Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Cell death in pancreatic cancer: from pathogenesis to therapy

Abstract

Pancreatic cancer is a devastating gastrointestinal cancer characterized by late diagnosis, limited treatment success and dismal prognosis. Exocrine tumours account for 95% of pancreatic cancers and the most common pathological type is pancreatic ductal adenocarcinoma (PDAC). The occurrence and progression of PDAC involve multiple factors, including internal genetic alterations and external inflammatory stimuli. The biology and therapeutic response of PDAC are further shaped by various forms of regulated cell death, such as apoptosis, necroptosis, ferroptosis, pyroptosis and alkaliptosis. Cell death induced by local or systemic treatments suppresses tumour proliferation, invasion and metastasis. However, unrestricted cell death or tissue damage might result in an inflammation-related immunosuppressive microenvironment, which is conducive to tumour progression or recurrence. The precise extent to which cell death affects PDAC is not yet well described. A growing body of preclinical and clinical studies document significant correlations between mutations (for example, in KRAS and TP53), stress responses (such as hypoxia and autophagy), metabolic reprogramming and chemotherapeutic responses. Here, we describe the molecular machinery of cell death, discuss the complexity and multifaceted nature of lethal signalling in PDAC cells, and highlight the challenges and opportunities for activating cell death pathways through precision oncology treatments.

Key points

  • The morbidity and mortality of pancreatic cancer continue to increase and constitute a major challenge for basic and applied oncology.

  • Regulated cell death occurs through apoptotic and non-apoptotic pathways, manifesting in different morphological, biochemical and genetic characteristics.

  • Regulated cell death plays a dual role in pancreatic cancer and has been shown to have both pro-tumorigenic and tumour-suppressive effects.

  • Endogenous damage-associated molecular patterns released from stressed, dying or dead cells play a key role in regulating inflammation and immune responses in the pancreatic tumour microenvironment.

  • A concerted preclinical and clinical evaluation is needed to determine whether therapies can induce adequate cell death responses.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Core molecular mechanisms of pancreatic cancer cell death.
Fig. 2: Modulation of cell death pathways in pancreatic cancer.
Fig. 3: TP53 in pancreatic cancer cell death.
Fig. 4: Hypoxia in pancreatic cancer cell death.
Fig. 5: Metabolism in pancreatic cancer cell death.
Fig. 6: Autophagy in pancreatic cancer cell death.
Fig. 7: Crosstalk between dying pancreatic cancer cells and immune cells.

Similar content being viewed by others

References

  1. Siegel, R. L., Miller, K. D. & Jemal, A. Cancer statistics, 2020. CA Cancer J. Clin. 70, 7–30 (2020).

    Article  PubMed  Google Scholar 

  2. Kleeff, J. et al. Pancreatic cancer. Nat. Rev. Dis. Prim. 2, 16022 (2016).

    Article  PubMed  Google Scholar 

  3. Rawla, P., Sunkara, T. & Gaduputi, V. Epidemiology of pancreatic cancer: global trends, etiology and risk factors. World J. Oncol. 10, 10–27 (2019).

    Article  PubMed Central  PubMed  Google Scholar 

  4. Sarantis, P., Koustas, E., Papadimitropoulou, A., Papavassiliou, A. G. & Karamouzis, M. V. Pancreatic ductal adenocarcinoma: treatment hurdles, tumor microenvironment and immunotherapy. World J. Gastrointest. Oncol. 12, 173–181 (2020).

    Article  PubMed Central  PubMed  Google Scholar 

  5. Tesfaye, A. A. & Philip, P. A. Adjuvant treatment of surgically resectable pancreatic ductal adenocarcinoma. Clin. Adv. Hematol. Oncol. 17, 54–63 (2019).

    PubMed  Google Scholar 

  6. Neoptolemos, J. P. et al. Therapeutic developments in pancreatic cancer: current and future perspectives. Nat. Rev. Gastroenterol. Hepatol. 15, 333–348 (2018).

    Article  PubMed  Google Scholar 

  7. Nevala-Plagemann, C., Hidalgo, M. & Garrido-Laguna, I. From state-of-the-art treatments to novel therapies for advanced-stage pancreatic cancer. Nat. Rev. Clin. Oncol. 17, 108–123 (2020).

    Article  PubMed  Google Scholar 

  8. Ho, W. J., Jaffee, E. M. & Zheng, L. The tumour microenvironment in pancreatic cancer - clinical challenges and opportunities. Nat. Rev. Clin. Oncol. 17, 527–540 (2020).

    Article  PubMed Central  PubMed  Google Scholar 

  9. Hosein, A. N., Brekken, R. A. & Maitra, A. Pancreatic cancer stroma: an update on therapeutic targeting strategies. Nat. Rev. Gastroenterol. Hepatol. 17, 487–505 (2020).

    Article  PubMed Central  PubMed  Google Scholar 

  10. Quinonero, F. et al. The challenge of drug resistance in pancreatic ductal adenocarcinoma: a current overview. Cancer Biol. Med. 16, 688–699 (2019).

    CAS  PubMed Central  PubMed  Google Scholar 

  11. Galluzzi, L. et al. Molecular mechanisms of cell death: recommendations of the Nomenclature Committee on Cell Death 2018. Cell Death Differ. 25, 486–541 (2018).

    Article  PubMed Central  PubMed  Google Scholar 

  12. Tang, D., Kang, R., Berghe, T. V., Vandenabeele, P. & Kroemer, G. The molecular machinery of regulated cell death. Cell Res. 29, 347–364 (2019).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  13. Hanahan, D. & Weinberg, R. A. Hallmarks of cancer: the next generation. Cell 144, 646–674 (2011).

    Article  CAS  PubMed  Google Scholar 

  14. Lee, S. Y. et al. Regulation of tumor progression by programmed necrosis. Oxid. Med. Cell. Longev. 2018, 3537471 (2018).

    PubMed Central  PubMed  Google Scholar 

  15. Chen, X., Kang, R., Kroemer, G. & Tang, D. Broadening horizons: the role of ferroptosis in cancer. Nat. Rev. Clin. Oncol. 18, 280–296 (2021).

    Article  CAS  PubMed  Google Scholar 

  16. Kerr, J. F., Wyllie, A. H., & Currie, A. R. Apoptosis: a basic biological phenomenon with wide-ranging implications in tissue kinetics. Br. J. Cancer 26, 239–257 (1972).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  17. Miller-Ocuin, J. L. et al. DNA released from neutrophil extracellular traps (NETs) activates pancreatic stellate cells and enhances pancreatic tumor growth. Oncoimmunology 8, e1605822 (2019).

    Article  PubMed Central  PubMed  Google Scholar 

  18. Miyatake, Y. et al. Visualising the dynamics of live pancreatic microtumours self-organised through cell-in-cell invasion. Sci. Rep. 8, 14054 (2018).

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  19. Carneiro, B. A. & El-Deiry, W. S. Targeting apoptosis in cancer therapy. Nat. Rev. Clin. Oncol. 17, 395–417 (2020).

    Article  PubMed Central  PubMed  Google Scholar 

  20. Li, P. et al. Cytochrome c and dATP-dependent formation of Apaf-1/caspase-9 complex initiates an apoptotic protease cascade. Cell 91, 479–489 (1997).

    Article  CAS  PubMed  Google Scholar 

  21. Du, C., Fang, M., Li, Y., Li, L. & Wang, X. Smac, a mitochondrial protein that promotes cytochrome c-dependent caspase activation by eliminating IAP inhibition. Cell 102, 33–42 (2000).

    Article  CAS  PubMed  Google Scholar 

  22. Susin, S. A. et al. Molecular characterization of mitochondrial apoptosis-inducing factor. Nature 397, 441–446 (1999).

    Article  CAS  PubMed  Google Scholar 

  23. Li, L. Y., Luo, X. & Wang, X. Endonuclease G is an apoptotic DNase when released from mitochondria. Nature 412, 95–99 (2001).

