Skip to main content
Erschienen in: Journal of Hematology & Oncology 1/2019

Open Access 01.12.2019 | Review

Ferroptosis, a new form of cell death: opportunities and challenges in cancer

verfasst von: Yanhua Mou, Jun Wang, Jinchun Wu, Dan He, Chunfang Zhang, Chaojun Duan, Bin Li

Erschienen in: Journal of Hematology & Oncology | Ausgabe 1/2019

Abstract

Ferroptosis is a novel type of cell death with distinct properties and recognizing functions involved in physical conditions or various diseases including cancers. The fast-growing studies of ferroptosis in cancer have boosted a perspective for its usage in cancer therapeutics. Here, we review the current findings of ferroptosis regulation and especially focus on the function of ncRNAs in mediating the process of cell ferroptotic death and on how ferroptosis was in relation to other regulated cell deaths. Aberrant ferroptosis in diverse cancer types and tissues were summarized, and we elaborated recent data about the novel actors of some “conventional” drugs or natural compounds as ferroptosis inducers in cancer. Finally, we deliberate future orientation for ferroptosis in cancer cells and current unsettled issues, which may forward the speed of clinical use of ferroptosis induction in cancer treatment.
Hinweise
Yanhua Mou and Jun Wang contributed equally to this work.
Abkürzungen
ACSL4
Acyl-CoA synthetase long-chain family member 4
AML
Acute myeloid leukemia
AMPK
Adenosine 5-monophosphate-activated protein kinase
ART
Artesunate
As-SLC7A11
Antisense lncRNAs
ATF4
Activating transcription factor 4
Atg5
Autophagy-related 5
BAP1
Breast cancer susceptibility gene 1-associated protein 1
BECN1
Beclin1
BID
BH3 interacting domain death agonist
CARS
Cysteinyl-tRNA synthetase
CD44v
CD44 variant
CDO1
Cysteine dioxygenase type 1
CHOP
C/EBP-homologous protein
CISD1
CDGSH iron sulfur domain 1
c-Myb
Cellular homolog
C-Myc
Cellular myelocytomatosis oncogene
CN-A
Cotylenin A
CoQ10
Coenzyme Q10
CTH
Cystathionine γ-lyase
Cul3
Cullin-3
Cys
Cysteine
Cys2
Cystine
DAMPs
Damage-associated molecular patterns
DDP4
Dipeptidyl-peptidase-4
DHA
Docosahexaenoic acid
DLBCL
Diffuse large B-cell lymphoma
DMT1
Divalent metal transporter 1
EGLN1
Egl nine homolog 1
ELAVL1
Embryonic lethal, abnormal vision, Drosophila-like 1
EMT
Epithelial-mesenchymal transition
ERK
Extracellular signal-regulated kinase
FADS2
Fatty acid desaturase 2
FIN
Ferroptosis inducing agents
Flt3
Fms-like tyrosine kinase 3
FPN
Ferroportin
FTH1
Ferritin heavy chain 1
FTL
Ferritin light chain
G3BP1
Ras GTPase-activating protein-binding protein 1
GBM
Glioblastoma multiforme
Gln
Glutamine
GLS2
Glutaminase 2
Glu
Glutamate
GLUT1
Glucose transporter 1
GOT1
Glutamic-oxaloacetic transaminase 1
GPX4
Glutathione peroxidase 4
GSC
Glioblastoma stem cells
GSH
Glutathione
HIF
Hypoxia-inducible transcriptional factor
HNCs
Head and neck cancers
HO-1
Heme oxygenase-1
HSF1
Heat shock factor-1
HSPB1
Heat shock protein beta-1
IPP
Isopentenyl pyrophosphate
IRE
Iron-responsive element
IREB2
Iron response element binding protein 2
Keap1
Keleh-like ECH-associated protein 1
LDL
Low-density lipoprotein
LncRNA
Long non-coding RNA
LPCAT3
Lysophosphatidylcholine acyltransferase 3
LSH
Lymphoid-specific helicase
MEK
Ras-mitogen-activated protein kinase kinase
MON-P53
Metal organic network-P53
MT-1G
Metallothionein-1 g
MUC1-C
Mucin 1 C-terminal
MVA
Mevalonate
NCOA4
Nuclear receptor coactivator 4
NFS1
Cysteine desulfurase
NOX
Nicotinamide adenine dinucleotide phosphate-oxidase
NQO1
Quinone oxidoreductase-1
Nrf2
Nuclear factor erythroid 2-related factor 2
PHKG2
Phosphorylase kinase gamma 2
PIK3C3
Phosphatidylinositol 3-kinase catalytic subunit type 3
PL-PUFAs
Phospholipid-polyunsaturated fatty acids
PtdIns3K
Phosphatidylinositol 3-kinase
PUFAs
Polyunsaturated fatty acids
PUMA
P53 upregulated modulator of apoptosis
RB
Retinoblastoma
ROS
Reactive oxygen species
RRM
RNA recognition motif
RSL3
Ras-selective lethal small molecule 3
SAT1
Spermidine/spermine N1-acetyltransferase 1
SCD1
Stearoyl-CoA desaturase 1
Se
Selenium
SLC1A5
Solute carrier family 1 member 5
SLC3A2
Solute carrier family 3 member 2
SLC7A11
Solute carrier family 7 member 11
SOD1
Superoxide dismutase 1
SQS
Squalene synthase
STEAP3
Six-transmembrane epithelial antigen of the prostate 3
TF
Transferrin
TFR1
Transferrin receptor 1
TICs
Tumor-initiating cells
TP53
Tumor suppressor P53
TRAIL
Tumor necrosis factor-related apoptosis-inducing ligand
UPR
Unfolded protein response
VDAC2/3
Voltage-dependent anion channel 2/3
xCT
System xc-
α-KG
α-ketoglutarate

Background

Ferroptosis was first proposed by Dixon as a novel cell death in 2012 [1]. Unlike autophagy and apoptosis, ferroptosis is an iron-dependent and reactive oxygen species (ROS)-reliant cell death with characteristics mainly of cytological changes, including decreased or vanished mitochondria cristae, a ruptured outer mitochondrial membrane, and a condensed mitochondrial membrane [26]. These cell abnormalities resulted from the loss of selective permeability of plasma membrane due to intense membrane lipid peroxidation and the occurrence of oxidative stress (Table 1) [7].
Table 1
The main features of ferroptosis, apoptosis, autophagy, necroptosis, and pyroptosis
Cell death
Ferroptosis
Apoptosis
Autophagy
Necroptosis
Pyroptosis
Biochemical features
Inhibition of xCT and reduced GSH, inhibition of GPX4. Iron accumulation and lipid peroxidation
Activation of caspases oligonucleosomal DNA fragmentation
Increased lysosomal activity
Drop in ATP levels; activation of RIP1, RIP3, and MLKL
Dependent on caspase-1 and proinflammatory cytokine releases
Morphological features
Small mitochondria with condensed mitochondrial membrane densities, reduction or vanishing of mitochondria crista, as well as outer mitochondrial membrane rupture
Plasma membrane blebbing; cellular and nuclear volume reduction; nuclear fragmentation
Formation of double-membraned autolysosomes
Plasma membrane rupture; organelle swelling; moderate chromatin condensation
Karyopyknosis, cell edema and membrane rupture
Key genes
GPX4, Nrf2, LSH, TFR1, xCT
Caspase, P53, Fas, Bcl-2, Bax
ATG5, ATG7, DRAM3, TFEB
LEF1, RIP1, RIP3
Caspase-1, IL-1β, IL-18
Regulatory pathways
xCT and Gpx4, MVA, HSF1-HSPB1, p62-Keap1-Nrf2 pathway, LSH signal pathway
Death receptor, Mitochondrial, Endoplasmic reticulum pathway
Csapase, P53, Bcl-2 mediated signaling pathway
PI3K-AKT-mTOR,MAPK-ERK1/2-mTOR signal pathway
TNFα,TNFR1,TLR3, TRAIL, FasL, ROS, PKC-MAPK-AP-1-mediated signaling pathway
Caspase-1, NLRP3-mediated signaling pathway.
Released DAMP
HMGB1
Ecto-CRT, HMGB1, and ATP
HMGB-1
DNA and IL-6
HMGB1, ATP, IL-1β, and IL-18
Immune features
Pro-inflammatory
Mostly anti-inflammatory
Mostly anti-inflammatory
Mostly pro-inflammatory
pro-inflammatory
Inducers
Erastin,DPI2, BSO, SAS, lanperisone, SRS, RSL3, DPI7, DPI10, FIN56, sorafenib, artemisinin
FASL, DCC, UNC5B
Rapamycin, lithium, sodium, valproate, carbamazepine, C2-ceramide, rapamycin
TNFa, zVAD-fmk, PAMPS
ZnO—NPs, Ivermectin
Inhibitors
Desferoxamine, vitamin E, U0126, ferrostatin-1, SRS, CA-1, cycloheximide, aminooxyacetic acid Liproxstatin-1 HCl
XIAP, c-IAP1, c-IAP2, ILP-2, ML-IAP/livin, NAIP, Z-VADFMK
3-ME, LY294002, wortmannin, PIK-III, compound 31, SAR 405, Vps34-In1,MRT68921, Spautin-1, Bafilomycin A1, hydrochloroquin
Nec-1, NSA, Kongensin- A
Necrosulfonami-de
Researches indicated that ferroptosis could be triggered by diverse physiological conditions and pathological stresses in humans and animals [8]. Ferroptosis is gradually accepted as an adaptive feature to eliminate the malignant cells. It plays a pivotal role in the depression of tumorigenesis by removing the cells that are deficient in key nutrients in the environment or damaged by infection or ambient stress [9]. Studies have shown that the classic oxidative stress pathway was an important causative factor to induce ferroptosis. Although cancer cells are under continuous oxidative stress with an exquisite balance between thiols and catalytic iron, ferroptosis does not often happen in the cancer development [10]. The underlying molecular mechanisms remain poorly understood. We herein review the occurrence and regulation of ferroptosis in various cancer cells. The opportunity and challenge of cancer treatment based on ferroptosis will be detailed, which was desired to prosper new strategies for cancer therapy of clinical value.

