Next Article in Journal
Trans-Regulation of Alternative PD-L1 mRNA Processing by CDK12 in Non-Small-Cell Lung Cancer Cells
Previous Article in Journal
Regulatory TR3-56 Cells in the Complex Panorama of Immune Activation and Regulation
Previous Article in Special Issue
Panobinostat Induced Spatial In Situ Biomarkers Predictive of Anti-PD-1 Efficacy in Mouse Mammary Carcinoma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Immunogenic Cell Stress and Death Sensitize Tumors to Immunotherapy

1
Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, 75006 Paris, France
2
Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, 94800 Villejuif, France
3
Department of Biology, Institut du Cancer Paris Cancer Research and Personalized Medicine (CARPEM), Hôpital Européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris (AP-HP), 75015 Paris, France
*
Authors to whom correspondence should be addressed.
Cells 2023, 12(24), 2843; https://doi.org/10.3390/cells12242843
Submission received: 5 December 2023 / Accepted: 13 December 2023 / Published: 15 December 2023
(This article belongs to the Special Issue Immunogenic Cell Stress and Death)
The efficacy of chemotherapy with cytotoxicants and that of targeted therapies with more sophisticated agents is limited due to the plasticity of malignant cells, which leads to the inevitable development of resistance. Immune checkpoint inhibitors (ICIs) that target inhibitory cytotoxic T lymphocyte associated protein 4 (CTLA4) or T cell exhaustion signals such as programmed cell death-1 (PDCD1, better known as PD-1) and programmed cell death-1 ligand 1 (CD-274, better known as PD-L1) occupy a central stage in the first-line adjuvant and neoadjuvant treatment of advanced neoplasms [1,2]. ICIs are now broadly used through the oncological spectrum.
Nevertheless, certain cytotoxic agents have the ability to induce durable disease control exceeding the clinical treatment phase in many cancer patients. Retrospectively, it appears that those agents that were empirically selected by clinicians for decades, due to their effectiveness, are particularly capable of triggering specific stress and death pathways in cancer cells, rendering them recognizable to the immune system [3,4]. In preclinical mouse models, treatment of tumors with drugs inducing immunogenic cell death (ICD) is only efficient if functional dendritic cells (DCs) and T lymphocytes are available [5,6,7]. In patients, the induction of ICD correlates with the recruitment of antigen-presenting cells (APCs) and cytotoxic T lymphocytes (CTLs) into the tumor bed, and tumor infiltration by DCs and CTLs is indeed a biomarker of favorable prognosis [8,9,10]. ICD inducers include conventional chemotherapeutic agents such as anthracyclines, oxaliplatin, and taxanes, as well as more disease-specific targeted agents from the group of tyrosine kinase inhibitors (TKIs) [11,12]. Moreover, ionizing irradiation [13,14], photodynamic therapy (PDT) [15,16], and oncolytic viruses [17,18] have been shown to induce ICD. In preclinical experimentation and in clinical routine, it appears that combinations of ICD inducers with immune checkpoint inhibitors (ICIs) are particularly efficient in mediating their therapeutic potential [19,20].
ICD facilitates the onset of anticancer immune responses via an increase in both the antigenicity as well as the adjuvanticity of malignant cells. Mechanistically, this involves the onset of coordinated premortem stress responses that can affect the antigenic makeup of cancer cells via genetic or epigenetic alterations of the transcriptome. Moreover, ICD-related cellular stress facilitates the emission of normally confined ‘danger associated molecular patterns’ (DAMPs) that act on the pattern recognition receptors (PRRs) expressed on professional antigen DCs [21,22,23,24]. Thus, immature DCs which express the ATP receptor P2Y2 are chemotactically attracted to the tumor bed by ATP secreted from cancer cells undergoing ICD [15,25]. The final approximation of DCs towards dying cancer cells is facilitated by tumor-emitted annexin A1 (ANXA1), which acts on the formyl peptide receptor-1 (FPR1) present on DCs [24]. Antigen transfer from tumor cells to DCs is mediated by an ‘eat-me’ signal, namely surface-exposed calreticulin, that appears on the plasma membrane of malignant cells and then acts on its receptor CD91, which is present on DCs [13]. The maturation of DCs is ignited via the ligation of Toll-like receptor 4 (TLR4) by the high mobility group box 1 (HMGB1) emanating from the nuclear compartment of dying cancer cells. DC maturation further drives the production of type-1 interferons, which in turn amplifies the synthesis of the C-X-C motif chemokine ligand 10 (CXCL10), thus stimulating T cell priming. In sum, the induction of ICD in malignant cells leads to a coordinated alteration of the cell surface and the local secretome, thereby facilitating the attraction, differentiation, and maturation of DCs so that they present tumor-associated antigens to T cells and, hence, initiate adaptive anticancer immunity [22,23,24]. Of note is that malignant cells and pathogenic viruses can subvert ICD-associated DAMP emission, thus blunting the immune response. Moreover, inherited or acquired defects in the perception of ICD by the host immune system can undermine immunosurveillance and provoke failure of cancer treatments.
The concept of ICD has transcended the realm of preclinical experimentation and is now used for drug discovery (to identify novel ICD inducers) for the design of clinical trials (to identify suitable drug combinations, particularly with ICIs). The present Special Issue, “Immunogenic Cell Stress and Death”, discusses different strategies for inducing anticancer immunity by facilitating the molecular crosstalk between cancer cells, DCs, and T lymphocytes. The role of ICD is reviewed in the context of colorectal, gastric, pancreatic, and hepatocellular cancer as well as multiple myeloma. Moreover, it is revealed that combination treatment with cetuximab plus cisplatin for the treatment of head and neck cancer induced traits of ICD. Interestingly, high doses of the non-immunogenic cell death inducer cisplatin blunted antitumor immunity in this context. Additional aspects of immunogenic cell stress and death cover the role of ICD-associated chemokines and chemokine receptors in the activation of CD8 T-cells and clinical applications thereof. Furthermore, light is shed on the role of tumor-associated macrophages (TAMs) in the response to dying cancer cells. Finally, three-dimensional organ-on-chip technology is introduced for modeling the tumor microenvironment (TME). At the mechanistic level, evidence is presented that targeting the unfolded protein response (UPR) can increase the efficacy of anticancer therapy.
In this Special Issue, ICD induction is discussed in different disease-relevant therapeutic approaches, novel immune signals are described, and the roles of specific immune cell subtypes are elucidated. Advanced in vitro systems help us understand the apical targets of ICD, the organellar genesis of ICD signals, and the complex cellular interplay in the tumor microenvironment. Altogether, an ever more sophisticated pipeline of preclinical ICD-relevant exploration will prepare the basis for the clinical implementation of ICD-based anticancer therapies.