    Article  CAS  PubMed  Google Scholar 

  24. Singh, R., Letai, A. & Sarosiek, K. Regulation of apoptosis in health and disease: the balancing act of BCL-2 family proteins. Nat. Rev. Mol. Cell Biol. 20, 175–193 (2019).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  25. Shimizu, S., Narita, M. & Tsujimoto, Y. Bcl-2 family proteins regulate the release of apoptogenic cytochrome c by the mitochondrial channel VDAC. Nature 399, 483–487 (1999).

    Article  CAS  PubMed  Google Scholar 

  26. Marzo, I. et al. Bax and adenine nucleotide translocator cooperate in the mitochondrial control of apoptosis. Science 281, 2027–2031 (1998).

    Article  CAS  PubMed  Google Scholar 

  27. Luo, X., Budihardjo, I., Zou, H., Slaughter, C. & Wang, X. Bid, a Bcl2 interacting protein, mediates cytochrome c release from mitochondria in response to activation of cell surface death receptors. Cell 94, 481–490 (1998).

    Article  CAS  PubMed  Google Scholar 

  28. Montero, J. & Letai, A. Why do BCL-2 inhibitors work and where should we use them in the clinic? Cell Death Differ. 25, 56–64 (2018).

    Article  CAS  PubMed  Google Scholar 

  29. Campani, D. et al. Bcl-2 expression in pancreas development and pancreatic cancer progression. J. Pathol. 194, 444–450 (2001).

    Article  CAS  PubMed  Google Scholar 

  30. Song, S., Wang, B., Gu, S., Li, X. & Sun, S. Expression of Beclin 1 and Bcl-2 in pancreatic neoplasms and its effect on pancreatic ductal adenocarcinoma prognosis. Oncol. Lett. 14, 7849–7861 (2017).

    PubMed Central  PubMed  Google Scholar 

  31. Miyamoto, Y. et al. Immunohistochemical analysis of Bcl-2, Bax, Bcl-X, and Mcl-1 expression in pancreatic cancers. Oncology 56, 73–82 (1999).

    Article  CAS  PubMed  Google Scholar 

  32. Holler, N. et al. Fas triggers an alternative, caspase-8-independent cell death pathway using the kinase RIP as effector molecule. Nat. Immunol. 1, 489–495 (2000).

    Article  CAS  PubMed  Google Scholar 

  33. Degterev, A. et al. Chemical inhibitor of nonapoptotic cell death with therapeutic potential for ischemic brain injury. Nat. Chem. Biol. 1, 112–119 (2005).

    Article  CAS  PubMed  Google Scholar 

  34. Weinlich, R., Oberst, A., Beere, H. M. & Green, D. R. Necroptosis in development, inflammation and disease. Nat. Rev. Mol. Cell Biol. 18, 127–136 (2017).

    Article  CAS  PubMed  Google Scholar 

  35. Galluzzi, L., Kepp, O., Chan, F. K. & Kroemer, G. Necroptosis: mechanisms and relevance to disease. Annu. Rev. Pathol. 12, 103–130 (2017).

    Article  CAS  PubMed  Google Scholar 

  36. Zhang, D. W. et al. RIP3, an energy metabolism regulator that switches TNF-induced cell death from apoptosis to necrosis. Science 325, 332–336 (2009).

    Article  CAS  PubMed  Google Scholar 

  37. He, S. et al. Receptor interacting protein kinase-3 determines cellular necrotic response to TNF-alpha. Cell 137, 1100–1111 (2009).

    Article  CAS  PubMed  Google Scholar 

  38. Cho, Y. S. et al. Phosphorylation-driven assembly of the RIP1-RIP3 complex regulates programmed necrosis and virus-induced inflammation. Cell 137, 1112–1123 (2009).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  39. Sun, L. et al. Mixed lineage kinase domain-like protein mediates necrosis signaling downstream of RIP3 kinase. Cell 148, 213–227 (2012).

    Article  CAS  PubMed  Google Scholar 

  40. Temkin, V., Huang, Q., Liu, H., Osada, H. & Pope, R. M. Inhibition of ADP/ATP exchange in receptor-interacting protein-mediated necrosis. Mol. Cell Biol. 26, 2215–2225 (2006).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  41. Los, M. et al. Activation and caspase-mediated inhibition of PARP: a molecular switch between fibroblast necrosis and apoptosis in death receptor signaling. Mol. Biol. Cell 13, 978–988 (2002).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  42. Gomes-Filho, S. M. et al. Aurora A kinase and its activator TPX2 are potential therapeutic targets in KRAS-induced pancreatic cancer. Cell Oncol. 43, 445–460 (2020).

    Article  CAS  Google Scholar 

  43. Xie, Y. et al. Inhibition of aurora kinase A induces necroptosis in pancreatic carcinoma. Gastroenterology 153, 1429–1443.e5 (2017).

    Article  CAS  PubMed  Google Scholar 

  44. Wang, J. et al. Phosphorylation-dependent regulation of ALDH1A1 by Aurora kinase A: insights on their synergistic relationship in pancreatic cancer. BMC Biol. 15, 10 (2017).

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  45. Kim, E. et al. Phase I study of the combination of alisertib (MLN8237) and gemcitabine in advanced solid tumors. J. Clin. Oncol. 33, 2526–2526 (2020).

    Article  Google Scholar 

  46. Cookson, B. T. & Brennan, M. A. Pro-inflammatory programmed cell death. Trends Microbiol. 9, 113–114 (2001).

    Article  CAS  PubMed  Google Scholar 

  47. Wang, Y. et al. Chemotherapy drugs induce pyroptosis through caspase-3 cleavage of a gasdermin. Nature 547, 99–103 (2017).

    Article  CAS  PubMed  Google Scholar 

  48. Cui, J. et al. MST1 suppresses pancreatic cancer progression via ROS-induced pyroptosis. Mol. Cancer Res. 17, 1316–1325 (2019).

    Article  CAS  PubMed  Google Scholar 

  49. Kayagaki, N. et al. Caspase-11 cleaves gasdermin D for non-canonical inflammasome signalling. Nature 526, 666–671 (2015).

    Article  CAS  PubMed  Google Scholar 

  50. Rogers, C. et al. Cleavage of DFNA5 by caspase-3 during apoptosis mediates progression to secondary necrotic/pyroptotic cell death. Nat. Commun. 8, 14128 (2017).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  51. Orning, P. et al. Pathogen blockade of TAK1 triggers caspase-8-dependent cleavage of gasdermin D and cell death. Science 362, 1064–1069 (2018).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  52. Taabazuing, C. Y., Okondo, M. C. & Bachovchin, D. A. Pyroptosis and apoptosis pathways engage in bidirectional crosstalk in monocytes and macrophages. Cell Chem. Biol. 24, 507–514.e4 (2017).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  53. Ding, J. et al. Pore-forming activity and structural autoinhibition of the gasdermin family. Nature 535, 111–116 (2016).

    Article  CAS  PubMed  Google Scholar 

  54. Liu, X. et al. Inflammasome-activated gasdermin D causes pyroptosis by forming membrane pores. Nature 535, 153–158 (2016).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  55. Sborgi, L. et al. GSDMD membrane pore formation constitutes the mechanism of pyroptotic cell death. EMBO J. 35, 1766–1778 (2016).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  56. Kang, R. et al. Lipid peroxidation drives gasdermin d-mediated pyroptosis in lethal polymicrobial sepsis. Cell Host Microbe 24, 97–108.e4 (2018).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  57. Riegman, M. et al. Ferroptosis occurs through an osmotic mechanism and propagates independently of cell rupture. Nat. Cell Biol. 22, 1042–1048 (2020).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  58. Dixon, S. J. et al. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell 149, 1060–1072 (2012).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  59. Xie, Y. et al. The tumor suppressor p53 limits ferroptosis by blocking DPP4 activity. Cell Rep. 20, 1692–1704 (2017).

    Article  CAS  PubMed  Google Scholar 

  60. Eling, N., Reuter, L., Hazin, J., Hamacher-Brady, A. & Brady, N. R. Identification of artesunate as a specific activator of ferroptosis in pancreatic cancer cells. Oncoscience 2, 517–532 (2015).