Ferroptosis, from a cancer perspective: an overview

Ferroptosis was first observed in oncogenic Ras-expressing human foreskin fibroblast cell line by a battery of small compounds considered as ferroptosis-inducing agents (FIN), including erastin and Ras-selective lethal small molecule 3 (RSL 3). With the following studies, the relationship of Ras oncoprotein with ferroptosis becomes agnostic. Some Ras WT cells including fibrosarcoma cells, kidney tubule cells, and T cells are vulnerable to erastin, but the RMS13 rhabdomyosarcoma cells with Ras mutation were resistant to erastin and RLS3. Indeed, the ferroptosis inducer artesunate/erastin can promote ferroptosis in a Ras-reliant way in pancreatic cancer or transformed fibroblastic cells, while in a Ras-independent manner in leukemia cells [6, 11].
Emerging evidence implicated that ferroptosis may be an adaptive process which was critical for eradicating the carcinogenic cells [1]. More clues for this role of ferroptosis can be derived from recent researches of the tumor suppressor P53 (TP53). The acetylation-defective mutant TP533KR lost the ability to induce cell senescence, apoptosis, and cell-cycle arrest, which were the main functions of TP53 in tumor suppression. Impressively, TP533KR can still hold the capacity of inhibiting tumorigenesis due to its ferroptosis induction [1214]. An argument does emerge that P53 expression may promote, limit, or detain the outset of ferroptosis in certain cells or conditions (Fig. 1). These opposite jobs of p53 in operating the process of ferroptotic cell death were executed by different mechanisms, including effects on metabolic genes transcription, post-translational regulation or by virtue of P53-P21 axis [15, 16]. The bidirectional regulation of ferroptosis by P53 in a cell-specific or context-dependent manner needs to be further investigated. Moreover, it is still obscure that what kind of role P53-target genes take part in manipulating of the ferroptotic cell death [17]?
Ferroptosis is programmed necrosis mainly triggered by extra-mitochondrial lipid peroxidation arising from an iron-dependent ROS accretion. Excessive iron originally from aberrant iron metabolism or maladjustment of two major redox systems (lipid peroxidation and thiols) was the main incentive factors of ROS production. Glutathione (GSH), a thiol-containing tripeptide, synthesis is determined by the constant import of cysteine (Cys2) by the cell surface Cys2/glutamate antiporter xCT (Fig. 2).
The activation of Ras-mitogen-activated protein kinase (MEK) signaling can attribute to the sensitivity of cancer cells to ferroptosis, resulting from its promoting iron abundance in cancer by governing the expression levels of the transferrin receptor and ferritin [2, 7, 8]. And the overactive Ras-MEK pathway may enhance ROS generation via inhibiting cystine (Cys2) uptake or mitochondrial voltage-dependent anion channel 2/3 (VDAC 2/3) and consequently sensitize cancer cells to ferroptosis [18, 19]. But scientist and skeptics argued that the conclusion is unreliable because the MEK inhibitor U0126 was used in these above studies. Compared with U0126, the special MEK1/2 inhibitor, PD0325901 cannot halt cell ferroptotic cell death induced by FIN. So MEK activity is not indispensable for ferroptosis [20, 21]. Other signal pathways were also pinpointed to regulate the process of cell ferroptotic death, e.g., Keleh-like ECH-associated protein 1 (Keap1)-nuclear factor erythroid 2-related factor 2 (Nrf2), lymphoid-specific helicase (LSH), Egl nine homolog 1 (EGLN1)/cellular myelocytomatosis oncogene (c-Myc), mevalonate (MVA), sulfur-transfer, mucin 1 C-terminal (MUC1-C)/system xc- (xCT) and heat shock factor-1 (HSF1)-heat shock protein beta-1 (HSPB1) pathway [5]. RNAi against Fms-like tyrosine kinase 3 (Flt3) saves cells from ferroptosis by limiting lipid peroxidation and inactivating p22phox which back the role for Flt3 kinase in ferroptosis. A new study indicates that adenosine 5′-monophosphate-activated protein kinase (AMPK)-mediated the phosphorylation of Beclin1 (BECN1) directly blocks the activity of system xc- and thus results in the occurrence of ferroptosis [22, 23]. Recent investigations have unraveled a perplexing network in the ferroptosis regulation which was shown in Fig. 3.
Interestingly, microRNA and long non-coding RNA (lncRNA) are increasingly recognized as the crucial mediators in the regulation of ferroptosis (Table 2). The cytosolic lncRNA P53RRA can promote ferroptosis via nuclear sequestration of P53. P53RRA interplays with the RNA recognition motif (RRM) domain of Ras GTPase-activating protein-binding protein 1 (G3BP1), resulting in the combination of G3BP1 with P53, which cause more P53 custody in the nucleus and less sequestration of p53 in the cytoplasm [24]. P53RRA increased the intracellular concentrations of iron and lipid ROS by means of the augmented P53 pathway. Taken together, the cytosolic P53RRA-G3BP1 is a novel mechanism for inducing ferroptosis in lung adenocarcinoma.
Table 2
Non-coding RNA associated with ferroptosis
ncRNA
Target
Cell lines
Mechanisms of action
Function
Ref
P53RRA
p53
A549, H522, SPCA1
Leads to higher retention of p53 in the nucleus, increases lipid ROS and iron concentrations
Promote
[24]
miR-9
GOT1
A375, G-361
Suppressed GOT1, which ultimately converts Glu to a-KG
Suppress
[25]
miR-137
SLC1A5
A375, G-361
Suppressed SLC1A5, resulting in decreased Gln uptake and malondialdehyde (MDA) accumulation
Suppress
[26]
miR-375
SLC7A11
MCF7
Effectively suppress the expression of SLC7A11
Promote
[27]
miR-27a
EJ/T24, RT112
[28]
miR-26b
CAL-27, Tca8113VC
[29]
As-SLC7A11
A433, OVCA429, TOV112D
[30]
miR-7
Nrf2
SH-SY5Y
Activates Nrf2 pathway by targeting Keap1 expression
Suppress
[31]
miR-200a
MCF-10A, MDA-MB-231
[32]
miR-101
HUVECs
Activate Nrf2 signaling by directly targeting Cul3
[34]
miR-455
hFOB1.19
[35]
miR-153
SH-SY5Y
Directly target Nrf2 and downregulate expression of Nrf2
Promote
[36]
miR-142-5p
miR-27a
miR-144
K562, SH-SY5Y
Reduced levels of Nrf2, decreased GSH
[37]
miR-93
MCF-10A, T47D
Decreased protein expression of Nrf2 and Nrf2- regulated genes
[38]
miR-34a
NRK-52E, HK-2
Decreased levels of Sirt1, which is required for the activation of Nrf2 system
[39]
miR-28
MCF-7
Inhibits Nrf2 expression through a Keap1- independent manner
[33]
miR-365-1
BEAS-2B, A549, 3 T3-L1
Decreased protein expression of Nrf2 genes
[40]
miR-193b
miR-29-b1
miR-20a
FPN
Huh7, NSCLC
Represses FPN expression by directly targeting the FPN 39UTR
Promote
[41]
miR-485-3p
HepG2, K562
[42]
miR-210
TFR
MCF7
Decreases the uptake of transferrin by inhibiting the expression of TFR
Suppress
[43]
miR-152
TFR1
SK-HEP1, HepG2
Effectively inhibit the expression level of TFR1
[44]
miR-200b
FTH
MDA-MB-231
Effectively inhibit the expression levels of FTH
[45]
miR-Let-7d
DMT1-IRE
K562, HEL
Reduces iron accumulation and simultaneously regulates the expression level of DMT1-IRE
[46]
miR-3595
ACSL4
HSC-T6
Inhibits the expression of mRNA and protein in ACSL4
Suppress
[47]
miR-205
HepG2
[48]
miR-224-5P
3 T3-L1
[49]
miR-19b-3p
CaCO2
Suppression the expression of ACSL4
[50]
miR-130a-3p
miR-150-5p
miR-144-3p
miR-16-5p
miR-7a-5p
miR-17-5p
MiR-206
ROS
Adult mongrel dogs
Increased the production of ROS by targeting SOD1
Promote
[51]
miR-155
Capan-2, Aspc-1
Increases ROS levels through inhibiting Foxo3a expression
[52]
miR-25
Rats
Restraining ROS level by targeting NOX4.
Suppress
[53]
miR-448-3p
Mice
Reduced NOX2- dependent ROS production
[54]
In melanoma cell lines G-361 and A375, miR-9 can inhibit ferroptosis via targeting glutamic-oxaloacetic transaminase 1 (GOT1), an enzyme via glutaminolysis converting glutamine (Gln) ultimately to α-ketoglutarate (α-KG), which can promote ROS accumulation and thus irritate ferroptosis [25]. Knockdown miR-9 elevated the level of GOT1 and a-KG, which subsequently increased the sensitivity of cells to erastin- and RSL3-induced ferroptosis. Intriguingly, the other study showed that miR-137 impedes ferroptosis through directly suppressing solute carrier family 1 member 5 (SLC1A5), which is a major receptor for Gln uptake [26]. Knockdown of miR-137 can enhance the antitumor activity of erastin by enhancing ferroptosis.
As a specific light-chain submission of the Cys2/glutamate (Glu) antiporter, solute carrier family 7 member 11 (SLC7A11) plays a critical role in the negative regulation of ferroptosis. Researches showed that miR-375, miR-27a, miR-26b, and As-SLC7A11 (antisense lncRNAs) could suppress the transcription of SLC7A11 mRNA and impair the strength of its protein [2730]. So, it is plausible that these miRNAs can promote ferroptosis by targeting SLC7A11.
The Nrf2 is the vital inhibitor of ferroptosis due to its ability to inhibit cellular iron uptake, limiting ROS production, and upregulating SLC7A11. First, miR-7 and miR-200a readily induce the activation of the Nrf2 pathway by repressing Keap1 expression [31, 32]. Contrastingly, miR-28 suppresses Nrf2 expression in a Keap1-independent way [33]. Second, both miR-101 and miR-455 can promote Nrf2 nuclear accumulation by targeting Cullin-3 (Cul3) [34, 35]. Finally, miR-153, miR-142-5p, miR-27a [36], miR-144 [37], miR-93 [38], miR-34a [39], miR-365-1, miR-193b, and miR-29-b1 [40] can decrease Nrf2 level through different mechanisms. These results indicated that miRNA might modify ferroptosis by means of regulating the expression of Nrf2.
It was confirmed that iron overload could contribute to ferroptosis in cancer. The iron metabolism-related genes, such as transferrin (TF), transferrin receptor 1 (TFR1), ferroportin (FPN), divalent metal transporter 1 (DMT1), ferritin heavy chain 1 (FTH1), and ferritin light chain (FTL), were the critical mediators in the ferroptosis procedure. Based on the existing research findings, miRNAs were also involved in the regulation of iron export, storage, utilization, and uptake. MiR-20a and miR-485-3p can reduce iron output by targeting FPN genes [41, 42]. MiR-210 and miR-152 inhibit the expression level of TFR, thereby reducing the uptake of TF [43, 44]. Concurrently, the expressions of miR-200b and miR-Let-7d effectively reduce iron accumulation by inhibiting the expression of FTH and DMT1-iron-responsive element (IRE), respectively [45, 46].
Beyond that, previous studies have confirmed that ROS generation requires the activation of polyunsaturated fatty acids (PUFAs) by Acyl-CoA synthetase long-chain family member 4 (ACSL4) and lysophosphatidylcholine acyltransferase 3 (LPCAT3). MiR-3595 [47], miR-205 [48], miR-224-5P [49], miR-19b-3p, miR-130a-3p, miR-150-5p, miR-144-3p, miR-16-5p, miR-7a-5p, and miR-17-5p [50] can decrease the expression of ACSL4. It is conceivable but not yet demonstrated that these miRNAs can regulate ferroptosis by targeting ACSL4.
Ultimately, ROS is the indispensable molecule in the process of ferroptosis. Previous studies have confirmed that miRNAs are closely related to redox signaling and ROS production, and we can further speculate that miRNAs can regulate ferroptosis by regulating the expression of ROS. MiR-206 significantly induces ROS accumulation by binding to the mRNA of superoxide dismutase 1 (SOD1) [51]. MiR-155 increases the generation of ROS by inducing Foxo3a deficiency [52]. Also, miR-25 and miR-448-3p have been proved to reduce ROS level by targeting the nicotinamide adenine dinucleotide phosphate-oxidase (NOX) [53, 54].

Ferroptosis, as an expanding network of programmed cell death in cancer

Apart from apoptosis and autophagy, other programmed cell death such as programmed necrosis was discovered. Ferroptosis, as well as necroptosis, parthanatos, and pyroptosis, are all belonging to the programmed necrosis, which was carried out by a specific program of genetically encoded cellular machinery demolishing the cell in an ordered fashion [9]. Over the latest 5 years, an astonishing boost in our perception of ferroptosis has been seen, and it was linked with apoptosis, autophagy, and other programmed necrosis which are tentatively investigated.

Ferroptosis and apoptosis: switch, synergism, or antagonism?

Recently, a growing research suggested the interconnection of ferroptosis and apoptosis. Besides preventing tumorigenesis by cell-cycle arrest and cell apoptosis induction, the canonical tumor suppressor protein P53 can also induce ferroptosis in certain conditions. Zheng et al. designed a novel type of P53 complex named metal organic network-P53 (MON-P53), which is tannic acid integrated with ferric ions forming MON on the external of the P53 plasmid [55]. When MON-P53 was internalized, ferric ions can induce Fenton reaction which will cause ROS generation. In vivo and vitro experiment, the dominant ferroptotic cell death, apart from cell apoptosis, was observed in the MON-P53-treated cells. The tumor growth was suppressed, and the life span of tumor-bearing mice was also prolonged. Therefore, this approach will direct a ferroptosis/apoptosis hybrid anti-cancer therapy [9].
Conversely, ferroptotic agents such as artesunate and erastin can induce the unfolded protein response (UPR) which sequentially promotes the expression of P53 upregulated modulator of apoptosis (PUMA) via C/EBP-homologous protein (CHOP) in the P53-independent way [56]. PUMA can enhance the function of apoptotic agent tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) in promoting cell apoptosis (Fig. 4). This observation of ferroptotic agent-mediated sensitization to TRAIL-induced apoptosis implies ferroptosis inducer combined with TRAIL can strongly augment the tumoricidal efficacy. Another opinion is that metabolic or other alterations related to the irritation of ferroptosis biochemically block apoptosis occurrence. Cells that were subjected to ferroptosis due to cysteine (Cys) deprivation have about 10% of the normal level of intracellular glutathione (GSH). The power of reduced GSH may be necessary for the cascade activation of caspases-3 and-8. Therefore, cells’ lack of GSH cannot activate caspases correctly [10, 57].

