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:

Targeting ferroptosis as a vulnerability in cancer

Abstract

Ferroptosis is an iron-dependent form of regulated cell death that is triggered by the toxic build-up of lipid peroxides on cellular membranes. In recent years, ferroptosis has garnered enormous interest in cancer research communities, partly because it is a unique cell death modality that is mechanistically and morphologically different from other forms of cell death, such as apoptosis, and therefore holds great potential for cancer therapy. In this Review, we summarize the current understanding of ferroptosis-inducing and ferroptosis defence mechanisms, dissect the roles and mechanisms of ferroptosis in tumour suppression and tumour immunity, conceptualize the diverse vulnerabilities of cancer cells to ferroptosis, and explore therapeutic strategies for targeting ferroptosis in cancer.

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: Ferroptosis-driving and ferroptosis defence mechanisms.
Fig. 2: Ferroptosis as a vulnerability in cancer.
Fig. 3: Role of ferroptosis in antitumour immunity.

Similar content being viewed by others

References

  1. Dixon, S. J. et al. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell 149, 1060–1072 (2012). The landmark publication that introduces the concept of ferroptosis.

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Stockwell, B. R. et al. Ferroptosis: a regulated cell death nexus linking metabolism, redox biology, and disease. Cell 171, 273–285 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Jiang, X., Stockwell, B. R. & Conrad, M. Ferroptosis: mechanisms, biology and role in disease. Nat. Rev. Mol. Cell Biol. 22, 266–282 (2021).

    PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Doll, S. et al. FSP1 is a glutathione-independent ferroptosis suppressor. Nature 575, 693–698 (2019). Together with ref.5, this study identifies the first GPX4-independent mechanism in ferroptosis defence.

    CAS  PubMed  Google Scholar 

  7. Kraft, V. A. et al. GTP cyclohydrolase 1/tetrahydrobiopterin counteract ferroptosis through lipid remodeling. ACS Cent. Sci. 6, 41–53 (2019).

    PubMed  PubMed Central  Google Scholar 

  8. Soula, M. et al. Metabolic determinants of cancer cell sensitivity to canonical ferroptosis inducers. Nat. Chem. Biol. 16, 1351–1360 (2020). Together with ref.7, this paper reports a GPX4-independent ferroptosis defence mechanism involving GCH1 and BH4.

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Mao, C. et al. DHODH-mediated ferroptosis defence is a targetable vulnerability in cancer. Nature 593, 586–590 (2021). This study identifies the first mitochondria-localized ferroptosis defence mechanism and proposes to target ferroptosis vulnerabilities in cancer based on imbalanced ferroptosis defences.

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Ingold, I. et al. Selenium utilization by GPX4 is required to prevent hydroperoxide-induced ferroptosis. Cell 172, 409–422.e21 (2018).

    CAS  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).

    PubMed  PubMed Central  Google Scholar 

  12. Wiernicki, B. et al. Excessive phospholipid peroxidation distinguishes ferroptosis from other cell death modes including pyroptosis. Cell Death Dis. 11, 922 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  14. Hassannia, B., Vandenabeele, P. & Berghe, T. V. Targeting ferroptosis to iron out cancer. Cancer Cell 35, 830–849 (2019).

    CAS  PubMed  Google Scholar 

  15. Jiang, L. et al. Ferroptosis as a p53-mediated activity during tumour suppression. Nature 520, 57–62 (2015). This study represents the first prominent publication that links ferroptosis to tumour suppression.

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Zhang, Y. et al. BAP1 links metabolic regulation of ferroptosis to tumour suppression. Nat. Cell Biol. 20, 1181–1192 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Yi, J., Zhu, J., Wu, J., Thompson, C. B. & Jiang, X. Oncogenic activation of PI3K-AKT-mTOR signaling suppresses ferroptosis via SREBP-mediated lipogenesis. Proc. Natl Acad. Sci. USA 117, 31189–31197 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Ubellacker, J. M. et al. Lymph protects metastasizing melanoma cells from ferroptosis. Nature 585, 113–118 (2020). This paper reports that melanoma cells exposed to the lymphatic environment can escape from ferroptosis, promoting their subsequent metastasis through the blood.

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Angeli, J. P. F., Krysko, D. V. & Conrad, M. Ferroptosis at the crossroads of cancer-acquired drug resistance and immune evasion. Nat. Rev. Cancer 19, 405–414 (2019).

    Google Scholar 

  20. Wu, J. et al. Intercellular interaction dictates cancer cell ferroptosis via NF2–YAP signalling. Nature 572, 402–406 (2019). This study shows that inactivation of tumour suppressors in the the E-cadherin–NF2–Hippo pathway sensitizes tumours to ferroptosis, presenting an example of how genetic mutations in certain tumour types can induce a vulnerability to ferroptosis.

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Hangauer, M. J. et al. Drug-tolerant persister cancer cells are vulnerable to GPX4 inhibition. Nature 551, 247–250 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Viswanathan, V. S. et al. Dependency of a therapy-resistant state of cancer cells on a lipid peroxidase pathway. Nature 547, 453–457 (2017). Together with ref.21, this study shows that certain therapy resistant cancer cells are vulnerable to ferroptosis.

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Zou, Y. et al. Plasticity of ether lipids promotes ferroptosis susceptibility and evasion. Nature 585, 603–608 (2020). This study identifies ePLs as additional substrates for lipid peroxidation to drive ferroptosis, and further establishes their relevance to ferroptosis susceptibility in tumours.

    CAS  PubMed  PubMed Central  Google Scholar 

  24. 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).

