4.5 Article

Cancer testis antigen PRAME: An anti-cancer target with immunomodulatory potential

Journal

JOURNAL OF CELLULAR AND MOLECULAR MEDICINE
Volume 25, Issue 22, Pages 10376-10388

Publisher

WILEY
DOI: 10.1111/jcmm.16967

Keywords

breast cancer; immune activation; immune checkpoints; immunotherapy; PRAME

Funding

  1. Qatar Biomedical Research Institute [VR94]
  2. Qatar National Research Fund [QRLP10-G-1803024]
  3. Qatar National Library

Ask authors/readers for more resources

PRAME in breast cancer is associated with poorer prognosis, especially in immune-unfavourable tumors. PRAME overexpression in cancer cells inhibits T cell activation and cytolytic potential, and reduces the expression of immune checkpoints and their ligands.
PReferentially expressed Antigen in Melanoma (PRAME) is a cancer testis antigen with restricted expression in somatic tissues and re-expression in poor prognostic solid tumours. PRAME has been extensively investigated as a target for immunotherapy, however, its role in modulating the anti-tumour immune response remains largely unknown. Here, we show that PRAME tumour expression is associated with worse survival in the TCGA breast cancer cohort, particularly in immune-unfavourable tumours. Using direct and indirect co-culture models, we found that PRAME overexpressing MDA-MB-468 breast cancer cells inhibit T cell activation and cytolytic potential, which could be partly restored by silencing of PRAME. Furthermore, silencing of PRAME reduced expression of several immune checkpoints and their ligands, including PD-1, LAG3, PD-L1, CD86, Gal-9 and VISTA. Interestingly, silencing of PRAME induced cancer cell killing to levels similar to anti-PD-L1 atezolizumab treatment. Comprehensive analysis of soluble inflammatory mediators and cancer cell expression of immune-related genes showed that PRAME tumour expression can suppress the expression and secretion of multiple pro-inflammatory cytokines, and mediators of T cell activation, differentiation and cytolysis. Together, our data indicate that targeting of PRAME offers a potential, novel dual therapeutic approach to specifically target tumour cells and regulate immune activation in the tumour microenvironment.

Authors

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

Reviews

Primary Rating

4.5
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available