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.
summary
In recent years, research in the field of ferroptosis in cancer has risen steeply and this is in part owing to its potential to be targeted. In this Review, Lei et al. provide an up-to-date synthesis of the roles and mechanisms of ferroptosis in tumour growth and progression including its function in tumour immunity, highlighting it as a vulnerability that can be exploited for cancer therapy.
Introduction
Ferroptosis, a term coined by Stockwell and colleagues in 20121, refers to an iron-dependent form of regulated cell death driven by an overload of lipid peroxides on cellular membranes. It is morphologically and mechanistically distinct from apoptosis and other types of regulated cell death; for example, morphologically, cells undergoing ferroptosis do not exhibit typical apoptotic features (such as chromatin condensation and apoptotic body formation), but are characterized by shrunken mitochondria and reduced numbers of mitochondrial cristae1,2. The lethal accumulation of lipid peroxides is a cardinal feature of ferroptosis3 and involves an antagonism between ferroptosis execution and ferroptosis defence systems in cells; ferroptosis occurs when ferroptosis-promoting cellular activities significantly override the antioxidant-buffering capabilities provided by ferroptosis defence systems 4–10 (Fig. 1a). This mechanistic feature is markedly different from several other forms of regulated cell death that centre on cell death executioner proteins (such as caspase-mediated apoptosis, mixed lineage kinase domain-like protein [MLKL]-mediated necroptosis, and gasdermin D-mediated pyroptosis)11. Finally, ferroptotic cells exhibit distinctive oxidized phospholipid (PL) profiles which differ from cells undergoing other forms of cell death12,13.
Figure 1. Ferroptosis-driving and -defence mechanisms.

a. Ferroptosis reflects an antagonism between prerequisites for ferroptosis and ferroptosis-defence systems. The prerequisites for ferroptosis consist of polyunsaturated fatty acid-containing phospholipid (PUFA-PL) synthesis and peroxidation, iron metabolism and mitochondrial metabolism. Ferroptosis-defence systems mainly include the glutathione peroxidase 4 (GPX4)–reduced glutathione (GSH) system, the ferroptosis suppressor protein-1 (FSP1)–ubiquinol (CoQH2) system, the dihydroorotate dehydrogenase (DHODH)–CoQH2 system, and the GTP cyclohydroxylase-1 (GCH1)– tetrahydrobiopterin (BH4) system. When ferroptosis-promoting cellular activities significantly exceed the detoxification capabilities provided by ferroptosis defence systems, a lethal accumulation of lipid peroxides on cellular membranes lead to subsequent membrane rupture and ferroptotic cell death.
b. Acyl-coenzyme A synthetase long chain family member 4 (ACSL4) and lysophosphatidylcholine acyltransferase 3 (LPCAT3) mediate the synthesis of PUFA-PLs, which are susceptible to peroxidation through both nonenzymatic and enzymatic mechanisms. Iron initiates the nonenzymatic Fenton reaction and acts as an essential cofactor for arachidonate lipoxygenases (ALOXs) and cytochrome P450 oxidoreductase (POR), which promote lipid peroxidation, and mitochondrial metabolism promotes the generation of reactive oxygen species (ROS), ATP, and/or PUFA-PLs. Excessive accumulation of lipid peroxides on cellular membranes can trigger ferroptosis. Cells have evolved at least 4 defence systems with different subcellular localizations to detoxify lipid peroxides and thus protect cells against ferroptosis, wherein cytosolic GPX4 (GPX4cyto) cooperates with FSP1 on the plasma membrane (and other non-mitochondrial membranes) and mitochondrial GPX4 (GPX4mito) with DHODH in the mitochondria to neutralize lipid peroxides. The subcellular compartment in which the GCH1–BH4 system operates remains to be defined. CoQ, coenzyme Q (also known as ubiquinone).
Ferroptosis as a unique cell death mechanism has sparked great interest in the cancer research community, as targeting ferroptosis might provide new therapeutic opportunities in treating cancers that are refractory to conventional therapies. In recent years, substantial progress has been achieved in understanding the role of ferroptosis in tumour biology and cancer therapy. On one hand, multiple cancer-associated signalling pathways have been shown to govern ferroptosis in cancer cells14. The engagement of ferroptosis in the activities of several tumour suppressors, such as p53 and BRCA1-associated protein 1 (BAP1), establishes ferroptosis as a natural barrier to cancer development15,16, whereas oncogene-mediated or oncogenic signalling-mediated ferroptosis evasion contributes to tumour initiation, progression, metastasis and therapeutic resistance17–19. On the other hand, the distinctive metabolism of cancer cells, their high load of reactive oxygen species (ROS), and their specific mutations render some of them intrinsically susceptible to ferroptosis, thereby exposing vulnerabilities that could be therapeutically targetable in certain cancer types20–24. Furthermore, some cancer cells appear to be particularly dependent on ferroptosis defence systems to survive under metabolic and oxidative stress conditions; consequently, disruption of those defences would be fatal to such cancer cells while sparing normal cells9. These recent data suggest that ferroptosis represents a targetable vulnerability of cancer in certain contexts. Ferroptosis has also been recognized as a critical cell death response triggered by a variety of cancer therapies, including radiotherapy (RT), immunotherapy, chemotherapy, and targeted therapies25–28. Thus, ferroptosis inducers (FINs) hold great potential in cancer therapy (Box 1), especially in combination with conventional therapies25,29,30.
Box 1. Ferroptosis inducers.
Ferroptosis inducers (FINs) are compounds or treatments that induce ferroptosis in cells, and can be categorized into at least four classes based on their mechanisms of action170. Class I FINs act by inhibiting solute carrier family 7 member 11 (SLC7A11)-mediated cystine uptake and restricting intracellular cysteine and glutathione levels82,170. Erastin is the most widely used class I FIN in cell culture studies, although its application in vivo is limited by its poor metabolic stability and water solubility170. Its analogue imidazole ketone erastin (IKE) has improved potency, solubility, and metabolic stability171, and has been shown to induce ferroptosis and suppress tumour growth in vivo 29. Sulfasalazine, a US Food and Drug Administration (FDA)-approved anti-inflammatory agent with class I FIN activity172,173, can inhibit tumour growth in diverse preclinical models97,173. Although sulfasalazine exhibits relatively poor metabolic stability and potency170, its activity in patients with glioblastoma has been demonstrated174 and its combination with radiotherapy is being tested in a clinical trial for glioblastoma (NCT04205357161). Sorafenib, another clinically approved agent is a multi-kinase inhibitor with activity against SLC7A11144; although it has been shown to induce ferroptosis, it also induces other nonferroptotic cellular effects144,170, and its classification as a FIN was challenged by a recent study175. In addition, cyst(e)inase degrades extracellular cystine and cysteine and has been shown to act as a potent and tolerable FIN to suppress tumour growth in vivo, when used either as a treatment alone or in combination with immune checkpoint inhibitors28,176.
Class II FINs operate by blocking the enzymatic activity of glutathione peroxidase 4 (GPX4), and include RSL3, ML162, and ML210. RSL3 and ML162 directly inhibit the catalytic activity of GPX4 by covalently binding to the selenocysteine residue of GPX4 through their electrophile chloroacetamide moiety4,165; however, their application in vivo is limited by their low solubility and poor pharmacokinetics165,170. ML210 is a nitroisoxazole-containing compound that is converted to α-nitroketoxime (known as JKE-1674) in cells and subsequently forms a nitrile-oxide electrophile, which covalently inhibits GPX4 with remarkable selectivity165. Notably, JKE-1674 exhibits improved pharmacokinetic properties165, although its effect on tumour growth remains to be investigated. Moreover, altretamine (an FDA-approved anticancer agent) and withaferin A (a natural anticancer agent suitable for in vivo treatment) have been shown to inhibit GPX4 activity or decrease GPX4 levels, providing an alternative for targeting GPX4 in vivo166,167.
Finally, class III FINs, such as FIN5636, act by depleting both GPX4 protein and ubiquinone. Class IV FINs, such as FINO2177, act by oxidizing iron and indirectly inactivating GPX4. However, these FINs have not been assessed in vivo.
With the exponential growth in ferroptosis research in the past 4 years, iterative insights into how to target ferroptosis in cancer are critically needed. In this Review, we summarize the current understanding of the regulatory networks of ferroptosis, including the prerequisites for ferroptosis and ferroptosis defence systems, thoroughly analyse the mechanistic bases of ferroptosis in tumour biology and synthesize a conceptual framework for how to target ferroptosis as a vulnerability in cancer therapy. Lastly, we highlight several key questions and challenges for future studies.
Ferroptosis prerequisites
The crux of ferroptosis execution is iron-catalysed peroxidation of polyunsaturated fatty acid (PUFA)-containing PLs (PUFA-PLs), which, when exceeding the buffering capability of ferroptosis defence systems (see the next section), can lead to lethal accumulation of lipid peroxides on cellular membranes and subsequent membrane rupture, resulting in ferroptotic cell death. As outlined in this section, PUFA-PL synthesis and peroxidation, iron metabolism, and mitochondrial metabolism constitute the main prerequisites driving ferroptosis3,31 (Fig. 1a).
PUFA-PL synthesis and peroxidation.
Acyl-coenzyme A (CoA) synthetase long chain family member 4 (ACSL4) and lysophosphatidylcholine acyltransferase 3 (LPCAT3) are critical mediators of PUFA-PL synthesis13,32,33 (Fig. 1b). ACSL4 catalyses the ligation of free PUFAs, such as arachidonic acids and adrenic acids, with CoA to generate PUFA-CoAs (such as arachidonic acid-CoA or adrenic acid-CoA)32,33, which are subsequently re-esterified and incorporated into PLs by LPCAT3 to form PUFA-PLs (such as arachidonic acid-phosphatidylethanolamine [PE] or adrenic acid-PE)13,32. Acetyl-CoA carboxylase (ACC)-catalysed carboxylation of acetyl-CoA generates malonyl-CoA, which is required for the synthesis of some PUFAs (likely at the elongation steps) and therefore for ferroptosis32,34–36. Inactivation of ACSL4, LPCAT3, or ACC blocks or attenuates ferroptosis13,33,37. Interestingly, p53-mediated ferroptosis, in some contexts, seems to be independent of ACSL438, although how ferroptosis occurs in the absence of ACSL4-mediated PUFA-PL synthesis remains unclear. Recent findings also revealed that PUFA-containing ether PLs (PUFA-ePLs) synthesized in peroxisomes act as additional substrates for lipid peroxidation, favouring ferroptosis onset23,39,40.
PUFA-PLs are particularly susceptible to peroxidation due to the presence of bis-allylic moieties in PUFAs41. The peroxidation of PUFA-PLs is primarily catalysed by nonenzymatic autoxidation driven by the Fenton reaction with iron as a catalyst41–43 (Fig. 1b). Cytochrome P450 oxidoreductase (POR)- or arachidonate lipoxygenase (ALOX)-mediated enzymatic reactions have also been shown to promote lipid peroxidation44–48; although the ability of POR to promote lipid peroxidation appears to be indirect via the generation of hydrogen peroxide (H2O2)46 and the role of ALOXs in ferroptosis has been challenged by other studies, including the lack of rescue of glutathione peroxidase 4 (Gpx4; part of a ferroptosis defence mechanism) knockout phenotypes by Alox15 deletion in mice49. Monounsaturated fatty acids (MUFAs), such as oleic acid and palmitoleic acid, are not readily peroxidised owing to their lack of bis-allylic moieties50. In contrast to PUFAs, MUFAs restrain lipid peroxidation and ferroptosis by displacing PUFAs from PLs in cellular membranes; inactivation of enzymes involved in MUFA-PL synthesis, such as stearoyl CoA desaturase 1 (SCD1) and ACSL3, sensitizes cancer cells to ferroptosis50–52.
Iron metabolism.
Iron governs ferroptosis not only by initiating the nonenzymatic Fenton reaction for direct peroxidation of PUFA-PLs41,43, but also by acting as an essential cofactor for enzymes that participate in lipid peroxidation (such as ALOX and POR)44,45. Cells normally maintain a relatively stable pool of labile iron through orchestrated regulation of iron uptake, utilization, storage, and export53. Imbalanced regulation of these iron metabolism processes can promote or suppress ferroptosis, depending on whether the level of the intracellular labile iron pool is increased or decreased, respectively. For example, intracellular iron is stored mostly as inert iron in ferritin, whereas autophagic degradation of ferritin (ferritinophagy) releases iron stored in ferritin into the labile iron pool; consequently, blockade of nuclear receptor coactivator 4 (NCOA4)-mediated ferritinophagy decreases the level of the labile iron pool and suppresses ferroptosis54,55. Conversely, enhancement of ferritinophagy by inhibition of cytosolic glutamate oxaloacetate transaminase 1 (GOT1) increases the labile iron pool and promotes ferroptosis (although exactly how GOT1 inhibition promotes ferritinophagy-mediated labile iron release remains to be further studied)56. We refer readers to a recent excellent review for detailed discussion of iron metabolism in ferroptosis53.
Mitochondrial metabolism.
