You are on page 1of 9

The official journal of

INTERNATIONAL FEDERATION OF PIGMENT CELL SOCIETIES · SOCIETY FOR MELANOMA RESEARCH

PIGMENT CELL & MELANOMA


Research
The role of ferroptosis in melanoma

Ronan Talty | Marcus Bosenberg

DOI: 10.1111/pcmr.13009
Volume 35, Issue 1, Pages 18–25

If you wish to order reprints of this article,


please see the guidelines here

EMAIL ALERTS
Receive free email alerts and stay up-to-date on what is published
in Pigment Cell & Melanoma Research – click here

Submit your next paper to PCMR online at http://mc.manuscriptcentral.com/pcmr

Subscribe to PCMR and stay up-to-date with the only journal committed to publishing
basic research in melanoma and pigment cell biology

As a member of the IFPCS or the SMR you automatically get online access to PCMR. Sign up as
a member today at www.ifpcs.org or at www.societymelanomaresarch.org

To take out a personal subscription, please click here


More information about Pigment Cell & Melanoma Research at www.pigment.org
| |
Received: 2 June 2021    Revised: 2 August 2021    Accepted: 12 August 2021

DOI: 10.1111/pcmr.13009

REVIEW

The role of ferroptosis in melanoma

Ronan Talty1  | Marcus Bosenberg1,2,3

1
Department of Pathology, Yale School of
Medicine, New Haven, CT, USA Abstract
2
Department of Dermatology, Yale School of Melanoma is the deadliest form of skin cancer. Although treatment with targeted
Medicine, New Haven, CT, USA
therapies and immune checkpoint inhibitors has dramatically improved survival in ad-
3
Department of Immunobiology, Yale School
of Medicine, New Haven, CT, USA
vanced melanoma, many patients do not benefit from these therapies or relapse after
an initial period of response. Thus, future outcomes in these categories of melanoma
Correspondence
Dr. Marcus Bosenberg, Department of
patients will depend on the identification of novel therapeutic targets and methods
Dermatology, Yale School of Medicine, P.O. to enhance existing targeted therapy and immunotherapy regimens. Ferroptosis is a
Box 208023, New Haven, CT 06520-­8 023,
USA.
newly identified form of iron-­dependent regulated cell death that is morphologically,
Email: Marcus.bosenberg@yale.edu biochemically, and genetically distinct from apoptosis, autophagy, pyroptosis, and

Funding information
necroptosis. Dysregulation of ferroptosis has been linked to the development of sev-
National Cancer Institute, Grant/Award eral forms of cancer. This review examines ferroptosis in the context of melanoma. It
Number: F30CA254246- ­01A1
presents an overview of ferroptosis biology, summarizes and interprets the current
literature, and poses several outstanding questions and areas of future direction.

KEYWORDS

cell death, ferroptosis, immunotherapy, iron, melanoma, skin neoplasms

1 | I NTRO D U C TI O N processes including lung fibrosis, ischemia–­


reperfusion injury,
stroke, traumatic brain injury, Alzheimer's disease, and Huntington's
Melanoma is the deadliest form of skin cancer, and it causes an esti- disease. In addition, ineffective ferroptosis is associated with tum-
mated 9,000 deaths in the United States annually (Siegel et al., 2019). origenesis, and dysregulation of ferroptosis has been linked to the
Until recently, once distant metastasis occurred, median survival and development of breast, cervical, colorectal, gastric, hepatocellu-
three-­year overall survival dropped to 8 months and less than 10%, lar, lung, ovarian, prostate, and renal carcinomas, lymphoma, and
respectively (Wilson et al., 2019). Now, treatment with dual BRAF melanoma (Lu et al., 2017). Comprehensive reviews of ferroptosis
and MEK-­
t argeted therapies and α-­PD-­1 and α-­C TLA-­4 immune have been published recently (Friedmann Angeli et al., 2019; Jiang
checkpoint inhibitors dramatically improves survival in these cases et al., 2021; Stockwell & Jiang, 2020). This review examines current
(Robert et al., 2019; Schachter et al., 2017). Responses to these data that support roles for ferroptosis in melanoma pathogenesis,
treatments, however, remain variable. Some patients do not benefit metastasis, and treatment.
at all, and others relapse after an initial period of response. Thus, fu-
ture outcomes in these categories of melanoma patients will depend
on the identification of novel therapeutic targets and methods to 2 | FE R RO P TOS I S OV E RV I E W
enhance existing targeted and immunotherapy regimens.
Ferroptosis is a newly identified form of iron-­dependent regu- The first ferroptosis-­inducing compounds, eradicator of RAS-­and
lated cell death that is morphologically, biochemically, and geneti- ST-­
expressing cells (erastin) and Ras Selective Lethal 3 (RSL3),
cally distinct from apoptosis, autophagy, pyroptosis, and necroptosis were identified prior to the first description of ferroptosis itself in
(Dixon et al., 2012). It is implicated in a growing list of disease high-­
throughput screens for small molecules that effectively kill

© 2021 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd

|
18     
wileyonlinelibrary.com/journal/pcmr Pigment Cell Melanoma Res. 2022;35:18–25.
TALTY and BOSENBERG |
      19

