Open Access

Advances in the study of regulators of ferroptosis in head and neck squamous cell carcinoma (Review)

  • Authors:
    • Mengyuan Yang
    • Rongrong Guo
    • Xin Chen
    • Guohua Song
    • Fang Zhang
  • View Affiliations

  • Published online on: April 13, 2023     https://doi.org/10.3892/ijmm.2023.5248
  • Article Number: 45
  • Copyright: © Yang 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

Head and neck squamous cell carcinoma (HNSCC), a common malignancy of the head and neck, is associated with a rapid progression, a high mortality rate and unsatisfactory curative effects. The treatment efficacy is unsatisfactory due to chemotherapeutic drug resistance, the lack of ideal therapeutic agents, as well as the absence of clinical prognostic models. Thus, the identification of novel potential therapeutic targets for its diagnosis and treatment is vital. Ferroptosis is an iron‑dependent regulatory cell death mode different from traditional cell death modes, such as apoptosis and autophagy, and has notable therapeutic potential in cancer treatment. The study of ferroptosis in HNSCC is expected to solve this bottleneck problem. In the present review, the findings, characteristics and regulatory mechanisms of ferroptosis are summarized, with emphasis on the factors and drugs that regulate ferroptosis in HNSCC, in order to provide theoretical basis for the targeted therapy of ferroptosis in HNSCC.

1. Introduction

Head and neck squamous cell carcinoma (HNSCC) is a general term for squamous epithelial malignancies originating in the nasal cavity, oral cavity, pharynx and larynx, accounting for >90% of head and neck cancers, with rapid progression, extensive infiltration and a poor prognosis (1). The current treatment strategies for HNSCC are mainly based on radiotherapy, chemotherapy and surgery (2). However, patients with advanced or metastatic HNSCC are highly susceptible to recurrence and metastasis following surgery. These patients are usually treated with cisplatin-based concurrent radiotherapy and targeted drug-induced chemotherapy represented by cetuximab; the efficacy of this treatment however, is hampered by the widespread development of cisplatin resistance, thus rendering cisplatin-based chemotherapeutic regimens ineffective. Previous studies have shown that promoting ferroptosis in tumor cells is a more effective method of reducing cisplatin resistance in cancer cells (3), and it can enhance the sensitivity of tumor cells to radiotherapy and chemotherapeutic drugs (4).

Ferroptosis is a regulated form of cell death that differs from the traditional cell death modality, depending on lipid peroxidation, which is induced by iron ions and reactive oxygen species (5). Initially, researchers found that ferroptosis can cause multiple tissue damage via oxidative stress pathways, which are associated with the pathogenesis of several degenerative diseases, such as Alzheimer's disease (6), ischemia-reperfusion injury (7) and osteoarthritis (8). Based on the ability of ferroptosis to induce damage to multiple tissues, it has been suggested that ferroptosis has immense therapeutic potential in the treatment of cancer with active abnormal proliferation, and the induction of ferroptosis in tumor cells may lead to the development of novel treatment strategies (9).

Therefore, the present review summarizes the mechanisms of action of ferroptosis and its regulators in HNSCC, and also discusses the drugs regulated by ferroptosis in HNSCC, in order to provide a theoretical basis for the further use of ferroptosis in the treatment of HNSCC.

2. Data collection methods

Up to October 2022, the 'PubMed', 'Springer', 'Web of Science' and 'CNKI' databases were searched for original research articles and reviews on the progress of ferroptosis in HNSCC. The terms for the search included the following: Ferroptosis, ferroptosis and HNSCC, regulatory mechanism of ferroptosis, iron metabolism, ferroptosis inducers, ferroptosis inhibitors, ferroptosis and glutathione peroxidase 4 (GPX4), ferroptosis and System Xc, and ferroptosis and iron (the detailed search strategies are presented in Table I).

Table I

Summary of the search strategy used in the present study.

Table I

Summary of the search strategy used in the present study.

ItemsSpecification
Date of searchOctober 31, 2022
Databases and other sources searched'PubMed' 'Springer' 'Web of Science' 'CNKI'
Search terms used'ferroptosis' (title/abstract)
'ferroptosis' and 'HNSCC' (title/abstract)
'regulatory mechanism of ferroptosis' (title/abstract)
'iron metabolism' (title/abstract)
'ferroptosis inducers' (title/abstract)
'ferroptosis inhibitors' (title/abstract)
'ferroptosis' and 'GPX4' (title/abstract)
'ferroptosis' and 'System Xc' (title/abstract)
'ferroptosis' and 'Iron' (title/abstract)
Timeframe2008-2022
Inclusion and exclusion criteriaFocus was placed on original articles and reviews in the English language about the regulatory factor of ferroptosis in head and neck squamous cell carcinoma; articles that had no information about HNSCC and ferroptosis were excluded

[i] CNKI, China National Knowledge Infrastructure; HNSCC, head and neck squamous cell carcinoma; GPX4, glutathione peroxidase 4.

3. The discovery and characteristics of ferroptosis

Ferroptosis is a regulated form of cell death which differs from apoptosis, necrosis and autophagy, which can be inhibited by iron chelators and antioxidants (10), characterized by the excessive accumulation of lipid peroxides and reactive oxygen species (ROS), discovered by the Stockwell laboratory at Columbia University in 2012 (11,12). The study of ferroptosis has been gradually applied to a variety of diseases, such as tissue ischemia-reperfusion, neurological diseases, acute renal failure and tumors (13).

Ferroptosis is caused by unrestricted lipid peroxidation and plasma membrane rupture, the two main features of which are iron ion accumulation and lipid peroxidation. Morphologically, ferroptosis is manifested by cell membrane fracture and effacement, mitochondrial atrophy, reduction or even the disappearance of mitochondrial ridges, and increased cell membrane density; biologically, it is manifested by the increased generation of ROSs, iron ion aggregation, the activation of mitogen-activated protein kinases (MAPKs), and the inhibition of cysteine-glutamate reverse transporter by decreasing cystine uptake and depleting glutathione (GSH); in immunologically, it is manifested by the release of damage-associated molecular patterns, which promote inflammatory responses (14).

4. Regulatory mechanisms of ferroptosis

It has been demonstrated that the sensitivity of cells to ferroptosis is associated with multiple regulatory mechanisms, mainly including three pathways: The regulation of iron metabolism, the regulation of the system Xc/GSH/GPX4 anti-oxidant system and the regulation of lipid peroxidation, which can directly affect the sensitivity of cells to ferroptosis (14) (Fig. 1).

Regulation of iron metabolism

Iron is an essential trace metal element with redox activity in the human body; increased levels of iron and/or iron-binding proteins and dysregulated iron metabolism lead to a risk of carcinogenesis and promote tumor growth (15).

Iron in the body circulation is usually present and is transported as Fe3+, stored in endosomes with transferrin (TF) in the presence of TF receptor 1, and is reduced to Fe2+ in the presence of iron oxidoreductase (STEAP3). When Fe2+ is in excess, it can be transferred to the cell plasma and eventually stored in ferritin to form labile iron pool. When the buffering capacity of ferritin is exceeded, a large amount of divalent iron ions (Fe2+) catalyze hydrogen peroxide (H2O2), which generates toxic hydroxyl radicals (·OH) and thus triggers cellular lipid peroxidation (16). It has been found that the concentration of H2O2 and iron ions in a large number of tumor cells is generally low; thus, when the disorder of iron metabolism causes an increase of free iron in cells, iron catalyzes the production of ROS through the Fenton reaction, and ROS further promotes lipid peroxidation and causes lipid peroxide aggregation, which induces ferroptosis (17).

Cancer cells are more iron-dependent compared to normal cells (18), and some cancer cells exhibit iron ion aggregation. Thus, the regulation of ferroptosis via iron homeostasis can effectively kill cancer cells by increasing iron uptake, decreasing iron storage and limiting iron efflux to promote ferroptosis; and by preventing ferroptosis through iron chelators and antioxidants (19).

Cellular iron accumulation is one of the typical markers of ferroptosis, and the detection of changes in iron ions can indicate whether ferroptosis has occurred. The iron ion content is positively associated with ferroptosis, and by using the Fe2+ detection probe FerroOrange, flow cytometry or confocal microscopy to detect intracellular iron content, the orange fluorescence of FerroOrange is enhanced in cells in which ferroptosis occurs (20).

Regulation of the System Xc−/GSH/GPX4 antioxidant system

Ferroptosis can be induced by exogenous or endogenous pathways. The exogenous pathway initiates ferroptosis mainly by inhibiting the functional activity of System Xc-. System Xc is a critical intracellular antioxidant system consisting of two subunits: Solute carrier family 7 member 11 (SLC7A11; xCT) and solute carrier family 3 member 2 (SLC3A2; CD98hc). SLC7A11 is responsible for the major transport activities, including cystine uptake and glutamate excretion; SLC3A2 functions as a chaperone protein. Cystine is exchanged into the cell at a 1:1 ratio with glutamate and is rapidly reduced to cysteine, which is involved in the synthesis of intracellular GSH and GPX4 (21).

The depletion of GSH, a key co-factor for the function of GPX4, disrupts cellular redox homeostasis, leading to the accumulation of ROS and ultimately inducing the onset of ferroptosis. Erastin can reduce GSH synthesis by targeting the inhibition of System Xc-, which reduces the cystine entering the cell and ultimately induces the onset of ferroptosis (22).

The endogenous pathway then activates ferroptosis by reducing the activity of GPX4. The basic function of GPX family members is H2O2 reduction at the expense of GSH, and GPX4 is the only enzyme that reduces cholesterol hydroperoxide and esterifies oxidized fatty acids (23). GPX4 is also a key regulatory component of the ferroptosis mechanism, and its reduced activity is the hallmark event for the onset of ferroptosis (24).

Regulation of lipid peroxidation

Normal cells tend to maintain a dynamic redox balance, whereas cancer cells are usually in an oxidative stress state with higher metabolic levels than normal cells, accompanied by ROS accumulation (25). The production of ROS is associated with damage to proteins, carbohydrates, lipids and nucleotides, which can eventually lead to the development of malignancies. The primary indicator of ferroptosis is the excessive accumulation of lipid peroxides and ROS, which is currently detected in laboratory mainly by the specific fluorescent probes, C11 BODIPY 581/591 and 2′,7′-dichlorodihydrofluorescein diacetate (H2DCFH-DA) (26).

In the process of ferroptosis, polyunsaturated fatty acids (PUFAs) can be oxidized by intracellular ROS to produce unstable lipid hydroperoxides and lipid peroxides, reaching the lethal amounts and can lead to the destruction of bilayer lipid structures, thus inducing ferroptosis (27). The level of intracellular PUFAs determines the development of ferroptosis (28,29). The enzymatic reaction of lipid peroxidation contains three key enzymes: Long-chain lipid acyl coenzyme A synthesis 4 (ACSL4) (30), lysophosphatidylcholine acyltransferase 3 (LPCAT3) (31) and arachidonic acid 15-lipoxygenase (ALOX15). Among these, ACSL4 catalyzes the conversion of free PUFAs into polyunsaturated fatty acids-acetyl coenzyme A (PUFA CoA), which is subsequently esterified by LPCAT3 into phospholipid-type polyunsaturated fatty acids (PL-PUFA), and finally ALOX15 participates in the peroxidation process of membrane phospholipids to lipid peroxides (32). In summary, disorders of lipid metabolism are closely related to cellular ferroptosis, and the accumulation of lipid oxides and ROS is necessary for ferroptosis to occur.

5. Ferroptosis regulators in head and neck squamous cell carcinoma

Cisplatin, one of the most widely used platinum compounds, is the clinical first-line chemotherapeutic agent used in the treatment of HNSCC (33). Although ~80% of patients with HNSCC are initially sensitive to this treatment, the resistance rate in subsequent treatment is as high as 70% (34), which severely diminishes the efficacy of cisplatin (35), and chemoresistance as a major cause of treatment failure in HNSCC can no longer be underestimated.

Recent research has identified some drugs that can reverse chemoresistance in vitro; however, the majority of these drugs have cytotoxic defects that limit their clinical application, and thus, the search for a novel methods of chemotherapy sensitization has become a hot topic and difficult issue in HNSCC research (2). Previous studies have reported that the resistance of HNSCC to cisplatin can be reduced by inducing ferroptosis without affecting normal tissues (36-38). This may provide a new strategy with which to improve the post-operative survival of patients with HNSCC. Thus, exploring ferroptosis regulators in HNSCC is critical for improving the prognosis of patients with HNSCC.

Currently, in HNSCC, studies on ferroptosis have focused on the System Xc-GSH-GPX4 pathway, which induces ferroptosis by directly or indirectly disrupting intracellular redox homeostasis (39,40) (Table II and Fig. 2).

Table II

Summary of ferroptosis regulators in HNSCC.

Table II

Summary of ferroptosis regulators in HNSCC.

MechanismCompound(Refs.)
Activation/inhibition of ferroptosis through regulation of SLC7A11miR-34c-3p(58)
circRNA FNDC3B(62)
NRF2(68)
SOCS1(75)
IL-6(78)
Activation/inhibition of ferroptosis through the regulation of GPX4RSL3(90)
EMP1(95)
PER1(105)
Cav-1(112)
Regulation of ferroptosis through iron metabolic pathwaysFTH1(75)

[i] HNSCC, head and neck squamous cell carcinoma; SLC7A11, solute carrier family 7 member 11; GPX4, glutathione peroxidase 4; circRNA, circular RNA; NRF2, nuclear factor erythroid 2-related factor 2; SOCS1, suppressor of cytokine signaling 1; IL-6, interleukin 6; RSL3, RAS-selective lethal 3; EMP1, epithelial membrane protein 1; PER1, period circadian regulator 1 (period 1); Cav-1, caveolin-1; FTH1, ferritin heavy chain.

Activation/inhibition of ferroptosis through the regulation of SLC7A11

SLC7A11 expression has been found to be higher than normal in a variety of cancer types (21,41,42) SLC7A11 can inhibit the occurrence of ferroptosis, and its expression level is positively associated with the clinical stage of patients with HNSCC (43); in human papillomavirus (HPV)-positive patients with HNSCC, the expression level of SLC7A11 has been found to be lower than that HPV-negative patients, and positive patients may be more responsive to radiotherapy and chemotherapy (44). It is hypothesized that the ferroptosis marker gene, SLC7A11, plays a crucial role in the development of HNSCC, and targeting SLC7A11 may selectively kill tumor cells while preserving normal cells (45).