Several metabolic processes in mitochondria have important roles in triggering ferroptosis31 (Fig. 1b). First, the generation of mitochondrial ROS is critical for lipid peroxidation and ferroptosis onset. Mitochondria are a major source of cellular ROS, in which electron leakage from electron transport chain complexes I and III generates superoxides, followed by their conversion to H2O2 by superoxide dismutase57. H2O2 can then react with labile iron via the Fenton reaction to generate hydroxyl radicals, which subsequently drive PUFA-PL peroxidation57,58. Moreover, electron transport and proton pumping in mitochondria are important for ATP production59,60, which also promotes ferroptosis35,37. Specifically, under ATP-depleted conditions, AMP-activated protein kinase (AMPK) phosphorylates and inactivates ACC, thereby suppressing PUFA-PL synthesis and blocking ferroptosis; in contrast, with sufficient energy (i.e., ATP), AMPK cannot be activated efficiently and ACC is activated, thus promoting PUFA-PL synthesis and ferroptosis35,37. Finally, the role of mitochondria in biosynthetic pathways in cellular metabolism also contributes to ferroptosis. Ferroptosis requires the tricarboxylic acid (TCA) cycle and various anaplerotic reactions that fuel the TCA cycle (such as glutaminolysis) in mitochondria59, which drive ferroptosis likely through promoting ROS, ATP, and/or PUFA-PL generation61–63. Therefore, the multifaceted functions of mitochondria in bioenergetics, biosynthesis, and ROS generation drive mitochondrial lipid peroxidation and ferroptosis31.
Ferroptosis defence mechanisms
Ferroptosis defence mechanisms involve cellular antioxidant systems that directly neutralize lipid peroxides. Studies of ferroptosis defence mechanisms have been one of the most exciting and rapidly evolving areas in ferroptosis research in the past 2 years. As discussed below, there are at least 4 such ferroptosis defence systems with distinctive subcellular localizations (Fig. 1).
The GPX4–GSH system.
GPX4 belongs to the GPX protein family64,65 and is the only GPX member capable of converting PL hydroperoxides to PL alcohols66,67. Genetic ablation or pharmacological inhibition of GPX4 induces unchecked lipid peroxidation and triggers potent ferroptosis under many in vitro and in vivo conditions4,49,66. GPX4 consists of 3 isoforms with distinctive subcellular localizations, namely cytosolic, mitochondrial, and nuclear GPX4s. These isoforms are encoded by the same GPX4 gene with different transcription initiation sites, resulting in the addition of a mitochondrial or nuclear localization sequence to the N-terminus of the GPX4 protein68–71. Until very recently, it was believed that only cytosolic GPX4 had a role in defending against ferroptosis, because cytosolic GPX4, but not the mitochondrial or nuclear isoform, is required for embryonic development72–75 and because reexpression of cytosolic GPX4, but not mitochondrial or nuclear GPX4, significantly suppresses cell death induced by Gpx4 deletion in mouse embryonic fibroblasts76. However, recent studies suggest that both cytosolic and mitochondrial GPX4 are important in defending against ferroptosis in different subcellular compartments (see below)9. The potential role of nuclear GPX4 in the regulation of ferroptosis remains to be studied.
Reduced glutathione (GSH), the cofactor used by GPX4, is a thiol-containing tripeptide that is derived from glycine, glutamate, and cysteine, with cysteine being the rate-limiting precursor77,78. Most cancer cells obtain intracellular cysteine primarily through system xc−-mediated uptake of cystine (an oxidized dimeric form of cysteine), followed by cystine reduction to cysteine in the cytosol79,80. Solute carrier family 7 member 11 (SLC7A11; also known as xCT) is the transporter subunit in system xc− 81,82. Removing cystine from culture media or pharmacologically blocking SLC7A11-mediated cystine transport with erastin or other FINs induces potent ferroptosis in many cancer cell lines1,15,16,82. The SLC7A11–GSH–GPX4 axis is believed to constitute the major cellular system defending against ferroptosis (Fig. 1b). However, upon GPX4 inactivation, some cancer cell lines remain resistant to ferroptosis22, suggesting the existence of additional ferroptosis defence mechanisms.
The FSP1–CoQH2 system.
While it was initially believed that GPX4 was the only ferroptosis defence system, this paradigm shifted following recent studies revealing that ferroptosis suppressor protein-1 (FSP1; also known as AIFM2) operates independently of GPX4 to defend against ferroptosis5,6. FSP1 localizes on the plasma membrane (as well as other subcellular compartments) and its plasma membrane localization appears to be both necessary and sufficient for the function of FSP1 in suppressing ferroptosis5,6. FSP1 functions as an NAD(P)H-dependent oxidoreductase capable of reducing ubiquinone (also known as coenzyme Q or CoQ)83,84 to ubiquinol (CoQH2)5,6 (Fig. 1b). Apart from its well-known function in mitochondrial electron transport, CoQH2 can also trap lipid peroxyl radicals, thereby suppressing lipid peroxidation and ferroptosis. Therefore, it has been proposed that FSP1 exerts its potent antiferroptosis activity through generating the nonmitochondrial CoQH2 pool as radical-trapping antioxidants5,6. It should be noted that CoQ is mainly synthesized in mitochondria83 but has been detected in nonmitochondrial membranes, including the plasma membrane85–88. The sources of the nonmitochondrial CoQ utilized by FSP1 for ferroptosis defence remain to be established.
The DHODH–CoQH2 system.
A recent study uncovered a mitochondria-localized defence system mediated by dihydroorotate dehydrogenase (DHODH) that can compensate for GPX4 loss to detoxify mitochondrial lipid peroxidation9. DHODH is an enzyme involved in pyrimidine synthesis that can reduce CoQ to CoQH2 in the inner mitochondrial membrane (Fig. 1b). When GPX4 is acutely inactivated, the flux through DHODH is significantly increased, resulting in enhanced CoQH2 generation that neutralises lipid peroxidation and defends against ferroptosis in mitochondria9. Consequently, inactivation of both mitochondrial GPX4 and DHODH unleashes potent mitochondrial lipid peroxidation and triggers robust ferroptosis. Importantly, while mitochondrial GPX4 and DHODH can compensate for each other to suppress mitochondrial lipid peroxidation, cytosolic GPX4 and FSP1 fail to do so9, ostensibly because they are not localized in mitochondria and therefore cannot detoxify lipid peroxides accumulated in the inner mitochondrial membrane, highlighting the importance of compartmentalization in ferroptosis defence.
These recent studies suggest a model whereby ferroptosis defence systems can be divided into 2 major divisions, the GPX4 system and the CoQH2 system, with each of these further subdivided into arms located in nonmitochondrial and mitochondrial compartments (i.e., cytosolic and mitochondrial GPX4 in the GPX4 system, versus FSP1 and DHODH in the CoQH2 system). The rationale for this division in ferroptosis defence is likely derived from the significant need to mitigate lipid peroxides generated in mitochondria (see subsection on mitochondrial metabolism) and the double-membrane structure of mitochondria (which prohibits cytosol- or other compartment-localized defence systems from entering into mitochondria). Further research is required to substantiate this compartmentalization model in ferroptosis regulation and to reconcile with some conflicting data from previous studies. For example, cytosolic GPX4 has also been found to significantly localize in the intermembrane space of mitochondria74. The abundance of cytosolic GPX4 in mitochondria and its potential role in suppressing lipid peroxidation within mitochondria remain to be clarified by future investigations.
Another open question concerns potential roles of other mitochondrial enzymes that produce CoQH2, most notably electron transport chain complexes I and II, in ferroptosis regulation. However, unlike DHODH inactivation, inactivation of complex I or II by pharmacological inhibition does not appear to affect GPX4 inactivation-induced ferroptosis or even promotes resistance to cystine starvation-induced ferroptosis62. Conceivably, the presumed antiferroptosis function of complex I or II (by producing mitochondrial CoQH2) might be offset by its proferroptosis functions (through producing ROS or ATP; see subsection on mitochondrial metabolism). The potential role of additional CoQH2-producing enzymes localized in mitochondria, such as electron transfer flavoprotein dehydrogenase (ETFDH), in ferroptosis regulation awaits further research.
The GCH1–BH4 system.
Recent studies identified GTP cyclohydroxylase-1 (GCH1) as another critical regulator of ferroptosis7,8. Tetrahydrobiopterin (BH4) is a cofactor of aromatic amino acid hydroxylases and other enzymes, and GCH1 mediates the rate-limiting reaction in the BH4 biosynthesis pathway89. BH4 is another radical-trapping antioxidant capable of trapping lipid peroxyl radicals, and its function in inhibiting ferroptosis appears to be independent of its role as a cofactor8. It was proposed that GCH1 suppresses ferroptosis through generating BH4 as a radical-trapping antioxidant as well as via GCH1-mediated production of CoQH2 and PLs containing two PUFA tails7,8 (whereas most PUFA-PLs tend to exhibit an asymmetric structure, with a PUFA tail at the sn-2 position and a saturated fatty acid (SFA) tail at the sn-1 position) (Fig. 1b). The subcellular compartment wherein the GCH1–BH4 system operates remains to be defined.
Ferroptosis evasion fuels tumours
Accumulating evidence indicates that ferroptosis is a critical tumour suppression mechanism. Several tumour-suppressor and oncogenic signalling pathways have been shown to promote or suppress ferroptosis, respectively (Boxes 2 and 3). Tumours have evolved at least 3 mechanisms to evade ferroptosis and to facilitate tumour development and metastasis, including limiting PUFA-PL synthesis and peroxidation, restricting labile iron availability, and upregulating cellular defence systems against ferroptosis.
Box 2. Ferroptosis regulation by tumour suppressors.
p53, the most frequently mutated tumour suppressor178, has an important role in regulating ferroptosis15,179. p53 inhibits solute carrier family 7 member 11 (SLC7A11) expression through binding directly to the SLC7A11 promoter or via interacting with ubiquitin-specific processing protease 7 (USP7) to reduce histone H2B monoubiquitination levels on the SLC7A11 promoter, sensitizing cancer cells to ferroptosis in an arachidonate 12-lipoxygenase (ALOX12)-dependent manner15,38,142,180. p53 can also promote ferroptosis by modulating additional metabolic targets181–183. Importantly, some p53 mutants cannot induce apoptosis, senescence or cell cycle arrest, yet can promote ferroptosis to mediate tumour suppression in vivo15. In contrast, other p53 mutants lacking ferroptosis regulatory activity lose their tumour-suppression function183,184. Note that p53 appears to play a dual role in ferroptosis regulation depending on the cellular context, as it can exert an antiferroptosis effect under certain conditions185,186.
The tumour suppressor BRCA1-associated protein 1 (BAP1) encodes a nuclear deubiquitinating enzyme (DUB) that reduces histone H2A ubiquitination (H2Aub) on chromatin187,188. BAP1 is frequently mutated in several sporadic cancers, including uveal melanoma, mesothelioma, and renal cell carcinoma (RCC)189. BAP1 reduces H2Aub occupancy on the SLC7A11 promoter and represses SLC7A11 expression in a DUB-dependent manner, thereby inhibiting cystine uptake and promoting ferroptosis, which partly mediates the tumour-suppressive effect of BAP1 in xenograft models16. In addition, the ability of BAP1 to restrain SLC7A11 expression and promote ferroptosis is compromised in cells with cancer-associated BAP1 mutations16.
Kelch-like ECH associated protein 1 (KEAP1) is a tumour suppressor frequently mutated in non-small-cell lung cancer190,191. It encodes a substrate adaptor that targets the transcription factor nuclear factor erythroid 2-related factor 2 (NRF2) for proteasomal degradation192,193. KEAP1 inactivation in cancer leads to NRF2 protein stabilization192,193. As a master regulator of antioxidant defence, NRF2 promotes the transcription of many genes governing ferroptosis suppression, such as SLC7A1199,101,194. In addition, ARF is a well-established tumour suppressor that is critical for p53 activation in response to oncogenic stress195. Interestingly, ARF was recently shown to inhibit NRF2 transcriptional activity independently of KEAP1 or p53100. In ARF-mutated tumours, NRF2 and its downstream target genes (such as SLC7A11) are activated, enabling cancer cells to evade ferroptosis under oxidative stress100. Therefore, ferroptosis activation by KEAP1 or ARF due to NRF2 suppression is likely a tumour-suppressive mechanism.
Most studies have focused on ferroptosis regulation mediated by tumour suppressors through SLC7A11. The control of ferroptosis by other tumour suppressors through other mechanisms remains to be explored.
Box 3. Ferroptosis regulation by oncogenes.
In line with the concept that ferroptosis is a natural tumour-suppressor mechanism, several oncogene-driven cancers have evolved mechanisms to escape from ferroptosis.
The PI3K oncogenic signalling pathway is one of the most frequently mutated pathways in human cancers196,197. It was recently demonstrated that cancer cells carrying PIK3CA activating mutations or PTEN deletion are generally resistant to ferroptosis17. Oncogenic activation of the PI3K signalling pathway promotes the activation of mTOR complex 1 (mTORC1)197, which inhibits ferroptosis through at least 3 parallel mechanisms198: promoting glutathione peroxidase 4 (GPX4) protein synthesis120, suppressing autophagy-dependent ferroptosis199 and boosting monounsaturated fatty acid-containing phospholipid (MUFA-PL) synthesis by increasing stearoyl coenzyme A desaturase 1 (SCD1) expression17. Thus, aberrant activation of mTORC1 allows cancer cells to evade ferroptosis, which might partly account for the oncogenic activity caused by mutations in the PI3K pathway.
