首页> 美国卫生研究院文献>Neuro-Oncology >OS09.3 Synergistic activity of NKG2D-based chimeric antigen receptor (CAR)-T cells and radiotherapy against glioma
【2h】

OS09.3 Synergistic activity of NKG2D-based chimeric antigen receptor (CAR)-T cells and radiotherapy against glioma

机译:OS09.3基于NKG2D的嵌合抗原受体(CAR)-T细胞的协同活性和对神经胶质瘤的放射治疗

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。
获取外文期刊封面目录资料

摘要

>Introduction: Glioblastoma is the most common primary brain tumor in adults and virtually always lethal despite a multimodal treatment regimen including surgery, chemotherapy and radiotherapy. Therefore, novel treatment modalities are needed. Adoptive immunotherapy with genetically engineered T cells that express a chimeric antigen receptor (CAR) to recognize and eliminate tumors in a MHC-independent manner is an emerging strategy that has led to remarkable responses in hematologic malignancies. CARs that target the natural-killer group 2-member D (NKG2D) system elegantly use the promiscuous binding properties of the NKG2D receptor that binds to several tumor-associated ligands. NKG2D-based CAR T cells have never been investigated against glioma and it is unknown if this strategy can overcome the challenges of this tumor entity and if it could even be implemented in established conventional treatment regimens. >Material and Methods: CAR T cells were generated by retroviral transduction of splenocytes to express a NKG2D-based CAR (chNKG2D) or wildtype NKG2D (wtNKG2D). For in vitro studies, murine glioma cells (GL-261, SMA-560, SMA-540, SMA-407) were co-cultured with chNKG2D or wtNKG2D T cells and we assessed the cytolytic activity and cytokine production. For in vivo studies, we used GL-261 cells syngeneic to C57BL/6 mice and monitored tumor growth by magnetic resonance imaging. We tracked CAR T cells in vivo with fluorescence molecular tomography (FMT), flow cytometry and immunohistochemistry. To study the combination of radiotherapy with CAR T cells, mice received a single subtherapeutic dose of local irradiation. >Results: In all murine glioma cell lines, chNKG2D T cells had a significantly higher specific cytolytic activity compared to wtNKG2D T cells. Furthermore, chNKG2D T cells produced more IFN-γ. In vivo, intravenously injected chNKG2D T cells migrated to the orthotopic tumor site, were tolerated without toxicities, prolonged the survival and cured a fraction of tumor-bearing mice. This anti-tumor effect was even more pronounced in case of intratumoral CAR T cell administration. Survivors were long-term protected against tumor re-challenge. Mechanistically, this was not the result of a classical immune memory response, but rather due to local persistence of chNKG2D T cells. Radiotherapy augmented the effect of chNKG2D T cell therapy already after a single application of a subtherapeutic dose. It promoted migration of CAR T cells to the tumor site and the effector function of NKG2D-based CAR T cells. >Conclusion: We provide the first systematical preclinical assessment of NKG2D-based CAR T cells against glioma. This strategy was effective, tolerated without toxicities, conferred long-term protection and was synergistic with radiotherapy. These findings could provide a rationale to test this immunotherapeutic strategy also in human glioma patients.
机译:>简介:成胶质细胞瘤是成人中最常见的原发性脑肿瘤,尽管采取了包括外科手术,化学疗法和放射疗法在内的多种治疗方法,但实际上几乎总是致命的。因此,需要新颖的治疗方式。用表达嵌合抗原受体(CAR)的基因工程T细胞以不依赖MHC的方式识别和消除肿瘤的过继免疫疗法是一种新兴策略,已引起血液系统恶性肿瘤的显着反应。靶向2号天然杀伤分子D(NKG2D)系统的CAR优雅地使用了与几种与肿瘤相关的配体结合的NKG2D受体的混杂结合特性。从未对基于NKG2D的CAR T细胞针对神经胶质瘤进行过研究,并且未知该策略能否克服该肿瘤实体的挑战,甚至是否可以在既定的常规治疗方案中实施。 >材料和方法:CAR T细胞是通过逆转录病毒转导脾细胞产生的,以表达基于NKG2D的CAR(chNKG2D)或野生型NKG2D(wtNKG2D)。为了进行体外研究,将鼠神经胶质瘤细胞(GL-261,SMA-560,SMA-540,SMA-407)与chNKG2D或wtNKG2D T细胞共培养,我们评估了细胞溶解活性和细胞因子的产生。对于体内研究,我们使用了与C57BL / 6小鼠同系的GL-261细胞,并通过磁共振成像监测了肿瘤的生长。我们通过荧光分子层析成像(FMT),流式细胞仪和免疫组织化学追踪了体内的CAR T细胞。为了研究放疗与CAR T细胞的组合,小鼠接受了单一治疗剂量的局部照射。 >结果:在所有鼠神经胶质瘤细胞系中,与wtNKG2D T细胞相比,chNKG2D T细胞具有明显更高的比细胞溶解活性。此外,chNKG2D T细胞产生更多的IFN-γ。在体内,静脉注射的chNKG2D T细胞迁移到原位肿瘤部位,可以耐受,没有毒性,延长了存活期,治愈了一部分荷瘤小鼠。在肿瘤内注射CAR T细胞的情况下,这种抗肿瘤作用甚至更加明显。幸存者受到长期保护,免受肿瘤再攻击。从机制上讲,这不是经典免疫记忆反应的结果,而是由于chNKG2D T细胞的局部持久性。单次应用亚治疗剂量后,放射疗法已经增强了chNKG2D T细胞疗法的效果。它促进了CAR T细胞向肿瘤部位的迁移以及基于NKG2D的CAR T细胞的效应功能。 >结论:我们提供了基于NKG2D的CAR T细胞针对神经胶质瘤的首次系统性临床前评估。该策略有效,可耐受且无毒性,可长期保护,并且与放疗协同。这些发现可以为在人脑胶质瘤患者中测试这种免疫治疗策略提供理论依据。

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