4.7 Article

CAR T cell therapy for breast cancer: harnessing the tumor milieu to drive T cell activation

Journal

JOURNAL FOR IMMUNOTHERAPY OF CANCER
Volume 6, Issue -, Pages -

Publisher

BMJ PUBLISHING GROUP
DOI: 10.1186/s40425-018-0347-5

Keywords

Chimeric antigen receptor; Genetic engineering; Inverted cytokine receptor; T cell therapy; Breast cancer

Funding

  1. NIH-NCI [P01 CA094237, P50 CA126752, P50 CA186784, P30 CA125123]
  2. Pancreatic Cancer Action Network Translational Research Grant [16-65-LEEN]
  3. American Cancer Society [MRSG-14-197-01-LIB]
  4. Cancer Prevention and Research Institute of Texas [RR170024]
  5. V Foundation for Cancer Research [T2016-006]
  6. Elsa U. Pardee Foundation
  7. National Pancreas Foundation
  8. Adrienne Helis Malvin Medical Research Foundation
  9. Baylor College of Medicine

Ask authors/readers for more resources

Background: The adoptive transfer of T cells redirected to tumor via chimeric antigen receptors (CARs) has produced clinical benefits for the treatment of hematologic diseases. To extend this approach to breast cancer, we generated CAR T cells directed against mucin1 (MUC1), an aberrantly glycosylated neoantigen that is overexpressed by malignant cells and whose expression has been correlated with poor prognosis. Furthermore, to protect our tumor-targeted cells from the elevated levels of immune-inhibitory cytokines present in the tumor milieu, we co-expressed an inverted cytokine receptor linking the IL4 receptor exodomain with the IL7 receptor endodomain (4/7ICR) in order to transform the suppressive IL4 signal into one that would enhance the anti-tumor effects of our CAR T cells at the tumor site. Methods: First (1G - CD3 zeta) and second generation (2G - 41BB.CD3 zeta) MUC1-specific CARs were constructed using the HMFG2 scFv. Following retroviral transduction transgenic expression of the CAR +/- ICR was assessed by flow cytometry. In vitro CAR/ICR T cell function was measured by assessing cell proliferation and short-and long-term cytotoxic activity using MUC1+ MDA MB 468 cells as targets. In vivo anti-tumor activity was assessed using IL4-producing MDA MB 468 tumor-bearing mice using calipers to assess tumor volume and bioluminescence imaging to track T cells. Results: In the IL4-rich tumor milieu, 1G CAR. MUC1 T cells failed to expand or kill MUC1+ tumors and while co-expression of the 4/7ICR promoted T cell expansion, in the absence of co-stimulatory signals the outgrowing cells exhibited an exhausted phenotype characterized by PD-1 and TIM3 upregulation and failed to control tumor growth. However, by co-expressing 2G CAR. MUC1 (signal 1 - activation + signal 2 - co-stimulation) and 4/7ICR (signal 3 - cytokine), transgenic T cells selectively expanded at the tumor site and produced potent and durable tumor control in vitro and in vivo. Conclusions: Our findings demonstrate the feasibility of targeting breast cancer using transgenic T cells equipped to thrive in the suppressive tumor milieu and highlight the importance of providing transgenic T cells with signals that recapitulate physiologic TCR signaling - [activation (signal 1), co-stimulation (signal 2) and cytokine support (signal 3)] - to promote in vivo persistence and memory formation.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available