Skip to main content
Erschienen in: Experimental Hematology & Oncology 1/2023

Open Access 01.12.2023 | Review

Ferroptosis: a novel regulated cell death participating in cellular stress response, radiotherapy, and immunotherapy

verfasst von: Xiaogang Zheng, Xiaodong Jin, Fei Ye, Xiongxiong Liu, Boyi Yu, Zheng Li, Ting Zhao, Weiqiang Chen, Xinguo Liu, Cuixia Di, Qiang Li

Erschienen in: Experimental Hematology & Oncology | Ausgabe 1/2023

Abstract

Background

Ferroptosis is a regulated cell death mode triggered by iron-dependent toxic membrane lipid peroxidation. As a novel cell death modality that is morphologically and mechanistically different from other forms of cell death, such as apoptosis and necrosis, ferroptosis has attracted extensive attention due to its association with various diseases. Evidence on ferroptosis as a potential therapeutic strategy has accumulated with the rapid growth of research on targeting ferroptosis for tumor suppression in recent years.

Methods

We summarize the currently known characteristics and major regulatory mechanisms of ferroptosis and present the role of ferroptosis in cellular stress responses, including ER stress and autophagy. Furthermore, we elucidate the potential applications of ferroptosis in radiotherapy and immunotherapy, which will be beneficial in exploring new strategies for clinical tumor treatment.

Result and conclusion

Based on specific biomarkers and precise patient-specific assessment, targeting ferroptosis has great potential to be translated into practical new approaches for clinical cancer therapy, significantly contributing to the prevention, diagnosis, prognosis, and treatment of cancer.
Hinweise

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
8-OHG
8-Hydroxyguanosine
ACSL4
Acyl-CoA synthetase long-chain family member 4
ALOXs
Lipoxygenases
AML
Acute myeloid leukemia
ATM
Ataxia telangiectasia mutated protein
BH4
Tetrahydrobiopterin
cGAS
Cyclic GMP–AMP synthase
CoQ10
Coenzyme Q10
DAMPs
Damage-associated molecular patterns
DHODH
Dihydroorotate dehydrogenase
DMT1
Ferrous ion membrane transport protein 1
FPN
Ferroportin
FSP1
Ferroptosis suppressor protein 1/AIFM2
GCH1
GTP cyclohydrolase 1
GCL
Glutamate–cysteine ligase
GGT1
Gamma-glutamyltransferase 1
GSS
Glutathione synthase
GPX4
Glutathione peroxidase 4
GR
Glutathione reductase
GSH
Glutathione
GSSG
Oxidized glutathione disulfide
HLF
Hepatic leukemia factor
HMGB1
High mobility group protein B1
IFNγ
Interferon-gamma
KRAS (G12D)
Kirsten rat sarcoma viral oncogene homolog-G12D mutation
LPCAT3
Lysophosphatidylcholine acyltransferase 3
MVBs
Multivesicular bodies
PD-L1
Programmed cell death-ligand 1
PERK
PKR-like ER kinase
POR
Cytochrome P450 oxidoreductase
PROM2
Prominin-2
PUFA
Polyunsaturated fatty acid
PUFA-PLs
Polyunsaturated fatty-acid-containing phospholipids
PUFA-PL-OH
PUFA phospholipid alcohols
PUFA-PL-OOH
Phospholipid with peroxidized polyunsaturated fatty acyl tail
ROS
Reactive oxygen species
RT-MPs
Irradiated tumor cell-released microparticles
STEAP3
Six-transmembrane epithelial antigen of the prostate 3
STING
Stimulator of interferon genes protein
TFRC
Transferrin receptor
TGFβ
Transforming growth factor β
TNBC
Triple negative breast cancer
TNFα
Tumor necrosis factor-alpha

Background

Ferroptosis is a novel regulated cell death (RCD) mode caused by damage to lipid membranes due to the accumulation of lipid peroxides and reactive oxygen species (ROS) produced by iron metabolism, which was first observed and named by Stockwell in 2012 [1, 2]. Ferroptosis is driven by lethal lipid peroxidation resulting from imbalances in cellular metabolism and redox homeostasis [2]. More and more studies have shown that ferroptosis is involved in stress processes such as endoplasmic reticulum stress and autophagy, and ferroptosis is associated with many diseases, including degenerative diseases, cancer, acute kidney injury, I/R injury of the heart, liver, and kidney, as well as other diseases such as acute myeloid leukemia, age-related macular degeneration (AMD), psoriasis, and hemolytic disorders [1, 3]. Induction of ferroptosis by experimental small-molecule compounds or clinical drugs is emerging as an effective antitumor strategy for various types of cancer, especially in iron-rich and higher mitochondrial abundance tissues such as hepatocellular carcinoma (HCC), pancreatic ductal adenocarcinoma (PDAC), breast cancer and non-small cell lung carcinoma (NSCLC) [4, 5]. Here we summarize the role of ferroptosis in cellular stress responses, as well as its potential role in cancer radiotherapy and immunotherapy.

Hallmarks and mechanisms of ferroptosis

Hallmarks of ferroptosis

Most organisms rely on oxygen as the final electron acceptor in redox-based metabolic processes, and one of the key issues in cell fate determination is the response of cells to oxidative stress. In the process of oxidative stress, irreversible oxidative damage of the lipid membrane bilayer may lead to ferroptosis. Mitochondria are the core sites for aerobic respiration and energy metabolism in mammals. Through the citric acid cycle and oxidative phosphorylation, substrates such as glucose, lipids and proteins are oxidized to produce ATP, which regulates the material cycle and energetic metabolism of organisms. During electron transport and oxidative phosphorylation, some electrons reduce oxygen to form superoxide, a key source of reactive oxygen species (ROS). Imbalance in ROS homeostasis is a critical trigger for lipid peroxidation and ferroptosis induction.
Morphological features of ferroptosis include loss of cell membrane integrity. On the subcellular level, mitochondria usually show morphological abnormalities in the case of ferroptosis. The primary manifestation is the shrinkage of the mitochondrial membrane, resulting in the increase of membrane density and the decrease of inner mitochondrial membrane cristae [1]. In a few cases, the cell morphology becomes rounded, and the number of autophagic vacuoles increases as well.
Different from known cell death modes such as apoptosis, necroptosis, pyroptosis, and cuproptosis, there is usually no change in nuclear morphology, DNA fragmentation, or Caspase signal transmission during ferroptosis, and this process cannot be reversed by the inhibitors of the above-mentioned RCD modes [3, 6, 7]. In addition, ferroptosis induction is usually accompanied by iron and ROS accumulation, lipid peroxidation, and mitogen-activated protein kinase (MAPK) activation. In contrast, when ferroptosis occurs, cystine-glutamate antiporter (system Xc) inhibition, NADPH oxidation, glutathione (GSH) depletion, and loss of mitochondrial membrane potential (ΔΨm) are common [8].

Mechanisms of ferroptosis

Intracellular iron accumulation and lipid peroxidation are two major biochemical events that induce ferroptosis (Fig. 1). Multiple organelles, including mitochondria, ER, Golgi apparatus, and lysosome, are involved in regulating iron metabolism and redox balance, suggesting that a dynamic signaling network controls and enforces ferroptosis. Ferroptotic cell death requires three conditions [9], including (1) the need for redox-active iron to drive the peroxidation reaction; (2) the need for polyunsaturated fatty acid phospholipids (PUFA-PLs) as substrates for the peroxidation reaction; (3) the malfunction of the homeostatic repair system that needs to remove lipid peroxidation products. In addition, MAPK and autophagy intensity also regulate ferroptosis [10, 11]. Conversely, lipid peroxidation and ferroptosis can be alleviated by maintaining iron homeostasis, scavenging free radicals, repairing peroxidative damage and stress response, and so on [8, 12].

Iron homeostasis and ferroptosis

Iron is involved in the synthesis of DNA, heme, and Fe–S clusters, the formation of various enzymes such as lipoxygenase (LOX), xanthine oxidase, NADPH oxidase, and the active site of mitochondrial complexes I and III [13, 14]. The extracellular circulating iron exists in the form of ferritin-bound Fe3+, which is transferred into cells via transferrin receptor (TFRC), and then delivered to the endosome and catalyzed by the reductase six-transmembrane epithelial antigen of the prostate 3 (STEAP3) into Fe2+. SLC11A2/DMT1 releases Fe2+ into the cytoplasm and stores it in ferritin or forms a labile iron pool. Ferritin consists of two subunits, light chain 1 (FTL1) and heavy chain 1 (FTH1), with a highly conserved spherical shell three-dimensional structure that accommodates iron in a safe, soluble, and bioavailable form, preventing iron from being oxidized by ROS and causing oxidative damage. Excess Fe2+ is converted into Fe3+ by the transmembrane exporter ferroportin/SLC40A1 (FPN) and transported to the extracellular circulation [15]. If the iron homeostasis mechanism fails, the Fenton reaction (Fe2+ + H2O2 → Fe3+ + HO + OH·) induced by iron generates ROS and induces the activation of iron-containing enzymes such as LOX, resulting in the damage of protein, lipid, and DNA [16]. The oxidation potential of OH· generated by the Fenton reaction is as high as 2.73 V, which has a strong oxidization ability and plays a significant role in membrane peroxidation.
Epidemiological studies have demonstrated that high dietary iron intake increases the risk of HCC and breast cancer. In vitro studies indicated that silencing TFRC can inhibit Erastin-induced ferroptosis. Deferoxamine (iron chelator) blocks cells from ferroptosis [1], while exogenous iron supplementation promotes ferroptosis induced by Erastin in NSCLC [17]. Iron-regulating proteins ACO1 and IREB2 regulate ferroptosis at the translational level [18]. Increasing the biosynthesis of Fe–S clusters reduces iron concentration and inhibits ferroptosis. Overexpression or inhibition of FPN1 can negatively or positively regulate Erastin-induced ferroptosis in ectopic endometrial stromal cells (EESCs) [19]. In vivo, IREB2 significantly upregulates the expression of FTL1 and FTH1 and inhibits Erastin-induced ferroptosis. Yue et al. developed a co-activated catalytic nanoplatform (CACN) that disassembles under the trigger of ATP and releases Fe3O4 nanoparticles in mouse breast cancer cells (4T1) or human embryonic kidney cells (HEK293), which lead to ferroptosis through the ROS-triggered lipid peroxidation. The disassembly of CACN also accurately releases doxorubicin (Dox), promotes the Fenton reaction and enhances ferroptosis [20]. Mitochondrial enzyme heme oxygenase 1 (HMOX1) is also involved in the regulation of ferroptosis by catalyzing the degradation of heme to release iron, which leads to iron overload and induces lipid peroxidation [2123]. Overexpression of HMOX1 increases intracellular iron content and sensitizes tumor cells to ferroptosis by the Nrf2-HMOX1 axis. In contrast, knockdown, pharmacological inhibition, or degradation of HMOX1 prevents ferroptosis by reducing iron overload, ROS production, and lipid peroxidation [2426]. However, another study showed that as a cell stress-induced protective enzyme, slight up-regulation of HMOX1 exerts a protective effect against ferroptosis [27]. Other methods of increasing iron absorption, reducing iron storage, and limiting iron efflux can lead to iron accumulation and promote ferroptosis [18].

Lipid peroxidation and ferroptosis

Lipid functions include energy storage, signal transmission, and biomembrane formation. Lipid peroxidation is one of the important features of ferroptosis, which often results in oxidative damage to biological membranes (plasma and inner organelle membranes), lipoproteins, and other lipid-containing molecules [28, 29]. PUFA-PLs are the primary substrates for lipid peroxidation. Acyl-CoA synthetase long-chain family member 4 (ACSL4) and lysophosphatidylcholine acyltransferase 3 (LPCAT3) catalyze the esterification and the incorporation of PUFAs into PLs. PUFA-PLs undergo oxygenase (ALOXs)-mediated peroxidation to form PL-PUFA-OOH, leading to ferroptosis. Inhibition of ACSL4 and LPCAT3 expression reduces the intracellular accumulation of lipid peroxide substrates and inhibits ferroptosis [30]. In mouse embryonic fibroblasts, knockout of ACSL4 inhibits lipid peroxidation and ferroptosis, while re-expression of ACSL4 after RSL3 treatment re-sensitizes cells to ferroptosis [31]. LPCAT3 is essential for ferroptosis execution in massive insertional mutagenesis of haploid KBM7 cells [32]. Lipid peroxidation produces a series of primary (lipid hydroperoxides) and secondary oxidation aldehydes, including propanal, hexanal, malondialdehyde (MDA), and 4-hydroxynonenal (4-HNE). Among them, MDA and 4-HNE have higher reactivity and toxicity, and are associated with cancer, diabetes, neurodegenerative and cardiovascular diseases [28]. MDA can exert its mutagenic potential by cross-linking with DNA or proteins [33]. 4-HNE can generate a variety of cytotoxic and genotoxic stresses, including cell cycle arrest, senescence, and programmed cell death (PCD) [28]. Thus, MDA and 4-HNE have become biomarkers of ferroptosis induced by lipid peroxidation due to iron homeostasis imbalance [34, 35]. Cells detoxify the lipid peroxides by blocking the Fenton reaction, scavenging free radicals, repairing damage caused by toxic oxidation products, upregulating the expression of antioxidant nuclear factor E2-related factor 2 (Nrf2), activating heat shock proteins (HSP), and so on [5, 36].

Redox system and ferroptosis

System XC

The system Xc is a transmembrane glutamate/cystine antiporter located on the cell surface. Structurally, system Xc consists of two subunits, the light chain SLC7A11 and the heavy chain SLC3A2, which carry out the transmembrane exchange of intracellular glutamate and extracellular cystine [35]. SLC7A11 is the core functional element of the system Xc, which is abundantly expressed in human tumors. Cells synthesize GSH using cystine, glutamate and glycine as substrates under the catalysis of glutamate–cysteine ligase (GCL) and glutathione synthase (GS). GSH acts as a free radical scavenger and a cofactor of GPX4 to alleviate oxidative stress and maintain redox homeostasis [37, 38]. Inhibiting the function of the system Xc by small-molecule drugs [class I ferroptosis inducers (FINs)] such as Erastin, sulfasalazine, sorafenib, or gene knockout can significantly reduce the production of intracellular GSH, thereby leading to lipid peroxidation and ferroptosis [1, 39].

GPX4

The selenoprotein glutathione peroxidase 4 (GPX4) is a core inhibitor of ferroptosis. In humans, the GPX4 gene is located on chromosome 19, which is about 4 kbp in length and consists of 7 exons and 6 introns [40]. The GPX4 gene has three different isomers, namely cytosolic isoform (c-GPX4), mitochondrial isoform (m-GPX4), and nuclear isoform (n-GPX4) [41]. The expression of each isoform is driven by different promoters and may play different roles in regulating signal transduction in different cell death modes. Pharmacological selenium (Se) induces changes in the expression of m-GPX4 and n-GPX4. n-GPX4 may be a necessary isoform induced by Se to counteract lipid peroxidation, and its underlying mechanism is that Se effectively inhibits GPX4-dependent ferroptosis by activating TFAP2c and Sp1-regulated gene expression [42]. With GSH as a cofactor, GPX4 reduces lipid peroxides (L-OOH) to non-toxic lipids (L-OH) so as to inhibit ferroptosis, and this may be hampered by ML210 or RSL3 [8]. GPX4 is essential for the utilization of membranes by living cells to enhance cellular plasticity and to use peroxides as second messengers in redox signaling [43, 44]. Decreased GPX4 activity leads to the accumulation of lipid peroxides (L-OOH) and the formation of lipid free radicals (L-O) catalyzed by iron, thereby resulting in cytotoxicity and cell death. Small molecule inhibitor (class II FINs) RSL3, ML162, ML210, or genetic inhibition of GPX4 leads to increased cytoplasmic ROS and promotes ferroptosis [39, 45]. DMOCPTL, a derivative of the natural product parthenolide, binds directly to the GPX4 and induces its ubiquitination, thereby inducing ferroptosis in triple-negative breast cancer (TNBC) cells [46]. Silencing GPX4 expression in gefitinib-resistant TNBC cells enhances the anti-cancer effect of gefitinib by promoting ferroptosis [47, 48]. In contrast, overexpression of GPX4 reduces cytoplasmic ROS levels and prevents ferroptosis [8, 4951].

Nrf2

The nuclear transcription factor Nrf2 forms a complex with Keap1 (Kelch-like ECH-associated protein 1) and is degraded by the ubiquitination pathway, keeping Nrf2 at a low level under normal physiological conditions. By competitively inhibiting Keap1, p62 prevents the degradation and promotes the nuclear translocation of Nrf2. In the nucleus, Nrf2 forms heterodimerization with Maf and binds to antioxidant response elements (AREs), and activates downstream genes (such as NQO1, HO-1, FTH1, etc.) that regulate iron and ROS metabolism via MT -1G, and thus inhibits ferroptosis induced by Erastin and sorafenib in HCC cells and xenograft models [5]. Activation of Nrf2 increases the degradation of lipid peroxides generated by 12/15-LOX through upregulation of CHAC1 and aldo–keto reductases AKR1C1/3, leading to ferroptosis resistance in melanoma cells. Inhibition of AKRs can re-sensitize drug-resistant melanoma cells to ferroptosis [52]. NRF2 knockdown or inhibition can decrease the expression of GPX4 and enhance the sensitivity of acute myeloid leukemia (AML) cells to ferroptosis inducers. NRF2 inhibition and GPX4 inhibition synergistically induce ferroptosis, effectively reducing the viability of AML cells [53]. On the other hand, recent studies have shown that up-regulation or stabilization of Nrf2 increases intracellular GSH levels, while only moderately inhibiting ferroptosis, because Nrf2 up-regulation results in collateral sensitivity via the expression of multidrug resistance protein 1 (MRP1), which mediates GSH efflux from cells. Thus Nrf2 increases both GSH synthesis and MRP1-mediated GSH efflux. Therefore, the high Nrf2 expression alone is a poor predictor of ferroptosis resistance in some cases, especially when MRP1-mediated GSH efflux counterbalances the protective effects of increased GSH synthesis [5456].

p53

Ferroptosis is considered an endogenous tumor suppressor mechanism downstream of p53 [57]. p53 can occupy the promoter region of the SLC7A11 gene, transcriptionally repress SLC7A11 expression, inhibit system Xc activity, lead to insufficient cystine uptake and GSH synthesis, increase lipid peroxidation, and cause ferroptosis [5860]. In addition, downregulation of SLC7A11 by p53 promotes the release of ALOX-12, which oxidizes membrane PUFAs, and exerts a pro-ferroptosis effect [61]. Moreover, p53 activates glutaminolysis by upregulating the expression of glutaminase 2 (GLS2) and enhances ALOX-15 by activating spermidine/spermine N1-acetyltransferase 1 (SAT1), jointly aggravating ROS-induced lipid peroxidation and promoting ferroptosis [6264].
In addition to the aforementioned antioxidant systems and mechanisms, reductive coenzyme Q10 (CoQ10) produced by extramitochondrial membranes protein ferroptosis suppressor protein 1 (FSP1) and inner mitochondrial membranes protein dihydroorotate dehydrogenase (DHODH), the reduced form of vitamin K (VKH2) generated by FSP1, and tetrahydrobiopterin (BH4) produced by GTP cyclohydrolase 1 (GCH1) are also endogenous antioxidants with a strong ability to reduce lipid peroxidation and resist ferroptosis [39, 6568]. Hepatic leukemia factor (HLF) promotes ferroptosis resistance by transactivating gamma-glutamyltransferase 1 (GGT1), thus driving the progression and cisplatin resistance of TNBC [69]. A variety of indole-containing natural products (such as DM10, Brucine, DIM), through inducing the increase of Fe2+ and lipid ROS accumulation, decreasing GSH levels, and down-regulating the expression of GPX4 and SLC7A11, have been demonstrated to be novel ferroptosis regulators, with the potential to prevent ferroptosis-related cancers [70].

