Journal Home > Volume 1 , Issue 1

Ferroptosis is a newly discovered form of cell death that is characterized by the accumulation of iron‐dependent lipid peroxidation. Research on ferroptosis has seen exponential growth over the past few years. Tumor cells are strongly dependent on iron for their growth, which makes them develop mechanisms to increase iron uptake and inhibit iron output, thereby completing iron accumulation. Ferroptosis can be induced or inhibited by various stresses through multiple mechanisms, making it stands at the crossroads of stresses related cancer cell fate determination. In this review, we give a brief summary of ferroptosis hallmarks and provide a systematic analysis of the current molecular mechanisms and regulatory networks of diverse stress conditions on ferroptosis. We also discuss the relationships between ferroptosis and cancer therapy responses to further understand potential targets and therapeutic strategies for cancer treatment.


menu
Abstract
Full text
Outline
About this article

Crosstalk between ferroptosis and stress—Implications in cancer therapeutic responses

Show Author's information Cheng Zhang1Jiao‐jiao Yu1Chen Yang1Shuang Shang1Xiao‐xi Lv1Bing Cui1( )Fang Hua1 ( )
CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China

Abstract

Ferroptosis is a newly discovered form of cell death that is characterized by the accumulation of iron‐dependent lipid peroxidation. Research on ferroptosis has seen exponential growth over the past few years. Tumor cells are strongly dependent on iron for their growth, which makes them develop mechanisms to increase iron uptake and inhibit iron output, thereby completing iron accumulation. Ferroptosis can be induced or inhibited by various stresses through multiple mechanisms, making it stands at the crossroads of stresses related cancer cell fate determination. In this review, we give a brief summary of ferroptosis hallmarks and provide a systematic analysis of the current molecular mechanisms and regulatory networks of diverse stress conditions on ferroptosis. We also discuss the relationships between ferroptosis and cancer therapy responses to further understand potential targets and therapeutic strategies for cancer treatment.

Keywords: stress, ferroptosis, tumor therapeutic response, crosstalk

References(159)

