[HTML][HTML] Optimal Population of FoxP3+ T Cells in Tumors Requires an Antigen Priming-Dependent Trafficking Receptor Switch

C Wang, JH Lee, CH Kim - PLoS One, 2012 - journals.plos.org
PLoS One, 2012journals.plos.org
FoxP3+ T cells populate tumors and regulate anti-tumor immunity. The requirement for
optimal population of FoxP3+ regulatory T cells in tumors remains unclear. We investigated
the migration requirement and stability of tumor-associated FoxP3+ T cells. We found that
only memory, but not naïve, FoxP3+ T cells are highly enriched in tumors. Almost all of the
tumor-infiltrating FoxP3+ T cells express Helios, an antigen associated either with thymus-
generated FoxP3+ T cells or activated T cells in the periphery. The tumor-infiltrating FoxP3+ …
FoxP3+ T cells populate tumors and regulate anti-tumor immunity. The requirement for optimal population of FoxP3+ regulatory T cells in tumors remains unclear. We investigated the migration requirement and stability of tumor-associated FoxP3+ T cells. We found that only memory, but not naïve, FoxP3+ T cells are highly enriched in tumors. Almost all of the tumor-infiltrating FoxP3+ T cells express Helios, an antigen associated either with thymus-generated FoxP3+ T cells or activated T cells in the periphery. The tumor-infiltrating FoxP3+ T cells largely lack CD62L and CCR7, two trafficking receptors required for T cell migration into secondary lymphoid tissues. Instead, the tumor infiltrating FoxP3+ T cells highly express memory/tumor-associated CCR8 and CXCR4. Antigen priming is required for induction of this trafficking receptor phenotype in FoxP3+ T cells and only antigen primed, but not antigen-inexperienced naive, FoxP3+ T cells can efficiently migrate into tumors. While the migration of FoxP3+ T cells into tumors was a readily detectable event, generation of induced FoxP3+ T cells within tumors was unexpectedly inefficient. Genetic marking of current and ex-FoxP3+ T cells revealed that tumor-infiltrating FoxP3+ T cells are highly stable and do not readily convert back to FoxP3 T cells. Taken together, our results indicate that population of tumors with thymus-generated FoxP3+ T cells requires an antigen priming-dependent trafficking receptor switch in lymphoid tissues.
PLOS