Abstract

Simple SummaryThe development of effective adoptive T-cell therapies (ATCs) to treat solid tumors has several challenges: the choice of a suitable target antigen, the generation of a specific T-cell receptor (TCR) directed against this target, and the hostile tumor microenvironment (TME). The cancer/testis antigen Preferentially Expressed Antigen in Melanoma (PRAME) is a promising target for ATCs since it is highly expressed in several solid tumor indications, while its expression in normal tissues is mainly restricted to the testis. Using our well-established high throughput TCR generation and characterization process, we identified a highly potent PRAME-specific TCR. To convert the inhibitory PD-1 signal in T-cells to an activating signal, we designed a chimeric receptor consisting of the extracellular domain of PD-1 and the signaling domain of 4-1BB. Combining this PD1-41BB receptor with our lead PRAME-TCR generated a very promising T-cell product with a favorable preclinical in vitro safety profile and enhanced in vitro and in vivo anti-tumor efficacy.The hostile tumor microenvironment (TME) is a major challenge for the treatment of solid tumors with T-cell receptor (TCR)-modified T-cells (TCR-Ts), as it negatively influences T-cell efficacy, fitness, and persistence. These negative influences are caused, among others, by the inhibitory checkpoint PD-1/PD-L1 axis. The Preferentially Expressed Antigen in Melanoma (PRAME) is a highly relevant cancer/testis antigen for TCR-T immunotherapy due to broad expression in multiple solid cancer indications. A TCR with high specificity and sensitivity for PRAME was isolated from non-tolerized T-cell repertoires and introduced into T-cells alongside a chimeric PD1-41BB receptor, consisting of the natural extracellular domain of PD-1 and the intracellular signaling domain of 4-1BB, turning an inhibitory pathway into a T-cell co-stimulatory pathway. The addition of PD1-41BB to CD8+ T-cells expressing the transgenic PRAME-TCR enhanced IFN-γ secretion, improved cytotoxic capacity, and prevented exhaustion upon repetitive re-challenge with tumor cells in vitro without altering the in vitro safety profile. Furthermore, a single dose of TCR-Ts co-expressing PD1-41BB was sufficient to clear a hard-to-treat melanoma xenograft in a mouse model, whereas TCR-Ts without PD1-41BB could not eradicate the PD-L1-positive tumors. This cutting-edge strategy supports development efforts to provide more effective TCR-T immunotherapies for the treatment of solid tumors.

Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call