4.5 Article

Preclinical evaluation of cancer immune therapy using patient-derived tumor antigen-specific T cells in a novel xenograft platform

期刊

出版社

WILEY
DOI: 10.1002/cti2.1246

关键词

cancer immunotherapy; checkpoint blockade; melanoma; patient-derived xenograft; T cells

资金

  1. National Institutes of Health [R01 CA131407, R43 CA224602, R21 CA205794, R01 CA204261, CA034196, R24 OD026440, AI132963, HL-70227]
  2. Center for Protein Therapeutics (CPT) grant

向作者/读者索取更多资源

The study established a novel in vivo model (Xenomimetic or 'X' mouse) to monitor the efficacy of human tumor-specific T cells in suppressing tumor growth and evaluating the therapeutic efficacy of immune-based therapies. Results showed that the model successfully predicted the effectiveness of immune-based therapies in inhibiting tumor progression and could potentially be used for preclinical evaluation of new immunotherapies.
Objectives. With a rapidly growing list of candidate immune-based cancer therapeutics, there is a critical need to generate highly reliable animal models to preclinically evaluate the efficacy of emerging immune-based therapies, facilitating successful clinical translation. Our aim was to design and validate a novel in vivo model (called Xenomimetic or 'X' mouse) that allows monitoring of the ability of human tumor-specific T cells to suppress tumor growth following their entry into the tumor. Methods. Tumor xenografts are established rapidly in the greater omentum of globally immunodeficient NOD-scid 11.2R gamma(null) (NSG) mice following an intraperitoneal injection of melanoma target cells expressing tumor neoantigen peptides, as well as green fluorescent protein and/or luciferase. Changes in tumor burden, as well as in the number and phenotype of adoptively transferred patient-derived tumor neoantigen-specific T cells in response to immunotherapy, are measured by imaging to detect fluorescence/luminescence and flow cytometry, respectively. Results. The tumors progress rapidly and disseminate in the mice unless patient-derived tumor-specific T cells are introduced. An initial T cell-mediated tumor arrest is later followed by a tumor escape, which correlates with the upregulation of the checkpoint molecules programmed cell death-1 (PD-1) and lymphocyte-activation gene 3 (LAG3) on T cells. Treatment with immune-based therapies that target these checkpoints, such as anti-PD-1 antibody (nivolumab) or interleukin-12 (IL-12), prevented or delayed the tumor escape. Furthermore, IL-12 treatment suppressed PD-1 and LAG3 upregulation on T cells. Conclusion. Together, these results validate the X-mouse model and establish its potential to preclinically evaluate the therapeutic efficacy of immune-based therapies.

作者

我是这篇论文的作者
点击您的名字以认领此论文并将其添加到您的个人资料中。

评论

主要评分

4.5
评分不足

次要评分

新颖性
-
重要性
-
科学严谨性
-
评价这篇论文

推荐

暂无数据
暂无数据