tumorigenic cells (Dolma et al., 2003; Yang & Stockwell, 2008). for the synthesis of reduced glutathione, which is used by GPX4 to
During their subsequent characterization, both erastin and RSL3 reduce lipid (hydro)peroxides. Inhibition of this antiporter by com-
were noted to increase cellular reactive oxygen species (ROS) and pounds like erastin, sulfasalazine, or sorafenib leads to decreased
elicit specific morphological responses like mitochondrial shrink- synthesis of reduced glutathione for use by GPX4, while com-
age in the absence of any features of other forms of cell death pounds like RSL3 and FIN56 inhibit GPX4 directly. In either case,
such as chromatin condensation, cytoplasmic swelling, or plasma lipid peroxides accumulate and trigger cell death by ferroptosis.
membrane rupture. Also, selective modifiers of other forms of cell The generation of lipid peroxides can occur through enzymatic and
death including inhibitors of caspases, calpains and cathepsins, cy- nonenzymatic pathways. Enzymatic lipid peroxidation is performed
clophilin D, RIPK1, and autophagy had no effect on the mode of mainly by lipoxygenases (LOXs) and NADPH oxidases (NOXs) that
cell death induced by these compounds (Dolma et al., 2003; Yang & often depend on iron as a cofactor, whereas nonenzymatic lipid
Stockwell, 2008). On the contrary, a distinct set of small molecules peroxidation occurs through an autoxidation reaction that uses free
consisting of deferoxamine (an iron chelator), trolox (a Vitamin E iron as a substrate for Fenton-­like chemistry (Conrad & Pratt, 2019).
analog), ebselen (a glutathione peroxidase mimetic), and U0126 (a Consequently, processes that increase the cellular availability of
MEK inhibitor) protected cells from erastin and RSL3-­induced cell free iron, such as ferritinophagy mediated by NCOA4, can promote
death. Two novel small molecules, named ferrostatin-­1 and liprox- ferroptosis, and iron chelators like deferoxamine (DFO), which re-
statin-­1, also inhibited this form of cell death with extremely high duce free iron, inhibit ferroptosis. Ferrostatin-­1, liproxstatin-­1, and
selectivity (Dixon et al., 2012; Friedmann Angeli et al., 2014). vitamin E act as radical-­trapping agents (RTAs) to inhibit the gener-
It is now known that ferroptosis occurs through three major ation of lipid peroxides. A broader list of ferroptosis inducers and
steps: reduced activity of the cysteine-­
glutamate antiporter inhibitors is presented in Table 1.
(System Xc-­), inhibition of glutathione peroxidase 4 (GPX4), and iron-­ Several other pathways are now recognized to promote ferro-
dependent generation of lipid peroxides. The relationship between ptosis resistance. First, ferroptosis suppressor protein 1 (FSP1), re-
these key processes is illustrated in Figure 1. Briefly, System Xc-­ is a named from apoptosis-­inducing mitochondria associated 2 (AIFM2),
cell surface antiporter comprised of SLC7A11 and SLC3A2 that im- conferred protection against ferroptosis in both a CRISPR-­
C as9
ports extracellular cystine in exchange for intracellular glutamate. loss of function screen with RSL3 treatment and a cDNA overex-
Under normal conditions, both of these amino acids are necessary pression screen in GPX4-­deficient cells (Bersuker et al., 2019; Doll

F I G U R E 1   An overview of ferroptosis, which is triggered by the toxic accumulation of lipid peroxides. These are generated through
a combination of iron-­mediated Fenton chemistry and the enzymes ALOX, NOX, and LPCAT. System Xc imports cystine into the cell to
support the synthesis of reduced glutathione (GSH). GSH is used by GPX4 to reduce lipid peroxides and prevent their accumulation. The
NRF2, hippo, mevalonate synthesis, and transsulfuration pathways are secondary pathways that can supplement the action of GPX4.
Ferroptosis inducers (red) and inhibitors (green) manipulate various points of these pathways. AA—­arachidonic acid; ACSL4—­acyl-­CoA
synthetase long-­chain family member 4; ALOX—­arachidonate lipoxygenase; Cys—­cysteine; CoQ—­coenzyme Q; FPN—­ferroportin; FSP1—­
ferroptosis suppressor 1; GCL—­glutamate cysteine ligase; Glu—­glutamate; GPX4—­glutathione peroxidase 4; GS—­glutamine synthetase;
GSH—­reduced glutathione; GS-­SG—­oxidized glutathione; LPCAT—­lysophosphatidylcholine acyl transferase; NCOA4—­nuclear receptor
coactivator 4; NOX—­NADPH oxidase; NRF2—­nuclear factor erythroid-­2-­related factor 2; PROM2—­prominin 2; PUFA—­polyunsaturated fatty
acid; RSL3—­R AS selective lethal 3; SLC3A2—­solute carrier family 3 member 2; SLC7A11—­solute carrier family 7 member 11; TAZ—­t afazzin;
TF—­transferrin; YAP—­yes-­associated protein
|
20       TALTY and BOSENBERG

TA B L E 1   A summary of common
Mechanism Compounds
ferroptosis inducers and inhibitors
Ferroptosis inducers organized by their mechanisms
FSP1-­CoQ10 -­NAD(P)H axis iFSP1
GPX4 inhibition Altretamine, DPIs, FIN56, JKE−1674, ML162, ML210, RSL3
GSH depletion Buthionine sulfoximine (BSO)
Iron overload Lapatinib, Salinomycin, Siramesine
Iron oxidation FINO2
System Xc-­inhibition Cyst(e)inase, DPI2, Erastin, Imidazole ketone erastin, Piperazine
erastin, Sorafenib, Sulfasalazine
Ferroptosis Inhibitors
Iron chelation Ciclopirox, Deferoxamine, Deferiprone
MEK inhibition U0126
Radical trapping a-­tocopherol, Ferrostatin-­1, Liproxstatin-­1

et al., 2019). FSP1 catalyzes the regeneration of CoQ10, which then acquired resistance sub-­
lines, established from patient biopsies.
traps lipid peroxyl radicals. As another example, the nuclear factor The cell lines clustered into four distinct classes according to their
erythroid-­2-­related factor 2 (NRF2) transcription factor is a master differentiation status: (1) melanocytic, (2) transitory, 3) neural crest
regulator of the antioxidant response. Many proteins responsible like, and 4) undifferentiated. Their analysis of these clusters revealed
for regulating iron metabolism (e.g., FTH, FTL, and SLC40A1) and consistent, specific dedifferentiation sequences that occur in re-
preventing lipid peroxidation (e.g., SLC7A11, GPX4, and FSP1) have sponse to BRAF inhibition, MAPK inhibition, and immunotherapy and
been identified as NRF2 target genes (Dodson et al., 2019). Other confer resistance. They paired their clusters with publicly available
pathways that regulate aspects of ferroptosis include hippo, meva- data from the Cancer Therapeutics Response Portal (CTRP), filtered
lonate synthesis, transsulfuration, and prominin2-­
exosome path- for drugs that exhibited cluster-­
specific sensitivity, and demon-
ways (Brown et al., 2019; Hayano et al., 2016; Warner et al., 2000; strated that dedifferentiation status positively correlated with sensi-
Wu et al., 2019). tivity to the ferroptosis inducers erastin, RSL3, ML162, and ML210.
After confirming these findings in their own cell lines, they tested
the combined effect of BRAF inhibitors and ferroptosis inducers in
3 |  M E L A N O M A D E D I FFE R E NTI ATI O N vitro and showed a dramatic decrease in the number of cells persist-
S TAT U S CO R R E L ATE S W ITH FE R RO P TOS I S ing after treatment. This effect, they reasoned, likely occurs because
S E N S ITI V IT Y each agent optimally targets melanoma cells at a distinct differentia-
tion status. Finally, they showed that the proinflammatory cytokines
Cellular dedifferentiation is a hallmark of cancer initiation, progres- TNF-­α and IFN-­γ induce dedifferentiation and potentiate lethality
sion, and metastasis, and it can also drive targeted therapy resist- upon treatment with ferroptosis inducers. These data suggest that
ance (Gupta et al., 2019). Melanoma cells, in particular, maintain a ferroptosis inducers could potentially promote antitumor immunity
highly plastic phenotype, possibly due to their neural crest origin, by killing dedifferentiated melanoma cells and preventing their im-
a transient, stem-­cell-­like embryonic cell population that differenti- munosuppressive actions. What remains unknown, however, is how
ates into diverse tissue types ranging from neurons and glia to en- dedifferentiation leads to increased ferroptosis sensitivity. It would
docrine cells and cartilage (Sauka-­Spengler & Bronner-­Fraser, 2008). be relevant to examine in subsequent experiments whether TNF-­α
In clinical practice, dedifferentiation allows melanomas to develop and IFN-­γ elicit downregulation of System Xc-­ or glutathione synthe-
resistance to both BRAF and MAPK inhibition associated with down- sis enzymes or whether an unrelated mechanism alters ferroptosis
regulation of MITF, the master regulator of melanocyte differentia- sensitivity.
tion (Hugo et al., 2015; Müller et al., 2014). Furthermore, decreased
MITF expression has been shown to enhance recruitment of immu-
nosuppressive myeloid immune cells into the tumor microenviron- 4 | TH E R A PY- ­R E S I S TA NT M E L A N O M A
ment to facilitate immune escape and tumor growth (Riesenberg C E LL S ACQ U I R E A G PX4 - ­D E PE N D E NT
et al., 2015). PE R S I S TE R S TATE
In 2018, Tsoi et al., reasoned that dedifferentiation might con-
fer susceptibility to novel therapies despite promoting resistance Efforts to understand mechanisms of therapy resistance by cancer
to many standard interventions. To examine this, they performed cells have widely focused on genetic mutations that confer drug
hierarchical clustering and principal component analysis of expres- resistance such as altered binding sites, compensatory signaling
sion profiles from 53 human melanoma cells lines, including paired pathways, and upregulation of efflux pumps. Emerging evidence,
TALTY and BOSENBERG |
      21

