Your browser doesn't support javascript.
loading
Intratumoral STING Activation with T-cell Checkpoint Modulation Generates Systemic Antitumor Immunity.
Ager, Casey R; Reilley, Matthew J; Nicholas, Courtney; Bartkowiak, Todd; Jaiswal, Ashvin R; Curran, Michael A.
Afiliação
  • Ager CR; Immunology Program, University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas.
  • Reilley MJ; Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, Texas.
  • Nicholas C; Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas.
  • Bartkowiak T; Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, Texas.
  • Jaiswal AR; Immunology Program, University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas.
  • Curran MA; Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, Texas.
Cancer Immunol Res ; 5(8): 676-684, 2017 08.
Article em En | MEDLINE | ID: mdl-28674082
ABSTRACT
Coordinated manipulation of independent immune regulatory pathways in the tumor microenvironment-including blockade of T-cell checkpoint receptors and reversal of suppressive myeloid programs-can render aggressive cancers susceptible to immune rejection. Elevated toxicity associated with combination immunotherapy, however, prevents translation of the most efficacious regimens. We evaluated T-cell checkpoint-modulating antibodies targeting CTLA-4, PD-1, and 4-1BB together with myeloid agonists targeting either STING or Flt3 in the TRAMP-C2 model of prostate cancer to determine whether low-dose intratumoral delivery of these agents could elicit systemic control of multifocal disease. Intratumoral administration of the STING agonist cyclic di-GMP (CDG) or Flt3 Ligand (Flt3L) augmented the therapeutic effect of systemic triple checkpoint modulation and promoted the cure of 75% of mice with bilateral TRAMP-C2; however, when all agents were administered locally, only CDG mobilized abscopal immunity. Combination efficacy correlated with globally enhanced ratios of CD8+ T cells to regulatory T cells (Treg), macrophages, and myeloid-derived suppressor cells, and downregulation of the M2 marker CD206 on tumor-associated macrophages. Flt3L improved CD8+ T-cell and dendritic cell infiltration of tumors, but was diminished in efficacy by concomitant Treg expansion. Although intratumoral CDG/checkpoint therapy invokes substantial ulceration at the injection site, reduced CDG dosing can preserve tissue integrity without sacrificing therapeutic benefit. For high-order combinations of T-cell checkpoint antibodies and local myeloid agonists, systemic antibody administration provides the greatest efficacy; however, local administration of CDG and antibody provides substantial systemic benefit while minimizing the potential for immune-related adverse events. Cancer Immunol Res; 5(8); 676-84. ©2017 AACR.
Assuntos

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Linfócitos do Interstício Tumoral / Proteínas de Membrana / Neoplasias Tipo de estudo: Prognostic_studies Limite: Animals / Humans Idioma: En Revista: Cancer Immunol Res Ano de publicação: 2017 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Linfócitos do Interstício Tumoral / Proteínas de Membrana / Neoplasias Tipo de estudo: Prognostic_studies Limite: Animals / Humans Idioma: En Revista: Cancer Immunol Res Ano de publicação: 2017 Tipo de documento: Article