Fulda S, Gorman AM, Hori O, Samali A. Cellular stress responses: cell survival and cell death. Int J Cell Biol. 2010;2010:214074.
Galluzzi L, Vitale I, Aaronson SA, Abrams JM, Adam D, Agostinis P, et al. Molecular mechanisms of cell death: recommendations of the Nomenclature Committee on Cell Death 2018. Cell Death Differ. 2018;25:486–541.
Milisav I, Poljsak B, Ribaric S. Reduced risk of apoptosis: mechanisms of stress responses. Apoptosis. 2017;22:265–83.
Murrow L, Debnath J. Autophagy as a stress‐response and quality‐control mechanism: implications for cell injury and human disease. Annu Rev Pathol. 2013;8:105–37.
Song X, Liu J, Kuang F, Chen X, Zeh HJ 3rd, Kang R, et al. PDK4 dictates metabolic resistance to ferroptosis by suppressing pyruvate oxidation and fatty acid synthesis. Cell Rep. 2021;34:108767.
Dierge E, Debock E, Guilbaud C, Corbet C, Mignolet E, Mignard L, et al. Peroxidation of n‐3 and n‐6 polyunsaturated fatty acids in the acidic tumor environment leads to ferroptosis‐mediated anticancer effects. Cell Metab. 2021;33:e5–1715.
Lee H, Zandkarimi F, Zhang Y, Meena JK, Kim J, Zhuang L, et al. Energy‐stress‐mediated AMPK activation inhibits ferroptosis. Nat Cell Biol. 2020;22:225–34.
Zou Y, Palte MJ, Deik AA, Li H, Eaton JK, Wang W, et al. A GPX4‐dependent cancer cell state underlies the clear‐cell morphology and confers sensitivity to ferroptosis. Nat Commun. 2019;10:1617.
Yi J, Zhu J, Wu J, Thompson CB, Jiang X. Oncogenic activation of PI3K‐AKT‐mTOR signaling suppresses ferroptosis via SREBP‐mediated lipogenesis. Proc Natl Acad Sci USA. 2020;117:31189–97.
Drayton RM, Dudziec E, Peter S, Bertz S, Hartmann A, Bryant HE, et al. Reduced expression of miRNA‐27a modulates cisplatin resistance in bladder cancer by targeting the cystine/glutamate exchanger SLC7A11. Clin Cancer Res. 2014;20:1990–2000.
Shibata Y, Yasui H, Higashikawa K, Miyamoto N, Kuge Y. Erastin, a ferroptosis‐inducing agent, sensitized cancer cells to X‐ray irradiation via glutathione starvation in vitro and in vivo. PLoS One. 2019;14:e0225931.
Wang W, Green M, Choi JE, Gijón M, Kennedy PD, Johnson JK, et al. CD8(+) T cells regulate tumour ferroptosis during cancer immunotherapy. Nature. 2019;569:270–4.
Dixon SJ, Lemberg KM, Lamprecht MR, Skouta R, Zaitsev EM, Gleason CE, et al. Ferroptosis: an iron‐dependent form of nonapoptotic cell death. Cell. 2012;149:1060–72.
Hassannia B, Vandenabeele P, Vanden Berghe T. Targeting ferroptosis to iron out cancer. Cancer Cell. 2019;35:830–49.
Dolma S, Lessnick SL, Hahn WC, Stockwell BR. Identification of genotype‐selective antitumor agents using synthetic lethal chemical screening in engineered human tumor cells. Cancer Cell. 2003;3:285–96.
Yagoda N, von Rechenberg M, Zaganjor E, Bauer AJ, Yang WS, Fridman DJ, et al. RAS‐RAF‐MEK‐dependent oxidative cell death involving voltage‐dependent anion channels. Nature. 2007;447:864–8.
Yang WS, Stockwell BR. Synthetic lethal screening identifies compounds activating iron‐dependent, nonapoptotic cell death in oncogenic‐RAS‐harboring cancer cells. Chem Biol. 2008;15:234–45.
Gao M, Monian P, Quadri N, Ramasamy R, Jiang X. Glutaminolysis and transferrin regulate ferroptosis. Mol Cell. 2015;59:298–308.
Shi H, Bencze KZ, Stemmler TL, Philpott CC. A cytosolic iron chaperone that delivers iron to ferritin. Science. 2008;320:1207–10.
Protchenko O, Baratz E, Jadhav S, Li F, Shakoury‐Elizeh M, Gavrilova O, et al. Iron chaperone poly rC binding protein 1 protects mouse liver from lipid peroxidation and steatosis. Hepatology. 2021;73:1176–93.
Mancias JD, Wang X, Gygi SP, Harper JW, Kimmelman AC. Quantitative proteomics identifies NCOA4 as the cargo receptor mediating ferritinophagy. Nature. 2014;509:105–9.
Quiles Del Rey M, Mancias JD. NCOA4‐mediated ferritinophagy: a potential link to neurodegeneration. Front Neurosci. 2019;13:238.
Gao M, Monian P, Pan Q, Zhang W, Xiang J, Jiang X. Ferroptosis is an autophagic cell death process. Cell Res. 2016;26:1021–32.
Hadian K, Stockwell BR. SnapShot: ferroptosis. Cell. 2020;181:1188.
