ArticlePDF AvailableLiterature Review

Radiotherapy-induced ferroptosis for cancer treatment

Authors:

Abstract and Figures

Ferroptosis is a regulated cell death mechanism controlled by iron, amino acid and reactive oxygen species metabolisms, which is very relevant for cancer therapy. Radiotherapy-induced ferroptosis is critical for tumor suppression and several preclinical studies have demonstrated that the combination of ionizing radiation with small molecules or nano-systems is effective in combating cancer growth and overcoming drug or ionizing radiation resistance. Here, we briefly overview the mechanisms of ferroptosis and the cross-talk existing between the cellular pathways activated by ferroptosis and those induced by radiotherapy. Lastly, we discuss the recently reported combinational studies involving radiotherapy, small molecules as well as nano-systems and report the recent findings achieved in this field for the treatment of tumors.
Content may be subject to copyright.
Radiotherapy-induced ferroptosis
for cancer treatment
Giovanni L. Beretta and Nadia Zaffaroni*
Molecular Pharmacology Unit, Department of Experimental Oncology, Fondazione IRCCS Istituto
Nazionale dei Tumori di Milano, Milan, Italy
Ferroptosis is a regulated cell death mechanism controlled by iron, amino acid and
reactive oxygen species metabolisms, which is very relevant for cancer therapy.
Radiotherapy-induced ferroptosis is critical for tumor suppression and several
preclinical studies have demonstrated that the combination of ionizing radiation
with small molecules or nano-systems is effective in combating cancer growth
and overcoming drug or ionizing radiation resistance. Here, we briey overview
the mechanisms of ferroptosis and the cross-talk existing between the cellular
pathways activated by ferroptosis and those induced by radiotherapy. Lastly, we
discuss the recently reported combinational studies involving radiotherapy, small
molecules as well as nano-systems and report the recent ndings achieved in this
eld for the treatment of tumors.
KEYWORDS
ferroptosis, radiotherapy, reactive oxygen species, nanomedicine, drug combinations,
gene signatures
1 Introduction
The accidental cell death (ACD) and the regulated cell death (RCD) govern the cell fate
(Tang et al., 2019). Necrosis is the best representative of ACD, which is a passive mechanism
allowing plasma membrane rupture, cytoplasm release and inducing inammation reaction.
Conversely, RCD is an active and regulated cell suicide, which can involve or not
inammation reaction, playing crucial functions in tissue homeostasis and in the
pathogenesis of several diseases (Galluzzi et al., 2018;Tang et al., 2019). So far, two
RCD categories are reported: apoptotic and non-apoptotic. Necroptosis, pyroptosis,
autophagy and ferroptosis belong to the non-apoptotic RCD and are classied according
to different molecular, morphological, biochemical and functional features (Galluzzi et al.,
2018). RCD pathways are implicated in physiologic processes regulating the development of
multicellular organisms and represent defense mechanisms against cancer transformation/
development as well as against pathogen infections (Galluzzi et al., 2018;Tang et al., 2019).
As suggested by its name, ferroptosis is stimulated by the lipid peroxidation provoked by the
iron accumulated into the cells. Critical for ferroptosis is the content of polyunsaturated-
fatty-acids (PUFA) composing the cellular membrane. PUFA represent a toxic reservoir that
in iron- and reactive oxygen species (ROS)-rich conditions are susceptible to peroxidation,
leading to membrane damage and cell death (Stockwell et al., 2017).
The induction of ferroptosis is a new interesting strategy for ghting cancer (Lei et al.,
2022). This strategy is based on the condition known as iron addiction, which is typical of
cancer cells that need higher levels of iron in comparison with healthy cells (Friedmann
Angeli et al., 2019). Iron addiction renders cancer cells more sensitive to iron and to iron-
induced ROS production (Fenton reaction) than normal cells (Torti and Torti, 2019).
Though ferroptosis induction proved efcacy in overcoming resistance to apoptosis
developed by tumors exposed to anticancer therapy, tumor cells challenged by
OPEN ACCESS
EDITED BY
Guo Chen,
China Pharmaceutical University, China
REVIEWED BY
Ana Cipak Gasparovic,
Rudjer Boskovic Institute, Croatia
Lixia Gao,
Chongqing University of Arts and
Sciences, China
*CORRESPONDENCE
Nadia Zaffaroni,
nadia.zaffaroni@istitutotumori.mi.it
RECEIVED 04 May 2023
ACCEPTED 05 June 2023
PUBLISHED 14 June 2023
CITATION
Beretta GL and Zaffaroni N (2023),
Radiotherapy-induced ferroptosis for
cancer treatment.
Front. Mol. Biosci. 10:1216733.
doi: 10.3389/fmolb.2023.1216733
COPYRIGHT
© 2023 Beretta and Zaffaroni. This is an
open-access article distributed under the
terms of the Creative Commons
Attribution License (CC BY). The use,
distribution or reproduction in other
forums is permitted, provided the original
author(s) and the copyright owner(s) are
credited and that the original publication
in this journal is cited, in accordance with
accepted academic practice. No use,
distribution or reproduction is permitted
which does not comply with these terms.
Frontiers in Molecular Biosciences frontiersin.org01
TYPE Review
PUBLISHED 14 June 2023
DOI 10.3389/fmolb.2023.1216733
ferroptosis inducers can evolve defense mechanisms that counteract
ferroptosis and in turn preserving cell vitality (Stockwell et al., 2017;
Hassannia et al., 2019;Zheng and Conrad, 2020). This implies that
drugs hitting cellular pathways involved in resistance to ferroptosis
potentiate pharmacological interventions (Liang et al., 2019).
Besides chemotherapy, tumors are treated by radiotherapy as
well. The exposure to ionizing radiation (IR) induces DNA damage
leading to cell death (Delaney et al., 2005;Jaffray, 2012). Besides
direct DNA damage, IR hits water molecules contained into the cells
favoring their radiolysis and, together with the activation of specic
enzymes, stimulate ROS production. ROS, including peroxides
(H
2
O
2
, ROOH), free radicals (HO,HO
2
,R,RO,NOand
NO
2
), singlet oxygen (
1
O
2
) and superoxide (O
2
), attack DNA,
lipids, and proteins (Azzam et al., 2012;Reisz et al., 2014). DNA
damages include nucleotide base damage, single strand breaks
(SSBs), and double strand breaks (DSBs) (Baidoo et al., 2013).
The cellular response to damaged DNA allows cell-cycle arrest,
cellular senescence, and RCD. Although apoptosis is the most
studied RCD induced by radiotherapy, other types of RCD are
reported in radiotherapy-treated cells, including ferroptosis (Lang
et al., 2019;Adjemian et al., 2020;Lei et al., 2020;Ye et al., 2020).
Since for the treatment of tumors radiotherapy is administered in
combination with chemotherapy, it is conceivable to study the
radiotherapy and ferroptosis pathways as well as their cross-talk
to set up combination strategies maximizing tumor response.
In this review we briey overview the cellular pathways
implicated in ferroptosis induced by radiotherapy and discuss the
potential combination strategies with small molecules and nano-
systems for enhancing radiotherapy antitumor activity.
2 General overview on ferroptosis
Compared to apoptosis, autophagy and necrosis, ferroptosis
shows peculiar properties. Morphological alterations of
mitochondria, including reduction in volume, increased density
of the mitochondrial membranes as well as reduced
mitochondrial cristae, characterize ferroptotic cells. Cells
undergoing ferroptosis are rounded and oating with intact
nuclei and uncondensed chromatin (Dixon et al., 2012;Stockwell
et al., 2017). Conversely, the typical features of apoptosis (e.g.,
chromatin condensation and the production of apoptotic bodies)
FIGURE 1
Cellular mechanisms of ferroptosis. The cellular pathways involving iron, amino acids and ROS metabolisms are reported. CIP, cellular iron pool; FR,
ferritin; CER, ceruloplasmin; TF, transferrin; TFR, transferrin receptor; FPN, ferroportin; ACSL4, acyl-CoA synthetase long-chain family member4;
LPCAT3, lysophosphatidylcholine acyltransferase 3; ALOXs, arachidonate lipoxygenase; GPX4, glutathione peroxidase 4; ASC, alanineserinecysteine
system; SLC7A11, solute carrier family 7 member 11; SLC3A2, solute carrier family 3 member 2; GLS, glutaminases; GSS, glutathione synthetase; GCL,
glutamate-cysteine ligase; PUFAs, polyunsaturated fatty acids; FSP1, ferroptosis suppressor protein 1; TXN, thioredoxin. AA, arachidonoyl; AdA, adrenic
acid; PE, phosphatidylethanolamines; RKIPI, Raf1 kinase inhibitory protein GSH, glutathione; GSSG, glutathione disulde. The gure is prepared using
tools from Servier Medical Art (http://www.servier.fr/servier-medical-art, accessed on March 2023).
Frontiers in Molecular Biosciences frontiersin.org02
Beretta and Zaffaroni 10.3389/fmolb.2023.1216733
as well as that of autophagy (e.g., the formation of autophagosomes)
are not reported in ferroptotic cells. Agents that inhibit apoptosis,
autophagy and necroptosis are ineffective on ferroptosis induction.
Therefore, the sensitivity to drugs that induce ferroptosis is
maintained by cells decient in apoptotic-related factors (e.g.,
BAX, BAK, MLKL, and RIPK1/3). On the contrary, antioxidants
and iron chelators inhibit ferroptosis (Vanden Berghe et al., 2010;
Lei et al., 2019).
Ferroptosis is governed by three main cellular mechanisms,
including i) iron metabolism, ii) amino acid metabolism and iii)
ROS metabolism (Figure 1)(Liang et al., 2019).
2.1 Iron metabolism
Iron transport systems regulate iron accumulation and in turn
ferroptosis induction. These transporters, including ceruloplasmin
(CER), transferrin (TF), transferrin receptor (TFR), ferritin (FR) and
ferroportin (FPN), impact on intracellular levels of iron. Adsorbed
Fe
2+
is oxidized to Fe
3+
by CER and in this form is captured by TF.
The interaction of TF with TFR favors the cellular uptake of iron.
Upon reduction to Fe
2+
by the sixtransmembrane epithelial antigen
of the prostate 3 (STEAP3), iron is bound to FR or stored into the
cellular iron pool (CIP). When cells are saturated by iron, exceeding
amounts of Fe
2+
are oxidized to Fe
3+
and pumped out of the cells by
FPN (Trujillo-Alonso et al., 2019). CIP is controlled by other two
factors: the nuclear receptor coactivator 4 (NCOA4), which is a
specic receptor favoring FR accumulation into the autophagosome,
and the iron-responsive element binding protein 2 (IREB2), which is
a transcription factor controlling the iron metabolism by regulating
the level of FR (Dixon et al., 2012;Mancias et al., 2014;Tang et al.,
2018). Interferences with the iron balance regulated by these
mechanisms (e.g., increased uptake or reduced export) stimulate
iron-mediated lipid peroxidation and ferroptosis (Yang and
Stockwell, 2008;Stockwell et al., 2017).
2.2 Amino acid metabolism
The exchange of cystine/cystathionine across the plasma
membranes depends on the red-ox state of the extracellular
compartment. Under reducing conditions, the intracellular
accumulation of cysteine is controlled by the
alanineserinecysteine (ASC) system. Conversely, oxidative
extracellular conditions stimulate the exchange of cystine/
cystathionine with glutamate mediated by Xctransporter system
(Doll and Conrad, 2017). Two subunits linked via a disulde bridge
compose the Xc, including the catalytic subunit solute carrier
family 7 member 11 (SLC7A11) and the regulatory subunit
solute carrier family 3 member 2 (SLC3A2). Intracellular
glutamate levels, which are under the control of the enzymatic
activity degrading glutamine (glutaminolysis) mediated by
glutaminases (GLS) 1 and 2, impact on Xcactivity. Diminished
cellular content of cysteine, which is stimulated by enhanced GLS
activity or reduced SLC7A11 levels as well as reduced activation of
spermidine/spermine N1 acetyltransferase 1 (SAT1), favors lipid
peroxidation and ferroptosis induction (Jiang et al., 2015;Jennis
et al., 2016;Ou et al., 2016;Zhang et al., 2018). Alterations in GLS,
SLC7A11 or SAT1 activities, which result in reduced intracellular
availability of cysteine, negatively impact on glutathione (GSH)
levels triggering ferroptosis (Shah et al., 2017). The synthesis of
GSH needs glutamate, cysteine, and glycine and is catalyzed by
glutamate-cysteine ligase (GCL) and glutathione synthase (GSS).
The erastin-mediated inhibition of Xc, which reduces cystine
uptake, or the inhibition of GSH biosynthesis via buthionine
sulfoximine, deplete intracellular GSH levels leading to ferroptosis.
2.3 ROS metabolism
Besides DNA damage, ROS stimulate ferroptosis by
provoking alterations in lipid metabolism (DHerde and
Krysko et al., 2017;Lin et al., 2018). The lipid peroxidation
occurring in PUFA is among the most important type of
cellular damage for ferroptosis induction, and cells with
high levels of PUFA are very sensitive to ferroptosis (Yang
et al., 2016;Yuan et al., 2016). The catalytic activity of two
enzymes, acyl-CoA synthetase long-chain family member 4
(ACSL4) and lysophosphatidylcholine acyltransferase 3
(LPCAT3), which allows the esterication and incorporation
of PUFA into membrane phospholipids, is crucial for
sensitizing cells to ferroptosis. The accumulation of lipid
peroxides enhances the formation of additional ROS that
increases biomacromolecule damage leading to cell
membrane destabilization and micelle formation, in turn
enhancing ferroptosis induction (Gaschler and Stockwell,
2017;Feng and Stockwell, 2018). Two proteins are critical
for ferroptosis induction as well, the small scaffolding
protein Raf1 kinase inhibitory protein (RKIP1) and the
seleno enzyme glutathione peroxidase 4 (GPX4) (Wenzel
et al., 2017;Tang et al., 2018;Liang et al., 2019). By
interacting with the iron-containing enzyme arachidonate
lipoxygenase 15 (ALOX15), RKIP1interferes with the
production of phospholipid alcohols regulating ferroptosis.