The RAS family of proto-oncogenes are frequently mutated in human cancers200,201. Notably, RAS-mutant cancer cells are characterized by high levels of intracellular cysteine and glutathione in response to oxidative stress, via mechanisms involving solute carrier family 7 member 11 (SLC7A11) upregulation103. Furthermore, pharmacological or genetic inhibition of SLC7A11 severely impairs mutant KRAS-induced tumour growth, suggesting that SLC7A11 is required for mutant KRAS-mediated tumourigenesis102,103. Therefore, SLC7A11-mediated cystine uptake and glutathione biosynthesis allow mutant RAS-driven tumours to evade ferroptosis under oxidative stress. It is also worth mentioning that KRAS mutation in lung cancer also increases Acyl-coenzyme A synthetase long chain family member 3 (ACSL3) expression to reprogram lipid metabolism, promoting MUFA-PL biosynthesis and ferroptosis resistance, and that these processes might be important for the development of mutant KRAS-driven lung cancer50,202.
Several noncanonical oncogenes also contribute to the escape of cancer cells from ferroptosis. Neural precursor cell expressed, developmentally down-regulated 4 (NEDD4), a ubiquitin ligase with oncogenic functions203, can be induced by the ferroptosis inducer erastin169. Erastin-induced ferroptosis requires voltage-dependent anion channel 2/3 (VDAC2/3)204; interestingly, VDAC2/3 expression was reduced in a NEDD4-dependent manner upon erastin treatment, thereby restricting erastin-induced ferroptosis169. Other proteins with oncogenic activities, such as DJ1205 and pyruvate dehydrogenase kinase 4 (PDK4)206, can also promote ferroptosis evasion through corresponding mechanisms145,149 (see section on targeting ferroptosis in cancer therapy). In all of these studies, inactivation of NEDD4, DJ1 or PDK4 promoted ferroptosis and enhanced the tumour-suppressive effects of ferroptosis inducers in xenograft models145,149,169. Whether ferroptosis evasion supports the oncogenic roles of these proteins remains to be further explored.
Suppression of PUFA-PL synthesis and peroxidation.
Downregulation of peroxidised PUFA-PL levels in cancer cells has been linked to ferroptosis evasion and enhanced tumour growth. Calcium-independent phospholipase A2β (iPLA2β), a member of the iPLA2 family, is overexpressed in some human cancers. iPLA2β was recently shown to facilitate cancer cell escape from ferroptosis by hydrolysing peroxidised PLs, and iPLA2β depletion sensitized cancer cells to ferroptosis and impaired xenograft tumour growth90,91. Another recent study revealed that the expression of the adipokine chemerin is frequently upregulated in renal cell carcinoma (RCC), and that chemerin may maintain xenograft RCC growth through downregulating the levels of peroxidised PUFA-PLs and promoting ferroptosis evasion92.
Recent findings highlight that ferroptosis evasion through modulating fatty acid metabolism also contributes to cancer metastasis. Melanoma generally undergoes regional metastasis through the lymphatic system prior to systemic metastasis through the blood. The lymphatic environment has been observed to promote the escape of cancer cells from oxidative stress and ferroptosis in vivo, partly due to abundant levels of oleic acid (a MUFA) and GSH and low levels of free iron in the lymphatic fluid; oleic acid protects melanoma cells from ferroptosis in an ACSL3-dependent manner18 (as discussed in above, ACSL3-mediated MUFA-PL synthesis suppresses ferroptosis by displacing PUFAs from PLs). Importantly, ferroptosis resistance conferred by the lymphatic environment on cancer cells contributes to their subsequent survival during metastasis through the blood18. Likewise, under conditions of hypercholesterolemia, cancer cells exhibit superior metastatic capabilities in vivo, which may be attributable to ferroptosis resistance caused by increased accumulation of lipid droplets and MUFAs in these cells93.
Restriction of the labile iron pool.
As an important cofactor in redox maintenance and iron homeostasis, iron–sulfur clusters (ISCs) and the associated regulatory pathways contribute to ferroptosis evasion in cancer cells by diminishing the labile iron pool94. The cysteine desulfurase NFS1 is involved in ISC biosynthesis by harvesting sulfur from cysteine. NFS1 was found to be overexpressed in lung cancers and to be required for lung tumour growth in vivo, likely due to its ability to limit labile iron levels and protect cancer cells from ferroptosis95. Frataxin, another protein involved in ISC assembly, is upregulated in different cancer types and was shown to promote cancer cell resistance to ferroptosis and, thus, xenograft tumour growth96. Likewise, another recent study revealed that CDGSH iron-sulfur domain-containing protein 2 (CISD2), a component of the iron–sulfur protein family that is highly expressed in head and neck cancers, enhances the ability of cancer cells to counteract ferroptosis in vitro, likely by modulating ISCs and decreasing free iron levels97. In addition, when breast cancer cells detach from the extracellular matrix, they can upregulate prominin 2 expression to promote the formation of ferritin-containing multivesicular bodies that export iron out of cells, thereby facilitating ferroptosis evasion in vitro98. In some of the examples discussed above, whether ferroptosis evasion in cancer cells indeed contributes to enhanced tumour growth or metastasis in vivo remains to be established.
Upregulation of ferroptosis defences.
Most current studies of these mechanisms have centred on SLC7A11, which is overexpressed in multiple human cancer types79,82. As discussed in Boxes 2 and 3, inactivation of tumour suppressors such as p53, BAP1, kelch-like ECH associated protein 1 (KEAP1) and ARF (p14), or activation of oncogenic KRAS, upregulates SLC7A11 expression, which can be dependent or independent of nuclear factor erythroid 2-related factor 2 (NRF2), conferring ferroptosis evasion and promoting tumour growth15,16,99–103. As a master regulator of antioxidant defence, the transcription factor NRF2 governs the transcription of many genes involved in GPX4–GSH-mediated ferroptosis defence, including SLC7A11, thereby facilitating the escape of cancer cells from ferroptosis. Consequently, NRF2 signalling is upregulated in many human cancer types99,101,104. Interestingly, a recent study showed that treatment with 27-hydroxycholesterol (27-HC) leads to downregulation of GPX4 levels in breast cancer cells, and that this effect is lost in breast cancer cells resistant to 27-HC, which likely endows such 27-HC-resistant breast cancer cells with increased resistance to ferroptosis and enhanced metastatic capability in vivo93.
It appears that tumours also exploit other ferroptosis defence mechanisms. For example, GCH1 is overexpressed in multiple cancer types, including breast, lung, and liver cancers, and its high expression correlates with ferroptosis resistance in vitro7. FSP1- and DHODH-mediated ferroptosis defence centres on CoQ, whose synthesis is derived from the mevalonate pathway83. Squalene monooxygenase (SQLE) is a key enzyme in the mevalonate pathway. A lack of SQLE expression in anaplastic lymphoma kinase (ALK)+ anaplastic large-cell lymphoma cells prevents cholesterol synthesis (thereby rendering such cancer cells cholesterol auxotrophic) and leads to the accumulation of the upstream metabolite squalene, a lipophilic antioxidant that protects cancer cells from ferroptosis in vivo105. This mechanism provides a unique advantage for ALK+ anaplastic large-cell lymphoma growth under conditions of oxidative stress105.
Ferroptosis vulnerability
Oncogenic mutations rewire cellular metabolic networks in cancer cells to fulfil their increased demand for nutrients and energy106,107; this reprogramming often exposes new metabolic liabilities in cancer cells, rendering some of them uniquely vulnerable to ferroptosis. In this section, we conceptualize 3 such ferroptosis-based vulnerabilities in cancer cells, namely those induced by metabolic reprogramming, genetic mutations, and an imbalance in ferroptosis defences (Fig. 2a–c).
Figure 2. Ferroptosis as a vulnerability in cancer.

a. Therapy-resistant cancer cells with specific cellular states are vulnerable to ferroptosis, for example cancer cells with a mesenchymal phenotype that are enriched in polyunsaturated fatty acids (PUFAs) owing to high expression of zinc finger E-box binding homeobox 1 (ZEB1), elongation of very long-chain fatty acid protein 5 (ELOVL5) or fatty acid desaturase 1 (FADS1). Similarly, dedifferentiated subtypes of melanoma cells are characterized by PUFA accumulation and a deficiency of reduced glutathione (GSH), which render these cells vulnerable to ferroptosis. In addition, certain cancer cells, such as clear-cell renal cell carcinoma (ccRCC), non-neuroendocrine (NE) small-cell lung cancer (SCLC), and triple-negative breast cancer (TNBC) cells are inherently susceptible to ferroptosis owing to their unique metabolic features, such as high levels of PUFA-containing ether phospholipids (ePLs).
b. Mutations in certain tumour suppressors or oncogenes render cancers vulnerable to ferroptosis. Inactivating mutations in any constituent of the tumour-suppressive E-cadherin– neurofibromin 2 (NF2)–Hippo pathway confers a vulnerability to ferroptosis by upregulating Yes-associated protein (YAP)- or transcriptional coactivator with PDZ-binding motif (TAZ)-mediated transcription of ferroptosis-promoting factors, such as acyl-coenzyme A synthetase long chain family member 4 (ACSL4), transferrin receptor 1 (TfR1) and NADPH oxidase 4 (NOX4). In ccRCC, mutation or loss of Von Hippel-Lindau (VHL) promotes hypoxia inducible factor (HIF)-dependent expression of hypoxia-inducible, lipid droplet-associated protein (HILPDA), rendering ccRCCs vulnerable to ferroptosis. Non-small-cell lung cancers (NSCLCs) with epidermal growth factor receptor (EGFR) mutations are vulnerable to ferroptosis because of their high dependence on cystine. In another example, isocitrate dehydrogenase 1 (IDH1)-mutated cancer cells with increased levels of the oncometabolite 2-hydroxyglutarate (2-HG) are sensitive to ferroptosis owing to their decreased glutathione peroxidase 4 (GPX4) levels. * indicates either a mutation or loss of the gene, dependent on the gene.
c. Vulnerability to ferroptosis is triggered by an imbalance between GPX4-dependent and GPX4-independent ferroptosis defence systems. Cancer cells with low expression of components of GPX4-independent systems (such as ferroptosis suppressor protein-1 (FSP1), dihydroorotate dehydrogenase (DHODH) or GTP cyclohydroxylase-1 (GCH1)) depend on GPX4 for survival and therefore are vulnerable to GPX4 inhibition. Conversely, cancer cells with low expression of GPX4 are sensitive to inactivation of components of GPX4-independent systems. The dashed lines indicate that the function of the indicated protein is diminished or blocked in the corresponding context. PLs, phospholipids.
Metabolic features of cancer cells.
Several studies revealed that therapy-refractory cancer cells in specific cellular states are unexpectedly sensitive to ferroptosis21,22,108. For example, cancer cells in a mesenchymal state, which are usually resistant to apoptosis induced by conventional therapies, are strongly dependent on GPX4, and this dependency is associated with high expression of zinc finger E-box binding homeobox 1 (ZEB1), a driver of epithelial-to-mesenchymal transition (EMT) and lipogenic factor109. PUFA-PL synthesis is enhanced in cancer cells in the mesenchymal state, possibly owing to the central role of ZEB1 in lipid metabolism (Fig. 2a). The increased PUFA-PL levels in such cancer cells render them dependent on GPX4 to detoxify lipid peroxides for survival; consequently, these cells are highly vulnerable to ferroptosis22. Consistently, certain recurrent breast cancer cells with mesenchymal features exhibit upregulated discoidin domain-containing receptor 2 (DDR2) expression, which may be driven by EMT transcription factors; increased DDR2 promotes ferroptosis sensitivity in cancer cells through the Hippo pathway (the role of which in ferroptosis will be further discussed below)110. Likewise, key enzymes involved in PUFA synthesis, such as elongation of very long-chain fatty acid protein 5 (ELOVL5) and fatty acid desaturase 1 (FADS1), were found to be selectively overexpressed in mesenchymal gastric cancer cells, rendering these cancer cells particularly susceptible to ferroptosis111 (Fig. 2a).
Similar to therapy-resistant mesenchymal-type cancer cells that depend on GPX4, drug-tolerant persister cancer cells also are highly sensitive to ferroptosis, perhaps because they also exhibit mesenchymal-like features21. Interestingly, CD44, a transmembrane glycoprotein that is highly expressed in mesenchymal cancer cells, mediates hyaluronate-dependent iron endocytosis112, which might promote ferroptosis sensitivity in mesenchymal cancer cells through increasing cellular iron load. Moreover, therapy-resistant dedifferentiated subsets of melanoma cells also show a vulnerability to ferroptosis, possibly because of PUFA accumulation and low levels of GSH108 (Fig. 2a). In sum, therapy-resistant cancer cells often exhibit altered metabolic states (such as increased PUFA-PLs associated with EMT) that confer a vulnerability to ferroptosis induction.
