Impaired STAT phosphorylation in T cells from melanoma patients in response to IL-2: association with clinical stage

R Mortarini, C Vegetti, A Molla, F Arienti… - Clinical cancer …, 2009 - AACR
R Mortarini, C Vegetti, A Molla, F Arienti, F Ravagnani, A Maurichi, R Patuzzo, M Santinami…
Clinical cancer research, 2009AACR
Purpose: To assess the extent of signal transducer and activator of transcription (STAT)
activation in response to interleukin 2 (IL-2) in melanoma patients' T cells, along with clinical
stage of tumor progression. Experimental Design: T lymphocytes from peripheral blood of
healthy donors and of American Joint Committee on Cancer stage I to IV melanoma patients,
as well as from metastatic lymph nodes of patients, were evaluated for responsiveness to IL-
2. CFSE assays and single-cell phospho-STAT–specific flow cytometry screening were …
Abstract
Purpose: To assess the extent of signal transducer and activator of transcription (STAT) activation in response to interleukin 2 (IL-2) in melanoma patients' T cells, along with clinical stage of tumor progression.
Experimental Design: T lymphocytes from peripheral blood of healthy donors and of American Joint Committee on Cancer stage I to IV melanoma patients, as well as from metastatic lymph nodes of patients, were evaluated for responsiveness to IL-2. CFSE assays and single-cell phospho-STAT–specific flow cytometry screening were used.
Results. T cells from advanced melanoma patients, in comparison with healthy donors, showed reduced proliferation to IL-2 and IL-15, but not to anti-CD3 monoclonal antibody. Impaired response occurred in CCR7+ and CCR7 T-cell subsets, but not in CD3 CD8+ natural killer (NK) cells, and was not explained by induction of apoptosis, increased cytokine consumption, or altered IL-2R subunit expression in patients' T lymphocytes. By phospho-specific flow cytometry, defective STAT1 and STAT5 activation in response to IL-2 was found mainly in T lymphocytes from peripheral blood and/or tumor site of American Joint Committee on Cancer stage III and IV patients, compared with stage I and II patients and to donors, and in melanoma antigen-specific T cells isolated from metastatic lymph nodes. At tumor site, impaired STAT activation in T cells did not correlate with frequency of CD4+ CD25+ Foxp3+ T cells. Serum from advanced melanoma patients inhibited IL-2–dependent STAT activation in donors' T cells and a neutralizing monoclonal antibody to transforming growth factor β1 counteracted such inhibition.
Conclusions: These results provide evidence for development of impaired STAT signaling in response to IL-2, along with clinical evolution of the disease, in melanoma patients' T cells.
AACR