首页> 美国卫生研究院文献>Proceedings of the National Academy of Sciences of the United States of America >Dual bioluminescence and near-infrared fluorescence monitoring to evaluate spherical nucleic acid nanoconjugate activity in vivo
【2h】

Dual bioluminescence and near-infrared fluorescence monitoring to evaluate spherical nucleic acid nanoconjugate activity in vivo

机译:双重生物发光和近红外荧光监测可评估体内球形核酸纳米共轭物的活性

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

RNA interference (RNAi)-based gene regulation platforms have shown promise as a novel class of therapeutics for the precision treatment of cancer. Techniques in preclinical evaluation of RNAi-based nanoconjugates have yet to allow for optimization of their gene regulatory activity. We have developed spherical nucleic acids (SNAs) as a blood–brain barrier-/blood–tumor barrier-penetrating nanoconjugate to deliver small interfering (si) and micro (mi)RNAs to intracranial glioblastoma (GBM) tumor sites. To identify high-activity SNA conjugates and to determine optimal SNA treatment regimens, we developed a reporter xenograft model to evaluate SNA efficacy in vivo. Engrafted tumors stably coexpress optical reporters for luciferase and a near-infrared (NIR) fluorescent protein (iRFP670), with the latter fused to the DNA repair protein O6-methylguanine-DNA-methyltransferase (MGMT). Using noninvasive imaging of animal subjects bearing reporter-modified intracranial xenografts, we quantitatively assessed MGMT knockdown by SNAs composed of MGMT-targeting siRNA duplexes (siMGMT-SNAs). We show that systemic administration of siMGMT-SNAs via single tail vein injection is capable of robust intratumoral MGMT protein knockdown in vivo, with persistent and SNA dose-dependent MGMT silencing confirmed by Western blotting of tumor tissue ex vivo. Analyses of SNA biodistribution and pharmacokinetics revealed rapid intratumoral uptake and significant intratumoral retention that increased the antitumor activity of coadministered temozolomide (TMZ). Our study demonstrates that dual noninvasive bioluminescence and NIR fluorescence imaging of cancer xenograft models represents a powerful in vivo strategy to identify RNAi-based nanotherapeutics with potent gene silencing activity and will inform additional preclinical and clinical investigations of these constructs.
机译:基于RNA干扰(RNAi)的基因调控平台已被证明是一种用于精确治疗癌症的新型疗法。基于RNAi的纳米缀合物的临床前评估技术尚未允许优化其基因调控活性。我们已经开发出球形核酸(SNA)作为穿透血脑屏障/血液-肿瘤屏障的纳米共轭物,以向颅内胶质母细胞瘤(GBM)肿瘤部位提供小的干扰(si)和微(mi)RNA。为了鉴定高活性的SNA缀合物并确定最佳的SNA治疗方案,我们开发了一种报告基因异种移植模型来评估SNA在体内的疗效。植入的肿瘤稳定地共表达荧光素酶和近红外(NIR)荧光蛋白(iRFP670)的光学报告基因,后者与DNA修复蛋白O 6 -甲基鸟嘌呤-DNA-甲基转移酶(MGMT)融合。使用携带报道基因修饰的颅内异种移植物的动物受试者的非侵入性成像,我们定量评估了由靶向MGMT的siRNA双链体(siMGMT-SNA)组成的SNA的MGMT敲低。我们显示,通过单尾静脉注射对siMGMT-SNAs进行全身给药,能够在体内增强肿瘤内MGMT蛋白的表达水平,并通过肿瘤组织的Western blotting证实了持久性和SNA剂量依赖性MGMT沉默。 SNA生物分布和药代动力学的分析表明,快速的肿瘤内摄取和显着的肿瘤内滞留增加了并用替莫唑胺(TMZ)的抗肿瘤活性。我们的研究表明,癌症异种移植模型的双重非侵入性生物发光和NIR荧光成像代表了一种有力的体内策略,可用于鉴定具有有效基因沉默活性的基于RNAi的纳米疗法,并将为这些构建物的临床前和临床研究提供参考。

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