    Article  PubMed Central  PubMed  Google Scholar 

  61. Zhu, S. et al. HSPA5 regulates ferroptotic cell death in cancer cells. Cancer Res. 77, 2064–2077 (2017).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  62. Tang, D. & Kroemer, G. Ferroptosis. Curr. Biol. 30, R1292–R1297 (2020).

    Article  CAS  PubMed  Google Scholar 

  63. Yang, W. S. et al. Regulation of ferroptotic cancer cell death by GPX4. Cell 156, 317–331 (2014).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  64. Zou, Y. et al. Plasticity of ether lipids promotes ferroptosis susceptibility and evasion. Nature 585, 603–608 (2020).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  65. Yuan, H., Li, X., Zhang, X., Kang, R. & Tang, D. Identification of ACSL4 as a biomarker and contributor of ferroptosis. Biochem. Biophys. Res. Commun. 478, 1338–1343 (2016).

    Article  CAS  PubMed  Google Scholar 

  66. Doll, S. et al. ACSL4 dictates ferroptosis sensitivity by shaping cellular lipid composition. Nat. Chem. Biol. 13, 91–98 (2017).

    Article  CAS  PubMed  Google Scholar 

  67. Kagan, V. E. et al. Oxidized arachidonic and adrenic PEs navigate cells to ferroptosis. Nat. Chem. Biol. 13, 81–90 (2017).

    Article  CAS  PubMed  Google Scholar 

  68. Dixon, S. J. et al. Human haploid cell genetics reveals roles for lipid metabolism genes in nonapoptotic cell death. ACS Chem. Biol. 10, 1604–1609 (2015).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  69. Chen, X., Li, J., Kang, R., Klionsky, D. J. & Tang, D. Ferroptosis: machinery and regulation. Autophagy https://doi.org/10.1080/15548627.2020.1810918 (2020).

    Article  PubMed Central  PubMed  Google Scholar 

  70. Hou, W. et al. Autophagy promotes ferroptosis by degradation of ferritin. Autophagy 12, 1425–1428 (2016).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  71. Li, J. et al. Tumor heterogeneity in autophagy-dependent ferroptosis. Autophagy https://doi.org/10.1080/15548627.2021.1872241 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  72. Doll, S. et al. FSP1 is a glutathione-independent ferroptosis suppressor. Nature 575, 693–698 (2019).

    Article  CAS  PubMed  Google Scholar 

  73. Bersuker, K. et al. The CoQ oxidoreductase FSP1 acts parallel to GPX4 to inhibit ferroptosis. Nature 575, 688–692 (2019).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  74. Kraft, V. A. N. et al. GTP cyclohydrolase 1/tetrahydrobiopterin counteract ferroptosis through lipid remodeling. ACS Cent. Sci. 6, 41–53 (2020).

    Article  CAS  PubMed  Google Scholar 

  75. Dai, E., Meng, L., Kang, R., Wang, X. & Tang, D. ESCRT-III-dependent membrane repair blocks ferroptosis. Biochem. Biophys. Res. Commun. 522, 415–421 (2020).

    Article  CAS  PubMed  Google Scholar 

  76. Zhang, Y. et al. Imidazole ketone erastin induces ferroptosis and slows tumor growth in a mouse lymphoma model. Cell Chem. Biol. 26, 623–633.e9 (2019).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  77. Sun, X. et al. Metallothionein-1G facilitates sorafenib resistance through inhibition of ferroptosis. Hepatology 64, 488–500 (2016).

    Article  CAS  PubMed  Google Scholar 

  78. Chen, X., Kang, R., Kroemer, G., & Tang, D. Targeting ferroptosis in pancreatic cancer: a double-edged sword. Trends Cancer https://doi.org/10.1016/j.trecan.2021.04.005 (2021).

    Article  PubMed  Google Scholar 

  79. Song, X. et al. JTC801 induces pH-dependent death specifically in cancer cells and slows growth of tumors in mice. Gastroenterology 154, 1480–1493 (2018).

    Article  CAS  PubMed  Google Scholar 

  80. Furlong, I. J., Ascaso, R., Lopez Rivas, A. & Collins, M. K. Intracellular acidification induces apoptosis by stimulating ICE-like protease activity. J. Cell Sci. 110, 653–661 (1997).

    Article  CAS  PubMed  Google Scholar 

  81. Wang, Y. Z. et al. Tissue acidosis induces neuronal necroptosis via ASIC1a channel independent of its ionic conduction. eLife 4, e05682 (2015).

    Article  PubMed Central  PubMed  Google Scholar 

  82. Nakamura, N., Matsuura, A., Wada, Y. & Ohsumi, Y. Acidification of vacuoles is required for autophagic degradation in the yeast, Saccharomyces cerevisiae. J. Biochem. 121, 338–344 (1997).

    Article  CAS  PubMed  Google Scholar 

  83. Galenkamp, K. M. O. et al. Golgi acidification by NHE7 regulates cytosolic pH homeostasis in pancreatic cancer cells. Cancer Discov. 10, 822–835 (2020).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  84. Zhu, S., Liu, J., Kang, R., Yang, M., & Tang, D. Targeting NF-κB-dependent alkaliptosis for the treatment of venetoclax-resistant acute myeloid leukemia cells. Biochem. Biophys. Res. Commun. 562, 55–61 (2021).

    Article  CAS  Google Scholar 

  85. Zheng, C. J., Yang, L. L., Liu, J. & Zhong, L. JTC-801 exerts anti-proliferative effects in human osteosarcoma cells by inducing apoptosis. J. Recept. Signal. Transduct. Res. 38, 133–140 (2018).

    Article  CAS  PubMed  Google Scholar 

  86. Buscail, L., Bournet, B. & Cordelier, P. Role of oncogenic KRAS in the diagnosis, prognosis and treatment of pancreatic cancer. Nat. Rev. Gastroenterol. Hepatol. 17, 153–168 (2020).

    Article  CAS  PubMed  Google Scholar 

  87. Weissmueller, S. et al. Mutant p53 drives pancreatic cancer metastasis through cell-autonomous PDGF receptor beta signaling. Cell 157, 382–394 (2014).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  88. Escobar-Hoyos, L. F. et al. Altered RNA splicing by mutant p53 activates oncogenic RAS signaling in pancreatic cancer. Cancer Cell 38, 198–211.e8 (2020).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  89. Collisson, E. A., Bailey, P., Chang, D. K. & Biankin, A. V. Molecular subtypes of pancreatic cancer. Nat. Rev. Gastroenterol. Hepatol. 16, 207–220 (2019).

    Article  PubMed  Google Scholar 

  90. Kern, S. E. et al. Oncogenic forms of p53 inhibit p53-regulated gene expression. Science 256, 827–830 (1992).

    Article  CAS  PubMed  Google Scholar 

  91. Miyashita, T. & Reed, J. C. Tumor suppressor p53 is a direct transcriptional activator of the human bax gene. Cell 80, 293–299 (1995).

    Article  CAS  PubMed  Google Scholar 

  92. Nakano, K. & Vousden, K. H. PUMA, a novel proapoptotic gene, is induced by p53. Mol. Cell 7, 683–694 (2001).

    Article  CAS  PubMed  Google Scholar 

  93. Oda, E. et al. Noxa, a BH3-only member of the Bcl-2 family and candidate mediator of p53-induced apoptosis. Science 288, 1053–1058 (2000).

    Article  CAS  PubMed  Google Scholar 

  94. Mihara, M. et al. p53 has a direct apoptogenic role at the mitochondria. Mol. Cell 11, 577–590 (2003).

    Article  CAS  PubMed  Google Scholar 

  95. Fiorini, C. et al. Mutant p53 stimulates chemoresistance of pancreatic adenocarcinoma cells to gemcitabine. Biochim. Biophys. Acta 1853, 89–100 (2015).