    PubMed  PubMed Central  Google Scholar 

  25. Lei, G. et al. The role of ferroptosis in ionizing radiation-induced cell death and tumor suppression. Cell Res. 30, 146–162 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Guo, J. et al. Ferroptosis: a novel anti-tumor action for cisplatin. Cancer Res. Treat. 50, 445 (2018).

    CAS  PubMed  Google Scholar 

  27. Sun, X. et al. Activation of the p62-Keap1-NRF2 pathway protects against ferroptosis in hepatocellular carcinoma cells. Hepatology 63, 173–184 (2016).

    CAS  PubMed  Google Scholar 

  28. Wang, W. et al. CD8+ T cells regulate tumour ferroptosis during cancer immunotherapy. Nature 569, 270–274 (2019). This paper reports that activated CD8+ T cells promote ferroptosis in tumours through inhibition of SLC7A11, and that combining ICIs with FINs represents a promising strategy in cancer therapy.

    CAS  PubMed  PubMed Central  Google Scholar 

  29. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Liang, C., Zhang, X., Yang, M. & Dong, X. Recent progress in ferroptosis inducers for cancer therapy. Adv. Mater. 31, 1904197 (2019).

    CAS  Google Scholar 

  31. Gan, B. Mitochondrial regulation of ferroptosis. J. Cell Biol. 220, e202105043 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  34. Lee, H., Zhuang, L. & Gan, B. Energy stress inhibits ferroptosis via AMPK. Mol. Cell. Oncol. 7, 1761242 (2020).

    PubMed  PubMed Central  Google Scholar 

  35. Li, C. et al. LKB1-AMPK axis negatively regulates ferroptosis by inhibiting fatty acid synthesis. Signal. Transduct. Target. Ther. 5, 187 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Shimada, K. et al. Global survey of cell death mechanisms reveals metabolic regulation of ferroptosis. Nat. Chem. Biol. 12, 497–503 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Cui, W., Liu, D., Gu, W. & Chu, B. Peroxisome-driven ether-linked phospholipids biosynthesis is essential for ferroptosis. Cell Death Differ. 28, 2536–2551 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Lee, H., Zhuang, L. & Gan, B. Ether phospholipids govern ferroptosis. J. Genet. Genomics 48, 517–519 (2021).

    CAS  PubMed  Google Scholar 

  41. Conrad, M. & Pratt, D. A. The chemical basis of ferroptosis. Nat. Chem. Biol. 15, 1137–1147 (2019).

    CAS  PubMed  Google Scholar 

  42. Shah, R., Shchepinov, M. S. & Pratt, D. A. Resolving the role of lipoxygenases in the initiation and execution of ferroptosis. ACS Cent. Sci. 4, 387–396 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Gaschler, M. M. & Stockwell, B. R. Lipid peroxidation in cell death. Biochem. Biophys. Res. Commun. 482, 419–425 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Yang, W. S. et al. Peroxidation of polyunsaturated fatty acids by lipoxygenases drives ferroptosis. Proc. Natl Acad. Sci. USA 113, E4966–E4975 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Koppula, P., Zhuang, L. & Gan, B. Cytochrome P450 reductase (POR) as a ferroptosis fuel. Protein Cell 12, 675–679 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Yan, B. et al. Membrane damage during ferroptosis is caused by oxidation of phospholipids catalyzed by the oxidoreductases POR and CYB5R1. Mol. Cell 81, 355–369.e10 (2021).

    CAS  PubMed  Google Scholar 

  47. Zou, Y. et al. Cytochrome P450 oxidoreductase contributes to phospholipid peroxidation in ferroptosis. Nat. Chem. Biol. 16, 302–309 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Wenzel, S. E. et al. PEBP1 wardens ferroptosis by enabling lipoxygenase generation of lipid death signals. Cell 171, 628–641.e26 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Angeli, J. P. F. et al. Inactivation of the ferroptosis regulator Gpx4 triggers acute renal failure in mice. Nat. Cell Biol. 16, 1180–1191 (2014). This study, together with ref.4, establishes the critical role of GPX4 in ferroptosis suppression.

    PubMed Central  Google Scholar 

  50. Magtanong, L. et al. Exogenous monounsaturated fatty acids promote a ferroptosis-resistant cell state. Cell Chem. Biol. 26, 420–432.e9 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Paton, C. M. & Ntambi, J. M. Biochemical and physiological function of stearoyl-CoA desaturase. Am. J. Physiol. Endocrinol. Metab. 297, E28–E37 (2009).

    CAS  PubMed  Google Scholar 

  52. Tesfay, L. et al. Stearoyl-coa desaturase 1 protects ovarian cancer cells from ferroptotic cell death. Cancer Res. 79, 5355–5366 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Chen, X., Yu, C., Kang, R. & Tang, D. Iron metabolism in ferroptosis. Front. Cell Dev. Biol. 8, 590226 (2020).

    PubMed  PubMed Central  Google Scholar 

  54. Gao, M. et al. Ferroptosis is an autophagic cell death process. Cell Res. 26, 1021–1032 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Kremer, D. M. et al. GOT1 inhibition promotes pancreatic cancer cell death by ferroptosis. Nat. Commun. 12, 4860 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Murphy, M. P. How mitochondria produce reactive oxygen species. Biochem. J. 417, 1–13 (2009).

    CAS  PubMed  Google Scholar 

  58. Zheng, J. & Conrad, M. The metabolic underpinnings of ferroptosis. Cell Metab. 32, 920–937 (2020).

    CAS  PubMed  Google Scholar 

  59. Friedman, J. R. & Nunnari, J. Mitochondrial form and function. Nature 505, 335–343 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Vasan, K., Werner, M. & Chandel, N. S. Mitochondrial metabolism as a target for cancer therapy. Cell Metab. 32, 341–352 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Gao, M. et al. Role of mitochondria in ferroptosis. Mol. Cell 73, 354–363.e3 (2019). This paper demonstrates the importance of mitochondria in promoting ferroptosis.

