Homeostatic proliferation plus regulatory T-cell depletion promotes potent rejection of B16 melanoma

J Kline, IE Brown, YY Zha, C Blank, J Strickler… - Clinical Cancer …, 2008 - AACR
J Kline, IE Brown, YY Zha, C Blank, J Strickler, H Wouters, L Zhang, TF Gajewski
Clinical Cancer Research, 2008AACR
Purpose: To investigate the antitumor efficacy of T-cell anergy reversal through homeostatic
proliferation and regulatory T-cell (Treg) depletion in a clinically relevant murine adoptive
immunotherapy model. Experimental Design: B16 melanoma cells were engineered to
express the model SIYRYYGL (SIY) antigen to enable immune monitoring. Tumor-specific T
cells expanded in tumor-challenged wild-type hosts but became hyporesponsive. To
examine whether lymphopenia-induced homeostatic proliferation could reverse tumor …
Abstract
Purpose: To investigate the antitumor efficacy of T-cell anergy reversal through homeostatic proliferation and regulatory T-cell (Treg) depletion in a clinically relevant murine adoptive immunotherapy model.
Experimental Design: B16 melanoma cells were engineered to express the model SIYRYYGL (SIY) antigen to enable immune monitoring. Tumor-specific T cells expanded in tumor-challenged wild-type hosts but became hyporesponsive. To examine whether lymphopenia-induced homeostatic proliferation could reverse tumor-induced T-cell anergy, total splenic T cells were transferred into lymphopenic RAG2−/− mice or control P14/RAG2−/− mice. Tumor growth was measured, and SIY-specific immune responses were monitored using ELISPOT and SIY/Kb tetramers. To determine whether Treg depletion could synergize with homeostatic proliferation, RAG2−/− mice received total or CD25-depleted T cells, followed or preceded by B16.SIY challenge. This approach was further investigated in wild-type mice lymphodepleted with sublethal total body irradiation.
Results: Adoptive transfer of total splenic T cells into RAG2−/− mice moderately affected the growth rate of B16.SIY. As Treg expansion occurred in tumor-bearing mice, CD25+ T cells were depleted from total T cells before adoptive transfer. Interestingly, transfer of CD25-depleted T cells into RAG2−/− mice resulted in potent rejection of B16 melanoma in both prophylactic and short-term preimplanted tumor settings and was associated with maintained T-cell effector function. Using a clinically applicable approach, wild-type mice were lymphodepleted using sublethal total body irradiation, which similarly supported tumor rejection upon transfer of CD25-depleted T cells.
Conclusions: Our results indicate that combined CD25 depletion and homeostatic proliferation support a potent antitumor immune response—an approach with potential for clinical translation.
AACR