Autophagy degrades ferritin to facilitate ferroptosis

Autophagy is a lysosome-dependent degradation pathway. The study provided data that the activation of autophagy pathway can degrade ferritin and then trigger ferroptosis in cancer cells [58]. Ferritinophagy, the autophagic turnover of ferritin, is critical to induce ferroptosis [59]. Further study indicated that BECN1 generation from the stimulation of upregulated embryonic lethal, abnormal vision, Drosophila-like 1 (ELAVL1) was responsible for the activation of autophagy in erastin- or sorafenib-irritated ferroptosis [60].
Autophagy can contribute to ferroptosis via the generation of lysosomal ROS and providing available labile iron via NCOA4-mediated ferritinophagy. And pharmacological blockage of autophagy weakens drug-induced ferroptosis in cancer cells. Genetic knockout of autophagy-related 5 (Atg5) and Atg7 limited erastin-induced ferroptosis by decreasing lipid peroxidation and intracellular ferrous iron levels. Significantly, blockage of nuclear receptor coactivator 4 (NCOA4), which was a choosy cargo receptor for ferritinophgay, suppresses ferritin degradation and inhibits ferroptosis. Contrarily, NCOA4 overexpression reinforces ferritin degradation and then drives ferroptosis. Autophagy supplies available labile iron via NCOA4-mediated ferritinophagy to the process of ferroptosis [6163]. These results unraveled the molecular interactions between ferroptosis and apoptosis, and ferroptosis is an autophagic cell death process [64]. But in the absence of ferroptosis, prolonged iron mediates ROS accumulation and triggers autophagy and then results in autosis. Ferroptosis and autophagy can motivate cell death occurrence separately in breast cancer cells with siramesine and lapatinib treatment [58]. But contrastly, Buccarelli et al. reported that the blockage of autophagy using quinacrine can enhance glioblastoma stem cells (GSC) sensitivity to temozolomide by means of ferroptotic cell death [65].
Recent research shows that SOCS1 is required for p53 activation and the regulation of cellular senescence. SOCS1 can regulate the expression of p53 target genes such as reducing the expression of the cystine transporter SLC7A11 and the levels of glutathione, and it therefore sensitize cells to ferroptosis [66].
Although the mechanism among diverse types of regulated cell death has distinctive morphological and biochemical traits, some crosstalk still prevails between regulators and components of these various processes [67, 68]. The pathway of ferroptosis was linked with that of oxytosis by transactivating BH3-interacting domain death agonist (BID), the pro-apoptotic member of Bcl-2 family proteins, which converged to mitochondrial damage [69]. MiR137 was identified as a central mediator among apoptosis, autophagy, and ferroptosis [26]. HSPB1 is a small heat-shock protein that is crucial in controlling autophagy and ferroptosis [70]. Erastin can simultaneously induce the ferroptosis and necroptosis in HL-60 cell line [6]. Therefore, explicating how these pathways of regulated cell death are interplayed at the molecular level and how these pathways could be mapped and integrated will advance new ways to systematical research on this field. Discerning the critical factors such as ncRNAs should enable these processes to be therapeutically targeted and would be highly desired.

Aberrant ferroptosis in diverse cancer types and tissues

The susceptibility of different types of cancer cells to ferroptosis was significantly different. NCI-60, a panel of different cancer cell lines from eight various tissue types, recommended by the US National Cancer Institute Developmental Therapeutics Program. Among them, diffuse large B cell lymphomas and Renal cell carcinoma are more susceptible to erastin-induced ferroptosis than other cancer cells from the six tissues (the breast, lung, colon, melanocytes, central nervous system, and ovary) [6]. Some argue that the sensitivity of different cell lines to ferroptosis is different because of the difference of their basic metabolic state. Numerous studies have confirmed the pivotal role of ferroptosis in killing cancer cells and suppressing cancer growth. Further investigations showed that chemotherapeutic drugs such as cytarabine/ara-C, cisplatin, doxorubicin/Adriamycin, and temozolomide combining with the ferroptosis inducer erastin gained a remarkable synergistic effect on their anti-tumor activity [3]. The prognosis is better than traditional chemotherapy alone. Here, we summarize the possible mechanism of ferroptosis in various cancer types and putative indictor of ferroptosis for clinical application.

Hepatocellular carcinoma

Ferroptosis was one of the underlying mechanisms in sorafenib treating HCC. HCC cells with the retinoblastoma (RB) protein deficiency had 2–3 times higher death rate more than that of cells with a normal level of RB protein [71]. This susceptibility of HCC with deactivated RB protein to ferroptosis was due to the augment of oxidative stress response in cells from increased reactive oxygen concentration in mitochondria. Metallothionein-1g (MT-1G) is a novel negative regulator of ferroptosis in HCC. MT-1G knockdown contributed to sorafenib-induced ferroptosis by increasing lipid peroxidation and GSH depletion. CDGSH iron sulfur domain 1 (CISD1) and ACSL4 inhibition promote erastin-induced ferroptosis in HCC. Low-density lipoprotein (LDL)–docosahexaenoic acid (DHA) nanoparticles cause cell death in HCC cells through the ferroptosis pathway. The p62-Keap1-Nrf2 pathway plays a vital role in saving HCC cells from ferroptosis, and Ras/Raf/MEK pathway is reported to be a critically important target for ferroptosis in treating HCC [72].

Colorectal cancer

Xie et al. reported that colorectal cancer was resistant to ferroptosis resulting from the inhibition of dipeptidyl-peptidase-4 (DDP4) activity by TP53 in a transcription-independent way [12]. P53 loss promotes DDP4 gathering to plasma-membrane and thus augments DDP4-dependent lipid peroxidation, eventually causing ferroptotic cell death.

Gastric cancer

Hao et al. found that erastin irritates ferroptosis in GC cells [73]. Ferrostatin-1 and liproxstatin-1 can reverse this effect. C-Myc increases the expression of cysteine dioxygenase type 1 (CDO1), facilitating ferroptosis occurrence. Mechanistically, CDO1 suppression leads to the resistance of GC cells to erastin-induced ferroptosis by restoring cellular glutathione peroxidase 4 (GPX4) expression and GSH levels, and also by decreasing ROS generation. Another study reported that the CD44 variant (CD44v) stabilizes xCT at the plasma membrane and increases Cys2 uptake for GSH synthesis, blocking the ROS-induced stress signaling, and thus confer to ferroptosis resistance in GC cell [74, 75].

Ovarian cancer

Ovarian cancer cells are characteristics of ferroptosis susceptibility because of excess iron overload by its tumor-initiating cells (TICs), which have overexpressed TFR1 and decreased iron efflux pump FPN level [76]. Artesunate (ART) can induce ferroptosis in a ROS-dependent way in ovarian cancer. Ferrostatin-1 can significantly reverse ART-induced cell ferroptosis, but transferrin pretreatment augments the ferroptosis of ovarian cancer cells induced by ART via enhancing cellular iron level [77].

Prostate adenocarcinoma

Erastin induces ferroptosis in Ras-carrying human prostate adenocarcinoma cells. The phosphorylation of HSF1-dependent HSPB1 contributes to the ferroptosis resistance to erastin through inhibiting lipid ROS accumulation and iron uptake [70]. HSPB1 inhibition specifically increased erastin-induced ferroptosis by facilitating iron accumulation from the upregulation of TFR1 and slight reduction of FTH1 expression.

Breast cancer

Ma et al. reported that siramesine and lapatinib induce ferroptosis by increasing iron-dependent ROS productions, and CDO1 overexpression can exacerbate ferroptotic cell death by the further accumulation of high-level ROS result from the decreased GSH levels in breast cancer cells [58]. In contrast, MUC1-C can upregulate the GSH expression by its formation of a complex with CD44v, which cause the cripple of ferroptosis in breast cancer cells [74].

Lung cancer

Ferroptosis of lung cancer cell was first induced by erastin in the K-ras mutated A549 cells [2]. And the following report shows that erastin sensitizes lung cancer cells to cisplatin in ferroptosis manner by GSH reduction and GPXs inactivation [19]. Cysteine desulfurase (NFS1), as an iron-sulfur cluster biosynthetic enzyme, can protect cells from ferroptosis under the high-oxygen tension by sustaining the iron-sulfur cofactors. Coinhibition of NFS1 and Cys transport can evoke ferroptosis in vitro and suppress tumor growth [20].

Rhabdomyosarcoma

High level of GSH biosynthesis is essential for RMS cells to grow and become multidrug resistant. GPX4 inhibition using RSL3 and erastin can induce ferroptosis in RMS13 cells by lessening GSH level [21]. Another possible reason for high susceptibility of RMS13 cell lines to erastin and RSL3 was related to its higher activity of intrinsic Ras/extracellular signal-regulated kinase (ERK). But contrastingly, the RMS13 cells with oncogenic Ras mutation were resistant to the oxidative stress-induced ferroptosis.

Hematological malignancies

Diffuse large B-cell lymphoma (DLBCL) cells were notably one of the most sensitive to ferroptosis inducer in the eight cell lines harvested from various tissues [78]. It is proved that the enhanced sensitivity might be due to its weakness in the sulfur transfer pathways, which causes more extracellular Cys and Cys2 required for cells survival [79]. Yu et al. reported low-dose erastin could remarkably increase the ability of cytarabine and doxorubicin to kill non-APL acute myeloid leukemia (AML) cells by irritating both necroptosis and ferroptosis [80].

Ferroptosis-induction cancer therapy

Since erastin, a novel compound is found in human tumor cells in 2003 [81]. It was first identified as a ferroptosis inducer in 2012 [8], several clinical drugs have also been found to hold a capacity of inducing ferroptosis in cancer cells.

Chemotherapeutic agents

Sulfasalazine

SSZ is recently recognized as a system xc- inhibitor [41]. xCT expression has circadian rhythm and the expression of TFR1 was affected by the circadian organization of molecular clock. Bmal1 and the clock regulate the circadian rhythm of xCT expression. The clock-controlled gene c-Myc rhythmically activated the transcription of the TfR1 gene. SSZ has been reported to disrupt the circadian rhythm of transferrin receptor 1 gene expression and thus it was plausible that SSZ may affect iron metabolism [8284]. Toyokuni et al. report that sulfasalazine inhibits Cys2 uptake via system xc-, resulting in ferroptosis in glioma cells [85]. Based on the circadian rhythm of xCT, SSZ has different effects on inducing ferroptosis at various times. But some argued that ferroptosis was not observed in the mouse embryonic fibroblasts treated with sulfasalazine [86]. One reasonable interpretation was the discrepancy of different cell lines on the sensitivity to ferroptosis.

Artesunate

ART and its derivatives can produce ROS and cause oxidative stress in cancer cells. In pancreatic ductal adenocarcinoma, head and neck cancers (HNCs), and ovarian cancer cells, the mechanism underlying the antitumor effect of ART was ferroptosis-induction [87]. However, because of the activation of Nrf2-antioxidant response element signaling pathway, the ferroptosis induction of artesunate can be partially attenuated in some cisplatin-resistant HNCs [88]. So Nrf2 inhibition via silencing Keap1 helps the reversal of ferroptosis resistance to artesunate in HNC cells.

Temozolomide

TMZ markedly induces system xc- expression via the activation of activating transcription factor 4 (ATF4) and Nrf2 pathway in glioblastoma multiforme (GBM) cells [89]. Cystathionine γ-lyase (CTH), an enzyme in the transsulfuration pathway, is induced after temozolomide treatment, which can supply Cys when system xc- is blocked. Based on the finding of erastin facilitating ferroptotic cell death to temozolomide, thus, the combination of TMZ and erastin maybe a promising therapy in GMB treatment [90].

Cisplatin

From the screening among five chemotherapeutic drugs, cisplatin was found as a ferroptosis-inducer. Cisplatin exerts its cytotoxic effects on A549 and HCT116 cells to undergo ferroptosis by reduced GSH depletion together with GPXs inactivation [19].