however, underscores the importance of non-­mutational mecha- prevented with iron chelators and other ferroptosis inhibitors.
nisms of resistance in cancer cells (Boumahdi & de Sauvage, 2020). Glutaminolysis and glutamine metabolism have also been implicated
Such changes in cell plasticity can enable them to either wholly resist in ferroptosis regulation in other ways. For example, the glutamate
initial treatment or maintain a surviving “persister” cell population importer (SLC1A5/SLC28A1), glutaminase (GLS), and glutamic-­
that can drive disease recurrence. Specific examples of this plasticity oxaloacetic transaminase-­1 (GOT1) collectively facilitate glutamine
include epithelial–­mesenchymal transition, neuroendocrine trans- uptake and its metabolism to glutamate and α-­ketoglutarate (α-­KG),
differentiation, and dedifferentiation-­based phenotypic discussed and inactivation of any of these genes protects cells from ferropto-
previously (Tsoi et al., 2018b). Cell identity conversions, chromatin sis induction (Gao et al., 2015). Additionally, the mutant Huntington
remodeling and epigenetic transformation, microenvironment con- (HTT) allele found in Huntington's disease has been linked to
tributions, and the existence of slow-­cycling cells are all proposed glutamate-­induced cytotoxicity, glutathione deficiency, disrupted
mechanisms for how these plasticity changes might occur in the set- iron metabolism, and lipid peroxidation (Johnson et al., 2012). This
ting of cancer treatment. link has prompted an extensive study of ferroptosis in the con-
Two studies implicated the central ferroptosis gene, GPX4, as text of Huntington's disease and other neurodegenerative condi-
a potential driver of cell plasticity. The first explored why high-­ tions like Alzheimer's disease and Parkinson's disease (Masaldan
mesenchymal cancer cell states confer resistance to multiple et al., 2019).
treatment modalities across diverse cancer lineages (Viswanathan MicroRNAs (miRNAs) have emerged as critical regulators of
et al., 2017). The group used publicly available gene expression diverse biological processes such as cell death, differentiation,
data sets to evaluate shared characteristics between multiple proliferation, and metabolism, and two studies also demonstrate
high-­
m esenchymal cancer cell lines. They demonstrated that that miRNAs can regulate ferroptosis by targeting glutamate me-
these cell lines exhibit increased synthesis of polyunsaturated tabolism. Luo et al., (2018) performed an unbiased screen in A375
lipids which serve as substrates for lipoxygenase enzymes, and and G-­361 melanoma cells transfected with miRNA overexpressing
this change in lipid production forces a dependence on the en- constructs and treated with erastin and RSL3 to discover miRNAs
zyme GPX4 to prevent the accumulation of lipid peroxides and that regulate ferroptosis. Overexpression of miR-­137 inhibited lipid
ferroptosis execution. This specifically occurred in BRAF-­m utant peroxidation, reduced iron accumulation, and rescued cells from
melanoma treated with TGFβ to upregulate AXL, downregulate ferroptosis. miR-­137 accomplishes this by suppressing expression
MITF, and induce targeted therapy and immunotherapy resis- of the SLC1A5 glutamine transporter component, and overexpres-
tance. Importantly, loss of GPX4 induced cell death in vitro in sion of SLC1A5 negates the ferroptosis protective effect of miR-­137.
these therapy-­resistant cells via ferroptosis and greatly impaired Importantly, miR-­137 overexpression in melanoma cells produced
tumor growth in vivo. The second study determined that a similar faster tumor growth, larger tumor volumes, and resistance to eras-
dependence on GPX4 for survival occurs in residual populations of tin in vivo. miR-­9 was also identified in this screen and similarly in-
cancer cells that persist after a broad range of drug treatments, in- hibited lipid peroxidation, reduced iron accumulation, and rescued
cluding A375 melanoma cells treated with vemurafenib (Hangauer cells from ferroptosis (Zhang et al., 2018). miR-­9, however, exerts no
et al., 2017). Once again, they showed that loss of GPX4 induces effect on SLC1A5. Instead, it negatively regulates GOT1 expression,
death of persister cells via ferroptosis in vitro and prevents tumor and overexpression of GOT1 negates the ferroptosis protective ef-
relapse after treatment in vivo. These studies implicate the ferro- fect of miR-­9. These studies confirm a role for miRNAs in ferroptosis
ptosis pathway as a molecular driver of cell plasticity and treat- regulation, implicate glutamine metabolism in melanoma progres-
ment resistance in the context of melanoma. Notably, it remains sion, and identify two potential therapeutic targets that can regu-
unknown whether immune checkpoint inhibitors also produce a late ferroptosis.
residual population of GPX4-­d ependent persister cells. Such in-
formation could help inform the use of ferroptosis modulators in
combination with current immunotherapy regimens for patients 6 | S E N S ITI V IT Y TO FE R RO P TOS I S
with metastatic melanoma. I N FLU E N C E S TH E M E TA S TATI C P OTE NTI A L
O F M E L A N O M A C E LL S