Similarly, GPX4 is a detoxifying enzyme catalyzing the
transformation of PUFA into non-toxic phospholipid
alcohols. The red-ox reaction involving the oxidation of
GSH into GSSG is required by GPX4 to accomplish its
catalytic activity, and cells showing high levels of GPX4 are
less susceptible to ferroptosis (Seibtetal.,2019). On the
contrary, compounds impairing the activity of GPX4 by
reducing its expression/activity are typical ferroptosis
inducers (Tang et al., 2018;Liang et al., 2019). Since the
activity of GPX4 needs selenocysteine tRNA, whose cellular
amount is controlled by the mevalonate (MVA) pathway, the
inhibition of MVA pathway favors ferroptosis by reducing the
availability of selenocysteine tRNA (Kryukov et al., 2003).
Another protein controlling GPX4 activity is the ferroptosis
suppressor protein 1 (FSP1). FSP1 inhibits GPX4 and its
expression is high in cells resistant to ferroptosis. Elevated
levels of FSP1 protect cells against compounds that induce
ferroptosis by targeting GPX4 (Bersuker et al., 2019;Doll et al.,
2019). Upon myristoylation, the cytoplasmic FSP1 moves to
the plasma membranes and in this peculiar cellular localization
catalyzes the reduction of coenzyme Q10. This behavior, in
presence of GSH as well as active GPX4, attenuates the
Frontiers in Molecular Biosciences frontiersin.org03
Beretta and Zaffaroni 10.3389/fmolb.2023.1216733
propagation of lipid peroxide production and reduces
phospholipid peroxidation attenuating ferroptosis (Doll
et al., 2019).
3 Cross-talk between radiotherapy and
ferroptosis in cancer
Besides direct damage of biomacromolecules (e.g., DNA,
proteins and lipids), radiotherapy generates ROS, which are the
most important molecules responsible for lipid peroxide
accumulation and ferroptosis. The relationship between
radiotherapy and ferroptosis is corroborated by several
evidence, including the specic staining (e.g., C11-BODIPY) as
well as the increased expression of specic markers (e.g., MDA, 4-
HNE and prostaglandin-endoperoxide synthase 2, PTGS2)
reecting lipid peroxidation observed in IR-exposed cancer cell
lines and tumor samples. Moreover, irradiated cells show
morphological alterations of mitochondria typical of ferroptosis
(Lang et al., 2019). Of note, these features depend on IR doses
administered. In support of the cross-talk between radiotherapy
and ferroptosis is the observation that treatment of cells with iron
chelators (e.g., deferoxamine) or with ferroptosis inhibitors (e.g.,
ferrostatin-1 and liproxstatin-1) before exposure to IR partially
rescue their survival, and that this nding is more evident in
comparison to what observed combining IR with compounds
that inhibits other RCD (Lei et al., 2021).
Three major pathways regulate IR-mediated ferroptosis
induction (Lang et al., 2019;Lei et al., 2020;Ye et al., 2020)
(Figure 2), including 1) ROS and ACSL4. Increased ROS
produced by IR are responsible for the formation of PUFA
radicals that, following the interaction with oxygen (Fenton
reaction), generate lipid hydroperoxides (PUFA-OOH) (Lei et al.,
2020). This pathway is powered by the IR-induced
ACSL4 expression (Figure 1) that, together with LPCAT3, favors
the synthesis of phospholipid containing PUFA, which are also liable
of peroxidation; 2) GSH and GPX4. IR exposure depletes GSH
leading to reduced activity/expression of GPX4 and in such a way
attenuating the GPX4-mediated detoxication functions leading to
increased toxic effects of lipid peroxides (Ye et al., 2020) and 3)
SLC7A11. Reduced levels of SLC7A11 favor ferroptosis by
downregulating cystine uptake and in turn GSH synthesis and
GPX4 functions (Figure 1). Through the activation of ataxia
telangiectasia mutated serine/threonine kinase (ATM), IR
represses SLC7A11 levels stimulating ferroptosis. Other studies
have underlined that the expression of SLC7A11 is increased
upon IR exposure leading to the interpretation that an adaptive
cellular response to IR rescues the SLC7A11 expression or that the
level of SLC7A11 upon IR exposure depends on a peculiar cellular
context (Xie et al., 2011;Lei et al., 2020).
The activation of the cellular pathways induced by the DNA
damage together with the stimulation of signaling pathways
associated with alterations of lipids contained into the cellular
membranes occurring upon radiotherapy exposure synergize each
FIGURE 2
Cellular pathways implicated in radiotherapy-induced ferroptosis. The gure reports the cross-talk occurring between ionizing radiation and
ferroptosis pathways, including ROS/ACSL4, GSH/GPX4, SLC7A11, ATM/ATR, and AMPK pathways. The gure is prepared using tools from Servier Medical
Art (http://www.servier.fr/servier-medical-art, accessed on March 2023).
Frontiers in Molecular Biosciences frontiersin.org04
Beretta and Zaffaroni 10.3389/fmolb.2023.1216733
other leading to enhanced tumor growth inhibition (Lei et al., 2020).
IR-induced DNA damage increases the expression of sensor
proteins, including ATM and ataxia telangiectasia and
Rad3 related serine/threonine kinase (ATR) that recognize the
altered DNA and stimulate DNA damage response signaling
cascades (DDR). DDR activate checkpoint kinases 1/2 (chk1/2)
and in turn stimulate the phosphorylation of p53 inducing cell-
cycle arrest. The block of the cell-cycle is required by the cells to test
the severity of the damage and decidetheir fate, 1) survive, upon
the activation of the DNA repair machine in case the damage can be
corrected, or 2) comit suicide, in case the damage is irreparable or
not correctly repaired, triggering RCD, including ferroptosis (Maier
et al., 2016;Huang and Zhou, 2020). Upon p53-mediated cell-cycle
arrest, irradiated cells mostly activate senescence (Bieging et al.,
2014;Maier et al., 2016). P53 mutation, which is a very common
condition in tumors, engage an alternative senescence checkpoint
protein, p16-retinoblastoma (RB) (Sabin and Anderson, 2011).
Senescence can coexists with apoptosis and in case of prolonged
p53 activation, IR preferentially stimulates both intrinsic (e.g.,
PUMA, BAX, NOXA, cytochrome C and caspase-9/3/7) and
extrinsic apoptosis (e.g., FAS/CD95, DR5, FAS ligands and
caspase-8) rather than senescence (Sheikh and Fornace, 2000;
Aubrey et al., 2018;Mijit et al., 2020). Besides the above
mentioned RCD, radiotherapy can induce autophagy and
necroptosis. Since autophagy has both pro-survival and pro-cell
death properties, controversial and not completely elucidated is its
role in IR response (Hu et al., 2016). Similarly, also the role of
necroptosis in radiation response is ambiguous (Adjemian et al.,
2020).
ATM activated by DNA damage increases p53 expression that
reduces the levels of SLC7A11 via a repressing interaction with
SLC7A11 promoter or by stimulating USP7-mediated proteasome
degradation of SLC7A11, in turn leading to ferroptosis (Kang et al.,
2019;Wang et al., 2019). Similarly, p53 induces ferroptosis by
stimulating the expression of SAT1, GLS2 or ferredoxin reductase
(FDXR) (Hu et al., 2010;Ou et al., 2016;Zhang et al., 2017). In
addition, p53 controls the expression of MDM2 that, via regulating
lipid metabolism and FSP1 expression, favors ferroptosis (Venkatesh
et al., 2020). Conversely, ferroptosis is attenuated upon p53-mediated
upregulation of p21. Metabolic stress induced by cystine deprivation
stimulates the p53-p21 axis preserving GSH levels and attenuating
ferroptosis (Tarangelo et al., 2018). Another protein involved in IR-
induced ferroptosis is the AMP-activated protein Kinase (AMPK) (Lei
et al., 2020;Ye et al., 2020). This protein can either stimulate or inhibit
ferroptosis. By stimulating the phosphorylation of beclin 1, which in
turn favors the downregulation of Xc, activated AMPK induces
ferroptosis (Song et al., 2018). In the other hand, AMPK activates
the biosynthesis of PUFA containing phospholipids inhibiting
ferroptosis (Lee et al., 2020). IR stimulates the expression of heme
oxygenase 1 (HMOX1) and FR, which trigger the release of iron and in
such a way the induction of ferroptosis (Chiang et al., 2018).
Conversely, the exposure to IR upregulates FR heavy chain (FTH1)
and reduces oxidative stress in turn attenuating ferroptosis and
promoting radiation resistance (Choudhary et al., 2020). These
ndings indicates an intersection between cellular pathways
implicated in DNA damage response and RCD, which
includes, besides ferroptosis, the immunogenic cell death (ICD)
mechanisms embracing apoptosis, necroptosis and autophagy.
Upon radiotherapy-mediated ICD stimulation, T-cells recruited into
the tumor microenvironment (TME) promote ferroptosis (Lang et al.,
2019).
3.1 Combination strategies for enhancing
ionizing radiation antitumor activity and for
overcoming radiation resistance
Radiation resistance of tumors is an urgent clinical problem
responsible for treatment failure. The understanding of the
molecular mechanisms subtending radiation resistance is critical
for setting up medical strategies, including drug combinations,
aimed at improving rediotherapy response (Table 1).
Radiotherapy is used for the clinical management of patients with
different tumor types including nasopharyngeal carcinoma (NPC) and
the development of radiation resistance is the main cause of treatment
failure in NPC-suffering patients. Huangetal.reportthatthesepatients
show increased expression of m6A mRNA demethylase fat mass and
obesity-associated protein (FTO) and that this feature correlates with
radiation resistance and poor prognosis (Huang et al., 2023). In support
of this observation is the nding that increased levels of FTO
characterize NPC cell lines resistant to IR (C666-1R, HONE1R)
compared to the corresponding sensitive counterparts (C666-1,
HONE1). NPC cells exposed to IR show morphological changes
typical of ferroptosis, increased MDA levels and reduced GSH
cellular content. The treatment with FB23-2 (a FTO inhibitor)
counteractsthisbehavior,which is reversed following FTO
overexpression or upon exposure to ferrostatin-1. Moreover, the IR
response of resistant cells is signicantly improved by FB23-2 treatment.
The exposure to IR and FB23-2 increases DNA damage in vitro and this
observation is corroborated in vivo in xenograft HNE1R-bearing mice.
TheauthorsspeculatethatalinkexistsbetweenFTOandOTU
deubiquitinase, ubiquitin aldehyde binding 1 (OTUB1). Molecularly,
via its demethylase activity, FTO produces m6A modication of the
OTUB1 transcript stimulating the expression of OTUB1 protein and its
interaction with SLC7A11, in turn activating SLC7A11 and leading to
ferroptosis inhibition and radiation resistance. These interpretations are
supported in vivo in xenograft HONE1R-bearing mice and PDX-mouse
models exposed to the combination erastin/IR. Compared to IR alone,
the combination with the ferroptosis inducer erastin enhances the
radiosensitivity of HONE1R tumors.
Another key protein implicated in radiation resistance by
inhibiting ferroptosis is angiopoietin-like 4 (ANGPTL4) (Zhang
et al., 2022). High levels of ANGPTL4 associate with poor
prognosis of patients suffering from lung adenocarcinoma and
adrenocortical carcinoma. A549 and H1299 lung cancer cell lines
cultured under hypoxic condition show an increased expression of
ANGPTL4 and radiation resistance. Moreover, compared to cells
cultured under physiologic conditions, hypoxic cells show higher
levels of ANGPTL4 accumulated into released exososmes. Upon
silencing of ANGPTL4, A549 cells restore the sensitivity to IR, while
reacquire IR resistance after ANGPTL4 ectopic expression. The
relationship between ANGPTL4 and ferroptosis is demonstrated in
ANGPTL4-overexpressing cells cultured under normoxia, which
show increased expression of GPX4, SLC7A11, FTH1, and FTL (e.g.,
reduced ferroptosis and enhanced radiation resistance). Conversely,
the above ferroptosis-associated proteins are decreased in
Frontiers in Molecular Biosciences frontiersin.org05
Beretta and Zaffaroni 10.3389/fmolb.2023.1216733
ANGPTL4-silenced cells cultured under hypoxic conditions (e.g.,
increased ferroptosis and radiosensitivity). In vivo experiments in
xenograft A549-bearing mice treated with ANGPTL4 enriched
exosomes and exposed to IR support in vitro data conrming the
relationship between radiation resistance and ferroptosis.
Radiation resistance accounts for the failure of radiotherapy in
hepatocellular carcinoma (HCC) as well and Chen and colleagues
have studied the role played by ferroptosis in radiosensitizing HCC
using a panel of radiosensitive (SK-Hep and HepG2) and
radioresistant (SK-Hep-1R and HepG2-1R) HCC cell lines (Chen
et al., 2023). By comparing the gene expression proles of SK-Hep
and SK-Hep-1R cells, SOCS2, and SMOX genes were identied as
differentially expressed. The expression of SOCS2 gene was reduced
in SK-Hep-1R, while SMOX was increased. Gene expression data
from GEPIA and HCCDB databases and Kaplan-Meier (K-M)
analysis showed that low expression of SOCS2 and high
expression of SMOX correlate with poor prognosis of HCC
patients. The radiation resistance observed in resistant cells is
overcome upon SOCS2 ectopic expression. Conversely, radiation
resistance is enhanced in SK-Hep and HepG2 cells following
SOCS2 silencing. In vivo experiments conrm the in vitro results.
Moreover, immunohistochemical staining of irradiated explanted
tumors shows an increased expression of SOCS2 paralleled by a
modulation of ferroptosis markers, including a downregulation of
GPX4 and SLC7A11, and an increased expression of 4-HNE. This
observation is conrmed in a set of clinical tissues in which the levels
of ferroptosis markers correlate with radiotherapy response. Co-
immunoprecipitation assays demonstrate a protein interaction
between SOCS2 and SLC7A11. Following the protein interaction,
the E3-ubiquitin ligase activity of SCOS2 allows ubiquitination and
proteasomal degradation of SLC7A11 leading to ferroptosis.
Iron homeostasis is controlled by copper (Cu) and elevated
levels of this micronutrient impact on tumorigenesis, resistance to
treatments and prognosis of HCC suffering patients (Yang et al.,
2022). HepG2 and MHCC-97H cells exposed to IR downregulates
the copper metabolism MURR1 domain 10 (COMMD10)
increasing the cellular accumulation of Cu and leading to
radiation resistance. These results are corroborated by gain and
TABLE 1 Combination strategies for enhancing ferroptosis induced by ionizi ng radiation.