Certain cancer types are also inherently susceptible to ferroptosis owing to other unique metabolic features (Fig. 2a). As described in a previous section, PUFA-ePLs synthesized in peroxisomes provide substrates for lipid peroxidation23. Clear-cell RCC (ccRCC) cells exhibit high levels of PUFA-ePLs, possibly owing to their high expression of alkylglycerone-phosphate synthase (AGPS, a key enzyme involved in PUFA-ePL synthesis), rendering these cells sensitive to ferroptosis23. Within small-cell lung cancer (SCLC), non-neuroendocrine SCLC cells are much more sensitive to ferroptosis than are neuroendocrine SCLC cells, at least partly because non-neuroendocrine SCLC cells overexpress ePL synthesis enzymes and have high levels of PUFA-ePLs113. Finally, triple-negative breast cancer (TNBC) cells were found to be susceptible to ferroptosis. This susceptibility has been ascribed to several metabolic features of these cells, including their abundant PUFA levels, elevated labile iron pool and attenuated GPX4–GSH defence system33,114. A vulnerability to ferroptosis uncovered in the abovementioned cancer types might provide new therapeutic opportunities for treating these highly intractable diseases.
Genetic mutations in cancer cells.
As discussed in Box 2, inactivation of tumour suppressors generally promotes ferroptosis resistance. However, in certain cases, tumour-suppressor mutations can confer an unexpected vulnerability to ferroptosis. The E-cadherin–neurofibromin 2 (NF2)–Hippo signalling axis presents a remarkable example of this type of vulnerability. Unlike other tumour suppressors (e.g., p53, BAP1, KEAP1 and ARF (p14), which promote ferroptosis; see Box 2), this tumour-suppressive axis suppresses ferroptosis20. E-cadherin-mediated intercellular interactions activate Hippo tumour-suppressive signalling in an NF2-dependent manner, repress the transcriptional activity of Yes-associated protein (YAP) or transcriptional coactivator with PDZ-binding motif (TAZ) and downregulate the expression of several ferroptosis-promoting factors, thereby suppressing ferroptosis (which clearly does not align with the tumour-suppressor function of NF2)20,115 (Fig. 2b). Consequently, inactivation of any component in the E-cadherin–NF2–Hippo pathway increases YAP or TAZ expression and/or activity, rendering cancer cells or tumours harbouring mutations in this pathway (such as NF2-mutant mesotheliomas) particularly susceptible to FINs20,115. The Von Hippel-Lindau (VHL) tumour suppressor is another such example. VHL is lost in the majority of ccRCCs116,117. Notably, VHL-deficient ccRCC cells are susceptible to ferroptosis, and restoration of wild-type VHL renders them insensitive to ferroptosis118. Several mechanisms might underlie this unique vulnerability in VHL-deficient ccRCCs, prominent among which is stabilization of hypoxia inducible factors (HIFs) induced by loss of VHL, which then promote PUFA synthesis through inducing the expression of hypoxia-inducible, lipid droplet-associated protein (HILPDA)24,118 (Fig. 2b).
Oncogene activation can also render cancer cells susceptible to ferroptosis. Non-small-cell lung cancer (NSCLC) cells with epidermal growth factor receptor (EGFR) mutations are highly dependent on cystine and sensitive to ferroptosis induced by SLC7A11 inhibition or cystine deprivation119 (Fig. 2b). Interestingly, cystine promotes cell survival in EGFR-mutant cancer cells through GSH-independent mechanisms119, which is in line with recent data showing that cyst(e)ine promotes GPX4 protein synthesis and suppresses ferroptosis partly via GSH-independent mechanisms119,120. As another example, isocitrate dehydrogenase 1 (IDH1)-mutated cancer cells exhibit increased levels of the oncometabolite 2-hydroxyglutarate (2-HG), which reduces GPX4 protein levels, sensitizing these cancer cells to ferroptosis121 (Fig. 2b). Collectively, it appears that oncogene activation and tumour-suppressor inactivation can either suppress or promote ferroptosis, depending on the context. Promotion of ferroptosis can create a vulnerability to ferroptosis for potential therapeutic targeting in corresponding cancer types.
Imbalanced ferroptosis defences.
Ferroptosis defence systems can be broadly divided into GPX4-dependent and GPX4-independent arms. Low expression or partial inactivation of one arm can render cancer cells highly dependent on the other for ferroptosis defence, and consequently highly susceptible to ferroptosis induced by the inactivation of the other defence arm (whereas normal cells with both arms intact might be unaffected by inhibition of one defence arm) (Fig. 2c). A therapeutic strategy targeting imbalanced ferroptosis defence would be similar to strategies targeting synthetic lethality; for instance, the use of poly(ADP-ribose) polymerase (PARP) inhibitors to treat BRCA1-deficient cancers, which is based on the concept that cells are dependent on 2 parallel DNA repair pathways, one involving PARP and the other requiring BRCA1122.
In support of this concept, analyses show that cancer cell lines with low expression of FSP1, DHODH or GCH1 are in general more vulnerable to GPX4 inhibitors than those with high expression of the corresponding gene (indeed, FSP1 was identified as the gene with the most striking such correlation in the Cancer Therapeutics Response Portal (CTRP))5–9. Furthermore, GPX4 genetic ablation reduced tumour growth more potently in FSP1 knockout xenograft tumours than in FSP1 wild-type counterparts, and this reduction in tumour growth was attributed to ferroptosis induction5. Due to the unsuitability of current GPX4 inhibitors for in vivo studies (see further discussion in Perspective), whether this observation also applies to GPX4 inhibitor treatment of tumours remains to be established. Conversely, cancer cells with low expression of GPX4 are more dependent on FSP1 or DHODH for ferroptosis defence, and therefore are more sensitive to ferroptosis induced by inactivation of FSP1 or DHODH than are those with high GPX4 expression5,6,9. DHODH inhibitors were found to suppress the growth of GPX4low xenograft tumours more potently than that of GPX4high ones9. Likewise, low expression of GPX4 (in concert with high expression of ALOX5) renders germinal centre B cell-like diffuse large B-cell lymphoma (DLBCL) cells vulnerable to ferroptosis induced by dimethyl fumarate (a compound approved for psoriasis treatment that depletes GSH through direct GSH succinylation)123. These recent findings suggest that the expression levels of genes involved in ferroptosis defence can be explored as biomarkers to select patients for cancer treatment with FINs that target other ferroptosis defence arms.
Ferroptosis in the microenvironment
Recent studies have also revealed that the tumour microenvironment (TME), particularly its immune cells, dictates whether tumour-cell ferroptosis will occur. CD8+ cytotoxic T cells, the major executors of antitumour immunity in the TME, secrete interferon-γ (IFNγ), which subsequently inhibits cystine uptake in cancer cells through downregulating SLC7A11 expression, thereby augmenting lipid peroxidation and ferroptosis in tumours28 (Fig. 3). Interestingly, IFNγ has also been shown to suppress SLC7A11-mediated cystine transport in macrophages124, suggesting that IFNγ can regulate SLC7A11 expression and/or activity in both cancer and non-cancer contexts. Furthermore, immune checkpoint inhibitors (ICIs) and cyst(e)inase together potentiated T cell-mediated antitumour immune responses by synergistically promoting tumour ferroptosis, suggesting that ferroptosis is important in T cell-mediated antitumour activity and that blocking SLC7A11-mediated cystine uptake in combination with ICIs is a potential therapeutic strategy for cancer28. Of note, ferroptotic cancer cells can release several immunostimulatory signals, such as high mobility group box 1 (HMGB1), calreticulin, ATP and PE, which promote dendritic cell maturation, increase the efficiency of macrophages in the phagocytosis of ferroptotic cancer cells and further enhance the infiltration of CD8+ T cells into tumours125–128 (Fig. 3). Specifically, early ferroptotic cells (after 1- or 3-hours of treatment with a GPX4 inhibitor) by releasing immunostimulatory signals can promote the phenotypic maturation of dendritic cells and elicit a vaccination-like effect to activate antitumour immunity127. Such evidence supports the concept that ferroptosis may act as a form of immunogenic cell death.
Figure 3. The role of ferroptosis in antitumour immunity.

Ferroptosis has a dual role to play in antitumour immunity, dependent on the nature of the immune cell. Boosting antitumour immunity, interferon-γ (IFNγ) secreted by CD8+ T cells promotes cancer cell ferroptosis by repressing solute carrier family 7 member 11(SLC7A11) expression in cancer cells. In turn, ferroptotic cancer cells release immunostimulatory signals that promote dendritic cell (DC) maturation and increase the efficiency of macrophages, particularly M1-like tumour-associated macrophages (TAMs), to phagocytose ferroptotic cancer cells. This further strengthens CD8+ T cell-mediated tumour suppression. In addition, several types of immunosuppressive cells, including regulatory T (Treg) cells, myeloid-derived suppressor cells (MDSCs) and M2-like TAMs, are impaired by ferroptosis induction mediated by inhibition of glutathione peroxidase 4 (GPX4) or N-acylsphingosine amidohydrolase 2 (ASAH2), thereby augmenting antitumour immunity. However, CD8+ T cells and some T helper (TH) cell subsets, such as T follicular helper (TFH) cells, are also susceptible to ferroptosis, which compromises the contribution of ferroptosis to antitumour immunity. The dashed lines indicate that the function of the indicated cell or protein is diminished or blocked in the corresponding context. TCR, T cell receptor.
Ferroptosis induction in some immunosuppressive cells can also augment antitumour immunity (Fig. 3). Regulatory T (Treg) cells, an immunosuppressive subset of CD4+ T cells that hinder protective immune surveillance against tumours, are relatively resistant to ferroptosis, possibly owing to GPX4 induction in activated Treg cells129,130. Correspondingly, Treg cell–specific deletion of Gpx4 triggers ferroptosis in Treg cells, contributing to antitumour immunity130. Similarly, myeloid-derived suppressor cells (MDSCs) with immunosuppressive functions exhibit resistance to ferroptosis that is driven by N-acylsphingosine amidohydrolase 2 (ASAH2)-mediated inhibition of the p53–heme oxygenase 1 (HMOX1) axis131; consequently, targeting ASAH2 to induce ferroptosis in MDSCs increases the activation of tumour-infiltrating cytotoxic CD8+ T cells and promotes tumour suppression131. In addition, tumour-associated macrophages (TAMs) predominantly display the M2-like phenotype to suppress antitumour immunity132. Notably, immunosuppressive M2-like TAMs are more vulnerable to ferroptosis induced by inhibition of GPX4 owing to their lack of inducible nitric oxide synthase (iNOS) expression and nitric oxide (NO•) generation (NO• can detoxify lipid peroxides) than are the M1-like TAMs that promote antitumour immunity126,133. Therefore, inducing ferroptosis in M2-like TAMs without affecting M1-like TAMs is a potential strategy to overcome the immunosuppressive TME and augment the effects of cancer immunotherapy133.
However, emerging evidence also indicates that ferroptosis has a tumour-promoting effect in the context of tumour immunity. Gpx4-deficient T cells derived from T cell–specific Gpx4 knockout mice rapidly accumulate lipid peroxides in cell membranes upon T-cell activation and subsequently undergo ferroptosis134. Furthermore, substantial lipid peroxides are detected in CD8+ T cells derived from tumours, but not those from lymph nodes, suggesting that ferroptosis may be a metabolic vulnerability of tumour-specific CD8+ T cells135. Ferroptosis in this context would presumably dampen antitumour immunity and promote tumour growth. Supporting this idea, CD36 mediates fatty acid uptake by tumour-infiltrating CD8+ T cells in the TME, and increased CD36 expression induces lipid peroxidation and ferroptosis in CD8+ T cells, thereby compromising antitumour immunity136. Genetic ablation of CD36 or blocking ferroptosis in CD8+ T cells effectively restores their antitumour activity, whereas CD8+ T cells treated with GPX4 inhibitors undergo ferroptosis and thus exert impaired antitumour effects in vivo136. Furthermore, T follicular helper (TFH) cells, a subset of CD4+ T cells that favours antitumour immunity, are highly susceptible to ferroptosis, and GPX4 expression is necessary for their survival and function137 (Fig. 3). However, it should be noted that Slc7a11 knockout in mice or cystine deprivation in vivo does not decrease the viability or antitumour effects of T cells138, which may explain the synergistic augmentation of antitumour immunity by targeting SLC7A11 in combination with ICIs28. The mechanisms underlying the differential effects of GPX4 deletion versus SLC7A11 deletion on T cell function remain elusive, but might relate to the low expression (and likely a non-essential role) of SLC7A11 in T cells139,140.
Targeting ferroptosis in cancer therapy
In recent years, FINs (Box 1) have garnered considerable interest in cancer research owing to their enormous therapeutic potential. Moreover, a variety of nanomaterials have been developed to induce ferroptosis locally or to enhance the activity of FINs30. Furthermore, growing evidence suggests that ferroptosis at least partly mediates the tumour-suppressive effects of several conventional cancer therapies, including RT25,141–143, chemotherapy26, targeted therapy27,144 and immunotherapy28, and that FINs could potentiate the efficacy of these therapies by boosting tumour ferroptosis. As discussed below, inducing ferroptosis with FINs could be a promising therapeutic strategy to eliminate cancers with specific characteristics (Table 1).
Table 1. Strategies for targeting ferroptosis in cancer therapy.
