1,368
Views
4
CrossRef citations to date
0
Altmetric
Editorial

Arsenic trioxide as an inducer of immunogenic cell death

ORCID Icon, , & ORCID Icon

Arsenic trioxide (ATO) is often combined with all-trans retinoic acid (ATRA) to treat promyelocytic leukemia (PML) with a rather high success rate. In mice, it has been documented that ATRA is much more efficient against PML developing in immunocompetent than in immunodeficient mice,Citation1,Citation2 pleading in favor of the idea that the antileukemic action of ATRA depends on the immune system. However, no such immune-dependent effects of ATO have been described in PML. Nonetheless, it has been shown that ATO increases lymphokine activated killer (LAK)-mediated cytotoxicity against human myeloma cellsCitation3 and enhances the efficacy of Bacille Calmette-Guérin (BCG) immunotherapy in a mouse model of bladder cancer.Citation4 Moreover, ATO has been demonstrated to deplete regulatory T cells in a mouse model of colon cancer.Citation5 Of note, in a recent paper published in Cellular and Molecular Immunology, Chen et al. demonstrate that ATO can trigger immunogenic cell death (ICD) in solid tumors.Citation6

The concept of ICD, initially established in cells undergoing apoptosis, has recently been extended to other variants of regulated cell death such as necroptosis, pyroptosis, and ferroptosis.Citation7–11 Canonical ICD triggers the emission of a set of danger associated molecular patterns (DAMPs), which act on specific pattern recognition receptors (PRRs) expressed by antigen presenting dendritic cells (DCs), thus stimulating phagocytosis of malignant cells and antigen presentation of tumor-associated antigens by DCs.Citation12,Citation13 Mature DCs facilitate cross-presentation of tumor antigens to cytotoxic T lymphocytes (CTL) as well as the education of memory T cells, altogether conferring efficacy to cancer therapies that last beyond treatment discontinuation.Citation12 Preclinical and clinical data support the notion that ICD inducers can be advantageously combined with additional immunotherapies such as immune checkpoint blockade targeting the PD-1/PD-L1 interaction.

In their work, Chen et al. discovered that in vitro cultures of malignant cells with ATO led to the generation of a whole cell vaccine that could be injected into mice to reduce cancer growth in prophylactic as well as in therapeutic settings.Citation6 These anticancer effects of ATO-treated cancer cells were lost or attenuated upon depletion of CD8+ (but not NK1.1+) T cells, as well as after blocking either interferon-ϒ (IFNϒ) or the Type-1 interferon receptor (IFNAR) with suitable antibodies. ATO-treated cells manifested several well-established hallmarks of ICD including the release of ATP and high-mobility group B1 (HMGB1) protein, the exposure of calreticulin (CALR) on the cell surface, the induction of cGAMP production, and the H151-repressible (and hence likely STING-dependent) induction of interferon-β1 (IFNβ1).Citation6

At the mechanistic level, the authors described that ATO induced biochemical characteristics of several cellular stress and death routines including autophagy, apoptosis, ferroptosis, necroptosis, and pyroptosis that all were blunted when ATO-induced oxidative stress was quenched by N-acetyl-L-cysteine. However, the knockout or knockdown of genes required for apoptosis (Bak, Bax), autophagy (Becn1), ferroptosis (Acsl4), necroptosis (Mlk1, Ripk3) and pyroptosis (Gsdmd, Gsdme) did not prevent ATO-induced cell killing, indicating that none among these pathways is indispensable for the lethal outcome of ATO treatment. In stark contrast, knockout of several among these effectors, in particular Acsl4 (involved in ferroptosis) and Mlk1 or Ripk3 (involved in necroptosis), fully abolished the capacity of ATO-treated TC-1 non-small cell lung cancer to induce prophylactic anti-TC-1 immune responses in mice. The knockout of Bak or Bax (both involved in apoptosis) or Becn1 (involved in autophagy) yielded a partial phenotype (i.e., attenuation but not abolition of vaccination), while the knockout of Gsdmd, Gsdme (involved in pyroptosis) failed to affect the capacity of ATO-treated TC-1 cells to induce a protective anti-TC-1 immune response. Hence, several among the ATO-triggered subroutines (autophagy, apoptosis, ferroptosis, necroptosis) contribute to the vaccination effect. Mechanistically, the authors showed that Acsl4, Bak, Bax,Becn1, Mkl1, and Ripk3 contributed to ATO-induced ATP release; Mkl1 and Ripk3 to HMGB1 release; Acsl4, Bak, Bax,Mlkl, and Ripk3 to CALR exposure; and Acsl4, Mlkl, and Ripk3 to extracellular cGAMP accumulation, IFNβ1 secretion and transcription of IFN-stimulated genes (ISG).Citation6

In a subsequent step, Chen et al. showed that intratumoral injection of ATO failed to suppress the outgrowth of TC-1 or MCA205 fibrosarcomas in vivo.Citation6 In contrast, immunization with an ATO-based whole-cell vaccine partially restrained the growth of established TC-1 or MCA205 tumors. Such therapeutic whole-cell vaccines required expression of Acsl4, Mlkl, and Ripk3. Of note, the failure of Ripk3−/− cells to act as a therapeutic vaccine could be restored when ATO treatment in vitro was combined with drugs that enhance extracellular ATP (the apyrase inhibitor ARL67156), ligate Toll-like receptor-4 (monophosphoryl lipid A) or activate STING (2ʹ3’-cGAMP). Of note, the ATO-based whole-cell vaccine increased its capacity to restrain tumor growth if combined with PD-1 blockade.Citation6 Although these latter effects appeared additive (rather than synergistic), they delineate a possible strategy for improving the efficacy of therapeutic vaccination with cells undergoing ICD.