Targeted agents

Sorafenib

Sorafenib was first identified as a ferroptosis-inducer in HCC cell lines [91]. System xc- inhibition and GSH depletion, the accomplices for ROS accumulation, were the main mechanism for sorafenib-induced ferroptosis. Haloperidol, as a sigma receptor 1 antagonist, can bolster erastin and sorafenib-induced ferroptosis by stimulating cellular iron accumulation, GSH depletion, and lipid peroxidation [92]. But the overactive p62-keap1-Nrf2 pathway will weaken the ferroptosis process, owing to the target genes of Nrf2 including heme oxygenase-1 (HO-1), FTH1, and quinone oxidoreductase-1 (NQO1) which can directly inhibit ROS accretion. Nrf2 inhibition using genetic tools or drugs could remarkably reinforce the anti-tumor effect of sorafenib [93].

Lapatinib and BAY87–2243

Lapatinib is a tyrosine kinase inhibitor. It can incite ferroptosis in breast cancer cells when it was used together with siramesine [58]. BAY87-2243, a robust inhibitor of NADH-coenzyme Q oxidoreductase, can promote ferroptosis in a dose-dependent manner on a series of BRaf (V600E) melanoma cell lines [94].

Others

Lanperisone

Lanperisone promotes ROS production to kill K-Ras-mutant mouse embryonic fibroblasts in ferroptotic ways. And it also induces lung cancer cell ferroptotic death by inhibiting Cys2 uptake in the mouse model [95].
Besides the clinically approved drugs, two antibiotics such as salinomycin and ironomycin can promote ferroptosis in colon cancer cells via interfering with iron metabolism allowing for ROS production [65]. Other natural compounds such as bromelain [50], baicalein, artenimol, artemisinin, cotylenin A (CN-A) [11], and various vitamins can regulate cell ferroptotic death by acting on the lipid peroxidation and ROS occurrence (Table 3). Numerous nanomaterials have also been prospered for ferroptosis-based cancer therapy. Most of them are iron-based nanomaterials, which can be used as carriers of certain genes, such as P53 and ACSL4, to inhibit or promote the expression of certain critical molecules correlated with ferroptosis [55, 94, 96].
Table 3
Drugs and compounds associated with ferroptosis
Category
Drugs/Compounds
Target
Mechanism
Application
Cell lines
Effect
Ref
Chemothe-rapeutic agents
Sulfasalazine
System xc-
Inhibit Cys2 uptake via system xc-
SASP enhanced ferroptosis induced by piperlongumine02 (PL)
BJeLR/HT1080
Induce
[8]
HT1080/Calu-1
[108]
HCT116, CX-1, PANC1
[22]
Artesunate
Fe
React with excess intracellular iron to promote the production of ROS
The cisplatin-resistant cancer cells were less sensitive to artesunate-induced ferroptosis
HN3, HN4, HN9
Induce
[88]
Panc-1, COLO357
[109]
BxPC-3, AsPC-1
[110]
TMZ
System xc-
TMZ induces xCT expression via Nrf2 and ATF4 activation pathway
The efficacy of TMZ can be potentiated after combination with erastin and SASP. SASP potentiates chemo-sensitivity of TMZ in xCT knockdown gliomas
F98, U251
Inhibit
[90]
u87-MG, GBM-n6, GBM-n15, a172, T98G
[89]
Cisplatin
GSH-GPXs
The depletion of reduced GSH and inactivation of GPXs
Erastin enhances the effect of cisplatin in NSCLCs.
A549, HCT116
Induce
[19]
A2780
[111]
Targeted agents
Sorafenib
System xc-
Inhibit system xc--mediated Cys2 import, leading to glutathione depletion and the iron-dependent accumulation of lipid ROS
DFX remarkably reduced the toxicity of sorafenib in an HCC cell line
HT-1080
Induce
[65]
Huh7
[71]
ACHN, PLC/PRF5
[92]
HSC-LX2, HSC
[60]
Lapatinib
Fe
Cause ferroptosis through iron transport disruption leading to increased ROS
Knockdown of FPN increased ferroptosis after siramesine and lapatinib treatment
SKBR3, MCF-7, MDA-MB-231
Induce
[58]
MCF-7, ZR-75-1
[18]
Others
Lanperisone
System xc-
Lanperisone-mediated induction of intracellular reactive oxygen species
The remarkable mechanistic similarities of LP as well as erastin underscore the potential of ROS-mediated therapies as a novel strategy to treat K-ras mutant tumors
K-ras-expressing MEFs
Induce
[95]
Artenimol artemisinin
_
Increase TFRC gene expression and ROS accumulation
Ferrostatin-1 and the iron chelator deferoxamine led to a significantly reduced cytotoxicity of artenimol
CCRF-CEM
Induce
[11]
Salinomycin
Ironomycin
Fe
Interacts with the iron
Against CSCs derived from breast human mammary epithelial cells
CSCs
Induce
[65]
Bromelain
ACSL-4
Effectively causes ferroptotic cell death by modulating ACSL-4 levels.
Increased erastin-induced ferroptosis in Kras mutant CRC cells
CT-116, DLD-1
Induce
[50]
Baicalein
LOX
Suppress both lipid peroxidation and iron accumulation; Selectively activate the Keap1-Nrf2 pathway and inhibit 12/15-LOX
PANC1, BxPc3
Inhibit
[11]
Cotylenin A
(CN-A)
_
The combined treatment with CN-A and PEITC synergistically increased ROS levels
CN-A plus PEITC inhibited the proliferation of gemcitabine-resistant PANC-1 cells
MIAPaCa-2, PANC-1
Induce
[11]
Vitamin E
LOX
Inhibits 15-lipox-ygenase via reduction of the enzyme’s non-heme iron from its active Fe3+ state to an inactive Fe2+ state
STHdhQ7/Q7
Inhibit
[112]
ATRA
LSH
Promotes ferroptosis through decreasing the expression of LSH
A549
Induce
[113]
Vitamin C
miR-93
Significantly increased Nrf2 mRNA and protein expression by decreasing miR-93
Prevents estrogen-induced breast tumor development
MCF-10A, T47D
Inhibit
[38]

Challenges

How does ferroptosis interact with other cell death at the molecular level and how could these pathways be mapped and integrated into the cellular events?
How does ncRNA regulate the process of ferroptotic cell death?
Can ferroptosis enhance the cell immunogenicity to the host and thereby evoke an adaptive immune response, as shown in necroptotic cell death?
Are the ferroptosis inducers effective in killing the cancer cells in pre-clinical or clinical trials?

Conclusion & perspective

Collectively, ferroptosis has taken a full expectation from us to provide a new approach in anti-tumor therapies. Current researches have mainly focused on the eradication of residual or resistant cancer cells, where ferroptotic cell death emerges to be a new cell death for this purpose (Fig. 5). Conspicuously, obtaining a mesenchymal cell state (e.g., epithelial-mesenchymal transition (EMT) or cancer stem cells) has been suggested to determine metastatic dissemination and chemo-resistance [97]. More recently, the cancer cells with the high-mesenchymal state have arisen as a vital mechanism of both acquired and de novo resistance to targeted therapies [98, 99]. This therapy-resistant mesenchymal cancer cells have bred a state of non-oncogene addition to GPX4, which inhibition will intuitively result in ferroptosis. Consistently, persistent cancer cells which are nominated to escape from conventional cytotoxic treatment via a dormant state tumor showed an identically selective dependency on the GPX4 pathway [100, 101]. Therefore, ferroptosis might be considered a viable therapeutic strategy to reverse therapy-resistance in cancer strategy.
Ferroptosis is a kind of programmed necrosis, which is accepted to be more immunogenic than apoptosis. Due to damage associated with molecular patterns (DAMPs) (e.g., HMGB1) release, ferroptosis was considered as a pro-inflammatory process [102]. By deliverying chemoattractant signals, ferroptosis hold the ability to recruit and activate immune cells at tumor sites, which provide the possibility of ferroptosis inducer as a suitable enhancer for anti-tumor immunotherapy treatment such as checkpoint-inhibitor [103]. Indeed, a large number of immune cells were observed inside the tumor mass when the mouse tumor xenografts underwent cell ferroptotic death induced by ultrasmall nanoparticles [104]. However, scientist skeptics argued that ferroptosis and necroinflammation did not have an unequivocal relationship [102].
Several studies of drug re-position suggest that “conventional” agents (i.e., SASP and artesunate) have antitumor therapeutic effects by activating ferroptosis [105]. As a chemo-sensitizer by ferroptosis-induction, erastin can be used with various drugs such as cisplatin, temozolomide, doxorubicin/adriamycin, and cytarabine/ara-C in different type cancers. Albeit there is limited knowledge of the elaborated mechanism in the ferroptosis pathway that is engaged by current ferroptosis inducers, ferroptosis may provide a new form of cell death for approaching the reversal of drug-resistance and boosting the host immune system. Although it was promising from the advantages of ferroptosis in cancer therapeutics, ferroptosis is still waiting for formal addressing in a pre-clinical setting and clinical achievability, partially due to the complexity of it observed in different contexts such as P53 or Ras-mutant cancer cells. Another challenge is that ferroptosis induction such as GPX4 inhibitor affects the development and function of nervous system and kidney, by causing GPX4 gene which is fundamental for embryonic development and some adult tissue homeostasis in mice [106]. In addition, another noticeable issue is that the occurrence of ferroptotic resistance, which was originally observed in the Hela cells with the erastin treatment. The resistance mechanism was the HSP27 overactivation by suppressing cytoskeleton-mediated iron absorption [107].

Acknowledgements

Not applicable

Funding

This work was supported by grants from the National Natural Science Foundation of China (81200366,81572281,81702278,81171841) and Province Natural Science Foundation of Hunan (No.14JJ6004) and the Key Subject Education Department of Hunan ([2012]594).

Availability of data and materials

The materials supporting the conclusion of this review have been included within the article.
This is not applicable for this review.
This is not applicable for this review.

Competing interests

The authors declare that they have no competing interests.

