Journal of Experimental & Clinical Cancer Research (Jun 2024)

PI3Kγ inhibition combined with DNA vaccination unleashes a B-cell-dependent antitumor immunity that hampers pancreatic cancer

  • Claudia Curcio,
  • Gianluca Mucciolo,
  • Cecilia Roux,
  • Silvia Brugiapaglia,
  • Alessandro Scagliotti,
  • Giorgia Guadagnin,
  • Laura Conti,
  • Dario Longo,
  • Demis Grosso,
  • Mauro Giulio Papotti,
  • Emilio Hirsch,
  • Paola Cappello,
  • Judith A. Varner,
  • Francesco Novelli

DOI
https://doi.org/10.1186/s13046-024-03080-1
Journal volume & issue
Vol. 43, no. 1
pp. 1 – 13

Abstract

Read online

Abstract Phosphoinositide-3-kinase γ (PI3Kγ) plays a critical role in pancreatic ductal adenocarcinoma (PDA) by driving the recruitment of myeloid-derived suppressor cells (MDSC) into tumor tissues, leading to tumor growth and metastasis. MDSC also impair the efficacy of immunotherapy. In this study we verify the hypothesis that MDSC targeting, via PI3Kγ inhibition, synergizes with α-enolase (ENO1) DNA vaccination in counteracting tumor growth. Mice that received ENO1 vaccination followed by PI3Kγ inhibition had significantly smaller tumors compared to those treated with ENO1 alone or the control group, and correlated with i) increased circulating anti-ENO1 specific IgG and IFNγ secretion by T cells, ii) increased tumor infiltration of CD8+ T cells and M1-like macrophages, as well as up-modulation of T cell activation and M1-like related transcripts, iii) decreased infiltration of Treg FoxP3+ T cells, endothelial cells and pericytes, and down-modulation of the stromal compartment and T cell exhaustion gene transcription, iv) reduction of mature and neo-formed vessels, v) increased follicular helper T cell activation and vi) increased “antigen spreading”, as many other tumor-associated antigens were recognized by IgG2c “cytotoxic” antibodies. PDA mouse models genetically devoid of PI3Kγ showed an increased survival and a pattern of transcripts in the tumor area similar to that of pharmacologically-inhibited PI3Kγ-proficient mice. Notably, tumor reduction was abrogated in ENO1 + PI3Kγ inhibition-treated mice in which B cells were depleted. These data highlight a novel role of PI3Kγ in B cell-dependent immunity, suggesting that PI3Kγ depletion strengthens the anti-tumor response elicited by the ENO1 DNA vaccine.