Therapeutic blockade of Foxp3 in experimental breast cancer models

Purpose Regulatory T cells (Tregs) impair the clinical benefit of cancer immunotherapy. To optimize the antitumor efficacy of therapeutic dendritic cell (DC) vaccines, we aimed to inhibit Foxp3, a transcription factor required for Treg function. Methods Mice bearing established syngeneic LM3 and 4T1...

Full description

Saved in:
Bibliographic Details
Published in:Breast cancer research and treatment 2017-11, Vol.166 (2), p.393-405
Main Authors: Moreno Ayala, Mariela A., Gottardo, María Florencia, Imsen, Mercedes, Asad, Antonela S., Bal de Kier Joffé, Elisa, Casares, Noelia, Lasarte, Juan José, Seilicovich, Adriana, Candolfi, Marianela
Format: Article
Language:eng
Subjects:
Online Access:Get full text
Tags: Add Tag
No Tags, Be the first to tag this record!
Description
Summary:Purpose Regulatory T cells (Tregs) impair the clinical benefit of cancer immunotherapy. To optimize the antitumor efficacy of therapeutic dendritic cell (DC) vaccines, we aimed to inhibit Foxp3, a transcription factor required for Treg function. Methods Mice bearing established syngeneic LM3 and 4T1 breast tumors were treated with antitumor DC vaccines and a synthetic peptide (P60) that has been shown to inhibit Foxp3. Results Treatment with P60 improved the therapeutic efficacy of DC vaccines in these experimental models. In addition, monotherapy with P60 inhibited tumor growth in immunocompetent as well as in immuno-compromised animals bearing established tumors. We found expression of Foxp3 in human and murine breast tumor cells. P60 inhibited IL-10 secretion in breast cancer cells that expressed Foxp3. Conclusions Our results suggest that Foxp3 blockade improves the therapeutic efficacy of DC vaccines by inhibition of Tregs and through a direct antitumor effect. This strategy could prove useful to neutralize the immunosuppressive microenvironment and to boost antitumor immunity in breast cancer.
ISSN:0167-6806
1573-7217