Tumor type Cell lines Pathway involved In vivo evaluation Combination strategy References
Nasopharyngeal carcinoma C666-1 FTO/OTUB1/SLC7A11 HONE1R IR/FB23-2 Huang et al. (2023)
HONE1
C666-1R
HONE1R
Lung cancer A549 ANGPL4/GPX4/SLC7A11 A549 IR/ANGPTL4 enriched exosomes Zhang et al. (2022)
H1299
Hepatocellular carcinoma SK-Hep SOCS2/SLC7A11 SK-Hep-1 SK-Hep-1R IR/SOCS2-overexpressing plasmid Chen et al. (2023)
HepG2
SK-Hep-1R
HepG2-1R
Hepatocellular carcinoma HepG2 COMMD10/HIF1α/
SLC7A11
HepG2 IR/Cu chelator Yang et al. (2022)
MHCC-97H
Oral squamous cell carcinoma SCC15-S GPX4 SCC15-R IR/Hyperbaric oxygen (HBO) Liu et al. (2022)
SCC15-R
Colorectal cancer MC38 ATF3-SLC7A11-GPX4 MC38 IR/niraparib Shen et al. (2022)
CT26
HT29
Esophageal squamous cell carcinoma KYSE30 SCD1 N.D. IR/MF-438 Luo et al. (2022)
KYSE70
KYSE140
KYSE150
KYSE410
KYSE450
KYSE510
N.D., not dened.
Frontiers in Molecular Biosciences frontiersin.org06
Beretta and Zaffaroni 10.3389/fmolb.2023.1216733
loss of function experiments performed in a panel of HCC cell lines
showing that increased COMMD10 levels parallel with reduced
cellular accumulation of Cu and radisensitivity. Conversely, reduced
expression of COMMD10 increases Cu accumulation leading to
radiation resistance. The role played by Cu in tumor growth is
conrmed in vivo in HepG2-bearing mice fed with Cu-rich food
upon irradiation. Compared to mice fed under normal conditions,
animals fed with Cu-rich food show increased tumor volume, which
is reduced following the administration of a Cu chelator. Moreover,
immunohistochemistry analysis of HCC clinical samples shows that
the levels of COMMD10 in patients sensitive to radiotherapy are
higher than that measured in samples from radioresistant subjects.
Proteomic analysis performed in HepG2 and COMMD10-depleted
HepG2 cells reveals a differential expression of genes implicated in
cell death. These ndings, together with the observation that the
treatment with ferrostatin-1 and liproxstatin-1 increases the growth
of COMMD10 overexperring cells, indicate that ferroptosis is
critical for radiosensitivity. Based on the observation that the
sequence of the promoter of SLC7A11 contains binding sites for
HIF1-α, the authors propose an interplay between COMMD10,
SLC7A11, and HIF1-αas a molecular mechanism supporting the
cellular response. By interacting with HIF1-α, COMMD10 impedes
HIF1-αnuclear translocation and reduces SLC7A11 expression. The
IR-mediated reduced expression of COMMD10 favors the cellular
accumulation of Cu that stabilizes HIF1-αand inhibits its ubiquitin
degradation. Following nuclear translocation, HIF1-αstimulates
CER and SLC7A11 transcription inhibiting ferroptosis.
The effect of the co-exposure of IR and hyperbaric oxygen
(HBO) on oral squamous cell carcinoma (OSCC) cells is studied
by Liu and co-workers (Liu et al., 2022). Compared to the exposure
to IR, the combination IR/HBO increases the cytotoxicity in SCC15-
S cells, which is only in minimal part mediated by apoptosis.
Conversely, the combination enhances the levels of ferroptosis
markers (iron, ROS and MDA). Upon IR exposure, cells increase
the levels of ACSL4 and SLC7A11 (two ferroptosis promoters) as
well as that of GPX4 (a ferroptosis blocker), the latter being the
major factor accounting for radiation resistance. The treatment with
IR/HBO is ineffective on ACSL4 and SLC7A11 levels, while reduces
the expression of GPX4 and in such a way shifts the equilibrium
towards the induction of ferroptosis. These ndings are
corroborated by the observation that the transfection of SCC15-S
cells with GPX4-overexpressing plasmid or the treatment with
ferrostatin-1 reverse the effects of the combination. The authors
also show that IR/HBO exposure sensitizes the radio-resistant
SCC15-R cells to IR. Although less sensitive to ferroptosis,
SCC15-R cells exposed to the combination signicantly reduce
the expression of GPX4 in turn favoring ferroptosis, and these
results are conrmed in vivo in xenograft SCC15-R-bearing mice.
Compared to mice treated with IR, animals exposed to IR/HBO
show a signicant reduction in tumor growth. In support of the
ferroptosis induction as a mechanism of tumor growth delay is the
observation that the treatment with ferrostatin-1 counteracts the
antitumor activity of IR/HBO exposure. A deeper investigation on
clinical samples (tumor and normal tissues as well as serum) from
38 OSCC patients shows that, compared to adjacent normal oral
tissues, the expression of GPX4 is increased in tumors. In addition,
the serum levels of GPX4 in cancer patients are increased in
comparison to healthy donors. Of note, high levels of GPX4 (e.g.,
reduced ferroptosis) are accompanied with poor chemo-
radiotherapy outcome.
Another interesting combination for sensitizing colorectal
cancer (CC) to IR is proposed by Shen and colleagues (Shen
et al., 2022). To protect themselves against DNA damages, cancer
cells stimulate poly (ADP-ribose) polymerase (PARP)-1 and PARP-
2 activities favoring the activation of DNA repair pathways. This
scenario supports the rational of combining radiotherapy with
PARP inhibitors (PARPi) to potentiate IR-mediated DNA
damage and in turn increasing cell death. Compared to CC cell
lines (murine MC38 and CT26 cells as well as human HT29 cells)
exposed to IR, cells exposed to the combination of the PARPi
niraparib with IR show increased levels of DNA DSBs. The
combination signicantly increases the cell death in vitro and
enhances antitumor effects in vivo. The exposure to PARPi
induces the cyclic GMP-AMP synthase (cGAS) and stimulator of
interferon genes (STING), allowing the activation of cGAS-STING-
TBK1-IRF3 signaling that stimulates IFNB1 transcription and the
release of IFNβ, CXCL10, CCL5, and MX1. These ndings are also
observed in vivo in M38 tumor-bearing mice. The role played by
cGAS is corroborated by the observation that cGAS-silenced
M38 cells are less sensitive to the combination treatment. The
levels of ferroptosis markers (increased MDA and PTGS2 levels,
reduced SLC7A11 and GPX4 expression) reect the induction of
ferroptosis as a mechanism of cell death. The analysis of the gene
expression of cGAS-silenced M38 cells in comparison to cGAS
normal expressing cells supports a critical role for the activating
transcription factor 3 (ATF3) and underlines the existence of a
ATF3-SLC7A11-GPX4 axis controlling ferroptosis induction upon
exposure to IR and niraparib. In addition, cGAS depletion in
M38 tumor-bearing mice abolishes the IR-induced inltration of
CD8+T, CD8+GZMB + T-cells leading to reduced antitumor
efcacy, thus corroborating the role of cGAS for the combination
efcacy. The analysis of tumor samples before and after
radiotherapy from 32 patients affected by CC reveals that
increased expression of cGAS, ATF3, and PTGS2 as well as an
high density of CD8+T-cells associate with a high disease-free
survival rate.
The enzyme stearoyl-CoA desaturase (SCD1) catalyzes the
formation of oleic acid and palmitoleic acid and plays a critical
role in IR response. Increased levels of SCD1 are observed in a panel
of esophageal squamous cell carcinoma (ESCC) cell lines and the
targeting of SCD1 by MF-438 is pursued by Luo and colleagues for
increasing IR potency (Luo et al., 2022). Cells treated with MF-438
reduce cell growth and the combined exposure to IR and subtoxic
concentrations of MF-438 results in synergistic antiproliferative
activity. The synergism is attenuated upon silencing of SCD1 as
well as following exposure to the ferroptosis inhibitor RLS3. The
MF-438-mediated inhibition of SCD1 increases lipid peroxidation,
ATP and HMGB1 release into the extracellular compartment, and
this behavior is enhanced upon exposure to the combination IR/MF-
438. Since similar results are observed in cells exposed to the
combination IR/RLS3, it is likely that the induction of ferroptosis
is the key mechanism of antitumor activity. These observations are
corroborated by the nding that cells exposed to exogenous oleic
acid or palmitoleic acid undergo ferroptosis. In vitro data are
conrmed in vivo in ESCC-bearing mice exposed to IR/MF-438.
The authors analyze the expression of SCD1 in ESCC patients from
Frontiers in Molecular Biosciences frontiersin.org07
Beretta and Zaffaroni 10.3389/fmolb.2023.1216733
GEPIA database and stratify them in high and low expression
groups. Compared to normal epithelium, SCD1 is signicantly
increased in tumor tissues and high SCD1 expressing patients
experience a shorter disease-free survival.
3.2 Nano-systems for enhancing ionizing
radiation antitumor activity
Another strategy for potentiating IR-mediated ferroptosis
induction implies the use of metal-based nanoparticles (NPs)
(Table 2). These NPs exploit the enhanced permeability retention
effect to selectively induce ferroptosis in tumors. The tumor
selectivity is also enhanced by the in local IR exposure, and these
features allow reduced treatment toxicity.
3.2.1 Iron-based nanosystems
Lin and coworkers have assembled hemin, PX-12 (a TRX-1
inhibitor) and human serum albumin to built HPNPs (Lin et al.,
2023).TheseNPsarestableinphysiologicsolutionandinbloodand
rapidly release PX-12 under acidic conditions (e.g., pH 5). Acidic
conditions, recapitulating acidic TME, favor the production of OH
by HPNPs and stimulate Fenton reaction. In vitro experiments
performed in mouse melanoma B16F10 cells and normal mouse
L929 broblasts show that HPNPs are better internalized in tumor
compared to normal cells. The combination HPNPs/radiotherapy
improves ROS production leading to increased cytotoxicity with
respect to HPNPs administered alone. Besides the increased ROS
production (mediated by both hemin and radiotherapy), the
combination HPNPs/radiotherapy implements MDA, reduces the
levels of antioxidants (GSH and TRX-1) and attenuates the
GPX4 activity, in turn stimulating ferroptosis. These ndings are
corroborated by the observation that the treatment with ferrostatin-1
counteracts this behavior. In vivo experiments in B16F10 tumor-suffering
mice demonstrate that HPNPs are biocompatible. Moreover, compared
to animals treated with HPNPs, the tumor growth of mice exposed to the
combination HPNPs/radiotherapy is signicantly reduced. Ex vivo
analysis evidences reduced GSH and GPX4 levels as well as increased
MDA content in tumors exposed to the combination with respect to
those treated with HPNPs alone.
To potentiate radiotherapy efcacy in breast cancer, Hou et al.
propose multifunctional NPs composed by a shell of platinum
decorated with hyaluronic acid (HA) encapsulating a core of
Fe(III)-polydopamine (FPH) (Hou et al. 2023). FPH are stable at
pH 7.4 and show photothermal properties upon 808 nm irradiation.
Conversely, in acidic conditions NPs dissociate and release Fe
3+
.The
red-ox reaction Fe
3+
/Fe
2+
converts GSH into GSSG allowing GSH
depletion and H
2
O
2
hydrolysis producing O
2
and OH(Fenton
reaction). Since the depletion of GSH is increased at 50°C, it is
conceivable that the photothermal-mediated hyperthermia
improves antitumor potency of FPH. An additional property of
FPH is their ability to produce O
2
by catalyzing the hydrolysis of
TABLE 2 Nano-systems for enhancing the antitumor activity of ionizing radiation.
Nano-system Type of metal Cell lines In vivo evaluation References
HPNPs Iron Mouse melanoma B16F10 cells Mouse melanoma B16F10 tumors Lin et al. (2023)
Mouse normal L929 broblasts
FPH Iron Mouse mammary carcinoma 4T1 cells Mouse mammary 4T1 tumors Hou et al. (2023)
Mouse RAW264.7 macrophages
Fe
2
O
3
@TA-Pt Iron Mouse mammary carcinoma 4T1 cells Mouse mammary 4T1 tumors Jiang et al. (2022)
Normal human HUVEC cells
GOD@FeN
4
-SAzyme Iron Mouse mammary carcinoma 4T1 cells Mouse mammary 4T1 tumors Zhu et al. (2022)
SPIONC Iron Human lung cancer NCI-H460 cells Human lung cancer NCI-H460 tumors Li et al. (2022)
iCoDMSN Cobalt Mouse mammary carcinoma 4T1 cells Mouse mammary 4T1 tumors Zhao et al. (2023)
Human breast cancer MCF-7 cells
Human lung cancer A549 cells
Human colorectal cancer Caco-2 cells
Human gastric carcinoma SGC-7901 cells
AGulX Gadolinium Human breast cancer MDA-MB-231 cells Human breast cancer MDA-MB-231 tumors Sun et al. (2022)
Human breast cancer MDA-MB-468 cells
3a Gold Human cervical carcinoma HeLa cells Human cervical carcinoma HeLa cells transplanted in
zebrash
Yang et al. (2022)
Human cervical carcinoma SiHa cells
PBmB-DOX Bismuth Mouse mammary carcinoma 4T1 cells Mouse mammary 4T1 tumors Hou et al. (2022)
Frontiers in Molecular Biosciences frontiersin.org08
Beretta and Zaffaroni 10.3389/fmolb.2023.1216733
H
2
O
2
by Pt nanoenzyme. In vitro experiments carried out in mouse
4T1 breast cancer cells and in RAW264.7 macrophage cells show a
preferential accumulation of FPH in cancer cells, likely dependent on
the interaction of HA with CD44. In addition, compared to the
treatment with FPH, near infrared radiation (NIR, 808 nm) or IR
alone, the exposure to FPH/NIR and FPH/IR signicantly potentiates
cytotoxicity in 4T1 cells. The improved cytotoxicity observed in cells
treated with the combinations correlates with increased depletion of
GSH levels and increased ROS and DNA damages. This behavior is
counteracted by the treatment with ferrostatin-1. In vivo experiments
performed in 4T1 tumor-bearing mice corroborate in vitro results and
demonstrate the biocompatibility of FPH. NIR absorbance, Pt-
mediated X-ray attenuation and enhanced permeability retention-
mediated tumor selectivity render FPH a very useful tool for imaging
as well.
Fe
2
O
3
@TA-Pt are NPs containing a core of Fe
2
O
3
covered by
platinum and tannic acid (TA-Pt) (Jiang et al., 2022). Fe
2
O
3
@TA-Pt
arestableinmouseserum,whileacidicpH(5.5),mimickingTME,favors
the disassembling of the TA-Pt envelop releasing Fe
2
O
3
. In presence of
H
2
O
2
, which is abundant in TME, Fe
3+
is converted in Fe
2+
generating O
2
and OH(Fenton reaction). Fe
2
O
3
@TA-Pt better accumulate into
4T1 cells with respect to normal HUVEC cells, and this observation
correlates with the increased cytotoxicity in tumor cells. The analysis of
the DNA damage upon NPs exposure reveals that, besides the ROS-
mediated DNA damage, Pt-DNA adducts, which reect the release of the
Pt by NPs, are observed. Additionally, Fe
2
O
3
@TA-Pt treatment enhances
the radiotherapy sensitivity of 4T1 cells and potentiates ferroptosis
induction, as demonstrated by the reduced levels of GSH and
GPX4 observed upon combination exposure. In vivo experiments in
4T1 tumor-bearing mice show the preferential accumulation of NPs in
liver and tumor. Moreover, the tumor volume of mice exposed to
radiotherapy upon treatment with Fe
2
O
3
@TA-Pt is signicantly
reduced with respect to that of animals singly treated with NPs or
radiotherapy. The combination Fe
2
O
3
@TA-Pt/IR is well tolerated with
nosignsoftoxicity,reducesthetumorrecurrenceaswellasthe
pulmonary metastasis and enhances the survival of mice.
Single-atom nanozymes (SAzymes) are enzyme-based drugs
containing a single metal atom in their active sites that are
interesting for anticancer therapy. Critical for the antitumor
properties of these nano-systems is the presence into the TME of
a specic enzymatic activity as well as H
2
O
2
.Zhu et al. (2022) have
engineered a SAzyme based on FeN
4
and glucose oxidase (GOD)
(GOD@FeN
4
-SAzyme) for radio-enzymatic therapy. The elevated
glucose level in tumor over the normal cells allows the production of
H
2
O
2
via GOD of the GOD@FeN
4
-SAzyme and this results in
sustained production of OHand O
2
as well as in GSH
depletion. The enzymatic cascade triggered by GOD is enhanced
by IR, which favor the conversion Fe
3+
/Fe
2+
implementing the
generation of OHand potentiating apoptosis and ferroptosis.
Cytotoxic experiments performed in 4T1 cells show that the
killing activity of GOD@FeN
4
-SAzyme is enhanced upon
exposure to IR. Compared to the treatment with GOD@FeN
4
-
SAzyme or IR separately, the combination GOD@FeN
4
-SAzyme/
IR signicantly increases the DNA damages (increased γ-H2AX
signals) as well as ferroptosis (increased lipid peroxidation and OH,
reduced GSH and GPX4 levels accompanied by mitochondria
membrane alterations). Besides ferroptosis, treated 4T1 cells show
apoptosis induction (e.g., PARP and caspase 3 activation).
Intravenous and intratumoral injection are used for in vivo
administration of GOD@FeN
4
-SAzyme in 4T1-tumor bearing
mice. Magnetic resonance imaging (MRI) reveals that GOD@
FeN
4
-SAzyme accumulate into the tumor. Compared to the
exposure to GOD@FeN
4
-SAzyme or IR, a signicant reduction of
tumor volume is observed in animals treated with the combination
GOD@FeN
4
-SAzyme/IR. Ex vivo investigation recapitulates in vitro
ndings (e.g., increased γ-H2Ax and reduced GSH). GOD@FeN
4
-
SAzyme are biocompatible and the combination is well tolerated
as well.
The pH-sensitive supramagnetic iron oxide nano-clusters (SPIONC)
are proposed by Li et al. (2022) for enhancing radiation sensitivity in lung
cancer. Under acidic conditions, which recapitulate TME and
intracellular compartment conditions, SPIONC decompose and
generate OHvia Fenton reaction. SPIONC efciently accumulate in
NCI-H460 cells. Compared to SPIONC or IR individual exposure, the
combination SPIONC/IR is more effective in inhibiting cell proliferation.
Cells exposed to the combination increase iron, lipid peroxides, ROS and
γ-H2AX levels. These features reect apoptosis (reduced expression of
Bcl2 and increased caspase 3 activation) and ferroptosis induction
(reduced expression of SLC7A11 and GPX4). The involvement of
ferroptosis in SPIONC/IR response of tumor cells is corroborated by
the observation that the pre-treatment with ferrostatin-1 attenuates
cytotoxicity. In vivo experiments in orthotopic mice model of NCI-
H460 cells in which SPIONC are injected by both intravenous and intra-
tracheal delivery show that intra-tracheal delivery is to prefer for MRI
analysis.Moreover,theexposuretothecombinationSPIONC/IRresults
in increased tumor volume inhibition and survival rate with respect to the
administration of SPIONC or IR separately. No important toxic side
effects are reported upon treatment. The analysis of explanted tumors
from euthanized mice conrms the molecular alterations observed in
vitro.
3.2.2 Nano-systems based on cobalt, gadolinium,
gold, and bismuth
Another ferroptosis-stimulating nano-system has been recently
proposed by Zhao and colleagues (Zhao et al., 2023). Starting from
the observation that high level of cobalt (Co) in tumors associates
with a good prognosis, the authors have engineered Co oxide
nanodots by assembling bovine serum albumin and CoCl
2
. These
nanodots are conjugated with iRGD peptides and encapsulated into
dentritic mesoporus silica nanoparticles (iCoDMSN). Acidic
conditions stimulate the release of Co
2+
by iCoDMSN and the
presence of iRGD favors their tumor penetration. iCoDMSN are
cytotoxic on a panel of different tumor cell lines, including murine
4T1 cells and human MCF-7 breast cancer cells, human A549 lung
cancer cells, human Caco-2 colorectal cancer cells and human SGC-
7901gastric carcinoma cells. Therefore, these nanostructures show
interesting photoacoustic imaging ability under 808 nm irradiation
and in vivo experiments in 4T1-tumor bearing mice show that
iCoDMSN are well tolerated (e.g., no important signs of liver and
kidney toxicity), preferentially co-localize with lysosomes and that
iRGD favors tumor accumulation. Proteomic studies performed on
4T1 cells exposed to iCoDMSN underline that ferroptosis pathways
play a critical role for cell response. These ndings are supported by
the increased lipid peroxidation, MDA and iron levels observed in
iCoDMSN-treated 4T1 cells, and by the observation that this
behavior is counteracted upon exposure to ferrostatin-1.
Frontiers in Molecular Biosciences frontiersin.org09
Beretta and Zaffaroni 10.3389/fmolb.2023.1216733
Proteomic analysis also underlines an increased level of HMOX1,
which controls Fe
2+
accumulation and ferroptosis induction by
increasing the expression of TFR as well as by reducing solute
carrier family 40 member 1 (SLC40A1). Upon treatment,
accumulated iCoDMSN perturb the KEAP1/NRF2/HMOX1 axis.
This axis is governed by the level of nuclear factor erythroid 2-
related factor 2 (NRF2, Bellezza et al., 2018), which is a transcription
factor for HMOX1. These results are conrmed in vivo in 4T1-
tumor bearing mice exposed to the combination iCoDMSN/
radiotherapy. Compared to mice treated with iCoDMSN, a
increased tumor volume inhibition and survival rate are observed
in mice treated with iCoDMSN/radiotherapy. Ex vivo analysis
conrms the results obtained in vitro (reduced KEAP1, increased
NRF2, HMOX1, iron, and MDA).
Aimed at overcoming radiation resistance and reducing
radiotherapy damages to normal tissues, Sun and colleagues have
proposed gadolinium (Gd)-based NPs (AGulX) (Sun et al., 2022).
AGulX are based on polysiloxane covering Gd entrapped by the
chelator dodecane tetraacetic acid (DOTA) moieties that functionalize
the polysiloxane. Upon IR exposure, AGulX produce secondary and
Auger electrons as well as free radicals. These NPs are under clinical
investigation in brain, lung and pancreatic cancers. In the study by Sun
et al., AGulX are evaluated in triple negative breast cancer cells (MDA-
MB-231 and MDA-MB-468 cell lines) in vitro and in vivo.Comparedto
the treatment with AGulX or IR, the combination AGulX/IR reduces cell
growth as well as cell migration and invasion capability. Additionally,
cells exposed to the combination enhance ROS production, DNA
damage (increased number of γ-H2AX foci) as well as G2/M cell-
cycle arrest. Molecularly, the combination stimulates the
phosphorylation of ATR and Chk1 (e.g., G2/M block) and reduces
the phosphorylation of ATM and Chk2 (e.g., reduced homologous
recombination repair ability). Moreover, by diminishing the activation
of the MRN-ATM-Chk2 axis, the combination also impairs the non
homologous end-joining, which is reected by the reduced
phosphorylation of p53 and BRCA1. Besides apoptosis induction
(e.g., PARP and caspase 3 activation), cells exposed to AGulX/IR
reduce the expression of NRF2 favoring ferroptosis by attenuating
SLC7A11 activity, in turn reducing GSH synthesis and GPX4 activity.
The increased levels of lipid perixidation and MDA support the
induction of ferroptosis as a mechanism of cell death. These ndings
are conrmed by the observation that ferroptosis is attenuated by the
siRNA-mediated silencing of NRF2, SLC7A11 and GPX4 as well as by
the treatment with ferrostatin-1. The in vitro results parallel the in vivo
observations in MDA-MB-231 tumor-bearing mice. No important signs
of toxicity are reported in treated animals.
A series of metal-biotin-conjugated nano-structures based
on different metals endowed with radiosensitizer properties is
proposed by Yang and colleagues (Yang et al., 2022). Among the
different nano-systems, the gold derivative 3a is selected for
further investigations. 3a contains a biotin moiety for favoring
tumor selectivity and uptake, a triply bonded dicarbon alkynyl
amide linker joining biotin to Au, which is hidden by a lipophilic
phosphine residue to increase membrane solubility. Compared
to the auranon (the reference), 3a shows similar
antiproliferative potency on human cervical carcinoma HeLa
and SiHa cells. Moreover, the tumor selectivity of 3a, which is
dependent on biotin, is supported by the observation that 3a
uptake is higher in Hela cells (expressing high levels of biotin
receptor) with respect to normal human cervical epithelial
H8 cells (expressing low levels of the receptor). Au-
containing compounds, including auranon, inhibit
thioredoxin reductases (TRXR) and 3a-treated cells show
reduced TRXR enzymatic activity. This nding is
corroborated by docking studies showing that Au binds the
selenium of the TRXR. The inhibition of the detoxication
properties of TRXR stimulates ROS production and favors
G2/M cell-cycle arrest as well as apoptosis (e.g., reduced
Bcl2, increased Bax and alteration of the mitochondria
membrane potential). Gene expression analysis performed on
HeLa cells exposed to 3a shows a differential expression of genes
involved in ferroptosis, includingTXNRD1,HMOX1,SLC7A11,
GCLM, FTH1, FTL, GPX1, GPX1P1, and GPX4. The exposure of
HeLa cells to IR following 3a treatment stimulates DNA damage
(increased γ-H2AX levels) and downregulates GPX4 expression,
leading to reduced cell survival. This behavior is counteracted by
the exposure to N-acetyl L-cysteine or ferrostatin-1. The
radiosensitizing properties of 3a are conrmed in vivo in
zebrash transplanted with Hela cells exposed to the
combination 3a/IR.
Along this way, Hou et al. have reported the synthesis and the
antitumor properties of PBmB-DOX NPs (Hou et al., 2022). PBmB-DOX
include a core of Bi
2
S
3
covered by PEGylated doxorubicin (DOX).
Moreover, PBmB-DOX contain-Mn-O- bonds that are sensitive to
high GSH level, which is typical of the TME, allowing tumor
selectivity and release of DOX under acidic conditions. The PBmB-
DOX disassembling mediated by high levels of GSH favors the release of
Mn
2+
that, besides stimulating Fenton reaction and potentiating DOX-
mediated antitumor activity, allows magnetic resonance contrast
enhancement. The downregulation of GSH reduces 4T1 cell
proliferation, while no important changes are evidenced in the growth
of normal PBmB-DOX-treated HUVECs cells. The depletion of GSH
upon treatment reduces the expression of GPX4 and increases lipid
peroxidation. PBmB-DOX are more potent than free DOX on 4T1 cells
and, compared to the exposure to IR or PBmB-DOX as single treatments,
the combination PBmB-DOX/IR increases the amount of γ-H2AX foci.
In vivo studies in 4T1 tumor-bearing mice demonstrate that PBmB-DOX
are biocompatible with no important toxic effects reported for major
organs, and that they preferentially accumulate into the tumor. Longer
circulation time in plasma is reported for PBmB-DOX with respect to free
DOX, and PBmB-DOX more efciently suppress tumor growth with
respect to free DOX. The exposure to IR upon PBmB-DOX treatment
signicantly improves antitumor activity with no important signs of
toxicity. Histological analysis of tumors explanted from mice shows
increased expression of γ-H2AX and reduced GPX4 levels following
the exposure to the combination. Lastly, a remarkable signal
enhancement in tumor is evidenced in MRI after 6 h post-injection of
PBmB-DOX, thus conrming the theranostic properties of the nano-
system.
4 Ionizing radiation-associated gene
signatures
Recently, results from investigations focused on the studies of
ferroptosis-associated gene signatures for predicting radiotherapy
patient outcomes have been reported (Table 3).
Frontiers in Molecular Biosciences frontiersin.org10
Beretta and Zaffaroni 10.3389/fmolb.2023.1216733
The combination of temozolomide and radiotherapy is used
for the treatment of malignant glioblastoma (GBM). Though
effective, the combination is not curative and the identication
of radiosensitive-associated biomarkers is an urgent need for
predicting prognosis and therapy outcome. In the study by Xie
et al. (2022), expression proles of genes involved in radiation
response and ferroptosis-associated pathways of GBM patients
and healthy subjects from The Cancer Genome Atlas (TCGA)
database are analyzed. Among the differentially expressed genes
(DEGs) intersecting the two pathways, seven genes (MAPK1,
ZEB1, MAP1LC3A, HSPB1, CA9, STAT3, and TNFAIP3)
overlap and the analysis of the protein-protein interaction
network indicates STAT3 as the hub gene. The application of
the Least Absolute Shrinkage and Selection Operator (LASSO)
and Cox regression analysis denes a risk score that straties
patients in low- and high-risk groups. Patients in the high-risk
group show low overall survival (OS) and high mortality.
Receiver Operating Characteristic (ROC) curve and K-M
analysis conrm the power of the signature in predicting
patientssurvival. The prognostic model is validated by data
from Chinese Glioma Genome Atlas (CGGA) database used as
an external independent validation cohort. Functional
enrichment analyses dened by Gene Ontology (GO) and
Kyoto Encyclopedia of Genes and Genomes (KEGG) as well
as immune cell inltration patterns analysis from single-sample
Gene Set Enrichment Analysis (ssGSEA) allow the identication
of the most represented pathways in high-risk group, including
IL-17, cytokine-cytokine receptor interaction, TNF signaling
pathways, DCs, macrophages, Tumor-inltrating lymphocytes
(TIL) and Treg cells. In vitro experiments performed in
glioblastoma U87 and U251 cells treated with the
combination erastin/IR support the relationship between
radiosensitivity and ferroptosis.
Gene expression proles of breast cancer and normal tissues
as well as the survival and clinical information from TCGA
database are analyzed by Liu and colleagues (Liu et al., 2022).
Among the DEGs associated with ferroptosis, SLC7A11 is the
most upregulated in tumors compared to normal tissues.
Numerous clinic-pathologic properties associate with
SLC7A11 levels, including the expression of estrogen
receptor (ER). ER-positive tissues show lower levels of
SLC7A11 (e.g., increased ferroptosis) with respect to ER-
negative samples. The univariate Cox regression for OS
model demonstrates that high SLC7A11 levels associate with
worse OS. In vitro experiments carried out in a panel of breast
cancer cell lines (ER-positive MCF7 and ZR-75-1 as well as ER-
negative MDA-MB-231) treated with ferrostatin-1 or erastin in
combination with IR support the critical role played by
SLC7A11 in regulating IR-induced ferroptosis in ER-positive
cells. The study also shows a positive correlation between the
expression of estrogen receptor 1 (ESR1) and SLC7A11 and the
analysis by K-M predicts poor prognosis for patients with high
levels of ESR1. Molecularly, IR exposure stimulates the
expression of ESR1 that, in turn, increases SLC7A11 levels
attenuating ferroptosis. This nding is supported by the
observationthatuponESR1/SLC7A11knockdowninER-
TABLE 3 Gene signatures associated to ionizing radiation.
Gene signature Tumor types Cell line validation Pathway involved References
MAPK1 Malignant glioblastoma U87 IL-17 Xie et al. (2022)
ZEB1 U251 Cytokine-cytokine receptor interaction
MAP1LC3A TNF signaling pathways
HSPB1 DCs
CA9 Macrophages
STAT3 TIL
TNFAIP3 Treg cells
SCL7A11 Breast cancer MCF7 SLC7A11/ESR1 Liu et al. (2022)
ZR-75-1 SLC7A11/NEDD4L
MDA-MB-231
ACSL3 Prostate cancer No Epithelialmesenchymal transition Feng et al. (2022)
EPAS1 Allograft rejection
FASN Fc gamma R-mediated phagocytosis
GSTP1 TGF beta signaling
LDHB ECM receptor interaction
NEDD4L Adipocytokine signaling
Androgen response
Notch signalling
Frontiers in Molecular Biosciences frontiersin.org11
Beretta and Zaffaroni 10.3389/fmolb.2023.1216733
positive cells, IR-induced ferroptosis is enhanced.
Immunoprecipitation assay revels that no direct protein-
protein interaction occurs between ESR1 and SLC7A11.
Conversely, a protein interaction involving SLC7A11 and the
E3 ubiquitin ligase neural precursor cell expressed
developmentally downregulated gene 4-like (NEDD4L) is
critical for stimulating the proteasome-mediated degradation
of SLC7A11. Based on these ndings, the authors suggest that
two pathways, including ESR1/SLC7A11 and SLC7A11/
NEDD4L, control SLC7A11 level and regulate ferroptosis
induced by IR exposure.
By analyzing expression data (mRNA and lncRNA) from
Gene Expression Omnibus (GEO) database of normal and
prostate cancer tissues of patients treated with radical
radiotherapy and intersecting them with ferroptosis-related
genes, Feng and co-workers construct a gene signature,
including ACSL3, EPAS1, FASN, GSTP1, LDHB, and
NEDD4L. This signature allows the denition of a
ferroptosis-related gene prognostic index (FGPI) useful for
predicting biochemical recurrence (BR) and radiation
resistance of prostate cancer suffering patients (Feng et al.,
2022). FGPI allows the stratication of the patients in high-
and low-risk groups. Although ROC curve poorly discriminates
BR patients from patients who do not experience BR, it
evidences that FGPI potentially reects radiation resistance.
Indeed, compared to no BR patients, a signicant higher
FGPI is observed for BR patients treated with radical
radiotherapy. The application of the K-M curve shows that
FGPI is an independent risk factor for biochemically relapse
(BCR) and metastasis-free survival (MFS) in patients treated
with radical radiotherapy. Moreover, compared to low-risk,
high-risk group patients treated with radical prostatectomy
are at higher risk of metastasis. The application of the
GeneMANIA database and ceRNA network assigns a critical
role to lnRNAPART1 in controlling ACSL3 and
EPAS1 expression via a intricate crosstalk involving
60 different miRNAs. Gene Set Enrichment Analysis (GSEA)
shows differences in pathway enrichment in high-risk
(epithelialmesenchymal transition, allograft rejection, Fc
gamma R-mediated phagocytosis, TGF beta signaling
pathway, and extracellular matrix receptor interaction) with
respect to low-risk patients (adipocytokine signaling pathway,
androgen response and notch signalling). Drug and
immunologic analysis as well as TME analysis resulting from
the application of dedicated softwares, which consider the
expression of ACSL3 and EPAS1, underline potential
sensitivity to nine drugs (OSI-027, OSI-930, PAC-1, PHA-
793887, PI-103, PIK-93, SNX-2112, TPCA-1, and UNC0638)
for high-risk patients. Compared to no BR patients, BR patients
group shows lower expression levels of METTL14, which predict
sensitivity to methylating agents, and higher expression of
PDCD1LG2 (PD-L2) and CD96. However, only CD96 is
signicantly associated with BCR-free survival. Regarding the
results of TME analysis, cancer-related broblasts,
macrophages, stromal score, immune score, estimate score,
and tumor purity are risk factors for BCR closely associated
to BCR-free survival.
5 Conclusion
Although radiotherapy is the rstchoiceforthetreatmentof
different tumors types, the development of radiation resistance
impairs its effectiveness and medical strategies aimed at
overcoming this drawback are urgent. Among these strategies,
the combination of IR with ferrptosis inducers proved to synergize
thus potentiating radiotherapy. Moreover, since the activation of
defense cellular pathways in response to IR exposure attenuates
radiotherapy effectiveness, deeper investigations aimed at studying
the involved pathways as well as at developing ferroptosis inducers
or novel combinatorial strategies are intriguing ways to pave for
combating radiation resistance. Besides the use of small molecules
inducing ferroptosis, the combination of IR with nano-systems
endowed with both diagnostic and therapeutic potential is
promising. In spite of their capability to overcome the
resistance to apoptosis developed by tumors exposed to
conventional chemotherapeutics, ferroptosis inducers show
drawbacks typical of small molecules, including low solubility,
limited tumor targeting, and toxic side effects that have often
impeded their clinical evaluation. These drawbacks have been
tackled by NPs that, functioning like a Trojan horse, localize
into the tumors via the enhanced permeability retention effect and
are entered into the therapeutics armamentarium for ghting
tumors. NPs ameliorate the circulation time of encapsulated
drugs and stimulate anticancer immunity at the tumor site.
Specic decoration aimed at targeting peculiar tumor-expressing
molecules as well as exploiting non physiologic conditions typical
of TME potentiate the tumor selectivity of NPs. Tumor targeting is
also in part guaranteed by the local irradiation of the tumor. NPs
are designed to disassemble themselves under peculiar conditions
(e.g., the acidic pH as well as the presence of specicenzymes),
ameliorating the selectivity of the cargo release. Therefore, the
magnetic properties of the metal composing the structure of the
NPs account for their theranostics potential. Among the
combinations including small molecules here reported, only IR/
niraparib is currently under investigation in phase I-II clinical
trials (https://www.clinicaltrials.gov)intriplenegativebreast
cancer (NCT03945721 and NCT04837209), pancreatic tumors
(NCT04409002), prostate cancer (NCT04194554,
NCT04037254), glioblastoma (NCT05666349) and head and
neck squamous cell carcinoma (NCT05784012) patients.
Regarding the clinical studies exploiting NPs, although all the
NPs considered have been tested in vivo showing interesting
antitumor prole, only AGluX is under phase I-II clinical
evaluation in patients with brain tumors and brain metastasis,
gynecologic cancers, non small cell lung cancers, and pancreatic
cancers (NCT04899908, NCT03308604, NCT02820454,
NCT04789486, NCT03818386, and NCT04784221. https://www.
clinicaltrials.gov). Lastly, the continuous implementation of the
gene signatures predicting IR response via ferroptosis-
mediated cell death is expected to positively impact on patient`s
health.
In conclusion, despite the preclinical success achieved with
the combination strategies, additional efforts and clinical
investigations are required in the future to demonstrate their
safety prole as well as their antitumor effectiveness.
Frontiers in Molecular Biosciences frontiersin.org12
Beretta and Zaffaroni 10.3389/fmolb.2023.1216733
Author contributions
GB and NZ wrote the review manuscript. All authors
contributed to the article and approved the submitted version.
Funding
The APC was funded by Ricerca Corrente from Italian Ministry
of Health.
Acknowledgments
This work was supported by Ricerca Corrente funds from Italian
Ministry of Health.
Conict of interest
The authors declare that the research was conducted in the
absence of any commercial or nancial relationships that could be
construed as a potential conict of interest.
Publishers note
All claims expressed in this article are solely those of the
authors and do not necessarily represent those of their
afliated organizations, or those of the publisher, the
editors and the reviewers. Any product that may be
evaluated in this article, or claim that may be made by
its manufacturer, is not guaranteed or endorsed by the
publisher.
References
Adjemian, S., Oltean, T., Martens, S., Wiernicki, B., Goossens, V., Vanden Berghe, T.,
et al. (2020). Ionizing radiation results in a mixture of cellular outcomes including
mitotic catastrophe, senescence, methuosis, and iron-dependent cell death. Cell Death
Dis. 11, 1003. doi:10.1038/s41419-020-03209-y
Aubrey, B. J., Kelly, G. L., Janic, A., Herold, M. J., and Strasser, A. (2018). How does
p53 induce apoptosis and how does this relate to p53-mediated tumour suppression?
Cell Death Differ. 25, 104113. doi:10.1038/cdd.2017.169
Azzam, E. I., Jay-Gerin, J. P., and Pain, D. (2012). Ionizing radiation-induce d
metabolic oxidative stress and prolonged cell injury. Cancer Lett. 327, 4860. doi:10.
1016/j.canlet.2011.12.012
Baidoo, K. E., Yong, K., and Brechbiel, M. W. (2013). Molecular pathways: Targeted
α-particle radiation therapy. Clin. Cancer Res. 19, 530537. doi:10.1158/1078-0432.
CCR-12-0298
Bellezza, I., Giambanco, I., Minelli, A., and Donato, R. (2018). Nrf2-Keap1 signaling
in oxidative and reductive stress. Biochim. Biophys. Acta Mol. Cell Res. 1865, 721733.
doi:10.1016/j.bbamcr.2018.02.010
Bersuker, K., Hendricks, J. M., Li, Z., Magtanong, L., Ford, B., Tang, P. H., et al. (2019).
The CoQ oxidoreductase FSP1 acts parallel to GPX4 to inhibit ferroptosis. Nature 575,
688692. doi:10.1038/s41586-019-1705-2
Bieging, K. T., Mello, S. S., and Attardi, L. D. (2014). Unravelling mechanisms of p53-
mediated tumour suppression. Nat. Rev. Cancer 14, 359370. doi:10.1038/nrc3711
Chen, Q., Zheng, W., Guan, J., Liu, H., Dan, Y., Zhu, L., et al. (2023). SOCS2-enhanced
ubiquitination of SLC7A11 promotes ferroptosis and radiosensitization in
hepatocellular carcinoma. Cell Death Differ. 30, 137151. doi:10.1038/s41418-022-
01051-7
Chiang, S. K., Chen, S. E., and Chang, L. C. (2018). A dual role of heme oxygenase-1 in
cancer cells. Int. J. Mol. Sci. 20, 39. doi:10.3390/ijms20010039
Choudhary, S., Burns, S. C., Mirsaan, H., Li, W., Vo, D. T., Qiao, M., et al. (2020).
Genomic analyses of early responses to radiation inglioblastoma reveal new alterations
at transcription,splicing, and translation levels. Sci. Rep. 10, 8979. doi:10.1038/s41598-
020-65638-1
DHerde, K., and Krysko, D. V. (2017). Ferroptosis: Oxidized PEs trigger death. Nat.
Chem. Biol. 13, 45. doi:10.1038/nchembio.2261
Delaney, G., Jacob, S., Featherstone, C., and Barton, M. (2005). The role of
radiotherapy in cancer treatment: Estimating optimal utilization from a review of
evidence-based clinical guidelines. Cancer 104, 11291137. doi:10.1002/cncr.21324
Dixon, S. J., Lemberg, K. M., Lamprecht, M. R., Skouta, R., Zaitsev, E. M., Gleason, C.
E., et al. (2012). Ferroptosis: An iron-dependent form of nonapoptotic cell death. Cell
149, 10601072. doi:10.1016/j.cell.2012.03.042
Doll, S., and Conrad, M. (2017). Iron and ferroptosis: A still ill-dened liaison.
IUBMB Life 69, 423434. doi:10.1002/iub.1616
Doll, S., Freitas, F. P., Shah, R., Aldrovandi, M., da Silva, M. C., Ingold, I., et al. (2019).
FSP1 is a glutathione-independent ferroptosis suppressor. Nature 575, 693698. doi:10.
1038/s41586-019-1707-0
Feng, D., Shi, X., Xiong, Q., Zhang, F., Li, D., Wei, W., et al. (2022). A ferroptosis-
related gene prognostic index associated with biochemical recurrence and radiation
resistance for patients with prostate cancer undergoing radical radiotherapy. Front. Cell
Dev. Biol. 10, 803766. doi:10.3389/fcell.2022.803766
Feng, H., and Stockwell, B. R. (2018). Unsolved mysteries: How does lipid
peroxidation cause ferroptosis? PLoS Biol. 16, e2006203. doi:10.1371/journal.pbio.
2006203
Friedmann Angeli, J. P., Krysko, D. V., and Conrad, M. (2019). Ferroptosis at the
crossroads of cancer-acquired drug resistance and immune evasion. Nat. Rev. Cancer
19, 405414. doi:10.1038/s41568-019-0149-1
Galluzzi, L., Vitale, I., Aaronson, S. A., Abrams, J. M., Adam, D., Agostinis, P., et al.
(2018). Molecular mechanisms of cell death: Recommendations of the nomenclature
committee on cell death. Cell Death Differ. 25, 486541. doi:10.1038/s41418-017-0012-4
Gaschler, M. M., and Stockwell, B. R. (2017). Lipid peroxidation in cell death.
Biochem. Biophys. Res. Commun. 482, 419425. doi:10.1016/j.bbrc.2016.10.086
Hassannia, B., Vandenabeele, P., and Vanden Berghe, T. (2019). Targeting ferroptosis
to iron out cancer. Cancer Cell 35, 830849. doi:10.1016/j.ccell.2019.04.002
Hou, M., Zhu, K., Hu, H., Zheng, S., Wu, Z., Ren, Y., et al. (2022). Rapid synthesis of
yolk-shell-like nanosystem for MR molecular and chemo-radio sensitization. J. Control
Release 347, 5567. doi:10.1016/j.jconrel.2022.04.033
Hou, Y. K., Zhang, Z. J., Li, R. T., Peng, J., Chen, S. Y., Yue, Y. R., et al. (2023).
Remodeling the tumor microenvironment with core-shell nanosensitizer featuring
dual-modal imaging and multimodal therapy for breast cancer. ACS Appl. Mater
Interfaces 15, 26022616. doi:10.1021/acsami.2c17691
Hu, L., Wang, H., Huang, L., Zhao, Y., and Wang, J. (2016). Crosstalk between
autophagy and intracellular radiation response (Review). Int. J. Oncol. 49, 22172226.
doi:10.3892/ijo.2016.3719
Hu, W., Zhang, C., Wu, R., Sun, Y., Levine, A., and Feng, Z. (2010). Glutaminase 2, a
novel p53 target gene regulating energy metabolism and antioxidant function. Proc.
Natl. Acad. Sci. U. S. A. 107, 74557460. doi:10.1073/pnas.1001006107
Huang, R. X., and Zhou, P. K. (2020). DNA damage response signaling pathways and
targets for radiotherapy sensitization in cancer. Signal Transduct. Target Ther. 5, 60.
doi:10.1038/s41392-020-0150-x
Huang, W. M., Li, Z. X., Wu, Y. H., Shi, Z. L., Mi, J. L., Hu, K., et al. (2023). m6A
demethylase FTO renders radioresistance of nasopharyngeal carcinoma via promoting
OTUB1-mediated anti-ferroptosis. Transl. Oncol. 27, 101576. doi:10.1016/j.tranon.
2022.101576
Jaffray, D. A. (2012). Image-guided radiotherapy: From current concept to future
perspectives. Nat. Rev. Clin. Oncol. 9, 688699. doi:10.1038/nrclinonc.2012.194
Jennis, M., Kung, C. P., Basu, S., Budina-Kolomets, A., Leu, J. I., Khaku, S., et al.
(2016). An African-specic polymorphism in the TP53 gene impairs p53 tumor
suppressor function in a mouse model. Genes Dev. 30, 918930. doi:10.1101/gad.
275891.115
Jiang, L., Kon, N., Li, T., Wang, S. J., Su, T., Hibshoosh, H., et al. (2015). Ferroptosis as
a p53-mediated activity during tumour suppression. Nature 520, 5762. doi:10.1038/
nature14344
Jiang, W., Wei, L., Chen, B., Luo, X., Xu, P., Cai, J., et al. (2022). Platinum prodrug
nanoparticles inhibiting tumor recurrence and metastasis by concurrent
chemoradiotherapy. J. Nanobiotechnology 20, 129. doi:10.1186/s12951-022-01322-y
Kang, R., Kroemer, G., and Tang, D. (2019). The tumor suppressor protein p53 and
the ferroptosis network. Free Radic. Biol. Med. 133, 162168. doi:10.1016/j.
freeradbiomed.2018.05.074
Frontiers in Molecular Biosciences frontiersin.org13
Beretta and Zaffaroni 10.3389/fmolb.2023.1216733
Kryukov, G. V., Castellano, S., Novoselov, S. V., Lobanov, A. V., Zehtab, O., Guigó, R.,
et al. (2003). Characterization of mammalian selenoproteomes. Science 300, 14391443.
doi:10.1126/science.1083516
Lang, X., Green, M. D., Wang, W., Yu, J., Choi, J. E., Jiang, L., et al. (2019).
Radiotherapy and immunotherapy promote tumoral lipid oxidation and ferroptosis
via synergistic repression of SLC7A11. Cancer Discov. 9, 16731685. doi:10.1158/2159-
8290.CD-19-0338
Lee, H., Zandkarimi, F., Zhang, Y., Meena, J. K., Kim, J., Zhuang, L., et al. (2020).
Energy-stress-mediated AMPK activation inhibits ferroptosis. Nat. Cell Biol. 22,
225234. doi:10.1038/s41556-020-0461-8
Lei, G., Mao, C., Yan, Y., Zhuang, L., and Gan, B. (2021). Ferroptosis, radiotherapy,
and combination therapeutic strategies. Protein Cell 12, 836857. doi:10.1007/s13238-
021-00841-y
Lei, G., Zhang, Y., Koppula, P., Liu, X., Zhang, J., Lin, S. H., et al. (2020). The role of
ferroptosis in ionizing radiation-induced cell death and tumor suppression. Cell Res. 30,
146162. doi:10.1038/s41422-019-0263-3
Lei, G., Zhuang, L., and Gan, B. (2022). Targeting ferroptosis as a vulnerability in
cancer. Nat. Rev. Cancer 22, 381396. doi:10.1038/s41568-022-00459-0
Lei,P.,Bai,T.,andSun,Y.(2019).Mechanismsofferroptosisandrelationswith
regulated cell death: A review. Front. Physiol. 10, 139. doi:10.3389/fphys.2019.
00139
Li,Y.,Yang,J.,Gu,G.,Guo,X.,He,C.,Sun,J.,etal.(2022).Pulmonarydelivery
of theranostic nanoclusters for lung cancer ferroptosis with enhanced
chemodynamic/radiation synergistic therapy. Nano Lett. 22, 963972. doi:10.
1021/acs.nanolett.1c03786
Liang,C.,Zhang,X.,Yang,M.,andDong,X.(2019).Recentprogressin
ferroptosis inducers for cancer therapy. Adv. Mater 31, e1904197. doi:10.1002/
adma.201904197
Lin, L. S., Song, J., Song, L., Ke, K., Liu, Y., Zhou, Z., et al. (2018). Simultaneous fenton-
like ion delivery and glutathione depletion by MnO2 -based nanoagent to enhance
chemodynamic therapy. Angew. Chem. Int. Ed. Engl. 57, 49024906. doi:10.1002/anie.
201712027
Lin, Y., Chen, X., Yu, C., Xu, G., Nie, X., Cheng, Y., et al. (2023). Radiotherapy-
mediated redox homeostasis-controllable nanomedicine for enhanced ferroptosis
sensitivity in tumor therapy. Acta Biomater. S1742-7061 (23), 300311. doi:10.1016/
j.actbio.2023.01.022
Liu, J., An, W., Zhao, Q., Liu, Z., Jiang, Y., Li, H., et al. (2022). Hyperbaric oxygen
enhances X-ray induced ferroptosis in oral squamous cell carcinoma cells. Oral Dis.
2022. doi:10.1111/odi.14461
Liu, R., Liu, L., Bian, Y., Zhang, S., Wang, Y., Chen, H., et al. (2022). The dual
regulation effects of ESR1/nedd4l on SLC7A11 in breast cancer under ionizing
radiation. Front. Cell Dev. Biol. 9, 772380. doi:10.3389/fcell.2021.772380
Luo, H., Wang, X., Song, S., Wang, Y., Dan, Q., and Ge, H. (2022). Targeting stearoyl-
coa desaturase enhances radiation induced ferroptosis and immunogenic cell death in
esophageal squamous cell carcinoma. Oncoimmunology 11, 2101769. doi:10.1080/
2162402X.2022.2101769
Maier, P., HartmannWenz, L,F., and Herskind, C. (2016). Cellular pathways in
response to ionizing radiation and their targetability for tumor radiosensitization. Int.
J. Mol. Sci. 17, 102. doi:10.3390/ijms17010102
Mancias, J. D., Wang, X., Gygi, S. P., Harper, J. W., and Kimmelman, A. C. (2014).
Quantitative proteomics identies NCOA4 as the cargo receptor mediating
ferritinophagy. Nature 509, 105109. PMID: 24695223. doi:10.1038/nature13148
Mijit, M., Caracciolo, V., Melillo, A., Amicarelli, F., and Giordano, A. (2020). Role of
p53 in the regulation of cellular senescence. Biomolecules 10, 420. doi:10.3390/
biom10030420
Ou, Y., Wang, S. J., Li, D., Chu, B., and Gu, W. (2016). Activation of SAT1 engages
polyamine metabolism with p53-mediated ferroptotic responses. Proc. Natl. Acad. Sci.
U. S. A. 113, E6806E6812. doi:10.1073/pnas.1607152113
Reisz,J.A.,Bansal,N.,Qian,J.,Zhao,W.,andFurdui,C.M.(2014).Effectsof
ionizing radiation on biological molecules-mechanisms of damage and emerging
methods of detection. Antioxid. Redox Signal 21, 260292. doi:10.1089/ars.2013.
5489
Sabin, R. J., and Anderson, R. M. (2011). Cellular Senescence - its role in cancer and
the response to ionizing radiation. Genome Integr. 2, 7. doi:10.1186/2041-9414-2-7
Seibt, T. M., Proneth, B., and Conrad, M. (2019). Role of GPX4 in ferroptosis and its
pharmacological implication. Free Radic. Biol. Med. 133, 144152. doi:10.1016/j.
freeradbiomed.2018.09.014
Shah, R., Margison, K., and Pratt, D. A. (2017). The potency of diarylamine radical-
trapping antioxidants as inhibitors of ferroptosis underscores the role of autoxidation in
the mechanism of cell death. ACS Chem. Biol. 12, 25382545. doi:10.1021/acschembio.
7b00730
Sheikh, M. S., and Fornace, A. J. (2000). Death and decoy receptors and p53-mediated
apoptosis. Leukemia 14, 15091513. doi:10.1038/sj.leu.2401865
Shen, D., Luo, J., Chen, L., Ma, W., Mao, X., Zhang, Y., et al. (2022). PARPi treatment
enhances radiotherapy-induced ferroptosis and antitumor immune responses via the
cGAS signaling pathway in colorectal cancer. Cancer Lett. 550, 215919. doi:10.1016/j.
canlet.2022.215919
Song, X., Zhu, S., Chen, P., Hou, W., Wen, Q., Liu, J., et al. (2018). AMPK-mediated
BECN1 phosphorylation promotes ferroptosis by directly blocking system Xc- activity.
Curr. Biol. 28, 23882399. doi:10.1016/j.cub.2018.05.094
Stockwell, B. R., Friedmann Angeli, J. P., Bayir, H., Bush, A. I., Conrad, M., Dixon, S.
J., et al. (2017). Ferroptosis: A regulated cell death nexus linking metabolism, redox
biology, and disease. Cell 171, 273285. doi:10.1016/j.cell.2017.09.021
Sun, H., Cai, H., Xu, C., Zhai, H., Lux, F., Xie, Y., et al. (2022). AGuIX nanoparticles
enhance ionizing radiation-induced ferroptosis on tumor cells by targeting the NRF2-
GPX4 signaling pathway. J. Nanobiotechnology 20, 449. doi:10.1186/s12951-022-
01654-9
Tang, D., Kang, R., Berghe, T. V., Vandenabeele, P., and Kroemer, G. (2019). The
molecular machinery of regulated cell death. Cell Res. 29, 347364. doi:10.1038/s41422-
019-0164-5
Tang, M., Chen, Z., Wu, D., and Chen, L. (2018). Ferritinophagy/ferroptosis: Iron-
related newcomers in human diseases. J. Cell Physiol. 233, 91799190. doi:10.1002/jcp.
26954
Tarangelo, A., Magtanong, L., Bieging-Rolett, K. T., Li, Y., Ye, J., Attardi, L. D., et al.
(2018). p53 suppresses metabolic stress-induced ferroptosis in cancer cells. Cell Rep. 22,
569575. doi:10.1016/j.celrep.2017.12.077
Torti, S. V., and Torti, F. M. (2019). Winning the war with iron. Nat. Nanotechnol. 14,
499500. doi:10.1038/s41565-019-0419-9
Trujillo-Alonso, V., Pratt, E. C., Zong, H., Lara-Martinez, A., Kaittanis, C., Rabie, M.
O., et al. (2019). FDA-approved ferumoxytol displays anti-leukaemia efcacy against
cells with low ferroportin levels. Nat. Nanotechnol. 14, 616622. doi:10.1038/s41565-
019-0406-1
Vanden Berghe, T., Vanlangenakker, N., Parthoens, E., Deckers, W., Devos, M.,
Festjens, N., et al. (2010). Necroptosis, necrosis and secondary necrosis converge on
similar cellular disintegration features. Cell Death Differ. 17, 922930. doi:10.1038/cdd.
2009.184
Venkatesh, D., OBrien, N. A., Zandkarimi, F., Tong, D. R., Stokes, M. E., Dunn, D. E.,
et al. (2020). MDM2 and MDMX promote ferroptosis by PPARα-mediated lipid
remodeling. Genes Dev. 34, 526543. doi:10.1101/gad.334219.119
Wang, Y., Yang, L., Zhang, X., Cui, W., Liu, Y., Sun, Q. R., et al. (2019). Epigenetic
regulation of ferroptosis by H2B monoubiquitination and p53. EMBO Rep. 20, e47563.
doi:10.15252/embr.201847563
Wenzel, S. E., Tyurina, Y. Y., Zhao, J., St Croix, C. M., Dar, H. H., Mao, G., et al.
(2017). PEBP1 wardens ferroptosis by enabling lipoxygenase generation of lipid death
signals. Cell 171, 628641. doi:10.1016/j.cell.2017.09.044
Xie,L.,Song,X.,Yu,J.,Guo,W.,Wei,L.,Liu,Y.,etal.(2011).Solutecarrier
protein family may involve in radiation-induced radioresistance of non-small cell
lung cancer. J. Cancer Res. Clin. Oncol. 137, 17391747. doi:10.1007/s00432-011-
1050-9
Xie, Y., Xiao, Y., Liu, Y., Lu, X., Wang, Z., Sun, S., et al. (2022). Construction of a novel
radiosensitivity- and ferroptosis-associated gene signature for prognosis prediction in
gliomas. J. Cancer 13, 26832693. doi:10.7150/jca.72893
Yang, M., Wu, X., Hu, J., Wang, Y., Wang, Y., Zhang, L., et al. (2022).
COMMD10 inhibits HIF1α/CP loop to enhance ferroptosis and radiosensitivity by
disrupting Cu-Fe balance in hepatocellular carcinoma. J. Hepatol. 76, 11381150.
doi:10.1016/j.jhep.2022.01.009
Yang, W. S., Kim, K. J., Gaschler, M. M., Patel, M., Shchepinov, M. S., and Stockwell,
B. R. (2016). Peroxidation of polyunsaturated fatty acids by lipoxygenases drives
ferroptosis. Proc. Natl. Acad. Sci. U. S. A. 113, E4966E4975. doi:10.1073/pnas.
1603244113
Yang, W. S., and Stockwell, B. R. (2008). Synthetic lethal screening identies
compounds activating iron-dependent, nonapoptotic cell death in oncogenic-
RAS-harboring cancer cells. Chem. Biol. 15, 234245. doi:10.1016/j.chembiol.
2008.02.010
Yang, Z., Huang, S., Liu, Y., Chang, X., Liang, Y., Li, X., et al. (2022). Biotin-Targeted
Au(I) radiosensitizer for cancer synergistic therapy by intervening with redox
homeostasis and inducing ferroptosis. J. Med. Chem. 65, 84018415. doi:10.1021/
acs.jmedchem.2c00300
Frontiers in Molecular Biosciences frontiersin.org14
Beretta and Zaffaroni 10.3389/fmolb.2023.1216733
Ye, L. F., Chaudhary, K. R., Zandkarimi, F., Harken, A. D., Kinslow, C. J., Upadhyayula, P.
S., et al. (2020). Radiation-induced lipid peroxidation triggers ferroptosis and synergizes with
ferroptosis inducers. ACS Chem. Biol. 15, 469484. doi:10.1021/acschembio.9b00939
Yuan, H., Li, X., Zhang, X., Kang, R., and Tang, D. (2016). Identication of ACS L4 as a
biomarker and contributor of ferroptosis. Biochem. Biophys. Res. Commun. 478,
13381343. doi:10.1016/j.bbrc.2016.08.124
Zhang,Y.,Liu,X.,Zeng,L.,Zhao,X.,Chen,Q.,Pan,Y.,etal.(2022).Exosomalprotein
angiopoietin-like 4 mediated radioresistance of lung cancer by inhibiting ferroptosis under
hypoxic microenvironment. Br.J.Cancer127, 17601772. doi:10.1038/s41416-022-01956-7
Zhang, Y., Qian, Y., Zhang, J., Yan, W., Jung, Y. S., Chen, M., et al. (2017). Ferredoxin
reductase is critical for p53-dependent tumor suppression via iron regulatory protein 2.
Genes Dev. 31, 12431256. doi:10.1101/gad.299388.117
Zhang, Y., Shi, J., Liu, X., Feng, L., Gong, Z., Koppula, P., et al. (2018). BAP1 links
metabolic regulation of ferroptosis to tumour suppression. Nat. Cell Biol. 20,
11811192. doi:10.1038/s41556-018-0178-0
Zhao, J., Chen, Y., Xiong, T., Han, S., Li, C., He, Y., et al. (2023). Clustered cobalt
nanodots initiate ferroptosis by upregulating heme oxygenase 1 for radiotherapy
sensitization. Small 19, e2206415. doi:10.1002/smll.202206415
Zheng, J., and Conrad, M. (2020). The metabolic underpinnings of ferroptosis. Cell
Metab. 32, 920937. doi:10.1016/j.cmet.2020.10.011
Zhu, X., Wu, J., Liu, R., Xiang, H., Zhang, W., Chang, Q., et al. (2022). Engineering
single-atom iron nanozymes with radiation-enhanced self-cascade catalysis and self-
supplied H2O2 for radio-enzymatic therapy. ACS Nano 16, 1884918862. doi:10.1021/
acsnano.2c07691
Frontiers in Molecular Biosciences frontiersin.org15
Beretta and Zaffaroni 10.3389/fmolb.2023.1216733
... Different nanoplatforms based on iron, cobalt, gadolinium, bismuth, and gold have already been established as effective radiosensitizers and synergize with radiotherapy. [18][19][20][21][22][23][24][25] Computational modeling of radioenhancement has actively been conducted to predict nanoparticle therapy outcome. To correctly explain the increase in biological efficacy, several models have been developed. ...
Article
Full-text available
A study on the radiophysical dose enhancement and the intrinsic biological sensitization by gold and iron nanoparticles in A549 cancer cells.
... Továbbá az MCC950 és a nekroszulfonamid piroptózis-inhibitorok gátolják a magas glükóz/magas zsírtartalom által kiváltott ferroptózist in vitro (56). A ferroptózist számos szövetben lehetséges terápiás targetként írták le, így pl. a májban (57), a szívizomban (58), az idegrendszerben (59), de szerepet tulajdonítanak neki a daganatok terápiájában is (60,61). A fer ro p tó zis gyógyszeres gátlása a legfrissebb tanulmányok szerint segíthet a szív I/R-károsodás mérséklésében is. ...
Article
Az intenzív kutatások ellenére továbbra sem rendelkezünk olyan kardioprotektív gyógyszerekkel, amelyek a szív iszkémia/reperfúziós (I/R) károsodásával járó infarktusméretet hatásosan csökkentenék. Ennek egyik magyarázata, hogy az I/R-t kísérő sejtelhalás összetett folyamata teljesen még nem tisztázott. A mechanizmus részletesebb megismerése javíthatja a kardioprotektív gyógyszerfejlesztések transzlálhatóságát. A klasszikus szabályozott (apoptózis, autofágia-mediálta sejthalál) és nem szabályozott (nekrózis) sejthalálfolyamatok mellett olyan programozott sejthalálformákat és mechanizmusokat ismertünk meg az elmúlt években, mint például a piroptózis, a PANoptózis vagy a ferroptózis. Jelen összefoglaló közleményünkben ezen folyamatokat kívánjuk röviden bemutatni a szív I/R károsodásában, valamint kitérünk a lehetséges modulálási stratégiákra is.
... Cell death may occur by various mechanisms such as apoptosis, necrosis, mitotic catastrophe, ferroptosis, senescence, and autophagy [92][93][94]. Apoptosis as a programmed cell death usually occurs in multicellular organisms under normal conditions to maintain homeostasis by eliminating damaged cells. It is mediated by intrinsic and extrinsic mechanisms that activate those members of the proteases family that lead to the initiation and amplification of proteolytic cascades: the intrinsic and extrinsic ones. ...
Article
Full-text available
Radiotherapy (RT) is a major part of cancer treatment. The reported variability in patient response to this modality can interfere with the continuation of best-possible care, promote side effects, and lead to long-term morbidity. Tools to predict a patient’s response to radiation could be highly useful in improving therapeutic outcomes while minimizing unnecessary and toxic exposure to radiation. This study investigates the potential of using molecular biomarkers as predictors of radiosensitivity in clinical practice. We review relative studies researching the positive correlation between various molecular biomarkers and patient radiosensitivity, including DNA damage response and repair proteins, inflammation and apoptosis markers, cell cycle regulators, and other biological markers. The clinical perspectives and applicability of these biomarkers in the prediction of radiosensitivity are also critically discussed. Conclusively, we underline the dynamics of molecular biomarkers to improve the efficacy and safety of radiotherapy in clinical practice and highlight the need for further research in this field. Identification of the most prominent markers is crucial for the personalization of therapies entailing ionizing radiation.
... Additionally, ferroptosis can be triggered by the degradation of ferritin, known as ferritinophagy [23]. Dysregulated ferroptosis is implicated in various pathological conditions and human diseases [15,24,25], making it a promising avenue for cancer therapeutics due to the increased iron levels and susceptibility to ferroptosis induction in cancer cells [26][27][28]. ...
Article
Full-text available
Ferroptosis, a regulated cell death dependent on iron, has garnered attention as a potential broad-spectrum anticancer approach in leukemia research. However, there has been limited ferroptosis research on ATL, an aggressive T-cell malignancy caused by HTLV-1 infection. Our study employs bioinformatic analysis, utilizing dataset GSE33615, to identify 46 ferroptosis-related DEGs and 26 autophagy-related DEGs in ATL cells. These DEGs are associated with various cellular responses, chemical stress, and iron-related pathways. Autophagy-related DEGs are linked to autophagy, apoptosis, NOD-like receptor signaling, TNF signaling, and the insulin resistance pathway. PPI network analysis revealed 10 hub genes and related biomolecules. Moreover, we predicted crucial miRNAs, transcription factors, and potential pharmacological compounds. We also screened the top 20 medications based on upregulated DEGs. In summary, our study establishes an innovative link between ATL treatment and ferroptosis, offering promising avenues for novel therapeutic strategies in ATL.
Article
Full-text available
Copper plays vital roles in numerous cellular processes and its imbalance can lead to oxidative stress and dysfunction. Recent research has unveiled a unique form of copper-induced cell death, termed cuproptosis, which differs from known cell death mechanisms. This process involves the interaction of copper with lipoylated tricarboxylic acid cycle enzymes, causing protein aggregation and cell death. Recently, a growing number of studies have explored the link between cuproptosis and cancer development. This review comprehensively examines the systemic and cellular metabolism of copper, including tumor-related signaling pathways influenced by copper. It delves into the discovery and mechanisms of cuproptosis and its connection to various cancers. Additionally, the review suggests potential cancer treatments using copper ionophores that induce cuproptosis, in combination with small molecule drugs, for precision therapy in specific cancer types.
Article
Full-text available
Ferroptosis is a non-apoptotic form of regulated cell death characterized by the lethal accumulation of iron-dependent membrane-localized lipid peroxides. It acts as an innate tumor suppressor mechanism and participates in the biological processes of tumors. Intriguingly, mesenchymal and dedifferentiated cancer cells, which are usually resistant to apoptosis and traditional therapies, are exquisitely vulnerable to ferroptosis, further underscoring its potential as a treatment approach for cancers, especially for refractory cancers. However, the impact of ferroptosis on cancer extends beyond its direct cytotoxic effect on tumor cells. Ferroptosis induction not only inhibits cancer but also promotes cancer development due to its potential negative impact on anticancer immunity. Thus, a comprehensive understanding of the role of ferroptosis in cancer is crucial for the successful translation of ferroptosis therapy from the laboratory to clinical applications. In this review, we provide an overview of the recent advancements in understanding ferroptosis in cancer, covering molecular mechanisms, biological functions, regulatory pathways, and interactions with the tumor microenvironment. We also summarize the potential applications of ferroptosis induction in immunotherapy, radiotherapy, and systemic therapy, as well as ferroptosis inhibition for cancer treatment in various conditions. We finally discuss ferroptosis markers, the current challenges and future directions of ferroptosis in the treatment of cancer.
Article
Ferroptosis is an iron-dependent programmed cell death mode that is distinct from other cell death modes, and radiation is able to stimulate cellular oxidative stress and induce the production of large amounts of reactive oxygen radicals, which in turn leads to the accumulation of lipid peroxide and the onset of ferroptosis. In this review, from the perspective of the role of ferroptosis in generating a radiation response following cellular irradiation, the relationship between ferroptosis induced by ionizing radiation stress and the response to ionizing radiation is reviewed, including the roles of MAPK and Nrf2 signaling pathways in ferroptosis, resulting from the oxidative stress response to ionizing radiation, the metabolic regulatory role of the p53 gene in ferroptosis, and regulatory modes of action of iron metabolism and iron metabolism-related regulatory proteins in promoting and inhibiting ferroptosis. It provides some ideas for the follow-up research to explore the specific mechanism and regulatory network of ferroptosis in response to ionizing radiation.
Article
Full-text available
High cobalt (Co) levels in tumors are associated with good clinical prognosis. An anticancer regimen that increases intratumoral Co through targeted nanomaterial delivery is proposed in this study. Bovine serum albumin and cobalt dichloride are applied to prepare cobaltous oxide nanodots using a facile biomineralization strategy. After iRGD peptide conjugation, the nanodots are loaded into dendritic mesoporous silica nanoparticles, generating a biocompatible product iCoDMSN. This nanocomposite accumulates in tumors after intravenous injection by deep tissue penetration and can be used for photoacoustic imaging. Proteomics research and molecular biology experiments reveal that iCoDMSN is a potent ferroptosis inducer in cancer cells. Mechanistically, iCoDMSNs upregulate heme oxygenase 1 (HMOX1), which increases transferrin receptors and reduces solute carrier family 40 member 1 (SLC40A1), resulting in Fe²⁺ accumulation and ferroptosis initiation. Furthermore, upregulated nuclear factor erythroid 2‐related factor 2 (NRF2), arising from the reduction in Kelch‐like ECH‐associated protein 1 (KEAP1) expression, is responsible for HMOX1 enhancement after iCoDMSN treatment. Owing to intensified ferroptosis, iCoDMSN acts as an efficient radiotherapy enhancer to eliminate cancer cells in vitro and in vivo. This study demonstrates a versatile Co‐based nanomaterial that primes ferroptosis by expanding the labile iron pool in cancer cells, providing a promising tumor radiotherapy sensitizer.
Article
Full-text available
Radiotherapy is a valid treatment for nasopharyngeal carcinoma (NPC), and radioresistance is the main cause of local NPC treatment failure. However, the underlying mechanisms and valuable markers of radioresistance for NPC remain have not been established. In this study, we observed that the m6A mRNA demethylase fat mass and obesity-associated protein (FTO) was significantly upregulated in radioresistant NPC tissues and cells relative to parental radiosensitive NPC tissues and cells. FTO enhances radioresistance by repressing radiation-induced ferroptosis in NPC. Mechanistically, FTO acts as an m6A demethylase to erase the m6A modification of the OTUB1 transcript and promote the expression of OTUB1, thereby inhibiting the ferroptosis of cells induced by radiation and finally triggering the radiotherapy resistance of NPC. Furthermore, our in vivo experiment results showed that the FTO inhibitor, FB23-2, and the ferroptosis activator, erastin, altered tumor responsiveness to radiotherapy in NPC cell lines and patient-derived xenografts. Our findings reveal, for the first time, that FTO enhances NPC radiotherapy resistance by withstanding radiation-induced ferroptosis, suggesting that FTO may serve as a potential therapeutic target and valuable prognostic biomarker in patients with NPC.
Article
Full-text available
In the frame of radiotherapy treatment of cancer, radioresistance remains a major issue that still needs solutions to be overcome. To effectively improve the radiosensitivity of tumors and reduce the damage of radiation to neighboring normal tissues, radiosensitizers have been given increasing attention in recent years. As nanoparticles based on the metal element gadolinium, AGuIX nanoparticles have been shown to increase the radiosensitivity of cancers. Although it is a rare nanomaterial that has entered preclinical trials, the unclear biological mechanism hinders its further clinical application. In this study, we demonstrated the effectiveness of AGuIX nanoparticles in the radiosensitization of triple-negative breast cancer. We found that AGuIX nanoparticles increased the level of DNA damage by compromising the homologous recombination repair pathway instead of the non-homologous end joining pathway. Moreover, the results showed that AGuIX nanoparticles induced apoptosis, but the degree of apoptosis ability was very low, which cannot fully explain their strong radiosensitizing effect. Ferroptosis, the other mode of cell death, was also discovered to play a significant role in radiation sensitization, and AGuIX nanoparticles may regulate the anti-ferroptosis system by inhibiting the NRF2-GSH-GPX4 signaling pathway. Graphical Abstract
Article
Full-text available
Background Hypoxia-mediated radioresistance is a major reason for the adverse radiotherapy outcome of non-small cell lung cancer (NSCLC) in clinical, but the underlying molecular mechanisms are still obscure. Methods Cellular and exosomal ANGPTL4 proteins under different oxygen status were examined. Colony survival, lipid peroxidation and hallmark proteins were employed to determine the correlation between ferroptosis and radioresistance. Gene regulations, western blot and xenograft models were used to explore the underlying mechanisms of the role of ANGPTL4 in radioresistance. Results ANGPTL4 had a much higher level in hypoxic NSCLC cells compared to normoxic cells. Up- or down- regulation of ANGPTL4 positively interrelated to the radioresistance of NSCLC cells and xenograft tumours. GPX4-elicited ferroptosis suppression and lipid peroxidation decrease were authenticated to be involved in the hypoxia-induced radioresistance. ANGPTL4 encapsulated in the exosomes from hypoxic cells was absorbed by neighbouring normoxic cells, resulting in radioresistance of these bystander cells in a GPX4-dependent manner, which was diminished when ANGPTL4 was downregulated in the donor exosomes. Conclusion Hypoxia-induced ANGPTL4 rendered radioresistance of NSCLC through at least two parallel pathways of intracellular ANGPTL4 and exosomal ANGPTL4, suggesting that ANGPTL4 might applicable as a therapeutic target to improve the therapeutic efficacy of NSCLC.
Article
Full-text available
Radioresistance is a principal culprit for the failure of radiotherapy in hepatocellular carcinoma (HCC). Insights on the regulation genes of radioresistance and underlying mechanisms in HCC are awaiting for profound investigation. In this study, the suppressor of cytokine signaling 2 (SOCS2) were screened out by RNA-seq and bioinformatics analyses as a potential prognosis predictor of HCC radiotherapy and then were determined to promote radiosensitivity in HCC both in vivo or in vitro. Meanwhile, the measurements of ferroptosis negative regulatory proteins of solute carrier family 7 member 11 (SLC7A11) and glutathione peroxidase 4 (GPX4), intracellular lipid peroxidation and Fe2+ concentration suggested that a high level of ferroptosis contributed to the radiosensitization of HCC. Moreover, SOCS2 and SLC7A11 were expressed oppositely in HCC clinical tissues and tumour xenografts with different radiosensitivities. Mechanistically, the N-terminal domain of SLC7A11 was specifically recognized by the SH2-structural domain of SOCS2. While the L162 and C166 of SOCS2-BOX region could bind elongin B/C compound to co-form a SOCS2/elongin B/C complex to recruit ubiquitin molecules. Herein, SOCS2 served as a bridge to transfer the attached ubiquitin to SLC7A11 and promoted K48-linked polyubiquitination degradation of SLC7A11, which ultimately led to the onset of ferroptosis and radiosensitization of HCC. In conclusion, it was demonstrated for the first time that high-expressed SOCS2 was one of the biomarkers predicting radiosensitivity of HCC by advancing the ubiquitination degradation of SLC7A11 and promoting ferroptosis, which indicates that targeting SOCS2 may enhance the efficiency of HCC radiotherapy and improve the prognosis of patients.
Article
Ferroptosis has received increasing attentions in cancer therapy owing to its unique advantages over apoptosis. However, ferroptosis is governed by the efficiency of reactive oxygen species (ROS) production and the tumor cell antioxidant microenvironment that compromises therapeutic efficacy of ferroptosis. It is of great significance to develop a strategy that can both achieve high-efficiency ROS production and modulate tumor cell antioxidant microenvironment to amplify ferroptosis. However, until now, such a strategy has rarely been realized. Here, we, for the first time, reported a radiotherapy -mediated redox homeostasis-controllable nanomedicine for amplifying ferroptosis sensitivity in tumor therapy. The nanomedicine is constructed by co-assembling a ferroptosis inducer hemin and a thioredoxin 1 (Trx-1) inhibitor 1-methylpropyl 2-imidazolyl disulfide (PX-12) with human serum albumin. For our nanomedicine, hemin converts H2O2 to ROS via Fenton reaction to induce ferroptosis while PX-12 effectively inhibits the activity of antioxidant Trx-1 to suppress ROS depletion, resulting in amplified ferroptosis. Particularly, combining radiotherapy with the nanomedicine, radiotherapy depletes the other key antioxidant glutathione and generates additional radiotherapy-induced ROS, further boosting the ferroptosis effect. Therefore, our strategy can simultaneously ensure efficient ROS production and regulation of tumor cell antioxidant microenvironment, thereby enhancing efficacy of ferroptosis in tumor therapy. Our work offers an innovative approach to amplify ferroptosis sensitivity against tumors by simultaneously promoting ROS production and regulating redox homeostasis. STATEMENT OF SIGNIFICANCE: : The antioxidants such as thioredoxin 1 (Trx-1) and glutathione (GSH) in tumor cells, are significantly upregulated by the innate cancer cellular redox homeostasis, severely restricting the reactive oxygen species (ROS)-based therapy and compromising the effect of Fenton reaction-induced ferroptosis against tumors. It is urgent to develop a strategy to simultaneously achieve Fenton reaction-induced ferroptosis and regulate the cancer cellular redox homeostasis against upregulated levels of Trx-1 and GSH. A radiotherapy-mediated redox homeostasis-regulatable nanomedicine was designed for amplifying ferroptosis sensitivity in tumor therapy, where the therapeutic efficacy of ferroptosis against tumors can be significantly amplified by integrating Fenton reaction-induced and radiotherapy-induced ferroptosis as well as PX-12-enabled inhibition of antioxidant Trx-1 and radiotherapy-induced downregulation of antioxidant GSH levels.
Article
To improve the efficiency of radiation therapy (RT) for breast cancer, a designable multifunctional core-shell nanocomposite of FeP@Pt is constructed using Fe(III)-polydopamine (denoted as FeP) as the core and platinum particles (Pt) as the shell. The hybrid structure is further covered with hyaluronic acid (HA) to give the final nanoplatform of FeP@Pt@HA (denoted as FPH). FPH exhibits good biological stability, prolongs blood circulation time, and is simultaneously endowed with tumor-targeting ability. With CD44-mediated endocytosis of HA, FPH can be internalized by cancer cells and activated by the tumor microenvironment (TME). The redox reaction between Fe3+ in FPH and endogenous glutathione (GSH) or/and hydrogen peroxide (H2O2) initiates ferroptosis therapy by promoting GSH exhaustion and •OH generation. Moreover, FPH has excellent photothermal conversion efficiency and can absorb near-infrared laser energy to promote the above catalytic reaction as well as to achieve photothermal therapy (PTT). Ferroptosis therapy and PTT are further accompanied by the catalase activity of Pt nanoshells to accelerate O2 production and the high X-ray attenuation coefficient of Pt for enhanced radiotherapy (RT). Apart from the therapeutic modalities, FPH exhibits dual-modal contrast enhancement in infrared (IR) thermal imaging and computed tomography (CT) imaging, offering potential in imaging-guided cancer therapy. In this article, the nanoplatform can remodel the TME through the production of O2, GSH- and H2O2-depletion, coenhanced PTT, ferroptosis, and RT. This multimodal nanoplatform is anticipated to shed light on the design of TME-activatable materials to enhance the synergism of treatment results and enable the establishment of efficient nanomedicine.
Article
Objective: The objective of this study was to investigate the combined effect of X-ray radiation (IR) and hyperbaric oxygen (HBO) on oral squamous cell carcinoma (OSCC) cells and to explore the possible molecular mechanism. Methods: The OSCC cells were treated with or without IR, together with or without HBO co-exposure. Cells were transfected with specific plasmids using Lipofectamine 2000. The cell varieties, apoptosis markers and ferroptosis markers were determined by using appropriate method. OSCC xenograft mice model were categorized into several subgroups according to the specific treatement. GPX4 expressions were determined by Immunohistochemistry (IHC) in OSCC tissues, and were tested by ELISA in serums from OSCC patients. Results: The co-exposure of IR and HBO significantly strengthened the cytotoxicity of IR on SCC15-S cells in ferroptosis dependent manner. The regulated GPX4/ferroptosis mediated the HBO function on re-sensitizing the radio-resistant OSCC cells to IR. In xenograft mice, co-exposure of IR and HBO can significantly reduce the tumor under IR activation compared with IR alone. Clinical data indicated that high GPX4 levels were associated with poor chemo-radiotherapy outcome. Conclusions: HBO could re-sensitize radio-resistant OSCC cells through GPX4/ferroptosis regulation. These results provide a potential therapeutic strategy for clinical radio-resistance.
Article
Single-atom nanozymes (SAzymes), with individually isolated metal atom as active sites, have shown tremendous potential as enzyme-based drugs for enzymatic therapy. However, using SAzymes in tumor theranostics remains challenging because of deficient enzymatic activity and insufficient endogenous H2O2. We develop an external-field-enhanced catalysis by an atom-level engineered FeN4-centered nanozyme (FeN4-SAzyme) for radio-enzymatic therapy. This FeN4-SAzyme exhibits peroxidase-like activity capable of catalyzing H2O2 into hydroxyl radicals and converting single-site FeII species to FeIII for subsequent glutathione oxidase-like activity. Density functional theory calculations are used to rationalize the origin of the single-site self-cascade enzymatic activity. Importantly, using X-rays can improve the overall single-site cascade enzymatic reaction process via promoting the conversion frequency of FeII/FeIII. As a H2O2 producer, natural glucose oxidase is further decorated onto the surface of FeN4-SAzyme to yield the final construct GOD@FeN4-SAzyme. The resulting GOD@FeN4-SAzyme not only supplies in situ H2O2 to continuously produce highly toxic hydroxyl radicals but also induces the localized deposition of radiation dose, subsequently inducing intensive apoptosis and ferroptosis in vitro. Such a synergistic effect of radiotherapy and self-cascade enzymatic therapy allows for improved tumor growth inhibition with minimal side effects in vivo. Collectively, this work demonstrates the introduction of external fields to enhance enzyme-like performance of nanozymes without changing their properties and highlights a robust therapeutic capable of self-supplying H2O2 and amplifying self-cascade reactions to address the limitations of enzymatic treatment.
Article
In cancer cells, poly(ADP-ribose) polymerase (PARP)-1 and PARP2 initiate and regulate DNA repair pathways to protect against DNA damage and cell death caused by radiotherapy or chemotherapy. Radiotherapy and PARP inhibitors (PARPis) have been combined in clinical trials, but their action mechanisms remain unclear. Here, we show that activated by ionizing radiation (IR) generated dsDNA, cyclic GMP-AMP synthase (cGAS) signaling promoted regulated cell death, specifically ferroptosis, via the activating transcription factor 3 (ATF3)–solute carrier family 7 member 11 axis and the antitumor immune response via the interferon-β-CD8⁺ T cell pathway. Niraparib, a widely used PARPi, augmented cGAS-mediated ferroptosis and immune activation. In colorectal cancer models, cGAS knockdown (KD) compromised IR-induced ferroptosis via downregulation of ATF3 (key ferroptosis regulator) expression. cGAS depletion reversed IR-induced infiltration of CD8⁺ T or CD8⁺GZMB⁺ T cells in the cGAS KD group. Survival analysis of paired tumor samples before and after standard radiotherapy revealed that high expression levels of cGAS, ATF3, and PTGS2 and high density of CD8⁺ T cells resulted in a significantly high disease-free survival rate in patients with rectal cancer. Therefore, PARPi treatment increases the cytoplasmic accumulation of dsDNA caused by IR, triggering the cGAS signaling-mediated tumor control in cancer cell lines and mouse xenograft models.