Ferroptosis and cellular stress response

Endoplasmic reticulum (ER) stress and ferroptosis

ER is a double-membrane organelle responsible for protein synthesis, folding, maturation, quality control and transport, and for maintaining cellular calcium homeostasis in eukaryotic cells [71]. Any abnormality in the ER protein quality control mechanism triggers a stress state named ER stress [72]. ER-associated degradation (ERAD) is the first line of quality control mechanisms that contribute to the ubiquitination and degradation of unfolded proteins through the proteasome pathway [73]. If ERAD fails, unfolded protein response (UPR) will be triggered to restore the function of the ER by inhibiting protein translation, synthesis, and restoration [7476]. If the steady-state reconstruction mechanism fails, intense or sustained ER stress will eventually lead to cell death [77]. The imbalance of ROS homeostasis may affect the correct folding of proteins and cause ER stress. Studies have shown that ER stress plays a critical role and might be the cross-link in the interactions between ferroptosis and other types of RCD [35, 78].
ER stress usually acts as a homeostatic protective mechanism that negatively regulates ferroptosis to maintain cell survival. Sorafenib inhibits system Xc and induces ER stress and ferroptosis [79]. PERK, which serves as one of the transmembrane signaling proteins during UPR, is also associated with the activation of Nrf2 [73]. In addition to the downstream signal of PERK, IRE1 and ATF4 are also deeply involved in the signal crosstalk of ferroptosis and apoptosis. In pancreatic ductal adenocarcinoma (PDAC) cells, Erastin induces phosphorylation of PERK and increases the expression of ATF4, and transcriptionally upregulates system Xc and HSPA5. HSPA5 forms a complex with GPX4 to prevent the latter from degradation, maintains cellular antioxidant capacity and inhibits ferroptosis. RNAi of HSPA5 or knockdown of ATF4 enhances Erastin-induced ferroptosis significantly [80]. Activation of the PERK-ATF4-HSPA5 pathway was also observed during dihydroartemisinin-induced ferroptosis in glioma cells [81]. In addition, the combined treatment of Erastin, RSL3 and ATF4 inhibitor promote ferroptosis of glioma cells [82].
However, it has also been reported that ER stress may act synergistically with ferroptosis to inhibit cellular activity in some cases. Erastin induces ER stress and activates the eIF2α-ATF4 pathway, and upregulates C/EBP-homologous protein (CHOP) and CHAC1, indicating that ER stress may be involved in the occurrence of ferroptosis. In prostate cancer cells, docetaxel (DTX) combined with overexpression of CHAC1 notably up-regulates GRP78 and CHOP, increasing intracellular lipid peroxidation while decreasing GPX4 protein, which significantly inhibits cell viability [83]. In mouse embryonic fibroblasts, Erastin and artesunate (ART) induce ER stress and promote p53 upregulated modulator of apoptosis (PUMA) expression via CHOP, promoting the synergistic lethal effect of Erastin/ART and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) [84]. ART induces ER stress and promotes the expression of PUMA through the PERK-eIF2α-ATF4-CHOP signaling axis, and PUMA participates in the synergistic interaction between ferroptosis and apoptosis [78]. In human intestinal epithelial cells (IEC), accumulation of iron and lipid radicals accompanied by the activation of the GRP78-PERK-ATF4-CHOP pathway was observed both in vitro and in vivo, while inhibition of PERK significantly inhibits ferroptosis [85]. These results suggest that the combined induction of ER stress and ferroptosis effectively improves the efficacy of tumor killing, and may be a novel strategy for cancer treatment.

Autophagy and ferroptosis

The imbalance of ROS homeostasis causes damage to biomacromolecules and organelles, leading to both bulk autophagy and selective autophagy. Ferritin degradation, iron chelation, lysosomal activity, p53 regulation, and the p62-Keap1-Nrf2 pathway are all involved in the activation of ferroptosis-induced autophagy [76] Studies have shown that autophagy plays an essential role in the induction of ferroptosis. Regulation of autophagy is considered to be one of the critical factors in targeting ferroptosis for antitumor therapy [86, 87]. Notably, key signaling markers of ferroptosis, including SLC7A11, GPX4, Nrf2, p53, HSPB1, CISD1, FANCD2, and ACSL4, are potential regulators of autophagy [86]. Conversely, lipid peroxides promote the formation of autophagosomes, while GPX4 over-expression inhibits ROS-mediated autophagy [88].

Macroautophagy in ferroptosis induction

Beclin-1 is involved in the ferroptosis-apoptosis interaction in human colon cancer cells, as evidenced by the fact that when co-treated with Erastin and TRAIL, Beclin-1 is cleaved by TRAIL-activated Caspase-8, leading to PUMA activation and driving apoptosis [89]. Beclin-1 can be phosphorylated by AMPK at Ser90/93/96 and forms the Beclin1-SLC7A11 complex, which inhibits the activity of system Xc and promotes lipid peroxidation. Knockdown of Beclin-1 attenuates the inhibitory effect on the system Xc of FINs (such as Erastin, sulfasalazine, and sorafenib) both in vitro and in vivo [90, 91]. Inhibition of CDGSH iron-sulfur domain 2 (CISD2) gene promotes Beclin-1involved autophagy, leading to iron accumulation, and enhances sorafenib-induced ferroptosis in HCC cells [92]. Deletion of pirin iron-binding nuclear protein (PIR) initiates autophagy through the binding of Beclin-1 to HMGB1, activates ACSL4 and promotes ferroptosis in human pancreatic cancer cells [93]. RSL3 promotes ferroptosis by mTOR inhibition and autophagy-dependent GPX4 degradation [94].
In acute lymphoblastic leukemia, knockdown of ATG5/ATG 7 or 3-MA treatment inhibits ferroptosis by blocking Hydnocarpin D (HD)-induced autophagy [95]. In malignant mesothelioma cells, NTP-activated Ringer’s lactate (PAL) induced ferroptosis is accompanied by autophagy features such as increases in LC3B-II, p62, and LAMP1 at the early stage [96]. Fingolimod (FTY720) induces ferroptosis and autophagy through the PP2A/AMPK pathway, and ferroptosis and autophagy promote each other and inhibit multiple myeloma effectively [97]. HMGB1 activates MAPKs (including MAPK/JNK and MAPK/p38) by up-regulating the expression of TFRC and positively regulates Erastin-induced ferroptosis in leukemia cells [98]. ATPR, a novel all-trans retinoic acid (ATRA) derivative, induces autophagy and regulates iron homeostasis to trigger ferroptosis in AML both in vivo and in vitro [99]. Inhibition of autophagy flux leads to accumulation of p62, promotes degradation of Keap1 and nucleus translocation of Nrf2, upregulates iron and ROS metabolism-related genes, and exerts anti-ferroptosis effect in HepG2 cells [100, 101]. The autophagy inhibitor chloroquine and the iron chelator deferoxamine attenuate Cd-induced lipid peroxidation and ferroptosis [102].
Some natural products also regulate ferroptosis by inducing autophagy. Amentoflavone (AF) activates the AMPK/mTOR-autophagy signaling pathway and triggers ferroptosis in glioma in vitro and in vivo [103]. Typhaneoside (TYP) triggers autophagic degradation of ferritin by activating AMPK signaling, resulting in ROS accumulation and ferroptosis in AML [104]. Quercetin induces transcription factor EB (TFEB)-mediated lysosomal activation and degrades ferritin, promoting ferroptosis [105]. In NSCLC, curcumin-induced autophagy leads to iron overload and mitochondrial membrane rupture, as well as up-regulation of ACSL4 and down-regulation of SLC7A11 and GPX4, resulting in GSH depletion and lipid peroxidation, ultimately leading to ferroptosis. Either chloroquine or siBeclin-1 attenuates curcumin-induced autophagy and subsequent ferroptosis [106]. 6-Gingerol inhibits the growth of NSCLC and solid tumors by inhibiting the expression of USP14 and promoting autophagy-dependent ferroptosis [107].
Nanomedicine-induced autophagy triggers ferroptosis as well. Iron-based nanomaterials can release Fe2+ or Fe3+ in acidic lysosomes and generate highly toxic ROS through the Fenton reaction [108]. Synthesized ultrasmall iron oxide nanoparticles (USIONPs) significantly upregulate ferroptosis in glioblastoma (GBM) cells via Beclin-1/Atg5-dependent autophagy. Overexpression of Beclin-1/ATG5 significantly promotes autophagy-induced ferroptosis, while autophagy or lysosome inhibitors can reverse autophagy-induced ferroptosis effectively [109]. In an in vitro and in vivo study, nanoparticle ferritin-bound Erastin and rapamycin (NFER) induce ferroptosis by down-regulating GPX4 and promoting lipid peroxide accumulation. Rapamycin potentiates autophagy-related ferroptosis and inhibits tumor progression [110]. MnO2@HMCu2-xS nanocomposites (HMCMs) exhibit photothermal (PT)-enhanced GPX4 inactivation and ferroptosis induction. Mn2+ generates ROS through the Fenton reaction and promotes the accumulation of lipid peroxides, while rapamycin promotes autophagy and further promotes the process of ferroptosis in human breast cancer models [111].
On the contrary, a few studies reported that autophagy inhibits ferroptosis activity, and inhibition of autophagy flux contributes to the induction of ferroptosis. Superparamagnetic iron oxide nanoparticles (SPIONs) lead to decreased autophagy activity but activate ferroptosis, thereby inhibiting the proliferation, invasion, drug resistance, and tumorigenicity of human ovarian cancer stem cells (HuOCSCs) [112]. In glioblastoma stem cells and neurodegenerative lysosomal storage diseases (LSD), inhibition of autophagic flux and autophagosome accumulation may lead to ferroptosis, suggesting an antagonistic relationship between autophagy and ferroptosis [10, 113]. As we know, low levels of autophagy help to remove damaged organelles and biomolecules and maintain cell survival, while intense and continuous autophagy usually causes autophagic cell death, namely type II PCD. Similarly, the regulation of autophagy on ferroptosis may also be related to the intensity of autophagy. Within a certain threshold, autophagy tends to inhibit ferroptosis, once the threshold is exceeded, autophagy switches to induce ferroptosis, thus playing an antitumor effect.

No evidence of a significant association between ferroptosis and macroautophagy

There are also studies showing that there is no apparent correlation between autophagy and ferroptosis, and their coexistence in space and time may just be a “coincidence”. The combination of siramesine and lapatinib induces both ferroptosis and autophagic death in breast cancer, but there is no evidence of a correlation between the two processes [114]. Chinese herbal medicines thymus vulgaris and arctium lappa inhibit the proliferation of leukemia and multiple myeloma cells by inducing autophagy, ferroptosis, and apoptosis. However, whether there is a direct relationship between autophagy and ferroptosis remains uncertain [115]. Betaine alkaloid ungeremine induces ferroptosis, autophagy, necrosis, and apoptosis in nine cancer cell lines by activating Caspases, altering mitochondrial membrane potential and increasing ROS, but the relationship between these cellular processes is not clear [116]. Autophagy, ferroptosis, anoikis, necrosis and ER stress all participate in the regulation and transduction of melanoma cell death, but the signaling crosstalk between autophagy and ferroptosis remains unclear [73]. In addition, autophagy activation and ferroptosis induction were also observed in some non-neoplastic diseases, such as heart failure and intracerebral hemorrhage, but there is no solid evidence of a connection between these two physiological processes [117, 118]. Therefore, more studies are still needed to elucidate the crosstalk mechanism between ferroptosis and autophagy for further understanding the function of ferroptosis in physiological processes.

Selective autophagy in ferroptosis induction

Selective autophagy, including nuclear receptor coactivator 4 (NCOA4) promoted ferritinophagy, RAB7A-dependent lipophagy, mitophagy, clockophagy, and chaperone-mediated autophagy (CMA), all play an important role in the modulation of ferroptosis [4].

Ferritinophagy

Ferritin can store up to 4500 iron atoms, which effectively avoids oxidative damage caused by iron overload in cells [98]. Ferritinophagy is a selective autophagy process that releases free iron from ferritin, leading to oxidative damage and ferroptosis via the Fenton reaction [119]. Knockdown of Atg5 inhibits Erastin-induced ferroptosis by reducing intracellular ferrous levels, suggesting that autophagy plays a critical role in ferritin degradation and ferroptosis induction [120, 121].
NCOA4-mediated ferritinophagy is the link between autophagy and ferroptosis [88, 121]. NCOA4 serves as a cargo receptor that targets ferritin to the lysosome and releases free iron by ferritinophagy. Knockdown of NCOA4 inhibits ferritin degradation and ferroptosis, whereas overexpression of NCOA4 increases ferritin degradation and promotes ferroptosis. In mouse embryonic fibroblasts, human pancreatic cancer cell lines (PANC1, PANC2.03) and human fibrosarcoma cell line (HT-1080), NCOA4 deficiency or Atg5 and Atg7 knockdown decreases intracellular labile ferrous and lipid peroxidation, thereby inhibiting Erastin-induced ferroptosis [120, 122, 123]. In glioblastoma, the knockdown of coatomer protein complex subunit zeta 1 (COPZ1) increases the expression of NCOA4, resulting in autophagic degradation of FTH1 and increased intracellular ferrous levels, thus leading to lipid peroxidation and ferroptosis [124].
In addition to the NCOA4-centered signaling axis, other signaling pathways also participate in regulating ferritinophagy. Erastin leads to ferroptosis by autophagic degradation of ferritin and induction of TFRC expression. Defects in autophagy genes BECN1 and LC3B effectively inhibit Erastin-induced ferroptosis [125]. Dihydroartemisinin promotes ferritinophagy by regulating the AMPK-mTOR-p70S6k signaling pathway, which supports the labile iron pool and ROS accumulation, resulting in the induction of ferroptosis in AML cells. Conversely, inhibition of autophagy suppresses ferroptosis in AML cells [126]. p62 promotes autophagic degradation of FPN and ferroptosis in cultured cells and xenograft mouse models [127]. In pancreatic MIN16 cells, NaAsO2-induced ferroptosis is dependent on mitochondrial reactive oxygen species (MtROS)-associated ferritinophagy [128]. Signal transducer and activator of transcription 3 (STAT3) mediates the expression and release of cathepsin B (CTSB), which is essential for lysosomal degradation of ferritin and induction ferroptosis in human pancreatic ductal adenocarcinoma (PDAC) cells [129]. Both testosterone and cisplatin can induce ferritinophagy-dependent ferroptosis, and the blockade of autophagy significantly alleviates ferroptosis [130, 131].

Lipophagy

Lipophagy is a type of selective autophagy that degrades lipid droplets (LDs) substrates by the lysosome. Free fatty acids are available as raw material for mitochondrial β-oxidation, and can be esterified to triglycerides and cholesterol to form LDs. Studies have shown that GTPase RAB7A mediates autophagic degradation of LDs increases the level of intracellular free fatty acids, and promotes RSL3-induced ferroptosis in hepatocytes. Knockout of RAB7A or Atg5 inhibits LDs degradation and reverses RSL3-induced ferroptosis in vivo and in vitro [132, 133]. Conversely, inhibition of lipophagy reduces lipid peroxidation substrates and prevents ferroptosis. Another study showed that genetically enhanced TPD52 promotes LDs formation, thereby decreasing free fatty acid levels and preventing lipid peroxidation-induced ferroptosis in hepatocytes [134].

Chaperone-mediated autophagy

CMA is also involved in the execution of ferroptosis. It has been shown that Erastin increases the level of lysosome-associated membrane protein-2a (Lamp-2a) and promotes CMA, which in turn promotes GPX4 degradation and stimulates ferroptosis, while inhibition of CMA stabilizes GPX4 and reduces ferroptosis [135]. HSC70 can also mediate CMA by interacting with Lamp-2a, knockout of HSPA8/HSC70 and Lamp-2a restores GPX4 levels and alleviates Erastin-induced ferroptosis [135, 136].

Clockophagy

Mechanistically, the core circadian clock regulator aryl hydrocarbon receptor nuclear translocator-like (ARNTL) inhibits ferroptosis by inhibiting the transcription of Egln2 and activating the pro-survival transcription factor HIF1A. In vivo and in vitro experiments on human fibrosarcoma cells have shown that FINs (RSL3 and FIN56) evoke p62-dependent autophagic degradation of ARNTL, negatively regulating HIF1 and preventing HIF1A-dependent fatty acid uptake and lipid storage, which promotes lipid peroxidation and eventually ferroptosis [137]. Another study showed that the knockout of SQSTM1 prevents the degradation of ARNTL by clockophagy, thereby exerting an anti-ferroptosis effect [138].

Mitophagy

Eukaryotic mitochondria generate most of the energy (ATP) through oxidative phosphorylation, which maintains the material circulation and energy metabolism of cells. Mitochondria are also major sites of ROS production if their structure is damaged, thus mitochondria are involved in regulating various forms of RCD [123, 139]. Fortunately, mitophagy can break down dysfunctional mitochondria, maintain ROS homeostasis and prevent apoptosis and lipid peroxidation-related ferroptosis [10, 11]. However, there is also a study showing that BAY 87-2243 induces mitophagy-dependent escalation of lipid ROS and ferroptosis in BRAF (V600E) melanoma cells both in vitro and in vivo [140]. Therefore, the role of mitophagy in the regulation of ferroptosis still needs to be further explored.

Role of ferroptosis in radiotherapy

Biological effects of ionizing radiation

Ionizing radiation directly causes DNA damage through the photoelectric effect and the Compton effect. Single-strand breaks (SSBs) can easily be repaired by the damage repair machinery in cells, while double-strand breaks (DSBs) usually lead to cell death, which is the core mechanism of ionizing radiation to kill tumors in the traditional sense. Another aspect that cannot be neglected is that ionizing radiation acts on water molecules to produce unstable free radicals, which causes damage to biological macromolecules such as DNA and protein through indirect effects. Ionizing radiation also activates autoimmunity, with potential advantages in combination with immunotherapy [141].
With the deepening of research, it has been found that ionizing radiation induces ferroptosis in apoptosis-resistant cells, or simultaneously induces mixed-RCD, including both apoptosis and ferroptosis, which will undoubtedly be more helpful in killing tumors and improving the tumor-suppressive effect [142]. Ionizing radiation causes water radiolysis to produce hydroxyl radicals (·OH), a highly reactive oxidant that serves as the main inducer of phospholipid peroxidation. By altering the intake and excretion of iron through FPN and TFRC, ionizing radiation also regulates iron metabolism and homeostasis [51]. The accumulation of labile iron in the cytoplasm produces a Fenton reaction with radiation-induced hydrogen peroxide (Fenton’s reagent), further increasing the level of ·OH. ·OH reacts with DNA, lipids, carbohydrates, and proteins, leading to DNA breaks, protein oxidation and glycation, and ferroptosis finally.

Radiotherapy induces ferroptosis in cancer cells through a variety of mechanisms

Up to now, the basic and applied research on radiation-induced ferroptosis mainly focuses on sparse ionizing radiations such as X- and γ-rays. Several studies have reported that ionizing radiation decreases the levels of SLC7A11, GSH, and GPX4 in cells [143, 144]. One of the mechanisms is that ionizing radiation activates cyclic GMP-AMP synthase (cGAS) signaling, which leads to autophagy-dependent ferroptosis [12] (Fig. 2). Another mechanism is radiation-induced ROS activates ATM and ACSL4 expression, then ATM mediates SLC7A11 downregulation, and ACSL4 increases PUFA-PLs formation for lethal lipid peroxidation. Therefore, ACSL4 ablation eliminates ionizing radiation-induced ferroptosis and promotes radioresistance [145]. In addition, ionizing radiation also induces tumor cells to release irradiated tumor cell-released microparticles (RT-MPs) and exosomes, thereby causing ferroptosis through the bystander effect. Interestingly, there is also evidence that the expression of SLC7A11 and GPX4 induced by ionizing radiation is a stress response, suggesting that oxidative stress caused by radiation serves as a double-edged sword, whether cells ultimately achieve homeostasis reconstruction or heading for ferroptosis, depending mainly on the extent and duration of membrane lipid damage.
Dense ionizing radiation such as heavy ions (especially carbon ions) has physical and biological advantages and exhibits excellent tumor-killing performance, and thus is more suitable for the treatment of locally non-proliferating tumors, tumors that are resistant to conventional radiation, and tumors located near critical tissues and organs that are difficult to surgically remove, especially chordoma, lung cancer, liver cancer, prostate cancer, bone and soft tissue sarcoma, etc. [141, 146149]. In addition, heavy ions cause cell death in various ways, inducing apoptosis, necrosis, autophagy, premature aging, accelerated differentiation, bystander effects, and ferroptosis, thus exerting a significant role in tumor inhibition. However, there still are relatively few studies focused on heavy ion-induced ferroptosis. Our recent study showed that ionizing radiation activates ER stress and induces ferroptosis accompanied by apoptosis through the PERK and p53-involved pathways, and that mixed-RCD plays a significant role in cancer suppression [150].