    CAS  PubMed  Google Scholar 

  63. Heldt, H. & Piechulla, B. 15-Lipids are membrane constituents and function as carbon stores. Plant Biochem. https://doi.org/10.1016/b978-0-12-384986-1.00015-6 (2011).

    Article  Google Scholar 

  64. Brigelius-Flohé, R. & Maiorino, M. Glutathione peroxidases. Biochim. Biophys. Acta 1830, 3289–3303 (2013).

    PubMed  Google Scholar 

  65. Brigelius-Flohé, R. & Flohé, L. Regulatory phenomena in the glutathione peroxidase superfamily. Antioxid. Redox Signal. 33, 498–516 (2020).

    PubMed  Google Scholar 

  66. Seibt, T. M., Proneth, B. & Conrad, M. Role of GPX4 in ferroptosis and its pharmacological implication. Free Radic. Biol. Med. 133, 144–152 (2019).

    CAS  PubMed  Google Scholar 

  67. Ursini, F., Maiorino, M., Valente, M., Ferri, L. & Gregolin, C. Purification from pig liver of a protein which protects liposomes and biomembranes from peroxidative degradation and exhibits glutathione peroxidase activity on phosphatidylcholine hydroperoxides. Biochim. Biophys. Acta 710, 197–211 (1982).

    CAS  PubMed  Google Scholar 

  68. Pushpa-Rekha, T. R., Burdsall, A. L., Oleksa, L. M., Chisolm, G. M. & Driscoll, D. M. Rat phospholipid-hydroperoxide glutathione peroxidase: cDNA cloning and identification of multiple transcription and translation start sites. J. Biol. Chem. 270, 26993–26999 (1995).

    CAS  PubMed  Google Scholar 

  69. Pfeifer, H. et al. Identification of a specific sperm nuclei selenoenzyme necessary for protamine thiol cross-linking during sperm maturation. FASEB J. 15, 1236–1238 (2001).

    CAS  PubMed  Google Scholar 

  70. Maiorino, M. et al. Distinct promoters determine alternative transcription of gpx-4 into phospholipid-hydroperoxide glutathione peroxidase variants. J. Biol. Chem. 278, 34286–34290 (2003).

    CAS  PubMed  Google Scholar 

  71. Moreno, S. G., Laux, G., Brielmeier, M., Bornkamm, G. W. & Conrad, M. Testis-specific expression of the nuclear form of phospholipid hydroperoxide glutathione peroxidase (PHGPx). Biol. Chem. 384, 635–643 (2003).

    CAS  PubMed  Google Scholar 

  72. Yant, L. J. et al. The selenoprotein GPX4 is essential for mouse development and protects from radiation and oxidative damage insults. Free Radic. Biol. Med. 34, 496–502 (2003).

    CAS  PubMed  Google Scholar 

  73. Conrad, M. et al. The nuclear form of phospholipid hydroperoxide glutathione peroxidase is a protein thiol peroxidase contributing to sperm chromatin stability. Mol. Cell. Biol. 25, 7637–7644 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  74. Liang, H. et al. Short form glutathione peroxidase 4 is the essential isoform required for survival and somatic mitochondrial functions. J. Biol. Chem. 284, 30836–30844 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Schneider, M. et al. Mitochondrial glutathione peroxidase 4 disruption causes male infertility. FASEB J. 23, 3233–3242 (2009).

    CAS  PubMed  Google Scholar 

  76. Imai, H. et al. Depletion of selenoprotein GPx4 in spermatocytes causes male infertility in mice. J. Biol. Chem. 284, 32522–32532 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Forman, H. J., Zhang, H. & Rinna, A. Glutathione: overview of its protective roles, measurement, and biosynthesis. Mol. Asp. Med. 30, 1–12 (2009).

    CAS  Google Scholar 

  78. Aquilano, K., Baldelli, S. & Ciriolo, M. R. Glutathione: new roles in redox signaling for an old antioxidant. Front. Pharmacol. 5, 196 (2014).

    PubMed  PubMed Central  Google Scholar 

  79. Koppula, P., Zhang, Y., Zhuang, L. & Gan, B. Amino acid transporter SLC7A11/xCT at the crossroads of regulating redox homeostasis and nutrient dependency of cancer. Cancer Commun. 38, 12 (2018).

    Google Scholar 

  80. Liu, X. et al. Cystine transporter regulation of pentose phosphate pathway dependency and disulfide stress exposes a targetable metabolic vulnerability in cancer. Nat. Cell Biol. 22, 476–486 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  81. Sato, H., Tamba, M., Ishii, T. & Bannai, S. Cloning and expression of a plasma membrane cystine/glutamate exchange transporter composed of two distinct proteins. J. Biol. Chem. 274, 11455–11458 (1999).

    CAS  PubMed  Google Scholar 

  82. Koppula, P., Zhuang, L. & Gan, B. Cystine transporter SLC7A11/xCT in cancer: ferroptosis, nutrient dependency, and cancer therapy. Protein Cell 12, 599–620 (2021).

    CAS  PubMed  Google Scholar 

  83. Stefely, J. A. & Pagliarini, D. J. Biochemistry of mitochondrial coenzyme Q biosynthesis. Trends Biochem. Sci. 42, 824–843 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  84. Crane, F. L. Discovery of ubiquinone (coenzyme Q) and an overview of function. Mitochondrion 7, S2–S7 (2007).

