首页> 美国卫生研究院文献>Viruses >Locally-Delivered T-Cell-Derived Cellular Vehicles Efficiently Track and Deliver Adenovirus Delta24-RGD to Infiltrating Glioma
【2h】

Locally-Delivered T-Cell-Derived Cellular Vehicles Efficiently Track and Deliver Adenovirus Delta24-RGD to Infiltrating Glioma

机译:本地交付的T细胞衍生的细胞载体有效地跟踪和交付腺病毒Delta24-RGD到浸润性胶质瘤

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

Oncolytic adenoviral vectors are a promising alternative for the treatment of glioblastoma. Recent publications have demonstrated the advantages of shielding viral particles within cellular vehicles (CVs), which can be targeted towards the tumor microenvironment. Here, we studied T-cells, often having a natural capacity to target tumors, for their feasibility as a CV to deliver the oncolytic adenovirus, Delta24-RGD, to glioblastoma. The Jurkat T-cell line was assessed in co-culture with the glioblastoma stem cell (GSC) line, MGG8, for the optimal transfer conditions of Delta24-RGD in vitro. The effect of intraparenchymal and tail vein injections on intratumoral virus distribution and overall survival was addressed in an orthotopic glioma stem cell (GSC)-based xenograft model. Jurkat T-cells were demonstrated to facilitate the amplification and transfer of Delta24-RGD onto GSCs. Delta24-RGD dosing and incubation time were found to influence the migratory ability of T-cells towards GSCs. Injection of Delta24-RGD-loaded T-cells into the brains of GSC-bearing mice led to migration towards the tumor and dispersion of the virus within the tumor core and infiltrative zones. This occurred after injection into the ipsilateral hemisphere, as well as into the non-tumor-bearing hemisphere. We found that T-cell-mediated delivery of Delta24-RGD led to the inhibition of tumor growth compared to non-treated controls, resulting in prolonged survival (p = 0.007). Systemic administration of virus-loaded T-cells resulted in intratumoral viral delivery, albeit at low levels. Based on these findings, we conclude that T-cell-based CVs are a feasible approach to local Delta24-RGD delivery in glioblastoma, although efficient systemic targeting requires further improvement.
机译:溶瘤腺病毒载体是治疗胶质母细胞瘤的有前途的替代方法。最近的出版物已经证明了在细胞载体(CVs)中屏蔽病毒颗粒的优势,这种载体可以针对肿瘤微环境。在这里,我们研究了通常具有天然能力靶向肿瘤的T细胞作为CV将溶瘤腺病毒Delta24-RGD递送至胶质母细胞瘤的可行性。与成胶质细胞瘤干细胞(GSC)系MGG8共培养评估Jurkat T细胞系的Delta24-RGD体外最佳转移条件。在基于原位神经胶质瘤干细胞(GSC)的异种移植模型中,研究了实质内和尾静脉注射对肿瘤内病毒分布和总体存活的影响。 Jurkat T细胞被证明可以促进Delta24-RGD的扩增和转移到GSC上。发现Delta24-RGD的剂量和孵育时间会影响T细胞向GSC迁移的能力。将装有Delta24-RGD的T细胞注射到带有GSC的小鼠的大脑中,导致向肿瘤的迁移以及病毒在肿瘤核心和浸润区内的扩散。这发生在注射到同侧半球以及非肿瘤半球中之后。我们发现,与未治疗的对照组相比,T细胞介导的Delta24-RGD递送导致肿瘤生长受到抑制,从而延长了生存期(p = 0.007)。病毒载T细胞的全身给药导致肿瘤内病毒传递,尽管水平较低。基于这些发现,我们得出结论,基于T细胞的CV是胶质母细胞瘤中局部Delta24-RGD递送的可行方法,尽管有效的全身靶向治疗需要进一步改善。

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