Author Contributions

Conceptualization, O.K. and G.K.; writing—original draft preparation, O.K.; writing—review and editing, G.K.; funding acquisition, O.K. and G.K. All authors have read and agreed to the published version of the manuscript.

Acknowledgments

O.K. receives funding from the DIM elicit (Ile de France), Association pour la recherche sur le cancer (ARC), and Institut National du Cancer (INCa). G.K. is supported by the Ligue contre le Cancer (équipe labellisée); Agence National de la Recherche (ANR)–Projets blancs; AMMICa US23/CNRS UMS3655; ARC; Cancéropôle Ile-de-France; Fondation pour la Recherche Médicale (FRM); a donation by Elior; Equipex Onco-Pheno-Screen; the European Joint Programme on Rare Diseases (EJPRD); the European Research Council (ICD-Cancer); European Union Horizon 2020 Projects Oncobiome and Crimson; Fondation Carrefour; 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.

Conflicts of Interest

O.K. and G.K. hold and have been holding research contracts with Daiichi Sankyo, Eleor, Kaleido, Lytix Pharma, PharmaMar, Osasuna Therapeutics, Samsara Therapeutics, Sanofi, Tollys, and Vascage. G.K. is on the Board of Directors of the Bristol Myers Squibb Foundation France. G.K. is a scientific co-founder of everImmune, Osasuna Therapeutics, Samsara Therapeutics, and Therafast Bio. O.K. is a scientific co-founder of Samsara Therapeutics. G.K. is on the scientific advisory boards of Hevolution, Institut Servier, and Longevity Vision Funds. G.K. is the inventor of patents covering therapeutic targeting of aging, cancer, cystic fibrosis, and metabolic disorders. GK’s brother, Romano Kroemer, was an employee of Sanofi and now consults for Boehringer-Ingelheim. The funders had no role in the design of the study; in the writing of the manuscript, or in the decision to publish the results.

