Abrogation of SRC homology region 2 domain-containing phosphatase 1 in tumor-specific T cells improves efficacy of adoptive immunotherapy by enhancing the …

IM Stromnes, C Fowler, CC Casamina… - The Journal of …, 2012 - journals.aai.org
IM Stromnes, C Fowler, CC Casamina, CM Georgopolos, MS McAfee, TM Schmitt, X Tan…
The Journal of Immunology, 2012journals.aai.org
T cell expression of inhibitory proteins can be a critical component for the regulation of
immunopathology owing to self-reactivity or potentially exuberant responses to pathogens,
but it may also limit T cell responses to some malignancies, particularly if the tumor Ag being
targeted is a self-protein. We found that the abrogation of Src homology region 2 domain-
containing phosphatase-1 (SHP-1) in tumor-reactive CD8+ T cells improves the therapeutic
outcome of adoptive immunotherapy in a mouse model of disseminated leukemia, with …
Abstract
T cell expression of inhibitory proteins can be a critical component for the regulation of immunopathology owing to self-reactivity or potentially exuberant responses to pathogens, but it may also limit T cell responses to some malignancies, particularly if the tumor Ag being targeted is a self-protein. We found that the abrogation of Src homology region 2 domain-containing phosphatase-1 (SHP-1) in tumor-reactive CD8+ T cells improves the therapeutic outcome of adoptive immunotherapy in a mouse model of disseminated leukemia, with benefit observed in therapy employing transfer of CD8+ T cells alone or in the context of also providing supplemental IL-2. SHP-1−/− and SHP-1+/+ effector T cells were expanded in vitro for immunotherapy. Following transfer in vivo, the SHP-1−/− effector T cells exhibited enhanced short-term accumulation, followed by greater contraction, and they ultimately formed similar numbers of long-lived, functional memory cells. The increased therapeutic effectiveness of SHP-1−/− effector cells was also observed in recipients that expressed the tumor Ag as a self-antigen in the liver, without evidence of inducing autoimmune toxicity. SHP-1−/− effector CD8+ T cells expressed higher levels of eomesodermin, which correlated with enhanced lysis of tumor cells. Furthermore, reduction of SHP-1 expression in tumor-reactive effector T cells by retroviral transduction with vectors that express SHP-1–specific small interfering RNA, a translatable strategy, also exhibited enhanced antitumor activity in vivo. These studies suggest that abrogating SHP-1 in effector T cells may improve the efficacy of tumor elimination by T cell therapy without affecting the ability of the effector cells to persist and provide a long-term response.
journals.aai.org