    CAS  PubMed  Google Scholar 

  85. Kalen, A., Norling, B., Appelkvist, E. L. & Dallner, G. Ubiquinone biosynthesis by the microsomal fraction from rat liver. Biochim. Biophys. Acta 926, 70–78 (1987).

    CAS  PubMed  Google Scholar 

  86. Turunen, M., Olsson, J. & Dallner, G. Metabolism and function of coenzyme Q. Biochim. Biophys. Acta 1660, 171–199 (2004).

    CAS  PubMed  Google Scholar 

  87. Takahashi, T., Okamoto, T., Mori, K., Sayo, H. & Kishi, T. Distribution of ubiquinone and ubiquinol homologues in rat tissues and subcellular fractions. Lipids 28, 803–809 (1993).

    CAS  PubMed  Google Scholar 

  88. Morre, D. J. & Morre, D. M. Non-mitochondrial coenzyme Q. Biofactors 37, 355–360 (2011).

    CAS  PubMed  Google Scholar 

  89. Thöny, B., Auerbach, G. & Blau, N. Tetrahydrobiopterin biosynthesis, regeneration and functions. Biochem. J. 347, 1–16 (2000).

    PubMed  PubMed Central  Google Scholar 

  90. Chen, D. et al. iPLA2β-mediated lipid detoxification controls p53-driven ferroptosis independent of GPX4. Nat. Commun. 12, 3644 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  91. Sun, W.-Y. et al. Phospholipase iPLA 2 β averts ferroptosis by eliminating a redox lipid death signal. Nat. Chem. Biol. 17, 465–476 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  92. Tan, S. K. et al. Obesity-dependent adipokine chemerin suppresses fatty acid oxidation to confer ferroptosis resistance. Cancer Discov. 11, 2072–2093 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  93. Liu, W. et al. Dysregulated cholesterol homeostasis results in resistance to ferroptosis increasing tumorigenicity and metastasis in cancer. Nat. Commun. 12, 5103 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  94. Sviderskiy, V. O., Terzi, E. M. & Possemato, R. in Ferroptosis in Health and Disease 215-237 (Springer, 2019).

  95. Alvarez, S. W. et al. NFS1 undergoes positive selection in lung tumours and protects cells from ferroptosis. Nature 551, 639–643 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  96. Du, J. et al. Identification of Frataxin as a regulator of ferroptosis. Redox Biol. 32, 101483 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  97. Kim, E. H., Shin, D., Lee, J., Jung, A. R. & Roh, J.-L. CISD2 inhibition overcomes resistance to sulfasalazine-induced ferroptotic cell death in head and neck cancer. Cancer Lett. 432, 180–190 (2018).

    CAS  PubMed  Google Scholar 

  98. Brown, C. W. et al. Prominin2 drives ferroptosis resistance by stimulating iron export. Dev. Cell 51, 575–586.e4 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  99. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  100. Chen, D. et al. NRF2 is a major target of ARF in p53-independent tumor suppression. Mol. Cell 68, 224–232.e4 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  101. Dodson, M., Castro-Portuguez, R. & Zhang, D. D. NRF2 plays a critical role in mitigating lipid peroxidation and ferroptosis. Redox Biol. 23, 101107 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  102. Hu, K. et al. Suppression of the SLC7A11/glutathione axis causes synthetic lethality in KRAS-mutant lung adenocarcinoma. J. Clin. Invest. 130, 1752–1766 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  103. Lim, J. K. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  104. de la Vega, M. R., Chapman, E. & Zhang, D. D. NRF2 and the hallmarks of cancer. Cancer Cell 34, 21–43 (2018).

    PubMed Central  Google Scholar 

  105. Garcia-Bermudez, J. et al. Squalene accumulation in cholesterol auxotrophic lymphomas prevents oxidative cell death. Nature 567, 118–122 (2019). This study shows that loss of SQLE expression in ALK+ anaplastic large-cell lymphomas leads to squalene accumulation and ferroptosis resistance, yet renders these tumours cholesterol auxotrophic.

    CAS  PubMed  PubMed Central  Google Scholar 

  106. Martinez-Outschoorn, U. E., Peiris-Pagés, M., Pestell, R. G., Sotgia, F. & Lisanti, M. P. Cancer metabolism: a therapeutic perspective. Nat. Rev. Clin. Oncol. 14, 11–31 (2017).

    CAS  PubMed  Google Scholar 

  107. Wolpaw, A. J. & Dang, C. V. Exploiting metabolic vulnerabilities of cancer with precision and accuracy. Trends Cell Biol. 28, 201–212 (2018).

    CAS  PubMed  Google Scholar 

  108. Tsoi, J. et al. Multi-stage differentiation defines melanoma subtypes with differential vulnerability to drug-induced iron-dependent oxidative stress. Cancer Cell 33, 890–904.e5 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  109. Krebs, A. M. et al. The EMT-activator Zeb1 is a key factor for cell plasticity and promotes metastasis in pancreatic cancer. Nat. Cell Biol. 19, 518–529 (2017).

    CAS  PubMed  Google Scholar 

  110. Lin, C.-C. et al. DDR2 upregulation confers ferroptosis susceptibility of recurrent breast tumors through the Hippo pathway. Oncogene 40, 2018–2034 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  111. Lee, J.-Y. et al. Polyunsaturated fatty acid biosynthesis pathway determines ferroptosis sensitivity in gastric cancer. Proc. Natl Acad. Sci. USA 117, 32433–32442 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  112. Müller, S. et al. CD44 regulates epigenetic plasticity by mediating iron endocytosis. Nat. Chem. 12, 929–938 (2020).

