...
首页> 外文期刊>International Journal of Nanomedicine >GEM-loaded magnetic albumin nanospheres modified with cetuximab for simultaneous targeting, magnetic resonance imaging, and double-targeted thermochemotherapy of pancreatic cancer cells
【24h】

GEM-loaded magnetic albumin nanospheres modified with cetuximab for simultaneous targeting, magnetic resonance imaging, and double-targeted thermochemotherapy of pancreatic cancer cells

机译:载有GEM的西妥昔单抗修饰的磁性白蛋白纳米球用于胰腺癌细胞的同时靶向,磁共振成像和双靶热化学疗法

获取原文

摘要

Background: Targeted delivery is a promising strategy to improve the diagnostic imaging and therapeutic effect of cancers. In this paper, novel cetuximab (C225)-conjugated, gemcitabine (GEM)-containing magnetic albumin nanospheres (C225-GEM/MANs) were fabricated and applied as a theranostic nanocarrier to conduct simultaneous targeting, magnetic resonance imaging (MRI), and double-targeted thermochemotherapy against pancreatic cancer cells. Methods: Fe3O4 nanoparticles (NPs) and GEM co-loaded albumin nanospheres (GEM/MANs) were prepared, and then C225 was further conjugated to synthesize C225-GEM/MANs. Their morphology, mean particle size, GEM encapsulation ratio, specific cell-binding ability, and thermal dynamic profiles were characterized. The effects of discriminating different EGFR-expressing pancreatic cancer cells (AsPC-1 and MIA PaCa-2) and monitoring cellular targeting effects were assessed by targeted MRI. Lastly, the antitumor efficiency of double/C225/magnetic-targeted and nontargeted thermochemotherapy was compared with chemotherapy alone using 3-(4, 5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) and flow cytometry (FCM) assay. Results: When treated with targeted nanospheres, AsPC-1 cells showed a significantly less intense MRI T2 signal than MIA PaCa-2 cells, while both cells had similar signal strength when incubated with nontargeted nanospheres. T2 signal intensity was significantly lower when magnetic and C225 targeting were combined, rather than used alone. The inhibitory and apoptotic rates of each thermochemotherapy group were significantly higher than those of the chemotherapy-alone groups. Additionally, both MTT and FCM analysis verified that double-targeted thermochemotherapy had the highest targeted killing efficiency among all groups. Conclusion: The C225-GEM/MANs can distinguish various EGFR-expressing live pancreatic cancer cells, monitor diverse cellular targeting effects using targeted MRI imaging, and efficiently mediate double-targeted thermochemotherapy against pancreatic cancer cells.
机译:背景:靶向递送是改善癌症的诊断成像和治疗效果的一种有前途的策略。在本文中,新型西妥昔单抗(C225)偶联,包含吉西他滨(GEM)的磁性白蛋白纳米球(C225-GEM / MANs)的制备和应用作为治疗诊断纳米载体进行同时靶向,磁共振成像(MRI)和双重针对胰腺癌细胞的靶向热化学疗法。方法:制备Fe3O4纳米颗粒(NPs)和GEM共载白蛋白纳米球(GEM / MANs),然后进一步偶联C225合成C225-GEM / MANs。表征了它们的形态,平均粒径,GEM包封率,比细胞结合能力和热动力学曲线。通过靶向MRI评估了区分不同表达EGFR的胰腺癌细胞(AsPC-1和MIA PaCa-2)和监测细胞靶向作用的效果。最后,将双重/ C225 /磁性靶向和非靶向热化学疗法与单独使用3-(4,5-二甲基-2-噻唑基)-2,5-二苯基-2H-四唑溴化物(MTT)的化疗的抗肿瘤效率进行了比较。流式细胞仪(FCM)分析。结果:当用靶向纳米球治疗时,AsPC-1细胞显示出比MIA PaCa-2细胞明显更低的MRI T2信号,而与非靶向纳米球一起孵育时,两种细胞的信号强度相似。当将磁性靶向和C225靶向相结合而不是单独使用时,T2信号强度显着降低。每个热化学疗法组的抑制率和凋亡率均显着高于单纯化学疗法组。此外,MTT和FCM分析均证实双靶热化学疗法在所有组中具有最高的靶杀灭效率。结论:C225-GEM / MANs可以区分各种表达EGFR的活胰腺癌细胞,使用靶向MRI成像监测多种细胞靶向作用,并有效介导针对胰腺癌细胞的双重靶向热化学疗法。

著录项

相似文献

  • 外文文献
  • 中文文献
  • 专利
获取原文

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