5 |  M I C RO R N A S R EG U L ATE FE R RO P TOS I S The role of oxidative stress in suppressing melanoma metastasis
I N M E L A N O M A BY TA RG E TI N G G LU TA M ATE is well documented, and the lymphatic system is the route of me-
M E TA B O LI S M tastasis for most melanoma cells (Gal et al., 2015; Lee et al., 2019;
Piskounova et al., 2015). Ubellacker et al., (2020) recently investi-
Long before the first description of ferroptosis, it had been noted gated this process and found that lymph specifically protects me-
that neurons treated with excess glutamate reduce their cystine tastasizing cells from ferroptosis. They obtained sets of efficiently
import, become glutathione deficient, accumulate peroxides, and metastasizing and inefficiently metastasizing patient melanoma
undergo cell death (Murphy et al., 1989). Glutamate-­induced cy- samples as well as YUMM1.7 and YUMM3.3 syngeneic mouse mela-
totoxicity is now suspected to occur via ferroptosis and can be noma models and tested their ability to form tumors when injected
|
22       TALTY and BOSENBERG

into mice under different conditions. For all cell lines, the group no- synthesis of reduced glutathione, and promotes lipid peroxidation
ticed that fewer cells were necessary to form tumors when injected and ferroptosis. They also evaluated whether expression of System
into lymph nodes than when injected intravenously. Melanoma cells Xc-­ subunits in human melanoma tissues correlates with immuno-
in the blood had greater ROS, lower ratios of glutathione to oxidized therapy outcomes. Reduced expression of SLC3A2 correlated with a
glutathione, and increased ferroptosis. Furthermore, pretreatment strong CD8+ T cell signature, IFN-­γ expression, and improved over-
of cells with ferroptosis inhibitors increased intravenous metastasis all patient survival.
but not intranodal metastasis. Loss of GPX4, on the other hand, re- More recently, Jiang, Lim, et al., (2021) showed that limiting tu-
duced the burden of tumors formed from intravenous injections but moral ferroptosis via upregulation of TYRO3 promotes resistance to
not intranodal or subcutaneous ones. Ultimately, the group identi- α-­PD-­1/PD-­L1 immune checkpoint inhibition. They performed an
fied oleic acid as a lymph component that directly protects cells from unbiased screen with receptor tyrosine kinase (RTK) antibodies that
ferroptosis. identified elevated TYRO3 expression in α-­PD-­1-­resistant cells. They
Sato et al. also interrogated the role of ferroptosis in melanoma then found in two publicly available gene expression databases,
pathogenesis and metastasis by generating System Xc-­-­deficient Tumor Immune Dysfunction and Exclusion (TIDE) and Prediction
B16F10 melanoma cells (Sato et al., 2020). Loss of System Xc-­ in of Clinical Outcomes from Genomics Profiles (PRECOG), that
these cells reduced cystine uptake, cellular glutathione, cell cycle higher TYRO3 expression correlated with shorter overall survival
progression, and proliferation in vitro, tumor spheroid formation in melanoma patients treated with α-­PD-­1 checkpoint inhibitors.
ex vivo, and subcutaneous tumor formation in vivo. Importantly, Overexpression of TYRO3 was sufficient to generate resistance to
none of these changes could be rescued by the ferroptosis inhib- α-­PD-­1 in mouse tumors, and knockout of TYRO3 in α-­PD-­1-­resistant
itor liproxstatin-­1. System Xc-­-­deficient cells also adhered poorly tumors conferred sensitivity to a-­PD-­1. In TYRO3-­overexpressing
to lung vascular endothelium in vitro, showed reduced migration cells, genes that block ferroptosis were upregulated (e.g., SLC40A1,
potential by in vitro scratch assays, and formed less metastases SLC7A11, SLC3A2, and GPX4), whereas genes that enhance ferro-
in vivo via tail vein, intrasplenic, IP, and footpad injections. These ptosis were downregulated (e.g., SLC5A1, TFRC). These cells were
studies collectively link ferroptosis susceptibility and metastatic also resistant to treatment with erastin in vitro, generated less lipid
potential in melanoma. Notably, neither study directly evaluated peroxides when treated with α-­PD-­1 in vivo, and reduced the M1/
the roles of immune cells on metastasis. New evidence suggests M2-­like macrophage ratio to create a protumor microenvironment.
this might be especially important in the context of lymph metas- Inhibition of TYRO3 promoted tumor ferroptosis and sensitized α-­
tases, and it will be important to investigate this in the future (Song PD-­1-­resistant tumors to α-­PD-­1 treatment. Figure 2. compares nor-
et al., 2020). mal CD8+ T cell–­tumor interaction after PD-­1 checkpoint blockade
with their interaction in the setting of TYRO3 overexpression by
tumor cells.
7 |   I M M U N E C H EC K P O I NT I N H I B ITI O N These reports highlight an exciting, new possibility for the im-
AC TI VATE S C D 8 + T C E LL S TO E N H A N C E provement of existing immunotherapy strategies by modulating
FE R RO P TOS I S I N T U M O R C E LL S ferroptosis. However, pathways that promote ferroptosis resistance
in tumors such as TYRO3 upregulation have already emerged, and
Inactivation of GPX4 in the brain and kidney led to the first experi- several unanswered questions remain. For example, what effect
mental hints that ferroptosis leads to a cellular immune response. does tumor cell ferroptosis have on immune cells other than CD8+
Neuron-­specific GPX4 loss induces strong immunoreactivity of T cells and how does ferroptosis shape the overall tumor microenvi-
brain tissue as measured by the degree of reactive gliosis (Seiler ronment? Jiang et al. showed that TYRO3-­overexpressing cells alter
et al., 2008). In the kidney, loss of GPX4 causes release of damage-­ macrophage polarization to favor immune suppression and tumor
associated molecular patterns (DAMPs), recruitment of mac- growth, but it remains unclear whether this also occurs with other
rophages, and massive tubular death and renal failure (Friedmann types of ferroptosis-­resistant cells. The relative sensitivities of spe-
Angeli et al., 2014). Given this, Wang et al., (2019) decided to in- cific immune cell populations in the tumor microenvironment to
vestigate a potential role for ferroptosis in antitumor immunity systemic ferroptosis inducers like RSL3 or imidazole ketone erastin
and cancer immunotherapy. First, they showed that treatment of are also unknown. Retrospective clinical studies that examine the
B16 melanoma xenografts with α-­PD-­1 immune checkpoint inhi- effects of iron status, thiazolidine use, and statin use on immuno-
bition elicits lipid peroxidation and ferroptosis in tumor cells but therapy outcomes in patients with metastatic melanoma could also
not tumor-­infiltrating lymphocytes. They then demonstrated, first prove insightful. Statins sensitize cells to ferroptosis through their
by treatment of an erastin-­resistant cell line and later by coad- modulation of mevalonate synthesis, whereas thiazolidines inhibit
ministration of the ferroptosis inhibitor liproxstatin-­1, that ferrop- the ferroptosis promoting enzyme ACSL4 and reduce lipid perox-
tosis is necessary for the antitumor efficacy of immunotherapy. ide production. If ferroptosis is a key component of the antitumor
Mechanistically, they determined that IFN-­γ secreted by CD8+ T action of immune checkpoint inhibition, it is possible that targeting
cells reduces the expression of the System Xc-­ subunits SLC3A2 and specific ferroptosis regulatory pathways could enhance therapeutic
SLC7A11. This impairs cystine uptake by tumor cells, reduces the responses.
TALTY and BOSENBERG |
      23