27-HC, 27-hydroxycholesterol; ALK, anaplastic lymphoma kinase; BSO, buthionine sulphoximine; BH4, tetrahydrobiopterin; ccRCC, clear cell renal cell carcinoma; CISD2, CDGSH iron-sulfur domain-containing protein 2; DHODH, dihydroorotate dehydrogenase; EGFR, epidermal growth factor receptor; EMT, epithelial-to-mesenchymal transition; FINs, ferroptosis inducers; FSP1, ferroptosis suppressor protein-1; GCH1, GTP cyclohydroxylase-1; GPX4, glutathione peroxidase 4; HMGCR, 3-hydroxy-3-methylglutaryl-coenzyme A reductase; ICIs, immune checkpoint inhibitors; IDH1, isocitrate dehydrogenase 1; iPLA2β, calcium-independent phospholipase A2β; KEAP1, kelch-like ECH associated protein 1; MT1G, metallothionein-1G; mTORC1, mTOR complex 1; NE, neuroendocrine; NCSLC, non-small-cell lung cancer; NEDD4, neural precursor cell expressed, developmentally down-regulated 4; NF2, neurofibromin 2; PD1, programmed cell death protein 1; PDK4, pyruvate dehydrogenase kinase 4; PDL1, PD1 ligand 1; PUFA-PL, polyunsaturated fatty acid-containing phospholipid; RCC, renal cell carcinoma; RT, radiotherapy; SCLC, small-cell lung cancer; SLC7A11, solute carrier family 7 member 11; SQLE, squalene monooxygenase; SQS, squalene synthase; TBH, tert-butyl-hydroperoxide; TNBC, triple-negative breast cancer; VHL, Von Hippel-Lindau.
| Strategy | Classification | Feature | Tumour type | Target or agent |
|---|---|---|---|---|
| Exploiting the vulnerability of tumours to ferroptosis | Cancer cells with specific metabolic features | EMT | Multiple cancers | GPX4 inhibitors, HMGCR inhibitors22,111 |
| Drug-tolerant persister | Multiple cancers | GPX4 inhibitors21 | ||
| Dedifferentiation | Melanoma | SLC7A11 inhibitors, GPX4 inhibitors108 | ||
| ccRCC | RCC | GPX4 inhibitors23 | ||
| Non-NE SCLC | SCLC | SLC7A11 inhibitors, GPX4 inhibitors113 | ||
| TNBC | Breast cancer | SLC7A11 inhibitors, GPX4 inhibitors114 | ||
| Cancer cells with certain genetic mutations | Mutations in E-cadherin–NF2–Hippo axis | Multiple cancer types | SLC7A11 inhibitors, GPX4 inhibitors20,115 | |
| VHL deficiency | RCC | GPX4 inhibitors, inhibition of glutathione synthesis (erastin, BSO)24,118 | ||
| EGFR mutation | NSCLC | Cysteine depletion119 | ||
| IDH1 mutation | Multiple cancer types | SLC7A11 inhibitors121 | ||
| Cancer cells with imbalanced ferroptosis defence | GPX4 low | Multiple cancer types | FSP1 inhibitors, DHODH inhibitors, GCH1 inhibitors, BH4 depletion5–9 | |
| FSP1, DHODH or GCH1 low | Multiple cancer types | SLC7A11 inhibitors, GPX4 inhibitors5–9 | ||
| Re-sensitizing resistant tumours to ferroptosis | Desuppression of PUFA-PL synthesis and peroxidation | iPLA2β overexpression | p53 wild-type cancers | iPLA2β inhibition + TBH90 |
| Adipokine chemerin upregulation | Renal cell carcinoma | Chemerin monoclonal antibody92 | ||
| Derestriction of the labile iron availability | NFS1 overexpression | Lung cancer | NFS1 inhibition + BSO, erastin or TBH95 | |
| Frataxin overexpression | Multiple cancer types | Frataxin inhibition + erastin96 | ||
| CISD2 overexpression | Head and neck cancer | Pioglitazone (CISD2 inhibitor) + sulfasalazine (SLC7A11 inhibitor)97 | ||
| Prominin2 upregulation | Breast cancer | Prominin2 inhibition + SLC7A11 inhibitors or GPX4 inhibitors98 | ||
| Preventing the upregulation of ferroptosis defence systems | SLC7A11 upregulation | KRAS mutation | SLC7A11 inhibitors102,103 | |
| GPX4 upregulation | Cancers chronically exposed to 27-HC | GPX4 inhibition93 | ||
| FSP1 or GCH1 high expression | Multiple cancer types | FSP1 inhibitors, GCH1 inhibitors, BH4 depletion5–8 | ||
| SQLE deficiency | ALK+ anaplastic large cell lymphoma | GPX4 inhibitors + inhibition of squalene synthesis (atorvastatin, zaragozic acid, SQS inhibition)105 | ||
| Abrogation of oncogenic activation-mediated ferroptosis resistance | PIK3CA activating mutations or PTEN deletion | Multiple cancer types | mTORC1 inhibitors + GPX4 inhibitors or SLC7A11 inhibitors17 | |
| NEDD4 overexpression | Melanoma | NEDD4 inhibition + SLC7A11 inhibitors169 | ||
| DJ1 overexpression | Multiple cancer types | DJ1 inhibition + SLC7A11 inhibitors or GPX4 inhibitors145 | ||
| PDK4 overexpression | Pancreatic ductal adenocarcinoma | PDK4 inhibitors + SLC7A11 inhibitors149 | ||
| Combining FINs with conventional cancer therapies | Sensitizing conventional cancer therapies | Chemotherapy, immunotherapy, or immunotherapy in combination with RT | Multiple cancer types | SLC7A11 inhibitors + cisplatin, doxorubicin, ICIs, or ICIs + RT26,28,143,153,154 |
| SLC7A11 or GPX4 induction upon RT | NSCLC | RT + SLC7A11 inhibitors or GPX4 inhibitors25 | ||
| GPX4 induction upon treatment with gemcitabine | Pancreatic ductal adenocarcinoma | Gemcitabine + SLC7A11 inhibitors152 | ||
| Overcoming resistance to conventional cancer therapies | Acquired radioresistance, intrinsic radioresistance (TP53 or KEAP1 mutation) | NSCLC | RT + SLC7A11 inhibitors or GPX4 inhibitors25,142,155 | |
| Acquired chemoresistance (cisplatin or docetaxel) | Head and neck cancer, ovarian cancer | SLC7A11 inhibitors + cisplatin or docetaxel157,158 | ||
| TYRO3-mediated anti-PD1 or PDL1 therapy resistance | Breast cancer | TYRO3 inhibitors + anti-PD1159 | ||
| MT1G-mediated sorafenib resistance | Hepatocellular carcinoma | MT1G inhibitors + sorafenib160 |
Exploiting ferroptosis vulnerabilities.
As outlined in the preceding section, ferroptosis represents a vulnerability in certain cancer types, and targeting these vulnerabilities by inducing ferroptosis provides opportunities for cancer treatment. Notably, in several cancer types (such as lung and breast cancers), cancer cells appear to be more sensitive to ferroptosis than their corresponding normal epithelial cells in vitro142. These data highlight the existence of appropriate therapeutic windows that would allow selective ferroptosis induction in tumours while sparing normal tissues.
Re-sensitizing resistant tumours to ferroptosis.
As previously discussed, diverse ferroptosis escape mechanisms confer ferroptosis resistance in cancer cells. Disrupting the mechanisms that drive ferroptosis evasion can re-sensitize ferroptosis-resistant cancer cells or tumours to ferroptosis. Ferroptosis resistance mediated by certain genes with oncogenic activities (see Box 3) can be overcome by inhibiting expression or activity of the protein products of the genes themselves. For example, DJ1 depletion by disrupting the activity of S-adenosyl homocysteine hydrolase (SAHH) in the transsulfuration pathway re-sensitizes xenograft tumours to FINs that block SLC7A11145. The transsulfuration pathway is constitutively active in conducting de novo cysteine synthesis in some cancer cells, whereas the expression of transsulfuration enzymes can be induced by cystine starvation in others145–147. Therefore, while cancer cells mainly rely on SLC7A11-mediated cystine uptake to generate intracellular cysteine, the transsulfuration pathway still significantly contributes to the intracellular cysteine pool in some cancer cells, and protects them from cystine starvation-induced ferroptosis146,147. Consequently, targeting enzymes involved in the transsulfuration pathway, such as the DJ1–SAHH axis, cystathionine β-synthase (CBS), and glycine N-methyltransferase (GNMT), can potentiate the susceptibility of tumours to FINs that block SLC7A11145,146,148. As another example, pyruvate dehydrogenase kinase 4 (PDK4) inhibition by accelerating pyruvate oxidation and stimulating fatty acid synthesis can re-sensitize xenograft tumours to FINs that block SLC7A11149.
Oncogene-induced ferroptosis resistance can also be mediated through downstream effectors that can be targeted to reverse ferroptosis resistance. For instance, PI3K promotes ferroptosis resistance mainly through mTOR complex 1 (mTORC1), and the combination of mTORC1 inhibitors with FINs that block SLC7A11 exerts potent tumour-suppressive effects in PIK3CA- or PTEN-mutant xenograft tumours17. Cysteine depletion by SLC7A11 inhibitors or cyst(e)inase exhibits striking efficacy against KRAS-mutant tumours, including those from genetically engineered mouse models, by impairing the enhanced ferroptosis defence (i.e., increased levels of intracellular cysteine and GSH) on which KRAS-mutant tumours depend102,103,150. Overall, targeting key oncogenic pathways that confer ferroptosis resistance represents an important strategy for cancer therapy.
Combining FINs with conventional cancer therapies.
It is increasingly appreciated that diverse forms of conventional cancer therapy can trigger ferroptosis. Therefore, boosting ferroptosis induced by these therapies (for instance with FINs) could further strengthen their therapeutic efficacy. RT induces ferroptosis through multiple parallel mechanisms151. In response to RT, cancer cells evolve adaptive responses, such as upregulating SLC7A11 or GPX4 expression, to antagonize RT-induced ferroptosis25. Consequently, FINs targeting either SLC7A11 or GPX4 in combination with RT can radiosensitise cancer cells or xenograft tumours by potentiating ferroptosis25,141. Similarly, gemcitabine, a chemotherapeutic agent, induces GPX4 expression and activity; GPX4 inhibition counteracts this effect, increasing the sensitivity of cancer cells and xenograft tumours to gemcitabine by inducing ferroptosis152. Likewise, FINs targeting SLC7A11 can sensitise cancer cells to chemotherapy (e.g., cisplatin and doxorubicin)153,154, immunotherapy (e.g., ICIs)28, and immunotherapy in combination with RT143.
Notably, for some cancer types with intrinsic or acquired therapy resistance, inducing ferroptosis with FINs could restore their sensitivity to conventional therapies. Mutations in the tumour suppressors TP53 or KEAP1 dampen RT-induced ferroptosis by upregulating SLC7A11 and other mechanisms, resulting in intrinsic radioresistance25,142. Cancer cells with acquired radioresistance also exhibit SLC7A11 upregulation and resistance to ferroptosis142,155,156. Consequently, reactivation of RT-induced ferroptosis by FINs can restore radiosensitivity in these cancer cells and xenograft tumours142,155. Likewise, chemoresistance in certain cancer types can be overcome by FINs. It was reported that cisplatin resistance in head and neck squamous cell carcinoma cells and corresponding xenograft tumours could be abrogated by adding FINs targeting SLC7A11157. In addition, FINs that inhibit SLC7A11 reversed docetaxel resistance in ovarian cancer cells in vitro158.
In the examples discussed above, FINs were combined with conventional therapies to overcome therapy resistance. Likewise, specific inhibitors can be combined with conventional therapies to boost ferroptosis induction in tumours and reverse therapy resistance. Tumours with high TYRO3 expression are resistant to anti-programmed cell death protein 1 (PD1) or programmed cell death 1 ligand 1 (PDL1) therapy, while metallothionein-1G (MT1G) induction renders cancer cells resistant to the multikinase inhibitor sorafenib. It was recently revealed that therapeutic resistance induced by these proteins is partly due to ferroptosis blockade and can be overcome by restoring ferroptosis through combining anti-PD1 or PDL1 therapy with TYRO3 inhibitors or sorafenib with MT1G inhibitors in vivo159,160.
Taken together, resistance to RT, chemotherapy, immunotherapy and other targeted therapies have been linked to ferroptosis resistance and this phenotype can be reversed by restoring ferroptosis with FINs or other inhibitors. It is important to note that the combination of FINs, especially class I FINs that inhibit SLC7A11, with conventional therapy appears to cause relatively limited toxic effects in normal cells142 and shows good tolerability in preclinical models25,28,141–143,159,160, further supporting the clinical application of ferroptosis-inducing combination therapeutic strategies. Several clinical trials are currently under way to test the efficacy and safety of FINs or anticancer drugs with ferroptosis-inducing activity, either alone or in combination with conventional therapy, in patients with cancer (e.g., NCT04205357161, NCT04092647162, NCT02559778163, NCT03247088164).