    Article  CAS  PubMed  Google Scholar 

  96. Izetti, P. et al. PRIMA-1, a mutant p53 reactivator, induces apoptosis and enhances chemotherapeutic cytotoxicity in pancreatic cancer cell lines. Invest. N. Drugs 32, 783–794 (2014).

    Article  CAS  Google Scholar 

  97. Chu, B. et al. ALOX12 is required for p53-mediated tumour suppression through a distinct ferroptosis pathway. Nat. Cell Biol. 21, 579–591 (2019).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  98. Jiang, L. et al. Ferroptosis as a p53-mediated activity during tumour suppression. Nature 520, 57–62 (2015).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  99. Badgley, M. A. et al. Cysteine depletion induces pancreatic tumor ferroptosis in mice. Science 368, 85–89 (2020).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  100. Jennis, M. et al. An African-specific polymorphism in the TP53 gene impairs p53 tumor suppressor function in a mouse model. Genes Dev. 30, 918–930 (2016).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  101. Ou, Y., Wang, S. J., Li, D. W., Chu, B. & Gu, W. Activation of SAT1 engages polyamine metabolism with p53-mediated ferroptotic responses. Proc. Natl Acad. Sci. USA 113, E6806–E6812 (2016).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  102. Gao, M. H., Monian, P., Quadri, N., Ramasamy, R. & Jiang, X. J. Glutaminolysis and transferrin regulate ferroptosis. Mol. Cell 59, 298–308 (2015).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  103. Tarangelo, A. et al. p53 Suppresses metabolic stress-induced ferroptosis in cancer cells. Cell Rep. 22, 569–575 (2018).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  104. Collisson, E. A. et al. A central role for RAF–>MEK–>ERK signaling in the genesis of pancreatic ductal adenocarcinoma. Cancer Discov. 2, 685–693 (2012).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  105. Bryant, K. L., Mancias, J. D., Kimmelman, A. C. & Der, C. J. KRAS: feeding pancreatic cancer proliferation. Trends Biochem. Sci. 39, 91–100 (2014).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  106. Kang, Y. W. et al. KRAS targeting antibody synergizes anti-cancer activity of gemcitabine against pancreatic cancer. Cancer Lett. 438, 174–186 (2018).

    Article  CAS  PubMed  Google Scholar 

  107. Yang, W. S. & Stockwell, B. R. Synthetic lethal screening identifies compounds activating iron-dependent, nonapoptotic cell death in oncogenic-RAS-harboring cancer cells. Chem. Biol. 15, 234–245 (2008).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  108. Hong, D. S. et al. KRAS(G12C) Inhibition with sotorasib in advanced solid tumors. N. Engl. J. Med. 383, 1207–1217 (2020).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  109. Kamerkar, S. et al. Exosomes facilitate therapeutic targeting of oncogenic KRAS in pancreatic cancer. Nature 546, 498–503 (2017).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  110. Tascilar, M. et al. The SMAD4 protein and prognosis of pancreatic ductal adenocarcinoma. Clin. Cancer Res. 7, 4115–4121 (2001).

    CAS  PubMed  Google Scholar 

  111. Ellenrieder, V. et al. Transforming growth factor beta1 treatment leads to an epithelial-mesenchymal transdifferentiation of pancreatic cancer cells requiring extracellular signal-regulated kinase 2 activation. Cancer Res. 61, 4222–4228 (2001).

    CAS  PubMed  Google Scholar 

  112. Rowland-Goldsmith, M. A., Maruyama, H., Kusama, T., Ralli, S. & Korc, M. Soluble type II transforming growth factor-β (TGF-β) receptor inhibits TGF-β signaling in COLO-357 pancreatic cancer cells in vitro and attenuates tumor formation. Clin. Cancer Res. 7, 2931–2940 (2001).

    CAS  PubMed  Google Scholar 

  113. Porcelli, L. et al. CAFs and TGF-beta signaling activation by mast cells contribute to resistance to gemcitabine/nabpaclitaxel in pancreatic cancer. Cancers 11, 330 (2019).

    Article  CAS  PubMed Central  Google Scholar 

  114. Giehl, K., Seidel, B., Gierschik, P., Adler, G. & Menke, A. TGFbeta1 represses proliferation of pancreatic carcinoma cells which correlates with Smad4-independent inhibition of ERK activation. Oncogene 19, 4531–4541 (2000).

    Article  CAS  PubMed  Google Scholar 

  115. David, C. J. et al. TGF-beta tumor suppression through a lethal EMT. Cell 164, 1015–1030 (2016).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  116. Viswanathan, V. S. et al. Dependency of a therapy-resistant state of cancer cells on a lipid peroxidase pathway. Nature 547, 453–457 (2017).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  117. Wu, J. et al. Intercellular interaction dictates cancer cell ferroptosis via NF2-YAP signalling. Nature 572, 402–406 (2019).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  118. Koong, A. C. et al. Pancreatic tumors show high levels of hypoxia. Int. J. Radiat. Oncol. Biol. Phys. 48, 919–922 (2000).

    Article  CAS  PubMed  Google Scholar 

  119. Carmeliet, P. et al. Role of HIF-1α in hypoxia-mediated apoptosis, cell proliferation and tumour angiogenesis. Nature 394, 485–490 (1998).

    Article  CAS  PubMed  Google Scholar 

  120. Bruick, R. K. Expression of the gene encoding the proapoptotic Nip3 protein is induced by hypoxia. Proc. Natl Acad. Sci. USA 97, 9082–9087 (2000).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  121. Sowter, H. M., Ratcliffe, P. J., Watson, P., Greenberg, A. H. & Harris, A. L. HIF-1-dependent regulation of hypoxic induction of the cell death factors BNIP3 and NIX in human tumors. Cancer Res. 61, 6669–6673 (2001).

    CAS  PubMed  Google Scholar 

  122. Giaccia, A., Siim, B. G. & Johnson, R. S. HIF-1 as a target for drug development. Nat. Rev. Drug Discov. 2, 803–811 (2003).

    Article  CAS  PubMed  Google Scholar 

  123. Akakura, N. et al. Constitutive expression of hypoxia-inducible factor-1alpha renders pancreatic cancer cells resistant to apoptosis induced by hypoxia and nutrient deprivation. Cancer Res. 61, 6548–6554 (2001).

    CAS  PubMed  Google Scholar 

  124. Chen, J. et al. Dominant-negative hypoxia-inducible factor-1 alpha reduces tumorigenicity of pancreatic cancer cells through the suppression of glucose metabolism. Am. J. Pathol. 162, 1283–1291 (2003).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  125. An, W. G. et al. Stabilization of wild-type p53 by hypoxia-inducible factor 1alpha. Nature 392, 405–408 (1998).

    Article  CAS  PubMed  Google Scholar 

  126. Okami, J., Simeone, D. M. & Logsdon, C. D. Silencing of the hypoxia-inducible cell death protein BNIP3 in pancreatic cancer. Cancer Res. 64, 5338–5346 (2004).

    Article  CAS  PubMed  Google Scholar 

  127. Abe, T. et al. Upregulation of BNIP3 by 5-aza-2’-deoxycytidine sensitizes pancreatic cancer cells to hypoxia-mediated cell death. J. Gastroenterol. 40, 504–510 (2005).

    Article  CAS  PubMed  Google Scholar 

  128. Li, C. et al. PINK1 and PARK2 suppress pancreatic tumorigenesis through control of mitochondrial iron-mediated immunometabolism. Dev. Cell 46, 441–455.e8 (2018).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  129. Humpton, T. J. et al. Oncogenic KRAS induces NIX-mediated mitophagy to promote pancreatic cancer. Cancer Discov. 9, 1268–1287 (2019).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  130. Yang, M. et al. Clockophagy is a novel selective autophagy process favoring ferroptosis. Sci. Adv. 5, eaaw2238 (2019).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  131. McKillop, I. H., Girardi, C. A. & Thompson, K. J. Role of fatty acid binding proteins (FABPs) in cancer development and progression. Cell Signal. 62, 109336 (2019).

