B7-H1 signaling is integrated during CD8 super(+) T cell priming and restrains effector differentiation

A promising strategy in tumor immunotherapy is the use of activated dendritic cells as vehicles for tumor vaccines with the goal of activating anti-tumor T cell responses. Current formulations for dendritic cell-based immunotherapies have limited effects on patient survival, providing motivation for...

Full description

Saved in:
Bibliographic Details
Published inCancer Immunology, Immunotherapy Vol. 63; no. 8; pp. 859 - 867
Main Authors Gibbons, Rachel M, Liu, Xin, Harrington, Susan M, Krco, Christopher J, Kwon, Eugene D, Dong, Haidong
Format Journal Article
LanguageEnglish
Published 01.08.2014
Online AccessGet full text

Cover

Loading…
More Information
Summary:A promising strategy in tumor immunotherapy is the use of activated dendritic cells as vehicles for tumor vaccines with the goal of activating anti-tumor T cell responses. Current formulations for dendritic cell-based immunotherapies have limited effects on patient survival, providing motivation for further investigation of ways to enhance dendritic cell priming of anti-tumor T cell responses. Using a brief in vitro priming model, we have found that B7-H1 expressed by activated dendritic cells is integrated during priming of naive CD8 super(+) T cells and functions to limit the differentiation of effector T cell responses. CD8 super(+) T cells primed by B7-H1-deficient dendritic cells exhibit increased production of IFN- gamma , enhanced target cell killing, and improved anti-tumor activity. Additionally, enhanced memory populations arise from CD8 super(+) T cells primed by B7-H1-deficient dendritic cells. Based on these findings, we suggest that early blockade of B7-H1 signaling should be investigated as a strategy to improve dendritic cell-based anti-tumor immunotherapy.
Bibliography:ObjectType-Article-1
SourceType-Scholarly Journals-1
content type line 23
ObjectType-Feature-2
ISSN:0340-7004
1432-0851
DOI:10.1007/s00262-014-1563-6