F I G U R E 2   CD8+ T cell–­tumor
interaction after PD-­1 checkpoint
blockade. (a) Under normal conditions,
CD8+ T cells release IFN-­γ which causes
tumor cells to downregulate SLC3A2
and SLC7A11, depletes their cysteine
stores, and triggers ferroptotic cell death.
(b) Overexpression of TYRO3 by tumor
cells causes them to upregulate SLC3A2,
SLC7A11, and GPX4 which confer
resistance to ferroptotic cell death and
checkpoint blockade

8 |  M U LTI PLE PATH WAYS C A N CO N FE R preclinical studies to directly examine the role of ferroptosis in
R E S I S TA N C E TO FE R RO P TOS I S I N models of melanoma metastasis.
MEL ANOMA

Given the therapeutic potential of ferroptosis regulators in 9 | CO N C LU S I O N A N D FU T U R E


melanoma, a better understanding of how resistance to these D I R EC TI O N S
compounds can develop will be critical for the development of ef-
fective clinical strategies. Two studies have explored resistance Although ferroptosis was only recently discovered as a distinct
to the ferroptosis inducer erastin. Although erastin is primarily mechanism of regulated cell death, accumulating evidence sug-
thought to induce ferroptosis by inhibiting System Xc-­, it also binds gests that regulation of ferroptosis plays multiple roles in melanoma
to the voltage-­dependent anion channels VDAC2 and VDAC3 to progression and therapeutic resistance. Multiple lines of evidence
alter the permeability of the outer mitochondrial membrane and support a role for ferroptotic pathways regulating the differentia-
decrease the rate of NADH oxidation. In the first study, the au- tion state of melanoma cells and resistance to particular therapeutic
thors observed a dramatic downregulation of VDAC2/3 in A375 agents. It is clear that ferroptosis inducers have strong therapeutic
melanoma cells just hours after treatment with erastin that con- potential in melanoma—­both as single agents to overcome resistance
tributes to erastin resistance (Yang et al., 2020). They then showed to conventional chemotherapy or targeted therapy and in combina-
that the E3 ligase NEDD4 recognizes, binds to, ubiquinates, and tion with targeted therapies or immune checkpoint inhibitors to aug-
degrades VDAC2/3 under such conditions. Furthermore, deple- ment antitumor immunity and increase the overall patient response
tion of NEDD4 by shRNA blocked erastin-­induced degradation of rates. The continued development of novel ferroptosis inducers
VDAC2/3 and increased the sensitivity of melanoma cells to fer- with improved pharmacokinetics such as engineered cyst(e)inase,
roptosis. A second study focused on resistance to erastin in four imidazole ketone erastin, and JKE-­1674 will further broaden thera-
human melanoma cell lines (A375, CHL-­1, C8161, and SK-­Mel-­5) peutic opportunities (Cramer et al., 2017; Eaton et al., 2020; Zhang
(Gagliardi et al., 2019). They showed that most melanoma cell lines et al., 2019). Several pathways of resistance to ferroptosis inducers
resist ferroptosis execution through the NRF2-­driven upregulation in melanoma, however, have already been identified and will be im-
of aldo–­keto reductases (AKRs), which catalyze the conversion of portant to keep in mind as clinical trials for ferroptosis inducers are
aldehydes and ketones to their corresponding alcohols to help re- designed. In the next several years, research on ferroptosis in mela-
duce lipid peroxide accumulation. The relevance of this pathway noma will undoubtedly continue to expand and will likely delineate
in vivo, however, is less clear as they found no associations be- the roles of additional potential regulators of ferroptosis, including
tween tumor NRF2 or AKR expression and survival in patients with iron metabolism, transsulfuration, hippo signaling, and mevalonate
metastatic melanoma. They did, however, identify SLC7A11 as a synthesis pathways. How current findings replicate across multiple
useful marker to predict the susceptibility of metastatic melanoma melanoma models and how well they translate to human patients
cells to ferroptosis. This specifically highlights the need for future with melanoma represent critical outstanding questions.
|
24       TALTY and BOSENBERG

AC K N OW L E D G E M E N T S Friedmann Angeli, J. P., Krysko, D. V., & Conrad, M. (2019). Ferroptosis at