Hou W, Xie Y, Song X, Sun X, Lotze MT, Zeh HJ, et al. Autophagy promotes ferroptosis by degradation of ferritin. Autophagy. 2016;12:1425–8.
Alvarez SW, Sviderskiy VO, Terzi EM, Papagiannakopoulos T, Moreira AL, Adams S, et al. NFS1 undergoes positive selection in lung tumours and protects cells from ferroptosis. Nature. 2017;551:639–43.
Abeysinghe RD, Roberts PJ, Cooper CE, MacLean KH, Hider RC, Porter JB. The environment of the lipoxygenase iron binding site explored with novel hydroxypyridinone iron chelators. J Biol Chem. 1996;271:7965–72.
Chen X, Yu C, Kang R, Tang D. Iron metabolism in ferroptosis. Front Cell Dev Biol. 2020;8:590226.
Yang WS, SriRamaratnam R, Welsch ME, Shimada K, Skouta R, Viswanathan VS, et al. Regulation of ferroptotic cancer cell death by GPX4. Cell. 2014;156:317–31.
Yang WS, Kim KJ, Gaschler MM, Patel M, Shchepinov MS, Stockwell BR. Peroxidation of polyunsaturated fatty acids by lipoxygenases drives ferroptosis. Proc Natl Acad Sci USA. 2016;113:E4966–75.
Hayano M, Yang WS, Corn CK, Pagano NC, Stockwell BR. Loss of cysteinyl‐tRNA synthetase (CARS) induces the transsulfuration pathway and inhibits ferroptosis induced by cystine deprivation. Cell Death Differ. 2016;23:270–8.
Kagan VE, Mao G, Qu F, Angeli JP, Doll S, Croix CS, et al. Oxidized arachidonic and adrenic PEs navigate cells to ferroptosis. Nat Chem Biol. 2017;13:81–90.
Son J, Lyssiotis CA, Ying H, Wang X, Hua S, Ligorio M, et al. Glutamine supports pancreatic cancer growth through a KRAS‐regulated metabolic pathway. Nature. 2013;496:101–5.
Zhou B, Liu J, Kang R, Klionsky DJ, Kroemer G, Tang D. Ferroptosis is a type of autophagy‐dependent cell death. Semin Cancer Biol. 2020;66:89–100.
Zhu J, Berisa M, Schworer S, Qin W, Cross JR, Thompson CB. Transsulfuration activity can support cell growth upon extracellular cysteine limitation. Cell Metab. 2019;30:865–76.
Yao P, Fox PL. Aminoacyl‐tRNA synthetases in medicine and disease. EMBO Mol Med. 2013;5:332–43.
Bersuker K, Hendricks JM, Li Z, Magtanong L, Ford B, Tang PH, et al. The CoQ oxidoreductase FSP1 acts parallel to GPX4 to inhibit ferroptosis. Nature. 2019;575:688–92.
Doll S, Freitas FP, Shah R, Aldrovandi M, da Silva MC, Ingold I, et al. FSP1 is a glutathione‐independent ferroptosis suppressor. Nature. 2019;575:693–8.
Roh JL, Kim EH, Jang H, Shin D. Nrf2 inhibition reverses the resistance of cisplatin‐resistant head and neck cancer cells to artesunate‐induced ferroptosis. Redox Biol. 2017;11:254–62.
Sun X, Ou Z, Chen R, Niu X, Chen D, Kang R, et al. Activation of the p62‐Keap1‐NRF2 pathway protects against ferroptosis in hepatocellular carcinoma cells. Hepatology. 2016;63:173–84.
Ma S, Dielschneider RF, Henson ES, Xiao W, Choquette TR, Blankstein AR, et al. Ferroptosis and autophagy induced cell death occur independently after siramesine and lapatinib treatment in breast cancer cells. PLoS One. 2017;12:e0182921.
Garten A, Grohmann T, Kluckova K, Lavery GG, Kiess W, Penke M. Sorafenib‐induced apoptosis in hepatocellular carcinoma is reversed by SIRT1. Int J Mol Sci. 2019;20:4048.
Lachaier E, Louandre C, Godin C, Saidak Z, Baert M, Diouf M, et al. Sorafenib induces ferroptosis in human cancer cell lines originating from different solid tumors. Anticancer Res. 2014;34:6417–22.
Jiang L, Kon N, Li T, Wang SJ, Su T, Hibshoosh H, et al. Ferroptosis as a p53‐mediated activity during tumour suppression. Nature. 2015;520:57–62.
Xie Y, Zhu S, Song X, Sun X, Fan Y, Liu J, et al. The tumor suppressor p53 limits ferroptosis by blocking DPP4 Activity. Cell Rep. 2017;20:1692–704.
Aubrey BJ, Kelly GL, Janic A, Herold MJ, Strasser A. How does p53 induce apoptosis and how does this relate to p53‐mediated tumour suppression?Cell Death Differ. 2018;25:104–3.
Kubbutat MH, Jones SN, Vousden KH. Regulation of p53 stability by Mdm2. Nature. 1997;387:299–303.
Lavin MF, Gueven N. The complexity of p53 stabilization and activation. Cell Death Differ. 2006;13:941–50.
Mihara M, Erster S, Zaika A, Petrenko O, Chittenden T, Pancoska P, et al. p53 has a direct apoptogenic role at the mitochondria. Mol Cell. 2003;11:577–90.