FINs enhance the radiosensitivity of cancer cells in vivo and in vitro

Inactivation of SLC7A11 or GPX4 by FINs (including sulfasalazine, RSL3, ML162, FIN56) enhances the radiosensitivity of cancer cells and xenograft tumors to X-rays, and usually portends a better response and prognosis after radiotherapy [145]. Erastin inhibits GPX4 expression and enhances the radiosensitivity of breast, cervical, and NSCLC to X-rays by inducing ferroptosis [151, 152]. Erastin also increases the sensitivity of breast cancer cells to γ-rays. In human lung adenocarcinoma and glioma patient models, FINs (IKE, RSL3, Sorafenib) and radiation induce ferroptosis synergistically, enhancing the tumor suppression effect of ionizing radiation [4]. Itraconazole enhances the radiosensitivity of nasopharyngeal carcinoma by inducing ferroptosis. In addition, overexpression of Collectrin (CLTRN) promotes ferroptosis by down-regulating GPX4, SLC7A11, and FTH1, thereby enhancing the radiosensitivity of HCC cells to X-rays both in vitro and in vivo [153]. These results suggest that FINs-induced ferroptosis act synergistically with ionizing radiation-induced apoptosis, providing new theoretical support for clinical medicine combined with radiotherapy in treating tumors.

Role of ferroptosis in immunotherapy

Cytotoxic T lymphocytes-driven immunity induces ferroptosis

Immune checkpoint blocking therapy has become one of the most prevalent cancer treatments in recent years, and the immune system can suppress tumors to some extent by inducing ferroptosis. Ferroptosis promoted by cytotoxic T lymphocytes is a potential antitumor mechanism, and targeting ferroptosis synergized with immune checkpoint inhibition is a potentially effective cancer therapy strategy [154]. In ovarian cancer and melanoma-bearing mouse models, interferon (IFN)-γ released by immunotherapy-activated CD8+ T cells down-regulates the expression of SLC7A11 and SLC3A2, inhibits cystine uptake in tumor cells, promotes lipid peroxidation and ferroptosis, thereby increasing the antitumor efficacy of immunotherapy (Fig. 3). In vivo, cystine/cysteine depletion combined with checkpoint blockade synergistically enhances T cell-mediated antitumor immunity and induces tumor cell ferroptosis [155]. Another study showed that IFN-γ inhibits the activity of system Xc via JAK-STAT signaling, increasing ROS levels and reducing mitochondrial membrane potential, sensitizing HCC cells to ferroptosis [156]. Nivolumab increases IFN-γ while decreasing SLC3A2 expression, exhibiting a better clinical prognosis of cancer. Hemoglobin plus photodynamic therapy (PDT) treatment releases IFN-γ from T lymphocytes, enhancing the sensitivity of tumor cells to ferroptosis and promoting tumor suppression [157]. It's worth noting that immunotherapy also sensitizes tumors to radiotherapy by promoting ferroptosis, which sheds light on a new treatment approach for tumors that combines radiotherapy with immunotherapy. Mechanistically, IFN-γ derived from immunotherapy-activated CD8+ T cells and radiotherapy-activated ATM synergistically inhibits SLC7A11, resulting in decreased cystine uptake and enhanced lipid peroxidation, and subsequently leading to ferroptosis to improve tumor suppression [143, 155].

Ferroptosis regulates antitumor immunity

With the increase of ferroptosis-related studies, it has been realized that ferroptosis is immunogenic and can activate innate immune [158, 159]. Moreover, damage-associated molecular patterns (DAMPs) or lipid metabolites released by ferroptotic cells activate the immune responses of adjacent immune cells and adaptive immunity-driven cytotoxic T lymphocytes, thus confirming that ferroptosis is a type of immunogenic cell death (ICD) [158, 160162]. Ferroptotic cells induce ICD by releasing ATP and HMGB1, or by regulating HSP70 and HSP90, contributing to the suppression of colon cancer [163]. The release of HMGB1 during ferroptosis leads to the up-regulation of tumor necrosis factor-alpha (TNFα) in macrophages and sustains the immune response [164]. In murine glioma or fibrosarcoma cells, ATP and HMGB1 released by ferroptotic cells can be phagocytosed by bone-marrow-derived dendritic cells (BMDCs), which subsequently mature to produce IL-6 and promote tumor suppression [165]. RSL3-induced ferroptosis leads to ICD in an ATP and P2X7-dependent manner, which promotes phenotypic maturation of BMDCs, and produces vaccine-like effects in immunocompetent mice [166, 167].
SAPE-OOH (1-Stearoyl-2′-15-HpETE-sn-glycero-3-phosphatidylethanolamine), a phospholipid that acts as an ‘eat-me’ signal on the surface of ferroptotic cells, causing TLR2-mediated phagocytosis in mouse mammary tumor [168]. In mouse xenografts of head and neck squamous cell carcinoma (HNSCC), ferroptosis significantly reduces the number of myeloid-derived suppressor cells (MDSCs) and tumor-associated M2-like macrophages (M2 TAMs), while increasing tumor-infiltrating CD4+ and CD8+ T cells in the tumor microenvironment, indicating that ferroptosis can enhance antitumor immune responses by reversing the immunosuppressive microenvironment [169, 170]. Immunogenic ferroptosis induced by BEBT-908 (PI3K inhibitor) leads to upregulation of major histocompatibility complex I (MHC-I) and activation of endogenous IFN-γ via STAT1, thereby activating the antitumor immune response and enhancing anti-PD1 therapy, effectively inhibits tumor growth [171]. The abnormality of ferroptosis regulator STEAP3 may also be involved in various immune-related signatures in the pathogenesis of HCC [172]. Overall, it is obvious that ferroptosis does have significant immunogenicity and may play a positive role in antitumor immunity.

Ferroptosis leads to dysfunction of immune-related cells

There are also studies demonstrating that ferroptosis of cancer cells is not immunogenic cell death, despite the release of immunogenicity-related DAMPs and cytokines [173].
On the one hand, ferroptosis may inhibit the survival of antitumor immune cells (including DC, NK, and CD8+T cells), resulting in immunodeficiency. Dendritic cells (DCs) are the most effective antigen-presenting cells required for T cell activation and maintenance of T cell-dependent immunity. Recent studies revealed that ferroptosis negatively regulates the maturation of DCs, leads to abnormal antigen-presentation, and inhibits the proliferation and activation of CD8+T cells, thereby hindering antitumor immunity in vitro and in vivo [174, 175]. One of the mechanisms is that peroxisome proliferator-activated receptor gamma (PPARG)-mediated ferroptosis of DCs elicits impaired production of cytokines (TNF and IL6), decreased expression of MCH-I, and inhibition of IFN-γ production by CD8+T cells, thus driving the formation of an immunosuppressive tumor microenvironment (TME). In addition, normal DCs loaded with ferroptotic cancer cells lose their ability to suppress tumor growth.
In lipid peroxides-rich TME, ferroptosis due to aberrant lipid accumulation reduces the antigen-presentation and antitumor ability of DCs, thereby promoting cancer progression [133, 176, 177]. Ferroptotic cancer cells may significantly induce the dysfunction of CD8+T and CD4+T cells, and disrupt the maturation and function of DCs, thus reducing the effectiveness of immune checkpoint inhibitor (ICI) therapy [175, 177].
NK cells play the role of antitumor immunity, and the defects of NK cells lead to increased rates of tumorigenesis and progression [178]. Lipid peroxidation-induced ferroptosis inhibits glucose metabolism of NK cells, resulting in NK cell dysfunction, while NRF2 activation alleviates ferroptosis and rescues antitumor activity of NK cells in vivo [179, 180].
Moreover, contrary to previous views, DAMPs released by ferroptotic cells may also exert immunosuppressive effects under certain circumstances. 8-OHG released from ferroptotic pancreatic cells activates the STING-dependent DNA sensor pathway, leading to tumor-associated macrophages (TAMs) infiltration and M2 polarization, promoting Kras-driven tumorigenesis and inflammation-related immunosuppression [162, 181, 182]. Prostaglandin E2 (PGE2) released by ferroptotic cancer cells serves as a critical immunosuppressor that can inhibit the antitumor function of DCs, NK cells, and cytotoxic T lymphocytes [183].
Ferroptosis of T lymphocytes inevitably leads to dysfunction of CD8+T and CD4+T cells [142, 184]. Lipid peroxide accumulation and ferroptosis caused by GPX4 deficiency impair the ability of T lymphocytes to eradicate viral or parasitic infections [184]. Conversely, overexpression of GPX4 or FSP1 inhibits the ferroptosis of T lymphocytes and maintains their immune effect [185]. In addition, immune checkpoint inhibitors sensitize cancer cells to ferroptosis, which leads to tumor immune escape by directly suppressing the function of immune cells [186, 187]. The fatty acid transporter CD36 enhances the sensitivity of CD8+ T cells to ferroptosis by promoting lipid peroxidation, impairing the production of cytotoxic cytokine and antitumor activity. Inhibition of ferroptosis by blocking CD36 restores the antitumor activity of CD8+ T cells [187].
On the other hand, some immunosuppressive immune cells, such as anti-inflammatory M2 TAMs and Tregs, require protection against ferroptosis to maintain cell viability, and ferroptosis induction in these cells may reverse their immunosuppressive function [178]. Tregs are essential for maintaining immune tolerance and suppressing antitumor immunity; thus an elevated proportion of Tregs is usually associated with poor prognosis and poor survival in cancer patients. Inhibition of Gpx4 in Tregs leads to lipid peroxides accumulation and ferroptosis, thereby enhancing the efficacy of immunotherapy in tumors with high Treg infiltration [188].
It is reasonable to believe that the immunogenicity of ferroptosis and its impact on antitumor immunity is closely related to tumor stage and TME. Exploring the relationship between ferroptosis and immunity will contribute to developing more effective cancer treatment strategies, and further studies are urgently needed.

Nanoparticles: ingenious attempts to target the combination of ferroptosis and immunomodulation

Currently, lots of studies have focused on the synergistic induction of ferroptosis and antitumor immunity by targeting nanoparticles to promote tumor inhibition (Fig. 4). In the mouse 4T1 metastasis model, the biomimetic magnetic nanoparticles Fe3O4-SAS@PLT trigger ferroptosis by inhibiting system Xc, which induces not only tumor-specific immune responses, but also effectively re-polarize macrophages from immunosuppressive M2 to antitumor M1, improving the efficacy of PD1 immune checkpoint blocking therapy and achieving tumor elimination [189]. Iron oxide-loaded nano-vaccines (IONVs)-mediated tumor microenvironment modification elevates ROS and induces ferroptosis, simultaneously activates M1 macrophages and promotes tumor antigens cross-presentation, completely eradicates aggressiveness and established tumors, and stimulates long-term protective antitumor immunity [190].
In addition to ferroptosis induction, hybrid core–shell vesicles (HCSVs)-induced Fenton reaction exposes calreticulin to tumor cells, leading to maturation of dendritic cells (DC) and tumor infiltration of cytotoxic T lymphocytes, thereby resulting in significant tumor suppression [191]. Ultrasmall single-crystal Fe nanoparticles (BCC-USINPs) simultaneously induce ferroptosis and ICD, promoting DC maturation and triggering adaptive T-cell infiltration by releasing immunogenic DMAPs, enabling ferroptosis-enhanced tumor immunotherapy [192]. Metal-phenolic networks (MPNs) nano-platform synergistically enhances ICD with ferroptosis and promotes primary and distant tumor killing in combination with DNAzyme-mediated PD-L1 inhibition [193]. A study of cancer cell membrane coated RSL3-loaded gold nanocages (C-RAuNC) revealed that RSL3-induced ferroptosis synergistically initiates antitumor immunity with photothermal therapy (PTT)-induced ICD, thereby inhibiting the growth and recurrence of osteosarcoma [194].
Nanoparticles containing ferritin and pH-sensitive molecular switch (FPBC@SN) release sorafenib and NLG919 in acidic cytoplasm. Sorafenib induces ferroptosis by blocking GSH synthesis, down-regulating GPX4, and increasing endogenous iron pools. NLG919 stimulates antitumor immunity by reducing indoleamine-2,3-dioxygenase (IDO)-regulated tryptophan metabolism, synergistically inhibiting cell growth and metastasis of breast cancer in vitro and in vivo [195]. Doxorubicin (DOX) and β-lapachone (LAP) are effective inducers of ICD, and they work synergistically with iron in tumor inhibition. Tumor-targeted delivery of DOX-Fe2+ complexes initiates antitumor immunity and induces ICD, effectively integrating tumor metabolism and immune response, exerting a potential antitumor tumor strategy [196, 197]. LAP-Fe2+ complexes synergistically trigger ferroptosis and ICD in tumor cells in a remotely controllable manner [198]. Biomimetic magnetosomes composed of Fe3O4 magnetic nanoclusters, TGF-β inhibitors, and PD-1 antibodies can achieve synergistic cancer treatment through ferroptosis and immunomodulatory [199]. In another study, nanocarriers of exosome inhibitors (GW4869) and Fe3+ enhance the response of melanoma to PD-L1 checkpoint blockade both in vivo and in vitro, representing an effective therapeutic strategy. Mechanistically, cooperation between GW4869 and Fe3+ in nano-units induces ferroptosis and antitumor immune responses against melanoma by reducing exosomes and attenuating exosomal PD-L1, and stimulating cytotoxic T lymphocytes and immune memory [200].
Overall, nanoparticles that simultaneously induce ferroptosis and ICD exhibit great potential in tumor suppression, which may bring new opportunities for the clinical treatment of tumors. However, the in vivo stability, active and passive targeting capabilities of nanoparticles, and their metabolic kinetics and toxicity, require further investigation.

Conclusions and perspectives

As a novel iron-dependent non-apoptotic RCD, ferroptosis has attracted extensive attention since its discovery. Ferroptosis is deeply involved in the regulation of cellular stress processes such as ER stress and autophagy, and affects the occurrence and development of cancer through a complex and sophisticated signal transduction network. Currently, targeting ferroptosis is emerging as a potentially effective approach for the treatment of various diseases, especially cancer and ischemic organ damage. Fortunately, available evidence suggests that ferroptosis and other known types of RCDs do not appear to be antagonistic to each other, but may coexist and interfere with each other, and act synergistically in cell fate determination. Therefore, the induction of mixed RCD can undoubtedly be an effective therapeutic strategy to break the bottleneck of the limitations of radiotherapy alone. In addition, the reciprocal regulatory feedback between ferroptosis and immune responses may further exert tumor suppressive effects. Furthermore, targeting ferroptosis using small molecule compounds, clinical drugs, and nanoparticles is also a potentially effective antitumor strategy in clinical practice, especially in iron-rich tumors that are prone to ferroptosis.
However, the existing understanding of ferroptosis remains insufficient. First, the execution mechanism of ferroptosis is still unclear. Lethal plasma membrane damage due to lipid peroxidation is the direct cause of ferroptosis; however, whether other unknown downstream molecules exist to directly regulate and execute ferroptosis, like a role BCL family proteins or Caspase-3 served for apoptosis, is still elusive. Therefore, cracking the execution mechanism of ferroptosis remains one of the critical issues in the next few years.
Second, the selective mechanism of ferroptosis remains poorly understood. At present, it is still at a loss when mentioned to selectively activate or inhibit ferroptosis in specific tissues and cells in different diseases. Whether pharmacological intervention can selectively inhibit tumor cells without causing severe toxic side effects on normal cells remains to be verified. Comprehensively understanding the susceptibility mechanism of cancer cells and immune cells to ferroptosis, selectively inhibiting or promoting ferroptosis of immune cells, and creating an antitumor immune microenvironment are still important issues to be solved urgently.
Third, definitive biomarkers for ferroptosis have not been identified. The search for predictive clinical biomarkers of ferroptosis, especially those that can be directly and rapidly detected in patient body fluids and biopsy samples, will be of fundamental importance in determining the physiological function and therapeutic potential of ferroptosis. Thus, finding biomarkers to facilitate the accurate detection of ferroptosis will also be an active area of research in the coming years.
Fourth, the clinical translation potential of targeted ferroptosis therapy needs to be validated. Based on the results obtained so far, approaches that combine FINs with other therapies, such as radiotherapy and immunotherapy, to improve the treatment effect have great potential for clinical translation. Unfortunately, most FINs exhibit poor pharmacology and pharmacokinetics in animal models, which directly limits their clinical application. Existing medicines, such as Sorafenib, Lapatinib, Sulfasalazine, Cisplatin, etc., can effectively induce ferroptosis [79, 201203]. However, a recent study showed that sorafenib is not qualified as an effective ferroptosis inducer, and ferroptosis induced by system Xc inhibitor can only be achieved in a subset of tumor cell lines [204]. Thus, under the premise of rigorous evaluation, innovative uses of these medicines in combination with other therapies to target ferroptosis may acquire great potential in specific types of tumor therapy. In addition, nanoparticles coated with specific ligands and carrying targeted drugs also have good potential for clinical translation. However, the stability, targeting capabilities, kinetics, and toxicity of nanocarriers still need to be intensively studied in vivo.
Addressing these unanswered questions promises to yield new insights, not only for ferroptosis research itself, but also for the biomedical frontiers associated with this novel RCD. It is believed that in the next few years, there will be exciting discoveries in the field of ferroptosis-related research. Based on specific biomarkers and precise patient-specific assessment, targeting ferroptosis will be translated into a practical new approach to clinical cancer therapy, significantly contributing to the prevention, diagnosis, prognosis, and treatment of cancer.

Acknowledgements

Not applicable.

Declarations

Not applicable.
Not applicable.

