...
首页> 外文期刊>Frontiers in Oncology >Tumor-Specific Delivery of Immune Checkpoint Inhibitors by Engineered AAV Vectors
【24h】

Tumor-Specific Delivery of Immune Checkpoint Inhibitors by Engineered AAV Vectors

机译:工程化AAV载体在肿瘤中的免疫检查点抑制剂的特异性递送

获取原文

摘要

Immune checkpoint inhibitors (ICIs) can block distinct receptors on T cells or tumor cells thus preventing T cell inactivation and tumor immune escape. While the clinical response to treatment with ICIs in cancer patients is impressive, this therapy is often associated with a number of immune-related adverse events. There is therefore a need to explore innovative strategies of tumor-specific delivery of ICIs. Delivery of therapeutic proteins on a genetic level can be accomplished with viral vectors including those derived from adeno-associated virus (AAV). Here, we assessed the tumor-targeted Her2-AAV, a receptor-targeted AAV vector binding to the tumor antigen Her2eu for cell entry, as vehicle for ICI gene delivery. Initially, we packaged the coding sequence of a scFv-Fc fusion protein directed against mouse programmed cell death protein-1 (PD-1) into Her2-AAV. Upon transduction of Her2eu ~(+) RENCA cells, AAV-encoded αPD-1 was readily detectable in the cell culture supernatant and revealed specific binding to its target antigen. In vivo , in BALB/c mice bearing subcutaneous RENCA-Her2eu tumors, Her2-AAV mediated specific gene delivery into tumor tissue upon intravenous administration as verified by luciferase gene transfer and in vivo imaging thus demonstrating unimpaired tumor-targeting by Her2-AAV vectors in immunocompetent animals. When delivering the αPD-1 gene, levels of ICI were similar in tumor tissue for Her2-AAV and AAV2 but substantially reduced in liver for Her2-AAV. When combined with chemotherapy a tendency for reduced progression of tumor growth was documented for Her2-AAV treated mice. To get closer to the clinical situation, AAV constructs that deliver the complete coding sequence of the therapeutic antibody nivolumab which is directed against human PD-1 were generated next. The AAV-Nivolumab constructs were expressed and released from transduced MDA-MB-453 cells in vitro and from RENCA-Her2eu cells upon intratumoral as well as intravenous administration in vivo . Antibody processing and expression levels were further improved through optimization of construct design. In conclusion, we provide proof-of-principle for redirecting the biodistribution of ICIs from liver and serum to tumor tissue by the use of engineered AAV vectors. This strategy can be easily combined with other types of immunotherapeutic concepts.
机译:免疫检查点抑制剂(ICI)可以阻断T细胞或肿瘤细胞上不同的受体,从而防止T细胞失活和肿瘤免疫逃逸。尽管癌症患者对ICIs治疗的临床反应令人印象深刻,但该疗法通常与许多免疫相关的不良事件有关。因此,需要探索ICI的肿瘤特异性递送的创新策略。可以使用包括从腺相关病毒(AAV)衍生的病毒载体在内的病毒载体完成遗传水平上的治疗性蛋白质的递送。在这里,我们评估了靶向肿瘤的Her2-AAV,这是一种受体靶向的AAV载体,可与进入细胞的肿瘤抗原Her2 / neu结合,作为ICI基因传递的载体。最初,我们将针对小鼠程序性细胞死亡蛋白-1(PD-1)的scFv-Fc融合蛋白的编码序列包装到Her2-AAV中。在转导Her2 / neu〜(+)RENCA细胞后,在细胞培养上清液中很容易检测到AAV编码的αPD-1,并揭示了与其靶抗原的特异性结合。在体内,在携带皮下RENCA-Her2 / neu肿瘤的BALB / c小鼠中,经荧光素酶基因转移和体内成像验证,静脉内给药后,Her2-AAV介导特异性基因传递到肿瘤组织中,从而证明了Her2-AAV对肿瘤的靶向作用不受影响免疫活性动物中的载体。当递送αPD-1基因时,Her2-AAV和AAV2的肿瘤组织中ICI水平相似,但Her2-AAV的肝脏中ICI水平显着降低。当与化学疗法组合时,已记录了Her2-AAV治疗的小鼠的肿瘤生长进程降低的趋势。为了更接近临床情况,接下来产生了递送抗人PD-1的治疗性抗体尼古鲁单抗的完整编码序列的AAV构建体。 AAV-Nivolumab构建体在肿瘤内以及体内静脉内给药后,从转导的MDA-MB-453细胞和RENCA-Her2 / neu细胞中表达和释放。通过优化构建体设计,进一步提高了抗体的加工和表达水平。总之,我们提供了使用工程化的AAV载体将ICI从肝脏和血清中的生物分布重定向到肿瘤组织的原理证明。这种策略可以很容易地与其他类型的免疫治疗概念相结合。

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