Kruse JP, Gu W. Modes of p53 regulation. Cell. 2009;137:609–22.
Mrakovcic M, Frohlich LF. p53‐mediated molecular control of autophagy in tumor cells. Biomolecules. 2018;8:14.
Kuang F, Liu J, Tang D, Kang R. Oxidative damage and antioxidant defense in ferroptosis. Front Cell Dev Biol. 2020;8:586578.
Sullivan MR, Vander Heiden MG. Determinants of nutrient limitation in cancer. Crit Rev Biochem Mol Biol. 2019;54:193–207.
Qi Y, Zhang X, Wu Z, Tian M, Chen F, Guan W, et al. Ferroptosis regulation by nutrient signalling. Nutr Res Rev. 2021​:1–13.
Dixon SJ, Patel DN, Welsch M, Skouta R, Lee ED, Hayano M, et al. Pharmacological inhibition of cystine‐glutamate exchange induces endoplasmic reticulum stress and ferroptosis. eLife. 2014;3:e02523.
Goji T, Takahara K, Negishi M, Katoh H. Cystine uptake through the cystine/glutamate antiporter xCT triggers glioblastoma cell death under glucose deprivation. J Biol Chem. 2017;292:19721–32.
Koppula P, Zhang Y, Shi J, Li W, Gan B. The glutamate/cystine antiporter SLC7A11/xCT enhances cancer cell dependency on glucose by exporting glutamate. J Biol Chem. 2017;292:14240–9.
Shin CS, Mishra P, Watrous JD, Carelli V, D'Aurelio M, Jain M, et al. The glutamate/cystine xCT antiporter antagonizes glutamine metabolism and reduces nutrient flexibility. Nat Commun. 2017;8:15074.
Liu X, Olszewski K, Zhang Y, Lim EW, Shi J, Zhang X, et al. Cystine transporter regulation of pentose phosphate pathway dependency and disulfide stress exposes a targetable metabolic vulnerability in cancer. Nat Cell Biol. 2020;22:476–86.
Koppula P, Zhuang L, Gan B. Cystine transporter SLC7A11/xCT in cancer: ferroptosis, nutrient dependency, and cancer therapy. Protein Cell. 2021;12:599–620.
Kang YP, Mockabee‐Macias A, Jiang C, Falzone A, Prieto‐Farigua N, Stone E, et al. Non‐canonical glutamate‐cysteine ligase activity protects against ferroptosis. Cell Metab. 2021;33:174–89.
Perez MA, Magtanong L, Dixon SJ, Watts JL. Dietary lipids induce ferroptosis in caenorhabditiselegans and human cancer cells. Dev Cell. 2020;54:447–54.
Magtanong L, Ko PJ, To M, Cao JY, Forcina GC, Tarangelo A, et al. Exogenous monounsaturated fatty acids promote a ferroptosis‐resistant cell state. Cell Chem Biol. 2019;26:420–32.
Du J, Zhou Y, Li Y, Xia J, Chen Y, Chen S, et al. Identification of Frataxin as a regulator of ferroptosis. Redox Biol. 2020;32:101483.
Brown CW, Amante JJ, Chhoy P, Elaimy AL, Liu H, Zhu LJ, et al. Prominin2 drives ferroptosis resistance by stimulating iron export. Dev Cell. 2019;51:575–86.
Yang L, Wang H, Yang X, Wu Q, An P, Jin X, et al. Auranofin mitigates systemic iron overload and induces ferroptosis via distinct mechanisms. Signal Transduct Target Ther. 2020;5:138.
Meng P, Zhang S, Jiang X, Cheng S, Zhang J, Cao X, et al. Arsenite induces testicular oxidative stress in vivo and in vitro leading to ferroptosis. Ecotoxicol Environ Saf. 2020;194:110360.
Stockwell BR, Friedmann Angeli JP, Bayir H, Bush AI, Conrad M, Dixon SJ, et al. Ferroptosis: a regulated cell death nexus linking metabolism, redox biology, and disease. Cell. 2017;171:273–85.
Cardoso BR, Hare DJ, Bush AI, Roberts BR. Glutathione peroxidase 4: a new player in neurodegeneration?Mol Psychiatry. 2017;22:328–5.
Yao Y, Chen Z, Zhang H, Chen C, Zeng M, Yunis J, et al. Selenium‐GPX4 axis protects follicular helper T cells from ferroptosis. Nat Immunol. 2021;22:1127–39.
Hu Q, Zhang Y, Lou H, Ou Z, Liu J, Duan W, et al. GPX4 and vitamin E cooperatively protect hematopoietic stem and progenitor cells from lipid peroxidation and ferroptosis. Cell Death Dis. 2021;12:706.
Richter K, Haslbeck M, Buchner J. The heat shock response: life on the verge of death. Mol Cell. 2010;40:253–66.
Sun X, Ou Z, Xie M, Kang R, Fan Y, Niu X, et al. HSPB1 as a novel regulator of ferroptotic cancer cell death. Oncogene. 2015;34:5617–25.
Wang N, Ma H, Li J, Meng C, Zou J, Wang H, et al. HSF1 functions as a key defender against palmitic acid‐induced ferroptosis in cardiomyocytes. J Mol Cell Cardiol. 2021;150:65–76.
