Your browser doesn't support javascript.
loading
Pyroptosis activates conventional type I dendritic cells to mediate the priming of highly functional anticancer T cells.
Inkol, Jordon M; Westerveld, Michael J; Verburg, Shayla G; Walsh, Scott R; Morrison, Jodi; Mossman, Karen L; Worfolk, Sarah M; Kallio, Kaslyn Lf; Phippen, Noah J; Burchett, Rebecca; Wan, Yonghong; Bramson, Jonathan; Workenhe, Samuel T.
Affiliation
  • Inkol JM; Department of Pathobiology, University of Guelph, Guelph, Ontario, Canada.
  • Westerveld MJ; Department of Pathobiology, University of Guelph, Guelph, Ontario, Canada.
  • Verburg SG; Department of Pathobiology, University of Guelph, Guelph, Ontario, Canada.
  • Walsh SR; Department of Pathobiology, University of Guelph, Guelph, Ontario, Canada.
  • Morrison J; Department of Biomedical Sciences, University of Guelph, Guelph, Ontario, Canada.
  • Mossman KL; Department of Medicine, McMaster University, Hamilton, Ontario, Canada.
  • Worfolk SM; Department of Pathobiology, University of Guelph, Guelph, Ontario, Canada.
  • Kallio KL; Department of Pathobiology, University of Guelph, Guelph, Ontario, Canada.
  • Phippen NJ; Department of Pathobiology, University of Guelph, Guelph, Ontario, Canada.
  • Burchett R; Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada.
  • Wan Y; Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada.
  • Bramson J; Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada.
  • Workenhe ST; Department of Pathobiology, University of Guelph, Guelph, Ontario, Canada sworkenh@uoguelph.ca.
J Immunother Cancer ; 12(4)2024 Apr 04.
Article in En | MEDLINE | ID: mdl-38580330
ABSTRACT

BACKGROUND:

Initiation of antitumor immunity is reliant on the stimulation of dendritic cells (DCs) to present tumor antigens to naïve T cells and generate effector T cells that can kill cancer cells. Induction of immunogenic cell death after certain types of cytotoxic anticancer therapies can stimulate T cell-mediated immunity. However, cytotoxic therapies simultaneously activate multiple types of cellular stress and programmed cell death; hence, it remains unknown what types of cancer cell death confer superior antitumor immunity.

METHODS:

Murine cancer cells were engineered to activate apoptotic or pyroptotic cell death after Dox-induced expression of procell death proteins. Cell-free supernatants were collected to measure secreted danger signals, cytokines, and chemokines. Tumors were formed by transplanting engineered tumor cells to specifically activate apoptosis or pyroptosis in established tumors and the magnitude of immune response measured by flow cytometry. Tumor growth was measured using calipers to estimate end point tumor volumes for Kaplan-Meier survival analysis.

RESULTS:

We demonstrated that, unlike apoptosis, pyroptosis induces an immunostimulatory secretome signature. In established tumors pyroptosis preferentially activated CD103+ and XCR1+ type I conventional DCs (cDC1) along with a higher magnitude and functionality of tumor-specific CD8+ T cells and reduced number of regulatory T cells within the tumor. Depletion of cDC1 or CD4+ and CD8+ T cells ablated the antitumor response leaving mice susceptible to a tumor rechallenge.

CONCLUSION:

Our study highlights that distinct types of cell death yield varying immunotherapeutic effect and selective activation of pyroptosis can be used to potentiate multiple aspects of the anticancer immunity cycle.
Subject(s)
Key words

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: CD8-Positive T-Lymphocytes / Neoplasms Limits: Animals Language: En Journal: J Immunother Cancer Year: 2024 Document type: Article Affiliation country: Canada

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: CD8-Positive T-Lymphocytes / Neoplasms Limits: Animals Language: En Journal: J Immunother Cancer Year: 2024 Document type: Article Affiliation country: Canada