Oncotarget

Research Papers:

An NCR1-based chimeric receptor endows T-cells with multiple anti-tumor specificities

Yair Tal, Shlomo Yaakobi, Miryam Horovitz-Fried, Einav Safyon, Benyamin Rosental, Angel Porgador and Cyrille J. Cohen _

PDF  |  HTML  |  Supplementary Files  |  How to cite

Oncotarget. 2014; 5:10949-10958. https://doi.org/10.18632/oncotarget.1919

Metrics: PDF 2677 views  |   HTML 3292 views  |   ?  


Abstract

Yair Tal1,*, Shlomo Yaakobi1,*, Miryam Horovitz-Fried1, Einav Safyon1, Benyamin Rosental2, Angel Porgador2, Cyrille J. Cohen1

1 Laboratory of Tumor Immunology and Immunotherapy, The Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan 52900, Israel

2 The Shraga Segal Department of Microbiology, Immunology and Genetics, and the National Institute for Biotechnology in the Negev, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel

* These authors contributed equally to this study

Correspondence:

Cyrille J. Cohen, email:

Keywords: NCR1, Tumor Immunotherapy, T-cells, T-cell engineering

Received: February 20, 2014 Accepted: April 24, 2014 Published: April 24, 2014

Abstract

The Ral (Ras-like) GTP-binding proteins (RalA and RalB), as effectors of the proto-oncogene Natural killer (NK) cells are an important component of the anti-tumor response. Tumor recognition by NK cells was found to be partly triggered by molecules termed natural cytotoxic receptors (NCRs). Adoptive transfer of genetically-engineered tumor-reactive T-lymphocytes can mediate remarkable tumor regressions mostly in melanoma and leukemia patients. Yet, the application of such treatments to other cancers is needed and dependent on the isolation of receptors that could facilitate efficient recognition of these malignancies. Herein, we aimed at combining NK tumor recognition capability with the genetic modification of T-cells to provide the latter with a means to recognize several tumors in a non-MHC restricted way.

Consequently, we generated and evaluated several chimeric receptors based on the extracellular domain of NCR1 (NKp46) fused to multiple signaling moieties and assess their antitumor activity when retrovirally expressed in T-cells. Following co-culture with different tumors, primary human T-lymphocytes expressing a chimeric NCR1 molecule recognized target cells derived from lung, cervical carcinoma, leukemia and pancreatic cancer. In addition, this receptor mediated an upregulation of surface activation markers and significant antitumor cytotoxicity both in vitro and in vivo. These results have meaningful implications for the immunotherapeutic treatment of cancer using gene-modified T-cells.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 1919