Perspective
Ferroptosis is a form of metabolically regulated cell death. Metabolism in cancer cells is substantially rewired to meet their increased bioenergetic and biosynthetic needs and to support their rapid proliferation. This metabolic reprogramming often induces unique metabolic features, such as an enrichment of PUFA-PLs, an overload of iron and imbalanced ferroptosis defence systems, that can create a targetable vulnerability to ferroptosis, and represents an exciting opportunity for the discovery of new therapeutic targets in cancer. In addition, for tumours that exhibit intrinsic or acquired ferroptosis resistance, targeting the underlying resistance mechanisms can restore their vulnerability to ferroptosis. Therapeutic efficacy can be further enhanced by combining FINs with conventional therapies that can induce ferroptosis; importantly, such combination therapies have shown good synergism and tolerability in preclinical models. However, thorough histological and pharmacological analyses remain imperative to evaluate potential toxic effects of FINs in normal tissues and to determine the optimal drug dosing and scheduling in patients. To realize the full potential of ferroptosis-inducing strategies in cancer therapy, several additional challenges remain to be tackled in future investigations.
GPX4 constitutes the most powerful defence against ferroptosis, and several types of therapy-resistant tumours are remarkably vulnerable to GPX4 inhibition22–24, highlighting the importance of GPX4 inhibitors in targeting ferroptosis vulnerability in cancer. The GPX4 covalent inhibitor ML210 and its recently designed derivatives (such as JKE-1674) are more selective than are RSL3 and ML162 (Box 1); however, most of these inhibitors have exhibited poor (or unclear) pharmacological properties in animal models, limiting their potential for clinical translation165. Developing and optimizing GPX4-targeting agents with improved pharmacokinetics and selectivity remains a major barrier to employing GPX4 inhibition in cancer therapy. In this regard, several currently available and in vivo-stable anticancer agents with GPX4-inhibitory activity, such as withaferin A and altretamine, may offer an alternative to address this problem166,167. In addition, considering that Gpx4 is an essential gene in the mouse49,72, it also remains to be determined whether pharmacological inhibition of GPX4 can selectively kill tumours without inducing extensive toxicity in normal tissues and intolerable side effects in patients.
In addition to cancer cells, several cell types in the TME, including immune cells that promote or suppress antitumour immunity, might also be susceptible to ferroptosis. Therefore, how to balance the ferroptosis vulnerabilities of cancer cells, antitumour immune cells and immunosuppressive cells remains a critical obstacle. To address this issue, it will be important to comprehensively understand the mechanisms underlying the differential sensitivities of cancer cells and various immune cells to ferroptosis.
Furthermore, there remains an urgent need to develop predictive biomarkers that can accurately predict tumour responses to ferroptosis induction, especially those that can be tested directly in patients’ body fluids and biopsy specimens. Such tools will be essential for the stratification of patients with cancer for treatment with ferroptosis-inducing therapies.
More in-depth understanding of ferroptosis mechanism will continue to provide key insights into targeting ferroptosis in cancer. The recently proposed compartmentalization model9 suggests that there might exist additional ferroptosis defence systems localized in other organelles. Another key question in ferroptosis research is whether lipid peroxidation on the plasma membrane represents the final step or there are additional as-yet-unknown downstream steps in triggering plasma membrane rupture and ferroptotic cell death. The observation that cytosolic GPX4 (which can detoxify lipid peroxides accumulated on the plasma membrane) fails to suppress ferroptosis in DHODH/GPX4 double inactivated cells9 indicates additional ferroptosis execution mechanisms downstream of lipid peroxidation on the plasma membrane. Understanding these fundamental questions might identify new targets in cancer therapy.
Finally, it is believed that proteins involved in PUFA-PL synthesis and peroxidation (such as ACSL4 or POR) play a passive role in ferroptosis execution by providing a constitutive supply of PUFA-PLs or ROS for lipid peroxidation (in contrast, cell death executioner proteins involved in other cell death mechanisms, such as caspase, MLKL, and gasdermin D, play an active role in inducing cell death in response to physiological cues and, correspondingly, their activation is tightly controlled by upstream signalling). This view was challenged by a very recent study showing that initial ferroptotic stress activates protein kinase C β isoform 2 (PKCβII), which then phosphorylates ACSL4 and promotes ACSL4-mediated PUFA-PL synthesis and subsequent ferroptosis, thereby forming a positive feedforward loop to amplify ferroptosis168. Attenuation of the PKCβII-ACSL4 signalling axis dampens the efficacy of immunotherapy by suppressing tumour ferroptosis168. This study will inspire future investigations aimed to understanding additional regulatory mechanisms involved in ferroptosis execution and their relevance to cancer therapy. We envision that the next few years will witness exciting new findings that will translate our mechanistic understanding of this intriguing cell death mechanism into effective cancer therapies.
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.
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 nonenzymatic reaction of labile iron and hydrogen peroxide (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
(Glutathione peroxidase). 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 2 subunits, including the transporter subunit SLC7A11 and the regulatory subunit SLC3A2.
- Ubiquinone
(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
- Hypercholesterolemia
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
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 sulphur from homocysteine to cysteine, leading to cysteine biosynthesis.
Footnotes
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.
References
- 1.Dixon SJ et al. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell 149, 1060–1072 (2012). This paper describe a new form of non-apoptotic cell death induced by the small molecule erastin, termed ferroptosis. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Stockwell BR et al. Ferroptosis: a regulated cell death nexus linking metabolism, redox biology, and disease. Cell 171, 273–285 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Jiang X, Stockwell BR & Conrad M Ferroptosis: mechanisms, biology and role in disease. Nature Reviews Molecular Cell Biology 22, 266–282 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Yang WS et al. Regulation of ferroptotic cancer cell death by GPX4. Cell 156, 317–331 (2014). This paper reports that GPX4 is a cardinal regulator of ferroptosis and that the small molecule RSL3 induces ferroptosis by directly inhibiting GPX4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Bersuker K et al. The CoQ oxidoreductase FSP1 acts parallel to GPX4 to inhibit ferroptosis. Nature 575, 688–692 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Doll S et al. FSP1 is a glutathione-independent ferroptosis suppressor. Nature 575, 693–698 (2019). Along with Bersuker et al. (2019), this paper reports a GPX4-independent ferroptosis defence mechanism in plasma membrane involving NADPH, FSP1 and ubiquinone. [DOI] [PubMed] [Google Scholar]
- 7.Kraft VA et al. GTP cyclohydrolase 1/tetrahydrobiopterin counteract ferroptosis through lipid remodeling. ACS Central Science 6, 41–53 (2019). Together with Soula et al. (2020), this paper reports a GPX4-independent ferroptosis defence mechanism involving GCH1 and BH4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Soula M et al. Metabolic determinants of cancer cell sensitivity to canonical ferroptosis inducers. Nature chemical biology 16, 1351–1360 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Mao C et al. DHODH-mediated ferroptosis defence is a targetable vulnerability in cancer. Nature 593, 586–590 (2021). This paper reports a targetable GPX4-independent ferroptosis defence mechanism in mitochondria involving NADPH, DHODH and ubiquinone. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Ingold I et al. Selenium utilization by GPX4 is required to prevent hydroperoxide-induced ferroptosis. Cell 172, 409–422. e421 (2018). [DOI] [PubMed] [Google Scholar]
- 11.Galluzzi L et al. Molecular mechanisms of cell death: recommendations of the Nomenclature Committee on Cell Death 2018. Cell Death & Differentiation 25, 486–541 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Wiernicki B et al. Excessive phospholipid peroxidation distinguishes ferroptosis from other cell death modes including pyroptosis. Cell Death & Disease 11, 922 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Kagan VE et al. Oxidized arachidonic and adrenic PEs navigate cells to ferroptosis. Nature chemical biology 13, 81–90 (2017). This paper identifies the specific relevance of phosphatidylethanolamine phospholipids to ferroptosis. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Hassannia B, Vandenabeele P & Berghe TV Targeting ferroptosis to iron out cancer. Cancer cell 35, 830–849 (2019). [DOI] [PubMed] [Google Scholar]
- 15.Jiang L et al. Ferroptosis as a p53-mediated activity during tumour suppression. Nature 520, 57–62 (2015). This study identified a new pattern of tumor suppression based on p53 regulation of cystine metabolism, ROS response and ferroptosis. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Zhang Y et al. BAP1 links metabolic regulation of ferroptosis to tumour suppression. Nature cell biology 20, 1181–1192 (2018). This paper reports that the tumour suppressor BAP1 acts by sensitizing cells to ferroptosis by mitigating SLC7A11 expression. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Yi J, Zhu J, Wu J, Thompson CB & Jiang X Oncogenic activation of PI3K-AKT-mTOR signaling suppresses ferroptosis via SREBP-mediated lipogenesis. Proceedings of the National Academy of Sciences 117, 31189–31197 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Ubellacker JM 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 and increases their survival during subsequent metastasis through the blood. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Angeli JPF, Krysko DV & Conrad M Ferroptosis at the crossroads of cancer-acquired drug resistance and immune evasion. Nature Reviews Cancer 19, 405–414 (2019). [DOI] [PubMed] [Google Scholar]
- 20.Wu J et al. Intercellular interaction dictates cancer cell ferroptosis via NF2–YAP signalling. Nature 572, 402–406 (2019). This paper reports that cell-to-cell contacts confer resistance to ferroptosis via the NF2–Hippo–YAP axis and reveals several malignant mutations in this axis as a ferroptosis vulnerability. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Hangauer MJ et al. Drug-tolerant persister cancer cells are vulnerable to GPX4 inhibition. Nature 551, 247–250 (2017). This study reports that drug-resistant persist cancer cells after targeted therapy are hypersensitive to GPX4 inhibition. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Viswanathan VS et al. Dependency of a therapy-resistant state of cancer cells on a lipid peroxidase pathway. Nature 547, 453–457 (2017). This paper reports that drug-resistant and metastasis-prone mesenchymal carcinoma cells are vulnerable to ferroptosis. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Zou Y et al. Plasticity of ether lipids promotes ferroptosis susceptibility and evasion. Nature 585, 603–608 (2020). This paper identifies PUFA-containing ether PLs (PUFA-ePLs) as additional substrates for lipid peroxidation and reports the peroxisome–ether-phospholipid axis-mediated ferroptosis vulnerability. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Zou Y et al. A GPX4-dependent cancer cell state underlies the clear-cell morphology and confers sensitivity to ferroptosis. Nature Communications 10, 1617 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Lei G et al. The role of ferroptosis in ionizing radiation-induced cell death and tumor suppression. Cell research 30, 146–162 (2020). Together with Lang et al. (2019) and Ye (2020), this study reports that ionizing radiation induces ferroptosis and that ionizing radiation synergizes with FINs, and suggests that clinically used radiotherapy works, at least partially, by inducing ferroptosis. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Guo J et al. Ferroptosis: a novel anti-tumor action for cisplatin. Cancer research and treatment: official journal of Korean Cancer Association 50, 445 (2018). [DOI] [PMC free article] [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). [DOI] [PMC free article] [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 combination of immune checkpoint inhibitors with FINs is a promising strategy cancer therapy. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Zhang Y et al. Imidazole ketone erastin induces ferroptosis and slows tumor growth in a mouse lymphoma model. Cell chemical biology 26, 623–633. e629 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Liang C, Zhang X, Yang M & Dong X Recent progress in ferroptosis inducers for cancer therapy. Advanced materials 31, 1904197 (2019). [DOI] [PubMed] [Google Scholar]
- 31.Gan B Mitochondrial regulation of ferroptosis. Journal of Cell Biology 220, e202105043 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Dixon SJ et al. Human haploid cell genetics reveals roles for lipid metabolism genes in nonapoptotic cell death. ACS chemical biology 10, 1604–1609 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Doll S et al. ACSL4 dictates ferroptosis sensitivity by shaping cellular lipid composition. Nature chemical biology 13, 91–98 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Lee H, Zhuang L & Gan B Energy stress inhibits ferroptosis via AMPK. Molecular & Cellular Oncology 7, 1761242 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Li C et al. LKB1-AMPK axis negatively regulates ferroptosis by inhibiting fatty acid synthesis. Signal transduction and targeted therapy 5, 187 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Shimada K et al. Global survey of cell death mechanisms reveals metabolic regulation of ferroptosis. Nature chemical biology 12, 497–503 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Lee H et al. Energy-stress-mediated AMPK activation inhibits ferroptosis. Nature cell biology 22, 225–234 (2020). Together with Li et al. (2020), this paper reports that energy stress suppresses ferroptosis through activation of AMPK pathway. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Chu B et al. ALOX12 is required for p53-mediated tumour suppression through a distinct ferroptosis pathway. Nature cell biology 21, 579–591 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Cui W, Liu D, Gu W & Chu B Peroxisome-driven ether-linked phospholipids biosynthesis is essential for ferroptosis. Cell Death & Differentiation 28, 2536–2551 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Lee H, Zhuang L & Gan B Ether phospholipids govern ferroptosis. Journal of Genetics and Genomics 48, 517–519 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Conrad M & Pratt DA The chemical basis of ferroptosis. Nature chemical biology 15, 1137–1147 (2019). [DOI] [PubMed] [Google Scholar]
- 42.Shah R, Shchepinov MS & Pratt DA Resolving the role of lipoxygenases in the initiation and execution of ferroptosis. ACS central science 4, 387–396 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Gaschler MM & Stockwell BR Lipid peroxidation in cell death. Biochemical and biophysical research communications 482, 419–425 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Yang WS et al. Peroxidation of polyunsaturated fatty acids by lipoxygenases drives ferroptosis. Proceedings of the National Academy of Sciences 113, E4966–E4975 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Koppula P, Zhuang L & Gan B Cytochrome P450 reductase (POR) as a ferroptosis fuel. Protein & Cell 12, 675–679 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Yan B et al. Membrane Damage during Ferroptosis Is Caused by Oxidation of Phospholipids Catalyzed by the Oxidoreductases POR and CYB5R1. Molecular Cell 81, 355–369. e310 (2021). [DOI] [PubMed] [Google Scholar]
- 47.Zou Y et al. Cytochrome P450 oxidoreductase contributes to phospholipid peroxidation in ferroptosis. Nature Chemical Biology 16, 302–309 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Wenzel SE et al. PEBP1 wardens ferroptosis by enabling lipoxygenase generation of lipid death signals. Cell 171, 628–641. e626 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Angeli JPF et al. Inactivation of the ferroptosis regulator Gpx4 triggers acute renal failure in mice. Nature cell biology 16, 1180–1191 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Magtanong L et al. Exogenous monounsaturated fatty acids promote a ferroptosis-resistant cell state. Cell chemical biology 26, 420–432. e429 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Paton CM & Ntambi JM Biochemical and physiological function of stearoyl-CoA desaturase. American Journal of Physiology-Endocrinology and Metabolism 297, E28–E37 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Tesfay L et al. Stearoyl-coa desaturase 1 protects ovarian cancer cells from ferroptotic cell death. Cancer research 79, 5355–5366 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Chen X, Yu C, Kang R & Tang D Iron metabolism in ferroptosis. Frontiers in Cell and Developmental Biology 8, 590226 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Gao M et al. Ferroptosis is an autophagic cell death process. Cell research 26, 1021–1032 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Hou W et al. Autophagy promotes ferroptosis by degradation of ferritin. Autophagy 12, 1425–1428 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Kremer DM et al. GOT1 inhibition promotes pancreatic cancer cell death by ferroptosis. Nature communications 12, 4860 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Murphy MP How mitochondria produce reactive oxygen species. Biochemical journal 417, 1–13 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Zheng J & Conrad M The Metabolic Underpinnings of Ferroptosis. Cell Metabolism 32, 920–937 (2020). [DOI] [PubMed] [Google Scholar]
- 59.Friedman JR & Nunnari J Mitochondrial form and function. Nature 505, 335–343 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Vasan K, Werner M & Chandel NS Mitochondrial metabolism as a target for cancer therapy. Cell metabolism 32, 341–352 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Gao M, Monian P, Quadri N, Ramasamy R & Jiang X Glutaminolysis and transferrin regulate ferroptosis. Molecular cell 59, 298–308 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Gao M et al. Role of mitochondria in ferroptosis. Molecular cell 73, 354–363. e353 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Heldt H & Piechulla B 15-Lipids are membrane constituents and function as carbon stores. Plant Biochemistry, 359–398 (2011).
