Oncotarget

Research Papers:

Immunotherapy of WAP-TNP mice with early stage mammary gland tumors

Michael Bruns and Wolfgang Deppert _

PDF  |  HTML  |  Supplementary Files  |  How to cite

Oncotarget. 2017; 8:67790-67804. https://doi.org/10.18632/oncotarget.18850

Metrics: PDF 1926 views  |   HTML 2085 views  |   ?  


Abstract

Michael Bruns1 and Wolfgang Deppert1,2

1Heinrich-Pette-Institute, Leibniz-Institute for Experimental Virology, 20251 Hamburg, Germany

2Institute for Tumor Biology, University Medical Center Hamburg-Eppendorf (UKE), University of Hamburg, 20246 Hamburg, Germany

Correspondence to:

Wolfgang Deppert, email: [email protected]

Keywords: transgenic breast cancer mouse model, SV40 T-antigen, LCMV NP T-cell epitope, adoptive transfer of low/high avidity CTLs, CTL exhaustion

Received: March 29, 2017     Accepted: June 05, 2017     Published: June 29, 2017

ABSTRACT

The SV40 transgenic BALB/c mouse based WAP-T/WAP-TNP model for triple-negative breast cancer allows the analysis of parameters influencing immunotherapeutic approaches. Except for WAP-TNP tumors expressing the immune-dominant LCMV NP-epitope within SV40 T-antigen (T-AgNP) which is not expressed by T-Ag of WAP-T tumors, the tumors are extremely similar. Comparative anti-PD1/PD-L1 immunotherapy of WAP-T and WAP-TNP mice supported the hypothesis that the immunogenicity of tumor antigen T-cell epitopes strongly influences the success of immune checkpoint blockade therapy, with highly immunogenic T-cell epitopes favoring rapid CTL exhaustion. Here we analyzed the immune response in NP8 mice during early times of tumor development. LCMV infection of lactating NP8 mice induced lifelong tumor protection by memory CTLs. Immunization with LCMV after involution and appearance of T-AgNP expressing parity-induced tumor progenitor cells could not cure the mice, as memory CTLs became exhausted. However, immunization significantly prolonged the time of tumor outgrowth. Elimination of exhausted CTLs and of immunosuppressive cells by sub-lethal γ-irradiation, followed by adoptive transfer of NP-epitope specific CTLs into NP8 tumor mice with early lesions, completely prevented tumor outgrowth, when lymphocytes obtained after injection of weakly immunogenic NP8 tumor-derived cells into BALB/c mice were transferred. Transfer of lymphocytes obtained after infection of BALB/c mice with highly immunogenic LCMV into such mice delayed tumor outgrowth for a significant period, but could not prevent it. We conclude that eliminating exhausted CTLs and immune-suppressive cells followed by transfer or generation of low-avidity tumor antigen-specific CTLs might be a promising approach for curative tumor immunotherapy.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 18850