Competing interests

The authors declare that they have no competing interests.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Dixon SJ, Lemberg KM, Lamprecht MR, Skouta R, Zaitsev EM, Gleason CE, et al. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell. 2012;149(5):1060–72.PubMedPubMedCentralCrossRef Dixon SJ, Lemberg KM, Lamprecht MR, Skouta R, Zaitsev EM, Gleason CE, et al. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell. 2012;149(5):1060–72.PubMedPubMedCentralCrossRef
2.
Zurück zum Zitat Jiang XJ, Stockwell BR, Conrad M. Ferroptosis: mechanisms, biology and role in disease. Nat Rev Mol Cell Bio. 2021;22(4):266–82.CrossRef Jiang XJ, Stockwell BR, Conrad M. Ferroptosis: mechanisms, biology and role in disease. Nat Rev Mol Cell Bio. 2021;22(4):266–82.CrossRef
4.
Zurück zum Zitat Zhou BR, Liu J, Kang R, Klionsky DJ, Kroemer G, Tang DL. Ferroptosis is a type of autophagy-dependent cell death. Semin Cancer Biol. 2020;66:89–100.PubMedCrossRef Zhou BR, Liu J, Kang R, Klionsky DJ, Kroemer G, Tang DL. Ferroptosis is a type of autophagy-dependent cell death. Semin Cancer Biol. 2020;66:89–100.PubMedCrossRef
5.
Zurück zum Zitat Sun X, Ou Z, Chen R, Niu X, Chen D, Kang R, et al. Activation of the p62-Keap1-NRF2 pathway protects against ferroptosis in hepatocellular carcinoma cells. Hepatology. 2016;63(1):173–84.PubMedCrossRef Sun X, Ou Z, Chen R, Niu X, Chen D, Kang R, et al. Activation of the p62-Keap1-NRF2 pathway protects against ferroptosis in hepatocellular carcinoma cells. Hepatology. 2016;63(1):173–84.PubMedCrossRef
6.
Zurück zum Zitat Tong X, Tang R, Xiao M, Xu J, Wang W, Zhang B, et al. Targeting cell death pathways for cancer therapy: recent developments in necroptosis, pyroptosis, ferroptosis, and cuproptosis research. J Hematol Oncol. 2022;15(1):174.PubMedPubMedCentralCrossRef Tong X, Tang R, Xiao M, Xu J, Wang W, Zhang B, et al. Targeting cell death pathways for cancer therapy: recent developments in necroptosis, pyroptosis, ferroptosis, and cuproptosis research. J Hematol Oncol. 2022;15(1):174.PubMedPubMedCentralCrossRef
7.
Zurück zum Zitat Tsvetkov P, Coy S, Petrova B, Dreishpoon M, Verma A, Abdusamad M, et al. Copper induces cell death by targeting lipoylated TCA cycle proteins. Science. 2022;375(6586):1254–61.PubMedPubMedCentralCrossRef Tsvetkov P, Coy S, Petrova B, Dreishpoon M, Verma A, Abdusamad M, et al. Copper induces cell death by targeting lipoylated TCA cycle proteins. Science. 2022;375(6586):1254–61.PubMedPubMedCentralCrossRef
8.
Zurück zum Zitat Yang WS, SriRamaratnam R, Welsch ME, Shimada K, Skouta R, Viswanathan VS, et al. Regulation of ferroptotic cancer cell death by GPX4. Cell. 2014;156(1–2):317–31.PubMedPubMedCentralCrossRef Yang WS, SriRamaratnam R, Welsch ME, Shimada K, Skouta R, Viswanathan VS, et al. Regulation of ferroptotic cancer cell death by GPX4. Cell. 2014;156(1–2):317–31.PubMedPubMedCentralCrossRef
9.
10.
Zurück zum Zitat Pierzynowska K, Rintz E, Gaffke L, Wegrzyn G. Ferroptosis and its modulation by autophagy in light of the pathogenesis of lysosomal storage diseases. Cells. 2021;10(2):365.PubMedPubMedCentralCrossRef Pierzynowska K, Rintz E, Gaffke L, Wegrzyn G. Ferroptosis and its modulation by autophagy in light of the pathogenesis of lysosomal storage diseases. Cells. 2021;10(2):365.PubMedPubMedCentralCrossRef
11.
Zurück zum Zitat Liu J, Kuang F, Kroemer G, Klionsky DJ, Kang R, Tang D. Autophagy-dependent ferroptosis: machinery and regulation. Cell Chem Biol. 2020;27(4):420–35.PubMedPubMedCentralCrossRef Liu J, Kuang F, Kroemer G, Klionsky DJ, Kang R, Tang D. Autophagy-dependent ferroptosis: machinery and regulation. Cell Chem Biol. 2020;27(4):420–35.PubMedPubMedCentralCrossRef
12.
Zurück zum Zitat Chen X, Kang R, Kroemer G, Tang D. Broadening horizons: the role of ferroptosis in cancer. Nat Rev Clin Oncol. 2021;18(5):280–96.PubMedCrossRef Chen X, Kang R, Kroemer G, Tang D. Broadening horizons: the role of ferroptosis in cancer. Nat Rev Clin Oncol. 2021;18(5):280–96.PubMedCrossRef
13.
Zurück zum Zitat Yien YY, Shi JH, Chen CY, Cheung JTM, Grillo AS, Shrestha R, et al. FAM210B is an erythropoietin target and regulates erythroid heme synthesis by controlling mitochondrial iron import and ferrochelatase activity. J Biol Chem. 2018;293(51):19797–811.PubMedPubMedCentralCrossRef Yien YY, Shi JH, Chen CY, Cheung JTM, Grillo AS, Shrestha R, et al. FAM210B is an erythropoietin target and regulates erythroid heme synthesis by controlling mitochondrial iron import and ferrochelatase activity. J Biol Chem. 2018;293(51):19797–811.PubMedPubMedCentralCrossRef
14.
Zurück zum Zitat Zohora F, Bidad K, Pourpak Z, Moin M. Biological and immunological aspects of iron deficiency anemia in cancer development: a narrative review. Nutr Cancer. 2018;70(4):546–56.PubMedCrossRef Zohora F, Bidad K, Pourpak Z, Moin M. Biological and immunological aspects of iron deficiency anemia in cancer development: a narrative review. Nutr Cancer. 2018;70(4):546–56.PubMedCrossRef
15.
Zurück zum Zitat Dixon SJ, Stockwell BR. The role of iron and reactive oxygen species in cell death. Nat Chem Biol. 2014;10(1):9–17.PubMedCrossRef Dixon SJ, Stockwell BR. The role of iron and reactive oxygen species in cell death. Nat Chem Biol. 2014;10(1):9–17.PubMedCrossRef
16.
Zurück zum Zitat Winterbourn CC. Toxicity of iron and hydrogen peroxide: the Fenton reaction. Toxicol Lett. 1995;82–83:969–74.PubMedCrossRef Winterbourn CC. Toxicity of iron and hydrogen peroxide: the Fenton reaction. Toxicol Lett. 1995;82–83:969–74.PubMedCrossRef
17.
Zurück zum Zitat Wu W, Geng ZX, Bai HR, Liu T, Zhang BM. Ammonium ferric citrate induced ferroptosis in non-small-cell lung carcinoma through the inhibition of GPX4-GSS/GSR-GGT axis activity. Int J Med Sci. 2021;18(8):1899–909.PubMedPubMedCentralCrossRef Wu W, Geng ZX, Bai HR, Liu T, Zhang BM. Ammonium ferric citrate induced ferroptosis in non-small-cell lung carcinoma through the inhibition of GPX4-GSS/GSR-GGT axis activity. Int J Med Sci. 2021;18(8):1899–909.PubMedPubMedCentralCrossRef
19.
Zurück zum Zitat Li Y, Zeng X, Lu D, Yin M, Shan M, Gao Y. Erastin induces ferroptosis via ferroportin-mediated iron accumulation in endometriosis. Hum Reprod. 2021;36(4):951–64.PubMedCrossRef Li Y, Zeng X, Lu D, Yin M, Shan M, Gao Y. Erastin induces ferroptosis via ferroportin-mediated iron accumulation in endometriosis. Hum Reprod. 2021;36(4):951–64.PubMedCrossRef
20.
Zurück zum Zitat Yue RY, Zhang C, Xu L, Wang YJ, Guan GQ, Lei LL, et al. Dual key co-activated nanoplatform for switchable MRI monitoring accurate ferroptosis-based synergistic therapy. Chem. 2022;8(7):1956–81.CrossRef Yue RY, Zhang C, Xu L, Wang YJ, Guan GQ, Lei LL, et al. Dual key co-activated nanoplatform for switchable MRI monitoring accurate ferroptosis-based synergistic therapy. Chem. 2022;8(7):1956–81.CrossRef
21.
Zurück zum Zitat Chang LC, Chiang SK, Chen SE, Yu YL, Chou RH, Chang WC. Heme oxygenase-1 mediates BAY 11-7085 induced ferroptosis. Cancer Lett. 2018;416:124–37.PubMedCrossRef Chang LC, Chiang SK, Chen SE, Yu YL, Chou RH, Chang WC. Heme oxygenase-1 mediates BAY 11-7085 induced ferroptosis. Cancer Lett. 2018;416:124–37.PubMedCrossRef
22.
Zurück zum Zitat Fang XX, Wang H, Han D, Xie EJ, Yang X, Wei JY, et al. Ferroptosis as a target for protection against cardiomyopathy. Proc Natl Acad Sci USA. 2019;116(7):2672–80.PubMedPubMedCentralCrossRef Fang XX, Wang H, Han D, Xie EJ, Yang X, Wei JY, et al. Ferroptosis as a target for protection against cardiomyopathy. Proc Natl Acad Sci USA. 2019;116(7):2672–80.PubMedPubMedCentralCrossRef
23.
Zurück zum Zitat Menon AV, Liu J, Tsai HP, Zeng L, Yang S, Asnani A, et al. Excess heme upregulates heme oxygenase 1 and promotes cardiac ferroptosis in mice with sickle cell disease. Blood. 2022;139(6):936–41.PubMedPubMedCentralCrossRef Menon AV, Liu J, Tsai HP, Zeng L, Yang S, Asnani A, et al. Excess heme upregulates heme oxygenase 1 and promotes cardiac ferroptosis in mice with sickle cell disease. Blood. 2022;139(6):936–41.PubMedPubMedCentralCrossRef
24.
Zurück zum Zitat Tang Z, Ju Y, Dai X, Ni N, Liu Y, Zhang D, et al. HO-1-mediated ferroptosis as a target for protection against retinal pigment epithelium degeneration. Redox Biol. 2021;43: 101971.PubMedPubMedCentralCrossRef Tang Z, Ju Y, Dai X, Ni N, Liu Y, Zhang D, et al. HO-1-mediated ferroptosis as a target for protection against retinal pigment epithelium degeneration. Redox Biol. 2021;43: 101971.PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Wu A, Feng B, Yu J, Yan L, Che L, Zhuo Y, et al. Fibroblast growth factor 21 attenuates iron overload-induced liver injury and fibrosis by inhibiting ferroptosis. Redox Biol. 2021;46: 102131.PubMedPubMedCentralCrossRef Wu A, Feng B, Yu J, Yan L, Che L, Zhuo Y, et al. Fibroblast growth factor 21 attenuates iron overload-induced liver injury and fibrosis by inhibiting ferroptosis. Redox Biol. 2021;46: 102131.PubMedPubMedCentralCrossRef
26.
Zurück zum Zitat Meng Z, Liang H, Zhao J, Gao J, Liu C, Ma X, et al. HMOX1 upregulation promotes ferroptosis in diabetic atherosclerosis. Life Sci. 2021;284: 119935.PubMedCrossRef Meng Z, Liang H, Zhao J, Gao J, Liu C, Ma X, et al. HMOX1 upregulation promotes ferroptosis in diabetic atherosclerosis. Life Sci. 2021;284: 119935.PubMedCrossRef
27.
Zurück zum Zitat Adedoyin O, Boddu R, Traylor A, Lever JM, Bolisetty S, George JF, et al. Heme oxygenase-1 mitigates ferroptosis in renal proximal tubule cells. Am J Physiol-Renal. 2018;314(5):F702–14.CrossRef Adedoyin O, Boddu R, Traylor A, Lever JM, Bolisetty S, George JF, et al. Heme oxygenase-1 mitigates ferroptosis in renal proximal tubule cells. Am J Physiol-Renal. 2018;314(5):F702–14.CrossRef
28.
Zurück zum Zitat Ayala A, Munoz MF, Arguelles S. Lipid peroxidation: production, metabolism, and signaling mechanisms of malondialdehyde and 4-hydroxy-2-nonenal. Oxid Med Cell Longev. 2014;2014: 360438.PubMedPubMedCentralCrossRef Ayala A, Munoz MF, Arguelles S. Lipid peroxidation: production, metabolism, and signaling mechanisms of malondialdehyde and 4-hydroxy-2-nonenal. Oxid Med Cell Longev. 2014;2014: 360438.PubMedPubMedCentralCrossRef
29.
Zurück zum Zitat Stockwell BR, Friedmann Angeli JP, Bayir H, Bush AI, Conrad M, Dixon SJ, et al. Ferroptosis: a regulated cell death nexus linking metabolism, redox biology, and disease. Cell. 2017;171(2):273–85.PubMedPubMedCentralCrossRef Stockwell BR, Friedmann Angeli JP, Bayir H, Bush AI, Conrad M, Dixon SJ, et al. Ferroptosis: a regulated cell death nexus linking metabolism, redox biology, and disease. Cell. 2017;171(2):273–85.PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Yang WS, Kim KJ, Gaschler MM, Patel M, Shchepinov MS, Stockwell BR. Peroxidation of polyunsaturated fatty acids by lipoxygenases drives ferroptosis. Proc Natl Acad Sci USA. 2016;113(34):E4966–75.PubMedPubMedCentralCrossRef Yang WS, Kim KJ, Gaschler MM, Patel M, Shchepinov MS, Stockwell BR. Peroxidation of polyunsaturated fatty acids by lipoxygenases drives ferroptosis. Proc Natl Acad Sci USA. 2016;113(34):E4966–75.PubMedPubMedCentralCrossRef
31.
Zurück zum Zitat Kagan VE, Mao GW, Qu F, Angeli JPF, Doll S, St Croix C, et al. Oxidized arachidonic and adrenic pes navigate cells to ferroptosis. Nat Chem Biol. 2017;13(1):81–90.PubMedCrossRef Kagan VE, Mao GW, Qu F, Angeli JPF, Doll S, St Croix C, et al. Oxidized arachidonic and adrenic pes navigate cells to ferroptosis. Nat Chem Biol. 2017;13(1):81–90.PubMedCrossRef
32.
Zurück zum Zitat Dixon SJ, Winter GE, Musavi LS, Lee ED, Snijder B, Rebsamen M, et al. Human haploid cell genetics reveals roles for lipid metabolism genes in nonapoptotic cell death. ACS Chem Biol. 2015;10(7):1604–9.PubMedPubMedCentralCrossRef Dixon SJ, Winter GE, Musavi LS, Lee ED, Snijder B, Rebsamen M, et al. Human haploid cell genetics reveals roles for lipid metabolism genes in nonapoptotic cell death. ACS Chem Biol. 2015;10(7):1604–9.PubMedPubMedCentralCrossRef
33.
Zurück zum Zitat Zager RA, Johnson AC, Hanson SY. Parenteral iron nephrotoxicity: potential mechanisms and consequences. Kidney Int. 2004;66(1):144–56.PubMedCrossRef Zager RA, Johnson AC, Hanson SY. Parenteral iron nephrotoxicity: potential mechanisms and consequences. Kidney Int. 2004;66(1):144–56.PubMedCrossRef
35.
36.
37.
38.
Zurück zum Zitat Musgrave WB, Yi H, Kline D, Cameron JC, Wignes J, Dey S, et al. Probing the origins of glutathione biosynthesis through biochemical analysis of glutamate–cysteine ligase and glutathione synthetase from a model photosynthetic prokaryote. Biochem J. 2013;450:63–72.PubMedCrossRef Musgrave WB, Yi H, Kline D, Cameron JC, Wignes J, Dey S, et al. Probing the origins of glutathione biosynthesis through biochemical analysis of glutamate–cysteine ligase and glutathione synthetase from a model photosynthetic prokaryote. Biochem J. 2013;450:63–72.PubMedCrossRef
40.
Zurück zum Zitat Savaskan NE, Ufer C, Kuhn H, Borchert A. Molecular biology of glutathione peroxidase 4: from genomic structure to developmental expression and neural function. Biol Chem. 2007;388(10):1007–17.PubMedCrossRef Savaskan NE, Ufer C, Kuhn H, Borchert A. Molecular biology of glutathione peroxidase 4: from genomic structure to developmental expression and neural function. Biol Chem. 2007;388(10):1007–17.PubMedCrossRef
41.
Zurück zum Zitat Savaskan NE, Borchert A, Brauer AU, Kuhn H. Role for glutathione peroxidase-4 in brain development and neuronal apoptosis: specific induction of enzyme expression in reactive astrocytes following brain injury. Free Radic Biol Med. 2007;43(2):191–201.PubMedCrossRef Savaskan NE, Borchert A, Brauer AU, Kuhn H. Role for glutathione peroxidase-4 in brain development and neuronal apoptosis: specific induction of enzyme expression in reactive astrocytes following brain injury. Free Radic Biol Med. 2007;43(2):191–201.PubMedCrossRef
42.
Zurück zum Zitat Alim I, Caulfield JT, Chen Y, Swarup V, Geschwind DH, Ivanova E, et al. Selenium drives a transcriptional adaptive program to block ferroptosis and treat stroke. Cell. 2019;177(5):1262-1279.e25.PubMedCrossRef Alim I, Caulfield JT, Chen Y, Swarup V, Geschwind DH, Ivanova E, et al. Selenium drives a transcriptional adaptive program to block ferroptosis and treat stroke. Cell. 2019;177(5):1262-1279.e25.PubMedCrossRef
43.
Zurück zum Zitat Angeli JPF, Conrad M. Selenium and GPX4, a vital symbiosis. Free Radic Biol Med. 2018;127:153–9.CrossRef Angeli JPF, Conrad M. Selenium and GPX4, a vital symbiosis. Free Radic Biol Med. 2018;127:153–9.CrossRef
45.
Zurück zum Zitat Eaton JK, Furst L, Ruberto RA, Moosmayer D, Hilpmann A, Ryan MJ, et al. Selective covalent targeting of GPX4 using masked nitrile-oxide electrophiles. Nat Chem Biol. 2020;16(5):497–506.PubMedPubMedCentralCrossRef Eaton JK, Furst L, Ruberto RA, Moosmayer D, Hilpmann A, Ryan MJ, et al. Selective covalent targeting of GPX4 using masked nitrile-oxide electrophiles. Nat Chem Biol. 2020;16(5):497–506.PubMedPubMedCentralCrossRef
46.
Zurück zum Zitat Ding Y, Chen X, Liu C, Ge W, Wang Q, Hao X, et al. Identification of a small molecule as inducer of ferroptosis and apoptosis through ubiquitination of GPX4 in triple negative breast cancer cells. J Hematol Oncol. 2021;14(1):19.PubMedPubMedCentralCrossRef Ding Y, Chen X, Liu C, Ge W, Wang Q, Hao X, et al. Identification of a small molecule as inducer of ferroptosis and apoptosis through ubiquitination of GPX4 in triple negative breast cancer cells. J Hematol Oncol. 2021;14(1):19.PubMedPubMedCentralCrossRef
47.
Zurück zum Zitat Song X, Wang X, Liu Z, Yu Z. Role of GPX4-mediated ferroptosis in the sensitivity of triple negative breast cancer cells to gefitinib. Front Oncol. 2020;10: 597434.PubMedPubMedCentralCrossRef Song X, Wang X, Liu Z, Yu Z. Role of GPX4-mediated ferroptosis in the sensitivity of triple negative breast cancer cells to gefitinib. Front Oncol. 2020;10: 597434.PubMedPubMedCentralCrossRef
48.
Zurück zum Zitat Liao M, Qin R, Huang W, Zhu HP, Peng F, Han B, et al. Targeting regulated cell death (RCD) with small-molecule compounds in triple-negative breast cancer: a revisited perspective from molecular mechanisms to targeted therapies. J Hematol Oncol. 2022;15(1):44.PubMedPubMedCentralCrossRef Liao M, Qin R, Huang W, Zhu HP, Peng F, Han B, et al. Targeting regulated cell death (RCD) with small-molecule compounds in triple-negative breast cancer: a revisited perspective from molecular mechanisms to targeted therapies. J Hematol Oncol. 2022;15(1):44.PubMedPubMedCentralCrossRef
49.
Zurück zum Zitat Shin D, Kim EH, Lee J, Roh JL. Nrf2 inhibition reverses resistance to GPX4 inhibitor-induced ferroptosis in head and neck cancer. Free Radic Biol Med. 2018;129:454–62.PubMedCrossRef Shin D, Kim EH, Lee J, Roh JL. Nrf2 inhibition reverses resistance to GPX4 inhibitor-induced ferroptosis in head and neck cancer. Free Radic Biol Med. 2018;129:454–62.PubMedCrossRef
50.
Zurück zum Zitat Jelinek A, Heyder L, Daude M, Plessner M, Krippner S, Grosse R, et al. Mitochondrial rescue prevents glutathione peroxidase-dependent ferroptosis. Free Radic Biol Med. 2018;117:45–57.PubMedCrossRef Jelinek A, Heyder L, Daude M, Plessner M, Krippner S, Grosse R, et al. Mitochondrial rescue prevents glutathione peroxidase-dependent ferroptosis. Free Radic Biol Med. 2018;117:45–57.PubMedCrossRef
51.
Zurück zum Zitat Zhang XH, Li X, Zheng CY, Yang CZ, Zhang R, Wang AL, et al. Ferroptosis, a new form of cell death defined after radiation exposure. Int J Radiat Biol. 2022;98(7):1201–9.PubMedCrossRef Zhang XH, Li X, Zheng CY, Yang CZ, Zhang R, Wang AL, et al. Ferroptosis, a new form of cell death defined after radiation exposure. Int J Radiat Biol. 2022;98(7):1201–9.PubMedCrossRef
52.
Zurück zum Zitat Gagliardi M, Cotella D, Santoro C, Cora D, Barlev NA, Piacentini M, et al. Aldo-keto reductases protect metastatic melanoma from er stress-independent ferroptosis. Cell Death Dis. 2019;10(12):902.PubMedPubMedCentralCrossRef Gagliardi M, Cotella D, Santoro C, Cora D, Barlev NA, Piacentini M, et al. Aldo-keto reductases protect metastatic melanoma from er stress-independent ferroptosis. Cell Death Dis. 2019;10(12):902.PubMedPubMedCentralCrossRef
53.
Zurück zum Zitat Liu X, Zhong S, Qiu K, Chen X, Wu W, Zheng J, et al. Targeting NRF2 uncovered an intrinsic susceptibility of acute myeloid leukemia cells to ferroptosis. Exp Hematol Oncol. 2023;12(1):47.PubMedPubMedCentralCrossRef Liu X, Zhong S, Qiu K, Chen X, Wu W, Zheng J, et al. Targeting NRF2 uncovered an intrinsic susceptibility of acute myeloid leukemia cells to ferroptosis. Exp Hematol Oncol. 2023;12(1):47.PubMedPubMedCentralCrossRef
54.
Zurück zum Zitat Cao JY, Poddar A, Magtanong L, Lumb JH, Mileur TR, Reid MA, et al. A genome-wide haploid genetic screen identifies regulators of glutathione abundance and ferroptosis sensitivity. Cell Rep. 2019;26(6):1544-1556.e8.PubMedPubMedCentralCrossRef Cao JY, Poddar A, Magtanong L, Lumb JH, Mileur TR, Reid MA, et al. A genome-wide haploid genetic screen identifies regulators of glutathione abundance and ferroptosis sensitivity. Cell Rep. 2019;26(6):1544-1556.e8.PubMedPubMedCentralCrossRef
55.
Zurück zum Zitat de Souza I, Monteiro LKS, Guedes CB, Silva MM, Andrade-Tomaz M, Contieri B, et al. High levels of NRF2 sensitize temozolomide-resistant glioblastoma cells to ferroptosis via ABCC1/MRP1 upregulation. Cell Death Dis. 2022;13(7):591.PubMedPubMedCentralCrossRef de Souza I, Monteiro LKS, Guedes CB, Silva MM, Andrade-Tomaz M, Contieri B, et al. High levels of NRF2 sensitize temozolomide-resistant glioblastoma cells to ferroptosis via ABCC1/MRP1 upregulation. Cell Death Dis. 2022;13(7):591.PubMedPubMedCentralCrossRef
56.
Zurück zum Zitat Hanssen KM, Haber M, Fletcher JI. Targeting multidrug resistance-associated protein 1 (MRP1)-expressing cancers: beyond pharmacological inhibition. Drug Resist Updates. 2021;59: 100795.CrossRef Hanssen KM, Haber M, Fletcher JI. Targeting multidrug resistance-associated protein 1 (MRP1)-expressing cancers: beyond pharmacological inhibition. Drug Resist Updates. 2021;59: 100795.CrossRef
58.