    PubMed  PubMed Central  Google Scholar 

  113. Bebber, C. M. et al. Ferroptosis response segregates small cell lung cancer (SCLC) neuroendocrine subtypes. Nat. Commun. 12, 2048 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  114. Verma, N. et al. Synthetic lethal combination targeting BET uncovered intrinsic susceptibility of TNBC to ferroptosis. Sci. Adv. 6, eaba8968 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  115. Yang, W.-H. et al. The hippo pathway effector TAZ regulates ferroptosis in renal cell carcinoma. Cell Rep. 28, 2501–2508.e4 (2019).

    CAS  PubMed  Google Scholar 

  116. Iliopoulos, O., Kibel, A., Gray, S. & Kaelin, W. G. Tumour suppression by the human von Hippel-Lindau gene product. Nat. Med. 1, 822–826 (1995).

    CAS  PubMed  Google Scholar 

  117. Kaelin, W. G. Jr The von Hippel–Lindau tumour suppressor protein: O2 sensing and cancer. Nat. Rev. Cancer 8, 865–873 (2008).

    CAS  PubMed  Google Scholar 

  118. Miess, H. et al. The glutathione redox system is essential to prevent ferroptosis caused by impaired lipid metabolism in clear cell renal cell carcinoma. Oncogene 37, 5435–5450 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  119. Poursaitidis, I. et al. Oncogene-selective sensitivity to synchronous cell death following modulation of the amino acid nutrient cystine. Cell Rep. 18, 2547–2556 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  120. Zhang, Y. et al. mTORC1 couples cyst (e) ine availability with GPX4 protein synthesis and ferroptosis regulation. Nat. Commun. 12, 1589 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  121. Wang, T.-X. et al. The oncometabolite 2-hydroxyglutarate produced by mutant IDH1 sensitizes cells to ferroptosis. Cell Death Dis. 10, 755 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  122. Kaelin, W. G. The concept of synthetic lethality in the context of anticancer therapy. Nat. Rev. Cancer 5, 689–698 (2005).

    CAS  PubMed  Google Scholar 

  123. Schmitt, A. et al. Dimethyl fumarate induces ferroptosis and impairs NF-κB/STAT3 signaling in DLBCL. Blood 138, 871–884 (2021).

    CAS  PubMed  Google Scholar 

  124. Sato, H., Fujiwara, K., Sagara, J. & Bannai, S. Induction of cystine transport activity in mouse peritoneal macrophages by bacterial lipopolysaccharide. Biochem. J. 310, 547–551 (1995).

    CAS  PubMed  PubMed Central  Google Scholar 

  125. Wen, Q., Liu, J., Kang, R., Zhou, B. & Tang, D. The release and activity of HMGB1 in ferroptosis. Biochem. Biophys. Res. Commun. 510, 278–283 (2019).

    CAS  PubMed  Google Scholar 

  126. Luo, X. et al. Oxygenated phosphatidylethanolamine navigates phagocytosis of ferroptotic cells by interacting with TLR2. Cell Death Differ. 28, 1971–1989 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    PubMed  PubMed Central  Google Scholar 

  128. Yu, B., Choi, B., Li, W. & Kim, D.-H. Magnetic field boosted ferroptosis-like cell death and responsive MRI using hybrid vesicles for cancer immunotherapy. Nat. Commun. 11, 3637 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  129. Li, C., Jiang, P., Wei, S., Xu, X. & Wang, J. Regulatory T cells in tumor microenvironment: new mechanisms, potential therapeutic strategies and future prospects. Mol. Cancer 19, 116 (2020).

    PubMed  PubMed Central  Google Scholar 

  130. Xu, C. et al. The glutathione peroxidase Gpx4 prevents lipid peroxidation and ferroptosis to sustain Treg cell activation and suppression of antitumor immunity. Cell Rep. 35, 109235 (2021).

    CAS  PubMed  Google Scholar 

  131. Zhu, H. et al. Asah2 represses the p53–Hmox1 axis to protect myeloid-derived suppressor cells from ferroptosis. J. Immunol. 206, 1395–1404 (2021).

    CAS  PubMed  Google Scholar 

  132. Xia, Y. et al. Engineering macrophages for cancer immunotherapy and drug delivery. Adv. Mater. 32, 2002054 (2020).

    CAS  Google Scholar 

  133. Kapralov, A. A. et al. Redox lipid reprogramming commands susceptibility of macrophages and microglia to ferroptotic death. Nat. Chem. Biol. 16, 278–290 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  134. Matsushita, M. et al. T cell lipid peroxidation induces ferroptosis and prevents immunity to infection. J. Exp. Med. 212, 555–568 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  135. Drijvers, J. M. et al. Pharmacologic screening identifies metabolic vulnerabilities of CD8+ T cells. Cancer Immunol. Res. 9, 184–199 (2020).

    PubMed  PubMed Central  Google Scholar 

  136. Ma, X. et al. CD36-mediated ferroptosis dampens intratumoral CD8+ T cell effector function and impairs their antitumor ability. Cell Metab. 33, 1001–1012.e5 (2021). This publication shows that ferroptosis in CD8+ T cells impairs their antitumour activity, leading to increased tumour growth, and suggests that the effect of ferroptosis on tumour growth can be context dependent.

    CAS  PubMed  PubMed Central  Google Scholar 

  137. Yao, Y. et al. Selenium–GPX4 axis protects follicular helper T cells from ferroptosis. Nat. Immunol. 22, 1127–1139 (2021).