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. 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.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Literatur
2.
Zurück zum Zitat Yagoda N, Von RM, Zaganjor E, Bauer AJ, Yang WS, Fridman DJ, et al. RAS-RAF-MEK-dependent oxidative cell death involving voltage-dependent anion channels. Nature. 2007;447:864–8.PubMedPubMedCentralCrossRef Yagoda N, Von RM, Zaganjor E, Bauer AJ, Yang WS, Fridman DJ, et al. RAS-RAF-MEK-dependent oxidative cell death involving voltage-dependent anion channels. Nature. 2007;447:864–8.PubMedPubMedCentralCrossRef
3.
Zurück zum Zitat Yu H, Guo P, Xie X, Wang Y, Chen G. Ferroptosis, a new form of cell death, and its relationships with tumourous diseases. J Cell Mol Med. 2017;21:648–57.PubMedCrossRef Yu H, Guo P, Xie X, Wang Y, Chen G. Ferroptosis, a new form of cell death, and its relationships with tumourous diseases. J Cell Mol Med. 2017;21:648–57.PubMedCrossRef
4.
Zurück zum Zitat Latunde-Dada GO. Ferroptosis: role of lipid peroxidation, iron and ferritinophagy. Biochim Biophys Acta. 2017;1861(8):1893.CrossRef Latunde-Dada GO. Ferroptosis: role of lipid peroxidation, iron and ferritinophagy. Biochim Biophys Acta. 2017;1861(8):1893.CrossRef
7.
Zurück zum Zitat Yang WS, Stockwell BR. Synthetic lethal screening identifies compounds activating iron-dependent, nonapoptotic cell death in oncogenic-RAS-harboring cancer cells. Chem Biol. 2008;15:234–45.PubMedPubMedCentralCrossRef Yang WS, Stockwell BR. Synthetic lethal screening identifies compounds activating iron-dependent, nonapoptotic cell death in oncogenic-RAS-harboring cancer cells. Chem Biol. 2008;15:234–45.PubMedPubMedCentralCrossRef
8.
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;149:1060–72.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;149:1060–72.PubMedPubMedCentralCrossRef
9.
10.
Zurück zum Zitat Ueda S, Nakamura H, Masutani H, Sasada T, Yonehara S, Takabayashi A, et al. Redox regulation of caspase-3(-like) protease activity: regulatory roles of thioredoxin and cytochrome c. J Immunol. 1998;161:6689–95.PubMed Ueda S, Nakamura H, Masutani H, Sasada T, Yonehara S, Takabayashi A, et al. Redox regulation of caspase-3(-like) protease activity: regulatory roles of thioredoxin and cytochrome c. J Immunol. 1998;161:6689–95.PubMed
11.
Zurück zum Zitat Ye J, Zhang R, Wu F, Zhai L, Wang K, Xiao M, et al. Non-apoptotic cell death in malignant tumor cells and natural compounds. Cancer Lett. 2018;420:210–27.PubMedCrossRef Ye J, Zhang R, Wu F, Zhai L, Wang K, Xiao M, et al. Non-apoptotic cell death in malignant tumor cells and natural compounds. Cancer Lett. 2018;420:210–27.PubMedCrossRef
12.
Zurück zum Zitat Xie Y, Zhu S, Song X, Sun X, Fan Y, Liu J, et al. The tumor suppressor p53 limits ferroptosis by blocking DPP4 activity. Cell Rep. 2017;20:1692–704.PubMedCrossRef Xie Y, Zhu S, Song X, Sun X, Fan Y, Liu J, et al. The tumor suppressor p53 limits ferroptosis by blocking DPP4 activity. Cell Rep. 2017;20:1692–704.PubMedCrossRef
13.
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;520:57–62.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;520:57–62.PubMedPubMedCentralCrossRef
14.
Zurück zum Zitat Kaiser AM, Attardi LD. Deconstructing networks of p53-mediated tumor suppression in vivo. Cell Death Differ. 2018;25:93–103.PubMedCrossRef Kaiser AM, Attardi LD. Deconstructing networks of p53-mediated tumor suppression in vivo. Cell Death Differ. 2018;25:93–103.PubMedCrossRef
15.
Zurück zum Zitat Tarangelo A, Magtanong L, Bieging-Rolett KT, Li Y, Ye J, Attardi LD, et al. p53 suppresses metabolic stress-induced ferroptosis in cancer cells. Cell Rep. 2018;22(3):569–75.PubMedPubMedCentralCrossRef Tarangelo A, Magtanong L, Bieging-Rolett KT, Li Y, Ye J, Attardi LD, et al. p53 suppresses metabolic stress-induced ferroptosis in cancer cells. Cell Rep. 2018;22(3):569–75.PubMedPubMedCentralCrossRef
16.
Zurück zum Zitat Aubrey BJ, Kelly GL, Janic A, Herold MJ, Strasser A. How does p53 induce apoptosis and how does this relate to p53-mediated tumour suppression? Cell Death Differ. 2017;25:104–13.PubMedPubMedCentralCrossRef Aubrey BJ, Kelly GL, Janic A, Herold MJ, Strasser A. How does p53 induce apoptosis and how does this relate to p53-mediated tumour suppression? Cell Death Differ. 2017;25:104–13.PubMedPubMedCentralCrossRef
17.
Zurück zum Zitat Gnanapradeepan K, Basu S, Barnoud T, Budina-Kolomets A, Kung CP, Murphy ME. The p53 tumor suppressor in the control of metabolism and ferroptosis. Front Endocrinol. 2018;9:124.CrossRef Gnanapradeepan K, Basu S, Barnoud T, Budina-Kolomets A, Kung CP, Murphy ME. The p53 tumor suppressor in the control of metabolism and ferroptosis. Front Endocrinol. 2018;9:124.CrossRef
18.
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: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:e2307.PubMedPubMedCentralCrossRef
19.
Zurück zum Zitat Guo J, Xu B, Han Q, Zhou H, Xia Y, Gong C, et al. Ferroptosis: a novel anti-tumor action for cisplatin. Cancer Res Treat. 2018;50:445–60.PubMedCrossRef Guo J, Xu B, Han Q, Zhou H, Xia Y, Gong C, et al. Ferroptosis: a novel anti-tumor action for cisplatin. Cancer Res Treat. 2018;50:445–60.PubMedCrossRef
20.
Zurück zum Zitat Alvarez SW, Sviderskiy VO, Terzi EM, Papagiannakopoulos T, Moreira AL, Adams S, et al. NFS1 undergoes positive selection in lung tumours and protects cells from ferroptosis. Nature. 2017;551:639–43.PubMedPubMedCentralCrossRef Alvarez SW, Sviderskiy VO, Terzi EM, Papagiannakopoulos T, Moreira AL, Adams S, et al. NFS1 undergoes positive selection in lung tumours and protects cells from ferroptosis. Nature. 2017;551:639–43.PubMedPubMedCentralCrossRef
22.
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;28:2388–99.e5.PubMedCrossRefPubMedCentral Song X, Zhu S, Chen P, Hou W, Wen Q, Liu J, et al. AMPK-mediated BECN1 phosphorylation promotes ferroptosis by directly blocking system xc(−) activity. Curr Biol. 2018;28:2388–99.e5.PubMedCrossRefPubMedCentral
24.
Zurück zum Zitat Mao C, Wang X, Liu Y, Wang M, Yan B, Jiang Y, et al. A G3BP1-interacting lncRNA promotes ferroptosis and apoptosis in cancer via nuclear sequestration of p53. Cancer Res. 2018;78:3484–96.PubMedCrossRefPubMedCentral Mao C, Wang X, Liu Y, Wang M, Yan B, Jiang Y, et al. A G3BP1-interacting lncRNA promotes ferroptosis and apoptosis in cancer via nuclear sequestration of p53. Cancer Res. 2018;78:3484–96.PubMedCrossRefPubMedCentral
25.
Zurück zum Zitat Zhang K, Wu L, Zhang P, Luo M, Du J, Gao T, et al. miR-9 regulates ferroptosis by targeting glutamic-oxaloacetic transaminase GOT1 in melanoma. Mol Carcinog. 2018;57:1566–76.PubMedCrossRef Zhang K, Wu L, Zhang P, Luo M, Du J, Gao T, et al. miR-9 regulates ferroptosis by targeting glutamic-oxaloacetic transaminase GOT1 in melanoma. Mol Carcinog. 2018;57:1566–76.PubMedCrossRef
26.
Zurück zum Zitat Luo M, Wu L, Zhang K, Wang H, Zhang T, Gutierrez L, et al. miR-137 regulates ferroptosis by targeting glutamine transporter SLC1A5 in melanoma. Cell Death Differ. 2018;25:1457–72.PubMedPubMedCentralCrossRef Luo M, Wu L, Zhang K, Wang H, Zhang T, Gutierrez L, et al. miR-137 regulates ferroptosis by targeting glutamine transporter SLC1A5 in melanoma. Cell Death Differ. 2018;25:1457–72.PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Wu Y, Sun X, Song B, Qiu X, Zhao J. MiR-375/SLC7A11 axis regulates oral squamous cell carcinoma proliferation and invasion. Cancer Med. 2017;6:1686–97.PubMedPubMedCentralCrossRef Wu Y, Sun X, Song B, Qiu X, Zhao J. MiR-375/SLC7A11 axis regulates oral squamous cell carcinoma proliferation and invasion. Cancer Med. 2017;6:1686–97.PubMedPubMedCentralCrossRef
28.
Zurück zum Zitat Drayton RM, Dudziec E, Peter S, Bertz S, Hartmann A, Bryant HE, et al. Reduced expression of miRNA-27a modulates cisplatin resistance in bladder cancer by targeting the cystine/glutamate exchanger SLC7A11. Clin Cancer Res. 2014;20:1990–2000.PubMedPubMedCentralCrossRef Drayton RM, Dudziec E, Peter S, Bertz S, Hartmann A, Bryant HE, et al. Reduced expression of miRNA-27a modulates cisplatin resistance in bladder cancer by targeting the cystine/glutamate exchanger SLC7A11. Clin Cancer Res. 2014;20:1990–2000.PubMedPubMedCentralCrossRef
29.
Zurück zum Zitat Liu XX, Li XJ, Zhang B, Liang YJ, Zhou CX, Cao DX, et al. MicroRNA-26b is underexpressed in human breast cancer and induces cell apoptosis by targeting SLC7A11. FEBS Lett. 2011;585:1363–7.PubMedCrossRef Liu XX, Li XJ, Zhang B, Liang YJ, Zhou CX, Cao DX, et al. MicroRNA-26b is underexpressed in human breast cancer and induces cell apoptosis by targeting SLC7A11. FEBS Lett. 2011;585:1363–7.PubMedCrossRef
30.
Zurück zum Zitat Yuan J, Liu Z, Song R. Antisense lncRNA As-SLC7A11 suppresses epithelial ovarian cancer progression mainly by targeting SLC7A11. Pharmazie. 2017;72:402–7.PubMed Yuan J, Liu Z, Song R. Antisense lncRNA As-SLC7A11 suppresses epithelial ovarian cancer progression mainly by targeting SLC7A11. Pharmazie. 2017;72:402–7.PubMed
31.
Zurück zum Zitat Kabaria S, Choi DC, Chaudhuri AD, Jain MR, Li H, Junn E. MicroRNA-7 activates Nrf2 pathway by targeting Keap1 expression. Free Radic Biol Med. 2015;89:548–56.PubMedPubMedCentralCrossRef Kabaria S, Choi DC, Chaudhuri AD, Jain MR, Li H, Junn E. MicroRNA-7 activates Nrf2 pathway by targeting Keap1 expression. Free Radic Biol Med. 2015;89:548–56.PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Eades G, Yang M, Yao Y, Zhang Y, Zhou Q. miR-200a regulates Nrf2 activation by targeting Keap1 mRNA in breast cancer cells. J Biol Chem. 2011;286:40725–33.PubMedPubMedCentralCrossRef Eades G, Yang M, Yao Y, Zhang Y, Zhou Q. miR-200a regulates Nrf2 activation by targeting Keap1 mRNA in breast cancer cells. J Biol Chem. 2011;286:40725–33.PubMedPubMedCentralCrossRef
33.
Zurück zum Zitat Yang M, Yao Y, Eades G, Zhang Y, Zhou Q. MiR-28 regulates Nrf2 expression through a Keap1-independent mechanism. Breast Cancer Res Treat. 2011;129:983–91.PubMedPubMedCentralCrossRef Yang M, Yao Y, Eades G, Zhang Y, Zhou Q. MiR-28 regulates Nrf2 expression through a Keap1-independent mechanism. Breast Cancer Res Treat. 2011;129:983–91.PubMedPubMedCentralCrossRef
34.