Zurück zum Zitat Jiang L, Kon N, Li T, Wang SJ, Su T, Hibshoosh H, et al. Ferroptosis as a p53-mediated activity during tumour suppression. Nature. 2015;520(7545):57–62.PubMedPubMedCentralCrossRef Jiang L, Kon N, Li T, Wang SJ, Su T, Hibshoosh H, et al. Ferroptosis as a p53-mediated activity during tumour suppression. Nature. 2015;520(7545):57–62.PubMedPubMedCentralCrossRef
59.
Zurück zum Zitat Wang SJ, Li DW, Ou Y, Jiang L, Chen Y, Zhao YM, et al. Acetylation is crucial for p53-mediated ferroptosis and tumor suppression. Cell Rep. 2016;17(2):366–73.PubMedPubMedCentralCrossRef Wang SJ, Li DW, Ou Y, Jiang L, Chen Y, Zhao YM, et al. Acetylation is crucial for p53-mediated ferroptosis and tumor suppression. Cell Rep. 2016;17(2):366–73.PubMedPubMedCentralCrossRef
60.
Zurück zum Zitat Liu YQ, Gu W. The complexity of p53-mediated metabolic regulation in tumor suppression. Semin Cancer Biol. 2022;85:4–32.PubMedCrossRef Liu YQ, Gu W. The complexity of p53-mediated metabolic regulation in tumor suppression. Semin Cancer Biol. 2022;85:4–32.PubMedCrossRef
61.
Zurück zum Zitat Chu B, Kon N, Chen D, Li T, Liu T, Jiang L, et al. ALOX12 is required for p53-mediated tumour suppression through a distinct ferroptosis pathway. Nat Cell Biol. 2019;21(5):579–91.PubMedPubMedCentralCrossRef Chu B, Kon N, Chen D, Li T, Liu T, Jiang L, et al. ALOX12 is required for p53-mediated tumour suppression through a distinct ferroptosis pathway. Nat Cell Biol. 2019;21(5):579–91.PubMedPubMedCentralCrossRef
62.
63.
Zurück zum Zitat Ou Y, Wang SJ, Li DW, Chu B, Gu W. Activation of SAT1 engages polyamine metabolism with p53-mediated ferroptotic responses. Proc Natl Acad Sci USA. 2016;113(44):E6806–12.PubMedPubMedCentralCrossRef Ou Y, Wang SJ, Li DW, Chu B, Gu W. Activation of SAT1 engages polyamine metabolism with p53-mediated ferroptotic responses. Proc Natl Acad Sci USA. 2016;113(44):E6806–12.PubMedPubMedCentralCrossRef
64.
Zurück zum Zitat Suzuki S, Tanaka T, Poyurovsky MV, Nagano H, Mayama T, Ohkubo S, et al. Phosphate-activated glutaminase (GLS2), a p53-inducible regulator of glutamine metabolism and reactive oxygen species. Proc Natl Acad Sci USA. 2010;107(16):7461–6.PubMedPubMedCentralCrossRef Suzuki S, Tanaka T, Poyurovsky MV, Nagano H, Mayama T, Ohkubo S, et al. Phosphate-activated glutaminase (GLS2), a p53-inducible regulator of glutamine metabolism and reactive oxygen species. Proc Natl Acad Sci USA. 2010;107(16):7461–6.PubMedPubMedCentralCrossRef
65.
Zurück zum Zitat Doll S, Freitas FP, Shah R, Aldrovandi M, da Silva MC, Ingold I, et al. FSP1 is a glutathione-independent ferroptosis suppressor. Nature. 2019;575(7784):693–8.PubMedCrossRef Doll S, Freitas FP, Shah R, Aldrovandi M, da Silva MC, Ingold I, et al. FSP1 is a glutathione-independent ferroptosis suppressor. Nature. 2019;575(7784):693–8.PubMedCrossRef
66.
Zurück zum Zitat Bersuker K, Hendricks JM, Li Z, Magtanong L, Ford B, Tang PH, et al. The CoQ oxidoreductase FSP1 acts parallel to GPX4 to inhibit ferroptosis. Nature. 2019;575(7784):688–92.PubMedPubMedCentralCrossRef Bersuker K, Hendricks JM, Li Z, Magtanong L, Ford B, Tang PH, et al. The CoQ oxidoreductase FSP1 acts parallel to GPX4 to inhibit ferroptosis. Nature. 2019;575(7784):688–92.PubMedPubMedCentralCrossRef
67.
Zurück zum Zitat Mishima E, Ito J, Wu Z, Nakamura T, Wahida A, Doll S, et al. A non-canonical vitamin K cycle is a potent ferroptosis suppressor. Nature. 2022;608(7924):778–83.PubMedPubMedCentralCrossRef Mishima E, Ito J, Wu Z, Nakamura T, Wahida A, Doll S, et al. A non-canonical vitamin K cycle is a potent ferroptosis suppressor. Nature. 2022;608(7924):778–83.PubMedPubMedCentralCrossRef
68.
Zurück zum Zitat Mao C, Liu X, Zhang Y, Lei G, Yan Y, Lee H, et al. Dhodh-mediated ferroptosis defence is a targetable vulnerability in cancer. Nature. 2021;593(7860):586–90.PubMedPubMedCentralCrossRef Mao C, Liu X, Zhang Y, Lei G, Yan Y, Lee H, et al. Dhodh-mediated ferroptosis defence is a targetable vulnerability in cancer. Nature. 2021;593(7860):586–90.PubMedPubMedCentralCrossRef
69.
Zurück zum Zitat Li H, Yang P, Wang J, Zhang J, Ma Q, Jiang Y, et al. HLF regulates ferroptosis, development and chemoresistance of triple-negative breast cancer by activating tumor cell-macrophage crosstalk. J Hematol Oncol. 2022;15(1):2.PubMedPubMedCentralCrossRef Li H, Yang P, Wang J, Zhang J, Ma Q, Jiang Y, et al. HLF regulates ferroptosis, development and chemoresistance of triple-negative breast cancer by activating tumor cell-macrophage crosstalk. J Hematol Oncol. 2022;15(1):2.PubMedPubMedCentralCrossRef
70.
Zurück zum Zitat Qin R, You FM, Zhao Q, Xie X, Peng C, Zhan G, et al. Naturally derived indole alkaloids targeting regulated cell death (RCD) for cancer therapy: from molecular mechanisms to potential therapeutic targets. J Hematol Oncol. 2022;15(1):133.PubMedPubMedCentralCrossRef Qin R, You FM, Zhao Q, Xie X, Peng C, Zhan G, et al. Naturally derived indole alkaloids targeting regulated cell death (RCD) for cancer therapy: from molecular mechanisms to potential therapeutic targets. J Hematol Oncol. 2022;15(1):133.PubMedPubMedCentralCrossRef
71.
Zurück zum Zitat Mollazadeh H, Atkin SL, Butler AE, Ruscica M, Sirtori CR, Sahebkar A. The effect of statin therapy on endoplasmic reticulum stress. Pharmacol Res. 2018;137:150–8.PubMedCrossRef Mollazadeh H, Atkin SL, Butler AE, Ruscica M, Sirtori CR, Sahebkar A. The effect of statin therapy on endoplasmic reticulum stress. Pharmacol Res. 2018;137:150–8.PubMedCrossRef
72.
Zurück zum Zitat Ron D, Walter P. Signal integration in the endoplasmic reticulum unfolded protein response. Nat Rev Mol Cell Biol. 2007;8(7):519–29.PubMedCrossRef Ron D, Walter P. Signal integration in the endoplasmic reticulum unfolded protein response. Nat Rev Mol Cell Biol. 2007;8(7):519–29.PubMedCrossRef
73.
Zurück zum Zitat Ashrafizadeh M, Mohammadinejad R, Tavakol S, Ahmadi Z, Roomiani S, Katebi M. Autophagy, anoikis, ferroptosis, necroptosis, and endoplasmic reticulum stress: potential applications in melanoma therapy. J Cell Physiol. 2019;234(11):19471–9.PubMedCrossRef Ashrafizadeh M, Mohammadinejad R, Tavakol S, Ahmadi Z, Roomiani S, Katebi M. Autophagy, anoikis, ferroptosis, necroptosis, and endoplasmic reticulum stress: potential applications in melanoma therapy. J Cell Physiol. 2019;234(11):19471–9.PubMedCrossRef
75.
Zurück zum Zitat Ruggiano A, Foresti O, Carvalho P. ER-associated degradation: protein quality control and beyond. J Cell Biol. 2014;204(6):868–78.CrossRef Ruggiano A, Foresti O, Carvalho P. ER-associated degradation: protein quality control and beyond. J Cell Biol. 2014;204(6):868–78.CrossRef
76.
Zurück zum Zitat Mahoney E, Byrd JC, Johnson AJ. Autophagy and ER stress play an essential role in the mechanism of action and drug resistance of the cyclin-dependent kinase inhibitor flavopiridol. Autophagy. 2013;9(3):434–5.PubMedPubMedCentralCrossRef Mahoney E, Byrd JC, Johnson AJ. Autophagy and ER stress play an essential role in the mechanism of action and drug resistance of the cyclin-dependent kinase inhibitor flavopiridol. Autophagy. 2013;9(3):434–5.PubMedPubMedCentralCrossRef
77.
Zurück zum Zitat Zhang G, Liu K, Ling X, Wang Z, Zou P, Wang X, et al. DBP-induced endoplasmic reticulum stress in male germ cells causes autophagy, which has a cytoprotective role against apoptosis in vitro and in vivo. Toxicol Lett. 2016;245:86–98.PubMedCrossRef Zhang G, Liu K, Ling X, Wang Z, Zou P, Wang X, et al. DBP-induced endoplasmic reticulum stress in male germ cells causes autophagy, which has a cytoprotective role against apoptosis in vitro and in vivo. Toxicol Lett. 2016;245:86–98.PubMedCrossRef
78.
Zurück zum Zitat Lee YS, Lee DH, Choudry HA, Bartlett DL, Lee YJ. Ferroptosis-induced endoplasmic reticulum stress: cross-talk between ferroptosis and apoptosis. Mol Cancer Res. 2018;16(7):1073–6.PubMedPubMedCentralCrossRef Lee YS, Lee DH, Choudry HA, Bartlett DL, Lee YJ. Ferroptosis-induced endoplasmic reticulum stress: cross-talk between ferroptosis and apoptosis. Mol Cancer Res. 2018;16(7):1073–6.PubMedPubMedCentralCrossRef
79.
Zurück zum Zitat Dixon SJ, Patel DN, Welsch M, Skouta R, Lee ED, Hayano M, et al. Pharmacological inhibition of cystine-glutamate exchange induces endoplasmic reticulum stress and ferroptosis. Elife. 2014;3: e02523.PubMedPubMedCentralCrossRef Dixon SJ, Patel DN, Welsch M, Skouta R, Lee ED, Hayano M, et al. Pharmacological inhibition of cystine-glutamate exchange induces endoplasmic reticulum stress and ferroptosis. Elife. 2014;3: e02523.PubMedPubMedCentralCrossRef
80.
Zurück zum Zitat Zhu S, Zhang Q, Sun X, Zeh HJ 3rd, Lotze MT, Kang R, et al. HSPA5 regulates ferroptotic cell death in cancer cells. Can Res. 2017;77(8):2064–77.CrossRef Zhu S, Zhang Q, Sun X, Zeh HJ 3rd, Lotze MT, Kang R, et al. HSPA5 regulates ferroptotic cell death in cancer cells. Can Res. 2017;77(8):2064–77.CrossRef
81.
Zurück zum Zitat Chen Y, Mi Y, Zhang X, Ma Q, Song Y, Zhang L, et al. Dihydroartemisinin-induced unfolded protein response feedback attenuates ferroptosis via PERK/ATF4/HSPA5 pathway in glioma cells. J Exp Clin Cancer Res. 2019;38(1):402.PubMedPubMedCentralCrossRef Chen Y, Mi Y, Zhang X, Ma Q, Song Y, Zhang L, et al. Dihydroartemisinin-induced unfolded protein response feedback attenuates ferroptosis via PERK/ATF4/HSPA5 pathway in glioma cells. J Exp Clin Cancer Res. 2019;38(1):402.PubMedPubMedCentralCrossRef
82.
Zurück zum Zitat Chen D, Fan Z, Rauh M, Buchfelder M, Eyupoglu IY, Savaskan N. ATF4 promotes angiogenesis and neuronal cell death and confers ferroptosis in a xCT-dependent manner. Oncogene. 2017;36(40):5593–608.PubMedPubMedCentralCrossRef Chen D, Fan Z, Rauh M, Buchfelder M, Eyupoglu IY, Savaskan N. ATF4 promotes angiogenesis and neuronal cell death and confers ferroptosis in a xCT-dependent manner. Oncogene. 2017;36(40):5593–608.PubMedPubMedCentralCrossRef
83.
Zurück zum Zitat He S, Zhang M, Ye Y, Zhuang J, Ma X, Song Y, et al. ChaC glutathione specific gamma-glutamylcyclotransferase 1 inhibits cell viability and increases the sensitivity of prostate cancer cells to docetaxel by inducing endoplasmic reticulum stress and ferroptosis. Exp Ther Med. 2021;22(3):997.PubMedPubMedCentralCrossRef He S, Zhang M, Ye Y, Zhuang J, Ma X, Song Y, et al. ChaC glutathione specific gamma-glutamylcyclotransferase 1 inhibits cell viability and increases the sensitivity of prostate cancer cells to docetaxel by inducing endoplasmic reticulum stress and ferroptosis. Exp Ther Med. 2021;22(3):997.PubMedPubMedCentralCrossRef
84.
Zurück zum Zitat Hong SH, Lee DH, Lee YS, Jo MJ, Jeong YA, Kwon WT, et al. Molecular crosstalk between ferroptosis and apoptosis: emerging role of er stress-induced p53-independent puma expression. Oncotarget. 2017;8(70):115164–78.PubMedPubMedCentralCrossRef Hong SH, Lee DH, Lee YS, Jo MJ, Jeong YA, Kwon WT, et al. Molecular crosstalk between ferroptosis and apoptosis: emerging role of er stress-induced p53-independent puma expression. Oncotarget. 2017;8(70):115164–78.PubMedPubMedCentralCrossRef
85.
Zurück zum Zitat Xu M, Tao J, Yang Y, Tan S, Liu H, Jiang J, et al. Ferroptosis involves in intestinal epithelial cell death in ulcerative colitis. Cell Death Dis. 2020;11(2):86.PubMedPubMedCentralCrossRef Xu M, Tao J, Yang Y, Tan S, Liu H, Jiang J, et al. Ferroptosis involves in intestinal epithelial cell death in ulcerative colitis. Cell Death Dis. 2020;11(2):86.PubMedPubMedCentralCrossRef
87.
Zurück zum Zitat Zhou Y, Shen Y, Chen C, Sui X, Yang J, Wang L, et al. The crosstalk between autophagy and ferroptosis: what can we learn to target drug resistance in cancer? Cancer Biol Med. 2019;16(4):630–46.PubMedPubMedCentralCrossRef Zhou Y, Shen Y, Chen C, Sui X, Yang J, Wang L, et al. The crosstalk between autophagy and ferroptosis: what can we learn to target drug resistance in cancer? Cancer Biol Med. 2019;16(4):630–46.PubMedPubMedCentralCrossRef
89.
Zurück zum Zitat Yoshida GJ. The interplay between apoptosis and ferroptosis mediated by er stress. Apoptosis. 2020;25(11–12):784–5.PubMedCrossRef Yoshida GJ. The interplay between apoptosis and ferroptosis mediated by er stress. Apoptosis. 2020;25(11–12):784–5.PubMedCrossRef
90.
Zurück zum Zitat Song X, Zhu S, Chen P, Hou W, Wen Q, Liu J, et al. AMPK-mediated BECN1 phosphorylation promotes ferroptosis by directly blocking system Xc− activity. Curr Biol. 2018;28(15):2388-2399.e5.PubMedPubMedCentralCrossRef Song X, Zhu S, Chen P, Hou W, Wen Q, Liu J, et al. AMPK-mediated BECN1 phosphorylation promotes ferroptosis by directly blocking system Xc activity. Curr Biol. 2018;28(15):2388-2399.e5.PubMedPubMedCentralCrossRef
92.
Zurück zum Zitat Li B, Wei S, Yang L, Peng X, Ma Y, Wu B, et al. CISD2 promotes resistance to sorafenib-induced ferroptosis by regulating autophagy in hepatocellular carcinoma. Front Oncol. 2021;11: 657723.PubMedPubMedCentralCrossRef Li B, Wei S, Yang L, Peng X, Ma Y, Wu B, et al. CISD2 promotes resistance to sorafenib-induced ferroptosis by regulating autophagy in hepatocellular carcinoma. Front Oncol. 2021;11: 657723.PubMedPubMedCentralCrossRef
93.
Zurück zum Zitat Hu NJ, Bai LL, Dai EY, Han L, Kang R, Li HJ, et al. Pirin is a nuclear redox-sensitive modulator of autophagy-dependent ferroptosis. Biochem Biophys Res Commun. 2021;536:100–6.PubMedCrossRef Hu NJ, Bai LL, Dai EY, Han L, Kang R, Li HJ, et al. Pirin is a nuclear redox-sensitive modulator of autophagy-dependent ferroptosis. Biochem Biophys Res Commun. 2021;536:100–6.PubMedCrossRef
94.
Zurück zum Zitat Liu Y, Wang Y, Liu J, Kang R, Tang DL. Interplay between MTOR and GPX4 signaling modulates autophagy-dependent ferroptotic cancer cell death. Cancer Gene Ther. 2021;28(1–2):55–63.PubMedCrossRef Liu Y, Wang Y, Liu J, Kang R, Tang DL. Interplay between MTOR and GPX4 signaling modulates autophagy-dependent ferroptotic cancer cell death. Cancer Gene Ther. 2021;28(1–2):55–63.PubMedCrossRef
95.
Zurück zum Zitat Lou S, Hong H, Maihesuti L, Gao H, Zhu Z, Xu L, et al. Inhibitory effect of hydnocarpin D on T-cell acute lymphoblastic leukemia via induction of autophagy-dependent ferroptosis. Exp Biol Med (Maywood). 2021;246(13):1541–53.PubMedCrossRef Lou S, Hong H, Maihesuti L, Gao H, Zhu Z, Xu L, et al. Inhibitory effect of hydnocarpin D on T-cell acute lymphoblastic leukemia via induction of autophagy-dependent ferroptosis. Exp Biol Med (Maywood). 2021;246(13):1541–53.PubMedCrossRef
96.
Zurück zum Zitat Jiang L, Zheng H, Lyu Q, Hayashi S, Sato K, Sekido Y, et al. Lysosomal nitric oxide determines transition from autophagy to ferroptosis after exposure to plasma-activated ringer’s lactate. Redox Biol. 2021;43: 101989.PubMedPubMedCentralCrossRef Jiang L, Zheng H, Lyu Q, Hayashi S, Sato K, Sekido Y, et al. Lysosomal nitric oxide determines transition from autophagy to ferroptosis after exposure to plasma-activated ringer’s lactate. Redox Biol. 2021;43: 101989.PubMedPubMedCentralCrossRef
97.
Zurück zum Zitat Zhong Y, Tian F, Ma H, Wang H, Yang W, Liu Z, et al. FTY720 induces ferroptosis and autophagy via PP2A/AMPK pathway in multiple myeloma cells. Life Sci. 2020;260: 118077.PubMedCrossRef Zhong Y, Tian F, Ma H, Wang H, Yang W, Liu Z, et al. FTY720 induces ferroptosis and autophagy via PP2A/AMPK pathway in multiple myeloma cells. Life Sci. 2020;260: 118077.PubMedCrossRef
98.
Zurück zum Zitat Arosio P, Ingrassia R, Cavadini P. Ferritins: a family of molecules for iron storage, antioxidation and more. Biochem Biophys Acta. 2009;1790(7):589–99.PubMedCrossRef Arosio P, Ingrassia R, Cavadini P. Ferritins: a family of molecules for iron storage, antioxidation and more. Biochem Biophys Acta. 2009;1790(7):589–99.PubMedCrossRef
99.
Zurück zum Zitat Du Y, Bao J, Zhang MJ, Li LL, Xu XL, Chen H, et al. Targeting ferroptosis contributes to ATPR-induced AML differentiation via ROS-autophagy-lysosomal pathway. Gene. 2020;755: 144889.PubMedCrossRef Du Y, Bao J, Zhang MJ, Li LL, Xu XL, Chen H, et al. Targeting ferroptosis contributes to ATPR-induced AML differentiation via ROS-autophagy-lysosomal pathway. Gene. 2020;755: 144889.PubMedCrossRef
100.
Zurück zum Zitat Zhao Y, Lu J, Mao A, Zhang R, Guan S. Autophagy inhibition plays a protective role in ferroptosis induced by alcohol via the p62-Keap1-Nrf2 pathway. J Agric Food Chem. 2021;69(33):9671–83.PubMedCrossRef Zhao Y, Lu J, Mao A, Zhang R, Guan S. Autophagy inhibition plays a protective role in ferroptosis induced by alcohol via the p62-Keap1-Nrf2 pathway. J Agric Food Chem. 2021;69(33):9671–83.PubMedCrossRef
101.
Zurück zum Zitat Sun X, Niu X, Chen R, He W, Chen D, Kang R, et al. Metallothionein-1g facilitates sorafenib resistance through inhibition of ferroptosis. Hepatology. 2016;64(2):488–500.PubMedCrossRef Sun X, Niu X, Chen R, He W, Chen D, Kang R, et al. Metallothionein-1g facilitates sorafenib resistance through inhibition of ferroptosis. Hepatology. 2016;64(2):488–500.PubMedCrossRef
102.
Zurück zum Zitat Zhao CJ, Yu D, He ZQ, Bao LJ, Feng LJ, Chen LT, et al. Endoplasmic reticulum stress-mediated autophagy activation is involved in cadmium-induced ferroptosis of renal tubular epithelial cells. Free Radic Bio Med. 2021;175:236–48.CrossRef Zhao CJ, Yu D, He ZQ, Bao LJ, Feng LJ, Chen LT, et al. Endoplasmic reticulum stress-mediated autophagy activation is involved in cadmium-induced ferroptosis of renal tubular epithelial cells. Free Radic Bio Med. 2021;175:236–48.CrossRef
103.
Zurück zum Zitat Chen Y, Li N, Wang H, Wang N, Peng H, Wang J, et al. Amentoflavone suppresses cell proliferation and induces cell death through triggering autophagy-dependent ferroptosis in human glioma. Life Sci. 2020;247: 117425.PubMedCrossRef Chen Y, Li N, Wang H, Wang N, Peng H, Wang J, et al. Amentoflavone suppresses cell proliferation and induces cell death through triggering autophagy-dependent ferroptosis in human glioma. Life Sci. 2020;247: 117425.PubMedCrossRef
104.
Zurück zum Zitat Zhu HY, Huang ZX, Chen GQ, Sheng F, Zheng YS. Typhaneoside prevents acute myeloid leukemia (AML) through suppressing proliferation and inducing ferroptosis associated with autophagy. Biochem Biophys Res Commun. 2019;516(4):1265–71.PubMedCrossRef Zhu HY, Huang ZX, Chen GQ, Sheng F, Zheng YS. Typhaneoside prevents acute myeloid leukemia (AML) through suppressing proliferation and inducing ferroptosis associated with autophagy. Biochem Biophys Res Commun. 2019;516(4):1265–71.PubMedCrossRef
105.
Zurück zum Zitat Wang ZX, Ma J, Li XY, Wu Y, Shi H, Chen Y, et al. Quercetin induces p53-independent cancer cell death through lysosome activation by the transcription factor EB and reactive oxygen species-dependent ferroptosis. Br J Pharmacol. 2021;178(5):1133–48.PubMedCrossRef Wang ZX, Ma J, Li XY, Wu Y, Shi H, Chen Y, et al. Quercetin induces p53-independent cancer cell death through lysosome activation by the transcription factor EB and reactive oxygen species-dependent ferroptosis. Br J Pharmacol. 2021;178(5):1133–48.PubMedCrossRef
106.
Zurück zum Zitat Tang X, Ding H, Liang M, Chen X, Yan Y, Wan N, et al. Curcumin induces ferroptosis in non-small-cell lung cancer via activating autophagy. Thorac Cancer. 2021;12(8):1219–30.PubMedPubMedCentralCrossRef Tang X, Ding H, Liang M, Chen X, Yan Y, Wan N, et al. Curcumin induces ferroptosis in non-small-cell lung cancer via activating autophagy. Thorac Cancer. 2021;12(8):1219–30.PubMedPubMedCentralCrossRef
107.
Zurück zum Zitat Tsai Y, Xia C, Sun Z. The inhibitory effect of 6-gingerol on ubiquitin-specific peptidase 14 enhances autophagy-dependent ferroptosis and anti-tumor in vivo and in vitro. Front Pharmacol. 2020;11: 598555.PubMedPubMedCentralCrossRef Tsai Y, Xia C, Sun Z. The inhibitory effect of 6-gingerol on ubiquitin-specific peptidase 14 enhances autophagy-dependent ferroptosis and anti-tumor in vivo and in vitro. Front Pharmacol. 2020;11: 598555.PubMedPubMedCentralCrossRef
108.
Zurück zum Zitat Ploetz E, Zimpel A, Cauda V, Bauer D, Lamb DC, Haisch C, et al. Metal-organic framework nanoparticles induce pyroptosis in cells controlled by the extracellular pH. Adv Mater. 2020;32(19): e1907267.PubMedCrossRef Ploetz E, Zimpel A, Cauda V, Bauer D, Lamb DC, Haisch C, et al. Metal-organic framework nanoparticles induce pyroptosis in cells controlled by the extracellular pH. Adv Mater. 2020;32(19): e1907267.PubMedCrossRef