    Article  CAS  PubMed  Google Scholar 

  132. Sinha, P. et al. Increased expression of epidermal fatty acid binding protein, cofilin, and 14-3-3-sigma (stratifin) detected by two-dimensional gel electrophoresis, mass spectrometry and microsequencing of drug-resistant human adenocarcinoma of the pancreas. Electrophoresis 20, 2952–2960 (1999).

    Article  CAS  PubMed  Google Scholar 

  133. Zou, Y. et al. A GPX4-dependent cancer cell state underlies the clear-cell morphology and confers sensitivity to ferroptosis. Nat. Commun. 10, 1617 (2019).

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  134. Lee, K. E. et al. Hif1a deletion reveals pro-neoplastic function of B cells in pancreatic neoplasia. Cancer Discov. 6, 256–269 (2016).

    Article  CAS  PubMed  Google Scholar 

  135. Criscimanna, A. et al. PanIN-specific regulation of Wnt signaling by HIF2alpha during early pancreatic tumorigenesis. Cancer Res. 73, 4781–4790 (2013).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  136. Zhao, T. et al. Inhibition of HIF-1α by PX-478 enhances the anti-tumor effect of gemcitabine by inducing immunogenic cell death in pancreatic ductal adenocarcinoma. Oncotarget 6, 2250–2262 (2015).

    Article  PubMed  Google Scholar 

  137. Schwartz, D. L. et al. Radiosensitization and stromal imaging response correlates for the HIF-1 inhibitor PX-478 given with or without chemotherapy in pancreatic cancer. Mol. Cancer Ther. 9, 2057–2067 (2010).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  138. Qin, C. et al. Metabolism of pancreatic cancer: paving the way to better anticancer strategies. Mol. Cancer 19, 50 (2020).

    Article  PubMed Central  PubMed  Google Scholar 

  139. Lim, J. K. M. et al. Cystine/glutamate antiporter xCT (SLC7A11) facilitates oncogenic RAS transformation by preserving intracellular redox balance. Proc. Natl Acad. Sci. USA 116, 9433–9442 (2019).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  140. Hong, Y. B. et al. Nuclear factor (erythroid-derived 2)-like 2 regulates drug resistance in pancreatic cancer cells. Pancreas 39, 463–472 (2010).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  141. Daher, B. et al. Genetic ablation of the cystine transporter xCT in PDAC cells inhibits mTORC1, growth, survival, and tumor formation via nutrient and oxidative stresses. Cancer Res. 79, 3877–3890 (2019).

    Article  CAS  PubMed  Google Scholar 

  142. Wang, L. et al. A pharmacological probe identifies cystathionine beta-synthase as a new negative regulator for ferroptosis. Cell Death Dis. 9, 1005 (2018).

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  143. Hayano, M., Yang, W. S., Corn, C. K., Pagano, N. C. & Stockwell, B. R. Loss of cysteinyl-tRNA synthetase (CARS) induces the transsulfuration pathway and inhibits ferroptosis induced by cystine deprivation. Cell Death Differ. 23, 270–278 (2016).

    Article  CAS  PubMed  Google Scholar 

  144. Yang, B. C. & Leung, P. S. Irisin is a positive regulator for ferroptosis in pancreatic cancer. Mol. Ther. Oncolytics 18, 457–466 (2020).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  145. Anandhan, A., Dodson, M., Schmidlin, C. J., Liu, P. & Zhang, D. D. Breakdown of an ironclad defense system: the critical role of NRF2 in mediating ferroptosis. Cell Chem. Biol. 27, 436–447 (2020).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  146. DeNicola, G. M. et al. Oncogene-induced Nrf2 transcription promotes ROS detoxification and tumorigenesis. Nature 475, 106–109 (2011).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  147. Chio, I. I. C. et al. NRF2 Promotes tumor maintenance by modulating mRNA translation in pancreatic cancer. Cell 166, 963–976 (2016).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  148. Lister, A. et al. Nrf2 is overexpressed in pancreatic cancer: implications for cell proliferation and therapy. Mol. Cancer 10, 37 (2011).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  149. Son, J. et al. Glutamine supports pancreatic cancer growth through a KRAS-regulated metabolic pathway. Nature 496, 101–105 (2013).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  150. Bott, A. J. et al. Glutamine anabolism plays a critical role in pancreatic cancer by coupling carbon and nitrogen metabolism. Cell Rep. 29, 1287–1298.e6 (2019).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  151. Chen, R. et al. Disrupting glutamine metabolic pathways to sensitize gemcitabine-resistant pancreatic cancer. Sci. Rep. 7, 7950 (2017).

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  152. Mukhopadhyay, S. et al. Undermining glutaminolysis bolsters chemotherapy while NRF2 promotes chemoresistance in KRAS-driven pancreatic cancers. Cancer Res. 80, 1630–1643 (2020).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  153. Schnelldorfer, T. et al. Glutathione depletion causes cell growth inhibition and enhanced apoptosis in pancreatic cancer cells. Cancer 89, 1440–1447 (2000).

    Article  CAS  PubMed  Google Scholar 

  154. Wang, K. et al. Branched-chain amino acid aminotransferase 2 regulates ferroptotic cell death in cancer cells. Cell Death Differ. 28, 1222–1236 (2021).

    Article  CAS  PubMed  Google Scholar 

  155. Shukla, S. K. et al. MUC1 and HIF-1alpha signaling crosstalk induces anabolic glucose metabolism to impart gemcitabine resistance to pancreatic cancer. Cancer Cell 32, 392 (2017).

    Article  CAS  PubMed  Google Scholar 

  156. Lee, H. et al. Energy-stress-mediated AMPK activation inhibits ferroptosis. Nat. Cell Biol. 22, 225–234 (2020).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  157. Song, X. et al. AMPK-mediated BECN1 phosphorylation promotes ferroptosis by directly blocking system Xc- activity. Curr. Biol. 28, 2388–2399.e5 (2018).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  158. Hu, M. et al. AMPK inhibition suppresses the malignant phenotype of pancreatic cancer cells in part by attenuating aerobic glycolysis. J. Cancer 10, 1870–1878 (2019).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  159. Liu, J., Song, N., Huang, Y. & Chen, Y. Irisin inhibits pancreatic cancer cell growth via the AMPK-mTOR pathway. Sci. Rep. 8, 15247 (2018).

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  160. Li, J. et al. Regulation and function of autophagy in pancreatic cancer. Autophagy https://doi.org/10.1080/15548627.2020.1847462 (2020).

    Article  PubMed Central  PubMed  Google Scholar 

  161. Piffoux, M., Eriau, E. & Cassier, P. A. Autophagy as a therapeutic target in pancreatic cancer. Br. J. Cancer 124, 333–344 (2021).

    Article  PubMed  Google Scholar 

  162. Levine, B. & Kroemer, G. Biological functions of autophagy genes: a disease perspective. Cell 176, 11–42 (2019).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  163. Kroemer, G., Marino, G. & Levine, B. Autophagy and the integrated stress response. Mol. Cell 40, 280–293 (2010).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  164. Bai, Y. et al. Lipid storage and lipophagy regulates ferroptosis. Biochem. Biophys. Res. Commun. 508, 997–1003 (2019).

    Article  CAS  PubMed  Google Scholar 

  165. Kinsey, C. G. et al. Protective autophagy elicited by RAF–>MEK–>ERK inhibition suggests a treatment strategy for RAS-driven cancers. Nat. Med. 25, 620–627 (2019).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  166. Liu, J. et al. Autophagy-dependent ferroptosis: machinery and regulation. Cell Chem. Biol. 27, 420–435 (2020).

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  167. Wu, Z. et al. Chaperone-mediated autophagy is involved in the execution of ferroptosis. Proc. Natl Acad. Sci. USA 116, 2996–3005 (2019).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  168. Mancias, J. D., Wang, X., Gygi, S. P., Harper, J. W. & Kimmelman, A. C. Quantitative proteomics identifies NCOA4 as the cargo receptor mediating ferritinophagy. Nature 509, 105–109 (2014).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  169. Xie, Y., Kuang, F., Liu, J., Tang, D. & Kang, R. DUSP1 blocks autophagy-dependent ferroptosis in pancreatic cancer. J. Pancreatol. 3, 154–160 (2020).

