来抑制肿瘤细胞的糖酵解。选择了水不溶性维生素E衍生物α-生育酚琥珀酸酯(α-TOS)来抑制OXPHOS。我们的数据表明,与单药给药相比,联合使用2-DG和α-TOS可以显着提高体外抗肿瘤效果。为了最大化治疗活性并最大程度减少不良副作用,在我们之前的工作基础上,开发了靶向叶酸受体(FR)的共递送纳米载体,可同时包封2-DG和α-TOS。用透射电子显微镜,动态光散射法和紫外可见分光光度计研究了负载α-TOS-2-DG和靶向FR的纳米粒子(TDF NPs)的形貌,尺寸分布和负载效率。发现TDF NP具有逐层形状,并且动态尺寸<100nm。 TDF NPs中最终的α-TOS和2-DG包封效率分别为94.3%±1.3%和61.7%±7.7%,载药量分别为8.9%±0.8%和13.2%±2.6%。在80小时内几乎没有发现α-TOS释放,而72小时内2-DG的释放持续且缓慢。 FR结合测定和荧光生物分布的结果表明,TDF NPs可以靶向在体内和体外高表达在肿瘤细胞上的FR。此外,体内抗肿瘤实验表明,TDF NP具有改善的生物学功能,且毒性较小。因此,我们的工作表明,共同递送的TDF NP在肿瘤治疗中具有巨大的潜力。

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