Your browser doesn't support javascript.
loading
p50 suppresses cytotoxic T lymphocyte effector function to regulate tumor immune escape and response to immunotherapy.
Lu, Chunwan; Klement, John D; Smith, Alyssa D; Yang, Dafeng; Waller, Jennifer L; Browning, Darren D; Munn, David H; Liu, Kebin.
Afiliación
  • Lu C; Department of Biochemistry and Molecular Biology, Augusta University, Augusta, Georgia, United States Clu@augusta.edu kliu@augusta.edu.
  • Klement JD; Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, Georgia, United States.
  • Smith AD; Charlie Norwood VA Medical Center, Augusta, Georgia, United States.
  • Yang D; Department of Biochemistry and Molecular Biology, Augusta University, Augusta, Georgia, United States.
  • Waller JL; Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, Georgia, United States.
  • Browning DD; Charlie Norwood VA Medical Center, Augusta, Georgia, United States.
  • Munn DH; Department of Biochemistry and Molecular Biology, Augusta University, Augusta, Georgia, United States.
  • Liu K; Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, Georgia, United States.
J Immunother Cancer ; 8(2)2020 10.
Article en En | MEDLINE | ID: mdl-33051343
ABSTRACT

BACKGROUND:

NF-κB is a key link between inflammation and cancer. Previous studies of NF-κB have largely focused on tumor cells, and the intrinsic function of NF-κB in T cells in tumor development and response to immunotherapy is largely unknown. We aimed at testing the hypothesis that NF-κB1 (p50) activation in T cells underlies human colon cancer immune escape and human cancer non-response to anti-PD-1 immunotherapy.

METHODS:

We screened NF-κB activation in human colon carcinoma and used mouse models to determine p50 function in tumor cells and immune cells. RNA-Seq was used to identify p50 target genes. p50 binding to target gene promoters were determined by electrophoresis mobility shift assay and chromatin immunoprecipitation. A p50 activation score was generated from gene expression profiling and used to link p50 activation to T-cell activation and function pre-nivolumab and post-nivolumab immunotherapy in human patients with cancer.

RESULTS:

p50 is the dominant form of NF-κB that is highly activated in immune cells in the human colorectal carcinoma microenvironment and neighboring non-neoplastic colon epithelial cells. Tumor cell intrinsic p50 signaling and T-cell intrinsic p50 signaling exert opposing functions in tumor growth control in vivo. Deleting Nfkb1 in tumor cells increased whereas in T cells decreased tumor growth in preclinical mouse models. Gene expression profiling identified Gzmb as a p50 target in T cells. p50 binds directly to a previously uncharacterized κB sequence at the Gzmb promoter in T cells, resulting in repression of Gzmb expression in tumor-infiltrating cytotoxic T lymphocytes (CTLs) to induce a dysfunctional CTL phenotype to promote tumor immune escape. p50 activation is inversely correlated with both GZMB expression and T-cell tumor infiltration in human colorectal carcinoma. Furthermore, nivolumab immunotherapy decreased p50 activation and increased GZMB expression in human patients with melanoma.

CONCLUSIONS:

Inflammation activates p50 that binds to the Gzmb promoter to repress granzyme B expression in T cells, resulting in CTL dysfunction to confer tumor immune escape and decreased response to anti-PD-1 immunotherapy.
Asunto(s)
Palabras clave

Texto completo: 1 Bases de datos: MEDLINE Asunto principal: Linfocitos T Citotóxicos / Inmunoterapia Tipo de estudio: Prognostic_studies Límite: Animals / Female / Humans Idioma: En Revista: J Immunother Cancer Año: 2020 Tipo del documento: Article

Texto completo: 1 Bases de datos: MEDLINE Asunto principal: Linfocitos T Citotóxicos / Inmunoterapia Tipo de estudio: Prognostic_studies Límite: Animals / Female / Humans Idioma: En Revista: J Immunother Cancer Año: 2020 Tipo del documento: Article