- 64.Brigelius-Flohé R & Maiorino M Glutathione peroxidases. Biochimica et Biophysica Acta (BBA)-General Subjects 1830, 3289–3303 (2013). [DOI] [PubMed] [Google Scholar]
- 65.Brigelius-Flohé R & Flohé L Regulatory phenomena in the glutathione peroxidase superfamily. Antioxidants & redox signaling 33, 498–516 (2020). [DOI] [PubMed] [Google Scholar]
- 66.Seibt TM, Proneth B & Conrad M Role of GPX4 in ferroptosis and its pharmacological implication. Free Radical Biology and Medicine 133, 144–152 (2019). [DOI] [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. Biochimica et Biophysica Acta (BBA)-Lipids and Lipid Metabolism 710, 197–211 (1982). [DOI] [PubMed] [Google Scholar]
- 68.Pushpa-Rekha TR, Burdsall AL, Oleksa LM, Chisolm GM & Driscoll DM Rat phospholipid-hydroperoxide glutathione peroxidase: cDNA cloning and identification of multiple transcription and translation start sites. Journal of Biological Chemistry 270, 26993–26999 (1995). [DOI] [PubMed] [Google Scholar]
- 69.Pfeifer H et al. Identification of a specific sperm nuclei selenoenzyme necessary for protamine thiol cross‐linking during sperm maturation. The FASEB Journal 15, 1236–1238 (2001). [PubMed] [Google Scholar]
- 70.Maiorino M et al. Distinct promoters determine alternative transcription of gpx-4 into phospholipid-hydroperoxide glutathione peroxidase variants. Journal of Biological Chemistry 278, 34286–34290 (2003). [DOI] [PubMed] [Google Scholar]
- 71.Moreno SG, Laux G, Brielmeier M, Bornkamm GW & Conrad M Testis-specific expression of the nuclear form of phospholipid hydroperoxide glutathione peroxidase (PHGPx). Biological Chemistry 384, 635–643 (2003). [DOI] [PubMed] [Google Scholar]
- 72.Yant LJ et al. The selenoprotein GPX4 is essential for mouse development and protects from radiation and oxidative damage insults. Free Radical Biology and Medicine 34, 496–502 (2003). [DOI] [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. Molecular and cellular biology 25, 7637–7644 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Liang H et al. Short form glutathione peroxidase 4 is the essential isoform required for survival and somatic mitochondrial functions. Journal of Biological Chemistry 284, 30836–30844 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Schneider M et al. Mitochondrial glutathione peroxidase 4 disruption causes male infertility. The FASEB journal 23, 3233–3242 (2009). [DOI] [PubMed] [Google Scholar]
- 76.Imai H et al. Depletion of selenoprotein GPx4 in spermatocytes causes male infertility in mice. Journal of Biological Chemistry 284, 32522–32532 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Forman HJ, Zhang H & Rinna A Glutathione: overview of its protective roles, measurement, and biosynthesis. Molecular aspects of medicine 30, 1–12 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Aquilano K, Baldelli S & Ciriolo MR Glutathione: new roles in redox signaling for an old antioxidant. Frontiers in pharmacology 5, 196 (2014). [DOI] [PMC free article] [PubMed] [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 Communications 38, 12 (2018). [DOI] [PMC free article] [PubMed] [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. Nature cell biology 22, 476–486 (2020). [DOI] [PMC free article] [PubMed] [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. Journal of Biological Chemistry 274, 11455–11458 (1999). [DOI] [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). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Stefely JA & Pagliarini DJ Biochemistry of mitochondrial coenzyme Q biosynthesis. Trends in biochemical sciences 42, 824–843, doi: 10.1016/j.tibs.2017.06.008 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Crane FL Discovery of ubiquinone (coenzyme Q) and an overview of function. Mitochondrion 7, S2–S7 (2007). [DOI] [PubMed] [Google Scholar]
- 85.Kalen A, Norling B, Appelkvist EL & Dallner G Ubiquinone biosynthesis by the microsomal fraction from rat liver. Biochim Biophys Acta 926, 70–78, doi: 10.1016/0304-4165(87)90183-8 (1987). [DOI] [PubMed] [Google Scholar]
- 86.Turunen M, Olsson J & Dallner G Metabolism and function of coenzyme Q. Biochim Biophys Acta 1660, 171–199, doi: 10.1016/j.bbamem.2003.11.012 (2004). [DOI] [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, doi: 10.1007/BF02536234 (1993). [DOI] [PubMed] [Google Scholar]
- 88.Morre DJ & Morre DM Non-mitochondrial coenzyme Q. Biofactors 37, 355–360, doi: 10.1002/biof.156 (2011). [DOI] [PubMed] [Google Scholar]
- 89.Thöny B, Auerbach G & Blau N Tetrahydrobiopterin biosynthesis, regeneration and functions. Biochemical Journal 347, 1–16 (2000). [PMC free article] [PubMed] [Google Scholar]
- 90.Chen D et al. iPLA2β-mediated lipid detoxification controls p53-driven ferroptosis independent of GPX4. Nature Communications 12, 3644 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Sun W-Y et al. Phospholipase iPLA 2 β averts ferroptosis by eliminating a redox lipid death signal. Nature Chemical Biology 17, 465–476 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Tan SK et al. Obesity-dependent adipokine chemerin suppresses fatty acid oxidation to confer ferroptosis resistance. Cancer discovery 11, 2072–2093 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Liu W et al. Dysregulated cholesterol homeostasis results in resistance to ferroptosis increasing tumorigenicity and metastasis in cancer. Nature communications 12, 5103 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Sviderskiy VO, Terzi EM & Possemato R in Ferroptosis in Health and Disease 215–237 (Springer, 2019). [Google Scholar]
- 95.Alvarez SW et al. NFS1 undergoes positive selection in lung tumours and protects cells from ferroptosis. Nature 551, 639–643 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Du J et al. Identification of Frataxin as a regulator of ferroptosis. Redox biology 32, 101483 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Kim EH, Shin D, Lee J, Jung AR & Roh J-L CISD2 inhibition overcomes resistance to sulfasalazine-induced ferroptotic cell death in head and neck cancer. Cancer letters 432, 180–190 (2018). [DOI] [PubMed] [Google Scholar]
- 98.Brown CW et al. Prominin2 drives ferroptosis resistance by stimulating iron export. Developmental Cell 51, 575–586. e574 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Anandhan A, Dodson M, Schmidlin CJ, Liu P & Zhang DD Breakdown of an ironclad defense system: the critical role of NRF2 in mediating ferroptosis. Cell chemical biology 27, 436–447 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Chen D et al. NRF2 is a major target of ARF in p53-independent tumor suppression. Molecular cell 68, 224–232. e224 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Dodson M, Castro-Portuguez R & Zhang DD NRF2 plays a critical role in mitigating lipid peroxidation and ferroptosis. Redox biology 23, 101107 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Hu K et al. Suppression of the SLC7A11/glutathione axis causes synthetic lethality in KRAS-mutant lung adenocarcinoma. The Journal of Clinical Investigation 130, 1752–1766 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Lim JK et al. Cystine/glutamate antiporter xCT (SLC7A11) facilitates oncogenic RAS transformation by preserving intracellular redox balance. Proceedings of the National Academy of Sciences 116, 9433–9442 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.de la Vega MR, Chapman E & Zhang DD NRF2 and the hallmarks of cancer. Cancer cell 34, 21–43 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Garcia-Bermudez J et al. Squalene accumulation in cholesterol auxotrophic lymphomas prevents oxidative cell death. Nature 567, 118–122 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Martinez-Outschoorn UE, Peiris-Pagés M, Pestell RG, Sotgia F & Lisanti MP Cancer metabolism: a therapeutic perspective. Nature reviews Clinical oncology 14, 11–31 (2017). [DOI] [PubMed] [Google Scholar]
- 107.Wolpaw AJ & Dang CV Exploiting metabolic vulnerabilities of cancer with precision and accuracy. Trends in cell biology 28, 201–212 (2018). [DOI] [PMC free article] [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. e895 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Krebs AM et al. The EMT-activator Zeb1 is a key factor for cell plasticity and promotes metastasis in pancreatic cancer. Nature cell biology 19, 518–529 (2017). [DOI] [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). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Lee J-Y et al. Polyunsaturated fatty acid biosynthesis pathway determines ferroptosis sensitivity in gastric cancer. Proceedings of the National Academy of Sciences 117, 32433–32442 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Müller S et al. CD44 regulates epigenetic plasticity by mediating iron endocytosis. Nature Chemistry 12, 929–938 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Bebber CM et al. Ferroptosis response segregates small cell lung cancer (SCLC) neuroendocrine subtypes. Nature communications 12, 2048 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Verma N et al. Synthetic lethal combination targeting BET uncovered intrinsic susceptibility of TNBC to ferroptosis. Science advances 6, eaba8968 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Yang W-H et al. The hippo pathway effector TAZ regulates ferroptosis in renal cell carcinoma. Cell reports 28, 2501–2508. e2504 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Iliopoulos O, Kibel A, Gray S & Kaelin WG Tumour suppression by the human von Hippel-Lindau gene product. Nature medicine 1, 822–826 (1995). [DOI] [PubMed] [Google Scholar]
- 117.Kaelin WG Jr The von Hippel–Lindau tumour suppressor protein: O 2 sensing and cancer. Nature Reviews Cancer 8, 865–873 (2008). [DOI] [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). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Poursaitidis I et al. Oncogene-selective sensitivity to synchronous cell death following modulation of the amino acid nutrient cystine. Cell reports 18, 2547–2556 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Zhang Y et al. mTORC1 couples cyst (e) ine availability with GPX4 protein synthesis and ferroptosis regulation. Nature Communications 12, 1589 (2021). This study uncovers a ferroptosis modulatory mechanism that coordinates GPX4 protein synthesis with cyst(e)ine availability and suggests the combination of mTORC1 inhibitors with FINs for cancer therapy. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Wang T-X et al. The oncometabolite 2-hydroxyglutarate produced by mutant IDH1 sensitizes cells to ferroptosis. Cell death & disease 10, 755 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Kaelin WG The concept of synthetic lethality in the context of anticancer therapy. Nature reviews cancer 5, 689–698 (2005). [DOI] [PubMed] [Google Scholar]
- 123.Schmitt A et al. Dimethyl fumarate induces ferroptosis and impairs NF-κB/STAT3 signaling in DLBCL. Blood, The Journal of the American Society of Hematology 138, 871–884 (2021). [DOI] [PubMed] [Google Scholar]
- 124.Sato H, Fujiwara K, Sagara J. i. & Bannai S Induction of cystine transport activity in mouse peritoneal macrophages by bacterial lipopolysaccharide. Biochemical Journal 310, 547–551 (1995). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Wen Q, Liu J, Kang R, Zhou B & Tang D The release and activity of HMGB1 in ferroptosis. Biochemical and biophysical research communications 510, 278–283 (2019). [DOI] [PubMed] [Google Scholar]
- 126.Luo X et al. Oxygenated phosphatidylethanolamine navigates phagocytosis of ferroptotic cells by interacting with TLR2. Cell Death & Differentiation 28, 1971–1989 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Efimova I et al. Vaccination with early ferroptotic cancer cells induces efficient antitumor immunity. Journal for immunotherapy of cancer 8, e001369 (2020). [DOI] [PMC free article] [PubMed] [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. Nature communications 11, 3637 (2020). [DOI] [PMC free article] [PubMed] [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. Molecular cancer 19, 1–23 (2020). [DOI] [PMC free article] [PubMed] [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 Reports 35, 109235 (2021). [DOI] [PubMed] [Google Scholar]
- 131.Zhu H et al. Asah2 Represses the p53–Hmox1 Axis to Protect Myeloid-Derived Suppressor Cells from Ferroptosis. The Journal of Immunology 206, 1395–1404 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Xia Y et al. Engineering macrophages for cancer immunotherapy and drug delivery. Advanced Materials 32, 2002054 (2020). [DOI] [PubMed] [Google Scholar]