Distéfano AM, Martin MV, Córdoba JP, Bellido AM, D'ippólito S, Colman SL, et al. Heat stress induces ferroptosis‐like cell death in plants. J Cell Biol. 2017;216:463–76.
Hattori K, Naguro I, Okabe K, Funatsu T, Furutani S, Takeda K, et al. ASK1 signalling regulates brown and beige adipocyte function. Nat Commun. 2016;7:11158.
Gon Y, Hashimoto S, Matsumoto K, Nakayama T, Takeshita I, Horie T. Cooling and rewarming‐induced IL‐8 expression in human bronchial epithelial cells through p38 MAP kinase‐dependent pathway. Biochem Biophys Res Commun. 1998;249:156–60.
Hattori K, Ishikawa H, Sakauchi C, Takayanagi S, Naguro I, Ichijo H. Cold stress‐induced ferroptosis involves the ASK1‐p38 pathway. EMBO Rep. 2017;18:2067–78.
Burchfield SR, Woods SC, Elich MS. Effects of cold stress on tumor growth. Physiol Behav. 1978;21:537–40.
Sun Y, Zhao D, Wang G, Wang Y, Cao L, Sun J, et al. Recent progress of hypoxia‐modulated multifunctional nanomedicines to enhance photodynamic therapy: opportunities, challenges, and future development. Acta Pharm Sin B. 2020;10:1382–96.
Fuhrmann DC, Mondorf A, Beifuss J, Jung M, Brune B. Hypoxia inhibits ferritinophagy, increases mitochondrial ferritin, and protects from ferroptosis. Redox Biol. 2020;36:101670.
Li Z, Jiang L, Chew SH, Hirayama T, Sekido Y, Toyokuni S. Carbonic anhydrase 9 confers resistance to ferroptosis/apoptosis in malignant mesothelioma under hypoxia. Redox Biol. 2019;26:101297.
Kenny DJ, Plichta DR, Shungin D, Koppel N, Hall AB, Fu B, et al. Cholesterol metabolism by uncultured human gut bacteria influences host cholesterol level. Cell Host Microbe. 2020;28:245–57.
Yang M, Chen P, Liu J, Zhu S, Kroemer G, Klionsky DJ, et al. Clockophagy is a novel selective autophagy process favoring ferroptosis. Sci Adv. 2019;5:eaaw2238.
Gatenby RA, Gillies RJ. Why do cancers have high aerobic glycolysis?Nat Rev Cancer. 2004;4:891–9.
Green DR, Galluzzi L, Kroemer G. Cell biology. Metabolic control of cell death. Science. 2014;345:1250256.
Hardie DG, Ross FA, Hawley SA. AMPK: a nutrient and energy sensor that maintains energy homeostasis. Nat Rev Mol Cell Biol. 2012;13:251–62.
Herzig S, Shaw RJ. AMPK: guardian of metabolism and mitochondrial homeostasis. Nat Rev Mol Cell Biol. 2018;19:121–35.
Li C, Dong X, Du W, Shi X, Chen K, Zhang W, et al. LKB1‐AMPK axis negatively regulates ferroptosis by inhibiting fatty acid synthesis. Signal Transduct Target Ther. 2020;5:187.
Song X, Zhu S, Chen P, Hou W, Wen Q, Liu J, et al. AMPK‐mediated BECN1 phosphorylation promotes ferroptosis by directly blocking system Xc(‐) activity. Curr Biol. 2018;28:2388–99.
Zhao Y, Li M, Yao X, Fei Y, Lin Z, Li Z, et al. HCAR1/MCT1 regulates tumor ferroptosis through the lactate‐mediated AMPK‐SCD1 activity and its therapeutic implications. Cell Rep. 2020;33:108487.
Herrmann AG, Deighton RF, Le Bihan T, McCulloch MC, Searcy JL, Kerr LE, et al. Adaptive changes in the neuronal proteome: mitochondrial energy production, endoplasmic reticulum stress, and ribosomal dysfunction in the cellular response to metabolic stress. J Cereb Blood Flow Metab. 2013;33:673–83.
Yang D, Wang Q, Wei G, Wu J, Zhu YC, Zhu Q, et al. Smyd3‐PARP16 axis accelerates unfolded protein response and vascular aging. Aging. 2020;12:21423–45.
Lee YS, Lee DH, Choudry HA, Bartlett DL, Lee YJ. Ferroptosis‐induced endoplasmic reticulum stress: cross‐talk between ferroptosis and apoptosis. Mol Cancer Res. 2018;16:1073–6.
Lee AS. Glucose‐regulated proteins in cancer: molecular mechanisms and therapeutic potential. Nat Rev Cancer. 2014;14:263–76.
Zhu S, Zhang Q, Sun X, Zeh HJ 3rd, Lotze MT, Kang R, et al. HSPA5 regulates ferroptotic cell death in cancer cells. Cancer Res. 2017;77:2064–77.
Hu K, Li K, Lv J, Feng J, Chen J, Wu H, et al. Suppression of the SLC7A11/glutathione axis causes synthetic lethality in KRAS‐mutant lung adenocarcinoma. J Clin Invest. 2020;130:1752–66.