    CAS  PubMed  Google Scholar 

  138. Arensman, M. D. et al. Cystine–glutamate antiporter xCT deficiency suppresses tumor growth while preserving antitumor immunity. Proc. Natl Acad. Sci. USA 116, 9533–9542 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  139. Gmünder, H., Eck, H. P. & Dröge, W. Low membrane transport activity for cystine in resting and mitogenically stimulated human lymphocyte preparations and human T cell clones. Eur. J. Biochem. 201, 113–117 (1991).

    PubMed  Google Scholar 

  140. Pacheco, R. et al. Glutamate released by dendritic cells as a novel modulator of T cell activation. J. Immunol. 177, 6695–6704 (2006).

    CAS  PubMed  Google Scholar 

  141. Ye, L. F. et al. Radiation-induced lipid peroxidation triggers ferroptosis and synergizes with ferroptosis inducers. ACS Chem. Biol. 15, 469–484 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  142. Lei, G. et al. Ferroptosis as a mechanism to mediate p53 function in tumor radiosensitivity. Oncogene 40, 3533–3547 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  143. Lang, X. et al. Radiotherapy and immunotherapy promote tumoral lipid oxidation and ferroptosis via synergistic repression of SLC7A11. Cancer Discov. 9, 1673–1685 (2019). Together with ref.25 and ref.141, this study reports that ionizing radiation induces ferroptosis and proposes to combine radiotherapy with FINs in cancer treatment.

    CAS  PubMed  PubMed Central  Google Scholar 

  144. Dixon, S. J. et al. Pharmacological inhibition of cystine–glutamate exchange induces endoplasmic reticulum stress and ferroptosis. eLife 3, e02523 (2014).

    PubMed  PubMed Central  Google Scholar 

  145. Cao, J. et al. DJ-1 suppresses ferroptosis through preserving the activity of S-adenosyl homocysteine hydrolase. Nat. Commun. 11, 1251 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  146. Zhu, J. et al. Transsulfuration activity can support cell growth upon extracellular cysteine limitation. Cell Metab. 30, 865–876.e5 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  147. Liu, N., Lin, X. & Huang, C. Activation of the reverse transsulfuration pathway through NRF2/CBS confers erastin-induced ferroptosis resistance. Br. J. Cancer 122, 279–292 (2020).

    CAS  PubMed  Google Scholar 

  148. Wang, L. et al. A pharmacological probe identifies cystathionine β-synthase as a new negative regulator for ferroptosis. Cell Death Dis. 9, 1025 (2018).

    PubMed  PubMed Central  Google Scholar 

  149. Song, X. et al. PDK4 dictates metabolic resistance to ferroptosis by suppressing pyruvate oxidation and fatty acid synthesis. Cell Rep. 34, 108767 (2021).

    CAS  PubMed  Google Scholar 

  150. Badgley, M. A. et al. Cysteine depletion induces pancreatic tumor ferroptosis in mice. Science 368, 85–89 (2020). Using genetically engineered mouse models, this study shows that inducing tumour ferroptosis by blocking SLC7A11-mediated cystine transport is a promising therapeutic strategy in pancreatic cancer.

    CAS  PubMed  PubMed Central  Google Scholar 

  151. Lei, G., Mao, C., Yan, Y., Zhuang, L. & Gan, B. Ferroptosis, radiotherapy, and combination therapeutic strategies. Protein Cell 12, 836–857 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  153. Yu, Y. et al. The ferroptosis inducer erastin enhances sensitivity of acute myeloid leukemia cells to chemotherapeutic agents. Mol. Cell. Oncol. 2, e1054549 (2015).

    PubMed  PubMed Central  Google Scholar 

  154. Yamaguchi, H. et al. Caspase-independent cell death is involved in the negative effect of EGF receptor inhibitors on cisplatin in non-small cell lung cancer cells. Clin. Cancer Res. 19, 845–854 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  155. Pan, X. et al. Erastin decreases radioresistance of NSCLC cells partially by inducing GPX4‑mediated ferroptosis. Oncol. Lett. 17, 3001–3008 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  156. Xie, L. et al. Solute carrier protein family may involve in radiation-induced radioresistance of non-small cell lung cancer. J. Cancer Res. Clin. Oncol. 137, 1739 (2011).

    CAS  PubMed  Google Scholar 

  157. Roh, J.-L., Kim, E. H., Jang, H. J., Park, J. Y. & Shin, D. Induction of ferroptotic cell death for overcoming cisplatin resistance of head and neck cancer. Cancer Lett. 381, 96–103 (2016).

    CAS  PubMed  Google Scholar 

  158. Zhou, H.-H. et al. Erastin reverses ABCB1-mediated docetaxel resistance in ovarian cancer. Front. Oncol. 9, 1398 (2019).

    PubMed  PubMed Central  Google Scholar 

  159. Jiang, Z. et al. TYRO3 induces anti–PD-1/PD-L1 therapy resistance by limiting innate immunity and tumoral ferroptosis. J. Clin. Invest. 131, e139434 (2021).

    PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  161. US National Library of Medicine. ClinicalTrials.gov https://ClinicalTrials.gov/show/NCT04205357 (2019).

  162. US National Library of Medicine. ClinicalTrials.gov https://ClinicalTrials.gov/show/NCT04092647 (2019).

  163. US National Library of Medicine. ClinicalTrials.gov https://ClinicalTrials.gov/show/NCT02559778 (2015).

  164. US National Library of Medicine. ClinicalTrials.gov https://ClinicalTrials.gov/show/NCT03247088 (2017).

  165. Eaton, J. K. et al. Selective covalent targeting of GPX4 using masked nitrile-oxide electrophiles. Nat. Chem. Biol. 16, 497–506 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  166. Hassannia, B. et al. Nano-targeted induction of dual ferroptotic mechanisms eradicates high-risk neuroblastoma. J. Clin. Invest. 128, 3341–3355 (2018).