List of Contributions

  • Chiaravalli, M.; Spring, A.; Agostini, A.; Piro, G.; Carbone, C.; Tortora, G. Immunogenic Cell Death: An Emerging Target in Gastrointestinal Cancers. Cells 2022, 11, 3033.
  • De Azevedo, J.; Mourtada, J.; Bour, C.; Devignot, V.; Schultz, P.; Borel, C.; Pencreach, E.; Mellitzer, G.; Gaiddon, C.; Jung, A.C. The EXTREME Regimen Associating Cetuximab and Cisplatin Favors Head and Neck Cancer Cell Death and Immunogenicity with the Induction of an Anti-Cancer Immune Response. Cells 2022, 11, 2866.
  • Johnstone, M.; Vinaixa, D.; Turi, M.; Morelli, E.; Anderson, K.C.; Gulla, A. Promises and Challenges of Immunogenic Chemotherapy in Multiple Myeloma. Cells 2022, 11, 2519.
  • Krysko, D.V.; Demuynck, R.; Efimova, I.; Naessens, F.; Krysko, O.; Catanzaro, E. In Vitro Veritas: From 2D Cultures to Organ-on-a-Chip Models to Study Immunogenic Cell Death in the Tumor Microenvironment. Cells 2022, 11, 3705.
  • Roussot, N.; Ghiringhelli, F.; Rebe, C. Tumor Immunogenic Cell Death as a Mediator of Intratumor CD8 T-Cell Recruitment. Cells 2022, 11, 3672.
  • Rufo, N.; Yang, Y.; De Vleeschouwer, S.; Agostinis, P. The “Yin and Yang” of Unfolded Protein Response in Cancer and Immunogenic Cell Death. Cells 2022, 11, 2899.
  • Tatarova, Z.; Blumberg, D.C.; Bensen, A.; Mills, G.B.; Jonas, O. Panobinostat Induced Spatial In Situ Biomarkers Predictive of Anti-PD-1 Efficacy in Mouse Mammary Carcinoma. Cells 2023, 12, 308.
  • Vanmeerbeek, I.; Govaerts, J.; Laureano, R.S.; Sprooten, J.; Naulaerts, S.; Borras, D.M.; Laoui, D.; Mazzone, M.; Van Ginderachter, J.A.; Garg, A.D. The Interface of Tumour-Associated Macrophages with Dying Cancer Cells in Immuno-Oncology. Cells 2022, 11, 3890.

