首页> 外文学位 >Enhancing Immunity to Glioma: Modulating the Adaptive Immune Response in the Tumor Microenvironment.
【24h】

Enhancing Immunity to Glioma: Modulating the Adaptive Immune Response in the Tumor Microenvironment.

机译:增强对神经胶质瘤的免疫力:调节肿瘤微环境中的适应性免疫反应。

获取原文
获取原文并翻译 | 示例

摘要

This thesis describes the utilization of two different immunotherapeutic strategies to target glioblastoma (GBM). The first strategy created a novel GBM target by inducing NY-ESO-1 antigen expression with the demethylating agent, decitabine, and targeting that antigen with engineered T cells. Specifically, we utilized human GBM cell cultures to induce expression of the antigen. We evaluated NY-ESO-1 TCR-transduced T cell-mediated GBM tumor cytolysis in a series of in vitro cytotoxicity assays. Following this, we examined the application of this therapy using an intracranially-implanted xenograft model. Our studies demonstrated that decitabine could effectively upregulate NY-ESO-1 both in vivo and in vitro. Engineered T cells were able to induce tumor cytolysis in vitro and were able to traffic to and target tumor in vivo. Tumor-bearing mice receiving adoptive transfer of these engineered T cells demonstrated significantly increased survival over mice that received non-transduced T cells. By inducing expression of a novel target on GBM, we were able to generate a highly specific, anti-GBM immune response. This strategy represented a clinically translatable therapeutic technique for treating patients with GBM.;The second strategy focused on using existing GBM targets to generate an endogenous immune response in a syngeneic, immune competent mouse model. Briefly, we administered an autologous tumor lysate-pulsed dendritic cell (DC) vaccine to produce a glioma-specific immune response. In our studies, the vaccination appeared to be capable of inducing T cell infiltration into tumors; however, in large, established tumors, this infiltrating response was not sufficient to increase mouse survival and provide significant therapeutic benefit. We described the role of the negative costimulatory pathway, programmed death-1/ligand-1 (PD-1/PD-L1) in mitigating T cell activation and memory in a series of in vitro and in vivo studies. We noted that PD-1 blockade with PD-1 mAb was not sufficient to produce a T cell infiltrate. However, when administered with DC vaccination, PD-1 blockade activated the vaccine-generated T cell response in the tumor microenvironment. We found that T cells with PD-1 mAb were able to mediate significant tumor cytolysis when compared to T cells without PD-1 blockade in vitro . The adjuvant administration of PD-1 mAb with the DC vaccine resulted in significant survival benefit over DC vaccine alone in mice bearing large, established gliomas. Additionally, this dual treatment resulted in the increased expression of integrin homing and immunologic memory markers on T cells infiltrating tumor. These findings were corroborated in samples from patient GBM, with PD-1 blockade enhancing the T cell-mediated GBM cytolysis. These findings provided us with a means to both generate and enhance a tumor-specific response.;The mechanism underlying this PD-1/PD-L1-mediated suppression was not fully understood. As such, we proceeded to identify a PD-L1-expressing tumor infiltrating myeloid (TIM) cell population that appeared to dominantly regulate the PD-1/PD-L1 signaling mechanism. Importantly, we determined the role that these cells play in inhibiting the immune response using a series of in vitro and in vivo studies utilizing TIM depletion and PD-1 mAb treatment strategies. We found that depletion of TIMs in both human GBM cultures and murine glioma abolished PD-1/PD-L1-mediated inhibition of T cell activation. Targeting TIMs with colony stimulating factor-1 receptor inhibitor (CSF-1Ri) reduced