the crossroads of cancer-­acquired drug resistance and immune eva-
This work was funded by grants to Marcus Bosenberg and Ronan
sion. Nature Reviews Cancer, 19.
Talty (NIH F30CA254246-­01A1) from the National Cancer Institute Friedmann Angeli, J. P., Schneider, M., Proneth, B., Tyurina, Y. Y.,
of the National Institute of Health. The content is solely the respon- Tyurin, V. A., Hammond, V. J., Herbach, N., Aichler, M., Walch, A.,
sibility of the authors and does not necessarily represent the offi- Eggenhofer, E., Basavarajappa, D., Rådmark, O., Kobayashi, S., Seibt,
T., Beck, H., Neff, F., Esposito, I., Wanke, R., Förster, H. … Conrad, M.
cial views of funding agencies. The figures in this work were created
(2014). Inactivation of the ferroptosis regulator Gpx4 triggers acute
with BioRender. renal failure in mice. Nature Cell Biology, 16, 1180–­1191.
Gagliardi, M., Cotella, D., Santoro, C., Corà, D., Barlev, N. A., Piacentini,
C O N FL I C T S O F I N T E R E S T M., & Corazzari, M. (2019). Aldo-­keto reductases protect metastatic
The authors declare no conflicts of interest. melanoma from ER stress-­ independent ferroptosis. Cell Death &
Disease, 10, 902. https://doi.org/10.1038/s4141​9-­019-­2143-­7
Gal, K. L., Ibrahim, M. X., Wiel, C., Sayin, V. I., Akula, M. K., Karlsson,
ORCID C., Dalin, M. G., Akyürek, L. M., Lindahl, P., Nilsson, J. et al (2015).
Ronan Talty  https://orcid.org/0000-0002-8742-3799 Antioxidants can increase melanoma metastasis in mice. Science
Translational Medicine, 7, 308re8-­3 08re8.
Gao, M., Monian, P., Quadri, N., Ramasamy, R., & Jiang, X. (2015).
REFERENCES
Glutaminolysis and transferrin regulate ferroptosis. Molecular Cell,
Bersuker, K., Hendricks, J. M., Li, Z., Magtanong, L., Ford, B., Tang, P. 59, 298–­3 08. https://doi.org/10.1016/j.molcel.2015.06.011
H., Roberts, M. A., Tong, B., Maimone, T. J., Zoncu, R., Bassik, M. Gupta, P. B., Pastushenko, I., Skibinski, A., Blanpain, C., & Kuperwasser,
C., Nomura, D. K., Dixon, S. J., & Olzmann, J. A. (2019). The CoQ C. (2019). Phenotypic plasticity: Driver of cancer initiation, progres-
oxidoreductase FSP1 acts parallel to GPX4 to inhibit ferroptosis. sion, and therapy resistance. Cell Stem Cell, 24, 65–­78. https://doi.
Nature, 575, 688–­692. https://doi.org/10.1038/s4158​6 - ­019-­1705-­2 org/10.1016/j.stem.2018.11.011
Boumahdi, S., & de Sauvage, F. J. (2020). The great escape: Tumour cell Hangauer, M. J., Viswanathan, V. S., Ryan, M. J., Bole, D., Eaton, J. K.,
plasticity in resistance to targeted therapy. Nat. Rev. Drug Discov., 19, Matov, A., Galeas, J., Dhruv, H. D., Berens, M. E., Schreiber, S. L.,
39–­56. https://doi.org/10.1038/s4157​3- ­019- ­0 044-­1 McCormick, F., & McManus, M. T. (2017). Drug-­tolerant persister
Brown, C. W., Amante, J. J., Chhoy, P., Elaimy, A. L., Liu, H., Zhu, L. J., cancer cells are vulnerable to GPX4 inhibition. Nature, 551, 247–­250.
Baer, C. E., Dixon, S. J., & Mercurio, A. M. (2019). Prominin2 drives https://doi.org/10.1038/natur​e24297
ferroptosis resistance by stimulating iron export. Developmental Cell, Hayano, M., Yang, W. S., Corn, C. K., Pagano, N. C., & Stockwell, B. R.
51, 575–­586.e4. https://doi.org/10.1016/j.devcel.2019.10.007 (2016). Loss of cysteinyl-­tRNA synthetase (CARS) induces the trans-
Conrad, M., & Pratt, D. A. (2019). The chemical basis of ferroptosis. sulfuration pathway and inhibits ferroptosis induced by cystine
Nature Chemical Biology, 15, 1137–­ 1147. https://doi.org/10.1038/ deprivation. Cell Death and Differentiation, 23, 270–­278. https://doi.
s4158​9-­019-­0 408-­1 org/10.1038/cdd.2015.93
Cramer, S. L., Saha, A., Liu, J., Tadi, S., Tiziani, S., Yan, W., Triplett, K., Hugo, W., Shi, H., Sun, L., Piva, M., Song, C., Kong, X., Moriceau, G., Hong,
Lamb, C., Alters, S. E., Rowlinson, S., Zhang, Y. J., Keating, M. J., A., Dahlman, K. B., Johnson, D. B., Sosman, J. A., Ribas, A., & Lo, R.
Huang, P., DiGiovanni, J., Georgiou, G., & Stone, E. (2017). Systemic S. (2015). Non-­genomic and immune evolution of melanoma acquiring
depletion of L-­c yst(e)ine with cyst(e)inase increases reactive oxygen MAPKi resistance. Cell, 162, 1271–­ 1285. https://doi.org/10.1016/j.
species and suppresses tumor growth. Nature Medicine, 23, 120–­127. cell.2015.07.061
https://doi.org/10.1038/nm.4232 Jiang, X., Stockwell, B. R., & Conrad, M. (2021). Ferroptosis: Mechanisms,
Dixon, S. J., Lemberg, K. M., Lamprecht, M. R., Skouta, R., Zaitsev, E. biology and role in disease. Nature Reviews Molecular Cell Biology, 22,
M., Gleason, C. E., Patel, D. N., Bauer, A. J., Cantley, A. M., Yang, 266–­282. https://doi.org/10.1038/s4158​0 -­020-­0 0324​-­8
W. S., Morrison, B., & Stockwell, B. R. (2012). Ferroptosis: An iron-­ Jiang, Z., Lim, S. O., Yan, M., Hsu, J. L., Yao, J., Wei, Y., Chang, S. S.,
dependent form of nonapoptotic cell death. Cell, 149, 1060–­1072. Yamaguchi, H., Lee, H. H., Ke, B., Hsu, J.-­M., Chan, L.-­C ., Hortobagyi,
https://doi.org/10.1016/j.cell.2012.03.042 G. N., Yang, L., Lin, C., Yu, D., & Hung, M.-­C . (2021). TYRO3 induces
Dodson, M., Castro-­Portuguez, R., & Zhang, D. D. (2019). NRF2 plays anti–­PD-­ 1/PD-­L1 therapy resistance by limiting innate immunity
a critical role in mitigating lipid peroxidation and ferroptosis. Redox and tumoral ferroptosis. J Clin Invest, 131(8), e139434. https://doi.
Biology, 23. https://doi.org/10.1016/j.redox.2019.101107 org/10.1172/JCI13​9434
Doll, S., Freitas, F. P., Shah, R., Aldrovandi, M., da Silva, M. C., Ingold, Johnson, W. M., Wilson-­ Delfosse, A. L., & Mieyal, J. J. (2012).
I., Grocin, A. G., Xavier da Silva, T. N., Panzilius, E., Scheel, C. H., Dysregulation of glutathione homeostasis in neurodegenerative dis-
Mourão, A., Buday, K., Sato, M., Wanninger, J., Vignane, T., Mohana, eases. Nutrients, 4, 1399–­1440. https://doi.org/10.3390/nu410​1399
V., Rehberg, M., Flatley, A., Schepers, A. …Conrad, M. (2019). FSP1 is Lee, C.-­K ., Jeong, S.-­H., Jang, C., Bae, H., Kim, Y. H., Park, I., Kim, S.
a glutathione-­independent ferroptosis suppressor. Nature, 575, 693–­ K., & Koh, G. Y. (2019). Tumor metastasis to lymph nodes requires
698. https://doi.org/10.1038/s4158​6-­019-­1707-­0 YAP-­dependent metabolic adaptation. Science (80-­.). 363, 644–­6 49.
Dolma, S., Lessnick, S. L., Hahn, W. C., & Stockwell, B. R. (2003). https://doi.org/10.1126/scien​ce.aav0173
Identification of genotype-­selective antitumor agents using synthetic Lu, B., Chen, X. B., Ying, M. D., He, Q. J., Cao, J., & Yang, B. (2017). The
lethal chemical screening in engineered human tumor cells. Cancer role of ferroptosis in cancer development and treatment response.
Cell, 3, 285–­296. https://doi.org/10.1016/S1535​-­6108(03)00050​-­3 Front. Pharmacol, 8, 992. https://doi.org/10.3389/fphar.2017.00992
Eaton, J. K., Furst, L., Ruberto, R. A., Moosmayer, D., Hilpmann, A., Ryan, Luo, M., Wu, L., Zhang, K., Wang, H., Zhang, T., Gutierrez, L., O’Connell,
M. J., Zimmermann, K., Cai, L. L., Niehues, M., Badock, V., Kramm, D., Zhang, P., Li, Y., Gao, T., Ren, W., & Yang, Y. (2018). miR-­137 reg-
A., Chen, S., Hillig, R. C., Clemons, P. A., Gradl, S., Montagnon, C., ulates ferroptosis by targeting glutamine transporter SLC1A5 in
Lazarski, K. E., Christian, S., Bajrami, B. …Schreiber, S. L. (2020). melanoma. Cell Death and Differentiation, 25, 1457–­1472. https://doi.
Selective covalent targeting of GPX4 using masked nitrile-­ oxide org/10.1038/s4141​8-­017-­0 053-­8
electrophiles. Nature Chemical Biology, 16, 497–­ 506. https://doi. Masaldan, S., Bush, A. I., Devos, D., Rolland, A. S., & Moreau, C. (2019).
org/10.1038/s4158​9-­020-­0501-­5 Striking while the iron is hot: Iron metabolism and ferroptosis in
TALTY and BOSENBERG |
      25