References

  1. Sharma, P.; Goswami, S.; Raychaudhuri, D.; Siddiqui, B.A.; Singh, P.; Nagarajan, A.; Liu, J.; Subudhi, S.K.; Poon, C.; Gant, K.L.; et al. Immune checkpoint therapy-current perspectives and future directions. Cell 2023, 186, 1652–1669. [Google Scholar] [CrossRef] [PubMed]
  2. Garbe, C.; Dummer, R.; Amaral, T.; Amaria, R.N.; Ascierto, P.A.; Burton, E.M.; Dreno, B.; Eggermont, A.M.M.; Hauschild, A.; Hoeller, C.; et al. Neoadjuvant immunotherapy for melanoma is now ready for clinical practice. Nat. Med. 2023, 29, 1310–1312. [Google Scholar] [CrossRef] [PubMed]
  3. Galassi, C.; Klapp, V.; Yamazaki, T.; Galluzzi, L. Molecular determinants of immunogenic cell death elicited by radiation therapy. In Immunological Reviews; John Wiley & Sons: Hoboken, NJ, USA, 2023. [Google Scholar] [CrossRef]
  4. Kroemer, G.; Galassi, C.; Zitvogel, L.; Galluzzi, L. Immunogenic cell stress and death. Nat. Immunol. 2022, 23, 487–500. [Google Scholar] [CrossRef]
  5. Casares, N.; Pequignot, M.O.; Tesniere, A.; Ghiringhelli, F.; Roux, S.; Chaput, N.; Schmitt, E.; Hamai, A.; Hervas-Stubbs, S.; Obeid, M.; et al. Caspase-dependent immunogenicity of doxorubicin-induced tumor cell death. J. Exp. Med. 2005, 202, 1691–1701. [Google Scholar] [CrossRef] [PubMed]
  6. de Mingo Pulido, A.; Gardner, A.; Hiebler, S.; Soliman, H.; Rugo, H.S.; Krummel, M.F.; Coussens, L.M.; Ruffell, B. TIM-3 Regulates CD103+ Dendritic Cell Function and Response to Chemotherapy in Breast Cancer. Cancer Cell 2018, 33, 60–74.e66. [Google Scholar] [CrossRef] [PubMed]
  7. Ashour, D.; Arampatzi, P.; Pavlovic, V.; Forstner, K.U.; Kaisho, T.; Beilhack, A.; Erhard, F.; Lutz, M.B. IL-12 from endogenous cDC1, and not vaccine DC, is required for Th1 induction. JCI Insight 2020, 5, e135143. [Google Scholar] [CrossRef]
  8. Senovilla, L.; Vitale, I.; Martins, I.; Tailler, M.; Pailleret, C.; Michaud, M.; Galluzzi, L.; Adjemian, S.; Kepp, O.; Niso-Santano, M.; et al. An immunosurveillance mechanism controls cancer cell ploidy. Science 2012, 337, 1678–1684. [Google Scholar] [CrossRef]
  9. Broz, M.L.; Binnewies, M.; Boldajipour, B.; Nelson, A.E.; Pollack, J.L.; Erle, D.J.; Barczak, A.; Rosenblum, M.D.; Daud, A.; Barber, D.L.; et al. Dissecting the tumor myeloid compartment reveals rare activating antigen-presenting cells critical for T cell immunity. Cancer Cell 2014, 26, 638–652. [Google Scholar] [CrossRef]
  10. Stoll, G.; Iribarren, K.; Michels, J.; Leary, A.; Zitvogel, L.; Cremer, I.; Kroemer, G. Calreticulin expression: Interaction with the immune infiltrate and impact on survival in patients with ovarian and non-small cell lung cancer. Oncoimmunology 2016, 5, e1177692. [Google Scholar] [CrossRef]
  11. Liu, P.; Zhao, L.; Pol, J.; Levesque, S.; Petrazzuolo, A.; Pfirschke, C.; Engblom, C.; Rickelt, S.; Yamazaki, T.; Iribarren, K.; et al. Crizotinib-induced immunogenic cell death in non-small cell lung cancer. Nat. Commun. 2019, 10, 1486. [Google Scholar] [CrossRef]
  12. Kang, X.; Zhang, Y.; Song, J.; Wang, L.; Li, W.; Qi, J.; Tang, B.Z. A photo-triggered self-accelerated nanoplatform for multifunctional image-guided combination cancer immunotherapy. Nat. Commun. 2023, 14, 5216. [Google Scholar] [CrossRef] [PubMed]
  13. Obeid, M.; Panaretakis, T.; Joza, N.; Tufi, R.; Tesniere, A.; van Endert, P.; Zitvogel, L.; Kroemer, G. Calreticulin exposure is required for the immunogenicity of gamma-irradiation and UVC light-induced apoptosis. Cell Death Differ. 2007, 14, 1848–1850. [Google Scholar] [CrossRef] [PubMed]
  14. Deutsch, E.; Chargari, C.; Galluzzi, L.; Kroemer, G. Optimising efficacy and reducing toxicity of anticancer radioimmunotherapy. Lancet Oncol. 2019, 20, e452–e463. [Google Scholar] [CrossRef] [PubMed]
