首页> 外文期刊>journal for immunotherapy of cancer >Chimeric antigen receptor T cells engineered to recognize the P329G-mutated Fc part of effector-silenced tumor antigen-targeting human IgG1 antibodies enable modular targeting of solid tumors
【24h】

Chimeric antigen receptor T cells engineered to recognize the P329G-mutated Fc part of effector-silenced tumor antigen-targeting human IgG1 antibodies enable modular targeting of solid tumors

机译:嵌合抗原受体 T 细胞经过工程改造,可识别效应沉默肿瘤抗原靶向人 IgG1 抗体的 P329G 突变的 Fc 部分,从而实现实体瘤的模块化靶向

获取原文
获取原文并翻译 | 示例
获取外文期刊封面目录资料

摘要

Background Chimeric antigen receptor (CAR) T cell therapy has proven its clinical utility in hematological malignancies. Optimization is still required for its application in solid tumors. Here, the lack of cancer-specific structures along with tumor heterogeneity represent a critical barrier to safety and efficacy. Modular CAR T cells indirectly binding the tumor antigen through CAR-adaptor molecules have the potential to reduce adverse events and to overcome antigen heterogeneity. We hypothesized that a platform utilizing unique traits of clinical grade antibodies for selective CAR targeting would come with significant advantages. Thus, we developed a P329G-directed CAR targeting the P329G mutation in the Fc part of tumor-targeting human antibodies containing P329G L234A/L235A (LALA) mutations for Fc silencing. Methods A single chain variable fragment-based second generation P329G-targeting CAR was retrovirally transduced into primary human T cells. These CAR T cells were combined with IgG1 antibodies carrying P329G LALA mutations in their Fc part targeting epidermal growth factor receptor (EGFR), mesothelin (MSLN) or HER2/neu. Mesothelioma, pancreatic and breast cancer cell lines expressing the respective antigens were used as target cell lines. Efficacy was evaluated in vitro and in vivo in xenograft mouse models. Results Unlike CD16-CAR T cells, which bind human IgG in a non-selective manner, P329G-targeting CAR T cells revealed specific effector functions only when combined with antibodies carrying P329G LALA mutations in their Fc part. P329G-targeting CAR T cells cannot be activated by an excess of human IgG. P329G-directed CAR T cells combined with a MSLN-targeting P329G-mutated antibody mediated pronounced in vitro and in vivo antitumor efficacy in mesothelioma and pancreatic cancer models. Combined with a HER2-targeting antibody, P329G-targeting CAR T cells showed substantial in vitro activation, proliferation, cytokine production and cytotoxicity against HER2-expressing breast cancer cell lines and induced complete tumor eradication in a breast cancer xenograft mouse model. The ability of the platform to target multiple antigens sequentially was shown in vitro and in vivo. Conclusions P329G-targeting CAR T cells combined with antigen-binding human IgG1 antibodies containing the P329G Fc mutation mediate pronounced in vitro and in vivo effector functions in different solid tumor models, warranting further clinical translation of this concept.
机译:背景 嵌合抗原受体 (CAR) T 细胞疗法已证明其在血液系统恶性肿瘤中的临床实用性。其在实体瘤中的应用仍需优化。在这里,缺乏癌症特异性结构以及肿瘤异质性是安全性和有效性的关键障碍。通过CAR接头分子间接结合肿瘤抗原的模块化CAR T细胞有可能减少不良事件并克服抗原异质性。我们假设,利用临床级抗体的独特特征进行选择性CAR靶向的平台将具有显着的优势。因此,我们开发了一种靶向 P329G 的靶向 CAR,靶向含有 P329G L234A/L235A (LALA) 突变的肿瘤靶向人抗体的 Fc 部分的 P329G 突变,用于 Fc 沉默。方法 将基于单链可变片段的第二代靶向P329G的CAR逆转录病毒转导至原代人T细胞中。这些 CAR T 细胞与在其 Fc 部分携带 P329G LALA 突变的 IgG1 抗体结合,靶向表皮生长因子受体 (EGFR)、间皮素 (MSLN) 或 HER2/neu。使用表达相应抗原的间皮瘤、胰腺癌和乳腺癌细胞系作为靶细胞系。在异种移植小鼠模型中评估了体外和体内的疗效。结果 与以非选择性方式结合人IgG的CD16-CAR T细胞不同,靶向P329G的CAR T细胞只有在与Fc部分携带P329G LARA突变的抗体结合时才显示出特异性的效应功能。靶向 P329G 的 CAR T 细胞不能被过量的人 IgG 激活。P329G 定向的 CAR T 细胞与靶向 MSLN 的 P329G 突变抗体联合介导,在间皮瘤和胰腺癌模型中具有明显的体外和体内抗肿瘤功效。结合 HER2 靶向抗体,靶向 P329G 的 CAR T 细胞对表达 HER2 的乳腺癌细胞系表现出显著的体外活化、增殖、细胞因子产生和细胞毒性,并在乳腺癌异种移植小鼠模型中诱导肿瘤完全根除。该平台在体外和体内依次靶向多种抗原的能力得到了证明。结论 靶向P329G的CAR T细胞与含有P329G Fc突变的抗原结合人IgG1抗体在不同实体瘤模型中具有显著的体外和体内效应功能,为进一步临床转化这一概念提供了依据。

著录项

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