the TIM population significantly and altered the remaining TIMs such that they demonstrated increased expression of chemotactic factors. While treatment with CSF-1Ri in conjunction with DC vaccine did not alter PD-L1 expression on remaining TIMs, we did note that there was increased TIL infiltration with this dual treatment significantly over DC vaccine alone. These findings suggested that TIMs exert inhibitory effects in the tumor microenvironment in a manner not restricted only to the PD-1/PD-L1 signaling mechanism. We found that the combined treatment of CSF-1Ri and PD-1 mAb with DC vaccination both increased TIL infiltration and activation in the tumor microenvironment. These findings were therapeutically relevant, with tumor-bearing mice receiving all three treatments showing a significant increase in survival over mice receiving each treatment alone. The studies outlined herein elucidated the role that TIMs play in dominantly mediating the PD-1/PD-L1 signaling mechanism to restrict TIL activation, as well as the ability to manipulate this population pharmacologically with clinically accessible agents.;In conclusion, this thesis demonstrates two distinct strategies to generate and enhance an immune response against GBM. In our first strategy, we utilized the adoptive transfer of engineered T cells to selectively target an antigen whose expression we artificially induced in GBM. This technique was largely effective. However, we were interested in directly targeting antigens already expressed by GBM. To that end, we described the utility of DC vaccination in generating an immune response. Further, we delineated the inhibitory mechanisms employed by TIMs in the tumor microenvironment and developed a therapeutic adjuvant to administer with DC vaccination. (Abstract shortened by UMI.).
机译:本文描述了两种不同的免疫治疗策略针对胶质母细胞瘤(GBM)的利用。第一种策略是通过用去甲基化剂地西他滨诱导NY-ESO-1抗原表达,并用工程化T细胞靶向该抗原,从而创建了一种新型的GBM靶标。具体而言,我们利用人GBM细胞培养物诱导抗原表达。我们在一系列体外细胞毒性试验中评估了NY-ESO-1 TCR介导的T细胞介导的GBM肿瘤细胞溶解。之后,我们使用颅内植入异种移植模型检查了该疗法的应用。我们的研究表明,地西他滨可以在体内和体外有效上调NY-ESO-1。工程改造的T细胞能够在体外诱导肿瘤细胞溶解,并能够在体内转运到肿瘤并靶向肿瘤。接受这些工程化T细胞过继转移的荷瘤小鼠比接受非转导T细胞的小鼠存活率显着提高。通过在GBM上诱导新靶标的表达,我们能够产生高度特异性的抗GBM免疫应答。该策略代表了一种可治疗GBM的临床可翻译治疗技术。第二个策略着重于利用现有的GBM靶标在同基因,具有免疫能力的小鼠模型中产生内源性免疫应答。简而言之,我们施用了自体肿瘤裂解物脉冲树突细胞(DC)疫苗,以产生神经胶质瘤特异性免疫反应。在我们的研究中,接种疫苗似乎能够诱导T细胞浸润到肿瘤中。但是,在已确定的大型肿瘤中,这种浸润反应不足以增加小鼠存活率并提供显着的治疗益处。在一系列体外和体内研究中,我们描述了负性共刺激途径,程序性死亡-1 /配体-1(PD-1 / PD-L1)在减轻T细胞活化和记忆中的作用。我们注意到用PD-1 mAb阻断PD-1不足以产生T细胞浸润。但是,当进行DC疫苗接种时,PD-1阻断在肿瘤微环境中激活了疫苗产生的T细胞应答。我们发现与不具有PD-1阻断作用的T细胞相比,具有PD-1单克隆抗体的T细胞能够介导显着的肿瘤细胞溶解。 PD-1 mAb与DC疫苗的佐剂给药在携带大的,已建立的神经胶质瘤的小鼠中比单独使用DC疫苗产生了显着的生存获益。另外,这种双重治疗导致T细胞浸润性肿瘤中整联蛋白归巢和免疫记忆标记物的表达增加。这些发现在来自患者GBM的样品中得到证实,PD-1阻断增强了T细胞介导的GBM细胞溶解。这些发现为我们提供了一种产生和增强肿瘤特异性反应的手段。该PD-1 / PD-L1介导的抑制的机制尚不完全清楚。因此,我们着手确定表达PD-L1的肿瘤浸润性髓样细胞(TIM)细胞群似乎主要调节PD-1 / PD-L1信号传导机制。重要的是,我们使用TIM耗竭和PD-1 mAb治疗策略进行了一系列体外和体内研究,确定了这些细胞在抑制免疫反应中的作用。我们发现在人类GBM文化和小鼠神经胶质瘤中TIM的耗竭消除了PD-1 / PD-L1介导的T细胞活化抑制作用。用集落刺激因子-1受体抑制剂(CSF-1Ri)靶向TIMs可以显着减少TIM种群,并改变剩余的TIMs,从而证明其趋化因子表达增加。虽然用CSF-1Ri联合DC疫苗治疗不会改变剩余TIM上的PD-L1表达,但我们确实注意到,与单独使用DC疫苗相比,这种双重治疗显着增加了TIL的浸润。这些发现表明TIMs以不仅限于PD-1 / PD-L1信号传导机制的方式在肿瘤微环境中发挥抑制作用。我们发现,将CSF-1Ri和PD-1 mAb与DC疫苗联合治疗可增加TIL浸润和在肿瘤微环境中的活化。这些发现与治疗有关,接受三种治疗的荷瘤小鼠的存活率明显高于仅接受每种治疗的小鼠。本文概述的研究阐明了TIMs在主要介导PD-1 / PD-L1信号传导机制以限制TIL活化中发挥的作用,以及通过临床上可利用的药物在药理上操纵这一人群的能力。产生和增强针对GBM的免疫反应的两种不同策略。在我们的第一个策略中,我们利用工程化T细胞的过继转移来选择性靶向在GBM中人工诱导表达的抗原。该技术非常有效。但是,我们对直接靶向GBM已表达的抗原感兴趣。为此,我们描述了DC疫苗接种在产生免疫反应中的效用。此外,我们描述了TIMs在肿瘤微环境中所采用的抑制机制,并开发了治疗佐剂与DC疫苗一起给药。 (摘要由UMI缩短。)。

著录项

  • 作者

    Antonios, Joseph Paul.;

  • 作者单位

    University of California, Los Angeles.;

  • 授予单位 University of California, Los Angeles.;
  • 学科 Neurosciences.;Immunology.;Oncology.
  • 学位 Ph.D.
  • 年度 2016
  • 页码 142 p.
  • 总页数 142
  • 原文格式 PDF
  • 正文语种 eng
  • 中图分类
  • 关键词

  • 入库时间 2022-08-17 11:48:16

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