    Article  Google Scholar 

  170. Li, C. et al. Mitochondrial DNA stress triggers autophagy-dependent ferroptotic death. Autophagy 17, 948–960 (2021).

    Article  CAS  PubMed  Google Scholar 

  171. Liu, Y., Wang, Y., Liu, J., Kang, R. & Tang, D. Interplay between MTOR and GPX4 signaling modulates autophagy-dependent ferroptotic cancer cell death. Cancer Gene Ther. 28, 55–63 (2021).

    Article  CAS  PubMed  Google Scholar 

  172. Gao, H. et al. Ferroptosis is a lysosomal cell death process. Biochem. Biophys. Res. Commun. 503, 1550–1556 (2018).

    Article  CAS  PubMed  Google Scholar 

  173. Yamamoto, K. et al. Autophagy promotes immune evasion of pancreatic cancer by degrading MHC-I. Nature 581, 100–105 (2020).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  174. Zeh, H. J. et al. A randomized phase II preoperative study of autophagy inhibition with high-dose hydroxychloroquine and gemcitabine/Nab-paclitaxel in pancreatic cancer patients. Clin. Cancer Res. 26, 3126–3134 (2020).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  175. Aghdassi, A. et al. Heat shock protein 70 increases tumorigenicity and inhibits apoptosis in pancreatic adenocarcinoma. Cancer Res. 67, 616–625 (2007).

    Article  CAS  PubMed  Google Scholar 

  176. Mori-Iwamoto, S. et al. Proteomics finding heat shock protein 27 as a biomarker for resistance of pancreatic cancer cells to gemcitabine. Int. J. Oncol. 31, 1345–1350 (2007).

    CAS  PubMed  Google Scholar 

  177. Ghadban, T. et al. HSP90 is a promising target in gemcitabine and 5-fluorouracil resistant pancreatic cancer. Apoptosis 22, 369–380 (2017).

    Article  CAS  PubMed  Google Scholar 

  178. Srinivasan, S. R. et al. Heat shock protein 70 (Hsp70) suppresses RIP1-dependent apoptotic and necroptotic cascades. Mol. Cancer Res. 16, 58–68 (2018).

    Article  CAS  PubMed  Google Scholar 

  179. Weinberg, F. et al. Mitochondrial metabolism and ROS generation are essential for Kras-mediated tumorigenicity. Proc. Natl Acad. Sci. USA 107, 8788–8793 (2010).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  180. Storz, P. KRas, ROS and the initiation of pancreatic cancer. Small GTPases 8, 38–42 (2017).

    Article  CAS  PubMed  Google Scholar 

  181. Hingorani, S. R. et al. Preinvasive and invasive ductal pancreatic cancer and its early detection in the mouse. Cancer Cell 4, 437–450 (2003).

    Article  CAS  PubMed  Google Scholar 

  182. Leinwand, J. & Miller, G. Regulation and modulation of antitumor immunity in pancreatic cancer. Nat. Immunol. 21, 1152–1159 (2020).

    Article  CAS  PubMed  Google Scholar 

  183. Lowenfels, A. B. et al. Pancreatitis and the risk of pancreatic cancer. International Pancreatitis Study Group. N. Engl. J. Med. 328, 1433–1437 (1993).

    Article  CAS  PubMed  Google Scholar 

  184. Carriere, C., Young, A. L., Gunn, J. R., Longnecker, D. S. & Korc, M. Acute pancreatitis markedly accelerates pancreatic cancer progression in mice expressing oncogenic Kras. Biochem. Biophys. Res. Commun. 382, 561–565 (2009).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  185. Philip, B. et al. A high-fat diet activates oncogenic Kras and COX2 to induce development of pancreatic ductal adenocarcinoma in mice. Gastroenterology 145, 1449–1458 (2013).

    Article  CAS  PubMed  Google Scholar 

  186. Guerra, C. et al. Chronic pancreatitis is essential for induction of pancreatic ductal adenocarcinoma by K-Ras oncogenes in adult mice. Cancer Cell 11, 291–302 (2007).

    Article  CAS  PubMed  Google Scholar 

  187. Pushalkar, S. et al. The pancreatic cancer microbiome promotes oncogenesis by induction of innate and adaptive immune suppression. Cancer Discov. 8, 403–416 (2018).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  188. Riquelme, E. et al. Tumor microbiome diversity and composition influence pancreatic cancer outcomes. Cell 178, 795–806.e12 (2019).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  189. Aykut, B. et al. The fungal mycobiome promotes pancreatic oncogenesis via activation of MBL. Nature 574, 264–267 (2019).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  190. Nejman, D. et al. The human tumor microbiome is composed of tumor type-specific intracellular bacteria. Science 368, 973–980 (2020).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  191. Sendler, M. et al. NLRP3 inflammasome regulates development of systemic inflammatory response and compensatory anti-inflammatory response syndromes in mice with acute pancreatitis. Gastroenterology 158, 253–269.e14 (2020).

    Article  CAS  PubMed  Google Scholar 

  192. Liu, Y., Wang, Y., Liu, J., Kang, R. & Tang, D. The circadian clock protects against ferroptosis-induced sterile inflammation. Biochem. Biophys. Res. Commun. 525, 620–625 (2020).

    Article  CAS  PubMed  Google Scholar 

  193. Seifert, L. et al. The necrosome promotes pancreatic oncogenesis via CXCL1 and Mincle-induced immune suppression. Nature 532, 245–249 (2016).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  194. Ando, Y. et al. Necroptosis in pancreatic cancer promotes cancer cell migration and invasion by release of CXCL5. PLoS One 15, e0228015 (2020).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  195. Wang, W. et al. RIP1 kinase drives macrophage-mediated adaptive immune tolerance in pancreatic cancer. Cancer Cell 34, 757–774.e7 (2018).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  196. Colbert, L. E. et al. Pronecrotic mixed lineage kinase domain-like protein expression is a prognostic biomarker in patients with early-stage resected pancreatic adenocarcinoma. Cancer 119, 3148–3155 (2013).

    Article  CAS  PubMed  Google Scholar 

  197. Dai, E. et al. Ferroptotic damage promotes pancreatic tumorigenesis through a TMEM173/STING-dependent DNA sensor pathway. Nat. Commun. 11, 6339 (2020).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  198. Fang, X. et al. The HMGB1-AGER-STING1 pathway mediates the sterile inflammatory response to alkaliptosis. Biochem. Biophys. Res. Commun. 560, 165–171 (2021).

    Article  CAS  PubMed  Google Scholar 

  199. Dai, E. et al. Autophagy-dependent ferroptosis drives tumor-associated macrophage polarization via release and uptake of oncogenic KRAS protein. Autophagy 16, 2069–2083 (2020).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  200. Das, S., Shapiro, B., Vucic, E. A., Vogt, S. & Bar-Sagi, D. Tumor cell-derived IL1beta promotes desmoplasia and immune suppression in pancreatic cancer. Cancer Res. 80, 1088–1101 (2020).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  201. Daley, D. et al. NLRP3 signaling drives macrophage-induced adaptive immune suppression in pancreatic carcinoma. J. Exp. Med. 214, 1711–1724 (2017).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  202. Chen, X., Yu, C., Kang, R., Kroemer, G. & Tang, D. Cellular degradation systems in ferroptosis. Cell Death Differ. 28, 1135–1148 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  203. Rosenfeldt, M. T. et al. p53 status determines the role of autophagy in pancreatic tumour development. Nature 504, 296–300 (2013).

    Article  CAS  PubMed  Google Scholar 

  204. Efimova, I. et al. Vaccination with early ferroptotic cancer cells induces efficient antitumor immunity. J. Immunother. Cancer 8, e001369 (2020).

    Article  PubMed Central  PubMed  Google Scholar 

  205. Ansari, D., Gustafsson, A. & Andersson, R. Update on the management of pancreatic cancer: surgery is not enough. World J. Gastroenterol. 21, 3157–3165 (2015).