    PubMed  PubMed Central  Google Scholar 

  167. Woo, J. H. et al. Elucidating compound mechanism of action by network perturbation analysis. Cell 162, 441–451 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  168. Zhang, H.-L. et al. PKCβII phosphorylates ACSL4 to amplify lipid peroxidation to induce ferroptosis. Nat. Cell Biol. 24, 88–89 (2022). This study challenges the view that PUFA-PL synthesis plays a passive role in ferroptosis execution and suggests a dynamic regulation of ferroptosis execution by upstream signalling.

    CAS  PubMed  Google Scholar 

  169. Yang, Y. et al. Nedd4 ubiquitylates VDAC2/3 to suppress erastin-induced ferroptosis in melanoma. Nat. Commun. 11, 433 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  170. Feng, H. & Stockwell, B. R. Unsolved mysteries: how does lipid peroxidation cause ferroptosis? PLoS Biol. 16, e2006203 (2018).

    PubMed  PubMed Central  Google Scholar 

  171. Larraufie, M.-H. et al. Incorporation of metabolically stable ketones into a small molecule probe to increase potency and water solubility. Bioorg. Med. Chem. Lett. 25, 4787–4792 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  172. Plosker, G. L. & Croom, K. F. Sulfasalazine. Drugs 65, 1825–1849 (2005).

    CAS  PubMed  Google Scholar 

  173. Gout, P., Buckley, A., Simms, C. & Bruchovsky, N. Sulfasalazine, a potent suppressor of lymphoma growth by inhibition of the xc− cystine transporter: a new action for an old drug. Leukemia 15, 1633–1640 (2001).

    CAS  PubMed  Google Scholar 

  174. Robert, S. M. et al. SLC7A11 expression is associated with seizures and predicts poor survival in patients with malignant glioma. Sci. Transl. Med. 7, 289ra286 (2015).

    Google Scholar 

  175. Zheng, J. et al. Sorafenib fails to trigger ferroptosis across a wide range of cancer cell lines. Cell Death Dis. 12, 698 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  176. Cramer, S. L. et al. Systemic depletion of L-cyst(e)ine with cyst(e)inase increases reactive oxygen species and suppresses tumor growth. Nat. Med. 23, 120–127 (2017).

    CAS  PubMed  Google Scholar 

  177. Gaschler, M. M. et al. FINO 2 initiates ferroptosis through GPX4 inactivation and iron oxidation. Nat. Chem. Biol. 14, 507–515 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  178. Bykov, V. J., Eriksson, S. E., Bianchi, J. & Wiman, K. G. Targeting mutant p53 for efficient cancer therapy. Nat. Rev. Cancer 18, 89–102 (2018).

    CAS  PubMed  Google Scholar 

  179. Bieging, K. T., Mello, S. S. & Attardi, L. D. Unravelling mechanisms of p53-mediated tumour suppression. Nat. Rev. Cancer 14, 359–370 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  180. Wang, Y. et al. Epigenetic regulation of ferroptosis by H2B monoubiquitination and p53. EMBO Rep. 20, e47563 (2019).

    PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  182. Zhang, Y. et al. Ferredoxin reductase is critical for p53-dependent tumor suppression via iron regulatory protein 2. Genes Dev. 31, 1243–1256 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  183. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  184. Wang, S.-J. et al. Acetylation is crucial for p53-mediated ferroptosis and tumor suppression. Cell Rep. 17, 366–373 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  187. Carbone, M. et al. BAP1 and cancer. Nat. Rev. Cancer 13, 153–159 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  188. Ventii, K. H. et al. BRCA1-associated protein-1 is a tumor suppressor that requires deubiquitinating activity and nuclear localization. Cancer Res. 68, 6953–6962 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  189. Carbone, M. et al. Biological mechanisms and clinical significance of BAP1 mutations in human cancer. Cancer Discov. 10, 1103–1120 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  190. Cancer Genome Atlas Research Network. Comprehensive genomic characterization of squamous cell lung cancers. Nature 489, 519 (2012).

    Google Scholar 

  191. Collisson, E. et al. Comprehensive molecular profiling of lung adenocarcinoma: the cancer genome atlas research network. Nature 511, 543–550 (2014).

    CAS  Google Scholar 

  192. Baird, L. & Yamamoto, M. The molecular mechanisms regulating the KEAP1-NRF2 pathway. Mol. Cell. Biol. 40, e00099-20 (2020).

    PubMed  PubMed Central  Google Scholar 

  193. Kensler, T. W., Wakabayashi, N. & Biswal, S. Cell survival responses to environmental stresses via the Keap1-Nrf2-ARE pathway. Annu. Rev. Pharmacol. Toxicol. 47, 89–116 (2007).

    CAS  PubMed  Google Scholar 

  194. Sasaki, H. et al. Electrophile response element-mediated induction of the cystine/glutamate exchange transporter gene expression. J. Biol. Chem. 277, 44765–44771 (2002).

    CAS  PubMed  Google Scholar 

  195. Sherr, C. J. Divorcing ARF and p53: an unsettled case. Nat. Rev. Cancer 6, 663–673 (2006).

    CAS  PubMed  Google Scholar 

  196. Fruman, D. A. et al. The PI3K pathway in human disease. Cell 170, 605–635 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  197. Zhang, Y. et al. A pan-cancer proteogenomic atlas of PI3K/AKT/mTOR pathway alterations. Cancer Cell 31, 820–832.e3 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  198. Lei, G., Zhuang, L. & Gan, B. mTORC1 and ferroptosis: regulatory mechanisms and therapeutic potential. BioEssays 43, e2100093 (2021).