  15. Garg, A.D.; Krysko, D.V.; Verfaillie, T.; Kaczmarek, A.; Ferreira, G.B.; Marysael, T.; Rubio, N.; Firczuk, M.; Mathieu, C.; Roebroek, A.J.; et al. A novel pathway combining calreticulin exposure and ATP secretion in immunogenic cancer cell death. EMBO J. 2012, 31, 1062–1079. [Google Scholar] [CrossRef] [PubMed]
  16. Gomes-da-Silva, L.C.; Zhao, L.; Bezu, L.; Zhou, H.; Sauvat, A.; Liu, P.; Durand, S.; Leduc, M.; Souquere, S.; Loos, F.; et al. Photodynamic therapy with redaporfin targets the endoplasmic reticulum and Golgi apparatus. EMBO J. 2018, 37, e98354. [Google Scholar] [CrossRef] [PubMed]
  17. Angelova, A.L.; Grekova, S.P.; Heller, A.; Kuhlmann, O.; Soyka, E.; Giese, T.; Aprahamian, M.; Bour, G.; Ruffer, S.; Cziepluch, C.; et al. Complementary induction of immunogenic cell death by oncolytic parvovirus H-1PV and gemcitabine in pancreatic cancer. J. Virol. 2014, 88, 5263–5276. [Google Scholar] [CrossRef] [PubMed]
  18. Ma, J.; Ramachandran, M.; Jin, C.; Quijano-Rubio, C.; Martikainen, M.; Yu, D.; Essand, M. Characterization of virus-mediated immunogenic cancer cell death and the consequences for oncolytic virus-based immunotherapy of cancer. Cell Death Dis. 2020, 11, 48. [Google Scholar] [CrossRef]
  19. Voorwerk, L.; Slagter, M.; Horlings, H.M.; Sikorska, K.; van de Vijver, K.K.; de Maaker, M.; Nederlof, I.; Kluin, R.J.C.; Warren, S.; Ong, S.; et al. Immune induction strategies in metastatic triple-negative breast cancer to enhance the sensitivity to PD-1 blockade: The TONIC trial. Nat. Med. 2019, 25, 920–928. [Google Scholar] [CrossRef]
  20. Xu, J.; Xu, N.; Bai, Y.; Liu, R.; Mao, C.; Sui, H.; Wang, X.; Jiang, Q.; Dou, Y. Anti-PD-1 antibody HX008 combined with oxaliplatin plus capecitabine for advanced gastric or esophagogastric junction cancer: A multicenter, single-arm, open-label, phase Ib trial. Oncoimmunology 2020, 10, 1864908. [Google Scholar] [CrossRef]
  21. Zitvogel, L.; Perreault, C.; Finn, O.J.; Kroemer, G. Beneficial autoimmunity improves cancer prognosis. Nat. Rev. Clin. Oncol. 2021, 18, 591–602. [Google Scholar] [CrossRef]
  22. Apetoh, L.; Ghiringhelli, F.; Tesniere, A.; Obeid, M.; Ortiz, C.; Criollo, A.; Mignot, G.; Maiuri, M.C.; Ullrich, E.; Saulnier, P.; et al. Toll-like receptor 4-dependent contribution of the immune system to anticancer chemotherapy and radiotherapy. Nat. Med. 2007, 13, 1050–1059. [Google Scholar] [CrossRef] [PubMed]
  23. Sistigu, A.; Yamazaki, T.; Vacchelli, E.; Chaba, K.; Enot, D.P.; Adam, J.; Vitale, I.; Goubar, A.; Baracco, E.E.; Remedios, C.; et al. Cancer cell-autonomous contribution of type I interferon signaling to the efficacy of chemotherapy. Nat. Med. 2014, 20, 1301–1309. [Google Scholar] [CrossRef]
  24. Vacchelli, E.; Ma, Y.; Baracco, E.E.; Sistigu, A.; Enot, D.P.; Pietrocola, F.; Yang, H.; Adjemian, S.; Chaba, K.; Semeraro, M.; et al. Chemotherapy-induced antitumor immunity requires formyl peptide receptor 1. Science 2015, 350, 972–978. [Google Scholar] [CrossRef] [PubMed]
  25. Moriya, T.; Kitagawa, K.; Hayakawa, Y.; Hemmi, H.; Kaisho, T.; Ueha, S.; Ikebuchi, R.; Yasuda, I.; Nakanishi, Y.; Honda, T.; et al. Immunogenic tumor cell death promotes dendritic cell migration and inhibits tumor growth via enhanced T cell immunity. iScience 2021, 24, 102424. [Google Scholar] [CrossRef] [PubMed]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Kepp, O.; Kroemer, G. Immunogenic Cell Stress and Death Sensitize Tumors to Immunotherapy. Cells 2023, 12, 2843. https://doi.org/10.3390/cells12242843

AMA Style

Kepp O, Kroemer G. Immunogenic Cell Stress and Death Sensitize Tumors to Immunotherapy. Cells. 2023; 12(24):2843. https://doi.org/10.3390/cells12242843

Chicago/Turabian Style

Kepp, Oliver, and Guido Kroemer. 2023. "Immunogenic Cell Stress and Death Sensitize Tumors to Immunotherapy" Cells 12, no. 24: 2843. https://doi.org/10.3390/cells12242843

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop