Skip to main content

Volume 3 Supplement 2

30th Annual Meeting and Associated Programs of the Society for Immunotherapy of Cancer (SITC 2015)

  • Poster presentation
  • Open access
  • Published:

Ex vivo conditioning with IL-12 decreases T cell sensitivity to intratumoral INF-γ-induced apoptosis following adoptive transfer

Background

In order to induce significant tumor regression T cells must effectively recognize and kill target cells. Secretion of IFN-γ is considered a key effector function of activated CD8+ T cells via induction of apoptosis. Thus programming T cells to secrete high levels of IFN-γ after adoptive transfer could represent a therapeutically effective anti-cancer intervention.

Methods

We previously demonstrated that naïve CD8+ T cells exposed to IL-12 during antigenic priming (PmelAg+12) provided superior anti-tumor activity after transfer when compared to cells activated in the presence of antigen alone (PmelAg). In this setting, tumor regression was associated with sustained levels of intra-tumoral IFN-γ. Expression analysis using total tumor RNA showed elevated expression of IFN-γ responsive genes such as IP-10, MCP-1, MIG, and MIP-1α. Even without IL-12 stimulation during ex vivo antigenic priming, Pmel cells were able to initially reach the tumor and secrete high levels of IFN-γ. However, by day 7 after adoptive transfer tumors in mice that received PmelAg were significantly larger than those in mice injected with PmelAg+12. Failure to maintain intra-tumoral levels of IFN-γ was associated with a decrease in the frequency of tumor infiltrating PmelAg. We hypothesized that high levels of IFN-γ had a detrimental effect on PmelAg, via induction of apoptosis. IFN-γ is a multifunctional cytokine that induces a variety of contrasting cell responses such as proliferation or cell death. The cellular response to an IFN-γ stimulus depends on the specific receptor being activated, with IFN-γR1 inducing proliferation and IFN-γR2 inducing apoptosis.

Results

We tested the hypothesis that the ability of T cells to survive in vivo after adoptive transfer was dependent on their susceptibility to IFN-γ-induced apoptosis. Real time PCR revealed that the expression levels of IFN-γR1 and IFN-γR2 immediately following antigen or antigen+ IL-12 priming were similar, though by 4d post adoptive transfer the tumor-infiltrating Pmel cells stimulated with antigen alone had 10 fold higher levels of IFN-γR2 than tumor associated PmelAg+IL-12.

Conclusions

These results suggest that the enhanced anti-tumor activity of PmelAg+IL-12 might be due to their decreased sensitivity to IFN-γ-induced apoptosis. Thus inhibiting IFN-γ-induced activation induced cell death (AICD) by down-regulating IFN-γR2 expression on T cells may represent a novel mechanism by which IL-12 enhances anti-tumor activity.

Author information

Authors and Affiliations

Authors

Rights and permissions

This article is published under license to BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Diaz-Montero, C.M., Tannenbaum, C., Rayman, P. et al. Ex vivo conditioning with IL-12 decreases T cell sensitivity to intratumoral INF-γ-induced apoptosis following adoptive transfer. j. immunotherapy cancer 3 (Suppl 2), P12 (2015). https://doi.org/10.1186/2051-1426-3-S2-P12

Download citation

  • Published:

  • DOI: https://doi.org/10.1186/2051-1426-3-S2-P12

Keywords