Oncotarget

Priority Research Papers:

Enhanced immunotherapy by combining a vaccine with a novel murine GITR ligand fusion protein

Y. Maurice Morillon II, Scott A. Hammond, Nicholas M. Durham, Jeffrey Schlom and John W. Greiner _

PDF  |  HTML  |  Supplementary Files  |  How to cite

Oncotarget. 2017; 8:73469-73482. https://doi.org/10.18632/oncotarget.20703

Metrics: PDF 1934 views  |   HTML 2517 views  |   ?  


Abstract

Y. Maurice Morillon II1, Scott A. Hammond2, Nicholas M. Durham2, Jeffrey Schlom1,* and John W. Greiner1,*

1 Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA

2 MedImmune LLC, Gaithersburg, MD, USA

* These authors have contributed equally to this work

Correspondence to:

Jeffrey Schlom, email:

Keywords: poxvirus vaccines, rMVA/rF-CEA-TRICOM, mGITRL-FP, CD4+FoxP3+ regulatory T cells, immunotherapy

Received: July 04, 2017 Accepted: August 03, 2017 Published: September 07, 2017

Abstract

Immunotherapy was significantly enhanced in a murine tumor model by combining a vaccine with a fusion protein designed to target the glucocorticoid-induced tumor necrosis factor (TNF) receptor related gene (GITR) on the surface of T cells. The recombinant poxvirus-based vaccine platform included Modified Vaccinia virus Ankara (rMVA) and fowlpox (rF) vectors as the driver immunogens both engineered to express the human carcinoembryonic antigen (CEA) and three murine costimulatory molecules B7.1, ICAM-1, LFA-3 (designated TRICOM). In previous studies, mice expressing human CEA as a transgene (CEA.Tg mice) vaccinated with rMVA/rF-CEA-TRICOM overcame CEA immune tolerance by inducing anti-CEA‒specific immunity and regression of CEA-expressing tumors. The murine GITR ligand fusion protein (mGITRL-FP) consisted of a mouse IgG2a Fc region, a yeast-derived coiled GCN4 pII and the extracellular GITR-binding domain of murine GITR ligand. The design maximized valency and the potential to agonize the GITR receptor. Combined treatment of the vaccine and mGITRL-FP mediated a more robust tumor regression, leading to sustained improvement in overall survival. The enhanced immunotherapeutic effect was linked to the generation of a strong CD8+ T cell antitumor immune response. A treatment schedule with mGITRL-FP administered prior to the priming rMVA-CEA-TRICOM vaccination was of paramount importance. The mechanism of action for the enhanced antitumor effects resided in the depletion of immune cells, particularly FoxP3+ regulatory T cells, that express high GITR levels following activation. The results provide evidence that targeting GITR with mGITRL-FP in concert with a cancer vaccine represents a potential novel approach to more effective immunotherapy.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 20703