109.
Zurück zum Zitat Wen J, Chen H, Ren Z, Zhang P, Chen J, Jiang S. Ultrasmall iron oxide nanoparticles induced ferroptosis via Beclin1/ATG5-dependent autophagy pathway. Nano convergence. 2021;8(1):10.PubMedPubMedCentralCrossRef Wen J, Chen H, Ren Z, Zhang P, Chen J, Jiang S. Ultrasmall iron oxide nanoparticles induced ferroptosis via Beclin1/ATG5-dependent autophagy pathway. Nano convergence. 2021;8(1):10.PubMedPubMedCentralCrossRef
110.
Zurück zum Zitat Li Y, Wang X, Yan J, Liu Y, Yang R, Pan D, et al. Nanoparticle ferritin-bound erastin and rapamycin: a nanodrug combining autophagy and ferroptosis for anticancer therapy. Biomater Sci. 2019;7(9):3779–87.PubMedCrossRef Li Y, Wang X, Yan J, Liu Y, Yang R, Pan D, et al. Nanoparticle ferritin-bound erastin and rapamycin: a nanodrug combining autophagy and ferroptosis for anticancer therapy. Biomater Sci. 2019;7(9):3779–87.PubMedCrossRef
111.
Zurück zum Zitat An P, Gao Z, Sun K, Gu D, Wu H, You C, et al. Photothermal-enhanced inactivation of glutathione peroxidase for ferroptosis sensitized by an autophagy promotor. ACS Appl Mater Interfaces. 2019;11(46):42988–97.PubMedCrossRef An P, Gao Z, Sun K, Gu D, Wu H, You C, et al. Photothermal-enhanced inactivation of glutathione peroxidase for ferroptosis sensitized by an autophagy promotor. ACS Appl Mater Interfaces. 2019;11(46):42988–97.PubMedCrossRef
112.
Zurück zum Zitat Huang Y, Lin J, Xiong Y, Chen J, Du X, Liu Q, et al. Superparamagnetic iron oxide nanoparticles induce ferroptosis of human ovarian cancer stem cells by weakening cellular autophagy. J Biomed Nanotechnol. 2020;16(11):1612–22.PubMedCrossRef Huang Y, Lin J, Xiong Y, Chen J, Du X, Liu Q, et al. Superparamagnetic iron oxide nanoparticles induce ferroptosis of human ovarian cancer stem cells by weakening cellular autophagy. J Biomed Nanotechnol. 2020;16(11):1612–22.PubMedCrossRef
113.
Zurück zum Zitat Buccarelli M, Marconi M, Pacioni S, De Pascalis I, D’Alessandris QG, Martini M, et al. Inhibition of autophagy increases susceptibility of glioblastoma stem cells to temozolomide by igniting ferroptosis. Cell Death Dis. 2018;9(8):841.PubMedPubMedCentralCrossRef Buccarelli M, Marconi M, Pacioni S, De Pascalis I, D’Alessandris QG, Martini M, et al. Inhibition of autophagy increases susceptibility of glioblastoma stem cells to temozolomide by igniting ferroptosis. Cell Death Dis. 2018;9(8):841.PubMedPubMedCentralCrossRef
114.
Zurück zum Zitat Ma S, Dielschneider RF, Henson ES, Xiao W, Choquette TR, Blankstein AR, et al. Ferroptosis and autophagy induced cell death occur independently after siramesine and lapatinib treatment in breast cancer cells. PLoS ONE. 2017;12(8): e0182921.PubMedPubMedCentralCrossRef Ma S, Dielschneider RF, Henson ES, Xiao W, Choquette TR, Blankstein AR, et al. Ferroptosis and autophagy induced cell death occur independently after siramesine and lapatinib treatment in breast cancer cells. PLoS ONE. 2017;12(8): e0182921.PubMedPubMedCentralCrossRef
115.
Zurück zum Zitat Adham NA, Hegazy MEF, Naqishbandi AM, Efferth T. Induction of apoptosis, autophagy and ferroptosis by thymus vulgaris and Arctium lappa extract in leukemia and multiple myeloma cell lines. Molecules. 2020;25(21):5016.CrossRef Adham NA, Hegazy MEF, Naqishbandi AM, Efferth T. Induction of apoptosis, autophagy and ferroptosis by thymus vulgaris and Arctium lappa extract in leukemia and multiple myeloma cell lines. Molecules. 2020;25(21):5016.CrossRef
116.
Zurück zum Zitat Mbaveng AT, Bitchagno GTM, Kuete V, Tane P, Efferth T. Cytotoxicity of ungeremine towards multi-factorial drug resistant cancer cells and induction of apoptosis, ferroptosis, necroptosis and autophagy. Phytomedicine. 2019;60: 152832.PubMedCrossRef Mbaveng AT, Bitchagno GTM, Kuete V, Tane P, Efferth T. Cytotoxicity of ungeremine towards multi-factorial drug resistant cancer cells and induction of apoptosis, ferroptosis, necroptosis and autophagy. Phytomedicine. 2019;60: 152832.PubMedCrossRef
117.
Zurück zum Zitat Chen XQ, Xu SD, Zhao CX, Liu B. Role of TLR4/NADPH oxidase 4 pathway in promoting cell death through autophagy and ferroptosis during heart failure. Biochem Biophys Res Commun. 2019;516(1):37–43.PubMedCrossRef Chen XQ, Xu SD, Zhao CX, Liu B. Role of TLR4/NADPH oxidase 4 pathway in promoting cell death through autophagy and ferroptosis during heart failure. Biochem Biophys Res Commun. 2019;516(1):37–43.PubMedCrossRef
118.
Zurück zum Zitat Li Q, Weiland A, Chen X, Lan X, Han X, Durham F, et al. Ultrastructural characteristics of neuronal death and white matter injury in mouse brain tissues after intracerebral hemorrhage: coexistence of ferroptosis, autophagy, and necrosis. Front Neurol. 2018;9:581.PubMedPubMedCentralCrossRef Li Q, Weiland A, Chen X, Lan X, Han X, Durham F, et al. Ultrastructural characteristics of neuronal death and white matter injury in mouse brain tissues after intracerebral hemorrhage: coexistence of ferroptosis, autophagy, and necrosis. Front Neurol. 2018;9:581.PubMedPubMedCentralCrossRef
119.
Zurück zum Zitat Liu L, Li L, Li M, Luo Z. Autophagy-dependent ferroptosis as a therapeutic target in cancer. ChemMedChem. 2021;16(19):2942–50.PubMedCrossRef Liu L, Li L, Li M, Luo Z. Autophagy-dependent ferroptosis as a therapeutic target in cancer. ChemMedChem. 2021;16(19):2942–50.PubMedCrossRef
120.
Zurück zum Zitat Hou W, Xie Y, Song X, Sun X, Lotze MT, Zeh HJ 3rd, et al. Autophagy promotes ferroptosis by degradation of ferritin. Autophagy. 2016;12(8):1425–8.PubMedPubMedCentralCrossRef Hou W, Xie Y, Song X, Sun X, Lotze MT, Zeh HJ 3rd, et al. Autophagy promotes ferroptosis by degradation of ferritin. Autophagy. 2016;12(8):1425–8.PubMedPubMedCentralCrossRef
121.
Zurück zum Zitat Mancias JD, Wang X, Gygi SP, Harper JW, Kimmelman AC. Quantitative proteomics identifies NCOA4 as the cargo receptor mediating ferritinophagy. Nature. 2014;509(7498):105–9.PubMedPubMedCentralCrossRef Mancias JD, Wang X, Gygi SP, Harper JW, Kimmelman AC. Quantitative proteomics identifies NCOA4 as the cargo receptor mediating ferritinophagy. Nature. 2014;509(7498):105–9.PubMedPubMedCentralCrossRef
122.
124.
Zurück zum Zitat Zhang Y, Kong Y, Ma Y, Ni S, Wikerholmen T, Xi K, et al. Loss of COPZ1 induces NCOA4 mediated autophagy and ferroptosis in glioblastoma cell lines. Oncogene. 2021;40(8):1425–39.PubMedPubMedCentralCrossRef Zhang Y, Kong Y, Ma Y, Ni S, Wikerholmen T, Xi K, et al. Loss of COPZ1 induces NCOA4 mediated autophagy and ferroptosis in glioblastoma cell lines. Oncogene. 2021;40(8):1425–39.PubMedPubMedCentralCrossRef
125.
Zurück zum Zitat Park E, Chung SW. ROS-mediated autophagy increases intracellular iron levels and ferroptosis by ferritin and transferrin receptor regulation. Cell Death Dis. 2019;10(11):822.PubMedPubMedCentralCrossRef Park E, Chung SW. ROS-mediated autophagy increases intracellular iron levels and ferroptosis by ferritin and transferrin receptor regulation. Cell Death Dis. 2019;10(11):822.PubMedPubMedCentralCrossRef
126.
Zurück zum Zitat Du J, Wang TT, Li YC, Zhou Y, Wang X, Yu XX, et al. DHA inhibits proliferation and induces ferroptosis of leukemia cells through autophagy dependent degradation of ferritin. Free Radic Bio Med. 2019;131:356–69.CrossRef Du J, Wang TT, Li YC, Zhou Y, Wang X, Yu XX, et al. DHA inhibits proliferation and induces ferroptosis of leukemia cells through autophagy dependent degradation of ferritin. Free Radic Bio Med. 2019;131:356–69.CrossRef
127.
128.
Zurück zum Zitat Wei S, Qiu T, Yao X, Wang N, Jiang L, Jia X, et al. Arsenic induces pancreatic dysfunction and ferroptosis via mitochondrial ROS-autophagy-lysosomal pathway. J Hazard Mater. 2020;384: 121390.PubMedCrossRef Wei S, Qiu T, Yao X, Wang N, Jiang L, Jia X, et al. Arsenic induces pancreatic dysfunction and ferroptosis via mitochondrial ROS-autophagy-lysosomal pathway. J Hazard Mater. 2020;384: 121390.PubMedCrossRef
129.
Zurück zum Zitat Gao H, Bai YS, Jia YY, Zhao YA, Kang RI, Tang DL, et al. Ferroptosis is a lysosomal cell death process. Biochem Biophys Res Commun. 2018;503(3):1550–6.PubMedCrossRef Gao H, Bai YS, Jia YY, Zhao YA, Kang RI, Tang DL, et al. Ferroptosis is a lysosomal cell death process. Biochem Biophys Res Commun. 2018;503(3):1550–6.PubMedCrossRef
130.
Zurück zum Zitat Kumar R, Mendonca J, Owoyemi O, Boyapati K, Thomas N, Kanacharoen S, et al. Supraphysiologic testosterone induces ferroptosis and activates immune pathways through nucleophagy in prostate cancer. Can Res. 2021;81(23):5948–62.CrossRef Kumar R, Mendonca J, Owoyemi O, Boyapati K, Thomas N, Kanacharoen S, et al. Supraphysiologic testosterone induces ferroptosis and activates immune pathways through nucleophagy in prostate cancer. Can Res. 2021;81(23):5948–62.CrossRef
131.
Zurück zum Zitat Jian B, Pang J, Xiong H, Zhang W, Zhan T, Su Z, et al. Autophagy-dependent ferroptosis contributes to cisplatin-induced hearing loss. Toxicol Lett. 2021;350:249–60.PubMedCrossRef Jian B, Pang J, Xiong H, Zhang W, Zhan T, Su Z, et al. Autophagy-dependent ferroptosis contributes to cisplatin-induced hearing loss. Toxicol Lett. 2021;350:249–60.PubMedCrossRef
132.
Zurück zum Zitat Schroeder B, Schulze RJ, Weller SG, Sletten AC, Casey CA, McNiven MA. The small GTPase Rab7 as a central regulator of hepatocellular lipophagy. Hepatology. 2015;61(6):1896–907.PubMedCrossRef Schroeder B, Schulze RJ, Weller SG, Sletten AC, Casey CA, McNiven MA. The small GTPase Rab7 as a central regulator of hepatocellular lipophagy. Hepatology. 2015;61(6):1896–907.PubMedCrossRef
133.
Zurück zum Zitat Bai YS, Meng LJ, Han L, Jia YY, Zhao YA, Gao H, et al. Lipid storage and lipophagy regulates ferroptosis. Biochem Biophys Res Commun. 2019;508(4):997–1003.PubMedCrossRef Bai YS, Meng LJ, Han L, Jia YY, Zhao YA, Gao H, et al. Lipid storage and lipophagy regulates ferroptosis. Biochem Biophys Res Commun. 2019;508(4):997–1003.PubMedCrossRef
134.
Zurück zum Zitat Garcia-Bermudez J, Baudrier L, Bayraktar EC, Shen Y, La K, Guarecuco R, et al. Squalene accumulation in cholesterol auxotrophic lymphomas prevents oxidative cell death. Nature. 2019;567(7746):118–22.PubMedPubMedCentralCrossRef Garcia-Bermudez J, Baudrier L, Bayraktar EC, Shen Y, La K, Guarecuco R, et al. Squalene accumulation in cholesterol auxotrophic lymphomas prevents oxidative cell death. Nature. 2019;567(7746):118–22.PubMedPubMedCentralCrossRef
135.
Zurück zum Zitat Wu ZM, Geng Y, Lu XJ, Shi YY, Wu GW, Zhang MM, et al. Chaperone-mediated autophagy is involved in the execution of ferroptosis. Proc Natl Acad Sci USA. 2019;116(8):2996–3005.PubMedPubMedCentralCrossRef Wu ZM, Geng Y, Lu XJ, Shi YY, Wu GW, Zhang MM, et al. Chaperone-mediated autophagy is involved in the execution of ferroptosis. Proc Natl Acad Sci USA. 2019;116(8):2996–3005.PubMedPubMedCentralCrossRef
136.
Zurück zum Zitat Shimada K, Skouta R, Kaplan A, Yang WS, Hayano M, Dixon SJ, et al. Global survey of cell death mechanisms reveals metabolic regulation of ferroptosis. Nat Chem Biol. 2016;12(7):497–503.PubMedPubMedCentralCrossRef Shimada K, Skouta R, Kaplan A, Yang WS, Hayano M, Dixon SJ, et al. Global survey of cell death mechanisms reveals metabolic regulation of ferroptosis. Nat Chem Biol. 2016;12(7):497–503.PubMedPubMedCentralCrossRef
137.
Zurück zum Zitat Yang M, Chen P, Liu J, Zhu S, Kroemer G, Klionsky DJ, et al. Clockophagy is a novel selective autophagy process favoring ferroptosis. Sci Adv. 2019;5(7): eaaw2238.PubMedPubMedCentralCrossRef Yang M, Chen P, Liu J, Zhu S, Kroemer G, Klionsky DJ, et al. Clockophagy is a novel selective autophagy process favoring ferroptosis. Sci Adv. 2019;5(7): eaaw2238.PubMedPubMedCentralCrossRef
138.
Zurück zum Zitat Liu J, Yang M, Kang R, Klionsky DJ, Tang D. Autophagic degradation of the circadian clock regulator promotes ferroptosis. Autophagy. 2019;15(11):2033–5.PubMedPubMedCentralCrossRef Liu J, Yang M, Kang R, Klionsky DJ, Tang D. Autophagic degradation of the circadian clock regulator promotes ferroptosis. Autophagy. 2019;15(11):2033–5.PubMedPubMedCentralCrossRef
139.
Zurück zum Zitat Chipuk JE, Mohammed JN, Gelles JD, Chen YY. Mechanistic connections between mitochondrial biology and regulated cell death. Dev Cell. 2021;56(9):1221–33.PubMedPubMedCentralCrossRef Chipuk JE, Mohammed JN, Gelles JD, Chen YY. Mechanistic connections between mitochondrial biology and regulated cell death. Dev Cell. 2021;56(9):1221–33.PubMedPubMedCentralCrossRef
140.
Zurück zum Zitat Basit F, van Oppen LM, Schockel L, Bossenbroek HM, van Emst-de Vries SE, Hermeling JC, et al. Mitochondrial complex I inhibition triggers a mitophagy-dependent ROS increase leading to necroptosis and ferroptosis in melanoma cells. Cell Death Dis. 2017;8(3): e2716.PubMedPubMedCentralCrossRef Basit F, van Oppen LM, Schockel L, Bossenbroek HM, van Emst-de Vries SE, Hermeling JC, et al. Mitochondrial complex I inhibition triggers a mitophagy-dependent ROS increase leading to necroptosis and ferroptosis in melanoma cells. Cell Death Dis. 2017;8(3): e2716.PubMedPubMedCentralCrossRef
141.
Zurück zum Zitat Kamada T, Tsujii H, Blakely EA, Debus J, De Neve W, Durante M, et al. Carbon ion radiotherapy in Japan: an assessment of 20 years of clinical experience. Lancet Oncol. 2015;16(2):E93–100.PubMedCrossRef Kamada T, Tsujii H, Blakely EA, Debus J, De Neve W, Durante M, et al. Carbon ion radiotherapy in Japan: an assessment of 20 years of clinical experience. Lancet Oncol. 2015;16(2):E93–100.PubMedCrossRef
142.
Zurück zum Zitat Adjemian S, Oltean T, Martens S, Wiernicki B, Goossens V, Vanden Berghe T, et al. Ionizing radiation results in a mixture of cellular outcomes including mitotic catastrophe, senescence, methuosis, and iron-dependent cell death. Cell Death Dis. 2020;11(11):1003.PubMedPubMedCentralCrossRef Adjemian S, Oltean T, Martens S, Wiernicki B, Goossens V, Vanden Berghe T, et al. Ionizing radiation results in a mixture of cellular outcomes including mitotic catastrophe, senescence, methuosis, and iron-dependent cell death. Cell Death Dis. 2020;11(11):1003.PubMedPubMedCentralCrossRef
143.
Zurück zum Zitat Lang XT, Green MD, Wang WM, Yu JL, Choi JE, Jiang L, et al. Radiotherapy and immunotherapy promote tumoral lipid oxidation and ferroptosis via synergistic repression of SLC7A11. Cancer Discov. 2019;9(12):1673–85.PubMedPubMedCentralCrossRef Lang XT, Green MD, Wang WM, Yu JL, Choi JE, Jiang L, et al. Radiotherapy and immunotherapy promote tumoral lipid oxidation and ferroptosis via synergistic repression of SLC7A11. Cancer Discov. 2019;9(12):1673–85.PubMedPubMedCentralCrossRef
144.
Zurück zum Zitat Zhang XH, Xing XS, Liu HW, Feng JD, Tian MX, Chang SQ, et al. Ionizing radiation induces ferroptosis in granulocyte-macrophage hematopoietic progenitor cells of murine bone marrow. Int J Radiat Biol. 2020;96(5):584–95.PubMedCrossRef Zhang XH, Xing XS, Liu HW, Feng JD, Tian MX, Chang SQ, et al. Ionizing radiation induces ferroptosis in granulocyte-macrophage hematopoietic progenitor cells of murine bone marrow. Int J Radiat Biol. 2020;96(5):584–95.PubMedCrossRef
145.
Zurück zum Zitat Lei G, Zhang YL, Koppula P, Liu XG, Zhang J, Lin SH, et al. The role of ferroptosis in ionizing radiation-induced cell death and tumor suppression. Cell Res. 2020;30(2):146–62.PubMedPubMedCentralCrossRef Lei G, Zhang YL, Koppula P, Liu XG, Zhang J, Lin SH, et al. The role of ferroptosis in ionizing radiation-induced cell death and tumor suppression. Cell Res. 2020;30(2):146–62.PubMedPubMedCentralCrossRef
146.
Zurück zum Zitat Mohamad O, Yamada S, Durante M. Clinical indications for carbon ion radiotherapy. Clin Oncol. 2018;30(5):317–29.CrossRef Mohamad O, Yamada S, Durante M. Clinical indications for carbon ion radiotherapy. Clin Oncol. 2018;30(5):317–29.CrossRef
147.
Zurück zum Zitat Mohamad O, Makishima H, Kamada T. Evolution of carbon ion radiotherapy at the national institute of radiological sciences in Japan. Cancers. 2018;10(3):66.PubMedPubMedCentralCrossRef Mohamad O, Makishima H, Kamada T. Evolution of carbon ion radiotherapy at the national institute of radiological sciences in Japan. Cancers. 2018;10(3):66.PubMedPubMedCentralCrossRef
148.
Zurück zum Zitat Malouff TD, Mahajan A, Krishnan S, Beltran C, Seneviratne DS, Trifiletti DM. Carbon ion therapy: a modern review of an emerging technology. Front Oncol. 2020;10:82.PubMedPubMedCentralCrossRef Malouff TD, Mahajan A, Krishnan S, Beltran C, Seneviratne DS, Trifiletti DM. Carbon ion therapy: a modern review of an emerging technology. Front Oncol. 2020;10:82.PubMedPubMedCentralCrossRef
149.
Zurück zum Zitat Mohamad O, Sishc BJ, Saha J, Pompos A, Rahimi A, Story MD, et al. Carbon ion radiotherapy: a review of clinical experiences and preclinical research, with an emphasis on DNA damage/repair. Cancers. 2017;9(6):66.PubMedPubMedCentralCrossRef Mohamad O, Sishc BJ, Saha J, Pompos A, Rahimi A, Story MD, et al. Carbon ion radiotherapy: a review of clinical experiences and preclinical research, with an emphasis on DNA damage/repair. Cancers. 2017;9(6):66.PubMedPubMedCentralCrossRef
150.
Zurück zum Zitat Zheng XG, Liu BT, Liu XX, Li P, Zhang PC, Ye F, et al. Perk regulates the sensitivity of hepatocellular carcinoma cells to high-let carbon ions via either apoptosis or ferroptosis. J Cancer. 2022;13(2):669–80.PubMedPubMedCentralCrossRef Zheng XG, Liu BT, Liu XX, Li P, Zhang PC, Ye F, et al. Perk regulates the sensitivity of hepatocellular carcinoma cells to high-let carbon ions via either apoptosis or ferroptosis. J Cancer. 2022;13(2):669–80.PubMedPubMedCentralCrossRef
151.
Zurück zum Zitat Zhang Z, Lu M, Chen C, Tong X, Li Y, Yang K, et al. Holo-lactoferrin: the link between ferroptosis and radiotherapy in triple-negative breast cancer. Theranostics. 2021;11(7):3167–82.PubMedPubMedCentralCrossRef Zhang Z, Lu M, Chen C, Tong X, Li Y, Yang K, et al. Holo-lactoferrin: the link between ferroptosis and radiotherapy in triple-negative breast cancer. Theranostics. 2021;11(7):3167–82.PubMedPubMedCentralCrossRef
152.
Zurück zum Zitat Pan X, Lin Z, Jiang D, Yu Y, Yang D, Zhou H, et al. Erastin decreases radioresistance of NSCLC cells partially by inducing GPX4-mediated ferroptosis. Oncol Lett. 2019;17(3):3001–8.PubMedPubMedCentral Pan X, Lin Z, Jiang D, Yu Y, Yang D, Zhou H, et al. Erastin decreases radioresistance of NSCLC cells partially by inducing GPX4-mediated ferroptosis. Oncol Lett. 2019;17(3):3001–8.PubMedPubMedCentral
153.
Zurück zum Zitat Yuan Y, Cao W, Zhou HB, Qian HX, Wang HG. CLTRN, regulated by NRF1/RAN/DLD protein complex, enhances radiation sensitivity of hepatocellular carcinoma cells through ferroptosis pathway. Int J Radiat Oncol. 2021;110(3):859–71.CrossRef Yuan Y, Cao W, Zhou HB, Qian HX, Wang HG. CLTRN, regulated by NRF1/RAN/DLD protein complex, enhances radiation sensitivity of hepatocellular carcinoma cells through ferroptosis pathway. Int J Radiat Oncol. 2021;110(3):859–71.CrossRef
155.
Zurück zum Zitat Wang W, Green M, Choi JE, Gijon M, Kennedy PD, Johnson JK, et al. CD8+ T cells regulate tumour ferroptosis during cancer immunotherapy. Nature. 2019;569(7755):270–4.PubMedPubMedCentralCrossRef Wang W, Green M, Choi JE, Gijon M, Kennedy PD, Johnson JK, et al. CD8+ T cells regulate tumour ferroptosis during cancer immunotherapy. Nature. 2019;569(7755):270–4.PubMedPubMedCentralCrossRef
156.
Zurück zum Zitat Kong R, Wang N, Han W, Bao W, Lu J. IFNgamma-mediated repression of system Xc− drives vulnerability to induced ferroptosis in hepatocellular carcinoma cells. J Leukoc Biol. 2021;110(2):301–14.PubMedCrossRef Kong R, Wang N, Han W, Bao W, Lu J. IFNgamma-mediated repression of system Xc drives vulnerability to induced ferroptosis in hepatocellular carcinoma cells. J Leukoc Biol. 2021;110(2):301–14.PubMedCrossRef
157.
Zurück zum Zitat Xu T, Ma Y, Yuan Q, Hu H, Hu X, Qian Z, et al. Enhanced ferroptosis by oxygen-boosted phototherapy based on a 2-in-1 nanoplatform of ferrous hemoglobin for tumor synergistic therapy. ACS Nano. 2020;14(3):3414–25.PubMedCrossRef Xu T, Ma Y, Yuan Q, Hu H, Hu X, Qian Z, et al. Enhanced ferroptosis by oxygen-boosted phototherapy based on a 2-in-1 nanoplatform of ferrous hemoglobin for tumor synergistic therapy. ACS Nano. 2020;14(3):3414–25.PubMedCrossRef