Poursaitidis I, Wang X, Crighton T, Labuschagne C, Mason D, Cramer SL, et al. Oncogene‐selective sensitivity to synchronous cell death following modulation of the amino acid nutrient cystine. Cell Rep. 2017;18:2547–56.
Qin Y, Pei Z, Feng Z, Lin P, Wang S, Li Y, et al. Oncogenic activation of YAP signaling sensitizes ferroptosis of hepatocellular carcinoma via ALOXE3‐mediated lipid peroxidation accumulation. Front Cell Dev Biol. 2021;9:751593.
Hao S, Yu J, He W, Huang Q, Zhao Y, Liang B, et al. Cysteine dioxygenase 1 mediates erastin‐induced ferroptosis in human gastric cancer cells. Neoplasia. 2017;19:1022–32.
Kang R, Kroemer G, Tang D. The tumor suppressor protein p53 and the ferroptosis network. Free Radic Biol Med. 2019;133:162–8.
Wang SJ, Li D, Ou Y, Jiang L, Chen Y, Zhao Y, et al. Acetylation is crucial for p53‐mediated ferroptosis and tumor suppression. Cell Rep. 2016;17:366–73.
Zhang Y, Shi J, Liu X, Feng L, Gong Z, Koppula P, et al. BAP1 links metabolic regulation of ferroptosis to tumour suppression. Nat Cell Biol. 2018;20:1181–92.
Wu J, Minikes AM, Gao M, Bian H, Li Y, Stockwell BR, et al. Intercellular interaction dictates cancer cell ferroptosis via NF2‐YAP signalling. Nature. 2019;572:402–6.
Ciccia A, Elledge SJ. The DNA damage response: making it safe to play with knives. Mol Cell. 2010;40:179–204.
Chen PH, Tseng WH, Chi JT. The intersection of DNA damage response and ferroptosis—a rationale for combination therapeutics. Biology. 2020;9:187.
Negrini S, Gorgoulis VG, Halazonetis TD. Genomic instability—an evolving hallmark of cancer. Nat Rev Mol Cell Biol. 2010;11:220–8.
Chen PH, Wu J, Ding CC, Lin CC, Pan S, Bossa N, et al. Kinome screen of ferroptosis reveals a novel role of ATM in regulating iron metabolism. Cell Death Differ. 2020;27:1008–22.
Li C, Zhang Y, Liu J, Kang R, Klionsky DJ, Tang D. Mitochondrial DNA stress triggers autophagy‐dependent ferroptotic death. Autophagy. 2021;17:948–60.
Kastenhuber ER, Lowe SW. Putting p53 in context. Cell. 2017;170:1062–78.
Marusyk A, Janiszewska M, Polyak K. Intratumor heterogeneity: the Rosetta Stone of therapy resistance. Cancer Cell. 2020;37:471–84.
Sato R, Semba T, Saya H, Arima Y. Concise review: stem cells and epithelial‐mesenchymal transition in cancer: biological implications and therapeutic targets. Stem Cells. 2016;34:1997–2007.
Byers LA, Diao L, Wang J, Saintigny P, Girard L, Peyton M, et al. An epithelial‐mesenchymal transition gene signature predicts resistance to EGFR and PI3K inhibitors and identifies Axl as a therapeutic target for overcoming EGFR inhibitor resistance. Clin Cancer Res. 2013;19:279–90.
Wu J, Chen J, Xi Y, Wang F, Sha H, Luo L, et al. High glucose induces epithelial‐mesenchymal transition and results in the migration and invasion of colorectal cancer cells. Exp Ther Med. 2018;16:222–30.
Ye LY, Chen W, Bai XL, Xu XY, Zhang Q, Xia XF, et al. Hypoxia‐induced epithelial‐to‐mesenchymal transition in hepatocellular carcinoma induces an immunosuppressive tumor microenvironment to promote metastasis. Cancer Res. 2016;76:818–30.
Zhang H, Deng T, Liu R, Ning T, Yang H, Liu D, et al. CAF secreted miR‐522 suppresses ferroptosis and promotes acquired chemo‐resistance in gastric cancer. Mol Cancer. 2020;19:43.
Mao C, Liu X, Zhang Y, Lei G, Yan Y, Lee H, et al. DHODH‐mediated ferroptosis defence is a targetable vulnerability in cancer. Nature. 2021;593:586–90.
Ding Y, Chen X, Liu C, Ge W, Wang Q, Hao X, et al. Identification of a small molecule as inducer of ferroptosis and apoptosis through ubiquitination of GPX4 in triple negative breast cancer cells. J Hematol Oncol. 2021;14:19.
Schmitt A, Xu W, Bucher P, Grimm M, Konantz M, Horn H, et al. Dimethyl fumarate induces ferroptosis and impairs NF‐kappaB/STAT3 signaling in DLBCL. Blood. 2021;138:871–4.
Chen TC, Chuang JY, Ko CY, Kao TJ, Yang PY, Yu CH, et al. AR ubiquitination induced by the curcumin analog suppresses growth of temozolomide‐resistant glioblastoma through disrupting GPX4‐mediated redox homeostasis. Redox Biol. 2020;30:101413.
Chen L, Li X, Liu L, Yu B, Xue Y, Liu Y. Erastin sensitizes glioblastoma cells to temozolomide by restraining xCT and cystathionine‐gamma‐lyase function. Oncol Rep. 2015;33:1465–74.
Ye J, Jiang X, Dong Z, Hu S, Xiao M. Low‐concentration PTX And RSL3 inhibits tumor cell growth synergistically by inducing ferroptosis in mutant p53 hypopharyngeal squamous carcinoma. Cancer Manag Res. 2019;11:9783–92.
Tagde A, Singh H, Kang MH, Reynolds CP. The glutathione synthesis inhibitor buthionine sulfoximine synergistically enhanced melphalan activity against preclinical models of multiple myeloma. Blood Cancer J. 2014;4:e229.
Madden EC, Gorman AM, Logue SE, Samali A. Tumour cell secretome in chemoresistance and tumour recurrence. Trends Cancer. 2020;6:489–505.
Kim EK, Jang M, Song MJ, Kim D, Kim Y, Jang HH. Redox‐mediated mechanism of chemoresistance in cancer cells. Antioxidants. 2019;8:471.
Xie Y, Hou W, Song X, Yu Y, Huang J, Sun X, et al. Ferroptosis: process and function. Cell Death Differ. 2016;23:369–79.
Chen Y, Li L, Lan J, Cui Y, Rao X, Zhao J, et al. CRISPR screens uncover protective effect of PSTK as a regulator of chemotherapy‐induced ferroptosis in hepatocellular carcinoma. Mol Cancer. 2022;21:11.
Yang C, Zhang Y, Lin S, Liu Y, Li W. Suppressing the KIF20A/NUAK1/Nrf2/GPX4 signaling pathway induces ferroptosis and enhances the sensitivity of colorectal cancer to oxaliplatin. Aging. 2021;13:13515–34.
Liu Q, Wang K. The induction of ferroptosis by impairing STAT3/Nrf2/GPx4 signaling enhances the sensitivity of osteosarcoma cells to cisplatin. Cell Biol Int. 2019;43:1245–56.
Tadokoro T, Ikeda M, Ide T, Deguchi H, Ikeda S, Okabe K, et al. Mitochondria‐dependent ferroptosis plays a pivotal role in doxorubicin cardiotoxicity. JCI Insight. 2020;5:e132747.
Guo J, Xu B, Han Q, Zhou H, Xia Y, Gong C, et al. Ferroptosis: a novel anti‐tumor action for cisplatin. Cancer Res Treat. 2018;50:445–60.
Deng S, Wu D, Li L, Li J, Xu Y. TBHQ attenuates ferroptosis against 5‐fluorouracil‐induced intestinal epithelial cell injury and intestinal mucositis via activation of Nrf2. Cell Mol Biol Lett. 2021;26:48.
Hu Z, Zhang H, Yi B, Yang S, Liu J, Hu J, et al. VDR activation attenuate cisplatin induced AKI by inhibiting ferroptosis. Cell Death Dis. 2020;11:73.
Maier P, Hartmann L, Wenz F, Herskind C. Cellular pathways in response to ionizing radiation and their targetability for tumor radiosensitization. Int J Mol Sci. 2016;17:102.
Azzam EI, Jay‐Gerin JP, Pain D. Ionizing radiation‐induced metabolic oxidative stress and prolonged cell injury. Cancer Lett. 2012;327:48–60.
Sleire L, Skeie BS, Netland IA, Førde HE, Dodoo E, Selheim F, et al. Drug repurposing: sulfasalazine sensitizes gliomas to gamma knife radiosurgery by blocking cystine uptake through system Xc‐, leading to glutathione depletion. Oncogene. 2015;34:5951–9.
Rodman SN, Spence JM, Ronnfeldt TJ, Zhu Y, Solst SR, O'Neill RA, et al. Enhancement of radiation response in breast cancer stem cells by inhibition of thioredoxin‐ and glutathione‐dependent metabolism. Radiat Res. 2016;186:385–95.
Lei G, Zhang Y, Koppula P, Liu X, Zhang J, Lin SH, et al. The role of ferroptosis in ionizing radiation‐induced cell death and tumor suppression. Cell Res. 2020;30:146–62.
Niu L, Liu L, Yang S, Ren J, Lai PBS, Chen GG. New insights into sorafenib resistance in hepatocellular carcinoma: responsible mechanisms and promising strategies. Biochim Biophys Acta, Rev Cancer. 2017;1868:564–70.
Sun X, Niu X, Chen R, He W, Chen D, Kang R, et al. Metallothionein‐1G facilitates sorafenib resistance through inhibition of ferroptosis. Hepatology. 2016;64:488–500.
Bai T, Lei P, Zhou H, Liang R, Zhu R, Wang W, et al. Sigma‐1 receptor protects against ferroptosis in hepatocellular carcinoma cells. J Cell Mol Med. 2019;23:7349–59.
Masoud V, Pages G. Targeted therapies in breast cancer: new challenges to fight against resistance. World J Clin Oncol. 2017;8:120–34.
Ma S, Henson ES, Chen Y, Gibson SB. Ferroptosis is induced following siramesine and lapatinib treatment of breast cancer cells. Cell Death Dis. 2016;7:e2307.