Altogether, the aforementioned data support the idea that ATO, which is likely the first antineoplastic chemotherapy that was used in the world (in particular in China), can stimulate several facets of ICD. Importantly, ATO activates a mixed pattern of stress and death pathways, many of which contribute to the immunogenic effects of ATO-treated cancer cells (). Future studies must explore the possibility that such a pleiotropic pattern of ICD might be more efficient in yielding therapeutic cancer vaccines than ICD relying on single or dual-cell stress/death subroutine(s). In other words, would a mixture of four stress/death pathways (autophagy, apoptosis, ferroptosis, and necroptosis) generate a more efficient ICD-based vaccine that only involves one or combinations of two or three pathways? The answer to this question will have implications for the future clinical development of therapeutic vaccines.

Figure 1. ATO induces canonical and non-canonical traits of immunogenic cell death. (a) Reactive oxygen species (ROS) induced by arsenic trioxide (ATO) trigger both canonical and non-canonical immunogenic cell death (ICD) responses such as the onset of autophagy and apoptosis as well the execution of necroptosis and ferroptosis, respectively. ATO-induced ICD facilitates the emission of danger associated molecular patterns (DAMP), including the exposure of calreticulin (CALR), the release of adenosine triphosphate (ATP), the exodus of high mobility group box 1 (HMGB1), the liberation of cyclic guanosine monophosphate–adenosine monophosphate (cGAMP) as well as Type I interferon (Type I IFN) responses. ICD-associated DAMPs act on dendritic cells (DC) and stimulate antigen presentation to T cells. T cell priming ultimately leads to clonal expansion of cytotoxic T lymphocytes (CTL) and the education of memory T cells, altogether inducing anticancer immune responses. ICD inducers can be advantageously combined with immune checkpoint blockade leveraging the full potential of T cell-mediated adaptive immunity. (b) Mechanistic effects on ICD induction of knockout (KO) or knockdown (KD) of essential genes in cell stress and cell death routines.

Figure 1. ATO induces canonical and non-canonical traits of immunogenic cell death. (a) Reactive oxygen species (ROS) induced by arsenic trioxide (ATO) trigger both canonical and non-canonical immunogenic cell death (ICD) responses such as the onset of autophagy and apoptosis as well the execution of necroptosis and ferroptosis, respectively. ATO-induced ICD facilitates the emission of danger associated molecular patterns (DAMP), including the exposure of calreticulin (CALR), the release of adenosine triphosphate (ATP), the exodus of high mobility group box 1 (HMGB1), the liberation of cyclic guanosine monophosphate–adenosine monophosphate (cGAMP) as well as Type I interferon (Type I IFN) responses. ICD-associated DAMPs act on dendritic cells (DC) and stimulate antigen presentation to T cells. T cell priming ultimately leads to clonal expansion of cytotoxic T lymphocytes (CTL) and the education of memory T cells, altogether inducing anticancer immune responses. ICD inducers can be advantageously combined with immune checkpoint blockade leveraging the full potential of T cell-mediated adaptive immunity. (b) Mechanistic effects on ICD induction of knockout (KO) or knockdown (KD) of essential genes in cell stress and cell death routines.

Acknowledgments

OK receives grants by the DIM ELICIT initiative of the Ile de France and Institut National du Cancer (INCa). GK is supported by the Ligue contre le Cancer (équipe labellisée); Agence National de la Recherche (ANR) – Projets blancs; AMMICa US23/CNRS UMS3655; Association pour la recherche sur le cancer (ARC); Cancéropôle Ile-de-France; Fondation pour la Recherche Médicale (FRM); a donation by Elior; Equipex Onco-Pheno-Screen; European Joint Programme on Rare Diseases (EJPRD); European Research Council (ICD-Cancer), European Union Horizon 2020 Projects Oncobiome and Crimson; Fondation Carrefour; Institut National du Cancer (INCa); Institut Universitaire de France; LabEx Immuno-Oncology (ANR-18-IDEX-0001); a Cancer Research ASPIRE Award from the Mark Foundation; the RHU Immunolife; Seerave Foundation; SIRIC Stratified Oncology Cell DNA Repair and Tumor Immune Elimination (SOCRATE); and SIRIC Cancer Research and Personalized Medicine (CARPEM). This study contributes to the IdEx Université de Paris ANR-18-IDEX-0001.

Data availability statement

All data that led to the conclusions in this manuscript have been included and all sources have been described.

Disclosure statement

OK is a cofounder of Samsara Therapeutics. GK has been holding research contracts with Daiichi Sankyo, Eleor, Kaleido, Lytix Pharma, PharmaMar, Osasuna Therapeutics, Samsara Therapeutics, Sanofi, Tollys, and Vascage. GK has been consulting for Reithera. GK is on the Board of Directors of the Bristol Myers Squibb Foundation France. GK is a scientific co-founder of EverImmune, Osasuna Therapeutics, Samsara Therapeutics, and Therafast Bio. GK is the inventor of patents covering therapeutic targeting of aging, cancer, cystic fibrosis, and metabolic disorders. GK’s wife, Laurence Zitvogel, has held research contracts with 9 Meters Biopharma, Daiichi Sankyo, Pilege, was on the Board of Directors of Transgene, is a cofounder of EverImmune, and holds patents covering the treatment of cancer and the therapeutic manipulation of the microbiota. GK’s brother, Romano Kroemer, was an employee of Sanofi and now consults for Boehringer-Ingelheim.

References

  • Westervelt P, Pollock JL, Oldfather KM, Walter MJ, Ma MK, Williams A, DiPersio JF, Ley TJ. Adaptive immunity cooperates with liposomal all-trans-retinoic acid (ATRA) to facilitate long-term molecular remissions in mice with acute promyelocytic leukemia. Proc Natl Acad Sci U S A. 2002;99(14):9468–3. doi:10.1073/pnas.132657799.
  • de The H, Pandolfi PP, Chen Z. Acute promyelocytic Leukemia: a paradigm for oncoprotein-targeted cure. Cancer Cell. 2017;32(5):552–560. doi:10.1016/j.ccell.2017.10.002.
  • Deaglio S, Canella D, Baj G, Arnulfo A, Waxman S, Malavasi F. Evidence of an immunologic mechanism behind the therapeutical effects of arsenic trioxide (As(2)O(3)) on myeloma cells. Leuk Res. 2001;25(3):227–235. doi:10.1016/S0145-2126(00)00105-3.
  • Mao MH, Huang HB, Zhang XL, Li K, Liu YL, Wang P. Additive antitumor effect of arsenic trioxide combined with intravesical bacillus Calmette-Guerin immunotherapy against bladder cancer through blockade of the IER3/Nrf2 pathway. Biomed Pharmacother. 2018;107:1093–1103. doi:10.1016/j.biopha.2018.08.057.
  • Thomas-Schoemann A, Batteux F, Mongaret C, Nicco C, Chereau C, Annereau M, Dauphin A, Goldwasser F, Weill B, Lemare F, et al. Arsenic trioxide exerts antitumor activity through regulatory T cell depletion mediated by oxidative stress in a murine model of colon cancer. J Immunol. 2012;189(11):5171–5177. doi:10.4049/jimmunol.1103094.
  • Chen J, Jin Z, Zhang S, Zhang X, Li P, Yang H, Ma Y. Arsenic trioxide elicits prophylactic and therapeutic immune responses against solid tumors by inducing necroptosis and ferroptosis. Cell Mol Immunol. 2022;20(1):51–64. doi:10.1038/s41423-022-00956-0.
  • Stoll G, Ma Y, Yang H, Kepp O, Zitvogel L, Kroemer G. Pro-necrotic molecules impact local immunosurveillance in human breast cancer. Oncoimmunology. 2017;6(4):e1299302. doi:10.1080/2162402X.2017.1299302.
  • Yang H, Ma Y, Chen G, Zhou H, Yamazaki T, Klein C, Pietrocola F, Vacchelli E, Souquere S, Sauvat A, et al. Contribution of RIP3 and MLKL to immunogenic cell death signaling in cancer chemotherapy. Oncoimmunology. 2016;5(6):e1149673. doi:10.1080/2162402X.2016.1149673.
  • Lu Y, Xu F, Wang Y, Shi C, Sha Y, He G, Yao Q, Shao K, Sun W, Du J, et al. Cancer immunogenic cell death via photo-pyroptosis with light-sensitive Indoleamine 2,3-dioxygenase inhibitor conjugate. Biomaterials. 2021;278:121167. doi:10.1016/j.biomaterials.2021.121167.
  • Kepp O, Kroemer G. Is ferroptosis immunogenic? The devil is in the details! Oncoimmunology. 2022;11(1):2127273. doi:10.1080/2162402X.2022.2127273.
  • Tang D, Kepp O, Kroemer G. Ferroptosis becomes immunogenic: implications for anticancer treatments. Oncoimmunology. 2020;10(1):1862949. doi:10.1080/2162402X.2020.1862949.
  • Kroemer G, Galassi C, Zitvogel L, Galluzzi L. Immunogenic cell stress and death. Nat Immunol. 2022;23(4):487–500. doi:10.1038/s41590-022-01132-2.
  • Krysko DV, Garg AD, Kaczmarek A, Krysko O, Agostinis P, Vandenabeele P. Immunogenic cell death and DAMPs in cancer therapy. Nat Rev Cancer. 2012;12(12):860–875. doi:10.1038/nrc3380.