In HNSCC, multiple factors regulate the expression of SLC7A11 and thus the occurrence of ferroptosis, and hotspot factors currently studied include non-coding RNAs (ncRNAs), nuclear factor erythroid 2-related factor 2 (NRF2), suppressor of cytokine signaling 1 (SOCS1) and interleukin (IL)-6, whose regulatory mechanisms are specified below.

ncRNAs

ncRNAs account for ~98% of the human transcribed genome and are involved in a number of physiological and pathological processes, including cancer, rendering them one of the hotspots of current research (46). It has been found that ncRNAs, particularly microRNAs (miRNAs/miRs) and circular RNAs (circRNAs), are closely related to the biological process of ferroptosis, and they can regulate ferroptosis in tumor cells through transcriptional or post-transcriptional levels by mechanisms involving glutamine catabolism, mitochondria-related proteins, iron metabolism, glutathione metabolism, amino acid metabolism, lipid peroxidation, cell cycle and the p53 signaling pathway (47). Therefore, ncRNAs and downstream target genes associated with cellular ferroptosis may become novel clinical markers and therapeutic targets for HNSCC.

a) miRNAs

It has been found that miRNAs can inhibit ferroptosis in tumor cells by regulating the glutamine catabolic pathway (48-50). miR-137 and miR-9 and other miRNAs achieve the inhibition of ferroptosis in tumor cells by inhibiting a link in the glutamine catabolic pathway, further revealing the molecular mechanisms of the miRNA regulation of ferroptosis in tumor cells, and promoting tumor cells by targeting related miRNAs glutamine metabolism can improve the sensitivity of tumor cells to ferroptosis (48,51). In addition, miRNAs can inhibit tumor cell ferroptosis by regulating mitochondria-related proteins. miRNA expression differences in tumor cells can affect mitochondrial function and energy metabolism, which in turn can achieve the regulation of tumor cell ferroptosis. Mitochondrial transferrin mainly mediates the entry of Fe2+ into mitochondria to maintain the coordinated intracellular distribution and utilization of iron (52,53).

Previous studies have demonstrated that miRNAs are key regulators of HNSCC carcinogenesis (54-56), and they have attracted widespread attention in HNSCC. Shen et al (57) revealed that miR-34c-3p is closely associated with the spread, migration, invasion and apoptosis of HNSCC. As illustrated in Fig. 2, miR-34c-3p inhibits the expression of SLC7A11 by binding to the 3′ non-coding region of SLC7A11, which inhibits the transport process of System Xc. The reduction of cysteine affects the synthesis of cysteine and glutathione, which inhibits the expression of GPX4 and ultimately induces lipid peroxidation and ROS generation, leading to ferroptosis in HNSCC and inhibiting HNSCC cell growth (58).

b) circRNAs

circRNA, which account for >90% of the human transcriptome, but have minimal potential to encode proteins, are involved in the regulation of ferroptosis in tumor cells by adsorbing certain miRNAs, functioning as competing endogenous RNAs to regulate the expression of relevant miRNA target genes and thus, and their involvement in cancer ferroptosis (59). The potential regulatory mechanisms include mitochondria-related proteins, iron metabolism, glutathione metabolism and lipid peroxidation (60).

Some circRNAs have been reported to regulate head and neck tumorigenesis (61). As also illustrated in Fig. 2, Yang et al (62) in HNSCC found that circRNA FNDC3B further upregulated the expression level of SLC7A11 by adsorbing miR-520d-5p, inhibited the occurrence of ferroptosis, induced epithelial-mesenchymal transition, and promoted HNSCC cell proliferation.

These studies have proven that ncRNA-based therapy may be used for cancer treatment by regulating ferroptosis; however, there is no mature technical means available for applying ncRNA-mediated tumor therapy to clinical practice. The in-depth study of the molecular mechanisms of the ferroptosis regulation of tumor cells by ncRNAs may provide potential targets and novel therapeutic strategies for cancer treatment. Thus far, circFNDC3B and miR-34c-3p have only been studied in HNSCC.

NRF2

The redox state of the cell is determined by the balance between the production of reactive substances and the subsequent reduction of a series of antioxidant defense systems. Excess reactive substances react to generate lipid peroxides, which are associated with the progression of a number of diseases, including cancer, diabetes, cardiovascular disease and liver disease (63). The intracellular antioxidant defense system is used to control the level of reactive substances.

NRF2, the central transcriptional regulator in the oxidative stress response, plays a key role in cellular antioxidant responses, redox homeostasis and metabolic homeostasis, and is involved in lipid peroxidation and free iron accumulation (64); numerous factors of the ferroptosis cascade are target genes of NRF2 (65), indicating its critical role in mediating the ferroptosis response. When oxidative stress occurs, NRF2 activates and induces the expression of antioxidant genes to eliminate ROS, and if excessive ROS generation causes peroxidation, it initiates a cellular suicide program, in which ferroptosis is the most critical. Furthermore, a number of antioxidants such as GSH, heme oxygenase 1 and NAD(P)H quinone dehydrogenase 1 are downstream genes of NRF2 (66). Cancer and neurodegenerative diseases highly associated with oxidative stress render NRF2 a crucial therapeutic target for these diseases (64,67).

As illustrated in Fig. 2, in HNSCC, the high expression of NRF2 induces the high expression of SLC7A11 by directly binding to the promoter region of SLC7A11, which in turn induces System Xc to transport intracellular glutamate away and extracellular cystine in, and elevated cystine leads to an increase in glutathione, which in turn increases the expression of GPX4. In turn, the high expression of GPX4 suppresses ROS and lipid peroxidation levels, ultimately inhibiting the sensitivity of HNSCC cells to ferroptosis and leading to increased patient resistance to radiation therapy (68). Indeed, in other types of cancer, such as colorectal (69), lung (70), stomach (71) and ovarian cancers (72), NRF2 also functions as an oncogenic transcription factor, and cancer cells frequently exhibit the overexpression of NRF2, which plays a key role in counteracting environmental or intracellular stress (21,73).

Therefore, controlling the progression of HNSCC by modulating the NRF2/SLC7A11/ferroptosis pathway provides a potential therapeutic target for enhancing the sensitivity of HNSCC to radiotherapy, and exploring the combination of NRF2 inhibitors with radiotherapy remains a direction for future research.

SOCS1

SOCS1 is considered as a negative feedback regulator of cytokine signaling and is also involved in the formation of the E3 ubiquitin ligase complex (74), which plays critical regulatory roles in both cell growth and proliferation. As illustrated in Fig. 2, in HNSCC, SOCS1 functions as a 'ferroptosis activator' similar to miR-34c-3p by suppressing the transcription of SLC7A11 and thus the uptake of cystine (75).

IL-6

Smoking and betel nut chewing are two risk factors for HNSCC, both of which upregulate the expression of the pro-inflammatory cytokine, IL-6 (76), and oxidative stress caused by an imbalance of oxidants and antioxidants is considered to underlie the development of HNSCC after smoking (77). As also demonstrated in Fig. 2, Li et al found that IL-6 accumulation was associated with lipid peroxidation, which inhibited HNSCC ferroptosis in vitro and in vivo through the upregulation of SLC7A11 expression (78). In addition, IL-6 is secreted by immune cells or tumor cells and can promote tumor cell proliferation, epithelial-mesenchymal transition (EMT) through the activation of signal transducer and transcriptional activator 3 pathways, thus promoting cancer progression and inducing chemotherapy resistance, which is associated with a poor prognosis of patients with HNSCC (76,79). Thus, IL-6 can function as an inhibitor of ferroptosis that drives tumor progression, and it can potentially be used as a tumor prevention and therapeutic target.

Regulation of ferroptosis through the GPX4 pathway

Compared with normal tissues, GPX4 expression levels are significantly elevated in a variety of tumor tissues, such as triple-negative breast (80), gastric (81) and esophageal (82) cancer, leading to the hypothesis that GPX4 may be an oncogene. It has been shown to be a key regulator of ferroptosis; the overexpression or knockdown of GPX4 can affect the cell lethality of 12 ferroptosis inducers (83). GPX4 can affect tumor stem cell production and the EMT process by regulating ROS production (84,85). Viswanathan et al (86) demonstrated that GPX4 inhibitors in the majority of tumors can induce the ferroptosis of drug-resistant cells and prevent tumor recurrence; the combination of the targeted therapy of tumors with GPX4 inhibitors has provide new insight into resolving the issue of drug resistance in cancer cells (87).

In HNSCC, GPX4 is strongly associated with the poor prognosis of patients (88). A previous study demonstrated that the knockdown of GPX4 decreased cell viability, although the level of the caspase activity marker was not increased, indicating that non-apoptotic cell death occurred, which was reversed following pre-treatment with ferrostatin-1, suggesting that cells die via the ferroptosis pathway (89). Thus, in HNSCC, GPX4 can function as an inhibitor of ferroptosis and its downregulation induces tumor cell death; i.e., the modification of GPX4 expression may be a novel therapeutic approach for HNSCC.

In HNSCC, various genes such as RAS-selective lethal 3 (RSL3), epithelial membrane protein 1 (EMP1), period circadian regulator 1 (period 1; PER1) and caveolin-1 (Cav-1) regulate the occurrence of ferroptosis by regulating GPX4 expression and their specific regulatory mechanisms are described below.

RSL3

RSL3, an inhibitor of GPX4 that inhibits cysteine and glutamate transporter proteins, is a ferroptosis activator. As illustrated in Fig. 2, RSL3 functions as a 'ferroptosis activator' in HNSCC by downregulating GPX4 expression, inducing an increase in ROS generation, and catalyzing the lipid peroxidation of highly expressed unsaturated fatty acids in the cell membrane (90).

EMP1

EMP1 is a member of the epithelial membrane protein family, encoded by the peripheral myelin protein family, and is involved in the proliferation, migration and differentiation of tumor cells (91). It is mainly expressed in the squamous epithelium and exhibits differential expression levels in various normal tissues, such as esophagus, stomach and gallbladder (92). EMP1 plays a key role in cell adhesion, proliferation, apoptosis, tumor formation and ferroptosis (93), and its decreased expression is associated with the poor prognosis of patients with certain malignancies and is an independent predictor of ferroptosis (94).

As also illustrated in Fig. 2, Wang et al (95) reported that the overexpression of EMP1 in HNSCC cells intensified the downregulation of GPX4 by the ferroptosis activator, RSL3, increasing the levels of ROS and lipid peroxidation in a time- and concentration-dependent manner, ultimately inducing the development of ferroptosis. In addition, EMP1 enhances the sensitivity of the tumor-targeting drug, gefitinib, and can be used as a biomarker of tumor chemotherapeutic resistance (96). Overall, EMP1 inhibits the malignant progression of HNSCC by activating ferroptosis in HNSCC and provides new markers which may be used to overcome the chemoresistance of tumors, contributing to the development of future therapeutic agents for HNSCC.

PER1

Period genes are present in almost all types of human tissues and organs and are involved in regulating a variety of important physiological and biochemical processes in organisms, and their abnormal expression is an important factor in the development of many diseases, including cancer (97). PER1 is one of the core genes of Period and is involved in regulating important physiological processes, such as apoptosis, autophagy, DNA damage repair and ferroptosis (98); its decreased expression in a variety of cancers, including gastric cancer (99), colon cancer (100), HNSCC (101) and non-small cell lung cancers (102) predicts that PER1 may be a key tumor suppressor gene (103).

In HNSCC, the decreased expression of PER1 is significantly associated with the TNM stage and poor prognosis of patients (104). PER1 negatively correlates with the expression of GPX4, a key regulator of ferroptosis, as shown in Fig. 2, which combine to form the PER1/GPX4 negative feedback pathway, thereby inducing the ferroptosis of cancer cells (105). In summary, the upregulation of PER1 gene expression can inhibit the proliferation, invasion and migration of HNSCC, which plays a similar role as RSL3 as a 'ferroptosis activator'; the in-depth study of this mechanism may provide a novel therapeutic approach for the treatment of HNSCC.

Cav-1

Caveolin is mainly composed of lipids and proteins, among which, Cav-1 plays a major biological role (106), functioning as an oncogene in a variety of tumors, including HNSCC, liver, colon, breast, kidney and lung cancer (107-109). It plays a critical regulatory role in substance transport, endothelial infiltration and tumorigenesis (110). Deng et al (111) demonstrated that Cav-1 was also involved in the regulation of ferroptosis. As demonstrated in Fig. 2, in HNSCC, Cav-1 is highly expressed and inhibits ferroptosis by upregulating GPX4 expression and suppressing ROS and lipid peroxidation. Lu et al (112) found that after knocking down Cav-1 in HNSCC, GPX4 expression was reduced, ferroptosis was activated, and cell proliferation, invasion and migration were all inhibited, suggesting that ferroptosis was negatively regulated by Cav-1. Overall, in HNSCC, Cav-1 functions as an inhibitor of ferroptosis and is likely to be a diagnostic marker of HNSCC (112).

Regulation of ferroptosis through iron metabolic pathways

Ferritin, the most critical form of iron storage in humans, consists of two polypeptide chains: The ferritin heavy chain (FTH1) and the light chain, which are essential for maintaining iron homeostasis and preventing iron overload, and also play a role in regulating the tumor microenvironment and immune metabolism (113). FTH1 is the major functional subunit of iron storage protein with iron oxidase activity that effectively reduces Fe2+ toxicity (114). Moreover, FTH1 is closely associated with the development of HNSCC, as illustrated in Fig. 2, where FTH1 expression levels are upregulated in HNSCC compared to normal tissues (75), which further inhibits the Fenton reaction by downregulating the expression of ferrous ions, thereby inhibiting cellular production of ROS and lipid peroxide aggregation, and ultimately negatively regulating ferroptosis (114). The further study of iron metabolic pathways may aid in the better understanding of the development of ferroptosis and may provide lead to the development of novel cancer therapeutics.

6. Therapeutic modalities regulating ferroptosis in head and neck squamous carcinoma

Drug treatment

Currently, drugs that have been experimentally validated to modulate ferroptosis in HNSCC include dyclonine and paclitaxel (PTX), and ferroptosis-based drug studies may alleviate current resistance to radiotherapy and chemotherapy in HNSCC and increase the sensitivity of the drugs.

Dyclonine, a covalent inhibitor of the cancer cell resistance gene, ALDH3A1 (115), plays a key role in inducing oxidative damage and cell death by promoting the accumulation of lipid peroxides in GSH-resistant cancer cells, and can reduce resistance to targeted therapy with the SLC7A11 inhibitor, sulfasalazine (116).

PTX is a paclitaxel-like substance extracted from plants that exerts anti-mitotic and apoptosis-inducing effects (117); it also induces cellular autophagy (118), downregulates glutamine degradation-related genes (119), affects glutamine catabolism and shifts metabolic reprogramming to oxidative phosphorylation, which in turn regulates the onset of ferroptosis (90,120).

At this stage, the FDA has approved a number of drugs for clinical use that inhibit the development of epithelial-derived tumors, such as ovarian, endometrial and squamous lung cancers through ferroptosis, including GPX4 inhibitors (altretamine), iron ion activators (lapatinib) and SLC7A11 inhibitors (sorafenib) (121). However, although these drugs have not yet been used in the clinical treatment of patients with HNSCC, HNSCC also serves as a tumor of epithelial origin in which these drugs are likely to play a similar role; thus, further research is required into the use of these drugs in HNSCC.

Emerging therapies

Currently, the conventional treatment for HNSCC is surgical resection with drug therapy; however, the, treatment efficacy is poor. Photodynamic therapy (PDT) is one of the emerging therapies for HNSCC. With the participation of photosensitizers and the action of light, the body cells generate reactive oxygen radicals through electron transfer and energy transfer, and eventually interact with oxygen molecules. The generation of large amounts of intracellular ROS leads to lipid peroxidation, causing damage to the structure and function of cell and organelle membranes (122,123).

The non-invasive nature of PDT allows organ function to be preserved in patients with HNSCC. However, PDT is hampered by hypoxia in the tumor microenvironment (TME) due to high intracellular oxygen consumption and tumor vascular distortion. Therefore, increasing oxygen production in the TME may be a new approach to enhancing the efficacy of PDT. The study by Zhu et al (124) in HNSCC found that ferroptosis significantly enhanced the efficacy of PDT and enhanced the anticancer effect through the generation of ROS and a sustained supply of O2 through the Fenton reaction (124). In other words, ferroptosis offers new hope for overcoming PDT resistance associated with hypoxia in the treatment of HNSCC.

7. Conclusions and future perspectives

HNSCC is the most common malignant tumors of the head and neck, with a recurrence and metastasis rate >65% and a 5-year survival rate <50% (125). Although combined surgical resection and radiotherapy have improved the survival rate of patients with HNSCC to a certain extent over the past 20 years, in the majority of patients, first-line drugs used for HNSCC, such as platinum-based drugs, 5-fluorouracil, polyene paclitaxel and cetuximab have minimal effects (2). Therefore, the effective treatment of patients with HNSCC, and the improvement of the quality of life and prognosis of patients are issues that still need to be addressed. Several new therapeutic approaches have been developed for HNSCC, among which PDT is one of the emerging therapies widely used in the clinical treatment of HNSCC, and its treatment mechanism is consistent with ferroptosis; the combination of ferroptosis and PDT may lead to the development of a novel strategy for more effective HNSCC treatment in the future (124).

Since its discovery in 2012, numerous studies have demonstrated that ferroptosis plays a critical role as a novel cell death modality in various diseases (126). Although a number of pathways have been demonstrated to be involved in ferroptosis, to date, the mechanisms of ferroptosis in vivo are mainly focused on three pathways: Iron metabolism, the System Xc/GSH/GPX4 antioxidant system and lipid metabolism. Among these, the pathway involved in regulating HNSCC is mainly the System Xc/GSH/GPX4 pathway, which has been widely studied in various types of cancer, such as liver cancer and non-small cell lung cancer, and drugs targeting ferroptosis have been used in clinical trials (127).

There are numerous factors in the System Xc-/GSH/GPX4 pathway that can affect the occurrence of HNSCC, and in the present review, the factors that regulate ferroptosis in HNSCC have been described, based on existing studies. Among these, NRF2 has been most comprehensively studied in ferroptosis and HNSCC, and a large number of studies have shown that NRF2 exerts a more pronounced inhibitory effect in HNSCC compared with other influencing factors, and further clinical trials are required to verify its role in regulating ferroptosis in HNSCC (128). Likewise, a number of researchers have explored ncRNAs for factors that can affect HNSCC by regulating ferroptosis; however, the research is not yet mature and further investigations are warranted (129).

At this stage, the FDA has approved a number of drugs for clinical use that inhibit the development of epithelial-derived tumors, such as ovarian, endometrial and squamous lung cancers by targeting ferroptosis (130,131). While these factors that target ferroptosis have also been shown to be effective in in vitro studies on HNSCC, all of these studies need to be explored in more depth with regard to their effects on the development and progression of HNSCC to demonstrate whether they can also be applied to the clinical treatment of HNSCC. However, the emergence of these targeted agents is certainly a glimmer of hope for the current dilemma facing HNSCC.

Overall, ferroptosis studies help to address the major issues of HNSCC and provide novel therapeutic targets and strategies for the diagnosis and clinical treatment of HNSCC.

Availability of data and materials

Not applicable.

Authors' contributions

MY drafted the manuscript, and prepared the figures and tables. RG and XC participated in the literature search and in the analysis of the data to be included in the review. GS and FZ edited and revised the manuscript. All authors have read and approved the final version of the 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.

Acknowledgments

Not applicable.

Funding

The present study was supported by the National Natural Science Foundation of China (grant no. 31772551).

References

1 

Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A and Bray F: Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 71:209–249. 2021. View Article : Google Scholar : PubMed/NCBI

2 

Johnson DE, Burtness B, Leemans CR, Lui VWY, Bauman JE and Grandis JR: Head and neck squamous cell carcinoma. Nat Rev Dis Primers. 6:922020. View Article : Google Scholar : PubMed/NCBI

3 

Roh JL, Kim EH, Jang HJ, Park JY and Shin D: Induction of ferroptotic cell death for overcoming cisplatin resistance of head and neck cancer. Cancer Lett. 381:96–103. 2016. View Article : Google Scholar : PubMed/NCBI

4 

Gorrini C, Harris IS and Mak TW: Modulation of oxidative stress as an anticancer strategy. Nat Rev Drug Discov. 12:931–947. 2013. View Article : Google Scholar : PubMed/NCBI

5 

Chen X, Comish PB, Tang D and Kang R: Characteristics and biomarkers of ferroptosis. Front Cell Dev Biol. 9:6371622021. View Article : Google Scholar : PubMed/NCBI

6 

Masaldan S, Belaidi AA, Ayton S and Bush AI: Cellular senescence and iron dyshomeostasis in Alzheimer's disease. Pharmaceuticals (Basel). 12:932019. View Article : Google Scholar : PubMed/NCBI

7 

Guan X, Li X, Yang X, Yan J, Shi P, Ba L, Cao Y and Wang P: The neuroprotective effects of carvacrol on ischemia/reperfusion-induced hippocampal neuronal impairment by ferroptosis mitigation. Life Sci. 235:1167952019. View Article : Google Scholar : PubMed/NCBI

8 

Lv Z, Han J, Li J, Guo H, Fei Y, Sun Z, Dong J, Wang M, Fan C, Li W, et al: Single cell RNA-seq analysis identifies ferroptotic chondrocyte cluster and reveals TRPV1 as an anti-ferroptotic target in osteoarthritis. EBioMedicine. 84:1042582022. View Article : Google Scholar : PubMed/NCBI

9 

Lei G, Zhuang L and Gan B: Targeting ferroptosis as a vulnerability in cancer. Nat Rev Cancer. 22:381–396. 2022. View Article : Google Scholar : PubMed/NCBI

10 

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

11 

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

12 

Stockwell BR, Friedmann Angeli JP, Bayir H, Bush AI, Conrad M, Dixon SJ, Fulda S, Gascón S, Hatzios SK, Kagan VE, et al: Ferroptosis: A regulated cell death nexus linking metabolism, redox biology, and disease. Cell. 171:273–285. 2017. View Article : Google Scholar : PubMed/NCBI

13 

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

14 

Yan HF, Zou T, Tuo QZ, Xu S, Li H, Belaidi AA and Lei P: Ferroptosis: Mechanisms and links with diseases. Signal Transduct Target Ther. 6:492021. View Article : Google Scholar : PubMed/NCBI

15 

Liang W and Ferrara N: Iron metabolism in the tumor microenvironment: Contributions of innate immune cells. Front Immunol. 11:6268122021. View Article : Google Scholar : PubMed/NCBI

16 

Shen Z, Song J, Yung BC, Zhou Z, Wu A and Chen X: Emerging strategies of cancer therapy based on ferroptosis. Adv Mater. 30:e17040072018. 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 

Manz DH, Blanchette NL, Paul BT, Torti FM and Torti SV: Iron and cancer: Recent insights. Ann N Y Acad Sci. 1368:149–161. 2016. View Article : Google Scholar : PubMed/NCBI

19 

Galaris D, Barbouti A and Pantopoulos K: Iron homeostasis and oxidative stress: An intimate relationship. Biochim Biophys Acta Mol Cell Res. 1866:1185352019. View Article : Google Scholar : PubMed/NCBI

20 

Yang Y, Lin Y, Wang M, Yuan K, Wang Q, Mu P, Du J, Yu Z, Yang S, Huang K, et al: Targeting ferroptosis suppresses osteocyte glucolipotoxicity and alleviates diabetic osteoporosis. Bone Res. 10:262022. View Article : Google Scholar : PubMed/NCBI

21 

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 :

22 

Tan Y, Huang Y, Mei R, Mao F, Yang D, Liu J, Xu W, Qian H and Yan Y: HucMSC-derived exosomes delivered BECN1 induces ferroptosis of hepatic stellate cells via regulating the xCT/GPX4 axis. Cell Death Dis. 13:3192022. View Article : Google Scholar : PubMed/NCBI

23 

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

24 

Seibt TM, Proneth B and Conrad M: Role of GPX4 in ferroptosis and its pharmacological implication. Free Radic Biol Med. 133:144–152. 2019. View Article : Google Scholar

25 

Luo C, Sun J, Liu D, Sun B, Miao L, Musetti S, Li J, Han X, Du Y, Li L, et al: Self-assembled redox dual-responsive prodrug-nano-system formed by single thioether-bridged paclitaxel-fatty acid conjugate for cancer chemotherapy. Nano Lett. 16:5401–5408. 2016. View Article : Google Scholar : PubMed/NCBI

26 

Yang Y, Karakhanova S, Hartwig W, D'Haese JG, Philippov PP, Werner J and Bazhin AV: Mitochondria and mitochondrial ROS in cancer: Novel targets for anticancer therapy. J Cell Physiol. 231:2570–2581. 2016. View Article : Google Scholar : PubMed/NCBI

27 

Yang WS and Stockwell BR: Ferroptosis: Death by lipid peroxidation. Trends Cell Biol. 26:165–176. 2016. View Article : Google Scholar :

28 

Lee JY, Kim WK, Bae KH, Lee SC and Lee EW: Lipid metabolism and ferroptosis. Biology (Basel). 10:1842021.PubMed/NCBI

29 

Lee H, Zandkarimi F, Zhang Y, Meena JK, Kim J, Zhuang L, Tyagi S, Ma L, Westbrook TF, Steinberg GR, et al: Energy-stress-mediated AMPK activation inhibits ferroptosis. Nat Cell Biol. 22:225–234. 2020. View Article : Google Scholar : PubMed/NCBI

30 

Doll S, Proneth B, Tyurina YY, Panzilius E, Kobayashi S, Ingold I, Irmler M, Beckers J, Aichler M, Walch A, et al: ACSL4 dictates ferroptosis sensitivity by shaping cellular lipid composition. Nat Chem Biol. 13:91–98. 2017. View Article : Google Scholar :

31 

Liu J, Kang R and Tang D: Signaling pathways and defense mechanisms of ferroptosis. FEBS J. 289:7038–7050. 2022. View Article : Google Scholar

32 

Gan B: ACSL4, PUFA, and ferroptosis: New arsenal in anti-tumor immunity. Signal Transduct Target Ther. 7:1282022. View Article : Google Scholar : PubMed/NCBI

33 

Hung CC, Chien CY, Chu PY, Wu YJ, Lin CS, Huang CJ, Chan LP, Wang YY, Yuan SF, Hour TC and Chen JY: Differential resistance to platinum-based drugs and 5-fluorouracil in p22phox-overexpressing oral squamous cell carcinoma: Implications of alternative treatment strategies. Head Neck. 39:1621–1630. 2017. View Article : Google Scholar : PubMed/NCBI

34 

Kallunki T, Olsen OD and Jäättelä M: Cancer-associated lysosomal changes: Friends or foes? Oncogene. 32:1995–2004. 2013. View Article : Google Scholar

35 

Pan ST, Qin Y, Zhou ZW, He ZX, Zhang X, Yang T, Yang YX, Wang D, Qiu JX and Zhou SF: Plumbagin induces G2/M arrest, apoptosis, and autophagy via p38 MAPK- and PI3K/Akt/mTOR-mediated pathways in human tongue squamous cell carcinoma cells. Drug Des Devel Ther. 9:1601–1626. 2015.PubMed/NCBI

36 

Chen X, Kang R, Kroemer G and Tang D: Broadening horizons: The role of ferroptosis in cancer. Nat Rev Clin Oncol. 18:280–296. 2021. View Article : Google Scholar : PubMed/NCBI

37 

Yu W, Chen Y, Putluri N, Coarfa C, Robertson MJ, Putluri V, Stossi F, Dubrulle J, Mancini MA, Pang JC, et al: Acquisition of cisplatin resistance shifts head and neck squamous cell carcinoma metabolism toward neutralization of oxidative stress. Cancers (Basel). 12:16702020. View Article : Google Scholar : PubMed/NCBI

38 

Han L, Li L and Wu G: Induction of ferroptosis by carnosic acid-mediated inactivation of Nrf2/HO-1 potentiates cisplatin responsiveness in OSCC cells. Mol Cell Probes. 64:1018212022. View Article : Google Scholar : PubMed/NCBI

39 

Lu B, Chen XB, Ying MD, He QJ, Cao J and Yang B: The role of ferroptosis in cancer development and treatment response. Front Pharmacol. 8:9922018. View Article : Google Scholar : PubMed/NCBI

40 

Feng H and Stockwell BR: Unsolved mysteries: How does lipid peroxidation cause ferroptosis? PLoS Biol. 16:e20062032018. View Article : Google Scholar : PubMed/NCBI

41 

Hu K, Li K, Lv J, Feng J, Chen J, Wu H, Cheng F, Jiang W, Wang J, Pei H, et al: Suppression of the SLC7A11/glutathione axis causes synthetic lethality in KRAS-mutant lung adenocarcinoma. J Clin Invest. 130:1752–1766. 2020. View Article : Google Scholar :

42 

Ji X, Qian J, Rahman SMJ, Siska PJ, Zou Y, Harris BK, Hoeksema MD, Trenary IA, Heidi C, Eisenberg R, et al: xCT (SLC7A11)-mediated metabolic reprogramming promotes non-small cell lung cancer progression. Oncogene. 37:5007–5019. 2018. View Article : Google Scholar : PubMed/NCBI