Zurück zum Zitat Kim JH, Lee KS, Lee DK, Kim J, Kwak SN, Ha KS, et al. Hypoxia-responsive microRNA-101 promotes angiogenesis via heme oxygenase-1/vascular endothelial growth factor axis by targeting cullin 3. Antioxid Redox Signal. 2014;21:2469–82.PubMedPubMedCentralCrossRef Kim JH, Lee KS, Lee DK, Kim J, Kwak SN, Ha KS, et al. Hypoxia-responsive microRNA-101 promotes angiogenesis via heme oxygenase-1/vascular endothelial growth factor axis by targeting cullin 3. Antioxid Redox Signal. 2014;21:2469–82.PubMedPubMedCentralCrossRef
35.
Zurück zum Zitat Xu D, Zhu H, Wang C, Zhu X, Liu G, Chen C, et al. microRNA-455 targets cullin 3 to activate Nrf2 signaling and protect human osteoblasts from hydrogen peroxide. Oncotarget. 2017;8:59225–34.PubMedPubMedCentral Xu D, Zhu H, Wang C, Zhu X, Liu G, Chen C, et al. microRNA-455 targets cullin 3 to activate Nrf2 signaling and protect human osteoblasts from hydrogen peroxide. Oncotarget. 2017;8:59225–34.PubMedPubMedCentral
36.
Zurück zum Zitat Narasimhan M, Patel D, Vedpathak D, Rathinam M, Henderson G, Mahimainathan L. Identification of novel microRNAs in post-transcriptional control of Nrf2 expression and redox homeostasis in neuronal, SH-SY5Y cells. PLoS One. 2012;7:e51111.PubMedPubMedCentralCrossRef Narasimhan M, Patel D, Vedpathak D, Rathinam M, Henderson G, Mahimainathan L. Identification of novel microRNAs in post-transcriptional control of Nrf2 expression and redox homeostasis in neuronal, SH-SY5Y cells. PLoS One. 2012;7:e51111.PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Sangokoya C, Telen MJ, Chi JT. microRNA miR-144 modulates oxidative stress tolerance and associates with anemia severity in sickle cell disease. Blood. 2010;116:4338–48.PubMedPubMedCentralCrossRef Sangokoya C, Telen MJ, Chi JT. microRNA miR-144 modulates oxidative stress tolerance and associates with anemia severity in sickle cell disease. Blood. 2010;116:4338–48.PubMedPubMedCentralCrossRef
38.
Zurück zum Zitat Singh B, Ronghe AM, Chatterjee A, Bhat NK, Bhat HK. MicroRNA-93 regulates NRF2 expression and is associated with breast carcinogenesis. Carcinogenesis. 2013;34:1165–72.PubMedPubMedCentralCrossRef Singh B, Ronghe AM, Chatterjee A, Bhat NK, Bhat HK. MicroRNA-93 regulates NRF2 expression and is associated with breast carcinogenesis. Carcinogenesis. 2013;34:1165–72.PubMedPubMedCentralCrossRef
39.
Zurück zum Zitat Zhang Y, Tao X, Yin L, Xu L, Xu Y, Qi Y, et al. Protective effects of dioscin against cisplatin-induced nephrotoxicity via the microRNA-34a/sirtuin 1 signalling pathway. Br J Pharmacol. 2017;174:2512–27.PubMedPubMedCentralCrossRef Zhang Y, Tao X, Yin L, Xu L, Xu Y, Qi Y, et al. Protective effects of dioscin against cisplatin-induced nephrotoxicity via the microRNA-34a/sirtuin 1 signalling pathway. Br J Pharmacol. 2017;174:2512–27.PubMedPubMedCentralCrossRef
40.
Zurück zum Zitat Chorley BN, Campbell MR, Wang X, Karaca M, Sambandan D, Bangura F, et al. Identification of novel NRF2-regulated genes by ChIP-Seq: influence on retinoid X receptor alpha. Nucleic Acids Res. 2012;40:7416–29.PubMedPubMedCentralCrossRef Chorley BN, Campbell MR, Wang X, Karaca M, Sambandan D, Bangura F, et al. Identification of novel NRF2-regulated genes by ChIP-Seq: influence on retinoid X receptor alpha. Nucleic Acids Res. 2012;40:7416–29.PubMedPubMedCentralCrossRef
41.
Zurück zum Zitat Babu KR, Muckenthaler MU. miR-20a regulates expression of the iron exporter ferroportin in lung cancer. J Mol Med (Berl). 2016;94:347–59.CrossRef Babu KR, Muckenthaler MU. miR-20a regulates expression of the iron exporter ferroportin in lung cancer. J Mol Med (Berl). 2016;94:347–59.CrossRef
42.
Zurück zum Zitat Sangokoya C, Doss JF, Chi JT. Iron-responsive miR-485-3p regulates cellular iron homeostasis by targeting ferroportin. PLoS Genet. 2013;9:e1003408.PubMedPubMedCentralCrossRef Sangokoya C, Doss JF, Chi JT. Iron-responsive miR-485-3p regulates cellular iron homeostasis by targeting ferroportin. PLoS Genet. 2013;9:e1003408.PubMedPubMedCentralCrossRef
43.
Zurück zum Zitat Yoshioka Y, Kosaka N, Ochiya T, Kato T. Micromanaging iron homeostasis: hypoxia-inducible micro-RNA-210 suppresses iron homeostasis-related proteins. J Biol Chem. 2012;287:34110–9.PubMedPubMedCentralCrossRef Yoshioka Y, Kosaka N, Ochiya T, Kato T. Micromanaging iron homeostasis: hypoxia-inducible micro-RNA-210 suppresses iron homeostasis-related proteins. J Biol Chem. 2012;287:34110–9.PubMedPubMedCentralCrossRef
44.
Zurück zum Zitat Kindrat I, Tryndyak V, de Conti A, Shpyleva S, Mudalige TK, Kobets T, et al. MicroRNA-152-mediated dysregulation of hepatic transferrin receptor 1 in liver carcinogenesis. Oncotarget. 2016;7:1276–87.PubMedCrossRef Kindrat I, Tryndyak V, de Conti A, Shpyleva S, Mudalige TK, Kobets T, et al. MicroRNA-152-mediated dysregulation of hepatic transferrin receptor 1 in liver carcinogenesis. Oncotarget. 2016;7:1276–87.PubMedCrossRef
45.
Zurück zum Zitat Shpyleva SI, Tryndyak VP, Kovalchuk O, Starlarddavenport A, Chekhun VF, Beland FA, et al. Role of ferritin alterations in human breast cancer cells. Breast Cancer Res Treat. 2011;126:63–71.PubMedCrossRef Shpyleva SI, Tryndyak VP, Kovalchuk O, Starlarddavenport A, Chekhun VF, Beland FA, et al. Role of ferritin alterations in human breast cancer cells. Breast Cancer Res Treat. 2011;126:63–71.PubMedCrossRef
46.
Zurück zum Zitat Andolfo I, De Falco L, Asci R, Russo R, Colucci S, Gorrese M, et al. Regulation of divalent metal transporter 1 (DMT1) non-IRE isoform by the microRNA let-7d in erythroid cells. Haematologica. 2010;95:1244–52.PubMedPubMedCentralCrossRef Andolfo I, De Falco L, Asci R, Russo R, Colucci S, Gorrese M, et al. Regulation of divalent metal transporter 1 (DMT1) non-IRE isoform by the microRNA let-7d in erythroid cells. Haematologica. 2010;95:1244–52.PubMedPubMedCentralCrossRef
47.
Zurück zum Zitat Wu X, Zhi F, Lun W, Deng Q, Zhang W. Baicalin inhibits PDGF-BB-induced hepatic stellate cell proliferation, apoptosis, invasion, migration and activation via the miR-3595/ACSL4 axis. Int J Mol Med. 2018;41:1992–2002.PubMedPubMedCentral Wu X, Zhi F, Lun W, Deng Q, Zhang W. Baicalin inhibits PDGF-BB-induced hepatic stellate cell proliferation, apoptosis, invasion, migration and activation via the miR-3595/ACSL4 axis. Int J Mol Med. 2018;41:1992–2002.PubMedPubMedCentral
48.
Zurück zum Zitat Cui M, Xiao ZL, Sun BD, Wang Y, Zheng MY, Ye LH, et al. Involvement of cholesterol in hepatitis B virus X protein-induced abnormal lipid metabolism of hepatoma cells via up-regulating miR-205-targeted ACSL4. Biochem Biophys Res Commun. 2014;445:651–5.PubMedCrossRef Cui M, Xiao ZL, Sun BD, Wang Y, Zheng MY, Ye LH, et al. Involvement of cholesterol in hepatitis B virus X protein-induced abnormal lipid metabolism of hepatoma cells via up-regulating miR-205-targeted ACSL4. Biochem Biophys Res Commun. 2014;445:651–5.PubMedCrossRef
49.
Zurück zum Zitat Peng Y, Xiang H, Chen C, Zheng R, Chai J, Peng J, et al. MiR-224 impairs adipocyte early differentiation and regulates fatty acid metabolism. Int J Biochem Cell Biol. 2013;45:1585–93.PubMedCrossRef Peng Y, Xiang H, Chen C, Zheng R, Chai J, Peng J, et al. MiR-224 impairs adipocyte early differentiation and regulates fatty acid metabolism. Int J Biochem Cell Biol. 2013;45:1585–93.PubMedCrossRef
50.
Zurück zum Zitat Park S, Oh J, Kim M, Jin E-J. Bromelain effectively suppresses Kras-mutant colorectal cancer by stimulating ferroptosis. Anim Cells Syst. 2018;22:334–40.CrossRef Park S, Oh J, Kim M, Jin E-J. Bromelain effectively suppresses Kras-mutant colorectal cancer by stimulating ferroptosis. Anim Cells Syst. 2018;22:334–40.CrossRef
51.
Zurück zum Zitat Zhang Y, Zheng S, Geng Y, Xue J, Wang Z, Xie X, et al. MicroRNA profiling of atrial fibrillation in canines: miR-206 modulates intrinsic cardiac autonomic nerve remodeling by regulating SOD1. PLoS One. 2015;10:e0122674.PubMedPubMedCentralCrossRef Zhang Y, Zheng S, Geng Y, Xue J, Wang Z, Xie X, et al. MicroRNA profiling of atrial fibrillation in canines: miR-206 modulates intrinsic cardiac autonomic nerve remodeling by regulating SOD1. PLoS One. 2015;10:e0122674.PubMedPubMedCentralCrossRef
52.
Zurück zum Zitat Peng W, Zhu CF, Ma MZ, Gang C, Ming S, Zeng ZL, et al. Micro-RNA-155 is induced by K-Ras oncogenic signal and promotes ROS stress in pancreatic cancer. Oncotarget. 2015;6:21148–58. Peng W, Zhu CF, Ma MZ, Gang C, Ming S, Zeng ZL, et al. Micro-RNA-155 is induced by K-Ras oncogenic signal and promotes ROS stress in pancreatic cancer. Oncotarget. 2015;6:21148–58.
53.
Zurück zum Zitat Varga ZV, Kupai K, Szucs G, Gaspar R, Paloczi J, Farago N, et al. MicroRNA-25-dependent up-regulation of NADPH oxidase 4 (NOX4) mediates hypercholesterolemia-induced oxidative/nitrative stress and subsequent dysfunction in the heart. J Mol Cell Cardiol. 2013;62:111–21.PubMedCrossRef Varga ZV, Kupai K, Szucs G, Gaspar R, Paloczi J, Farago N, et al. MicroRNA-25-dependent up-regulation of NADPH oxidase 4 (NOX4) mediates hypercholesterolemia-induced oxidative/nitrative stress and subsequent dysfunction in the heart. J Mol Cell Cardiol. 2013;62:111–21.PubMedCrossRef
54.
Zurück zum Zitat Kyrychenko S, Kyrychenko V, Badr MA, Ikeda Y, Sadoshima J, Shirokova N. Pivotal role of miR-448 in the development of ROS-induced cardiomyopathy. Cardiovasc Res. 2015;108:324–34.PubMedPubMedCentralCrossRef Kyrychenko S, Kyrychenko V, Badr MA, Ikeda Y, Sadoshima J, Shirokova N. Pivotal role of miR-448 in the development of ROS-induced cardiomyopathy. Cardiovasc Res. 2015;108:324–34.PubMedPubMedCentralCrossRef
55.
Zurück zum Zitat Zheng DW, Lei Q, Zhu JY, Fan JX, Li CX, Li C, et al. Switching apoptosis to ferroptosis: metal-organic network for high-efficiency anticancer therapy. Nano Lett. 2017;17:284–91.PubMedCrossRef Zheng DW, Lei Q, Zhu JY, Fan JX, Li CX, Li C, et al. Switching apoptosis to ferroptosis: metal-organic network for high-efficiency anticancer therapy. Nano Lett. 2017;17:284–91.PubMedCrossRef
56.
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;16:1073–6.PubMedCrossRefPubMedCentral Lee YS, Lee DH, Choudry HA, Bartlett DL, Lee YJ. Ferroptosis-induced endoplasmic reticulum stress: cross-talk between ferroptosis and apoptosis. Mol Cancer Res. 2018;16:1073–6.PubMedCrossRefPubMedCentral
57.
Zurück zum Zitat Hentze H, Schmitz I, Latta M, Krueger A, Krammer PH, Wendel A. Glutathione dependence of caspase-8 activation at the death-inducing signaling complex. J Biol Chem. 2002;277:5588–95.PubMedCrossRef Hentze H, Schmitz I, Latta M, Krueger A, Krammer PH, Wendel A. Glutathione dependence of caspase-8 activation at the death-inducing signaling complex. J Biol Chem. 2002;277:5588–95.PubMedCrossRef
58.
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: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:e0182921.PubMedPubMedCentralCrossRef
59.
Zurück zum Zitat Tang M, Chen Z, Wu D, Chen L. Ferritinophagy/ferroptosis: iron-related newcomers in human diseases. J Cell Physiol. 2018;233:9179–90.PubMedCrossRef Tang M, Chen Z, Wu D, Chen L. Ferritinophagy/ferroptosis: iron-related newcomers in human diseases. J Cell Physiol. 2018;233:9179–90.PubMedCrossRef
60.
Zurück zum Zitat Zhang Z, Yao Z, Wang L, Ding H, Shao J, Chen A, et al. Activation of ferritinophagy is required for the RNA-binding protein ELAVL1/HuR to regulate ferroptosis in hepatic stellate cells. Autophagy. 2018;14:2083–103.PubMedCrossRefPubMedCentral Zhang Z, Yao Z, Wang L, Ding H, Shao J, Chen A, et al. Activation of ferritinophagy is required for the RNA-binding protein ELAVL1/HuR to regulate ferroptosis in hepatic stellate cells. Autophagy. 2018;14:2083–103.PubMedCrossRefPubMedCentral
61.
Zurück zum Zitat Abdelmonsif DA, Sultan AS, El-Hadidy WF, Abdallah DM. Targeting AMPK, mTOR and β-catenin by combined metformin and aspirin therapy in HCC: an appraisal in Egyptian HCC patients. Mol Diagn Ther. 2017;22(5):1–13. Abdelmonsif DA, Sultan AS, El-Hadidy WF, Abdallah DM. Targeting AMPK, mTOR and β-catenin by combined metformin and aspirin therapy in HCC: an appraisal in Egyptian HCC patients. Mol Diagn Ther. 2017;22(5):1–13.
62.
Zurück zum Zitat Yoshida GJ. Therapeutic strategies of drug repositioning targeting autophagy to induce cancer cell death: from pathophysiology to treatment. J Hematol Oncol. 2017;10(1):67.PubMedPubMedCentralCrossRef Yoshida GJ. Therapeutic strategies of drug repositioning targeting autophagy to induce cancer cell death: from pathophysiology to treatment. J Hematol Oncol. 2017;10(1):67.PubMedPubMedCentralCrossRef
63.
65.
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;9:841.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;9:841.PubMedPubMedCentralCrossRef
66.
Zurück zum Zitat Saintgermain E, Lian M, Vernier M. SOCS1 regulates senescence and ferroptosis by modulating the expression of p53 target genes. Aging. 2017;9(10):2137–62.CrossRef Saintgermain E, Lian M, Vernier M. SOCS1 regulates senescence and ferroptosis by modulating the expression of p53 target genes. Aging. 2017;9(10):2137–62.CrossRef
68.
Zurück zum Zitat Dong T, Liao D, Liu X, Lei X. Using small molecules to dissect non-apoptotic programmed cell death: necroptosis, Ferroptosis, and Pyroptosis. Chembiochem. 2015;16:2557–61.PubMedCrossRef Dong T, Liao D, Liu X, Lei X. Using small molecules to dissect non-apoptotic programmed cell death: necroptosis, Ferroptosis, and Pyroptosis. Chembiochem. 2015;16:2557–61.PubMedCrossRef
69.
Zurück zum Zitat Neitemeier S, Jelinek A, Laino V, Hoffmann L, Eisenbach I, Eying R, et al. BID links ferroptosis to mitochondrial cell death pathways. Redox Biol. 2017;12:558–70.PubMedPubMedCentralCrossRef Neitemeier S, Jelinek A, Laino V, Hoffmann L, Eisenbach I, Eying R, et al. BID links ferroptosis to mitochondrial cell death pathways. Redox Biol. 2017;12:558–70.PubMedPubMedCentralCrossRef
70.
71.
Zurück zum Zitat Louandre C, Marcq I, Bouhlal H, Lachaier E, Godin C, Saidak Z, et al. The retinoblastoma (Rb) protein regulates ferroptosis induced by sorafenib in human hepatocellular carcinoma cells. Cancer Lett. 2015;356:971–7.PubMedCrossRef Louandre C, Marcq I, Bouhlal H, Lachaier E, Godin C, Saidak Z, et al. The retinoblastoma (Rb) protein regulates ferroptosis induced by sorafenib in human hepatocellular carcinoma cells. Cancer Lett. 2015;356:971–7.PubMedCrossRef
72.
Zurück zum Zitat Nie J, Lin B, Zhou M, Wu L, Zheng T. Role of ferroptosis in hepatocellular carcinoma. J Cancer Res Clin Oncol. 2018;144:2329–37.PubMedCrossRef Nie J, Lin B, Zhou M, Wu L, Zheng T. Role of ferroptosis in hepatocellular carcinoma. J Cancer Res Clin Oncol. 2018;144:2329–37.PubMedCrossRef
73.
Zurück zum Zitat Hao S, Yu J, He W, Huang Q, Zhao Y, Liang B, et al. Cysteine dioxygenase 1 mediates erastin-induced ferroptosis in human gastric cancer cells. Neoplasia. 2017;19:1022–32.PubMedPubMedCentralCrossRef Hao S, Yu J, He W, Huang Q, Zhao Y, Liang B, et al. Cysteine dioxygenase 1 mediates erastin-induced ferroptosis in human gastric cancer cells. Neoplasia. 2017;19:1022–32.PubMedPubMedCentralCrossRef
74.
Zurück zum Zitat Masanori H, Hidekazu T, Hasan R, Maroof A, Yozo S, Li Y, et al. Functional interactions of the cystine/glutamate antiporter, CD44v and MUC1-C oncoprotein in triple-negative breast cancer cells. Oncotarget. 2016;7:11756–69. Masanori H, Hidekazu T, Hasan R, Maroof A, Yozo S, Li Y, et al. Functional interactions of the cystine/glutamate antiporter, CD44v and MUC1-C oncoprotein in triple-negative breast cancer cells. Oncotarget. 2016;7:11756–69.
75.
Zurück zum Zitat Ishimoto T, Nagano O, Yae T, Tamada M, Motohara T, Oshima H, et al. CD44 variant regulates redox status in cancer cells by stabilizing the xCT subunit of system xc and thereby promotes tumor growth. Cancer Cell. 2011;19(3):387–400.PubMedCrossRef Ishimoto T, Nagano O, Yae T, Tamada M, Motohara T, Oshima H, et al. CD44 variant regulates redox status in cancer cells by stabilizing the xCT subunit of system xc and thereby promotes tumor growth. Cancer Cell. 2011;19(3):387–400.PubMedCrossRef
76.
Zurück zum Zitat Basuli D, Tesfay L, Deng Z, Paul B, Yamamoto Y, Ning G, et al. Iron addiction: a novel therapeutic target in ovarian cancer. Oncogene. 2017;36:4089–99.PubMedPubMedCentralCrossRef Basuli D, Tesfay L, Deng Z, Paul B, Yamamoto Y, Ning G, et al. Iron addiction: a novel therapeutic target in ovarian cancer. Oncogene. 2017;36:4089–99.PubMedPubMedCentralCrossRef
77.
Zurück zum Zitat Greenshields AL, Shepherd TG, Hoskin DW. Contribution of reactive oxygen species to ovarian cancer cell growth arrest and killing by the anti-malarial drug artesunate. Mol Carcinog. 2016;56:75–93.PubMedCrossRef Greenshields AL, Shepherd TG, Hoskin DW. Contribution of reactive oxygen species to ovarian cancer cell growth arrest and killing by the anti-malarial drug artesunate. Mol Carcinog. 2016;56:75–93.PubMedCrossRef
78.
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;156:317–31.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;156:317–31.PubMedPubMedCentralCrossRef
79.
Zurück zum Zitat Kinowaki Y, Kurata M, Ishibashi S, Ikeda M, Tatsuzawa A, Yamamoto M, et al. Glutathione peroxidase 4 overexpression inhibits ROS-induced cell death in diffuse large B-cell lymphoma. Lab Investig. 2018;98:609–19.PubMedCrossRef Kinowaki Y, Kurata M, Ishibashi S, Ikeda M, Tatsuzawa A, Yamamoto M, et al. Glutathione peroxidase 4 overexpression inhibits ROS-induced cell death in diffuse large B-cell lymphoma. Lab Investig. 2018;98:609–19.PubMedCrossRef
80.
Zurück zum Zitat Yu Y, Xie Y, Cao L, Yang L, Yang M, Lotze MT, et al. The ferroptosis inducer erastin enhances sensitivity of acute myeloid leukemia cells to chemotherapeutic agents. Mol Cell Oncol. 2015;2:e1054549.PubMedPubMedCentralCrossRef Yu Y, Xie Y, Cao L, Yang L, Yang M, Lotze MT, et al. The ferroptosis inducer erastin enhances sensitivity of acute myeloid leukemia cells to chemotherapeutic agents. Mol Cell Oncol. 2015;2:e1054549.PubMedPubMedCentralCrossRef
81.
Zurück zum Zitat Dolma S, Lessnick SL, Hahn WC, Stockwell BR. Identification of genotype-selective antitumor agents using synthetic lethal chemical screening in engineered human tumor cells. Cancer Cell. 2003;3:285–96.PubMedCrossRef Dolma S, Lessnick SL, Hahn WC, Stockwell BR. Identification of genotype-selective antitumor agents using synthetic lethal chemical screening in engineered human tumor cells. Cancer Cell. 2003;3:285–96.PubMedCrossRef
82.
Zurück zum Zitat Yoshida GJ. Metabolic reprogramming: the emerging concept and associated therapeutic strategies. J Exp Clin Cancer Res. 2015;34(1):1–10.CrossRef Yoshida GJ. Metabolic reprogramming: the emerging concept and associated therapeutic strategies. J Exp Clin Cancer Res. 2015;34(1):1–10.CrossRef
83.
Zurück zum Zitat Okazaki F, Matsunaga N, Hamamura K, Suzuki K, Nakao T, Okazaki H, et al. Administering xCT inhibitors based on circadian clock improves antitumor effects. Cancer Res. 2017;77(23):0720.2017.CrossRef Okazaki F, Matsunaga N, Hamamura K, Suzuki K, Nakao T, Okazaki H, et al. Administering xCT inhibitors based on circadian clock improves antitumor effects. Cancer Res. 2017;77(23):0720.2017.CrossRef
85.
Zurück zum Zitat Toyokuni S, Ito F, Yamashita K, Okazaki Y, Akatsuka S. Iron and thiol redox signaling in cancer: an exquisite balance to escape ferroptosis. Free Radic Biol Med. 2017;108:610–26.PubMedCrossRef Toyokuni S, Ito F, Yamashita K, Okazaki Y, Akatsuka S. Iron and thiol redox signaling in cancer: an exquisite balance to escape ferroptosis. Free Radic Biol Med. 2017;108:610–26.PubMedCrossRef
86.
Zurück zum Zitat Yamaguchi Y, Kasukabe T, Kumakura S. Piperlongumine rapidly induces the death of human pancreatic cancer cells mainly through the induction of ferroptosis. Int J Oncol. 2018;52(3):1011–22.PubMed Yamaguchi Y, Kasukabe T, Kumakura S. Piperlongumine rapidly induces the death of human pancreatic cancer cells mainly through the induction of ferroptosis. Int J Oncol. 2018;52(3):1011–22.PubMed
87.
Zurück zum Zitat Roh JL, Kim EH, Jang HJ, Park JY, Shin D. Induction of ferroptotic cell death for overcoming cisplatin resistance of head and neck cancer. Cancer Lett. 2016;381:96–103.PubMedCrossRef Roh JL, Kim EH, Jang HJ, Park JY, Shin D. Induction of ferroptotic cell death for overcoming cisplatin resistance of head and neck cancer. Cancer Lett. 2016;381:96–103.PubMedCrossRef
88.
Zurück zum Zitat Roh JL, Kim EH, Jang H, Shin D. Nrf2 inhibition reverses the resistance of cisplatin-resistant head and neck cancer cells to artesunate-induced ferroptosis. Redox Biol. 2017;11:254–62.PubMedCrossRef Roh JL, Kim EH, Jang H, Shin D. Nrf2 inhibition reverses the resistance of cisplatin-resistant head and neck cancer cells to artesunate-induced ferroptosis. Redox Biol. 2017;11:254–62.PubMedCrossRef
