Your browser doesn't support javascript.
loading
Intra-tumoral delivery of CXCL11 via a vaccinia virus, but not by modified T cells, enhances the efficacy of adoptive T cell therapy and vaccines.
Moon, Edmund K; Wang, Liang-Chuan S; Bekdache, Kheng; Lynn, Rachel C; Lo, Albert; Thorne, Stephen H; Albelda, Steven M.
Afiliação
  • Moon EK; Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
  • Wang LS; Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
  • Bekdache K; current address: Preclinical Pharmacology, Incyte Corporation, Wilmington, DE, USA.
  • Lynn RC; Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
  • Lo A; Ovarian Cancer Research Center, Department of Obstetrics and Gynecology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
  • Thorne SH; Department of Biomedical Sciences, School of Veterinary Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
  • Albelda SM; University of Pittsburgh Cancer Institute, and Departments of Surgery and Immunology, University of Pittsburgh, Pittsburgh, PA, USA.
Oncoimmunology ; 7(3): e1395997, 2018.
Article em En | MEDLINE | ID: mdl-29399394
ABSTRACT
T cell trafficking into tumors depends on a "match" between chemokine receptors on effector cells (e.g., CXCR3 and CCR5) and tumor-secreted chemokines. There is often a chemokine/chemokine receptor "mismatch", with tumors producing minute amounts of chemokines, resulting in inefficient targeting of effectors to tumors. We aimed to alter tumors to produce higher levels of CXCL11, a CXCR3 ligand, to attract more effector cells following immunotherapy. Mice bearing established subcutaneous tumors were studied. In our first approach, we used modified chimeric antigen receptor (CAR)-transduced human T cells to deliver CXCL11 (CAR/CXCL11) into tumors. In our second approach, we intravenously (iv) administered a modified oncolytic vaccinia virus (VV) engineered to produce CXCL11 (VV.CXCL11). The effect of these treatments on T cell trafficking into the tumors and anti-tumor efficacy after subsequent CAR T cell injections or anti-tumor vaccines was determined. CAR/CXCL11 and VV.CXCL11 significantly increased CXCL11 protein levels within tumors. For CAR/CXCL11, injection of a subsequent dose of CAR T cells did not result in increased intra-tumoral trafficking, and appeared to decrease the function of the injected CAR T cells. In contrast, VV.CXCL11 increased the number of total and antigen-specific T cells within tumors after CAR T cell injection or vaccination and significantly enhanced anti-tumor efficacy. Both approaches were successful in increasing CXCL11 levels within the tumors; however, only the vaccinia approach was successful in recruiting T cells and augmenting anti-tumor efficacy. VV.CXCL11 should be considered as a potential approach to augment adoptive T cell transfer or vaccine immunotherapy.
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Idioma: En Ano de publicação: 2018 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Idioma: En Ano de publicação: 2018 Tipo de documento: Article