1. Academic Validation
  2. Arsenic trioxide elicits prophylactic and therapeutic immune responses against solid tumors by inducing necroptosis and ferroptosis

Arsenic trioxide elicits prophylactic and therapeutic immune responses against solid tumors by inducing necroptosis and ferroptosis

  • Cell Mol Immunol. 2023 Jan;20(1):51-64. doi: 10.1038/s41423-022-00956-0.
Jinfeng Chen 1 2 Ziqi Jin 1 2 Shuqing Zhang 1 2 Xiao Zhang 1 2 Peipei Li 1 2 Heng Yang 1 2 3 Yuting Ma 4 5 6 7
Affiliations

Affiliations

  • 1 Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 10005, China.
  • 2 Suzhou Institute of Systems Medicine, Suzhou, Jiangsu, 215123, China.
  • 3 National Key Laboratory of Medical Immunology, Shanghai, 200433, China.
  • 4 Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 10005, China. [email protected].
  • 5 Suzhou Institute of Systems Medicine, Suzhou, Jiangsu, 215123, China. [email protected].
  • 6 National Key Laboratory of Medical Immunology, Shanghai, 200433, China. [email protected].
  • 7 Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 211166, China. [email protected].
Abstract

Boosting tumor immunosurveillance with vaccines has been proven to be a feasible and cost-effective strategy to fight Cancer. Although major breakthroughs have been achieved in preventative tumor vaccines targeting oncogenic viruses, limited advances have been made in curative vaccines for virus-irrelevant malignancies. Accumulating evidence suggests that preconditioning tumor cells with certain cytotoxic drugs can generate whole-cell tumor vaccines with strong prophylactic activities. However, the immunogenicity of these vaccines is not sufficient to restrain the outgrowth of existing tumors. In this study, we identified arsenic trioxide (ATO) as a wide-spectrum cytotoxic and highly immunogenic drug through multiparameter screening. ATO preconditioning could generate whole-cell tumor vaccines with potent antineoplastic effects in both prophylactic and therapeutic settings. The tumor-preventive or tumor-suppressive benefits of these vaccines relied on CD8+ T cells and type I and II interferon signaling and could be linked to the release of immunostimulatory danger molecules. Unexpectedly, following ATO-induced oxidative stress, multiple cell death pathways were activated, including Autophagy, Apoptosis, Necroptosis, and Ferroptosis. CRISPR‒Cas9-mediated knockout of cell death executors revealed that the absence of Rip3, Mlkl, or Acsl4 largely abolished the efficacy of ATO-based prophylactic and therapeutic Cancer vaccines. This therapeutic failure could be rescued by coadministration of danger molecule analogs. In addition, PD-1 blockade synergistically improved the therapeutic efficacy of ATO-based Cancer vaccines by augmenting local IFN-γ production.

Keywords

Arsenic trioxide; Ferroptosis; Immunosurveillance; Necroptosis; Tumor vaccine.

Figures
Products