    Article  PubMed Central  PubMed  Google Scholar 

  206. Plunkett, W. et al. Gemcitabine: metabolism, mechanisms of action, and self-potentiation. Semin. Oncol. 22, 3–10 (1995).

    CAS  PubMed  Google Scholar 

  207. Conroy, T. et al. FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N. Engl. J. Med. 364, 1817–1825 (2011).

    Article  CAS  PubMed  Google Scholar 

  208. Von Hoff, D. D. et al. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N. Engl. J. Med. 369, 1691–1703 (2013).

    Article  CAS  Google Scholar 

  209. Modi, S., Kir, D., Banerjee, S. & Saluja, A. Control of apoptosis in treatment and biology of pancreatic cancer. J. Cell Biochem. 117, 279–288 (2016).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  210. Geller, L. T. et al. Potential role of intratumor bacteria in mediating tumor resistance to the chemotherapeutic drug gemcitabine. Science 357, 1156–1160 (2017).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  211. Philippou, Y., Sjoberg, H., Lamb, A. D., Camilleri, P. & Bryant, R. J. Harnessing the potential of multimodal radiotherapy in prostate cancer. Nat. Rev. Urol. 17, 321–338 (2020).

    Article  PubMed  Google Scholar 

  212. Lang, X. et al. Radiotherapy and immunotherapy promote tumoral lipid oxidation and ferroptosis via synergistic repression of SLC7A11. Cancer Discov. 9, 1673–1685 (2019).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  213. Cao, J. Y. et al. A genome-wide haploid genetic screen identifies regulators of glutathione abundance and ferroptosis sensitivity. Cell Rep. 26, 1544–1556.e8 (2019).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  214. Xiao, J. et al. Radiation causes tissue damage by dysregulating inflammasome-gasdermin D signaling in both host and transplanted cells. PLoS Biol. 18, e3000807 (2020).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  215. Chen, X. et al. Toll-like receptor 2 and Toll-like receptor 4 exhibit distinct regulation of cancer cell stemness mediated by cell death-induced high-mobility group Box 1. EBioMedicine 40, 135–150 (2019).

    Article  PubMed Central  PubMed  Google Scholar 

  216. Golan, T. et al. Maintenance olaparib for germline BRCA-mutated metastatic pancreatic cancer. N. Engl. J. Med. 381, 317–327 (2019).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  217. Iqbal, J. et al. The incidence of pancreatic cancer in BRCA1 and BRCA2 mutation carriers. Br. J. Cancer 107, 2005–2009 (2012).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  218. Barbie, D. A. et al. Systematic RNA interference reveals that oncogenic KRAS-driven cancers require TBK1. Nature 462, 108–112 (2009).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  219. Scholl, C. et al. Synthetic lethal interaction between oncogenic KRAS dependency and STK33 suppression in human cancer cells. Cell 137, 821–834 (2009).

    Article  CAS  PubMed  Google Scholar 

  220. Steckel, M. et al. Determination of synthetic lethal interactions in KRAS oncogene-dependent cancer cells reveals novel therapeutic targeting strategies. Cell Res. 22, 1227–1245 (2012).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  221. Yamaguchi, Y., Kasukabe, T. & Kumakura, S. Piperlongumine rapidly induces the death of human pancreatic cancer cells mainly through the induction of ferroptosis. Int. J. Oncol. 52, 1011–1022 (2018).

    CAS  PubMed  Google Scholar 

  222. Chen, X., Kang, R., Kroemer, G. & Tang, D. Ferroptosis in infection, inflammation, and immunity. J. Exp. Med. 218, e20210518 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  223. Hu, N. et al. Pirin is a nuclear redox-sensitive modulator of autophagy-dependent ferroptosis. Biochem. Biophys. Res. Commun. 536, 100–106 (2021).

    Article  CAS  PubMed  Google Scholar 

  224. Liu, L. et al. NUPR1 is a critical repressor of ferroptosis. Nat. Commun. 12, 647 (2021).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  225. Kuang, F., Liu, J., Xie, Y., Tang, D. & Kang, R. MGST1 is a redox-sensitive repressor of ferroptosis in pancreatic cancer cells. Cell Chem. Biol. 28, 765–775.e5 (2021).

    Article  CAS  PubMed  Google Scholar 

  226. Akimoto, M., Maruyama, R., Kawabata, Y., Tajima, Y. & Takenaga, K. Antidiabetic adiponectin receptor agonist AdipoRon suppresses tumour growth of pancreatic cancer by inducing RIPK1/ERK-dependent necroptosis. Cell Death Dis. 9, 804 (2018).

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  227. Santofimia-Castano, P. et al. Ligand-based design identifies a potent NUPR1 inhibitor exerting anticancer activity via necroptosis. J. Clin. Invest. 129, 2500–2513 (2019).

    Article  PubMed Central  PubMed  Google Scholar 

  228. Galluzzi, L., Humeau, J., Buque, A., Zitvogel, L. & Kroemer, G. Immunostimulation with chemotherapy in the era of immune checkpoint inhibitors. Nat. Rev. Clin. Oncol. 17, 725–741 (2020).

    Article  PubMed  Google Scholar 

  229. Torphy, R. J., Zhu, Y. & Schulick, R. D. Immunotherapy for pancreatic cancer: barriers and breakthroughs. Ann. Gastroenterol. Surg. 2, 274–281 (2018).

    Article  PubMed Central  PubMed  Google Scholar 

  230. O’Donnell, J. S., Teng, M. W. L. & Smyth, M. J. Cancer immunoediting and resistance to T cell-based immunotherapy. Nat. Rev. Clin. Oncol. 16, 151–167 (2019).

    Article  CAS  PubMed  Google Scholar 

  231. Clark, C. E. et al. Dynamics of the immune reaction to pancreatic cancer from inception to invasion. Cancer Res. 67, 9518–9527 (2007).

    Article  CAS  PubMed  Google Scholar 

  232. Bear, A. S., Vonderheide, R. H. & O’Hara, M. H. Challenges and opportunities for pancreatic cancer immunotherapy. Cancer Cell 38, 788–802 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  233. Moral, J. A. et al. ILC2s amplify PD-1 blockade by activating tissue-specific cancer immunity. Nature 579, 130–135 (2020).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  234. Huang, J. et al. CDK1/2/5 inhibition overcomes IFNG-mediated adaptive immune resistance in pancreatic cancer. Gut 70, 890–899 (2021).

    Article  CAS  PubMed  Google Scholar 

  235. Galluzzi, L. et al. Consensus guidelines for the definition, detection and interpretation of immunogenic cell death. J. Immunother. Cancer 8, e000337 (2020).

    Article  PubMed Central  PubMed  Google Scholar 

  236. Angelova, A. L. et al. Complementary induction of immunogenic cell death by oncolytic parvovirus H-1PV and gemcitabine in pancreatic cancer. J. Virol. 88, 5263–5276 (2014).

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  237. Lu, J. et al. Nano-enabled pancreas cancer immunotherapy using immunogenic cell death and reversing immunosuppression. Nat. Commun. 8, 1811 (2017).

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  238. Duewell, P. et al. RIG-I-like helicases induce immunogenic cell death of pancreatic cancer cells and sensitize tumors toward killing by CD8+ T cells. Cell Death Differ. 21, 1825–1837 (2014).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  239. Ye, J. et al. Assessing the magnitude of immunogenic cell death following chemotherapy and irradiation reveals a new strategy to treat pancreatic cancer. Cancer Immunol. Res. 8, 94–107 (2020).

    Article  CAS  PubMed  Google Scholar 

  240. Wang, W. et al. CD8+ T cells regulate tumour ferroptosis during cancer immunotherapy. Nature 569, 270–274 (2019).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  241. Wang, Q. et al. A bioorthogonal system reveals antitumour immune function of pyroptosis. Nature 579, 421–426 (2020).