    PubMed  Google Scholar 

  199. 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).

    CAS  PubMed  Google Scholar 

  200. Prior, I. A., Lewis, P. D. & Mattos, C. A comprehensive survey of Ras mutations in cancer. Cancer Res. 72, 2457–2467 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  201. Pylayeva-Gupta, Y., Grabocka, E. & Bar-Sagi, D. RAS oncogenes: weaving a tumorigenic web. Nat. Rev. Cancer 11, 761–774 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  202. Padanad, M. S. et al. Fatty acid oxidation mediated by acyl-CoA synthetase long chain 3 is required for mutant KRAS lung tumorigenesis. Cell Rep. 16, 1614–1628 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  203. Wang, X. et al. NEDD4-1 is a proto-oncogenic ubiquitin ligase for PTEN. Cell 128, 129–139 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  204. Yagoda, N. et al. RAS–RAF–MEK-dependent oxidative cell death involving voltage-dependent anion channels. Nature 447, 865–869 (2007).

    CAS  Google Scholar 

  205. Cao, J., Lou, S., Ying, M. & Yang, B. DJ-1 as a human oncogene and potential therapeutic target. Biochem. Pharmacol. 93, 241–250 (2015).

    CAS  PubMed  Google Scholar 

  206. Leclerc, D. et al. Oncogenic role of PDK4 in human colon cancer cells. Br. J. Cancer 116, 930–936 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank Amy Ninetto from the Research Medical Library at MD Anderson, for editing the manuscript, and apologize to colleagues whose work cannot be cited in this manuscript due to space limitations. B.G. is supported by The University of Texas MD Anderson Cancer Center, Emerson Collective Cancer Research Fund, and grants R01CA181196, R01CA244144 and R01CA247992 from the National Institutes of Health. The research from the authors’ lab has also been supported by the National Institutes of Health Cancer Center Support Grant P30CA016672 to The University of Texas MD Anderson Cancer Center.

Author information

Authors and Affiliations

Authors

Contributions

G.L. and B.G. researched data for the article and contributed substantially to discussion of the content. All authors wrote the article and reviewed and/or edited the manuscript before submission.

Corresponding author

Correspondence to Boyi Gan.

Ethics declarations

Competing interests

B.G. is an inventor on patent applications involving targeting ferroptosis in cancer therapy. G.L. and L.Z. declare no competing interests.

Peer review

Peer review information

Nature Reviews Cancer thanks Marcus Conrad, Xuejun Jiang and Silvia Von Karsted for their contribution to the peer review of this work.

Additional information

Publisher’s note

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

Related links

Cancer Therapeutics Response Portal: https://portals.broadinstitute.org/ctrp/

Glossary

Cristae

Folds of the inner mitochondrial membrane that extend into the matrix of a mitochondrion.

Ferroptosis inducers

(FINs). A compound or treatment that can induce ferroptosis by boosting ferroptosis-promoting mechanisms and/or suppressing ferroptosis defence mechanisms.

Polyunsaturated fatty acid

(PUFA). A fatty acid that contains more than one double bond and is required for cellular signalling and membrane fluidity.

Peroxisomes

Organelles that are important for β-oxidation of very-long-chain fatty acids and synthesis of ether phospholipids.

Fenton reaction

A non-enzymatic reaction of labile iron and H2O2 that generates hydroxide and hydroxyl radicals, which can subsequently induce lipid peroxidation.

Anaplerotic reactions

Metabolic reactions that replenish the supply of intermediates involved in the citric acid cycle.

GPX

A family of peroxidases that use reduced glutathione as their cofactor to reduce hydroperoxide species to their corresponding alcohols.

System xc

An antiporter that imports cystine and exports glutamate; it consists of two subunits, including the transporter subunit SLC7A11 and the regulatory subunit SLC3A2.

Ubiquinone

Also known as coenzyme Q or CoQ. A lipophilic molecule that is composed of a quinone head group linked to a polyisoprenoid lipid tail and acts as an electron transport carrier in mitochondria.

Ubiquinol

(CoQH2). The fully reduced form of ubiquinone.

Dihydroorotate dehydrogenase

(DHODH). An inner mitochondrial membrane-localized enzyme that oxidizes dihydroorotate to orotate for pyrimidine synthesis while reducing CoQ to CoQH2.

Hypercholesterolaemia

High levels of cholesterol in the blood.

Lipid droplets

Organelles with a phospholipid monolayer that are responsible for lipid storage, including PUFA storage.

Iron–sulfur clusters

(ISCs). Molecular ensembles of iron and sulfur that function as protein co-factors to regulate iron homeostasis and redox reactions in response to oxidative stress.

Mevalonate pathway

A metabolic pathway that synthesizes cholesterol, CoQ and steroid hormones.

Epithelial-to-mesenchymal transition

(EMT). A process by which epithelial cells gradually lose their cell polarity and intercellular adhesion properties and acquire mesenchymal-like phenotypes, including migratory and invasive properties.

Cyst(e)inase

Engineered enzymes that degrade extracellular cysteine and cystine.

Transsulfuration pathway

A metabolic pathway that transfers sulfur from homocysteine to cysteine, leading to cysteine biosynthesis.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lei, G., Zhuang, L. & Gan, B. Targeting ferroptosis as a vulnerability in cancer. Nat Rev Cancer 22, 381–396 (2022). https://doi.org/10.1038/s41568-022-00459-0

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41568-022-00459-0

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer