You are on page 1of 10

Cancer Letters 483 (2020) 127–136

Contents lists available at ScienceDirect

Cancer Letters
journal homepage: www.elsevier.com/locate/canlet

Mini-review

Ferroptosis, a novel pharmacological mechanism of anti-cancer drugs T


a,b,c a,b,c a,b,c a,b,c a,b,c
Yanwei Su , Bin Zhao , Liangfu Zhou , Zheyuan Zhang , Ying Shen ,
Huanhuan Lva,b,c, Luban Hamdy Hameed AlQudsyc, Peng Shangb,c,∗
a
School of Life Science, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China
b
Research & Development Institute of Northwestern Polytechnical University in Shenzhen Research & Development Institute of Northwestern Polytechnical University in
Shenzhen, Shenzhen, 518057, China
c
Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, School of Life Science, Northwestern Polytechnical University,
Xi'an, Shaanxi, 710072, China

ARTICLE INFO ABSTRACT

Keywords: Ferroptosis, a form of regulated cell death, is initiated by oxidative perturbations of the intracellular micro-
System xc- environment, which is under the constitutive control of glutathione peroxidase 4 (GPX4). Ferrous iron (Fe2+)
Glutathione peroxidase 4 accumulation and lipid peroxidation are critical events in the induction of ferroptosis, which is inhibited by iron
Iron chelators and lipophilic antioxidants. Ferroptosis terminates in mitochondrial dysfunction and toxic lipid per-
Ferritinophagy
oxidation. It plays a vital role in inhibiting cancer growth and proliferation. It can be induced in cancer cells, and
Reactive oxygen species
certain normal cells, by experimental compounds (e.g., erastin, Ras-selective lethal small molecule 3) or clinical
drugs. The purpose of this review is to summarize the various drugs (e.g., sulfasalazine, lanperisone, sorafenib,
fenugreek (trigonelline), acetaminophen, cisplatin, artesunate, combination of siramesine and lapatinib, fer-
umoxytol, and salinomycin (ironomycin)) that could induce ferroptosis in cancer cells and provide an overview
of current knowledge regarding the mechanisms underlying ferroptosis. In future, we anticipate the development
of more ferroptosis-inducing drugs, and the availability of such drugs for the clinical treatment of cancer.

1. Introduction RSL3 treatments do not trigger morphological changes or biochemical


processes consistent with apoptosis, like chromatin margination or poly
Ferroptosis is a form of cell death which is morphologically, bio- ADP-ribose polymerase (PARP) cleavage. Moreover, erastin- and RSL3-
chemically and genetically distinct from apoptosis, necrosis and au- induced cell deaths are not attenuated by apoptosis-, necrosis-, ne-
tophagy. Ferroptosis was discovered and identified by Dixon in 2012 croptosis-, or autophagy-inhibitors. However, antioxidants (e.g., vi-
[1]. It is characterized by an overwhelming, iron-dependent accumu- tamin E) and iron chelators (e.g., deferoxamine mesylate) block RSLs-
lation of lethal lipids and reactive oxygen species (ROS) [2]. In fer- induced cell death. Consequently, ferroptosis generally refers to an
roptotic cells, the mitochondria appear smaller and have increased iron-dependent, non-apoptotic form of regulated cell death (RCD)
mitochondrial membrane density, and reduced or vanished of mi- [5,6].
tochondrial cristae. The term ‘ferroptosis’ originated from the fact that Ferroptosis plays an important role in cancer therapy. As a recently
iron chelation, which lowers intracellular iron levels, prevents the identified condition that regulates cell death, ferroptosis is a novel
formation of reactive radical species like radical hydroxyl, and protects method for the destruction of cancer cells. In this review, we have
cells from death [1]. summarized the signaling pathways involved in ferroptosis, while fo-
Essentially, ferroptosis inducers were identified before the concept cusing on the mechanisms of ferroptosis-associated drugs against cancer
of ferroptosis. The first identified ferroptosis inducer was erastin, in cells to lay a foundation for cancer treatment.
2003, which is a synthetic lethal molecule expressing the engineered
mutant RAS oncogene in human foreskin fibroblasts (BJeLR) [3]. Era- 2. Mechanisms of ferroptosis
stin irreversibly binds to SLC7A11, and thereby inactivates it. Ras-se-
lective lethal small molecule (RSL)-3 and RSL5, which selectively killed Ferroptosis is a novel RCD with its unique morphological, bio-
BJeLR cells in a non-apoptotic manner, were later identified in 2008 in chemical and genetic hallmarks. Glutathione peroxidase 4 (GPX4) and
a high-throughput small molecule-screening study [4]. Erastin and system xc- are considered to be the primary signaling pathways


Corresponding author. Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen, 518057, China.
E-mail address: shangpeng@nwpu.edu.cn (P. Shang).

https://doi.org/10.1016/j.canlet.2020.02.015
Received 13 December 2019; Received in revised form 12 February 2020; Accepted 12 February 2020
0304-3835/ © 2020 Elsevier B.V. All rights reserved.
Y. Su, et al. Cancer Letters 483 (2020) 127–136

Fig. 1. Mechanisms of ferroptosis. The inhibition of


system xc- leads to GSH depletion and subsequent
inactivation of GPX4, finally leading to the accu-
mulation of lethal lipid peroxides and initiation of
ferroptosis in the presence of iron. Excessive
amounts of Fe2+ leads to the accumulation of lipid
ROS through the Fenton reaction, which in turn
causes ferroptosis. (GSH, glutathione; GS-SG, oxi-
dized glutathione; GPX4, glutathione peroxidase 4;
ROS, reactive oxygen species; PUFA, poly-
unsaturated fatty acids; NCOA4, nuclear receptor
co-activator 4; IREB2/IRP2, iron responsive element
binding protein 2; TFR, transferrin receptor; FPN,
ferroportin; DMT1, divalent metal transporter 1;
STEAP3, six-transmembrane epithelial antigen of
prostate 3)

associated with ferroptosis [1]. System xc- belongs to the family of cysteine is supplemented by system xc--mediated cysteine secreted from
heterodimeric amino acid transporters. SLC7A11, an amino acid the nearby somatic cells (e.g., fibroblasts, activated macrophages, or
transporter, functions to exchange L-cystine and L-glutamate [7]. Ad- dendritic cells) in their micro-environment [14]. Cancer cells expres-
ditionally, iron metabolism and lipid peroxidation are two essential sing the system xc- transporter directly take up cystine from their micro-
events in ferroptosis. Erastin, a small molecule acting as a ferroptosis environment. Therefore, system xc- transporter is a potential target for
inducer, inhibits the function of system xc- and depletes glutathione the treatment of cancers in which the growth and survival of cancer
(GSH), finally inactivating GPX4 [1]. Inactivation of GPX4 leads to the cells are heavily dependent on the uptake of amino acids.
accumulation of lipid peroxides, which further leads to an increase in Additionally, GPX4 uses GSH as a basic co-factor to catalyze the
ROS [8]. Circulating iron, ferric ion (Fe3+), is imported into cells by the reduction of hydrogen peroxide and organic peroxides, especially lipid
transferrin receptor (TFR), and subsequently converted to Fe2+ in the peroxides, to water and the corresponding alcohols. Therefore, the in-
endosome [9]. Excessive amounts of Fe2+ leads to the accumulation of hibition of system xc- leads to the depletion of GSH, and subsequent
lipid ROS through the Fenton reaction, which in turn causes ferroptosis inactivation of GPX4, finally leading to the accumulation of lethal lipid
(Fig. 1) [1]. peroxides and initiation of ferroptosis, in the presence of iron.
Inhibition of system xc--mediated GSH synthesis is a major mechanism
which induces the accumulation of lipid peroxides, and the occurrence
2.1. Inhibition of system xc- - glutathione/GPX4 axis triggers ferroptosis
of ferroptosis.
2.1.1. Inhibition of system xc- triggers ferroptosis
The first mechanism under consideration for the induction of fer- 2.1.2. Roles of GPX4 in ferroptosis
roptosis is the inhibition of system xc- (e.g., mediated by erastin), which Another molecular mechanism of ferroptosis is the direct inhibition
indirectly inhibits GPX4 [1,10]. Erastin, a classic ferroptosis agonist, of GPX4 through the loss of its activity, or by promoting its degradation.
inhibits the function of system xc-, thereby causing GSH depletion and GPX4 is a unique member of the selenium-dependent glutathione per-
GPX4 inactivation, and subsequently triggering ferroptosis (Fig. 1) oxidase family in mammals, with a pivotal role in the inhibition of lipid
[1,6,8]. peroxides during ferroptotic cell death [8]. GPX4 functions to remove
System xc-, a cystine/glutamate antiporter on the plasma mem- lipid peroxides from phospholipid membranes [15]. It reduces toxic
brane, imports extracellular cystine in exchange for intracellular glu- lipid peroxides to non-toxic lipid alcohols in the presence of GSH [8].
tamate. It plays a key role in maintaining the intracellular GSH levels The classical ferroptosis inducer RSL3 covalently targets the seleno-
and extracellular cystine/cysteine redox balance [7]. Furthermore, the cysteine in its active site, in an irreversible manner, thereby inhibiting
system xc- is a hetero-dimeric cell surface amino acid antiporter, which GPX4 enzyme activity [8,16]. Inactivation or loss of GPX4 leads to the
consists of the twelve-pass transmembrane transporter protein accumulation of lipid peroxides, which is considered as a lethal signal
SLC7A11, coupled to the single-pass transmembrane regulatory protein for ferroptosis (Fig. 1) [16,17]. GPX4 has been identified as a negative
SLC3A2, by a disulfide bond [11]. Cysteine is a precursor of GSH regulator of ferroptosis since it limits the production of lipid ROS [8].
synthesis, and cystine can be reduced by it. It is produced during me- However, the GPX4-mediated regulatory mechanism of ferroptosis is
thionine metabolism in tissues (such as liver), by trans-sulfuration still largely unknown.
pathways including the cleavage of cystathionine by gamma-cy-
stathionase to alpha-ketobutyrate and cysteine [12]. In contrast, certain 2.1.3. Roles of lipid peroxidation in ferroptosis
types of cancer cells, including leukemia and lymphomas, are unable to Lipid peroxidation is a key event in ferroptosis. The production of
generate cysteine. Likewise, to maintain growth, amino acids need to be ROS caused by excessive iron in turn causes an increase in lipid per-
taken from their micro-environment [13]. When the cysteine con- oxides. The production of lipid peroxides in cell membranes is also a
centrations are relatively low in in vivo circulation, intracellular source of lipid ROS, which in turn leads to ferroptosis [18]. The

128
Y. Su, et al. Cancer Letters 483 (2020) 127–136

overwhelming accumulation of lipid ROS leads to the execution of TFR, and reduce the chelation of ferritin during storage [31].
ferroptosis, which can be prevented by lipophilic antioxidants. Nicoti-
namide adenine dinucleotide phosphate (NAPDH) oxidases are re- 2.2.2. Ferritinophagy
sponsible for the accumulation of ROS in erastin-induced ferroptosis Essentially, ferroptosis is an autophagic cell death process [32]. It
[1]. Under normal conditions, fatty acid hydroperoxides are converted was previously suggested to be an autophagy-independent process [1].
to fatty acid alcohols by GPX4. However, the occurrence of ferroptosis However, by using various concentrations of erastin and measuring
inhibits the activity of GPX4. Accumulated fatty acid peroxides are ferroptosis at different time periods in two different cell types (MEF and
further catalyzed into lipid peroxide free radicals in the lipid-mediated HT1080 cells), Gao et al. [32] that the effect of autophagy-inhibition on
Fenton reaction. The most susceptible lipids are the polyunsaturated ferroptosis was more obvious in the early stages of ferroptosis when the
fatty acid-containing phospholipids (PUFA-PLs) which result in sub- induction was weak, and tended to be stable when induction was
sequent cell death [19]. These lipid free radicals extract protons from strong. Consequently, ferroptosis was previously considered to be in-
adjacent PUFAs, triggering a new round of lipid oxidation, further dependent of autophagy.
spreading oxidative damage from one lipid to another, and accelerating Intriguingly, autophagy plays an important role in ferroptosis,
the production of lipid ROS. PUFA oxidation and free radical-mediated through the regulation of cellular iron homeostasis and cellular ROS
damage ultimately results in the fragmentation of PUFA into a variety [32]. Increased autophagy degrades ferritin, which leads to increased
of products [20]. PUFA peroxides, as well as their final product re- iron levels, resulting in oxidative injury by Fenton reaction, an essential
actants, such as malondialdehydes and 4-hydroxynonenal, are dama- factor for ferroptosis [33,34]. This process named ferritinophagy has
ging to cells. Lipid peroxidation is the oxidative degradation of lipids, also been demonstrated to serve as a bridge between ferroptosis and
and it plays an important role in inducing ferroptosis by increasing li- autophagy [33]. Autophagy exerts positive effects on the regulation of
potoxicity [21]. ferroptosis. Remarkably, nuclear receptor coactivator 4 (NCOA4) is a
selective cargo receptor for the selective autophagic turnover of ferritin.
2.2. Activation of iron axis triggers ferroptosis Using NCOA4 as the cargo receptor, ferritinophagy recognizes ferritin
and delivers it for lysosomal degradation, resulting in the release of free
2.2.1. The roles of iron in ferroptosis iron, thereby increasing the iron levels (Fig. 1) [35].
Iron is an essential nutrient implicated in the synthesis of iron-sulfur
cluster (Fe–S), heme, and other cofactors, in all eukaryotes and most 3. Ferroptosis and cancer
prokaryotes. Ferroptosis is an iron-dependent, non-apoptotic form of
regulated cell death, which requires abundant available-cellular iron Cell death is essential for normal development, homeostasis, and
[9,22]. It has been verified that iron overload contributes to ferroptosis prevention of hyper-proliferative diseases like cancer. Despite success
in cancer cells. Iron metabolism-related proteins like transferrin (TF), in clinical cancer treatments, resistance to existing chemotherapeutic
transferrin receptor 1 (TFR1), ferroportin (FPN), divalent metal trans- drugs owing to genetic changes remains a problem [36]. Ferroptosis is
porter 1 (DMT1), ferritin heavy chain 1 (FTH1), and ferritin light chain associated with a variety of physiological and pathological processes
(FTL), are vital mediators of ferroptosis [9]. TFR1 mediates a great and diseases, especially the treatment of multiple types of cancers.
majority of the cellular iron uptake, by binding to iron-transferrin at the Numerous studies have confirmed that ferroptosis plays a key role in
cell surface, which is internalized by endocytosis through receptor- killing tumor cells and inhibiting tumor growth. Ferroptosis was iden-
binding. Iron, stored in the form of ferritin, is used up during low iron tified as the cause for the death of several types of tumorigenic cells like
levels. Degradation of ferritin in the lysosomes causes an increase in non-small cell lung cancer [37], breast cancer [38], leukemia [39],
iron levels, which further leads to the accumulation of intracellular pancreatic cancer [40], and hepatocellular carcinoma Table 1 [41].
ROS, and ultimately cell death [23]. Ferritin is the major intracellular Consequently, ferroptosis induction may be a novel therapeutic strategy
iron storage protein complex, and includes FTL1 and FTH1. Further- for cancer treatment.
more, FPN is the only iron efflux transporter in the plasma membrane, Fortunately, some clinical drugs approved by the U.S. Food and
which releases iron from cells. Summarizing, iron levels are actively Drug Administration (FDA) (e.g., sorafenib, sulfasalazine and artesu-
regulated in cells by TFR, which transports iron into cells, and FPN, nate) can induce ferroptosis in several cancer types, thereby making the
which exports iron from the cell to the outside (Fig. 1). application of ferroptosis in pre-clinical and clinical settings feasible
Ferroptosis, induced by erastin or cysteine deletion, can be pre- Table 1. Moreover, ferroptosis inducers (like erastin, piperazine erastin,
vented by TFRC gene silencing, and by the iron chelator, deferoxamine and RSL3) were found to prevent tumor growth in the xenograft model
(DFO) [4,24,25]. Conversely, supplementation with iron-bound trans- of HT-1080 cells in vivo [8]. Collectively, in the clinical setting, there is
ferrin or a bioavailable form of iron (e.g., ferric ammonium citrate), an urgent need for the discovery of ferroptosis-inducing drugs, for use
rather than other divalent metals, accelerates erastin-induced ferrop- in the treatment of tumors, especially drug-resistant tumors.
tosis [1,24]. Excess iron catalyzes the Fenton reaction, producing highly
reactive hydroxyl radicals, which enhance lipid ROS production, and 4. Ferroptosis-associated anti-cancer drugs
lead to ferroptosis [26].
Furthermore, iron regulatory proteins (IRPs), including IRP1 and 4.1. Sulfasalazine
IRP2, bind to the mRNA of iron response elements (IRE) and regulate
the expression of DMT1, TFR1, ferritin and FPN1 [27]. IRP1 is regu- Sulfasalazine, an azo bridge-linked anti-inflammatory agent, was
lated by an abnormal iron-sulfur cluster switch [28]. Iron response originally synthesized from an antibiotic, sulfapyridine, in 1940. It is
element binding protein 2 (IRE-BP2) regulates iron metabolism. It is commonly used to treat chronic inflammatory diseases like in-
considered as an important gene in the induction of ferroptosis [29]. flammatory bowel (Crohn's) disease and rheumatoid arthritis [42,43].
IRP2, an RNA-binding protein which controls the translation of a set of In Crohn's disease, 5-aminosalicylic acid and sulfasalazine appear to
mRNAs involved in iron homeostasis, reduces iron uptake by reducing have equivalent activity, each superior to sulfapyridine, while sulfa-
the expression of TFRs through the ubiquitin-proteasomal system in pyridine is the active component in rheumatoid arthritis treatment
iron-replete cells. It sequesters free Fe2+ by inducing ferritin expression [43].
[30]. In iron-depleted cells, IRP binds to IREs, which inhibit the In 2001, a study showed that sulfasalazine was a potent inhibitor of
translation of RNA elements in the mRNA of ferritin, TFR, and many system xc- [13]. Subsequently, Sulfasalazine was found to inhibit cy-
other transcripts, or prolong the half-life of mRNA. Consequently, iron- stine absorption, leading to the attenuation of GSH, and ultimately
starved cells increase the transferrin-bound iron absorption ability of resulting in the death of certain types of cancer cells, both in vitro and in

129
Y. Su, et al. Cancer Letters 483 (2020) 127–136

vivo [43]. Sulfasalazine induces ferroptosis by inhibiting the function of


Reference
system xc- (Fig. 2) [1]. Sulfasalazine has been reported to suppress drug

[77,81]
[10,41]
[1,13]

[37]

[38]

[39]
[96]
[49]

[41]
[52]
resistance in certain lung adenocarcinoma and breast cancer cells in
vitro [44,45]. Sulfasalazine is considered to be non-toxic, and therefore
it can be combined with traditional drugs to reduce drug resistance and

Mutationally-active KRas pancreatic ductal adenocarcinoma (PDAC) cell


side effects [46]. Okazaki and his colleagues demonstrated that the
combination of dyclonine and sulfasalazine co-operatively suppressed
the growth of head and neck squamous cell carcinoma or gastric tumors
highly expressing ALDH3A1, which were resistant to sulfasalazine
Hepatocellular Carcinoma Cells; head and neck cancer cells

monotherapy [47]. Moreover, Yamaguchi et al. [40] found that a triple

GSH, glutathione; ROS, reactive oxygen species; NRF2, The nuclear factor erythroid 2-related factor 2; IREB2/IRP2, Iron response element binding protein 2; TFRC, transferrin receptor.
combination treatment with piperlongumine, cotylenin A, and sulfa-
Lymphoma; small-cell lung cancer; Prostate Cancer

salazine was highly effective against pancreatic cancer.


Non-small cell lung cancer (NSCLC) cell lines

4.2. Lanperisone

The FDA-approved drug lanperisone (LP), a modified form of tol-


Hepatocellular Carcinoma Cells

perisone, which was originally developed as a muscle relaxant, has


been clinically tested and found to safely and effectively target Ras-
Primary hepatocytes cells
Kras-mutant tumor cells

mutant cancers [48]. The mechanism of cancer cell destruction by LP is


similar to the iron- and ROS-dependent mechanisms in K-ras mutant
Cancer/cancer cells

Breast cancer cells

Cancer stem cells

cells, which leads to oxidative stress and ultimately cell death [30,49].
Leukemia cells

Furthermore, it is possible that LP inhibits the function of system xc- or


other targets in the ferroptotic pathway (Fig. 2) [1]. In terms of the
underlying mechanism of action, LP appears to be similar to erastin. By
lines

binding to the mitochondrial voltage-dependent anion channels


(VDACs), erastin alters VDAC gating, leading to mitochondrial dys-
Inhibits the absorption of cystine by system xc-, causes GSH depletion; activates NRF2 against ferroptosis

function, production of ROS, and ultimately triggering ferroptosis [49].


Increases levels of IREB2/IRP2 and TFRC along with a rapid degradation of the iron storage protein
Decreases the expression of ferroportin and ferritin and increasing the expression of transferrin to

Whether LP interacts with VDAC in K-ras mutant cells, which leads to


mitochondrial dysfunction, remains undetermined. In in vivo studies, LP
was showed the ability to inhibit tumor growth in a KRAS-mutant
mouse model [49]. There are few studies on the ferroptosis of cancer
cells caused by LP. However, the specific mechanism needs more re-
Conjugates with GSH to form the Pt-GS complex causing extensive GSH depletion

search.
Promotes ferritin degradation and releases lysosomal iron, reacts with ROS

4.3. Acetaminophen
Inhibits the absorption of cystine by system xc-, causes GSH depletion
Inhibits the absorption of cystine by system xc-, causes GSH depletion

Acetaminophen (APAP) is widely used in the treatment of pain and


Two forms excess of iron produce harmful ROS and cell death

fever in humans. The highly reactive metabolite of acetaminophen is


Reacts rapidly with GSH causing extensive GSH depletion

the N-acetyl-p-benzoquinone imine (NAPQI). An early study had shown


that the early hypothermia caused by acetaminophen in mice was due
to the parent drug, and not its toxic reactive metabolite [50]. Owing to
the complexity of human thermoregulatory systems, the exact me-
chanism of hypothermia caused by acetaminophen is unclear [51].
Lorincz et al. [52] showed that, in addition to necroptosis and
apoptosis, ferroptosis was also involved in acetaminophen-induced cell
Possible mechanisms of ferroptosis

death in primary hepatocytes. However, this phenomenon was not


found in the hepatoma cell line, HepG2. It is well known that HepG2
cells practically do not express phase I enzymes, and therefore cannot
form the reactive metabolite NAPQI [53]. The highly reactive meta-
bolite NAPQI reacts rapidly with GSH, causing extensive GSH deple-
increases iron

tion, and increasing liver damage (Fig. 2) [54]. Thus, the cell death
Blocks NRF2

induced by APAP can be characterized by GSH depletion and GPX in-


ferritin

hibition. The process is independent of caspase, but involves ferroptosis


[55]. Excessive use of APAP can cause liver failure in patients, which is
aggravated by liver ferroptosis [56]. However, this hypothesis is yet to
System xc-,
System xc-
System xc-

be confirmed by in vivo and in vitro studies.


Targets
Drugs associated with ferroptosis.

NRF2
NRF2
GSH
GSH
Iron

Iron

Iron
Iron

4.4. Cisplatin
Salinomycin (ironomycin)
Siramesine and lapatinib
Fenugreek (trigonelline)

Cisplatin is an extremely effective and widely used anti-cancer drug


for the treatment of solid tumors [57]. Previously, the anti-cancer
mechanism of cisplatin was known to be primarily mediated by the
Acetaminophen
Sulfasalazine

Ferumoxytol

production of nuclear DNA adducts, leading to apoptosis [58]. Un-


Lanperisone

Artesunate
Sorafenib

Cisplatin

fortunately, many cancers often develop resistance in response to cis-


Table 1

Drugs

platin treatment [59]. Recent research has indicated that cisplatin is an


inducer of both ferroptosis and apoptosis in the non-small cell lung

130
Y. Su, et al. Cancer Letters 483 (2020) 127–136

Fig. 2. Inhibition of system xc--glutathione/GPX4


axis triggers ferroptosis. Sulfasalazine inhibits the
absorption of cystine by system xc-. Lanperisone
induces a non-apoptotic, iron-dependent death in
mouse KRAS-mutant tumor cells that may inhibit
the function of system xc- or other targets in the
ferroptotic pathway. The highly reactive metabolite
NAPQI of acetaminophen rapidly reacts with GSH
causing extensive GSH depletion. Cisplatin is con-
jugated with GSH to form the Pt-GS complex.
(NAPQI, N-acetyl-p-benzoquinone imine; GSH, glu-
tathione; GS-SG, oxidized glutathione; GPX4, glu-
tathione peroxidase 4; ROS, reactive oxygen species;
PUFA, polyunsaturated fatty acids)

cancer (NSCLC) cell lines A549 and HCT116 cells [37]. adaptor p62 protein prevents NRF2 degradation, and enhances sub-
Platinum compounds, including cisplatin, have a higher affinity for sequent NRF2 nuclear accumulation by the inactivation of Kelch-like
thiol-rich biomolecules. GSH is one of the most abundant non-protein ECH-associated protein 1 (Keap1) [41]. Additionally, NRF2 protein
thiols in cells. In the cytoplasm, the major part of intracellular cisplatin activates MafG, and induces multiple genes including quinone oxidor-
is found conjugated to GSH, to form the Pt-GS complex (Fig. 2) [60]. eductase 1 (NQO1), heme oxygenase-1 (HO1), and ferritin heavy chain
Similar to erastin, GSH depletion along with the inactivation of GPXs 1 (FTH1) [66]. Metallothionein-1G (MT-1G) is a new regulator of fer-
was the underlying mechanism of action of cisplatin. However, the roptosis in HCC cells [67]. Metallothioneins (MT) are a family of high-
inhibitory effect of cisplatin on GPXs was weaker than that of erastin on level small stress response proteins, which are induced by oxidative
GPXs. Additionally, Guo et al. [37] showed that the antitumor activity stress. They can effectively scavenge ROS [68]. On treatment with
of cisplatin combined with erastin was superior to that of cisplatin sorafenib, under the high expression of NRF2, the expression of MT-1G
alone. Considering the different mechanisms of these two inducers, (but not other MTs) in HCC cells was significantly up-regulated. MT-1G
combining erastin with cisplatin may be a better strategy to improve inhibits ferroptosis by blocking GSH depletion-mediated lipid perox-
the efficacy of cisplatin treatment [37,61]. Drug resistance was found to idation, a process that promotes sorafenib resistance in HCC cells [67].
affect cisplatin-induced apoptosis, but not cisplatin-induced ferroptosis. In summary, NRF2 is a master transcription factor which prevents
Therefore, ferroptosis provides a novel approach for the clinical ap- sorafenib-induced ferroptosis by regulating in redox and iron metabo-
plication of cisplatin, and a bright future for tumor resistance to cis- lism [67].
platin. Ferroptosis is a potential target in the treatment of HCC, thereby
opening new avenues for the optimum usage of sorafenib in tumors
4.5. Sorafenib [64]. Sorafenib is the first clinically approved anticancer drug, which
induces ferroptosis [65]. The combination of erastin and sorafenib may
Sorafenib, an inhibitor of oncogenic kinases, is an FDA-approved prove to be a promising therapeutic strategy for cancer treatment,
drug for the treatment of patients with advanced renal cell carcinoma especially in cases of sorafenib resistance [36]. Moreover, NRF2 and
(RCC), advanced hepatocellular carcinoma (HCC), and other solid tu- MT-1G inhibit sorafenib-induced ferroptosis, and the inhibition of these
mors [62]. The antitumor efficiency of sorafenib correlates with the regulators can increase the resistance to sorafenib, thereby providing a
inhibition of molecular components in the Raf-MEK-ERK signaling promising strategy for HCC treatment. Conversely, haloperidol pro-
pathway, and several receptor tyrosine kinases including the vascular motes sorafenib-induced ferroptosis, thereby presenting a novel
endothelial growth factor receptor (VEGFR), thereby inhibiting strategy for the treatment of HCC with sorafenib [69].
neoangiogenesis [63].
Recently, Louandre et al. [64] found that sorafenib had the ability 4.6. Fenugreek (trigonelline)
induce ferroptosis. However, the induction of ferroptosis was unrelated
to the RAF kinase inhibitory effect of sorafenib [65]. There are two Trigonelline, the main pharmacological ingredient of a traditional
main mechanisms by which sorafenib effects ferroptosis (Fig. 3). Like Chinese herbal medicine Trigonella foenum-graecum L. (fenugreek), is an
erastin, sorafenib inhibits system xc--mediated cystine import, leading alkaloid present in considerable amounts in coffee and fenugreek seed.
to endoplasmic reticulum stress (ER stress), GSH depletion and the iron- Fenugreek plays an important role in the treatment of diabetes, com-
dependent accumulation of lipid ROS [10]. Another mechanism is as- plications of diabetes, and central nervous system disorders.
sociated with the p62-Keap1-NRF2 pathway (Fig. 3) [41]. The nuclear Furthermore, trigonelline has been shown to reduce diabetic auditory
factor erythroid 2-related factor 2 (NRF2) is a key factor in determining neuropathy and platelet aggregation, by affecting β cell regeneration,
the therapeutic response of ferroptosis-targeted therapies in HCC cells. insulin secretion, activities of enzymes associated with glucose meta-
Under the treatment of sorafenib, the expression of the substrate bolism, ROS, axonal extension, and neuron excitability [70]. Recently,

131
Y. Su, et al. Cancer Letters 483 (2020) 127–136

Fig. 3. Inhibition of NRF2 pathway triggers ferrop-


tosis. Treatment with sorafenib effects ferroptosis by
two main mechanisms. First, sorafenib inhibits the
function of system xc- to induce ferroptosis. Second,
the p62-Keap1-NRF2 pathway inhibits sorafenib-
induced ferroptosis in HCC. Fenugreek (trigonel-
line) induces ferroptosis by inhibiting NRF2. (NRF2,
nuclear factor erythroid 2-related factor; Keap1,
Kelch-like ECH-associated protein 1; MT-1G,
Metallothionein-1G; NQO1, quinone oxidoreductase
1; HO1, heme oxygenase-1; FTH1, ferritin heavy
chain 1; GSH, glutathione; GPX4, glutathione per-
oxidase 4; ROS, reactive oxygen species) (The solid
line represents what has been reported, while the
dotted line is a prediction.)

trigonelline has been reported to act against NRF2 (Fig. 3) [71,72]. including ART, altered the mRNA levels of numerous iron-related
Inhibition of NRF2 by the alkaloid trigonelline was found to sig- genes, which contribute to cell death in cancer cells [81]. This process
nificantly enhance the anticancer activity of erastin and sorafenib in may also be involved in ferroptosis, and requires more research.
HCC cells and tumor xenograft models [41]. On inhibition of NRF2 by
trigonelline, the expression of MT-1G was blocked, which reduced the
content of GSH and further caused ferroptosis [41]. 4.8. Siramesine and lapatinib

Siramesine, a sigma-2 receptor ligand, is a lysosomotropic agent


4.7. Artesunate
that was originally used to treat depression [82]. Siramesine-treated
tumor cells displayed lysosomal membrane permeabilization, which
Artesunate (ART), a water soluble derivative of artemisinin, is a
lead to the release of cathepsin B and an increase in ROS, resulting in
first-line drug in the treatment of malaria [73]. ARTs are sesquiterpene
cell death [83]. Siramesine has earlier been shown to be well tolerated
lactones with a 1, 2, 4-trioxane core incorporating an endoperoxide
by humans. Lapatinib is a potent double tyrosine kinase inhibitor of the
linkage [74]. The mechanism of action of ART against malaria is still
epidermal growth factor receptor (EGFR, ErbB-1) and ErbB-2 [84]. Both
controversial. A widespread assumption is that ART derivatives are pro-
in vitro and in vivo studies demonstrated the ability of lapatinib to in-
drugs which are activated by the cleavage of the endoperoxide bridge,
hibit the proliferation of ErbB2 and EGFR-overexpressing cancer cells
in the presence of iron or heme, through Fenton reaction. This reaction
[85]. Although lapatinib presents a new treatment choice for ErbB2-
produces ROS and carbon-centered free radicals, which impose direct
positive cancer, lapatinib monotherapy frequently demonstrated only
toxicity on the intra-erythrocytic parasites [74,75].
modest activity in intermediate ErbB2-positive breast cancer cells [86].
Recent studies have revealed that ART selectively induces ferrop-
A recent study showed that the combination of siramesine and la-
tosis in mutationally-active KRAS pancreatic ductal adenocarcinoma
patinib acted synergistically and induced ferroptosis-mediated cell
(PDAC) cell lines [76]. This process is blocked by the iron chelator DFO,
death in breast cancer cells [26,38]. Ma et al. [38] found that the
and enhanced by exogenous lysosomal iron. In addition to the iron
combination of siramesine and lapatinib induced ferroptosis in MDA
chelator DFO, ROS inhibitors (e.g., trolox and ferrostatin-1) except
MB 231 and SKBR3 cells by lowering the expression of ferroportin and
necrostatin-1, were found to significantly inhibit ART-induced ferrop-
ferritin, and increasing the expression of transferrin, thereby increasing
tosis in PDAC cells. Moreover, ART is preferably accumulated in the
the free iron content. Subsequently, the excess iron catalyzes the Fenton
lysosomes, and activates lysosomal function via the promotion of ly-
reaction, which produces extremely active hydroxyl radicals, and raises
sosomal V-ATPase assembly. Furthermore, lysosomes function up-
ROS production, leading to ferroptosis (Fig. 4). However, this process is
stream of the mitochondria in the production of ROS. ART treatment
associated with autophagy, and further research is required to de-
activates lysosomal function, and promotes ferritin degradation [77].
termine if ferritinophagy is involved [1,38]. The ferroptosis triggered
Lysosomal iron has been shown to react with ROS, leading to the for-
by the combination of lapatinib and siramesine acts independent of the
mation of free radicals via Fenton reaction, which results in lysosomal
downstream targets (EGFR family members) and cathepsin B [38]. This
bursting and cell death [78]. It has been reported that artesunate se-
indicates the involvement of other targets of siramesine and lapatinib in
lectively killed head and neck cancer (HNC) cells by inducing the iron-
ferroptosis. Therefore, more in-depth research is required to explore the
dependent, ROS-accumulated ferroptosis. Artesunate induces ferrop-
targets of siramesine and lapatinib associated with ferroptosis. These
tosis via the accumulation of iron, leading to elevated lipid peroxides
results provide us with a novel way to induce ferroptosis - by altering
and reduced antioxidant capacity [79,80]. Surprisingly, artesunate also
the transport of iron-utilizing clinical drugs – aiding in the development
triggers ferritinophagy accompanied by the up-regulation of NCOA4
of new therapeutic strategies to overcome the apoptosis resistance of
(Fig. 4) [80].
breast cancer.
Additionally, a study showed that 10 artemisinin derivatives,

132
Y. Su, et al. Cancer Letters 483 (2020) 127–136

Fig. 4. Activation of iron axis triggers ferroptosis.


Artesunate treatment activates lysosomal function
and promotes ferritin degradation. Artesunate also
triggers ferritinophagy by up-regulating NCOA4.
The combination of siramesine and lapatinib in-
duces ferroptosis by decreasing the expression of
ferroportin and ferritin, and increasing the expres-
sion of transferrin, to increase the free iron level in
MDA MB 231 and SKBR3 cells. Ferumoxytol directly
increases intracellular iron in leukemia cells.
Salinomycin (Ironomycin) affects the rise in IREB2/
IRP2 and TFRC/CD71 levels as well as the rapid
degradation of the iron storage protein, ferritin.
More free iron is released, further leading to in-
tracellular ROS accumulation, which ultimately
leads to ferroptosis.(NCOA4, nuclear receptor co-
activator 4; IREB2/IRP2, iron responsive element
binding protein 2; TFRC/CD71, transferrin receptor;
TFR, transferrin receptor; FPN, ferroportin; DMT1,
divalent metal transporter 1; STEAP3, six-trans-
membrane epithelial antigen of prostate 3; ROS,
reactive oxygen species)

4.9. Ferumoxytol [95]. Salinomycin and ironomycin prevented the movement of iron
from the lumen to the cytosol, thereby triggering an iron depletion
Ferumoxytol (Feraheme), an FDA-approved super-paramagnetic reaction. This response was characterized by increased levels of iron
iron oxide nanoparticle with a carbohydrate shell composed of poly- responsive element binding protein 2 (IREB2/IRP2) and transferrin
glucose sorbitol carboxymethyl ether (PSC), is clinically used in the receptor (TFRC/CD71), and rapid degradation of the iron storage pro-
treatment of iron deficiency associated anemia in patients in the USA tein, ferritin (Fig. 4) [95,96]. Ironomycin was physically accumulated
[87]. Ferumoxytol is produced by AMAG pharmaceuticals, Inc. (Cam- in lysosomes, which induced iron loading in the lysosomes, and the
bridge, MA), with a colloidal particle size of 30 nm and a molecular production of ROS, by Fenton reaction. These effects of ironomycin -
weight of 750 kDa [88]. Ferumoxytol reduces potential toxicity by se- the induction of excess iron and ROS - imply the activation of ferrop-
parating bioactive iron cores from plasma components, until the iron- tosis [97].
PSC complex enters the reticuloendothelial system macrophages [89].
Ferumoxytol is a sterile liquid with a neutral pH which is isotonic with
5. Conclusions and perspectives
mannitol. Preliminary data indicates that it contains less free iron
compared to other intravenous iron preparations [88]. Therefore, fer-
In summary, the aforementioned drugs induce ferroptosis by in-
umoxytol can be administered quickly in relatively high doses [90].
hibiting the system xc- -glutathione/GPX4 axis or by regulating iron
Ferumoxytol has been widely considered as safe when given as a single
homeostasis. However, the exact mechanism of ferroptosis induction by
infusion.
these drugs needs further clinical validation. This review provides a
Recently, Trujillo-Alonso et al. [91] found that ferumoxytol showed
comprehensive summary of drugs that can induce ferroptosis, laying
anti-leukemic effect in vitro and in vivo. Low expression of ferroportin
the foundation for future clinical treatment of cancer Table 1.
results in the susceptibility of leukemia cells to ferumoxytol through an
Ferroptosis was previously thought to be a non-apoptotic, non-au-
increase in intracellular iron. Ferumoxytol's iron core consists of 5874
tophagy, non-necrotic RCD. However, there is growing evidence that
iron atoms, and is a mixture of the ferrous and ferric forms of iron.
ferroptosis is associated with autophagy [25]. Beclin 1 (BECN1) is a key
During Fenton reaction and in the presence of peroxides, the two forms
player of macroautophagy/autophagy. Song et al. [98] found that
of iron of the ferumoxytol overproduce harmful ROS which causes cell
BECN1 was a novel driver of ferroptosis in 2018. It promoted ferrop-
death (Fig. 4) [92]. Ferumoxytol treatment significantly reduces the
tosis by directly blocking system xc- activity by binding to SLC7A11
disease burden in the mouse leukemia model, as well as patient-derived
[99]. The specific link between ferroptosis and autophagy requires
xenografts of leukemia cells with low ferritin expression [39]. The
more in-depth research. Further studies are required to decipher if
mechanism of action of ferumoxytol in destroying cancer cells may be
BECN1 could link the two pathways of cell death. Therefore, further
associated with ferroptosis.
research is required to identify the differences and associations between
ferroptosis and other regulated cell death pathways (Fig. 5).
4.10. Salinomycin (ironomycin) There are many kinds of ferroptosis-inducers, which act by the in-
hibition of system xc-, or by inhibiting GSH, or by directly inhibiting
Salinomycin is a monocarboxylic polyether antibiotic isolated from GPX4, or by altering intracellular iron levels. Generally, ferroptosis is
the bacterial strain Streptomyces albus. It is a broad-spectrum anti- achieved in two ways, either by affecting lipid oxidation or affecting
bacterial agent against gram-positive bacteria, fungi and parasites iron metabolism. Depending on the type of cancer, physiological con-
[93,94]. Salinomycin is a selective agent for cancer stem cells (CSCs) dition, and even individual lifestyle, different cancer cells respond
through an elusive mechanism. Hamai and colleagues developed a differently to ferroptosis inducers and have different sensitivities to
synthetic derivative of salinomycin – ironomycin - which is at least 10- ferroptosis. Researchers need to explore more drugs targeting these two
fold more potent than salinomycin against CSCs both in vitro and in vivo important events in ferroptosis to fight cancer (Fig. 5). Simultaneously,

133
Y. Su, et al. Cancer Letters 483 (2020) 127–136

[9] K. Gkouvatsos, G. Papanikolaou, K. Pantopoulos, Regulation of iron transport and


the role of transferrin, Biochim. Biophys. Acta 1820 (2012) 188–202.
[10] S.J. Dixon, D. Patel, M. Welsch, R. Skouta, E. Lee, M. Hayano, A.G. Thomas,
C. Gleason, N. Tatonetti, B.S. Slusher, B.R. Stockwell, Pharmacological inhibition of
cystine-glutamate exchange induces endoplasmic reticulum stress and ferroptosis,
Elife 3 (2014).
[11] H. Sato, M. Tamba, T. Ishii, S. Bannai, Cloning and expression of a plasma mem-
brane cystine/glutamate exchange transporter composed of two distinct proteins, J.
Biol. Chem. 274 (1999) 11455–11458.
[12] S.P. Stabler, J. Lindenbaum, D.G. Savage, R.H. Allen, Elevation of serum cy-
stathionine levels in patients with cobalamin and folate-deficiency, Blood 81 (1993)
3404–3413.
[13] P.W. Gout, A.R. Buckley, C.R. Simms, N. Bruchovsky, Sulfasalazine, a potent sup-
pressor of lymphoma growth by inhibition of the x(c)- cystine transporter: a new
action for an old drug, Leukemia 15 (2001) 1633–1640.
[14] A.L. Edinger, C.B. Thompson, Antigen-presenting cells control T cell proliferation
by regulating amino acid availability, Proc. Natl. Acad. Sci. U. S. A. 99 (2002)
1107–1109.
Fig. 5. Perspective of ferroptosis. Many unsolved mysteries like the relationship [15] F. Ursini, M. Maiorino, M. Valente, L. Ferri, C. Gregolin, Purification from pig-liver
of a protein which protects liposomes and biomembranes from peroxidative de-
between ferroptosis and other RCD, and the relationship between cell hetero-
gradation and exhibits glutathione-peroxidase activity on phosphatidylcholine hy-
geneity and ferroptosis, are yet to be solved. There is an urgent need to discover droperoxides, Biochim. Biophys. Acta 710 (1982) 197–211.
new ferroptosis inducers and new effective combination regimens. Currently, [16] W.S. Yang, K.J. Kim, M.M. Gaschler, M. Patel, M.S. Shchepinov, B.R. Stockwell,
no accurate method or indicator exists to determine the occurrence of ferrop- Peroxidation of polyunsaturated fatty acids by lipoxygenases drives ferroptosis,
tosis in vivo. (RCD, regulated cell death) Proc. Natl. Acad. Sci. U. S. A. 113 (2016) E4966–E4975.
[17] V.E. Kagan, G. Mao, F. Qu, J.P. Angeli, S. Doll, C.S. Croix, H.H. Dar, B. Liu,
V.A. Tyurin, V.B. Ritov, A.A. Kapralov, A.A. Amoscato, J. Jiang, T. Anthonymuthu,
there is an urgent need to discover new ferroptosis inducers or effective D. Mohammadyani, Q. Yang, B. Proneth, J. Klein-Seetharaman, S. Watkins, I. Bahar,
J. Greenberger, R.K. Mallampalli, B.R. Stockwell, Y.Y. Tyurina, M. Conrad, H. Bayir,
combination regimens, as well as more effective and safe chemotherapy Oxidized arachidonic and adrenic PEs navigate cells to ferroptosis, Nat. Chem. Biol.
to better utilize ferroptosis to treat cancer (Fig. 5). 13 (2017) 81–90.
Ferroptosis is also involved in cancer development and therapeutic [18] G.O. Latunde-Dada, Ferroptosis: role of lipid peroxidation, iron and ferritinophagy,
Biochim. Biophys. Acta Gen. Subj. 1861 (2017) 1893–1900.
response and contributes to tumor suppression activities. Although [19] S. Doll, B. Proneth, Y.Y. Tyurina, E. Panzilius, S. Kobayashi, I. Ingold, M. Irmler,
many in vitro experiments have shown that ferroptosis can be detected J. Beckers, M. Aichler, A. Walch, H. Prokisch, D. Trumbach, G. Mao, F. Qu, H. Bayir,
by several measurements like cell viability, iron levels, and ROS levels, J. Fullekrug, C.H. Scheel, W. Wurst, J.A. Schick, V.E. Kagan, J.P. Angeli, M. Conrad,
ACSL4 dictates ferroptosis sensitivity by shaping cellular lipid composition, Nat.
there are no accurate methods or indicators to identify the presence of
Chem. Biol. 13 (2017) 91–98.
ferroptosis in vivo (Fig. 5). Further research is required to understand [20] Z.Y. Cheng, Y.Z. Li, What is responsible for the initiating chemistry of iron-mediated
the mechanisms of ferroptosis. A deeper understanding of ferroptosis is lipid peroxidation: an update (vol 107, pg 748, 2007), Chem. Rev. 107 (2007) 2165-
certainly beneficial to the treatment of related diseases. 2165.
[21] W.S. Yang, B.R. Stockwell, Ferroptosis: death by lipid peroxidation, Trends Cell
Biol. 26 (2016) 165–176.
Funding [22] M. Conrad, V.E. Kagan, H. Bayir, G.C. Pagnussat, B. Head, M.G. Traber,
B.R. Stockwell, Regulation of lipid peroxidation and ferroptosis in diverse species,
Genes Dev. 32 (2018) 602–619.
This work is supported by grants from the National Natural Science [23] T. Asano, M. Komatsu, Y. Yamaguchi-Iwai, F. Ishikawa, N. Mizushima, K. Iwai,
Foundation of China (51777171, 81803032), the Northwestern Distinct mechanisms of ferritin delivery to lysosomes in iron-depleted and iron-
Polytechnical University Foundation for Fundamental Research replete cells, Mol. Cell Biol. 31 (2011) 2040–2052.
[24] M. Gao, P. Monian, N. Quadri, R. Ramasamy, X. Jiang, Glutaminolysis and trans-
(3102018JGC012), Natural Science Foundation of Shaanxi Province ferrin regulate ferroptosis, Mol. Cell. 59 (2015) 298–308.
(NO. 2019JQ632), and Science and Technology Planning Project of [25] S.J. Dixon, K.M. Lemberg, M.R. Lamprecht, R. Skouta, E.M. Zaitsev, C.E. Gleason,
Shenzhen of China (JCYJ20170412140904406). D.N. Patel, A.J. Bauer, A.M. Cantley, W.S. Yang, B. Morrison, B.R. Stockwell,
Ferroptosis: an iron-dependent form of nonapoptotic cell death, Cell 149 (2012)
1060–1072.
Declaration of competing interest [26] S. Ma, R.F. Dielschneider, E.S. Henson, W. Xiao, T.R. Choquette, A.R. Blankstein,
Y. Chen, S.B. Gibson, Ferroptosis and autophagy induced cell death occur in-
dependently after siramesine and lapatinib treatment in breast cancer cells, PloS
The authors declare no conflict of interest. One 12 (2017) e0182921.
[27] G. Gao, J. Li, Y. Zhang, Y.Z. Chang, Cellular iron metabolism and regulation, Adv.
References Exp. Med. Biol. 1173 (2019) 21–32.
[28] D.J. Haile, T.A. Rouault, J.B. Harford, M.C. Kennedy, G.A. Blondin, H. Beinert,
R.D. Klausner, Cellular regulation of the iron-responsive element binding protein:
[1] S.J. Dixon, K.M. Lemberg, M.R. Lamprecht, R. Skouta, E.M. Zaitsev, C.E. Gleason, disassembly of the cubane iron-sulfur cluster results in high-affinity RNA binding,
D.N. Patel, A.J. Bauer, A.M. Cantley, W.S. Yang, B. Morrison 3rd, B.R. Stockwell, Proc. Natl. Acad. Sci. U. S. A. 89 (1992) 11735–11739.
Ferroptosis: an iron-dependent form of nonapoptotic cell death, Cell 149 (2012) [29] A.R. Bogdan, M. Miyazawa, K. Hashimoto, Y. Tsuji, Regulators of iron homeostasis:
1060–1072. new players in metabolism, cell death, and disease, Trends Biochem. Sci. 41 (2016)
[2] S.J. Dixon, B.R. Stockwell, The role of iron and reactive oxygen species in cell death, 274–286.
Nat. Chem. Biol. 10 (2014) 9–17. [30] J.C. Reed, M. Pellecchia, Ironing out cell death mechanisms, Cell 149 (2012)
[3] S. Dolma, S.L. Lessnick, W.C. Hahn, B.R. Stockwell, Identification of genotype-se- 963–965.
lective antitumor agents using synthetic lethal chemical screening in engineered [31] K. Pantopoulos, Iron metabolism and the IRE/IRP regulatory system: an update,
human tumor cells, Canc. Cell 3 (2003) 285–296. Ann. N. Y. Acad. Sci. 1012 (2004) 1–13.
[4] W.S. Yang, B.R. Stockwell, Synthetic lethal screening identifies compounds acti- [32] M.H. Gao, P. Monian, Q.H. Pan, W. Zhang, J. Xiang, X.J. Jiang, Ferroptosis is an
vating iron-dependent, nonapoptotic cell death in oncogenic-RAS-harboring cancer autophagic cell death process, Cell Res. 26 (2016) 1021–1032.
cells, Chem. Biol. 15 (2008) 234–245. [33] J.D. Mancias, X.X. Wang, S.P. Gygi, J.W. Harper, A.C. Kimmelman, Quantitative
[5] Y. Xie, W. Hou, X. Song, Y. Yu, J. Huang, X. Sun, R. Kang, D. Tang, Ferroptosis: proteomics identifies NCOA4 as the cargo receptor mediating ferritinophagy,
process and function, Cell Death Differ. 23 (2016) 369–379. Nature 509 (2014) 105.
[6] J.Y. Cao, S.J. Dixon, Mechanisms of ferroptosis, Cell. Mol. Life Sci. 73 (2016) [34] W. Hou, Y. Xie, X. Song, X. Sun, M.T. Lotze, H.J. Zeh 3rd, R. Kang, D. Tang,
2195–2209. Autophagy promotes ferroptosis by degradation of ferritin, Autophagy 12 (2016)
[7] R.J. Bridges, N.R. Natale, S.A. Patel, System xc(-) cystine/glutamate antiporter: an 1425–1428.
update on molecular pharmacology and roles within the CNS, Br. J. Pharmacol. 165 [35] W.E. Dowdle, B. Nyfeler, J. Nagel, R.A. Elling, S. Liu, E. Triantafellow, S. Menon,
(2012) 20–34. Z. Wang, A. Honda, G. Pardee, J. Cantwell, C. Luu, I. Cornella-Taracido,
[8] W.S. Yang, R. SriRamaratnam, M.E. Welsch, K. Shimada, R. Skouta, E. Harrington, P. Fekkes, H. Lei, Q. Fang, M.E. Digan, D. Burdick, A.F. Powers,
V.S. Viswanathan, J.H. Cheah, P.A. Clemons, A.F. Shamji, C.B. Clish, L.M. Brown, S.B. Helliwell, S. D'Aquin, J. Bastien, H. Wang, D. Wiederschain, J. Kuerth,
A.W. Girotti, V.W. Cornish, S.L. Schreiber, B.R. Stockwell, Regulation of ferroptotic P. Bergman, D. Schwalb, J. Thomas, S. Ugwonali, F. Harbinski, J. Tallarico,
cancer cell death by GPX4, Cell 156 (2014) 317–331. C.J. Wilson, V.E. Myer, J.A. Porter, D.E. Bussiere, P.M. Finan, M.A. Labow, X. Mao,

134
Y. Su, et al. Cancer Letters 483 (2020) 127–136

L.G. Hamann, B.D. Manning, R.A. Valdez, T. Nicholson, M. Schirle, M.S. Knapp, T.F. Greten, P.R. Galle, J.F. Seitz, I. Borbath, D. Haussinger, T. Giannaris, M. Shan,
E.P. Keaney, L.O. Murphy, Selective VPS34 inhibitor blocks autophagy and un- M. Moscovici, D. Voliotis, J. Bruix, S.I.S. Group, Sorafenib in advanced hepatocel-
covers a role for NCOA4 in ferritin degradation and iron homeostasis in vivo, Nat. lular carcinoma, N. Engl. J. Med. 359 (2008) 378–390.
Cell Biol. 16 (2014) 1069–1079. [64] C. Louandre, Z. Ezzoukhry, C. Godin, J.C. Barbare, J.C. Maziere, B. Chauffert,
[36] B. Lu, X.B. Chen, M.D. Ying, Q.J. He, J. Cao, B. Yang, The role of ferroptosis in A. Galmiche, Iron-dependent cell death of hepatocellular carcinoma cells exposed to
cancer development and treatment response, Front. Pharmacol. 8 (2017) 992. sorafenib, Int. J. Canc. 133 (2013) 1732–1742.
[37] J. Guo, B. Xu, Q. Han, H. Zhou, Y. Xia, C. Gong, X. Dai, Z. Li, G. Wu, Ferroptosis: a [65] E. Lachaier, C. Louandre, C. Godin, Z. Saidak, M. Baert, M. Diouf, B. Chauffert,
novel anti-tumor action for cisplatin, Canc. Res. Treat. 50 (2018) 445–460. A. Galmiche, Sorafenib induces ferroptosis in human cancer cell lines originating
[38] S. Ma, E.E. Henson, Y. Chen, S.B. Gibson, Ferroptosis is induced following sir- from different solid tumors, Anticancer Res. 34 (2014) 6417–6422.
amesine and lapatinib treatment of breast cancer cells, Cell Death Dis. 7 (2016). [66] T. Suzuki, H. Motohashi, M. Yamamoto, Toward clinical application of the Keap1-
[39] V. Trujillo-Alonso, E.C. Pratt, H.L. Zong, A. Lara-Martinez, C. Kaittanis, M.O. Rabie, Nrf2 pathway, Trends Pharmacol. Sci. 34 (2013) 340–346.
V. Longo, M.W. Becker, G.J. Roboz, J. Grimm, M.L. Guzman, FDA-approved fer- [67] X.F. Sun, X.H. Niu, R.C. Chen, W.Y. He, D. Chen, R. Kang, D.L. Tang,
umoxytol displays anti-leukaemia efficacy against cells with low ferroportin levels, Metallothionein-1G facilitates sorafenib resistance through inhibition of ferroptosis,
Nat. Nanotechnol. 14 (2019) 616. Hepatology 64 (2016) 488–500.
[40] Y. Yamaguchi, T. Kasukabe, S. Kumakura, Piperlongumine rapidly induces the [68] J. Datta, S. Majumder, H. Kutay, T. Motiwala, W. Frankel, R. Costa, H.C. Cha,
death of human pancreatic cancer cells mainly through the induction of ferroptosis, O.A. MacDougald, S.T. Jacob, K. Ghoshal, Metallothionein expression is suppressed
Int. J. Oncol. 52 (2018) 1011–1022. in primary human hepatocellular carcinomas and is mediated through inactivation
[41] X. Sun, Z. Ou, R. Chen, X. Niu, D. Chen, R. Kang, D. Tang, Activation of the p62- of CCAAT/enhancer binding protein alpha by phosphatidylinositol 3-kinase sig-
Keap1-NRF2 pathway protects against ferroptosis in hepatocellular carcinoma cells, naling cascade, Canc. Res. 67 (2007) 2736–2746.
Hepatology 63 (2016) 173–184. [69] J. Nie, B. Lin, M. Zhou, L. Wu, T. Zheng, Role of ferroptosis in hepatocellular car-
[42] U. Klotz, K. Maier, C. Fischer, K. Heinkel, Therapeutic efficacy of sulfasalazine and cinoma, J. Canc. Res. Clin. Oncol. 144 (2018) 2329–2337.
its metabolites in patients with ulcerative-colitis and crohns-disease, N. Engl. J. [70] J. Zhou, L. Chan, S. Zhou, Trigonelline: a plant alkaloid with therapeutic potential
Med. 303 (1980) 1499–1502. for diabetes and central nervous system disease, Curr. Med. Chem. 19 (2012)
[43] C. Wahl, S. Liptay, G. Adler, R.M. Schmid, Sulfasalazine: a potent and specific in- 3523–3531.
hibitor of nuclear factor kappa B, J. Clin. Invest. 101 (1998) 1163–1174. [71] U. Boettler, K. Sommerfeld, N. Volz, G. Pahlke, N. Teller, V. Somoza, R. Lang,
[44] J.D. Lay, C.C. Hong, J.S. Huang, Y.Y. Yang, C.Y. Pao, C.H. Liu, Y.P. Lai, G.M. Lai, T. Hofmann, D. Marko, Coffee constituents as modulators of Nrf2 nuclear translo-
A.L. Cheng, I.J. Su, S.E. Chuang, Sulfasalazine suppresses drug resistance and in- cation and ARE (EpRE)-dependent gene expression, J. Nutr. Biochem. 22 (2011)
vasiveness of lung adenocarcinoma cells expressing AXL, Canc. Res. 67 (2007) 426–440.
3878–3887. [72] D. Shin, E.H. Kim, J. Lee, J.L. Roh, Nrf2 inhibition reverses resistance to GPX4
[45] V.S. Narang, G.M. Pauletti, P.W. Gout, D.J. Buckley, A.R. Buckley, Sulfasalazine- inhibitor-induced ferroptosis in head and neck cancer, Free Radic. Biol. Med. 129
induced reduction of glutathione levels in breast cancer cells: enhancement of (2018) 454–462.
growth-inhibitory activity of Doxorubicin, Chemotherapy 53 (2007) 210–217. [73] A. Dondorp, F. Nosten, K. Stepniewska, N. Day, N. White, g. South, East Asian
[46] J. Guan, M. Lo, P. Dockery, S. Mahon, C.M. Karp, A.R. Buckley, S. Lam, P.W. Gout, Quinine Artesunate Malaria Trial, Artesunate versus quinine for treatment of severe
Y.Z. Wang, The x (c) (-) cystine/glutamate antiporter as a potential therapeutic falciparum malaria: a randomised trial, Lancet 366 (2005) 717–725.
target for small-cell lung cancer: use of sulfasalazine, Canc. Chemother. Pharmacol. [74] N. Klonis, D.J. Creek, L. Tilley, Iron and heme metabolism in Plasmodium falci-
64 (2009) 463–472. parum and the mechanism of action of artemisinins, Curr. Opin. Microbiol. 16
[47] S. Okazaki, S. Shintani, Y. Hirata, K. Suina, T. Semba, J. Yamasaki, K. Umene, (2013) 722–727.
M. Ishikawa, H. Saya, O. Nagano, Synthetic lethality of the ALDH3A1 inhibitor [75] C. Roussel, E. Caumes, M. Thellier, P.A. Ndour, P.A. Buffet, S. Jaureguiberry,
dyclonine and xCT inhibitors in glutathione deficiency-resistant cancer cells, Artesunate to treat severe malaria in travellers: review of efficacy and safety and
Oncotarget 9 (2018) 33832–33843. practical implications, J. Trav. Med. 24 (2017).
[48] K. Sakitama, Y. Ozawa, N. Aoto, H. Tomita, M. Ishikawa, Effects of a new centrally [76] N. Eling, L. Reuter, J. Hazin, A. Hamacher-Brady, N.R. Brady, Identification of ar-
acting muscle relaxant, NK433 (lanperisone hydrochloride) on spinal reflexes, Eur. tesunate as a specific activator of ferroptosis in pancreatic cancer cells, Oncoscience
J. Pharmacol. 337 (1997) 175–187. 2 (2015) 517–532.
[49] A.T. Shaw, M.M. Winslow, M. Magendantz, C. Ouyang, J. Dowdle, A. Subramanian, [77] N.D. Yang, S.H. Tan, S. Ng, Y. Shi, J. Zhou, K.S. Tan, W.S. Wong, H.M. Shen,
T.A. Lewis, R.L. Maglathin, N. Tolliday, T. Jacks, Selective killing of K-ras mutant Artesunate induces cell death in human cancer cells via enhancing lysosomal
cancer cells by small molecule inducers of oxidative stress, Proc. Natl. Acad. Sci. U. function and lysosomal degradation of ferritin, J. Biol. Chem. 289 (2014)
S. A. 108 (2011) 8773–8778. 33425–33441.
[50] T.E. Massey, R.M. Walker, T.F. Mcelligott, W.J. Racz, Acetaminophen-induced hy- [78] T. Kurz, B. Gustafsson, U.T. Brunk, Intralysosomal iron chelation protects against
pothermia in mice - evidence for a central action of the parent compound, oxidative stress-induced cellular damage, FEBS J. 273 (2006) 3106–3117.
Toxicology 25 (1982) 187–200. [79] J.L. Roh, E.H. Kim, H. Jang, D. Shin, Nrf2 inhibition reverses the resistance of
[51] M.S. Agier, N. Chollet, E. Bediou, M. Boyer Gervoise, A.P. Jonville-Bera, Transient cisplatin-resistant head and neck cancer cells to artesunate-induced ferroptosis,
major hypothermia associated with acetaminophen: a pediatric case report and Redox Biol. 11 (2017) 254–262.
literature review, Arch. Pediatr. (2019). [80] Z. Kong, R. Liu, Y. Cheng, Artesunate alleviates liver fibrosis by regulating ferrop-
[52] T. Lorincz, K. Jemnitz, T. Kardon, J. Mandl, A. Szarka, Ferroptosis is involved in tosis signaling pathway, Biomed. Pharmacother. 109 (2019) 2043–2053.
acetaminophen induced cell death, Pathol. Oncol. Res. 21 (2015) 1115–1121. [81] E. Ooko, M.E. Saeed, O. Kadioglu, S. Sarvi, M. Colak, K. Elmasaoudi, R. Janah,
[53] A.L. Roe, J.E. Snawder, R.W. Benson, D.W. Roberts, D.A. Casciano, HepG2 cells: an H.J. Greten, T. Efferth, Artemisinin derivatives induce iron-dependent cell death
in vitro model for P450-dependent metabolism of acetaminophen, Biochem. (ferroptosis) in tumor cells, Phytomedicine 22 (2015) 1045–1054.
Biophys. Res. Commun. 190 (1993) 15–19. [82] C. Heading, Siramesine H. Lundbeck, Curr. Opin. Invest. Drugs 2 (2001) 266–270.
[54] J.R. Mitchell, D.J. Jollow, W.Z. Potter, J.R. Gillette, B.B. Brodie, Acetaminophen- [83] M.S. Ostenfeld, N. Fehrenbacher, M. Hoyer-Hansen, C. Thomsen, T. Farkas,
induced hepatic necrosis. IV. Protective role of glutathione, J. Pharmacol. Exp. M. Jaattela, Effective tumor cell death by sigma-2 receptor ligand siramesine in-
Therapeut. 187 (1973) 211–217. volves lysosomal leakage and oxidative stress, Canc. Res. 65 (2005) 8975–8983.
[55] C.D. Williams, A. Farhood, H. Jaeschke, Role of caspase-1 and interleukin-1beta in [84] E.R. Wood, A.T. Truesdale, O.B. McDonald, D. Yuan, A. Hassell, S.H. Dickerson,
acetaminophen-induced hepatic inflammation and liver injury, Toxicol. Appl. B. Ellis, C. Pennisi, E. Horne, K. Lackey, K.J. Alligood, D.W. Rusnak, T.M. Gilmer,
Pharmacol. 247 (2010) 169–178. L. Shewchuk, A unique structure for epidermal growth factor receptor bound to
[56] A. Ramachandran, H. Jaeschke, Mechanisms of acetaminophen hepatotoxicity and GW572016 (Lapatinib): relationships among protein conformation, inhibitor off-
their translation to the human pathophysiology, J. Clin. Trans. Res. 3 (2017) rate, and receptor activity in tumor cells, Canc. Res. 64 (2004) 6652–6659.
157–169. [85] D.W. Rusnak, K. Lackey, K. Affleck, E.R. Wood, K.J. Alligood, N. Rhodes, B.R. Keith,
[57] D. Wang, S.J. Lippard, Cellular processing of platinum anticancer drugs, Nat. Rev. D.M. Murray, W.B. Knight, R.J. Mullin, T.M. Gilmer, The effects of the novel, re-
Drug Discov. 4 (2005) 307–320. versible epidermal growth factor receptor/ErbB-2 tyrosine kinase inhibitor,
[58] D.B. Zamble, S.J. Lippard, Cisplatin and DNA repair in cancer chemotherapy, GW2016, on the growth of human normal and tumor-derived cell lines in vitro and
Trends Biochem. Sci. 20 (1995) 435–439. in vivo, Mol. Canc. Therapeut. 1 (2001) 85–94.
[59] L. Amable, Cisplatin resistance and opportunities for precision medicine, [86] D. Wetterskog, K.K. Shiu, I. Chong, T. Meijer, A. Mackay, M. Lambros,
Pharmacol. Res. 106 (2016) 27–36. D. Cunningham, J.S. Reis-Filho, C.J. Lord, A. Ashworth, Identification of novel
[60] Y. Min, C.Q. Mao, S. Chen, G. Ma, J. Wang, Y. Liu, Combating the drug resistance of determinants of resistance to lapatinib in ERBB2-amplified cancers, Oncogene 33
cisplatin using a platinum prodrug based delivery system, Angew. Chem. 51 (2012) (2014) 966–976.
6742–6747. [87] M.H. Rosner, M. Auerbach, Ferumoxytol for the treatment of iron deficiency, Expet
[61] M. Sato, R. Kusumi, S. Hamashima, S. Kobayashi, S. Sasaki, Y. Komiyama, Rev. Hematol. 4 (2011) 399–406.
T. Izumikawa, M. Conrad, S. Bannai, H. Sato, The ferroptosis inducer erastin irre- [88] B.S. Spinowitz, A.T. Kausz, J. Baptista, S.D. Noble, R. Sothinathan, M.V. Bernardo,
versibly inhibits system xc- and synergizes with cisplatin to increase cisplatin's L. Brenner, B.J. Pereira, Ferumoxytol for treating iron deficiency anemia in CKD, J.
cytotoxicity in cancer cells, Sci. Rep. 8 (2018) 968. Am. Soc. Nephrol. : JASN (J. Am. Soc. Nephrol.) 19 (2008) 1599–1605.
[62] S.M. Wilhelm, L. Adnane, P. Newell, A. Villanueva, J.M. Llovet, M. Lynch, [89] Y. Shao, W. Luo, H. Xu, L. Zhang, Q. Guo, The efficacy of ferumoxytol for iron
Preclinical overview of sorafenib, a multikinase inhibitor that targets both Raf and deficiency anemia: a meta-analysis of randomized controlled trials, Acta Haematol.
VEGF and PDGF receptor tyrosine kinase signaling, Mol. Canc. Therapeut. 7 (2008) (2019) 1–7.
3129–3140. [90] B.S. Spinowitz, M.H. Schwenk, P.M. Jacobs, W.K. Bolton, M.R. Kaplan, C. Charytan,
[63] J.M. Llovet, S. Ricci, V. Mazzaferro, P. Hilgard, E. Gane, J.F. Blanc, A.C. de Oliveira, M. Galler, The safety and efficacy of ferumoxytol therapy in anemic chronic kidney
A. Santoro, J.L. Raoul, A. Forner, M. Schwartz, C. Porta, S. Zeuzem, L. Bolondi, disease patients, Kidney Int. 68 (2005) 1801–1807.

135
Y. Su, et al. Cancer Letters 483 (2020) 127–136

[91] S.V. Torti, F.M. Torti, Winning the war with iron, Nat. Nanotechnol. 14 (2019) [96] T.T. Mai, A. Hamai, A. Hienzsch, T. Caneque, S. Muller, J. Wicinski, O. Cabaud,
499–500. C. Leroy, A. David, V. Acevedo, A. Ryo, C. Ginestier, D. Birnbaum, E. Charafe-
[92] E. Birben, U.M. Sahiner, C. Sackesen, S. Erzurum, O. Kalayci, Oxidative stress and Jauffret, P. Codogno, M. Mehrpour, R. Rodriguez, Salinomycin kills cancer stem
antioxidant defense, World Allergy Organ. J. 5 (2012) 9–19. cells by sequestering iron in lysosomes, Nat. Chem. 9 (2017) 1025–1033.
[93] Y. Miyazaki, M. Shibuya, H. Sugawara, O. Kawaguchi, C. Hirsoe, Salinomycin, a [97] B. Zhao, X. Li, Y. Wang, P. Shang, Iron-dependent cell death as executioner of
new polyether antibiotic, J. Antibiot. (Tokyo) 27 (1974) 814–821. cancer stem cells, J. Exp. Clin. Canc. Res. 37 (2018).
[94] S. Zhou, F.F. Wang, E.T. Wong, E. Fonkem, T.C. Hsieh, J.M. Wu, E.X. Wu, [98] X.X. Song, S. Zhu, P. Chen, W. Hou, Q.R. Wen, J. Liu, Y.C. Xie, J.B. Liu,
Salinomycin: a novel anti-cancer agent with known anti-coccidial activities, Curr. D.J. Klionsky, G. Kroemer, M.T. Lotze, H.J. Zeh, R. Kang, D.L. Tang, AMPK-medi-
Med. Chem. 20 (2013) 4095–4101. ated BECN1 phosphorylation promotes ferroptosis by directly blocking system X-c
[95] A. Hamai, T. Caneque, S. Muller, T.T. Mai, A. Hienzsch, C. Ginestier, E. Charafe- (-) activity, Curr. Biol. 28 (2018) 2388-+.
Jauffret, P. Codogno, M. Mehrpour, R. Rodriguez, An iron hand over cancer stem [99] R. Kang, S. Zhu, H.J. Zeh, D.J. Klionsky, D.L. Tang, BECN1 is a new driver of fer-
cells, Autophagy 13 (2017) 1465–1466. roptosis, Autophagy 14 (2018) 2173–2175.

136

You might also like