    Article  CAS  PubMed  Google Scholar 

  242. Zhang, Z. et al. Gasdermin E suppresses tumour growth by activating anti-tumour immunity. Nature 579, 415–420 (2020).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  243. Yang, M. et al. TFAM is a novel mediator of immunogenic cancer cell death. Oncoimmunology 7, e1431086 (2018).

    Article  PubMed Central  PubMed  Google Scholar 

  244. Newton, K. et al. Activity of protein kinase RIPK3 determines whether cells die by necroptosis or apoptosis. Science 343, 1357–1360 (2014).

    Article  CAS  PubMed  Google Scholar 

  245. Buchbinder, J. H., Pischel, D., Sundmacher, K., Flassig, R. J. & Lavrik, I. N. Quantitative single cell analysis uncovers the life/death decision in CD95 network. PLoS Comput. Biol. 14, e1006368 (2018).

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  246. Krysko, D. V. et al. Immunogenic cell death and DAMPs in cancer therapy. Nat. Rev. Cancer 12, 860–875 (2012).

    Article  CAS  PubMed  Google Scholar 

  247. Galluzzi, L., Buque, A., Kepp, O., Zitvogel, L. & Kroemer, G. Immunogenic cell death in cancer and infectious disease. Nat. Rev. Immunol. 17, 97–111 (2017).

    Article  CAS  PubMed  Google Scholar 

  248. Hou, W. et al. Strange attractors: DAMPs and autophagy link tumor cell death and immunity. Cell Death Dis. 4, e966 (2013).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  249. Dey, P. et al. Oncogenic KRAS-driven metabolic reprogramming in pancreatic cancer cells utilizes cytokines from the tumor microenvironment. Cancer Discov. 10, 608–625 (2020).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  250. Markosyan, N. et al. Tumor cell-intrinsic EPHA2 suppresses anti-tumor immunity by regulating PTGS2 (COX-2). J. Clin. Invest. 129, 3594–3609 (2019).

    Article  PubMed Central  PubMed  Google Scholar 

  251. Zhang, Y. et al. Regulatory T-cell depletion alters the tumor microenvironment and accelerates pancreatic carcinogenesis. Cancer Discov. 10, 422–439 (2020).

    Article  PubMed Central  PubMed  Google Scholar 

  252. Daley, D. et al. T cells support pancreatic oncogenesis by restraining αβ T cell activation. Cell 166, 1485–1499.e15 (2016).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  253. Bayne, L. J. et al. Tumor-derived granulocyte-macrophage colony-stimulating factor regulates myeloid inflammation and T cell immunity in pancreatic cancer. Cancer Cell 21, 822–835 (2012).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  254. Daley, D. et al. Dectin 1 activation on macrophages by galectin 9 promotes pancreatic carcinoma and peritumoral immune tolerance. Nat. Med. 23, 556–567 (2017).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  255. Hegde, S. et al. Dendritic cell paucity leads to dysfunctional immune surveillance in pancreatic cancer. Cancer Cell 37, 289–307.e9 (2020).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  256. Beatty, G. L. et al. CD40 agonists alter tumor stroma and show efficacy against pancreatic carcinoma in mice and humans. Science 331, 1612–1616 (2011).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  257. Gunderson, A. J. et al. Bruton tyrosine kinase-dependent immune cell cross-talk drives pancreas cancer. Cancer Discov. 6, 270–285 (2016).

    Article  CAS  PubMed  Google Scholar 

  258. Tape, C. J. et al. Oncogenic KRAS regulates tumor cell signaling via stromal reciprocation. Cell 165, 910–920 (2016).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  259. Lesina, M. et al. Stat3/Socs3 activation by IL-6 transsignaling promotes progression of pancreatic intraepithelial neoplasia and development of pancreatic cancer. Cancer Cell 19, 456–469 (2011).

    Article  CAS  PubMed  Google Scholar 

  260. Chen, X., Comish, P., Tang, D. & Kang, R. Characteristics and biomarkers of ferroptosis. Front. Cell Dev. Biol. 9, 637162 (2021).

    Article  PubMed Central  PubMed  Google Scholar 

  261. Tang, D., Kang, R., Coyne, C. B., Zeh, H. J. & Lotze, M. T. PAMPs and DAMPs: signal 0s that spur autophagy and immunity. Immunol. Rev. 249, 158–175 (2012).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  262. Casares, N. et al. Caspase-dependent immunogenicity of doxorubicin-induced tumor cell death. J. Exp. Med. 202, 1691–1701 (2005).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  263. Michaud, M. et al. Autophagy-dependent anticancer immune responses induced by chemotherapeutic agents in mice. Science 334, 1573–1577 (2011).

    Article  CAS  PubMed  Google Scholar 

  264. Vacchelli, E. et al. Chemotherapy-induced antitumor immunity requires formyl peptide receptor 1. Science 350, 972–978 (2015).

    Article  CAS  PubMed  Google Scholar 

  265. Apetoh, L. et al. Toll-like receptor 4-dependent contribution of the immune system to anticancer chemotherapy and radiotherapy. Nat. Med. 13, 1050–1059 (2007).

    Article  CAS  PubMed  Google Scholar 

  266. Garg, A. D. et al. Pathogen response-like recruitment and activation of neutrophils by sterile immunogenic dying cells drives neutrophil-mediated residual cell killing. Cell Death Differ. 24, 832–843 (2017).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  267. Chiba, S. et al. Tumor-infiltrating DCs suppress nucleic acid-mediated innate immune responses through interactions between the receptor TIM-3 and the alarmin HMGB1. Nat. Immunol. 13, 832–842 (2012).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  268. Obeid, M. et al. Calreticulin exposure dictates the immunogenicity of cancer cell death. Nat. Med. 13, 54–61 (2007).

    Article  CAS  PubMed  Google Scholar 

  269. Panaretakis, T. et al. The co-translocation of ERp57 and calreticulin determines the immunogenicity of cell death. Cell Death Differ. 15, 1499–1509 (2008).

    Article  CAS  PubMed  Google Scholar 

  270. Fucikova, J. et al. Human tumor cells killed by anthracyclines induce a tumor-specific immune response. Cancer Res. 71, 4821–4833 (2011).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank Dave Primm (Department of Surgery, University of Texas Southwestern Medical Center) for his critical reading of the manuscript. G.K. is supported by the Ligue Contre le Cancer (Equipe Labellisée); Agence National de la Recherche (ANR) – Projets blancs; ANR under the frame of E-Rare-2, the ERA-Net for Research on Rare Diseases; Association pour la recherche sur le cancer; Cancéropôle Ile-de-France; Chancelerie des Universités de Paris (Legs Poix), Fondation pour la Recherche Médicale; a donation by Elior; European Research Area Network on Cardiovascular Diseases (ERA-CVD, MINOTAUR); Gustave Roussy Odyssea, the European Union Horizon 2020 Project Oncobiome; Fondation Carrefour; High-end Foreign Expert Program in China (GDW20171100085 and GDW20181100051), Institut National du Cancer; Inserm (HTE); Institut Universitaire de France; LeDucq Foundation; the LabEx Immuno-Oncology; the RHU Torino Lumière; the Seerave Foundation; the SIRIC Stratified Oncology Cell DNA Repair and Tumour Immune Elimination (SOCRATE); and the SIRIC Cancer Research and Personalized Medicine Program (CARPEM).

Author information

Authors and Affiliations

Authors

Contributions

All authors wrote the manuscript. G.K., D.T. and R.K. edited, reviewed and approved the manuscript before submission.

Corresponding authors

Correspondence to Rui Kang, Guido Kroemer or Daolin Tang.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information

Nature Reviews Gastroenterology & Hepatology thanks H. Jacob, who co-reviewed with A. Saluja; L. Buscail; and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Related links

US NIH ClinicalTrials.gov database: https://www.clinicaltrials.gov

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chen, X., Zeh, H.J., Kang, R. et al. Cell death in pancreatic cancer: from pathogenesis to therapy. Nat Rev Gastroenterol Hepatol 18, 804–823 (2021). https://doi.org/10.1038/s41575-021-00486-6

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41575-021-00486-6

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing