Open Access

Cross‑link between ferroptosis and nasopharyngeal carcinoma: New approach to radiotherapy sensitization (Review)

  • Authors:
    • Hai-Long Li
    • Nian-Hua Deng
    • Jia-Xin Xiao
    • Xiu-Sheng He
  • View Affiliations

  • Published online on: September 9, 2021     https://doi.org/10.3892/ol.2021.13031
  • Article Number: 770
  • Copyright: © Li et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Ferroptosis is a recently discovered special type of regulated cell death that is strongly associated with both homeostasis maintenance and cancer development. Previous studies have indicated that a number of small‑molecular agents inducing ferroptosis have great potential in the treatment of different types of cancer, including breast, pancreatic, prostate and head and neck cancer. However, the role of ferroptosis in nasopharyngeal carcinoma (NPC) has remained to be fully determined. To the best of our knowledge, no review of the currently available studies on this subject has been published to date. The metabolism and expression of specific genes that regulate ferroptosis may represent a promising radiosensitization target in cancer treatment. The aim of the present review was to describe the cross‑link between ferroptosis and NPC and to discuss the potential value of regulators and the possible mechanism underlying the role of ferroptosis in the radiosensitization of NPC, in the hope that linking the mechanism of ferroptosis with the development of NPC will accelerate the development of novel ferroptosis‑based targets and radiotherapy strategies in NPC.

Introduction

Nasopharyngeal carcinoma (NPC), an important type of head and neck cancer, is highly prevalent in East Africa and Asia, particularly in southern China (1,2). Due to the concealed location of NPC, surgical treatment is relatively difficult. According to the National Comprehensive Cancer Network guidelines, radiotherapy (RT) is the primary treatment of choice for NPC (3). RT uses an appropriate intensity of ionizing radiation (IR) to eliminate tumor cells (4,5). IR directly causes DNA damage and indirectly stimulates the production of reactive oxygen species (ROS) in tumor cells (6,7). However, according to statistics, 10–20% of patients with NPC suffer from recurrence after primary RT due to radiation resistance (8). Therefore, there is an urgent requirement to discover novel methods of radiosensitization for patients with resistance to RT.

Ferroptosis, a newly identified type of regulated cell death (RCD), was proposed by Dixon et al (9) in 2012. Unlike other types of RCD, ferroptosis is characterized by loss of lipid peroxidation repair ability and the accumulation of redox-active iron (10). Morphologically, the mitochondrial cristae decrease in number or disappear, the outer mitochondrial membrane ruptures and the mitochondrial membrane becomes condensed. Although the mechanism of ferroptosis has yet to be fully elucidated, ferroptosis has a key role in a number human diseases, such as ischemia/reperfusion injury (11), neurodegeneration (12) and various types of cancer, including NPC (13,14). From the perspective of radiosensitization and side effects of RT, the pharmacological modulation of ferroptosis (stimulation or inhibition) may be of significant clinical value.

The aim of the present review article was to discuss the potential molecular mechanisms of ferroptosis and the microRNAs (miRNAs/miRs) regulating ferroptosis in NPC, alongside the potential future directions and clinical value of ferroptosis research in RT for NPC.

Molecular mechanism of ferroptosis in cancer

Research has revealed three critical pathways involved in ferroptosis, which are the iron metabolism pathway, the polyunsaturated fatty acid (PUFA) metabolism pathway and the phospholipid hydroperoxidase glutathione peroxidase (GPX)4 metabolism pathway (Fig. 1) (15). Iron metabolism is a redox reaction of iron in the cytoplasm. During this process, ferric iron (Fe3+) is absorbed into the cytoplasm via transferrin and is then rapidly transformed into ferrous iron (Fe2+) and stored as ferritin or in the labile iron pool; however, Fe2+ is released due to the destruction of ferritin via ferritinophagy, a process mediated by nuclear receptor coactivator 4 (14,16). Finally, excessive Fe2+ is oxidized through the Fenton reaction by PUFA-containing phospholipids (PUFA-PL), generating a large amount of ROS and subsequently resulting in ferroptosis (17). PUFA-PL is the stress form of PUFA acetylated by acyl-CoA synthetase and activated by acyl-CoA synthetase long chain family member 4 (ACSL4) and lysophosphatidylcholine acyltransferase 3, and it represents the main lipid source of ROS (18,19). ROS are a group of molecules with partially reduced oxygen, including free radicals (HO• and RO•), peroxides (H2O2 and ROOH) and superoxide (O2•), which cause cell death by damaging DNA, RNA and lipid molecules (20). Therefore, PUFA metabolism has an oxidative role in ferroptosis. GPX4 metabolism is key for antagonizing lipid ROS due to the fact that GPX4 is able to catalyze the decomposition of H2O2 and complex lipid peroxides (21). Yang et al (22) reported that GPX4 is able to convert reduced glutathione (GSH) to oxidized glutathione (GSSG), leading to weakened Ras-selective lethal small molecule 3 (RSL3)- and erastin-induced ferroptosis. Mechanistically, several molecules are involved in GPX4 metabolism in ferroptosis. Among those, system Xc and GPX4 are considered as the main regulators of GPX4 metabolism, negatively regulating ferroptosis (23). System Xc is a membrane Na+-dependent cysteine-glutamate transporter, which is a disulphide-linked heterodimer composed of a light-chain subunit [solute carrier (SLC)7A11] and a heavy-chain subunit (SLC3A2) (24). Cysteine and glutamate are important elements in GSH synthesis, and GSH generation and maintenance are key to preventing oxidative damage due to lack of GPX4 (25). In addition, the mitochondrion is the most important organelle involved in ferroptosis, as the energy and electron transfer provided by the electron transfer chain are necessary in the process of ferroptosis (26). Mitochondrial voltage-dependent anion channels (VDACs), the transmembrane channels for transporting ion and metabolites, are widely distributed on the outer mitochondrial membrane (27). It was previously demonstrated that erastin is able to target VDAC2 on the outer mitochondrial membrane, resulting in lipid ROS release and slowing down the oxidation of NADH (28). NADH is mainly involved in material and energy metabolism in cells, which supplies the energy required for ATP synthesis through the oxidative phosphorylation process and for the conversion of GSSH to GSH (14,29). Therefore, VDACs and NADPH oxidase (NOX) are crucial positive regulators that promote ferroptosis, and altering outer mitochondrial membrane permeability by antitumor drugs may be a novel approach to tumor treatment. In summary, ferroptosis is a non-apoptotic type of cell death that is involved in several complex regulatory and three intersecting metabolic pathways.

Potential role of ferroptosis in NPC radiosensitization

RT-induced ferroptosis

A number of pharmacological studies have attempted to promote ferroptosis through various methods to improve the efficacy of RT (30,31). However, to date, the association between ferroptosis and RT has not been studied in depth. IR randomly causes oxidative damage in all intercellular spaces, including lipid membranes, and ferroptosis is caused by the accumulation of toxic lipid peroxidation products (31). Therefore, there may be an interesting connection between ferroptosis and IR. According to various studies, RT affects the four key regulators of ferroptosis, namely ROS, SLC7A11, ACSL4 and GPX4 (Fig. 2). Among those, ROS is considered the most important factor implicated in ferroptosis caused by RT. Ye et al (32) reported that IR acting synergistically with ferroptosis inducers increased ROS levels, leading to lipid oxidation in an in vitro study using the HT-1080 human fibrosarcoma cell line. However, regarding the expression of SLC7A11, the conclusions have been controversial among studies. An in vitro study involving the irradiation of ovarian cancer, melanoma and human fibrosarcoma cells demonstrated that the ataxia-telangiectasia mutated gene (ATM) activated by RT and IFN derived from activated CD8+ T cells synergistically inhibited the expression of SLC7A11 (30). However, Lei et al (33) indicated that IR markedly induced the expression of SLC7A11, GPX4 and ACSL4. Subsequently, the expression of SLC7A11 was considered as an adaptive response (34) and it likely involves activating transcription factor 4 and/or the transcription factor NF-E2-related factor 2 (NRF2), both of which are known to regulate SLC7A11 transcription and are largely activated by IR (3537). Therefore, it appears that RT is able to either repress or activate SLC7A11 expression, depending on the conditions. Mechanistically, there are three pathways involved in RT-induced ferroptosis (3033). First, RT is able to induce oxidative stress and then produce a large amount of ROS, leading to lipid peroxidation. In addition, RT may promote PUFA-PL biosynthesis by upregulating ACSL4 expression. Furthermore, RT also promotes GPX4-mediated ferroptosis through DNA damage and inhibition of GSH production. Taken together, these results indicated that the specific mechanisms of RT-induced ferroptosis require to be further explored and inducing ferroptosis to eliminate the radiation resistance of tumor cells may be a direction worthy of further investigation. These findings indicate the potential therapeutic value of targeting ferroptosis to enhance the radiosensitivity of NPC.

Crosstalk between ferroptosis and other types of RT-induced cell death

As described in the previous section, the major cellular effect triggered by RT is to damage DNA and induce ROS generation in cells. With regard to signaling, there appear to be interactions between ferroptosis and other types of radiation-induced cell death. RT damages DNA in the nucleus and, thus, activates ATM (38). ATM is the major regulator of the first step in DNA damage response sensing (39). ATM activation is able to sensitize AMP-activated protein kinase, which promotes Beclin 1 (BECN1)-mediated autophagy, while at the same time ferroptosis is promoted through regulating the ATM/GPX4 axis (31,40). A previous study indicated that RT is able to directly induce tumor cell necroptosis, which displays a certain overlap with apoptosis (41). The intrinsic apoptotic pathway is initiated by identifying double-strand breaks (DSBs) if DNA repair is not successful (42). Of note, ROS accumulation is able to prevent DNA repair and promote ferroptosis (7). In brief, multiple lines of evidence suggest that there is a close association between ferroptosis and other types of RT-induced cell death, particularly apoptosis, necroptosis and autophagic cell death.

Targeting ferroptosis to sensitize NPC to RT

RT currently remains the first choice of treatment for NPC. However, radiation resistance has come to represent a serious problem, as tumor cells are not sensitive to other forms of death, including apoptosis. Therefore, inducing ferroptosis of tumor cells may represent a good target for the radiosensitization of NPC. RT causes DNA DSBs in tumor cells. Furthermore, a large number of ROS are produced during the process of ferroptosis, which makes it difficult to repair the DNA double strand, thus further accelerating cell death (43). Acquired radioresistance is currently a long-standing challenge in RT for NPC. In view of this fact, the landing points for investigating the therapeutic relevance of ferroptosis in RT include the following: i) Whether the regulators of ferroptosis modulate radiosensitivity in NPC; and ii) how to target the regulators in NPC. These points are discussed below.

Iron

As the term suggests, the occurrence of ferroptosis requires high levels of intracellular iron (9). In the process of ferroptosis, numerous ROS-forming or -decomposing enzymes (cytochrome P450, xanthine oxidase, lipoxygenase, NOX, mitochondrial complex I and III, catalase and peroxidases) are iron-dependent (44,45). Imbalances in iron metabolism in cells lead to iron overload and ROS accumulation, resulting in Fenton oxidation reaction on the lipid membrane and, eventually, ferroptosis (46). Therefore, iron is able to amplify the production of ROS in ferroptosis (47,48). The conversion process between Fe3+ and Fe2+ is accompanied by the generation of energy, which benefits cellular energy metabolism (49). Similarly, it was demonstrated that reducing the level of intracellular iron via the tumor suppressor gene 3-hydroxybutyrate dehydrogenase type 2 inhibited the proliferation and metastasis of NPC cells (50). Furthermore, Xu et al (51) indicated that itraconazole was able to reduce the activity of NPC stem cells by increasing the concentration of intracellular iron in lysosomes and lipid peroxides. Therefore, the disruption of intracellular iron balance may affect NPC cell survival and proliferation (52). Of note, an in vivo study suggested that long-term treatment with iron-containing water improved the efficiency of RT for glioma in rats via ferroptosis (53). This method may also be applied to RT for NPC. When ferritin is degraded via ferritinophagy, it releases iron and promotes ferroptosis (54,55). Previous studies suggested that transferrin and its receptor promote ferroptotic cell death, whereas iron chelators inhibit this type of cell death (5658). Serum ferritin is the best single marker reflecting iron stores in vivo (59). Compared with that of healthy individuals, the serum ferritin level of patients with undifferentiated NPC was reported to be higher (60). It was previously indicated that serum ferritin levels may be valuable for predicting distant metastasis in patients with NPC following standard intensity-modulated RT and chemotherapy (61). Lactotransferrin (LTF), a member of the transferrin family, may negatively regulate the development and metastasis of NPC in vivo (62). It has been reported that LTF is highly expressed in NPC cells and overexpression of LTF inhibited the proliferation of NPC cells by modulating the MAPK/AKT pathway, which is an essential pathway for tumor radiosensitization (6366). Therefore, this may be a viable strategy for promoting radiosensitization of NPC through disrupting iron metabolism.

NRF2

NRF2 is considered as a main regulator of the antioxidant response in ferroptosis, as a number of its downstream target genes are responsible for preventing redox imbalance in cancer cells (67). Qiang et al (68) indicated that NRF2 serves a protective role in ferroptosis-mediated ischemia/reperfusion-induced acute lung injury by regulating SLC7A11 and activating STAT3. P62 may promote this process by preventing NRF2 degradation and then increasing NRF2 nuclear accumulation through inhibiting kelch-like ECH-associated protein 1 (KEAP1), which is able to regulate the expression of NRF2 via the ubiquitin-proteasome route (69,70). NRF2 was observed to be markedly upregulated in NPC tissues and may serve as an unfavorable prognostic biomarker in patients with NPC (71). An in vitro study suggested that NRF2 gene knockout enhanced the radiosensitivity of NPC cells, whereas silencing KEAP1 inhibited the radiosensitivity of NPC cells (72). In addition, Zhang et al (73) reported that lowering NRF2 levels and promoting ROS production sensitized NPC cells to RT. Huang et al (71) indicated that NRF2 expression was upregulated through the Raf kinase inhibitor protein/miR-450b-5p/NRF2/NAD(P)H:quinone oxidoreductase 1 axis, which improved the radioresistance of NPC. Another study also demonstrated that NRF2 promoted the proliferation of Epstein-Barr virus (EBV)-transformed B cells through the EBV-related proteins LMP1 and 2A and AKT signaling, which indicated that NRF2 may represent a potential molecular target for EBV-related diseases, including NPC (74).

GSH

GSH is able to protect lipid membranes by scavenging ROS (75). Under normal physiological conditions, the concentration of GSSG is 10–100 times lower compared with that of GSH; however, under conditions of severe oxidative stress, GSH is transformed into GSSG (76). GSH is an important cofactor of GPX4 and its biosynthesis is accomplished with the help of system Xc (77). The functions of GSH include inactivation of dangerous endogenous compounds and/or detoxification of exogenous compounds through the action of GPXs and GSH-S-transferases (78). Xu et al (51) demonstrated that NPC spheroids displayed a certain degree of ferroptosis resistance due to increased GSH levels. The metabolism of free radicals is disrupted and the effectiveness of the antioxidant defense system decreases significantly in patients with NPC. GSH has been indicated to have an antiapoptotic role in response to radiation via decreasing ROS production and inhibiting the MAPK pathway in NPC cells (79).

Fanconi anemia group D2 protein (FANCD2)

FANCD2, a negative regulator of ferroptosis, is able to repair DNA damage as a nuclear protein in bone marrow stromal cells (80). Knockout of FANCD2 may influence iron and GPX4 metabolism. In addition, an in vitro and in vivo study revealed that FANCD2 silencing enhanced the sensitivity of NPC cells to ionizing radiation (81). Recent results have demonstrated that FANCD2 expression is associated with the prognosis of NPC (82). Therefore, FANCD2 may be an effective target for radiosensitization, as well as a prognostic and diagnostic marker of NPC.

Heme oxygenase 1 (HO-1)

HO-1 may be regulated by NRF2 and endoplasmic reticulum-associated protein degradation and has a dual role in ferroptosis (10). On the one hand, increased expression of HO-1 may increase intracellular iron levels; on the other hand, HO-1 was able to attenuate erastin-induced ferroptosis in renal epithelial cells (83,84). A study on the association between NPC and HO-1 suggested that patients with low expression levels of HO-1 were more sensitive to RT compared with those with high expression levels of HO-1 (85). The results suggested that HO-1 may be a useful indicator for identifying patients with RT-sensitive NPC. Therefore, HO-1, as a regulator of ferroptosis, may also be an important target for radiosensitization.

p53

p53, a key tumor suppressor gene, is activated under different stress stimuli, including IR. p53 is able to transcriptionally inhibit SLC7A11 expression to impair cysteine import, ultimately promoting ferroptosis (86). p533KR, an acetylation-defective p53 mutant, is highly effective in repressing the expression of SLC711A, but not that of other already known p53 target genes (cell cycle-, apoptosis- or senescence-related genes) (87). However, it has also been reported that p53 may inhibit ferroptosis by the transcriptional activation of cyclin-dependent kinase (CDK) inhibitor 1A/p21 or inhibition of dipeptidyl-peptidase 4 activity (88). Therefore, p53 appears to have a dual role in ferroptosis. Previous studies have indicated that p53 also has a key role in regulating the occurrence and development of NPC, particularly in terms of its radiosensitivity. Wang et al (89) reported that activating the p53 signaling pathway via overexpressing miR-372 enhanced the radiosensitivity of NPC. Furthermore, a clinical study suggested that recombinant human adenovirus p53 promoted radiosensitivity in patients with recurrent NPC (90).

BECN1

BECN1, a key regulator of autophagy, is able to block the activity of system Xc via combining with SLC7A11 to promote ferroptosis in cancer cells (91,92). A randomized controlled trial indicated that BECN1 and hypoxia-inducible factor (HIF)-1α expression exhibited a positive association and that HIF-1α-associated high BECN1 expression promoted NPC cell survival after chemoradiotherapy (93). Of note, another previous study suggested that increasing HIF-1α stability promoted radiosensitivity in NPC (94). Therefore, there appears to be a close association among BECN1, HIF-1α and radiosensitivity in NPC.

miRNAs regulating ferroptosis

The major function of miRNAs is to bind to the 3′-untranslated region of target mRNAs and subsequently inhibit their expression (95). Previous studies have revealed that miRNAs have an important role in the regulation of ferroptosis. miR-182-5p and miR-324-3p were demonstrated to promote ferroptosis via targeting GPX4 in ischemia/reperfusion-induced renal injury and lung adenocarcinoma (96,97). miR-17-92 and miR-424-5p abrogated erastin- and RSL3-induced ferroptosis through targeting ACSL4 in human umbilical vein endothelial cells and ovarian cancer cells (98,99). In radioresistant cells, miR-7-5p restrained ferroptosis through downregulating mitoferrin and subsequently reducing iron levels (100). Furthermore, miR-9 and miR-137 promoted ferroptosis via reducing intracellular GSH levels; miR-9 inhibited the synthesis of GSH and miR-137 suppressed the expression of SLC1A5, which is a component of system Xc (101,102). To date, several studies have demonstrated that miRNAs regulating ferroptosis are associated with the proliferation, invasion, migration and apoptosis of NPC cells, particularly in terms of radiosensitivity regulation (Table I). For instance, miR-214 and miR-182-5p were indicated to contribute to radioresistance in NPC by regulating LTF and BNIP3 expression (103,104). However, miR-124 and miR-9 may promote radiosensitivity of NPC via targeting programmed cell death protein 6 (PDCD6) and suppressing the expression of junctional adhesion molecule A (JAMA) (105107). Hu et al (108) indicated that miR-214 enhanced radiosensitivity of colorectal cancer via inhibition of autophagy-related 12-mediated autophagy. Based on the results reported to date, miR-214 is considered to act as an oncogene in NPC, which is able to promote the proliferation of NPC cells and inhibit apoptosis by targeting BAX, LTF, Bcl-2-like protein 11, WW domain-containing oxidoreductase and phosphatase and tensin homolog (109112). The expression levels of miR-214 were indicated to be upregulated in NPC, particularly in metastasis-prone NPC tissues compared with those in normal nasopharyngeal epithelial tissues (112). Therefore, miR-214 may serve as a potential novel diagnostic and RT sensitization biomarker for NPC. miR-124 has been detected in copious amounts in the brain and it may participate in the pathogenesis of several disorders. Deng et al (113) suggested that miR-124 was able to radiosensitize glioblastoma multiforme cells by targeting CDK4. In addition, miR-124 has been reported to be downregulated in NPC (114). miR-124 may enhance cell radiosensitivity by targeting JAMA and PDCD6 (105). Current research suggested that miR-124 is able to suppress stem-like properties and enhance radiosensitivity in NPC cells by directly targeting JAMA (106). These results may provide novel insight into the molecular mechanisms underlying RT failure in NPC and enable the design of novel therapeutic approaches (115,116).

Table I.

Summary of miRNAs regulating ferroptosis in NPC.

Table I.

Summary of miRNAs regulating ferroptosis in NPC.

miRNATarget of ferroptosisFunction in NPCTarget gene/pathway in NPC(Refs.)
miR-214TFR1 TP53Proliferation (+); Apoptosis (−)WWOX, PTEN, Bim, Bax, LTF(115)
miR-182-5pGPX4Apoptosis (−)BNIP3(96)
miR-124 Fe2+Proliferation (−); Apoptosis (+); Invasion (−)Wnt, PDCD6, JAMA, Foxq1, NF-κB(116)
miR-9GSHProliferation (−); Apoptosis (+); Invasion (−)MDK, PDK/AKT, CXCR4, GSH(101)
miR-424-5pACSL4Proliferation (−); Apoptosis (+); Invasion (−)AKT3(99)
miR-7-5p Fe2+Apoptosis (−)ENO2(100)
miR-324-3pGPX4Proliferation (−); Apoptosis (+); Invasion (−)WNT2B, GLI3(97)

[i] (−) was used to indicate inhibition and (+) was used for promotion. miRNA/miR, microRNA; NPC, nasopharyngeal carcinoma; WWOX, WW domain-containing oxidoreductase; LTF, lactotransferrin; BNIP3, Bcl-2/adenovirus E1B 19kDa interacting protein 3; PDCD6, programmed cell death 6; MDK, midkine; ENO2, hypermethylation of the enolase gene.

Potential value of ferroptosis inducers for NPC radiosensitization

To date, a number of small-molecule compounds have been confirmed to induce ferroptosis, which are expected to be developed into novel antitumor small-molecule drugs. According to the different targets of small-molecule compounds, ferroptosis inducers may be divided into four categories as follows: i) Inhibition of system Xc; ii) inhibition of GPX4 activity; iii) degradation of GPX4 and coenzyme Q10; and iv) induction of lipid peroxide production (18). In NPC, ferroptosis may be induced by disulfiram/copper, itraconazole attenuates, cephalosporin antibiotics and cucurbitacin B (13,51,117,118). Among those, itraconazole is able to sequester iron in lysosomes, thereby causing ferroptosis and reversing the radiation resistance of NPC spheres. Therefore, whether ferroptosis inducers exert radiosensitizing effects and their potential value in RT for NPC warrants further investigation. An in vitro study indicated that the ferroptosis inducers imidazole ketone erastin and RSL3 act synergistically with radiation to promote ferroptotic cell death in a variety of tumor cell lines (32). In addition, another study suggested that erastin enhances the radiosensitivity of HeLa and NCI-H1975 adenocarcinoma cells via GSH depletion (119). Therefore, ferroptosis inducers may reduce the GSH concentration to enhance the radiosensitivity of radioresistant tumors, including NPC. A recent study indicated that the ferroptosis inducer erastin is able to trigger autophagy by increasing intracellular iron levels (120). Of note, a large number of studies have indicated that the activation of cytotoxic autophagy is able to enhance the sensitivity of tumor cells to RT (121). Therefore, the application of ferroptosis inducers to induce cytotoxic autophagy in NPC cells may be a promising method for radiosensitization.

Conclusions and future perspectives

The role of ferroptosis, a relatively newly identified type of cell death, has not been extensively investigated in NPC to date. The aim of the present review was to discuss the molecular mechanisms of ferroptosis in cancer. The metabolism of iron, PUFA and GPX4 have key roles in ferroptosis and there is a potential utility for the modulation of ferroptosis in the radiosensitization of NPC (51,122). Of note, the core regulators of ferroptosis, including miRNAs, serve important functions in RT for NPC. Radiation-resistant cells have been suggested to be more susceptible to ferroptosis due to their metabolic characteristics and cellular signaling pathways (123). Therefore, ferroptosis inducers may be of value in the radiosensitization of NPC. Itraconazole is a promising ferroptosis inducer for radiosensitization of NPC, which may reverse the radioresistance of NPC spheroids (51). In brief, targeting ferroptosis may provide a novel strategy to improve RT sensitivity of NPC.

However, several issues remain to be addressed, including elucidating the exact mechanism of action of ferroptosis, determining the possible association between autophagy and ferroptosis in the radiosensitization of NPC, determining how to use nanotechnology materials to target ferroptosis regulators in NPC to enhance RT sensitivity and discovering additional ferroptosis inducers and regulatory genes. These questions must be addressed and successfully resolved before ferroptosis may be applied in the clinical setting.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

Data sharing is not applicable.

Authors' contributions

HLL mainly took charge of researching the literature and writing the manuscript; XSH had a guiding role in the review and was involved in revising the manuscript critically for important intellectual content; NHD and JXX provided ideas in the revision process. All authors read and approved the final manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Chen YP, Chan ATC, Le QT, Blanchard P, Sun Y and Ma J: Nasopharyngeal carcinoma. Lancet. 394:64–80. 2019. View Article : Google Scholar : PubMed/NCBI

2 

Long M, Fu Z, Li P and Nie Z: Cigarette smoking and the risk of nasopharyngeal carcinoma: A meta-analysis of epidemiological studies. BMJ Open. 7:e0165822017. View Article : Google Scholar : PubMed/NCBI

3 

Liu YP, Lv X, Zou X, Hua YJ, You R, Yang Q, Xia L, Guo SY, Hu W, Zhang MX, et al: Minimally invasive surgery alone compared with intensity-modulated radiotherapy for primary stage I nasopharyngeal carcinoma. Cancer Commun (Lond). 39:752019. View Article : Google Scholar : PubMed/NCBI

4 

Jaffray DA: Image-guided radiotherapy: From current concept to future perspectives. Nat Rev Clin Oncol. 9:688–699. 2012. View Article : Google Scholar : PubMed/NCBI

5 

Liu G, Zeng X, Wu B, Zhao J and Pan Y: RNA-Seq analysis of peripheral blood mononuclear cells reveals unique transcriptional signatures associated with radiotherapy response of nasopharyngeal carcinoma and prognosis of head and neck cancer. Cancer Biol Ther. 21:139–146. 2020. View Article : Google Scholar : PubMed/NCBI

6 

Baidoo KE, Yong K and Brechbiel MW: Molecular pathways: Targeted α-particle radiation therapy. Clin Cancer Res. 19:530–537. 2013. View Article : Google Scholar : PubMed/NCBI

7 

Srinivas US, Tan BWQ, Vellayappan BA and Jeyasekharan AD: ROS and the DNA damage response in cancer. Redox Biol. 25:1010842019. View Article : Google Scholar : PubMed/NCBI

8 

Lee AWM, Ng WT, Chan JYW, Corry J, Mäkitie A, Mendenhall WM, Rinaldo A, Rodrigo JP, Saba NF, Strojan P, et al: Management of locally recurrent nasopharyngeal carcinoma. Cancer Treat Rev. 79:1018902019. View Article : Google Scholar : PubMed/NCBI

9 

Dixon SJ, Lemberg KM, Lamprecht MR, Skouta R, Zaitsev EM, Gleason CE, Patel DN, Bauer AJ, Cantley AM, Yang WS, et al: Ferroptosis: An iron-dependent form of nonapoptotic cell death. Cell. 149:1060–1072. 2012. View Article : Google Scholar : PubMed/NCBI

10 

Xu T, Ding W, Ji X, Ao X, Liu Y, Yu W and Wang J: Molecular mechanisms of ferroptosis and its role in cancer therapy. J Cell Mol Med. 23:4900–4912. 2019. View Article : Google Scholar : PubMed/NCBI

11 

Friedmann Angeli JP, Schneider M, Proneth B, Tyurina YY, Tyurin VA, Hammond VJ, Herbach N, Aichler M, Walch A, Eggenhofer E, et al: Inactivation of the ferroptosis regulator Gpx4 triggers acute renal failure in mice. Nat Cell Biol. 16:1180–1191. 2014. View Article : Google Scholar : PubMed/NCBI

12 

Raven EP, Lu PH, Tishler TA, Heydari P and Bartzokis G: Increased iron levels and decreased tissue integrity in hippocampus of Alzheimer's disease detected in vivo with magnetic resonance imaging. J Alzheimers Dis. 37:127–136. 2013. View Article : Google Scholar : PubMed/NCBI

13 

Huang S, Cao B, Zhang J, Feng Y, Wang L, Chen X, Su H, Liao S, Liu J, Yan J and Liang B: Induction of ferroptosis in human nasopharyngeal cancer cells by cucurbitacin B: molecular mechanism and therapeutic potential. Cell Death Dis. 12:2372021. View Article : Google Scholar : PubMed/NCBI

14 

Li Z, Chen L, Chen C, Zhou Y, Hu D, Yang J, Chen Y, Zhuo W, Mao M, Zhang X, et al: Targeting ferroptosis in breast cancer. Biomark Res. 8:582020. View Article : Google Scholar : PubMed/NCBI

15 

Xie Y, Hou W, Song X, Yu Y, Huang J, Sun X, Kang R and Tang D: Ferroptosis: Process and function. Cell Death Differ. 23:369–379. 2016. View Article : Google Scholar : PubMed/NCBI

16 

Kazan HH, Urfali-Mamatoglu C and Gunduz U: Iron metabolism and drug resistance in cancer. Biometals. 30:629–641. 2017. View Article : Google Scholar : PubMed/NCBI

17 

He YJ, Liu XY, Xing L, Wan X, Chang X and Jiang HL: Fenton reaction-independent ferroptosis therapy via glutathione and iron redox couple sequentially triggered lipid peroxide generator. Biomaterials. 241:1199112020. View Article : Google Scholar : PubMed/NCBI

18 

Torti SV and Torti FM: Iron and cancer: More ore to be mined. Nat Rev Cancer. 13:342–355. 2013. View Article : Google Scholar : PubMed/NCBI

19 

Liang C, Zhang X, Yang M and Dong X: Recent progress in ferroptosis inducers for cancer therapy. Adv Mater. 31:e19041972019. View Article : Google Scholar : PubMed/NCBI

20 

Lin LS, Song J, Song L, Ke K, Liu Y, Zhou Z, Shen Z, Li J, Yang Z, Tang W and Niu G: Simultaneous fenton-like ion delivery and glutathione depletion by MnO2-based nanoagent to enhance chemodynamic therapy. Angew Chem Int Ed Engl. 57:4902–4906. 2018. View Article : Google Scholar : PubMed/NCBI

21 

Wang H, Lin D, Yu Q, Li Z, Lenahan C, Dong Y, Wei Q and Shao A: A promising future of ferroptosis in tumor therapy. Front Cell Dev Biol. 9:6291502021. View Article : Google Scholar : PubMed/NCBI

22 

Yang WS, SriRamaratnam R, Welsch ME, Shimada K, Skouta R, Viswanathan VS, Cheah JH, Clemons PA, Shamji AF, Clish CB, et al: Regulation of ferroptotic cancer cell death by GPX4. Cell. 156:317–331. 2014. View Article : Google Scholar : PubMed/NCBI

23 

Stockwell BR, Jiang X and Gu W: Emerging mechanisms and disease relevance of ferroptosis. Trends Cell Biol. 30:478–490. 2020. View Article : Google Scholar : PubMed/NCBI

24 

Sato H, Tamba M, Ishii T and Bannai S: Cloning and expression of a plasma membrane cystine/glutamate exchange transporter composed of two distinct proteins. J Biol Chem. 274:11455–11458. 1999. View Article : Google Scholar : PubMed/NCBI

25 

Lo M, Ling V, Wang YZ and Gout PW: The xc-cystine/glutamate antiporter: A mediator of pancreatic cancer growth with a role in drug resistance. Br J Cancer. 99:464–472. 2008. View Article : Google Scholar : PubMed/NCBI

26 

Gao M, Yi J, Zhu J, Minikes AM, Monian P, Thompson CB and Jiang X: Role of mitochondria in ferroptosis. Mol Cell. 73:354–363.e3. 2019. View Article : Google Scholar : PubMed/NCBI

27 

Tang D, Chen X, Kang R and Kroemer G: Ferroptosis: Molecular mechanisms and health implications. Cell Res. 31:107–125. 2021. View Article : Google Scholar : PubMed/NCBI

28 

Yagoda N, von Rechenberg M, Zaganjor E, Bauer AJ, Yang WS, Fridman DJ, Wolpaw AJ, Smukste I, Peltier JM, Boniface J, et al: RAS-RAF-MEK-dependent oxidative cell death involving voltage-dependent anion channels. Nature. 447:864–868. 2007. View Article : Google Scholar : PubMed/NCBI

29 

Yang WH, Huang Z, Wu J, Ding CC, Murphy SK and Chi JT: A TAZ-ANGPTL4-NOX2 axis regulates ferroptotic cell death and chemoresistance in epithelial ovarian cancer. Mol Cancer Res. 18:79–90. 2020. View Article : Google Scholar : PubMed/NCBI

30 

Lang X, Green MD, Wang W, Yu J, Choi JE, Jiang L, Liao P, Zhou J, Zhang Q, Dow A, et al: Radiotherapy and immunotherapy promote tumoral lipid oxidation and ferroptosis via synergistic repression of SLC7A11. Cancer Discov. 9:1673–1685. 2019. View Article : Google Scholar : PubMed/NCBI

31 

Lei G, Mao C, Yan Y, Zhuang L and Gan B: Ferroptosis, radiotherapy, and combination therapeutic strategies. Protein Cell. Apr 23–2021.(Epub ahead of print). View Article : Google Scholar

32 

Ye LF, Chaudhary KR, Zandkarimi F, Harken AD, Kinslow CJ, Upadhyayula PS, Dovas A, Higgins DM, Tan H, Zhang Y, et al: Radiation-induced lipid peroxidation triggers ferroptosis and synergizes with ferroptosis inducers. ACS Chem Biol. 15:469–484. 2020. View Article : Google Scholar : PubMed/NCBI

33 

Lei G, Zhang Y, Koppula P, Liu X, Zhang J, Lin SH, Ajani JA, Xiao Q, Liao Z, Wang H and Gan B: The role of ferroptosis in ionizing radiation-induced cell death and tumor suppression. Cell Res. 30:146–162. 2020. View Article : Google Scholar : PubMed/NCBI

34 

Xie L, Song X, Yu J, Guo W, Wei L, Liu Y and Wang X: Solute carrier protein family may involve in radiation-induced radioresistance of non-small cell lung cancer. J Cancer Res Clin Oncol. 137:1739–1747. 2011. View Article : Google Scholar : PubMed/NCBI

35 

McDonald JT, Kim K, Norris AJ, Vlashi E, Phillips TM, Lagadec C, Della Donna L, Ratikan J, Szelag H, Hlatky L and McBride WH: Ionizing radiation activates the Nrf2 antioxidant response. Cancer Res. 70:8886–8895. 2010. View Article : Google Scholar : PubMed/NCBI

36 

Zong Y, Feng S, Cheng J, Yu C and Lu G: Up-regulated ATF4 expression increases cell sensitivity to apoptosis in response to radiation. Cell Physiol Biochem. 41:784–794. 2017. View Article : Google Scholar : PubMed/NCBI

37 

Koppula P, Zhuang L and Gan B: Cystine transporter SLC7A11/xCT in cancer: Ferroptosis, nutrient dependency, and cancer therapy. Protein Cell. 12:599–620. 2021. View Article : Google Scholar : PubMed/NCBI

38 

Maier P, Hartmann L, Wenz F and Herskind C: Cellular pathways in response to ionizing radiation and their targetability for tumor radiosensitization. Int J Mol Sci. 17:1022016. View Article : Google Scholar : PubMed/NCBI

39 

Stracker TH, Roig I, Knobel PA and Marjanović M: The ATM signaling network in development and disease. Front Genet. 4:372013. View Article : Google Scholar : PubMed/NCBI

40 

Sanli T, Steinberg GR, Singh G and Tsakiridis T: AMP-activated protein kinase (AMPK) beyond metabolism: Anovel genomic stress sensor participating in the DNA damage response pathway. Cancer Biol Ther. 15:156–169. 2014. View Article : Google Scholar : PubMed/NCBI

41 

Degterev A and Yuan J: Expansion and evolution of cell death programmes. Nat Rev Mol Cell Biol. 9:378–390. 2008. View Article : Google Scholar : PubMed/NCBI

42 

Gudkov AV and Komarova EA: The role of p53 in determining sensitivity to radiotherapy. Nat Rev Cancer. 3:117–129. 2003. View Article : Google Scholar : PubMed/NCBI

43 

Kirtonia A, Sethi G and Garg M: The multifaceted role of reactive oxygen species in tumorigenesis. Cell Mol Life Sci. 77:4459–4483. 2020. View Article : Google Scholar : PubMed/NCBI

44 

Halliwell B and Cross CE: Oxygen-derived species: Their relation to human disease and environmental stress. Environ Health Perspect. 102 (Suppl 10):S5–S12. 1994. View Article : Google Scholar : PubMed/NCBI

45 

Grivennikova VG and Vinogradov AD: Generation of superoxide by the mitochondrial Complex I. Biochim Biophys Acta. 1757:553–561. 2006. View Article : Google Scholar : PubMed/NCBI

46 

Hentze MW, Muckenthaler MU, Galy B and Camaschella C: Two to tango: Regulation of mammalian iron metabolism. Cell. 142:24–38. 2010. View Article : Google Scholar : PubMed/NCBI

47 

Gutteridge JM and Halliwell B: Iron toxicity and oxygen radicals. Baillieres Clin Haematol. 2:195–256. 1989. View Article : Google Scholar : PubMed/NCBI

48 

Doll S and Conrad M: Iron and ferroptosis: A still ill-defined liaison. IUBMB Life. 69:423–434. 2017. View Article : Google Scholar : PubMed/NCBI

49 

Andrews NC: Disorders of iron metabolism. N Engl J Med. 341:1986–1995. 1999. View Article : Google Scholar : PubMed/NCBI

50 

Li B, Liao Z, Mo Y, Zhao W, Zhou X, Xiao X, Cui W, Feng G, Zhong S, Liang Y, et al: Inactivation of 3-hydroxybutyrate dehydrogenase type 2 promotes proliferation and metastasis of nasopharyngeal carcinoma by iron retention. Br J Cancer. 122:102–110. 2020. View Article : Google Scholar : PubMed/NCBI

51 

Xu Y, Wang Q, Li X, Chen Y and Xu G: Itraconazole attenuates the stemness of nasopharyngeal carcinoma cells via triggering ferroptosis. Environ Toxicol. 36:257–266. 2021. View Article : Google Scholar : PubMed/NCBI

52 

Andrews NC: Forging a field: The golden age of iron biology. Blood. 112:219–230. 2008. View Article : Google Scholar : PubMed/NCBI

53 

Ivanov SD, Semenov AL, Kovan'ko EG and Yamshanov VA: Effects of iron ions and iron chelation on the efficiency of experimental radiotherapy of animals with gliomas. Bull Exp Biol Med. 158:800–803. 2015. View Article : Google Scholar : PubMed/NCBI

54 

Hou W, Xie Y, Song X, Sun X, Lotze MT, Zeh HJ III, Kang R and Tang D: Autophagy promotes ferroptosis by degradation of ferritin. Autophagy. 12:1425–1428. 2016. View Article : Google Scholar : PubMed/NCBI

55 

Park E and Chung SW: ROS-mediated autophagy increases intracellular iron levels and ferroptosis by ferritin and transferrin receptor regulation. Cell Death Dis. 10:8222019. View Article : Google Scholar : PubMed/NCBI

56 

Hong X, Roh W, Sullivan RJ, Wong KHK, Wittner BS, Guo H, Dubash TD, Sade-Feldman M, Wesley B, Horwitz E, et al: The lipogenic regulator SREBP2 induces transferrin in circulating melanoma cells and suppresses ferroptosis. Cancer Discov. 11:678–695. 2021. View Article : Google Scholar : PubMed/NCBI

57 

Feng H, Schorpp K, Jin J, Yozwiak CE, Hoffstrom BG, Decker AM, Rajbhandari P, Stokes ME, Bender HG, Csuka JM, et al: Transferrin receptor is a specific ferroptosis marker. Cell Rep. 30:3411–3423.e7. 2020. View Article : Google Scholar : PubMed/NCBI

58 

Yang WS and Stockwell BR: Synthetic lethal screening identifies compounds activating iron-dependent, nonapoptotic cell death in oncogenic-RAS-harboring cancer cells. Chem Biol. 15:234–245. 2008. View Article : Google Scholar : PubMed/NCBI

59 

Guagnozzi D, Severi C, Ialongo P, Viscido A, Patrizi F, Testino G, Vannella L, Labriola R, Strom R and Caprilli R: Ferritin as a simple indicator of iron deficiency in anemic IBD patients. Inflamm Bowel Dis. 12:150–151. 2006. View Article : Google Scholar : PubMed/NCBI

60 

Ma BB, Leungm SF, Hui EP, Mo F, Kwan WH, Zee B, Yuen J and Chan AT: Prospective validation of serum CYFRA 21-1, beta-2-microglobulin, and ferritin levels as prognostic markers in patients with nonmetastatic nasopharyngeal carcinoma undergoing radiotherapy. Cancer. 101:776–781. 2004. View Article : Google Scholar : PubMed/NCBI

61 

Chen X, Long X, Liang Z, Lei H, Li L, Qu S and Zhu X: Higher N stage and serum ferritin, but lower serum albumin levels are associated with distant metastasis and poor survival in patients with nasopharyngeal carcinoma following intensity-modulated radiotherapy. Oncotarget. 8:73177–73186. 2017. View Article : Google Scholar : PubMed/NCBI

62 

Zhang W, Fan S, Zou G, Shi L, Zeng Z, Ma J, Zhou Y, Li X, Zhang X, Li X, et al: Lactotransferrin could be a novel independent molecular prognosticator of nasopharyngeal carcinoma. Tumour Biol. 36:675–683. 2015. View Article : Google Scholar : PubMed/NCBI

63 

Zhou Y, Zeng Z, Zhang W, Xiong W, Wu M, Tan Y, Yi W, Xiao L, Li X, Huang C, et al: Lactotransferrin: A candidate tumor suppressor-deficient expression in human nasopharyngeal carcinoma and inhibition of NPC cell proliferation by modulating the mitogen-activated protein kinase pathway. Int J Cancer. 123:2065–2072. 2008. View Article : Google Scholar : PubMed/NCBI

64 

Zhang H, Feng X, Liu W, Jiang X, Shan W, Huang C, Yi H, Zhu B, Zhou W, Wang L, et al: Underlying mechanisms for LTF inactivation and its functional analysis in nasopharyngeal carcinoma cell lines. J Cell Biochem. 112:1832–1843. 2011. View Article : Google Scholar : PubMed/NCBI

65 

Deng M, Zhang W, Tang H, Ye Q, Liao Q, Zhou Y, Wu M, Xiong W, Zheng Y, Guo X, et al: Lactotransferrin acts as a tumor suppressor in nasopharyngeal carcinoma by repressing AKT through multiple mechanisms. Oncogene. 32:4273–4283. 2013. View Article : Google Scholar : PubMed/NCBI

66 

Song M, Bode AM, Dong Z and Lee MH: AKT as a therapeutic target for cancer. Cancer Res. 79:1019–1031. 2019. View Article : Google Scholar : PubMed/NCBI

67 

Dodson M, Castro-Portuguez R and Zhang DD: NRF2 plays a critical role in mitigating lipid peroxidation and ferroptosis. Redox Biol. 23:1011072019. View Article : Google Scholar : PubMed/NCBI

68 

Qiang Z, Dong H, Xia Y, Chai D, Hu R and Jiang H: Nrf2 and STAT3 alleviates ferroptosis-mediated IIR-ALI by regulating SLC7A11. Oxid Med Cell Longev. 2020:51469822020. View Article : Google Scholar : PubMed/NCBI

69 

Sun X, Ou Z, Chen R, Niu X, Chen D, Kang R and Tang D: Activation of the p62-Keap1-NRF2 pathway protects against ferroptosis in hepatocellular carcinoma cells. Hepatology. 63:173–184. 2016. View Article : Google Scholar : PubMed/NCBI

70 

Cloer EW, Goldfarb D, Schrank TP, Weissman BE and Major MB: NRF2 activation in cancer: From DNA to protein. Cancer Res. 79:889–898. 2019. View Article : Google Scholar : PubMed/NCBI

71 

Huang W, Shi G, Yong Z, Li J, Qiu J, Cao Y, Zhao Y and Yuan L: Downregulation of RKIP promotes radioresistance of nasopharyngeal carcinoma by activating NRF2/NQO1 axis via downregulating miR-450b-5p. Cell Death Dis. 11:5042020. View Article : Google Scholar : PubMed/NCBI

72 

Zhou J, Ding J, Ma X, Zhang M, Huo Z, Yao Y, Li D and Wang Z: The NRF2/KEAP1 pathway modulates nasopharyngeal carcinoma cell radiosensitivity via ROS elimination. Onco Targets Ther. 13:9113–9122. 2020. View Article : Google Scholar : PubMed/NCBI

73 

Zhang G, Wang W, Yao C, Ren J, Zhang S and Han M: Salinomycin overcomes radioresistance in nasopharyngeal carcinoma cells by inhibiting Nrf2 level and promoting ROS generation. Biomed Pharmacother. 91:147–154. 2017. View Article : Google Scholar : PubMed/NCBI

74 

Yun SM, Kim YS and Hur DY: LMP1 and 2A induce the expression of Nrf2 through Akt signaling pathway in Epstein-Barr virus-transformed B cells. Transl Oncol. 12:775–783. 2019. View Article : Google Scholar : PubMed/NCBI

75 

Hsu JL, Chou JW, Chen TF, Hsu JT, Su FY, Lan JL, Wu PC, Hu CM, Lee EY and Lee WH: Glutathione peroxidase 8 negatively regulates caspase-4/11 to protect against colitis. EMBO Mol Med. 12:e93862020. View Article : Google Scholar : PubMed/NCBI

