Journal of Experimental & Clinical Cancer Research (Jul 2024)

IL-33 stimulates the anticancer activities of eosinophils through extracellular vesicle-driven reprogramming of tumor cells

  • Adriana Rosa Gambardella,
  • Caterina Antonucci,
  • Cristiana Zanetti,
  • Francesco Noto,
  • Sara Andreone,
  • Davide Vacca,
  • Valentina Pellerito,
  • Chiara Sicignano,
  • Giuseppe Parrottino,
  • Valentina Tirelli,
  • Antonella Tinari,
  • Mario Falchi,
  • Adele De Ninno,
  • Luca Businaro,
  • Stefania Loffredo,
  • Gilda Varricchi,
  • Claudio Tripodo,
  • Claudia Afferni,
  • Isabella Parolini,
  • Fabrizio Mattei,
  • Giovanna Schiavoni

DOI
https://doi.org/10.1186/s13046-024-03129-1
Journal volume & issue
Vol. 43, no. 1
pp. 1 – 25

Abstract

Read online

Abstract Immune cell-derived extracellular vesicles (EV) affect tumor progression and hold promise for therapeutic applications. Eosinophils are major effectors in Th2-related pathologies recently implied in cancer. Here, we evaluated the anti-tumor activities of eosinophil-derived EV following activation with the alarmin IL-33. We demonstrate that IL-33-activated mouse and human eosinophils produce higher quantities of EV with respect to eosinophils stimulated with IL-5. Following incorporation of EV from IL-33-activated eosinophils (Eo33-EV), but not EV from IL-5-treated eosinophils (Eo5-EV), mouse and human tumor cells increased the expression of cyclin-dependent kinase inhibitor (CDKI)-related genes resulting in cell cycle arrest in G0/G1, reduced proliferation and inhibited tumor spheroid formation. Moreover, tumor cells incorporating Eo33-EV acquired an epithelial-like phenotype characterized by E-Cadherin up-regulation, N-Cadherin downregulation, reduced cell elongation and migratory extent in vitro, and impaired capacity to metastasize to lungs when injected in syngeneic mice. RNA sequencing revealed distinct mRNA signatures in Eo33-EV and Eo5-EV with increased presence of tumor suppressor genes and enrichment in pathways related to epithelial phenotypes and negative regulation of cellular processes in Eo33-EV compared to Eo5-EV. Our studies underscore novel IL-33-stimulated anticancer activities of eosinophils through EV-mediated reprogramming of tumor cells opening perspectives on the use of eosinophil-derived EV in cancer therapy.

Keywords