- 133.Kapralov AA et al. Redox lipid reprogramming commands susceptibility of macrophages and microglia to ferroptotic death. Nature chemical biology 16, 278–290 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Matsushita M et al. T cell lipid peroxidation induces ferroptosis and prevents immunity to infection. Journal of Experimental Medicine 212, 555–568 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Drijvers JM et al. Pharmacologic Screening Identifies Metabolic Vulnerabilities of CD8+ T Cells. Cancer immunology research 9, 184–199 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Ma X et al. CD36-mediated ferroptosis dampens intratumoral CD8+ T cell effector function and impairs their antitumor ability. Cell metabolism 33, 1001–1012.e1005. (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Yao Y et al. Selenium–GPX4 axis protects follicular helper T cells from ferroptosis. Nature Immunology 22, 1127–1139 (2021). [DOI] [PubMed] [Google Scholar]
- 138.Arensman MD et al. Cystine–glutamate antiporter xCT deficiency suppresses tumor growth while preserving antitumor immunity. Proceedings of the National Academy of Sciences 116, 9533–9542 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.GMÜNDER H, ECK HP & DRÖGE W Low membrane transport activity for cystine in resting and mitogenically stimulated human lymphocyte preparations and human T cell clones. European journal of biochemistry 201, 113–117 (1991). [DOI] [PubMed] [Google Scholar]
- 140.Pacheco R et al. Glutamate released by dendritic cells as a novel modulator of T cell activation. The Journal of Immunology 177, 6695–6704 (2006). [DOI] [PubMed] [Google Scholar]
- 141.Ye LF et al. Radiation-induced lipid peroxidation triggers ferroptosis and synergizes with ferroptosis inducers. ACS chemical biology 15, 469–484 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Lei G et al. Ferroptosis as a mechanism to mediate p53 function in tumor radiosensitivity. Oncogene 40, 3533–3547 (2021). This study identifies a previously unappreciated role of ferroptosis in p53-mediated radiosensitization and suggests the combination of radiotherapy and FINs for treatment of p53-mutant cancers. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Lang X et al. Radiotherapy and immunotherapy promote tumoral lipid oxidation and ferroptosis via synergistic repression of SLC7A11. Cancer discovery 9, 1673–1685 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Dixon SJ et al. Pharmacological inhibition of cystine–glutamate exchange induces endoplasmic reticulum stress and ferroptosis. Elife 3, e02523 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Cao J et al. DJ-1 suppresses ferroptosis through preserving the activity of S-adenosyl homocysteine hydrolase. Nature communications 11, 1251 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Zhu J et al. Transsulfuration activity can support cell growth upon extracellular cysteine limitation. Cell metabolism 30, 865–876. e865 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Liu N, Lin X & Huang C Activation of the reverse transsulfuration pathway through NRF2/CBS confers erastin-induced ferroptosis resistance. British journal of cancer 122, 279–292 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Wang L et al. A pharmacological probe identifies cystathionine β-synthase as a new negative regulator for ferroptosis. Cell death & disease 9, 1025 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Song X et al. PDK4 dictates metabolic resistance to ferroptosis by suppressing pyruvate oxidation and fatty acid synthesis. Cell Reports 34, 108767 (2021). [DOI] [PubMed] [Google Scholar]
- 150.Badgley MA et al. Cysteine depletion induces pancreatic tumor ferroptosis in mice. Science 368, 85–89 (2020). [DOI] [PMC free article] [PubMed] [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). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Zhu S et al. HSPA5 regulates ferroptotic cell death in cancer cells. Cancer research 77, 2064–2077 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Yu Y et al. The ferroptosis inducer erastin enhances sensitivity of acute myeloid leukemia cells to chemotherapeutic agents. Molecular & cellular oncology 2, e1054549 (2015). [DOI] [PMC free article] [PubMed] [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. Clinical cancer research 19, 845–854 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Pan X et al. Erastin decreases radioresistance of NSCLC cells partially by inducing GPX4‑mediated ferroptosis. Oncology letters 17, 3001–3008 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Xie L et al. Solute carrier protein family may involve in radiation-induced radioresistance of non-small cell lung cancer. Journal of cancer research and clinical oncology 137, 1739 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Roh J-L, Kim EH, Jang HJ, Park JY & Shin D Induction of ferroptotic cell death for overcoming cisplatin resistance of head and neck cancer. Cancer letters 381, 96–103 (2016). [DOI] [PubMed] [Google Scholar]
- 158.Zhou H-H et al. Erastin reverses ABCB1-mediated docetaxel resistance in ovarian cancer. Frontiers in oncology 9, 1398 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Jiang Z et al. TYRO3 induces anti–PD-1/PD-L1 therapy resistance by limiting innate immunity and tumoral ferroptosis. Journal of Clinical Investigation 131, e139434 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Sun X et al. Metallothionein‐1G facilitates sorafenib resistance through inhibition of ferroptosis. Hepatology 64, 488–500 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.US National Library of Medicine. ClinicalTrials.gov, https://ClinicalTrials.gov/show/NCT04205357 (2019). [DOI] [PubMed]
- 162.US National Library of Medicine. ClinicalTrials.gov, https://ClinicalTrials.gov/show/NCT04092647 (2019). [DOI] [PubMed]
- 163.US National Library of Medicine. ClinicalTrials.gov, https://ClinicalTrials.gov/show/NCT02559778 (2015). [DOI] [PubMed]
- 164.US National Library of Medicine. ClinicalTrials.gov, https://ClinicalTrials.gov/show/NCT03247088 (2017). [DOI] [PubMed]
- 165.Eaton JK et al. Selective covalent targeting of GPX4 using masked nitrile-oxide electrophiles. Nature chemical biology 16, 497–506 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Hassannia B et al. Nano-targeted induction of dual ferroptotic mechanisms eradicates high-risk neuroblastoma. The Journal of clinical investigation 128, 3341–3355 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Woo JH et al. Elucidating compound mechanism of action by network perturbation analysis. Cell 162, 441–451 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Zhang H-L et al. PKCβII phosphorylates ACSL4 to amplify lipid peroxidation to induce ferroptosis. Nature Cell Biology, 88–89 (2022). [DOI] [PubMed]
- 169.Yang Y et al. Nedd4 ubiquitylates VDAC2/3 to suppress erastin-induced ferroptosis in melanoma. Nature communications 11, 433 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Feng H & Stockwell BR Unsolved mysteries: How does lipid peroxidation cause ferroptosis? PLoS biology 16, e2006203 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Larraufie M-H et al. Incorporation of metabolically stable ketones into a small molecule probe to increase potency and water solubility. Bioorganic & medicinal chemistry letters 25, 4787–4792 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Plosker GL & Croom KF Sulfasalazine. Drugs 65, 1825–1849 (2005). [DOI] [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). [DOI] [PubMed] [Google Scholar]
- 174.Robert SM et al. SLC7A11 expression is associated with seizures and predicts poor survival in patients with malignant glioma. Science translational medicine 7, 289ra286–289ra286 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Zheng J et al. Sorafenib fails to trigger ferroptosis across a wide range of cancer cell lines. Cell Death & Disease 12, 698 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Cramer SL et al. Systemic depletion of L-cyst (e) ine with cyst (e) inase increases reactive oxygen species and suppresses tumor growth. Nature medicine 23, 120–127 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Gaschler MM et al. FINO 2 initiates ferroptosis through GPX4 inactivation and iron oxidation. Nature chemical biology 14, 507–515 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Bykov VJ, Eriksson SE, Bianchi J & Wiman KG Targeting mutant p53 for efficient cancer therapy. Nature Reviews Cancer 18, 89–102 (2018). [DOI] [PubMed] [Google Scholar]
- 179.Bieging KT, Mello SS & Attardi LD Unravelling mechanisms of p53-mediated tumour suppression. Nature Reviews Cancer 14, 359–370 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Wang Y et al. Epigenetic regulation of ferroptosis by H2B monoubiquitination and p53. EMBO reports 20, e47563 (2019). [DOI] [PMC free article] [PubMed] [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. Proceedings of the National Academy of Sciences 113, E6806–E6812 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Zhang Y et al. Ferredoxin reductase is critical for p53-dependent tumor suppression via iron regulatory protein 2. Genes & development 31, 1243–1256 (2017). [DOI] [PMC free article] [PubMed] [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 & development 30, 918–930 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Wang S-J et al. Acetylation is crucial for p53-mediated ferroptosis and tumor suppression. Cell reports 17, 366–373 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Tarangelo A et al. p53 suppresses metabolic stress-induced ferroptosis in cancer cells. Cell reports 22, 569–575 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Xie Y et al. The tumor suppressor p53 limits ferroptosis by blocking DPP4 activity. Cell reports 20, 1692–1704 (2017). [DOI] [PubMed] [Google Scholar]
- 187.Carbone M et al. BAP1 and cancer. Nature Reviews Cancer 13, 153–159 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Ventii KH et al. BRCA1-associated protein-1 is a tumor suppressor that requires deubiquitinating activity and nuclear localization. Cancer research 68, 6953–6962 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Carbone M et al. Biological mechanisms and clinical significance of BAP1 mutations in human cancer. Cancer discovery 10, 1103–1120 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Network CGAR Comprehensive genomic characterization of squamous cell lung cancers. Nature 489, 519 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Collisson E et al. Comprehensive molecular profiling of lung adenocarcinoma: The cancer genome atlas research network. Nature 511, 543–550 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Baird L & Yamamoto M The molecular mechanisms regulating the KEAP1-NRF2 pathway. Molecular and cellular biology 40, e00099–00020 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Kensler TW, Wakabayashi N & Biswal S Cell survival responses to environmental stresses via the Keap1-Nrf2-ARE pathway. Annu. Rev. Pharmacol. Toxicol 47, 89–116 (2007). [DOI] [PubMed] [Google Scholar]
- 194.Sasaki H et al. Electrophile response element-mediated induction of the cystine/glutamate exchange transporter gene expression. Journal of Biological Chemistry 277, 44765–44771 (2002). [DOI] [PubMed] [Google Scholar]
- 195.Sherr CJ Divorcing ARF and p53: an unsettled case. Nature Reviews Cancer 6, 663–673 (2006). [DOI] [PubMed] [Google Scholar]
- 196.Fruman DA et al. The PI3K pathway in human disease. Cell 170, 605–635 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Zhang Y et al. A pan-cancer proteogenomic atlas of PI3K/AKT/mTOR pathway alterations. Cancer cell 31, 820–832. e823 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198.Lei G, Zhuang L & Gan B mTORC1 and ferroptosis: Regulatory mechanisms and therapeutic potential. BioEssays, 2100093 (2021). [DOI] [PubMed]
- 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 Therapy 28, 55–63 (2021). [DOI] [PubMed] [Google Scholar]
- 200.Prior IA, Lewis PD & Mattos C A comprehensive survey of Ras mutations in cancer. Cancer research 72, 2457–2467 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Pylayeva-Gupta Y, Grabocka E & Bar-Sagi D RAS oncogenes: weaving a tumorigenic web. Nature Reviews Cancer 11, 761–774 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202.Padanad MS et al. Fatty acid oxidation mediated by Acyl-CoA synthetase long chain 3 is required for mutant KRAS lung tumorigenesis. Cell reports 16, 1614–1628 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.Wang X et al. NEDD4–1 is a proto-oncogenic ubiquitin ligase for PTEN. Cell 128, 129–139 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204.Yagoda N et al. RAS–RAF–MEK-dependent oxidative cell death involving voltage-dependent anion channels. Nature 447, 865–869 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.Cao J, Lou S, Ying M & Yang B DJ-1 as a human oncogene and potential therapeutic target. Biochemical pharmacology 93, 241–250 (2015). [DOI] [PubMed] [Google Scholar]
- 206.Leclerc D et al. Oncogenic role of PDK4 in human colon cancer cells. British journal of cancer 116, 930–936 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