76 

Koeberle SC, Gollowitzer A, Laoukili J, Kranenburg O, Werz O, Koeberle A and Kipp AP: Distinct and overlapping functions of glutathione peroxidases 1 and 2 in limiting NF-κB-driven inflammation through redox-active mechanisms. Redox Biol. 28:1013882020. View Article : Google Scholar : PubMed/NCBI

77 

Maiorino M, Conrad M and Ursini F: GPx4, lipid peroxidation, and cell death: Discoveries, rediscoveries, and open issues. Antioxid Redox Signal. 29:61–74. 2018. View Article : Google Scholar : PubMed/NCBI

78 

Nunes SC and Serpa J: Glutathione in ovarian cancer: A Double-edged sword. Int J Mol Sci. 19:18822018. View Article : Google Scholar : PubMed/NCBI

79 

Meng DF, Guo LL, Peng LX, Zheng LS, Xie P, Mei Y, Li CZ, Peng XS, Lang YH, Liu ZJ, et al: Antioxidants suppress radiation-induced apoptosis via inhibiting MAPK pathway in nasopharyngeal carcinoma cells. Biochem Biophys Res Commun. 527:770–777. 2020. View Article : Google Scholar : PubMed/NCBI

80 

Song X, Xie Y, Kang R, Hou W, Sun X, Epperly MW, Greenberger JS and Tang D: FANCD2 protects against bone marrow injury from ferroptosis. Biochem Biophys Res Commun. 480:443–449. 2016. View Article : Google Scholar : PubMed/NCBI

81 

Bao Y, Feng H, Zhao F, Zhang L, Xu S, Zhang C, Zhao C and Qin G: FANCD2 knockdown with shRNA interference enhances the ionizing radiation sensitivity of nasopharyngeal carcinoma CNE-2 cells. Neoplasma. 68:40–52. 2021. View Article : Google Scholar : PubMed/NCBI

82 

Xu S, Zhao F, Liang Z, Feng H, Bao Y, Xu W, Zhao C and Qin G: Expression of FANCD2 is associated with prognosis in patients with nasopharyngeal carcinoma. Int J Clin Exp Pathol. 12:3465–3473. 2019.PubMed/NCBI

83 

Suttner DM and Dennery PA: Reversal of HO-1 related cytoprotection with increased expression is due to reactive iron. FASEB J. 13:1800–1809. 1999. View Article : Google Scholar : PubMed/NCBI

84 

Adedoyin O, Boddu R, Traylor A, Lever JM, Bolisetty S, George JF and Agarwal A: Heme oxygenase-1 mitigates ferroptosis in renal proximal tubule cells. Am J Physiol Renal Physiol. 314:F702–F714. 2018. View Article : Google Scholar : PubMed/NCBI

85 

Shi L and Fang J: Implication of heme oxygenase-1 in the sensitivity of nasopharyngeal carcinomas to radiotherapy. J Exp Clin Cancer Res. 27:132008. View Article : Google Scholar : PubMed/NCBI

86 

Jiang L, Kon N, Li T, Wang SJ, Su T, Hibshoosh H, Baer R and Gu W: Ferroptosis as a p53-mediated activity during tumour suppression. Nature. 520:57–62. 2015. View Article : Google Scholar : PubMed/NCBI

87 

Wang SJ, Li D, Ou Y, Jiang L, Chen Y, Zhao Y and Gu W: Acetylation is crucial for p53-mediated ferroptosis and tumor suppression. Cell Rep. 17:366–373. 2016. View Article : Google Scholar : PubMed/NCBI

88 

Xie Y, Zhu S, Song X, Sun X, Fan Y, Liu J, Zhong M, Yuan H, Zhang L, Billiar TR, et al: The tumor suppressor p53 limits Ferroptosis by blocking DPP4 activity. Cell Rep. 20:1692–1704. 2017. View Article : Google Scholar : PubMed/NCBI

89 

Wang Z, Mao JW, Liu GY, Wang FG, Ju ZS, Zhou D and Wang RY: MicroRNA-372 enhances radiosensitivity while inhibiting cell invasion and metastasis in nasopharyngeal carcinoma through activating the PBK-dependent p53 signaling pathway. Cancer Med. 8:712–728. 2019. View Article : Google Scholar : PubMed/NCBI

90 

Ma WS, Ma JG and Xing LN: Efficacy and safety of recombinant human adenovirus p53 combined with chemoradiotherapy in the treatment of recurrent nasopharyngeal carcinoma. Anticancer Drugs. 28:230–236. 2017. View Article : Google Scholar : PubMed/NCBI

91 

Song X, Zhu S, Chen P, Hou W, Wen Q, Liu J, Xie Y, Liu J, Klionsky DJ, Kroemer G, et al: AMPK-mediated BECN1 phosphorylation promotes ferroptosis by directly blocking System Xc activity. Curr Biol. 28:2388–2399.e5. 2018. View Article : Google Scholar : PubMed/NCBI

92 

Kang R, Zhu S, Zeh HJ, Klionsky DJ and Tang D: BECN1 is a new driver of ferroptosis. Autophagy. 14:2173–2175. 2018. View Article : Google Scholar : PubMed/NCBI

93 

Wan XB, Fan XJ, Chen MY, Xiang J, Huang PY, Guo L, Wu XY, Xu J, Long ZJ, Zhao Y, et al: Elevated Beclin 1 expression is correlated with HIF-1alpha in predicting poor prognosis of nasopharyngeal carcinoma. Autophagy. 6:395–404. 2010. View Article : Google Scholar : PubMed/NCBI

94 

Wang Y, Chen W, Lian J, Zhang H, Yu B, Zhang M, Wei F, Wu J, Jiang J, Jia Y, et al: The lncRNA PVT1 regulates nasopharyngeal carcinoma cell proliferation via activating the KAT2A acetyltransferase and stabilizing HIF-1α. Cell Death Differ. 27:695–710. 2020. View Article : Google Scholar : PubMed/NCBI

95 

Majidinia M, Karimian A, Alemi F, Yousefi B and Safa A: Targeting miRNAs by polyphenols: Novel therapeutic strategy for aging. Biochem Pharmacol. 173:1136882020. View Article : Google Scholar : PubMed/NCBI

96 

Ding C, Ding X, Zheng J, Wang B, Li Y, Xiang H, Dou M, Qiao Y, Tian P and Xue W: miR-182-5p and miR-378a-3p regulate ferroptosis in I/R-induced renal injury. Cell Death Dis. 11:9292020. View Article : Google Scholar : PubMed/NCBI

97 

Deng SH, Wu DM, Li L, Liu T, Zhang T, Li J, Yu Y, He M, Zhao YY, Han R and Xu Y: miR-324-3p reverses cisplatin resistance by inducing GPX4-mediated ferroptosis in lung adenocarcinoma cell line A549. Biochem Biophys Res Commun. 549:54–60. 2021. View Article : Google Scholar : PubMed/NCBI

98 

Xiao FJ, Zhang D, Wu Y, Jia QH, Zhang L, Li YX, Yang YF, Wang H, Wu CT and Wang LS: miRNA-17-92 protects endothelial cells from erastin-induced ferroptosis through targeting the A20-ACSL4 axis. Biochem Biophys Res Commun. 515:448–454. 2019. View Article : Google Scholar : PubMed/NCBI

99 

Ma LL, Liang L, Zhou D and Wang SW: Tumor suppressor miR-424-5p abrogates ferroptosis in ovarian cancer through targeting ACSL4. Neoplasma. 68:165–173. 2021. View Article : Google Scholar : PubMed/NCBI

100 

Tomita K, Fukumoto M, Itoh K, Kuwahara Y, Igarashi K, Nagasawa T, Suzuki M, Kurimasa A and Sato T: MiR-7-5p is a key factor that controls radioresistance via intracellular Fe2+ content in clinically relevant radioresistant cells. Biochem Biophys Res Commun. 518:712–718. 2019. View Article : Google Scholar : PubMed/NCBI

101 

Zhang K, Wu L, Zhang P, Luo M, Du J, Gao T, O'Connell D, Wang G, Wang H and Yang Y: miR-9 regulates ferroptosis by targeting glutamic-oxaloacetic transaminase GOT1 in melanoma. Mol Carcinog. 57:1566–1576. 2018. View Article : Google Scholar : PubMed/NCBI

102 

Luo M, Wu L, Zhang K, Wang H, Zhang T, Gutierrez L, O'Connell D, Zhang P, Li Y, Gao T, et al: miR-137 regulates ferroptosis by targeting glutamine transporter SLC1A5 in melanoma. Cell Death Differ. 25:1457–1472. 2018. View Article : Google Scholar : PubMed/NCBI

103 

Qi YF, Yang Y, Zhang Y, Liu S, Luo B and Liu W: Down regulation of lactotransferrin enhanced radio-sensitivity of nasopharyngeal carcinoma. Comput Biol Chem. 90:1074262021. View Article : Google Scholar : PubMed/NCBI

104 

He W, Jin H, Liu Q and Sun Q: miR-182-5p contributes to radioresistance in nasopharyngeal carcinoma by regulating BNIP3 expression. Mol Med Rep. 23:1302021. View Article : Google Scholar : PubMed/NCBI

105 

Zhang Y, Zheng L, Lin S, Liu Y, Wang Y and Gao F: MiR-124 enhances cell radiosensitivity by targeting PDCD6 in nasopharyngeal carcinoma. Int J Clin Exp Pathol. 10:11461–11470. 2017.PubMed/NCBI

106 

Tian Y, Tian Y, Tu Y, Zhang G, Zeng X, Lin J, Ai M, Mao Z, Zheng R and Yuan Y: microRNA-124 inhibits stem-like properties and enhances radiosensitivity in nasopharyngeal carcinoma cells via direct repression of expression of JAMA. J Cell Mol Med. 24:9533–9544. 2020. View Article : Google Scholar : PubMed/NCBI

107 

Chen L, Zhou H and Guan Z: CircRNA_000543 knockdown sensitizes nasopharyngeal carcinoma to irradiation by targeting miR-9/platelet-derived growth factor receptor B axis. Biochem Biophys Res Commun. 512:786–792. 2019. View Article : Google Scholar : PubMed/NCBI

108 

Hu JL, He GY, Lan XL, Zeng ZC, Guan J, Ding Y, Qian XL, Liao WT, Ding YQ and Liang L: Inhibition of ATG12-mediated autophagy by miR-214 enhances radiosensitivity in colorectal cancer. Oncogenesis. 7:162018. View Article : Google Scholar : PubMed/NCBI

109 

Han JB, Huang ML, Li F, Yang R, Chen SM and Tao ZZ: MiR-214 mediates cell proliferation and apoptosis of nasopharyngeal carcinoma through targeting both WWOX and PTEN. Cancer Biother Radiopharm. 35:615–625. 2020. View Article : Google Scholar : PubMed/NCBI

110 

Zhang ZC, Li YY, Wang HY, Fu S, Wang XP, Zeng MS, Zeng YX and Shao JY: Knockdown of miR-214 promotes apoptosis and inhibits cell proliferation in nasopharyngeal carcinoma. PLoS One. 9:e861492014. View Article : Google Scholar : PubMed/NCBI

111 

He J, Tang Y and Tian Y: MicroRNA-214 promotes proliferation and inhibits apoptosis via targeting Bax in nasopharyngeal carcinoma cells. Mol Med Rep. 12:6286–6292. 2015. View Article : Google Scholar : PubMed/NCBI

112 

Deng M, Ye Q, Qin Z, Zheng Y, He W, Tang H, Zhou Y, Xiong W, Zhou M, Li X, et al: miR-214 promotes tumorigenesis by targeting lactotransferrin in nasopharyngeal carcinoma. Tumour Biol. 34:1793–1800. 2013. View Article : Google Scholar : PubMed/NCBI

113 

Deng X, Ma L, Wu M, Zhang G, Jin C, Guo Y and Liu R: miR-124 radiosensitizes human glioma cells by targeting CDK4. J Neurooncol. 114:263–274. 2013. View Article : Google Scholar : PubMed/NCBI

114 

Luo Y, Wang J, Wang F, Liu X, Lu J, Yu X, Ma X, Peng X and Li X: Foxq1 promotes metastasis of nasopharyngeal carcinoma by inducing vasculogenic mimicry via the EGFR signaling pathway. Cell Death Dis. 12:4112021. View Article : Google Scholar : PubMed/NCBI

115 

Lu J, Xu F and Lu H: LncRNA PVT1 regulates ferroptosis through miR-214-mediated TFR1 and p53. Life Sci. 260:1183052020. View Article : Google Scholar : PubMed/NCBI

116 

Bao WD, Zhou XT, Zhou LT, Wang F, Yin X, Lu Y, Zhu LQ and Liu D: Targeting miR-124/Ferroportin signaling ameliorated neuronal cell death through inhibiting apoptosis and ferroptosis in aged intracerebral hemorrhage murine model. Aging Cell. 19:e132352020. View Article : Google Scholar : PubMed/NCBI

117 

Li Y, Chen F, Chen J, Chan S, He Y, Liu W and Zhang G: Disulfiram/Copper induces antitumor activity against both nasopharyngeal cancer cells and cancer-associated fibroblasts through ROS/MAPK and Ferroptosis pathways. Cancers (Basel). 12:1382020. View Article : Google Scholar : PubMed/NCBI

118 

He X, Yao Q, Fan D, Duan L, You Y, Liang W, Zhou Z, Teng S, Liang Z, Hall DD, et al: Cephalosporin antibiotics specifically and selectively target nasopharyngeal carcinoma through HMOX1-induced ferroptosis. Life Sci. 277:1194572021. View Article : Google Scholar : PubMed/NCBI

119 

Shibata Y, Yasui H, Higashikawa K, Miyamoto N and Kuge Y: Erastin, a ferroptosis-inducing agent, sensitized cancer cells to X-ray irradiation via glutathione starvation in vitro and in vivo. PLoS One. 14:e02259312019. View Article : Google Scholar : PubMed/NCBI

120 

Li M, Wang X, Lu S, He C, Wang C, Wang L, Wang X, Ge P and Song D: Erastin triggers autophagic death of breast cancer cells by increasing intracellular iron levels. Oncol Lett. 20:572020.PubMed/NCBI

121 

Tam SY, Wu VW and Law HK: Influence of autophagy on the efficacy of radiotherapy. Radiat Oncol. 12:572017. View Article : Google Scholar : PubMed/NCBI

122 

Mou Y, Wang J, Wu J, He D, Zhang C, Duan C and Li B: Ferroptosis, a new form of cell death: opportunities and challenges in cancer. J Hematol Oncol. 12:342019. View Article : Google Scholar : PubMed/NCBI

123 

Friedmann Angeli JP, Krysko DV and Conrad M: Ferroptosis at the crossroads of cancer-acquired drug resistance and immune evasion. Nat Rev Cancer. 19:405–414. 2019. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2021
Volume 22 Issue 5

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Li H, Deng N, Xiao J and He X: Cross‑link between ferroptosis and nasopharyngeal carcinoma: New approach to radiotherapy sensitization (Review). Oncol Lett 22: 770, 2021
APA
Li, H., Deng, N., Xiao, J., & He, X. (2021). Cross‑link between ferroptosis and nasopharyngeal carcinoma: New approach to radiotherapy sensitization (Review). Oncology Letters, 22, 770. https://doi.org/10.3892/ol.2021.13031
MLA
Li, H., Deng, N., Xiao, J., He, X."Cross‑link between ferroptosis and nasopharyngeal carcinoma: New approach to radiotherapy sensitization (Review)". Oncology Letters 22.5 (2021): 770.
Chicago
Li, H., Deng, N., Xiao, J., He, X."Cross‑link between ferroptosis and nasopharyngeal carcinoma: New approach to radiotherapy sensitization (Review)". Oncology Letters 22, no. 5 (2021): 770. https://doi.org/10.3892/ol.2021.13031