Regression of renal cell carcinoma by T cell receptor-engineered T cells targeting a human endogenous retrovirus

AI Summary

The content describes the discovery of a human endogenous retrovirus (CT-RCC HERV-E) selectively expressed in clear cell renal cell carcinomas (ccRCC) and its use as an antigen for T cell-mediated killing. A novel T cell receptor (TCR) targeting a CT-RCC HERV-E-derived antigen specific to ccRCC was cloned and characterized for its antitumor activity. T cells modified with HERV-E TCR showed potent antitumor activity against ccRCC cells expressing CT-RCC HERV-E in vitro and in a murine xenograft model. Large-scale production of HERV-E T cells under good manufacturing conditions from healthy donors retained their antigen recognition and cytotoxicity. This study paves the way for evaluating HERV-E TCR-transduced T cell infusions in patients with metastatic ccRCC.

Background

We discovered a novel human endogenous retrovirus (CT-RCC HERV-E) that was selectively expressed in most clear cell renal cell carcinomas (ccRCC) and served as a source of antigens for T cell-mediated killing. Here, we described the cloning of a novel T cell receptor (TCR) targeting a CT-RCC HERV-E-derived antigen specific to ccRCC and characterized antitumor activity of HERV-E TCR-transduced T cells (HERV-E T cells).

Methods

We isolated a CD8+ T cell clone from a patient with immune-mediated regression of ccRCC post-allogeneic stem cell transplant that recognized the CT-RCC-1 HERV-E-derived peptide in an HLA-A11-restricted manner. We used 5’Rapid Amplification of cDNA Ends (RACE) to clone the full length HERV-E TCR and generated retrovirus encoding this TCR for transduction of T cells. We characterized HERV-E T cells for phenotype and function in vitro and in a murine xenograft model. Lastly, we implemented a good manufacturing practice-compliant method for scalable production of HERV-E T cells.

Results

The HLA-A11-restricted HERV-E-reactive TCR exhibited a CD8-dependent phenotype and demonstrated specific recognition of the CT-RCC-1 peptide. CD8+ T cells modified to express HERV-E TCR displayed potent antitumor activity against HLA-A11+ ccRCC cells expressing CT-RCC HERV-E compared with unmodified T cells. Killing by HERV-E T cells was lost when cocultured against HERV-E knockout ccRCC cells. HERV-E T cells induced regression of established ccRCC tumors in a murine model and improved survival of tumor-bearing mice. Large-scale production of HERV-E T cells under good manufacturing practice conditions generated from healthy donors retained specific antigen recognition and cytotoxicity against ccRCC.

Conclusions

This is the first report showing that human ccRCC cells can be selectively recognized and killed by TCR-engineered T cells targeting a HERV-derived antigen. These preclinical findings provided the foundation for evaluating HERV-E TCR-transduced T cell infusions in patients with metastatic ccRCC in a clinical trial (NCT03354390).

Leave a Reply