89.
Zurück zum Zitat Chen L, Li X, Liu L, Yu B, Xue Y, Liu Y. Erastin sensitizes glioblastoma cells to temozolomide by restraining xCT and cystathionine-γ-lyase function. Oncol Rep. 2015;33:1465–74.PubMedCrossRef Chen L, Li X, Liu L, Yu B, Xue Y, Liu Y. Erastin sensitizes glioblastoma cells to temozolomide by restraining xCT and cystathionine-γ-lyase function. Oncol Rep. 2015;33:1465–74.PubMedCrossRef
90.
Zurück zum Zitat Sehm T, Rauh M, Wiendieck K, Buchfelder M, Eyupoglu IY, Savaskan NE. Temozolomide toxicity operates in a xCT/SLC7a11 dependent manner and is fostered by ferroptosis. Oncotarget. 2016;7:74630–47.PubMedPubMedCentral Sehm T, Rauh M, Wiendieck K, Buchfelder M, Eyupoglu IY, Savaskan NE. Temozolomide toxicity operates in a xCT/SLC7a11 dependent manner and is fostered by ferroptosis. Oncotarget. 2016;7:74630–47.PubMedPubMedCentral
91.
Zurück zum Zitat Lachaier E, Louandre C, Godin C, Saidak Z, Baert M, Diouf M, et al. Sorafenib induces ferroptosis in human cancer cell lines originating from different solid tumors. Anticancer Res. 2014;34:6417–22.PubMed Lachaier E, Louandre C, Godin C, Saidak Z, Baert M, Diouf M, et al. Sorafenib induces ferroptosis in human cancer cell lines originating from different solid tumors. Anticancer Res. 2014;34:6417–22.PubMed
92.
Zurück zum Zitat Bai T, Wang S, Zhao Y, Zhu R, Wang W, Sun Y. Haloperidol, a sigma receptor 1 antagonist, promotes ferroptosis in hepatocellular carcinoma cells. Biochem Biophys Res Commun. 2017;491:919–25.PubMedCrossRef Bai T, Wang S, Zhao Y, Zhu R, Wang W, Sun Y. Haloperidol, a sigma receptor 1 antagonist, promotes ferroptosis in hepatocellular carcinoma cells. Biochem Biophys Res Commun. 2017;491:919–25.PubMedCrossRef
93.
Zurück zum Zitat Sauzay C, Louandre C, Bodeau S, Anglade F, Godin C, Saidak Z, et al. Protein biosynthesis, a target of sorafenib, interferes with the unfolded protein response (UPR) and ferroptosis in hepatocellular carcinoma cells. Oncotarget. 2018;9:8400–14.PubMedPubMedCentralCrossRef Sauzay C, Louandre C, Bodeau S, Anglade F, Godin C, Saidak Z, et al. Protein biosynthesis, a target of sorafenib, interferes with the unfolded protein response (UPR) and ferroptosis in hepatocellular carcinoma cells. Oncotarget. 2018;9:8400–14.PubMedPubMedCentralCrossRef
94.
95.
Zurück zum Zitat Shaw AT, Winslow MM, Magendantz M, Ouyang C, Dowdle J, Subramanian A, et al. Selective killing of K-ras mutant cancer cells by small molecule inducers of oxidative stress. Proc Natl Acad Sci U S A. 2011;108:8773–8.PubMedPubMedCentralCrossRef Shaw AT, Winslow MM, Magendantz M, Ouyang C, Dowdle J, Subramanian A, et al. Selective killing of K-ras mutant cancer cells by small molecule inducers of oxidative stress. Proc Natl Acad Sci U S A. 2011;108:8773–8.PubMedPubMedCentralCrossRef
96.
Zurück zum Zitat Bazak R, Houri M, Achy SE, Hussein W, Refaat T. Passive targeting of nanoparticles to cancer: a comprehensive review of the literature. Mol Clin Oncol. 2014;2:904–8.PubMedPubMedCentralCrossRef Bazak R, Houri M, Achy SE, Hussein W, Refaat T. Passive targeting of nanoparticles to cancer: a comprehensive review of the literature. Mol Clin Oncol. 2014;2:904–8.PubMedPubMedCentralCrossRef
98.
Zurück zum Zitat Salt MB, Bandyopadhyay S, Mccormick F. Epithelial-to-mesenchymal transition rewires the molecular path to PI3K-dependent proliferation. Cancer Discovery. 2014;4:186–99.PubMedCrossRef Salt MB, Bandyopadhyay S, Mccormick F. Epithelial-to-mesenchymal transition rewires the molecular path to PI3K-dependent proliferation. Cancer Discovery. 2014;4:186–99.PubMedCrossRef
99.
Zurück zum Zitat Rizos H, Menzies AM, Pupo GM, Carlino MS, Fung C, Hyman J, et al. BRAF inhibitor resistance mechanisms in metastatic melanoma: spectrum and clinical impact. Clin Cancer Res. 2014;20:1965.PubMedCrossRef Rizos H, Menzies AM, Pupo GM, Carlino MS, Fung C, Hyman J, et al. BRAF inhibitor resistance mechanisms in metastatic melanoma: spectrum and clinical impact. Clin Cancer Res. 2014;20:1965.PubMedCrossRef
100.
Zurück zum Zitat Galluzzi L, Vitale I, Aaronson SA, Abrams JM, Adam D, Agostinis P, et al. Molecular mechanisms of cell death: recommendations of the Nomenclature Committee on Cell Death 2018. Cell Death Differ. 2018;25:486–541.PubMedPubMedCentralCrossRef Galluzzi L, Vitale I, Aaronson SA, Abrams JM, Adam D, Agostinis P, et al. Molecular mechanisms of cell death: recommendations of the Nomenclature Committee on Cell Death 2018. Cell Death Differ. 2018;25:486–541.PubMedPubMedCentralCrossRef
101.
Zurück zum Zitat Ahern TP, Lash TL, Damkier P, Christiansen PM, Cronin-Fenton DP. Statins and breast cancer prognosis: evidence and opportunities. Lancet Oncol. 2014;15:e461–8.PubMedPubMedCentralCrossRef Ahern TP, Lash TL, Damkier P, Christiansen PM, Cronin-Fenton DP. Statins and breast cancer prognosis: evidence and opportunities. Lancet Oncol. 2014;15:e461–8.PubMedPubMedCentralCrossRef
102.
Zurück zum Zitat Proneth B, Conrad M. Ferroptosis and necroinflammation, a yet poorly explored link. Cell Death Differ. 2019;26:14–24.PubMedCrossRef Proneth B, Conrad M. Ferroptosis and necroinflammation, a yet poorly explored link. Cell Death Differ. 2019;26:14–24.PubMedCrossRef
103.
Zurück zum Zitat Garg AD, Agostinis P. Cell death and immunity in cancer: from danger signals to mimicry of pathogen defense responses. Immunol Rev. 2017;280:126–48.PubMedCrossRef Garg AD, Agostinis P. Cell death and immunity in cancer: from danger signals to mimicry of pathogen defense responses. Immunol Rev. 2017;280:126–48.PubMedCrossRef
104.
Zurück zum Zitat Kim SE, Zhang L, Ma K, Riegman M, Chen F, Ingold I, et al. Ultrasmall nanoparticles induce ferroptosis in nutrient-deprived cancer cells and suppress tumour growth. Nat Nanotechnol. 2016;11:977–85.PubMedPubMedCentralCrossRef Kim SE, Zhang L, Ma K, Riegman M, Chen F, Ingold I, et al. Ultrasmall nanoparticles induce ferroptosis in nutrient-deprived cancer cells and suppress tumour growth. Nat Nanotechnol. 2016;11:977–85.PubMedPubMedCentralCrossRef
105.
Zurück zum Zitat Yoshida GJ. Therapeutic strategies of drug repositioning targeting autophagy to induce cancer cell death: from pathophysiology to treatment. J Hematol Oncol. 2017;10:67.PubMedPubMedCentralCrossRef Yoshida GJ. Therapeutic strategies of drug repositioning targeting autophagy to induce cancer cell death: from pathophysiology to treatment. J Hematol Oncol. 2017;10:67.PubMedPubMedCentralCrossRef
106.
Zurück zum Zitat Liu H, Schreiber SL, Stockwell BR. Targeting dependency on the GPX4 lipid peroxide repair pathway for cancer therapy. Biochemistry. 2018;57:2059–60.PubMedCrossRef Liu H, Schreiber SL, Stockwell BR. Targeting dependency on the GPX4 lipid peroxide repair pathway for cancer therapy. Biochemistry. 2018;57:2059–60.PubMedCrossRef
107.
Zurück zum Zitat Razaghi A, Heimann K, Schaeffer PM, Gibson SB. Negative regulators of cell death pathways in cancer: perspective on biomarkers and targeted therapies. Apoptosis. 2018;23:93–112.PubMedCrossRef Razaghi A, Heimann K, Schaeffer PM, Gibson SB. Negative regulators of cell death pathways in cancer: perspective on biomarkers and targeted therapies. Apoptosis. 2018;23:93–112.PubMedCrossRef
108.
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
109.
Zurück zum Zitat Eling N, Reuter L, Hazin J, Hamacherbrady A, Brady NR. Identification of artesunate as a specific activator of ferroptosis in pancreatic cancer cells. Oncoscience. 2015;2:517–32.PubMedPubMedCentralCrossRef Eling N, Reuter L, Hazin J, Hamacherbrady A, Brady NR. Identification of artesunate as a specific activator of ferroptosis in pancreatic cancer cells. Oncoscience. 2015;2:517–32.PubMedPubMedCentralCrossRef
110.
Zurück zum Zitat Zhou L, Zhao B, Zhang L, Wang S, Dong D, Lv H, et al. Alterations in cellular iron metabolism provide more therapeutic opportunities for cancer. Int J Mol Sci. 2018;19:1545.PubMedCentralCrossRef Zhou L, Zhao B, Zhang L, Wang S, Dong D, Lv H, et al. Alterations in cellular iron metabolism provide more therapeutic opportunities for cancer. Int J Mol Sci. 2018;19:1545.PubMedCentralCrossRef
111.
Zurück zum Zitat Sato M, Kusumi R, Hamashima S, Kobayashi S, Sasaki S, Komiyama Y, et al. The ferroptosis inducer erastin irreversibly inhibits system xc- and synergizes with cisplatin to increase cisplatin's cytotoxicity in cancer cells. Sci Rep. 2018;8:968.PubMedPubMedCentralCrossRef Sato M, Kusumi R, Hamashima S, Kobayashi S, Sasaki S, Komiyama Y, et al. The ferroptosis inducer erastin irreversibly inhibits system xc- and synergizes with cisplatin to increase cisplatin's cytotoxicity in cancer cells. Sci Rep. 2018;8:968.PubMedPubMedCentralCrossRef
112.
Zurück zum Zitat Hinman A, Holst CR, Latham JC, Bruegger JJ, Ulas G, Mccusker KP, et al. Vitamin E hydroquinone is an endogenous regulator of ferroptosis via redox control of 15-lipoxygenase. PLoS One. 2018;13:e0201369.PubMedPubMedCentralCrossRef Hinman A, Holst CR, Latham JC, Bruegger JJ, Ulas G, Mccusker KP, et al. Vitamin E hydroquinone is an endogenous regulator of ferroptosis via redox control of 15-lipoxygenase. PLoS One. 2018;13:e0201369.PubMedPubMedCentralCrossRef
113.
Zurück zum Zitat Jiang Y, Mao C, Yang R, Yan B, Shi Y, Liu X, et al. EGLN1/c-Myc induced lymphoid-specific helicase inhibits ferroptosis through lipid metabolic gene expression changes. Theranostics. 2017;7:3293–305.PubMedPubMedCentralCrossRef Jiang Y, Mao C, Yang R, Yan B, Shi Y, Liu X, et al. EGLN1/c-Myc induced lymphoid-specific helicase inhibits ferroptosis through lipid metabolic gene expression changes. Theranostics. 2017;7:3293–305.PubMedPubMedCentralCrossRef
Metadaten
Titel
Ferroptosis, a new form of cell death: opportunities and challenges in cancer
verfasst von
Yanhua Mou
Jun Wang
Jinchun Wu
Dan He
Chunfang Zhang
Chaojun Duan
Bin Li
Publikationsdatum
01.12.2019
Verlag
BioMed Central
Erschienen in
Journal of Hematology & Oncology / Ausgabe 1/2019
Elektronische ISSN: 1756-8722
DOI
https://doi.org/10.1186/s13045-019-0720-y

Weitere Artikel der Ausgabe 1/2019

Journal of Hematology & Oncology 1/2019 Zur Ausgabe

Update Onkologie

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