4.7 Article

Adoptive cell therapy of triple negative breast cancer with redirected cytokine-induced killer cells

期刊

ONCOIMMUNOLOGY
卷 9, 期 1, 页码 -

出版社

TAYLOR & FRANCIS INC
DOI: 10.1080/2162402X.2020.1777046

关键词

Cytokine-induced killer cells (CIK); adoptive cells therapy (ACT); triple negative breast cancer (TNBC); TNBC mouse models; cetuximab

资金

  1. Fondazione AIRC [21354]
  2. University of Padova [BIRD180331/18]
  3. 5 per Mille 2018, Veneto Institute of Oncology IOV-IRCCS [BIGID219SOMM]
  4. Ministry of Health-Alliance Against Cancer (MoH-ACC) project Research project on CAR T cells for hematological malignancies and solid tumors
  5. 5 per Mille 2019 program [22759]

向作者/读者索取更多资源

Cytokine-Induced Killer (CIK) cells share several functional and phenotypical properties of both T and natural killer (NK) cells. They represent an attractive approach for cell-based immunotherapy, as they do not require antigen-specific priming for tumor cell recognition, and can be rapidly expandedin vitro. Their relevant expression of Fc gamma RIIIa (CD16a) can be exploited in combination with clinical-grade monoclonal antibodies (mAbs) to redirect their lytic activity in an antigen-specific manner. Here, we report the efficacy of this combined approach against triple negative breast cancer (TNBC), an aggressive tumor that still requires therapeutic options. Different primitive and metastatic TNBC cancer mouse models were established in NSG mice, either by implanting patient-derived TNBC samples or injecting MDA-MB-231 cells orthotopically or intravenously. The combined treatment consisted in the repeated intratumoral or intravenous injection of CIK cells and cetuximab. Tumor growth and metastasis were monitored by bioluminescence or immunohistochemistry, and survival was recorded. CIK cells plus cetuximab significantly restrained primitive tumor growth in mice, either in patient-derived tumor xenografts or MDA-MB-231 cell line models. Moreover, this approach almost completely abolished metastasis spreading and dramatically improved survival. The antigen-specific mAb favored tumor and metastasis tissue infiltration by CIK cells, and led to an enrichment of the CD16a(+)subset. Data highlight the potentiality of this novel immunotherapy strategy where a nonspecific cytotoxic cell population can be converted into tumor-specific effectors with clinical-grade antibodies, thus providing not only a therapeutic option for TNBC but also a valid alternative to more complex approaches based on chimeric antigen receptor-engineered cells.

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

4.7
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