neurodegeneration. Free Radical Biology and Medicine, 133, 221–­233. defines melanoma subtypes with differential vulnerability to drug-­
https://doi.org/10.1016/j.freer​adbio​med.2018.09.033 induced iron-­dependent oxidative stress. Cancer Cell, 33, 890–­904.
Müller, J., Krijgsman, O., Tsoi, J., Robert, L., Hugo, W., Song, C., Kong, X., e5. https://doi.org/10.1016/j.ccell.2018.03.017
Possik, P. A., Cornelissen-­Steijger, P. D. M., Foppen, M. H. G., Kemper, Tsoi, J., Robert, L., Paraiso, K., Galvan, C., Sheu, K. M., Lay, J., Wong, D.
K., Goding, C. R., McDermott, U., Blank, C., Haanen, J., Graeber, T. G., J. L., Atefi, M., Shirazi, R., Wang, X., Braas, D., Grasso, C. S., Palaskas,
Ribas, A., Lo, R. S., & Peeper, D. S. (2014). Low MITF/AXL ratio pre- N., Ribas, A., & Graeber, T. G. (2018b). Multi-­stage differentiation
dicts early resistance to multiple targeted drugs in melanoma. Nature defines melanoma subtypes with differential vulnerability to drug-­
Communications, 5. https://doi.org/10.1038/ncomm​s6712 induced iron-­dependent oxidative stress. Cancer Cell, 33, 890–­904.
Murphy, T. H., Miyamoto, M., Sastre, A., Schnaar, R. L., & Coyle, J. T. e5. https://doi.org/10.1016/j.ccell.2018.03.017
(1989). Glutamate toxicity in a neuronal cell line involves inhibition of Ubellacker, J. M., Tasdogan, A., Ramesh, V., Shen, B., Mitchell, E. C.,
cystine transport leading to oxidative stress. Neuron, 2, 1547–­1558. Martin-­Sandoval, M. S., Gu, Z., McCormick, M. L., Durham, A. B.,
https://doi.org/10.1016/0896-­6273(89)90043​-­3 Spitz, D. R., Zhao, Z., Mathews, T. P., & Morrison, S. J. (2020). Lymph
Piskounova, E., Agathocleous, M., Murphy, M. M., Hu, Z., Huddlestun, protects metastasizing melanoma cells from ferroptosis. Nature, 585,
S. E., Zhao, Z., Leitch, A. M., Johnson, T. M., DeBerardinis, R. J., & 113. https://doi.org/10.1038/s4158​6-­020-­2623-­z
Morrison, S. J. (2015). Oxidative stress inhibits distant metasta- Viswanathan, V. S., Ryan, M. J., Dhruv, H. D., Gill, S., Eichhoff, O. M.,
sis by human melanoma cells. Nature, 527, 186–­ 191. https://doi. Seashore-­Ludlow, B., Kaffenberger, S. D., Eaton, J. K., Shimada, K.,
org/10.1038/natur​e15726 Aguirre, A. J., Viswanathan, S. R., Chattopadhyay, S., Tamayo, P.,
Riesenberg, S., Groetchen, A., Siddaway, R., Bald, T., Reinhardt, J., Yang, W. S., Rees, M. G., Chen, S., Boskovic, Z. V., Javaid, S., Huang,
Smorra, D., Kohlmeyer, J., Renn, M., Phung, B., Aymans, P., Schmidt, C. …Schreiber, S. L. (2017). Dependency of a therapy-­resistant state
T., Hornung, V., Davidson, I., Goding, C. R., Jönsson, G., Landsberg, J., of cancer cells on a lipid peroxidase pathway. Nature, 547, 453–­457.
Tüting, T., & Hölzel, M. (2015). MITF and c-­Jun antagonism intercon- https://doi.org/10.1038/natur​e23007
nects melanoma dedifferentiation with pro-­inflammatory cytokine Wang, W., Green, M., Choi, J. E., Gijón, M., Kennedy, P. D., Johnson, J.
responsiveness and myeloid cell recruitment. Nature Communications, K., Liao, P., Lang, X., Kryczek, I., Sell, A., Xia, H., Zhou, J., Li, G., Li,
6. https://doi.org/10.1038/ncomm​s9755 J., Li, W., Wei, S., Vatan, L., Zhang, H., Szeliga, W. … Zou, W. (2019).
Robert, C., Grob, J. J., Stroyakovskiy, D., Karaszewska, B., Hauschild, CD8+ T cells regulate tumour ferroptosis during cancer immuno-
A., Levchenko, E., Chiarion Sileni, V., Schachter, J., Garbe, C., therapy. Nature, 569, 270–­ 274. https://doi.org/10.1038/s4158​
Bondarenko, I., Gogas, H., Mandalá, M., Haanen, J. B. A. G., Lebbé, 6-­019-­1170-­y
C., Mackiewicz, A., Rutkowski, P., Nathan, P. D., Ribas, A., Davies, Warner, G. J., Berry, M. J., Moustafa, M. E., Carlson, B. A., Hatfield, D. L.,
M. A. …Long, G. V. (2019). Five-­year outcomes with dabrafenib plus & Faust, J. R. (2000). Inhibition of selenoprotein synthesis by sele-
trametinib in metastatic melanoma. New England Journal of Medicine, nocysteine tRNA([Ser]Sec) lacking isopentenyladenosine. Journal of
381, 626–­636. https://doi.org/10.1056/NEJMo​a1904059 Biological Chemistry, 275, 28110–­28119. https://doi.org/10.1074/jbc.
Sato, M., Onuma, K., Domon, M., Hasegawa, S., Suzuki, A., Kusumi, R., Hino, M0012​8 0200