158.
Zurück zum Zitat Friedmann Angeli JP, Krysko DV, Conrad M. Ferroptosis at the crossroads of cancer-acquired drug resistance and immune evasion. Nat Rev Cancer. 2019;19(7):405–14.PubMedCrossRef Friedmann Angeli JP, Krysko DV, Conrad M. Ferroptosis at the crossroads of cancer-acquired drug resistance and immune evasion. Nat Rev Cancer. 2019;19(7):405–14.PubMedCrossRef
159.
Zurück zum Zitat Yatim N, Jusforgues-Saklani H, Orozco S, Schulz O, BarreiradaSilva R, ReiseSousa C, et al. RIPK1 and NF-kappaB signaling in dying cells determines cross-priming of CD8+ T cells. Science. 2015;350(6258):328–34.PubMedPubMedCentralCrossRef Yatim N, Jusforgues-Saklani H, Orozco S, Schulz O, BarreiradaSilva R, ReiseSousa C, et al. RIPK1 and NF-kappaB signaling in dying cells determines cross-priming of CD8+ T cells. Science. 2015;350(6258):328–34.PubMedPubMedCentralCrossRef
160.
Zurück zum Zitat Galluzzi L, Vitale I, Warren S, Adjemian S, Agostinis P, Martinez AB, et al. Consensus guidelines for the definition, detection and interpretation of immunogenic cell death. J Immunother Cancer. 2020;8(1): e000337.PubMedPubMedCentralCrossRef Galluzzi L, Vitale I, Warren S, Adjemian S, Agostinis P, Martinez AB, et al. Consensus guidelines for the definition, detection and interpretation of immunogenic cell death. J Immunother Cancer. 2020;8(1): e000337.PubMedPubMedCentralCrossRef
161.
Zurück zum Zitat Xiong H, Wang C, Wang Z, Lu H, Yao J. Self-assembled nano-activator constructed ferroptosis-immunotherapy through hijacking endogenous iron to intracellular positive feedback loop. J Control Release. 2021;332:539–52.PubMedCrossRef Xiong H, Wang C, Wang Z, Lu H, Yao J. Self-assembled nano-activator constructed ferroptosis-immunotherapy through hijacking endogenous iron to intracellular positive feedback loop. J Control Release. 2021;332:539–52.PubMedCrossRef
163.
Zurück zum Zitat Song J, Liu T, Yin Y, Zhao W, Lin Z, Yin Y, et al. The deubiquitinase OTUD1 enhances iron transport and potentiates host antitumor immunity. EMBO Rep. 2021;22(2): e51162.PubMedPubMedCentralCrossRef Song J, Liu T, Yin Y, Zhao W, Lin Z, Yin Y, et al. The deubiquitinase OTUD1 enhances iron transport and potentiates host antitumor immunity. EMBO Rep. 2021;22(2): e51162.PubMedPubMedCentralCrossRef
164.
Zurück zum Zitat Wen QR, Liu J, Kang R, Zhou BR, Tang D. The release and activity of HMGB1 in ferroptosis. Biochem Biophys Res Commun. 2019;510(2):278–83.PubMedCrossRef Wen QR, Liu J, Kang R, Zhou BR, Tang D. The release and activity of HMGB1 in ferroptosis. Biochem Biophys Res Commun. 2019;510(2):278–83.PubMedCrossRef
165.
Zurück zum Zitat Turubanova VD, Balalaeva IV, Mishchenko TA, Catanzaro E, Alzeibak R, Peskova NN, et al. Immunogenic cell death induced by a new photodynamic therapy based on photosens and photodithazine. J Immunother Cancer. 2019;7(1):350.PubMedPubMedCentralCrossRef Turubanova VD, Balalaeva IV, Mishchenko TA, Catanzaro E, Alzeibak R, Peskova NN, et al. Immunogenic cell death induced by a new photodynamic therapy based on photosens and photodithazine. J Immunother Cancer. 2019;7(1):350.PubMedPubMedCentralCrossRef
166.
Zurück zum Zitat Efimova I, Catanzaro E, Van der Meeren L, Turubanova VD, Hammad H, Mishchenko TA, et al. Vaccination with early ferroptotic cancer cells induces efficient antitumor immunity. J Immunother Cancer. 2020;8(2): e001369.PubMedPubMedCentralCrossRef Efimova I, Catanzaro E, Van der Meeren L, Turubanova VD, Hammad H, Mishchenko TA, et al. Vaccination with early ferroptotic cancer cells induces efficient antitumor immunity. J Immunother Cancer. 2020;8(2): e001369.PubMedPubMedCentralCrossRef
167.
168.
Zurück zum Zitat Luo X, Gong HB, Gao HY, Wu YP, Sun WY, Li ZQ, et al. Oxygenated phosphatidylethanolamine navigates phagocytosis of ferroptotic cells by interacting with TLR2. Cell Death Differ. 2021;28(6):1971–89.PubMedPubMedCentralCrossRef Luo X, Gong HB, Gao HY, Wu YP, Sun WY, Li ZQ, et al. Oxygenated phosphatidylethanolamine navigates phagocytosis of ferroptotic cells by interacting with TLR2. Cell Death Differ. 2021;28(6):1971–89.PubMedPubMedCentralCrossRef
169.
Zurück zum Zitat Li S, Huang Y. Ferroptosis: an iron-dependent cell death form linking metabolism, diseases, immune cell and targeted therapy. Clin Transl Oncol. 2021;24(1):1–12.PubMedPubMedCentralCrossRef Li S, Huang Y. Ferroptosis: an iron-dependent cell death form linking metabolism, diseases, immune cell and targeted therapy. Clin Transl Oncol. 2021;24(1):1–12.PubMedPubMedCentralCrossRef
170.
Zurück zum Zitat Zhao YY, Lian JX, Lan Z, Zou KL, Wang WM, Yu GT. Ferroptosis promotes anti-tumor immune response by inducing immunogenic exposure in HNSCC. Oral Dis. 2021;29(3):933–41.PubMedCrossRef Zhao YY, Lian JX, Lan Z, Zou KL, Wang WM, Yu GT. Ferroptosis promotes anti-tumor immune response by inducing immunogenic exposure in HNSCC. Oral Dis. 2021;29(3):933–41.PubMedCrossRef
171.
Zurück zum Zitat Fan F, Liu P, Bao R, Chen J, Zhou M, Mo Z, et al. A dual PI3K/HDAC inhibitor induces immunogenic ferroptosis to potentiate cancer immune checkpoint therapy. Cancer Res. 2021;81(24):6233–45.PubMedCrossRef Fan F, Liu P, Bao R, Chen J, Zhou M, Mo Z, et al. A dual PI3K/HDAC inhibitor induces immunogenic ferroptosis to potentiate cancer immune checkpoint therapy. Cancer Res. 2021;81(24):6233–45.PubMedCrossRef
172.
Zurück zum Zitat Yan Y, Liang Q, Xu Z, Huang J, Chen X, Cai Y, et al. Downregulated ferroptosis-related gene STEAP3 as a novel diagnostic and prognostic target for hepatocellular carcinoma and its roles in immune regulation. Front Cell Dev Biol. 2021;9: 743046.PubMedPubMedCentralCrossRef Yan Y, Liang Q, Xu Z, Huang J, Chen X, Cai Y, et al. Downregulated ferroptosis-related gene STEAP3 as a novel diagnostic and prognostic target for hepatocellular carcinoma and its roles in immune regulation. Front Cell Dev Biol. 2021;9: 743046.PubMedPubMedCentralCrossRef
173.
Zurück zum Zitat Wiernicki B, Maschalidi S, Pinney J, Adjemian S, Vanden Berghe T, Ravichandran KS, et al. Cancer cells dying from ferroptosis impede dendritic cell-mediated anti-tumor immunity. Nat Commun. 2022;13(1):3676.PubMedPubMedCentralCrossRef Wiernicki B, Maschalidi S, Pinney J, Adjemian S, Vanden Berghe T, Ravichandran KS, et al. Cancer cells dying from ferroptosis impede dendritic cell-mediated anti-tumor immunity. Nat Commun. 2022;13(1):3676.PubMedPubMedCentralCrossRef
174.
Zurück zum Zitat Han L, Bai LL, Qu CJ, Dai EY, Liu J, Kang R, et al. PPARG-mediated ferroptosis in dendritic cells limits antitumor immunity. Biochem Biophys Res Commun. 2021;576:33–9.PubMedCrossRef Han L, Bai LL, Qu CJ, Dai EY, Liu J, Kang R, et al. PPARG-mediated ferroptosis in dendritic cells limits antitumor immunity. Biochem Biophys Res Commun. 2021;576:33–9.PubMedCrossRef
176.
Zurück zum Zitat Rothe T, Gruber F, Uderhardt S, Ipseiz N, Rossner S, Oskolkova O, et al. 12/15-lipoxygenase-mediated enzymatic lipid oxidation regulates DC maturation and function. J Clin Investig. 2015;125(5):1944–54.PubMedPubMedCentralCrossRef Rothe T, Gruber F, Uderhardt S, Ipseiz N, Rossner S, Oskolkova O, et al. 12/15-lipoxygenase-mediated enzymatic lipid oxidation regulates DC maturation and function. J Clin Investig. 2015;125(5):1944–54.PubMedPubMedCentralCrossRef
177.
Zurück zum Zitat Cubillos-Ruiz JR, Silberman PC, Rutkowski MR, Chopra S, Perales-Puchalt A, Song M, et al. ER stress sensor XBP1 controls anti-tumor immunity by disrupting dendritic cell homeostasis. Cell. 2015;161(7):1527–38.PubMedPubMedCentralCrossRef Cubillos-Ruiz JR, Silberman PC, Rutkowski MR, Chopra S, Perales-Puchalt A, Song M, et al. ER stress sensor XBP1 controls anti-tumor immunity by disrupting dendritic cell homeostasis. Cell. 2015;161(7):1527–38.PubMedPubMedCentralCrossRef
178.
Zurück zum Zitat Xu H, Ye D, Ren M, Zhang H, Bi F. Ferroptosis in the tumor microenvironment: perspectives for immunotherapy. Trends Mol Med. 2021;27(9):856–67.PubMedCrossRef Xu H, Ye D, Ren M, Zhang H, Bi F. Ferroptosis in the tumor microenvironment: perspectives for immunotherapy. Trends Mol Med. 2021;27(9):856–67.PubMedCrossRef
179.
Zurück zum Zitat Dodson M, Castro-Portuguez R, Zhang DD. NRF2 plays a critical role in mitigating lipid peroxidation and ferroptosis. Redox Biol. 2019;23: 101107.PubMedPubMedCentralCrossRef Dodson M, Castro-Portuguez R, Zhang DD. NRF2 plays a critical role in mitigating lipid peroxidation and ferroptosis. Redox Biol. 2019;23: 101107.PubMedPubMedCentralCrossRef
180.
Zurück zum Zitat Poznanski SM, Singh K, Ritchie TM, Aguiar JA, Fan IY, Portillo AL, et al. Metabolic flexibility determines human NK cell functional fate in the tumor microenvironment. Cell Metab. 2021;33(6):1205-1220.e5.PubMedCrossRef Poznanski SM, Singh K, Ritchie TM, Aguiar JA, Fan IY, Portillo AL, et al. Metabolic flexibility determines human NK cell functional fate in the tumor microenvironment. Cell Metab. 2021;33(6):1205-1220.e5.PubMedCrossRef
181.
Zurück zum Zitat Dai EY, Han L, Liu J, Xie YC, Zeh HJ, Kang R, et al. Ferroptotic damage promotes pancreatic tumorigenesis through a TMEM173/STING-dependent DNA sensor pathway. Nat Commun. 2020;11(1):6339.PubMedPubMedCentralCrossRef Dai EY, Han L, Liu J, Xie YC, Zeh HJ, Kang R, et al. Ferroptotic damage promotes pancreatic tumorigenesis through a TMEM173/STING-dependent DNA sensor pathway. Nat Commun. 2020;11(1):6339.PubMedPubMedCentralCrossRef
182.
Zurück zum Zitat Dai E, Han L, Liu J, Xie Y, Kroemer G, Klionsky DJ, et al. Autophagy-dependent ferroptosis drives tumor-associated macrophage polarization via release and uptake of oncogenic KRAS protein. Autophagy. 2020;16(11):2069–83.PubMedPubMedCentralCrossRef Dai E, Han L, Liu J, Xie Y, Kroemer G, Klionsky DJ, et al. Autophagy-dependent ferroptosis drives tumor-associated macrophage polarization via release and uptake of oncogenic KRAS protein. Autophagy. 2020;16(11):2069–83.PubMedPubMedCentralCrossRef
184.
Zurück zum Zitat Matsushita M, Freigang S, Schneider C, Conrad M, Bornkamm GW, Kopf M. T cell lipid peroxidation induces ferroptosis and prevents immunity to infection. J Exp Med. 2015;212(4):555–68.PubMedPubMedCentralCrossRef Matsushita M, Freigang S, Schneider C, Conrad M, Bornkamm GW, Kopf M. T cell lipid peroxidation induces ferroptosis and prevents immunity to infection. J Exp Med. 2015;212(4):555–68.PubMedPubMedCentralCrossRef
185.
Zurück zum Zitat Drijvers JM, Gillis JE, Muijlwijk T, Nguyen TH, Gaudiano EF, Harris IS, et al. Pharmacologic screening identifies metabolic vulnerabilities of CD8+ T cells. Cancer Immunol Res. 2021;9(2):184–99.PubMedCrossRef Drijvers JM, Gillis JE, Muijlwijk T, Nguyen TH, Gaudiano EF, Harris IS, et al. Pharmacologic screening identifies metabolic vulnerabilities of CD8+ T cells. Cancer Immunol Res. 2021;9(2):184–99.PubMedCrossRef
186.
Zurück zum Zitat Shi L, Liu Y, Li M, Luo Z. Emerging roles of ferroptosis in the tumor immune landscape: from danger signals to anti-tumor immunity. FEBS J. 2022;289(13):3655–65.PubMedCrossRef Shi L, Liu Y, Li M, Luo Z. Emerging roles of ferroptosis in the tumor immune landscape: from danger signals to anti-tumor immunity. FEBS J. 2022;289(13):3655–65.PubMedCrossRef
187.
Zurück zum Zitat Ma X, Xiao L, Liu L, Ye L, Su P, Bi E, et al. CD36-mediated ferroptosis dampens intratumoral CD8+ T cell effector function and impairs their antitumor ability. Cell Metab. 2021;33(5):1001-1012.e5.PubMedPubMedCentralCrossRef Ma X, Xiao L, Liu L, Ye L, Su P, Bi E, et al. CD36-mediated ferroptosis dampens intratumoral CD8+ T cell effector function and impairs their antitumor ability. Cell Metab. 2021;33(5):1001-1012.e5.PubMedPubMedCentralCrossRef
188.
Zurück zum Zitat Xu C, Sun S, Johnson T, Qi R, Zhang S, Zhang J, et al. The glutathione peroxidase Gpx4 prevents lipid peroxidation and ferroptosis to sustain Treg cell activation and suppression of antitumor immunity. Cell Rep. 2021;35(11): 109235.PubMedCrossRef Xu C, Sun S, Johnson T, Qi R, Zhang S, Zhang J, et al. The glutathione peroxidase Gpx4 prevents lipid peroxidation and ferroptosis to sustain Treg cell activation and suppression of antitumor immunity. Cell Rep. 2021;35(11): 109235.PubMedCrossRef
189.
Zurück zum Zitat Jiang Q, Wang K, Zhang X, Ouyang B, Liu H, Pang Z, et al. Platelet membrane-camouflaged magnetic nanoparticles for ferroptosis-enhanced cancer immunotherapy. Small. 2020;16(22): e2001704.PubMedCrossRef Jiang Q, Wang K, Zhang X, Ouyang B, Liu H, Pang Z, et al. Platelet membrane-camouflaged magnetic nanoparticles for ferroptosis-enhanced cancer immunotherapy. Small. 2020;16(22): e2001704.PubMedCrossRef
190.
Zurück zum Zitat Ruiz-de-Angulo A, Bilbao-Asensio M, Cronin J, Evans SJ, Clift MJD, Llop J, et al. Chemically programmed vaccines: iron catalysis in nanoparticles enhances combination immunotherapy and immunotherapy-promoted tumor ferroptosis. iScience. 2020;23(9): 101499.PubMedPubMedCentralCrossRef Ruiz-de-Angulo A, Bilbao-Asensio M, Cronin J, Evans SJ, Clift MJD, Llop J, et al. Chemically programmed vaccines: iron catalysis in nanoparticles enhances combination immunotherapy and immunotherapy-promoted tumor ferroptosis. iScience. 2020;23(9): 101499.PubMedPubMedCentralCrossRef
191.
Zurück zum Zitat Yu B, Choi B, Li W, Kim DH. Magnetic field boosted ferroptosis-like cell death and responsive MRI using hybrid vesicles for cancer immunotherapy. Nat Commun. 2020;11(1):3637.PubMedPubMedCentralCrossRef Yu B, Choi B, Li W, Kim DH. Magnetic field boosted ferroptosis-like cell death and responsive MRI using hybrid vesicles for cancer immunotherapy. Nat Commun. 2020;11(1):3637.PubMedPubMedCentralCrossRef
192.
Zurück zum Zitat Liang H, Wu XY, Zhao GZ, Feng K, Ni KY, Sun XL. Renal clearable ultrasmall single-crystal Fe nanoparticles for highly selective and effective ferroptosis therapy and immunotherapy. J Am Chem Soc. 2021;143(38):15812–23.PubMedCrossRef Liang H, Wu XY, Zhao GZ, Feng K, Ni KY, Sun XL. Renal clearable ultrasmall single-crystal Fe nanoparticles for highly selective and effective ferroptosis therapy and immunotherapy. J Am Chem Soc. 2021;143(38):15812–23.PubMedCrossRef
193.
Zurück zum Zitat Liu P, Shi X, Peng Y, Hu J, Ding J, Zhou W. Anti-PD-L1 DNAzyme loaded photothermal Mn2+/Fe3+ hybrid metal-phenolic networks for cyclically amplified tumor ferroptosis-immunotherapy. Adv Healthc Mater. 2021;11: e2102315.PubMedCrossRef Liu P, Shi X, Peng Y, Hu J, Ding J, Zhou W. Anti-PD-L1 DNAzyme loaded photothermal Mn2+/Fe3+ hybrid metal-phenolic networks for cyclically amplified tumor ferroptosis-immunotherapy. Adv Healthc Mater. 2021;11: e2102315.PubMedCrossRef
194.
Zurück zum Zitat Zhang C, Liu HF, Gong M, Yang M, Yang ZQ, Xie YL, et al. Biomimetic gold nanocages for overcoming chemoresistance of osteosarcoma by ferroptosis and immunogenic cell death. Mater Design. 2021;210: 110087.CrossRef Zhang C, Liu HF, Gong M, Yang M, Yang ZQ, Xie YL, et al. Biomimetic gold nanocages for overcoming chemoresistance of osteosarcoma by ferroptosis and immunogenic cell death. Mater Design. 2021;210: 110087.CrossRef
195.
Zurück zum Zitat Zuo T, Fang T, Zhang J, Yang J, Xu R, Wang Z, et al. pH-sensitive molecular-switch-containing polymer nanoparticle for breast cancer therapy with ferritinophagy-cascade ferroptosis and tumor immune activation. Adv Healthc Mater. 2021;10(21): e2100683.PubMedCrossRef Zuo T, Fang T, Zhang J, Yang J, Xu R, Wang Z, et al. pH-sensitive molecular-switch-containing polymer nanoparticle for breast cancer therapy with ferritinophagy-cascade ferroptosis and tumor immune activation. Adv Healthc Mater. 2021;10(21): e2100683.PubMedCrossRef
196.
Zurück zum Zitat Yang J, Ma S, Xu R, Wei Y, Zhang J, Zuo T, et al. Smart biomimetic metal organic frameworks based on ROS-ferroptosis-glycolysis regulation for enhanced tumor chemo-immunotherapy. J Control Release. 2021;334:21–33.PubMedCrossRef Yang J, Ma S, Xu R, Wei Y, Zhang J, Zuo T, et al. Smart biomimetic metal organic frameworks based on ROS-ferroptosis-glycolysis regulation for enhanced tumor chemo-immunotherapy. J Control Release. 2021;334:21–33.PubMedCrossRef
197.
Zurück zum Zitat Xue CC, Li MH, Zhao Y, Zhou J, Hu Y, Cai KY, et al. Tumor microenvironment-activatable Fe-doxorubicin preloaded amorphous CaCO3 nanoformulation triggers ferroptosis in target tumor cells. Sci Adv. 2020;6(18): eaax1346.PubMedPubMedCentralCrossRef Xue CC, Li MH, Zhao Y, Zhou J, Hu Y, Cai KY, et al. Tumor microenvironment-activatable Fe-doxorubicin preloaded amorphous CaCO3 nanoformulation triggers ferroptosis in target tumor cells. Sci Adv. 2020;6(18): eaax1346.PubMedPubMedCentralCrossRef
198.
Zurück zum Zitat Xue CC, Li MH, Liu CH, Li YN, Fei Y, Hu Y, et al. NIR-actuated remote activation of ferroptosis in target tumor cells through a photothermally responsive iron-chelated biopolymer nanoplatform. Angew Chem Int Ed. 2021;60(16):8938–47.CrossRef Xue CC, Li MH, Liu CH, Li YN, Fei Y, Hu Y, et al. NIR-actuated remote activation of ferroptosis in target tumor cells through a photothermally responsive iron-chelated biopolymer nanoplatform. Angew Chem Int Ed. 2021;60(16):8938–47.CrossRef
199.
Zurück zum Zitat Zhang F, Li F, Lu GH, Nie WD, Zhang LJ, Lv YL, et al. Engineering magnetosomes for ferroptosis/immunomodulation synergism in cancer. ACS Nano. 2019;13(5):5662–73.PubMedCrossRef Zhang F, Li F, Lu GH, Nie WD, Zhang LJ, Lv YL, et al. Engineering magnetosomes for ferroptosis/immunomodulation synergism in cancer. ACS Nano. 2019;13(5):5662–73.PubMedCrossRef
200.
Zurück zum Zitat Wang G, Xie L, Li B, Sang W, Yan J, Li J, et al. A nanounit strategy reverses immune suppression of exosomal PD-L1 and is associated with enhanced ferroptosis. Nat Commun. 2021;12(1):5733.PubMedPubMedCentralCrossRef Wang G, Xie L, Li B, Sang W, Yan J, Li J, et al. A nanounit strategy reverses immune suppression of exosomal PD-L1 and is associated with enhanced ferroptosis. Nat Commun. 2021;12(1):5733.PubMedPubMedCentralCrossRef
201.
Zurück zum Zitat Ma S, Henson ES, Chen Y, Gibson SB. Ferroptosis is induced following siramesine and lapatinib treatment of breast cancer cells. Cell Death Dis. 2016;7(7): e2307.PubMedPubMedCentralCrossRef Ma S, Henson ES, Chen Y, Gibson SB. Ferroptosis is induced following siramesine and lapatinib treatment of breast cancer cells. Cell Death Dis. 2016;7(7): e2307.PubMedPubMedCentralCrossRef
202.
Zurück zum Zitat Gout PW, Buckley AR, Simms CR, Bruchovsky N. Sulfasalazine, a potent suppressor of lymphoma growth by inhibition of the Xc− cystine transporter: a new action for an old drug. Leukemia. 2001;15(10):1633–40.PubMedCrossRef Gout PW, Buckley AR, Simms CR, Bruchovsky N. Sulfasalazine, a potent suppressor of lymphoma growth by inhibition of the Xc cystine transporter: a new action for an old drug. Leukemia. 2001;15(10):1633–40.PubMedCrossRef
203.
Zurück zum Zitat Guo JP, Xu BF, Han Q, Zhou HX, Xia Y, Gong CW, et al. Ferroptosis: a novel anti-tumor action for cisplatin. Cancer Res Treat. 2018;50(2):445–60.PubMedCrossRef Guo JP, Xu BF, Han Q, Zhou HX, Xia Y, Gong CW, et al. Ferroptosis: a novel anti-tumor action for cisplatin. Cancer Res Treat. 2018;50(2):445–60.PubMedCrossRef
204.
Zurück zum Zitat Zheng J, Sato M, Mishima E, Sato H, Proneth B, Conrad M. Sorafenib fails to trigger ferroptosis across a wide range of cancer cell lines. Cell Death Dis. 2021;12(7):698.PubMedPubMedCentralCrossRef Zheng J, Sato M, Mishima E, Sato H, Proneth B, Conrad M. Sorafenib fails to trigger ferroptosis across a wide range of cancer cell lines. Cell Death Dis. 2021;12(7):698.PubMedPubMedCentralCrossRef
Metadaten
Titel
Ferroptosis: a novel regulated cell death participating in cellular stress response, radiotherapy, and immunotherapy
verfasst von
Xiaogang Zheng
Xiaodong Jin
Fei Ye
Xiongxiong Liu
Boyi Yu
Zheng Li
Ting Zhao
Weiqiang Chen
Xinguo Liu
Cuixia Di
Qiang Li
Publikationsdatum
01.12.2023
Verlag
BioMed Central
Erschienen in
Experimental Hematology & Oncology / Ausgabe 1/2023
Elektronische ISSN: 2162-3619
DOI
https://doi.org/10.1186/s40164-023-00427-w