Lorenzato A, Magrì A, Matafora V, Audrito V, Arcella P, Lazzari L, et al. Vitamin C restricts the emergence of acquired resistance to EGFR‐targeted therapies in colorectal cancer. Cancers. 2020;12:685.
Tsoi J, Robert L, Paraiso K, Galvan C, Sheu KM, Lay J, et al. Multi‐stage differentiation defines melanoma subtypes with differential vulnerability to drug‐induced iron‐dependent oxidative stress. Cancer Cell. 2018;33:890–904.
Friedmann Angeli JP, Krysko DV, Conrad M. Ferroptosis at the crossroads of cancer‐acquired drug resistance and immune evasion. Nat Rev Cancer. 2019;19:405–14.
Lauber K, Bohn E, Kröber SM, Xiao YJ, Blumenthal SG, Lindemann RK, et al. Apoptotic cells induce migration of phagocytes via caspase‐3‐mediated release of a lipid attraction signal. Cell. 2003;113:717–30.
Rothe T, Gruber F, Uderhardt S, Ipseiz N, Rössner S, Oskolkova O, et al. 12/15‐Lipoxygenase‐mediated enzymatic lipid oxidation regulates DC maturation and function. J Clin Invest. 2015;125:1944–54.
Luo X, Gong HB, Gao HY, Wu YP, Sun WY, Li ZQ, et al. Oxygenated phosphatidylethanolamine navigates phagocytosis of ferroptotic cells by interacting with TLR2. Cell Death Differ. 2021;28:1971–89.
Wen Q, Liu J, Kang R, Zhou B, Tang D. The release and activity of HMGB1 in ferroptosis. Biochem Biophys Res Commun. 2019;510:278–83.
Song R, Li T, Ye J, Sun F, Hou B, Saeed M, et al. Acidity‐activatable dynamic nanoparticles boosting ferroptotic cell death for immunotherapy of cancer. Adv Mater. 2021;33:e2101155.
Kapralov AA, Yang Q, Dar HH, Tyurina YY, Anthonymuthu TS, Kim R, et al. Redox lipid reprogramming commands susceptibility of macrophages and microglia to ferroptotic death. Nat Chem Biol. 2020;16:278–90.
Jiang Q, Wang K, Zhang X, Ouyang B, Liu H, Pang Z, et al. Platelet membrane‐camouflaged magnetic nanoparticles for ferroptosis‐enhanced cancer immunotherapy. Small. 2020;16:e2001704.
Golstein P, Griffiths GM. An early history of T cell‐mediated cytotoxicity. Nat Rev Immunol. 2018;18:527–35.
Lang X, Green MD, Wang W, Yu J, Choi JE, Jiang L, et al. Radiotherapy and immunotherapy promote tumoral lipid oxidation and ferroptosis via synergistic repression of SLC7A11. Cancer Discov. 2019;9:1673–85.
Xu T, Ma Y, Yuan Q, Hu H, Hu X, Qian Z, et al. Enhanced ferroptosis by oxygen‐boosted phototherapy based on a 2‐in‐1 nanoplatform of ferrous hemoglobin for tumor synergistic therapy. ACS Nano. 2020;14:3414–25.
Wang D, DuBois RN. The role of prostaglandin E(2) in tumor‐associated immunosuppression. Trends Mol Med. 2016;22:1–3.
Zelenay S, van der Veen AG, Böttcher JP, Snelgrove KJ, Rogers N, Acton SE, et al. Cyclooxygenase‐dependent tumor growth through evasion of immunity. Cell. 2015;162:1257–70.
Shan X, Li S, Sun B, Chen Q, Sun J, He Z, et al. Ferroptosis‐driven nanotherapeutics for cancer treatment. J Control Release. 2020;319:322–32.
Publication history
Copyright
Acknowledgements
Rights and permissions

Publication history

Received: 22 February 2022
Accepted: 15 April 2022
Published: 11 June 2022
Issue date: June 2022

Copyright

© 2022 The Authors.

Acknowledgements

This study was supported by grants from the National Key R&D Program of China (2017YFA0205400), the National Natural Science Foundation of China (81973344 to FH; 81874316 to BC and 82173379 to JJY), CAMS Innovation Fund for Medical Sciences (2021‐I2M‐1‐021 to FH and SS), Peking Union Medical College Graduate Innovation Fund (2019‐1007‐24 to CZ), Central Public‐interest Scientific Institution Basal Research Fund (2018PT35004), and Beijing Outstanding Young Scientist Program (BJJWZYJH01201910023028). The authors sincerely apologize for not being able to keep as many important primary papers in the References because of strict space constraints.

Rights and permissions

This is an open access article under the terms of the Creative Commons Attribution‐NonCommercial‐NoDerivs License, which permits use and distribution in any medium, provided the original work is properly cited, the use is non‐commercial and no modifications or adaptations are made.

Return