R., Kakihara, N., Kanda, Y., Osaki, M., Hamada, J., Bannai, S., Feederle, Wilson, M. A., Zhong, J., Rosenbaum, B. E., Utter, K., Moran, U.,
R., Buday, K., Angeli, J. P. F., Proneth, B., Conrad, M., Okada, F., & Sato, Darvishian, F., Polsky, D., Berman, R. S., Shapiro, R. L., Pavlick, A. C.,
H. (2020). Loss of the cystine/glutamate antiporter in melanoma abro- & Osman, I. (2019). Impact of initial stage on metastatic melanoma
gates tumor metastasis and markedly increases survival rates of mice. survival. Melanoma Research, 29, 281–­288. https://doi.org/10.1097/
International Journal of Cancer. https://doi.org/10.1002/ijc.33262 CMR.00000​0 0000​0 00526
Sauka-­Spengler, T., & Bronner-­Fraser, M. (2008). A gene regulatory net- Wu, J., Minikes, A. M., Gao, M., Bian, H., Li, Y., Stockwell, B. R., Chen,
work orchestrates neural crest formation. Nature Reviews Molecular Z. N., & Jiang, X. (2019). Intercellular interaction dictates cancer cell
Cell Biology, 9, 557–­568. https://doi.org/10.1038/nrm2428 ferroptosis via NF2–­YAP signalling. Nature, 572, 402–­4 06. https://
Schachter, J., Ribas, A., Long, G. V., Arance, A., Grob, J. J., Mortier, L., doi.org/10.1038/s4158​6-­019-­1426-­6
Daud, A., Carlino, M. S., McNeil, C., Lotem, M., Larkin, J., Lorigan, P., Yang, W. S., & Stockwell, B. R. (2008). Synthetic lethal screening identi-
Neyns, B., Blank, C., Petrella, T. M., Hamid, O., Zhou, H., Ebbinghaus, fies compounds activating iron-­dependent, nonapoptotic cell death
S., Ibrahim, N., & Robert, C. (2017). Pembrolizumab versus ipilimumab in oncogenic-­R AS-­harboring cancer cells. Chemistry & Biology, 15,
for advanced melanoma: Final overall survival results of a multicentre, 234–­245. https://doi.org/10.1016/j.chemb​iol.2008.02.010
randomised, open-­label phase 3 study (KEYNOTE-­0 06). Lancet, 390, Yang, Y., Luo, M., Zhang, K., Zhang, J., Gao, T., Connell, D. O., Yao, F.,
1853–­1862. https://doi.org/10.1016/S0140​-­6736(17)31601​-­X Mu, C., Cai, B., Shang, Y. & Chen, W. (2020). Nedd4 ubiquitylates
Seiler, A., Schneider, M., Förster, H., Roth, S., Wirth, E. K., Culmsee, C., VDAC2/3 to suppress erastin-­ induced ferroptosis in melanoma.
Plesnila, N., Kremmer, E., Rådmark, O., Wurst, W., Bornkamm, G. W., Nature Communications, 11. https://doi.org/10.1038/s4146​7-­020-­
Schweizer, U., & Conrad, M. (2008). Glutathione peroxidase 4 senses 14324​-­x
and translates oxidative stress into 12/15-­lipoxygenase dependent-­ Zhang, K., Wu, L., Zhang, P., Luo, M., Du, J., Gao, T., O’Connell, D., Wang,
and AIF-­mediated cell death. Cell Metabolism, 8, 237–­248. https:// G., Wang, H., & Yang, Y. (2018). miR-­9 regulates ferroptosis by target-
doi.org/10.1016/j.cmet.2008.07.005 ing glutamic-­oxaloacetic transaminase GOT1 in melanoma. Molecular
Siegel, R. L., Miller, K. D., & Jemal, A. (2019). Cancer statistics, 2019. CA: Carcinogenesis, 57, 1566–­1576. https://doi.org/10.1002/mc.22878
A Cancer Journal for Clinicians, 69, 7–­3 4. https://doi.org/10.3322/ Zhang, Y., Tan, H., Daniels, J. D., Zandkarimi, F., Liu, H., Brown, L. M.,
caac.21551 Uchida, K., O’Connor, O. A., & Stockwell, B. R. (2019). Imidazole ke-
Song, E., Mao, T., Dong, H., Boisserand, L. S. B., Antila, S., Bosenberg, tone erastin induces ferroptosis and slows tumor growth in a mouse
M., Alitalo, K., Thomas, J. L., & Iwasaki, A. (2020). VEGF-­C-­driven lymphoma model. Cell Chem. Biol., 26, 623–­ 633.e9. https://doi.
lymphatic drainage enables immunosurveillance of brain tumours. org/10.1016/j.chemb​iol.2019.01.008
Nature, 577, 689–­694. https://doi.org/10.1038/s4158​6-­019-­1912-­x
Stockwell, B. R., & Jiang, X. (2020). The chemistry and biology of ferro-
ptosis. Cell Chem Biol, 27, 365–­375. https://doi.org/10.1016/j.chemb​
How to cite this article: Talty, R., & Bosenberg, M. (2022).
iol.2020.03.013
The role of ferroptosis in melanoma. Pigment Cell & Melanoma
Tsoi, J., Robert, L., Paraiso, K., Galvan, C., Sheu, K. M., Lay, J., Wong, D.
J. L., Atefi, M., Shirazi, R., Wang, X., Braas, D., Grasso, C. S., Palaskas, Research, 35, 18–­25. https://doi.org/10.1111/pcmr.13009
N., Ribas, A., & Graeber, T. G. (2018a). Multi-­stage differentiation

You might also like