Weitere Artikel der Ausgabe 1/2023

Experimental Hematology & Oncology 1/2023 Zur Ausgabe

Erhöhtes Risiko fürs Herz unter Checkpointhemmer-Therapie

28.05.2024 Nebenwirkungen der Krebstherapie Nachrichten

Kardiotoxische Nebenwirkungen einer Therapie mit Immuncheckpointhemmern mögen selten sein – wenn sie aber auftreten, wird es für Patienten oft lebensgefährlich. Voruntersuchung und Monitoring sind daher obligat.

Costims – das nächste heiße Ding in der Krebstherapie?

28.05.2024 Onkologische Immuntherapie Nachrichten

„Kalte“ Tumoren werden heiß – CD28-kostimulatorische Antikörper sollen dies ermöglichen. Am besten könnten diese in Kombination mit BiTEs und Checkpointhemmern wirken. Erste klinische Studien laufen bereits.

Perioperative Checkpointhemmer-Therapie verbessert NSCLC-Prognose

28.05.2024 NSCLC Nachrichten

Eine perioperative Therapie mit Nivolumab reduziert das Risiko für Rezidive und Todesfälle bei operablem NSCLC im Vergleich zu einer alleinigen neoadjuvanten Chemotherapie um über 40%. Darauf deuten die Resultate der Phase-3-Studie CheckMate 77T.

Positiver FIT: Die Ursache liegt nicht immer im Dickdarm

27.05.2024 Blut im Stuhl Nachrichten

Immunchemischer Stuhltest positiv, Koloskopie negativ – in solchen Fällen kann die Blutungsquelle auch weiter proximal sitzen. Ein Forschungsteam hat nachgesehen, wie häufig und in welchen Lokalisationen das der Fall ist.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.