Back to Search Start Over

Guadecitabine increases response to combined anti-CTLA-4 and anti-PD-1 treatment in mouse melanoma in vivo by controlling T-cells, myeloid derived suppressor and NK cells

Authors :
Adriana Amaro
Francesco Reggiani
Daniela Fenoglio
Rosaria Gangemi
Anna Tosi
Alessia Parodi
Barbara Banelli
Valentina Rigo
Luca Mastracci
Federica Grillo
Alessandra Cereghetti
Aizhan Tastanova
Adhideb Ghosh
Fabio Sallustio
Laura Emionite
Antonio Daga
Tiziana Altosole
Gilberto Filaci
Antonio Rosato
Mitchell Levesque
Michele Maio
Ulrich Pfeffer
Michela Croce
EPigenetic Immune-oncology Consortium Airc (EPICA) consortium
Source :
Journal of Experimental & Clinical Cancer Research, Vol 42, Iss 1, Pp 1-24 (2023)
Publication Year :
2023
Publisher :
BMC, 2023.

Abstract

Abstract Background The combination of Programmed Cell Death 1 (PD-1) and Cytotoxic T-Lymphocyte Antigen 4 (CTLA-4) blockade has dramatically improved the overall survival rate for malignant melanoma. Immune checkpoint blockers (ICBs) limit the tumor’s immune escape yet only for approximately a third of all tumors and, in most cases, for a limited amount of time. Several approaches to overcome resistance to ICBs are being investigated among which the addition of epigenetic drugs that are expected to act on both immune and tumor cells. Guadecitabine, a dinucleotide prodrug of a decitabine linked via phosphodiester bond to a guanosine, showed promising results in the phase-1 clinical trial, NIBIT-M4 (NCT02608437). Methods We used the syngeneic B16F10 murine melanoma model to study the effects of immune checkpoint blocking antibodies against CTLA-4 and PD-1 in combination, with and without the addition of Guadecitabine. We comprehensively characterized the tumor’s and the host’s responses under different treatments by flow cytometry, multiplex immunofluorescence and methylation analysis. Results In combination with ICBs, Guadecitabine significantly reduced subcutaneous tumor growth as well as metastases formation compared to ICBs and Guadecitabine treatment. In particular, Guadecitabine greatly enhanced the efficacy of combined ICBs by increasing effector memory CD8+ T cells, inducing effector NK cells in the spleen and reducing tumor infiltrating regulatory T cells and myeloid derived suppressor cells (MDSC), in the tumor microenvironment (TME). Guadecitabine in association with ICBs increased serum levels of IFN-γ and IFN-γ-induced chemokines with anti-angiogenic activity. Guadecitabine led to a general DNA-demethylation, in particular of sites of intermediate methylation levels. Conclusions These results indicate Guadecitabine as a promising epigenetic drug to be added to ICBs therapy.

Details

Language :
English
ISSN :
17569966
Volume :
42
Issue :
1
Database :
Directory of Open Access Journals
Journal :
Journal of Experimental & Clinical Cancer Research
Publication Type :
Academic Journal
Accession number :
edsdoj.54d112fcea9d430e9f590e99dd740788
Document Type :
article
Full Text :
https://doi.org/10.1186/s13046-023-02628-x