The Inducible Costimulator (ICOS) Is Critical for the Development of Human TH17 Cells

CM Paulos, C Carpenito, G Plesa… - Science translational …, 2010 - science.org
CM Paulos, C Carpenito, G Plesa, MM Suhoski, A Varela-Rohena, TN Golovina, RG Carroll…
Science translational medicine, 2010science.org
Human T helper 17 (TH17) cells regulate host defense, autoimmunity, and tumor immunity.
Although cytokines that control human TH17 cell development have been identified, the
costimulatory molecules important for TH17 cell generation are unknown. Here, we found
that the inducible costimulator (ICOS) was critical for the differentiation and expansion of
human TH17 cells. Human cord blood contained a subset of CD161+ CD4+ T cells that were
recent emigrants from the thymus, expressed ICOS constitutively, and were imprinted as …
Human T helper 17 (TH17) cells regulate host defense, autoimmunity, and tumor immunity. Although cytokines that control human TH17 cell development have been identified, the costimulatory molecules important for TH17 cell generation are unknown. Here, we found that the inducible costimulator (ICOS) was critical for the differentiation and expansion of human TH17 cells. Human cord blood contained a subset of CD161+CD4+ T cells that were recent emigrants from the thymus, expressed ICOS constitutively, and were imprinted as TH17 cells through ICOS signaling. ICOS stimulation induced c-MAF, RORC2, and T-bet expression in these cells, leading to increased secretion of interleukin-21 (IL-21), IL-17, and interferon-γ (IFN-γ) compared with cells stimulated with CD28. Conversely, CD28 ligation abrogated ICOS costimulation, dampening RORC2 expression while promoting the expression of the aryl hydrocarbon receptor, which led to reduced secretion of IL-17 and enhanced production of IL-22 compared with cells stimulated with ICOS. Moreover, ICOS promoted the robust expansion of IL-17+IFN-γ+ human T cells, and the antitumor activity of these cells after adoptive transfer into mice bearing large human tumors was superior to that of cells expanded with CD28. The therapeutic effectiveness of ICOS-expanded cells was associated with enhanced functionality and engraftment in vivo. These findings reveal a vital role for ICOS signaling in the generation and maintenance of human TH17 cells and suggest that components of this pathway could be therapeutically targeted to treat cancer or chronic infection and, conversely, that interruption of this pathway may have utility in multiple sclerosis and other autoimmune syndromes. These findings have provided the rationale for designing new clinical trials for tumor immunotherapy.
AAAS