43 

Ma Z, Zhang H, Lian M, Yue C, Dong G, Jin Y, Li R, Wan H, Wang R, Wang Y, et al: SLC7A11, a component of cysteine/glutamate transporter, is a novel biomarker for the diagnosis and prognosis in laryngeal squamous cell carcinoma. Oncol Rep. 38:3019–3029. 2017. View Article : Google Scholar : PubMed/NCBI

44 

Hémon A, Louandre C, Lailler C, Godin C, Bottelin M, Morel V, François C, Galmiche A and Saidak Z: SLC7A11 as a biomarker and therapeutic target in HPV-positive head and neck squamous cell carcinoma. Biochem Biophys Res Commun. 533:1083–1087. 2020. View Article : Google Scholar : PubMed/NCBI

45 

Jyotsana N, Ta KT and DelGiorno KE: The role of cystine/glutamate antiporter SLC7A11/xCT in the pathophysiology of cancer. Front Oncol. 12:8584622022. View Article : Google Scholar : PubMed/NCBI

46 

Slaby O, Laga R and Sedlacek O: Therapeutic targeting of non-coding RNAs in cancer. Biochem J. 474:4219–4251. 2017. View Article : Google Scholar : PubMed/NCBI

47 

Xie B and Guo Y: Molecular mechanism of cell ferroptosis and research progress in regulation of ferroptosis by noncoding RNAs in tumor cells. Cell Death Discov. 7:1012021. View Article : Google Scholar : PubMed/NCBI

48 

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

49 

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

50 

Quirico L, Orso F, Cucinelli S, Paradzik M, Natalini D, Centonze G, Dalmasso A, La Vecchia S, Coco M, Audrito V, et al: miRNA-guided reprogramming of glucose and glutamine metabolism and its impact on cell adhesion/migration during solid tumor progression. Cell Mol Life Sci. 79:2162022. View Article : Google Scholar : PubMed/NCBI

51 

Wang J, Wang B, Ren H and Chen W: miR-9-5p inhibits pancreatic cancer cell proliferation, invasion and glutamine metabolism by targeting GOT1. Biochem Biophys Res Commun. 509:241–218. 2019. View Article : Google Scholar

52 

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

53 

Tomita K, Kuwahara Y, Takashi Y, Igarashi K, Nagasawa T, Nabika H, Kurimasa A, Fukumoto M, Nishitani Y and Sato T: Clinically relevant radioresistant cells exhibit resistance to H2O2 by decreasing internal H2O2 and lipid peroxidation. Tumour Biol. 40:10104283187992502018. View Article : Google Scholar

54 

Amaral AJ, Andrade J, Foxall RB, Matoso P, Matos AM, Soares RS, Rocha C, Ramos CG, Tendeiro R, Serra-Caetano A, et al: miRNA profiling of human naive CD4 T cells links miR-34c-5p to cell activation and HIV replication. EMBO J. 36:346–360. 2017. View Article : Google Scholar

55 

Zhang B, Li Y, Hou D, Shi Q, Yang S and Li Q: MicroRNA-375 inhibits growth and enhances radiosensitivity in oral squamous cell carcinoma by targeting insulin like growth factor 1 receptor. Cell Physiol Biochem. 42:2105–2117. 2017. View Article : Google Scholar : PubMed/NCBI

56 

Wang K, Jin J, Ma T and Zhai H: MiR-139-5p inhibits the tumorigenesis and progression of oral squamous carcinoma cells by targeting HOXA9. J Cell Mol Med. 21:3730–3740. 2017. View Article : Google Scholar : PubMed/NCBI

57 

Shen Y, Sun C, Zhao B, Guo H, Li J, Xia Y, Liu M, Piao S and Saiyin W: miR-34c-5p mediates the cellular malignant behaviors of oral squamous cell carcinoma through targeted binding of TRIM29. Ann Transl Med. 9:15372021. View Article : Google Scholar : PubMed/NCBI

58 

Sun K, Ren W, Li S, Zheng J, Huang Y, Zhi K and Gao L: MiR-34c-3p upregulates erastin-induced ferroptosis to inhibit proliferation in oral squamous cell carcinomas by targeting SLC7A11. Pathol Res Pract. 231:1537782022. View Article : Google Scholar : PubMed/NCBI

59 

Garikipati VNS, Verma SK, Cheng Z, Liang D, Truongcao MM, Cimini M, Yue Y, Huang G, Wang C, Benedict C, et al: Circular RNA CircFndc3b modulates cardiac repair after myocardial infarction via FUS/VEGF-A axis. Nat Commun. 10:43172019. View Article : Google Scholar : PubMed/NCBI

60 

Balihodzic A, Prinz F, Dengler MA, Calin GA, Jost PJ and Pichler M: Non-coding RNAs and ferroptosis: Potential implications for cancer therapy. Cell Death Differ. 29:1094–1106. 2022. View Article : Google Scholar : PubMed/NCBI

61 

Zhang X, Wang L, Li H, Zhang L, Zheng X and Cheng W: Crosstalk between noncoding RNAs and ferroptosis: New dawn for overcoming cancer progression. Cell Death Dis. 11:5802020. View Article : Google Scholar : PubMed/NCBI

62 

Yang J, Cao XH, Luan KF and Huang YD: Circular RNA FNDC3B protects oral squamous cell carcinoma cells from ferroptosis and contributes to the malignant progression by regulating miR-520d-5p/SLC7A11 axis. Front Oncol. 11:6727242021. View Article : Google Scholar

63 

Ayala A, Muñoz MF and Argüelles S: Lipid peroxidation: Production, metabolism, and signaling mechanisms of malondialdehyde and 4-hydroxy-2-nonenal. Oxid Med Cell Longev. 2014:3604382014. View Article : Google Scholar : PubMed/NCBI

64 

Song X and Long D: Nrf2 and ferroptosis: A new research direction for neurodegenerative diseases. Front Neurosci. 14:2672020. View Article : Google Scholar : PubMed/NCBI

65 

Zhang Q, Qu H, Chen Y, Luo X, Chen C, Xiao B, Ding X, Zhao P, Lu Y, Chen AF and Yu Y: Atorvastatin induces mitochondria-dependent ferroptosis via the modulation of Nrf2-xCT/GPx4 axis. Front Cell Dev Biol. 10:8060812022. View Article : Google Scholar : PubMed/NCBI

66 

Ma Q: Role of nrf2 in oxidative stress and toxicity. Annu Rev Pharmacol Toxicol. 53:401–426. 2013. View Article : Google Scholar : PubMed/NCBI

67 

Jelic MD, Mandic AD, Maricic SM and Srdjenovic BU: Oxidative stress and its role in cancer. J Cancer Res Ther. 17:22–28. 2021. View Article : Google Scholar : PubMed/NCBI

68 

Feng L, Zhao K, Sun L, Yin X, Zhang J, Liu C and Li B: SLC7A11 regulated by NRF2 modulates esophageal squamous cell carcinoma radiosensitivity by inhibiting ferroptosis. J Transl Med. 19:3672021. View Article : Google Scholar : PubMed/NCBI

69 

Yang J, Mo J, Dai J, Ye C, Cen W, Zheng X, Jiang L and Ye L: Cetuximab promotes RSL3-induced ferroptosis by suppressing the Nrf2/HO-1 signalling pathway in KRAS mutant colorectal cancer. Cell Death Dis. 12:10792021. View Article : Google Scholar : PubMed/NCBI

70 

Sánchez-Ortega M, Carrera AC and Garrido A: Role of NRF2 in lung cancer. Cells. 10:18792021. View Article : Google Scholar : PubMed/NCBI

71 

Farkhondeh T, Pourbagher-Shahri AM, Azimi-Nezhad M, Forouzanfar F, Brockmueller A, Ashrafizadeh M, Talebi M, Shakibaei M and Samarghandian S: Roles of Nrf2 in gastric cancer: Targeting for therapeutic strategies. Molecules. 26:31572021. View Article : Google Scholar : PubMed/NCBI

72 

Tossetta G, Fantone S, Montanari E, Marzioni D and Goteri G: Role of NRF2 in ovarian cancer. Antioxidants (Basel). 11:6632022. View Article : Google Scholar : PubMed/NCBI

73 

Wang XJ, Sun Z, Villeneuve NF, Zhang S, Zhao F, Li Y, Chen W, Yi X, Zheng W, Wondrak GT, et al: Nrf2 enhances resistance of cancer cells to chemotherapeutic drugs, the dark side of Nrf2. Carcinogenesis. 29:1235–1243. 2008. View Article : Google Scholar : PubMed/NCBI

74 

Ying J, Qiu X, Lu Y and Zhang M: SOCS1 and its potential clinical role in tumor. Pathol Oncol Res. 25:1295–1301. 2019. View Article : Google Scholar : PubMed/NCBI

75 

Hu ZW, Wen YH, Ma RQ, Chen L, Zeng XL, Wen WP and Sun W: Ferroptosis driver SOCS1 and suppressor FTH1 independently correlate with M1 and M2 macrophage infiltration in head and neck squamous cell carcinoma. Front Cell Dev Biol. 9:7277622021. View Article : Google Scholar : PubMed/NCBI

76 

Lee CH, Chang JS, Syu SH, Wong TS, Chan JY, Tang YC, Yang ZP, Yang WC, Chen CT, Lu SC, et al: IL-1β promotes malignant transformation and tumor aggressiveness in oral cancer. J Cell Physiol. 230:875–884. 2015. View Article : Google Scholar

77 

Zhao J, Dar HH, Deng Y, St Croix CM, Li Z, Minami Y, Shrivastava IH, Tyurina YY, Etling E, Rosenbaum JC, et al: PEBP1 acts as a rheostat between prosurvival autophagy and ferroptotic death in asthmatic epithelial cells. Proc Natl Acad Sci USA. 117:14376–14385. 2020. View Article : Google Scholar : PubMed/NCBI

78 

Li M, Jin S, Zhang Z, Ma H and Yang X: Interleukin-6 facilitates tumor progression by inducing ferroptosis resistance in head and neck squamous cell carcinoma. Cancer Lett. 527:28–40. 2022. View Article : Google Scholar

79 

Jin S, Yang X, Li J, Yang W, Ma H and Zhang Z: p53-targeted lincRNA-p21 acts as a tumor suppressor by inhibiting JAK2/STAT3 signaling pathways in head and neck squamous cell carcinoma. Mol Cancer. 18:382019. View Article : Google Scholar : PubMed/NCBI

80 

Ding Y, Chen X, Liu C, Ge W, Wang Q, Hao X, Wang M, Chen Y and Zhang Q: Identification of a small molecule as inducer of ferroptosis and apoptosis through ubiquitination of GPX4 in triple negative breast cancer cells. J Hematol Oncol. 14:192021. View Article : Google Scholar

81 

Zhao L, Peng Y, He S, Li R, Wang Z, Huang J, Lei X, Li G and Ma Q: Apatinib induced ferroptosis by lipid peroxidation in gastric cancer. Gastric Cancer. 24:642–654. 2021. View Article : Google Scholar : PubMed/NCBI

82 

Liu CC, Li HH, Lin JH, Chiang MC, Hsu TW, Li AF, Yen DH, Hsu HS and Hung SC: Esophageal cancer stem-like cells resist ferroptosis-induced cell death by active Hsp27-GPX4 pathway. Biomolecules. 12:482021. View Article : Google Scholar

83 

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

84 

Chatterjee R and Chatterjee J: ROS and oncogenesis with special reference to EMT and stemness. Eur J Cell Biol. 99:1510732020. View Article : Google Scholar : PubMed/NCBI

85 

Cheng FZY, Wang X, Dou J and Wu Z: Research progress on the role and mechanism of GPX4 in ferroptosis. Mod Oncol. 29:1254–1258. 2021.

86 

Viswanathan VS, Ryan MJ, Dhruv HD, Gill S, Eichhoff OM, Seashore-Ludlow B, Kaffenberger SD, Eaton JK, Shimada K, Aguirre AJ, et al: Dependency of a therapy-resistant state of cancer cells on a lipid peroxidase pathway. Nature. 547:453–457. 2017. View Article : Google Scholar : PubMed/NCBI

87 

Hangauer MJ, Viswanathan VS, Ryan MJ, Bole D, Eaton JK, Matov A, Galeas J, Dhruv HD, Berens ME, Schreiber SL, et al: Drug-tolerant persister cancer cells are vulnerable to GPX4 inhibition. Nature. 551:247–250. 2017. View Article : Google Scholar : PubMed/NCBI

88 

Lee JR, Roh JL, Lee SM, Park Y, Cho KJ, Choi SH, Nam SY and Kim SY: Overexpression of glutathione peroxidase 1 predicts poor prognosis in oral squamous cell carcinoma. J Cancer Res Clin Oncol. 143:2257–2265. 2017. View Article : Google Scholar : PubMed/NCBI

89 

Fukuda M, Ogasawara Y, Hayashi H, Okuyama A, Shiono J, Inoue K and Sakashita H: Down-regulation of glutathione peroxidase 4 in oral cancer inhibits tumor growth through SREBP1 signaling. Anticancer Res. 41:1785–1792. 2021. View Article : Google Scholar : PubMed/NCBI

90 

Ye J, Jiang X, Dong Z, Hu S and Xiao M: Low-concentration PTX and RSL3 inhibits tumor cell growth synergistically by inducing ferroptosis in mutant p53 hypopharyngeal squamous carcinoma. Cancer Manag Res. 11:9783–9792. 2019. View Article : Google Scholar : PubMed/NCBI

91 

Ahmat Amin MKB, Shimizu A and Ogita H: The pivotal roles of the epithelial membrane protein family in cancer invasiveness and metastasis. Cancers (Basel). 11:16202019. View Article : Google Scholar : PubMed/NCBI

92 

Liu Y, Ding Y, Nie Y and Yang M: EMP1 promotes the proliferation and invasion of ovarian cancer cells through activating the MAPK pathway. Onco Targets Ther. 13:2047–2055. 2020. View Article : Google Scholar : PubMed/NCBI

93 

Durgan J, Tao G, Walters MS, Florey O, Schmidt A, Arbelaez V, Rosen N, Crystal RG and Hall A: SOS1 and Ras regulate epithelial tight junction formation in the human airway through EMP1. EMBO Rep. 16:87–96. 2015. View Article : Google Scholar :

94 

Sun GG, Wang YD, Cui DW, Cheng YJ and Hu WN: EMP1 regulates caspase-9 and VEGFC expression and suppresses prostate cancer cell proliferation and invasion. Tumour Biol. 35:3455–3462. 2014. View Article : Google Scholar

95 

Wang Y, Zhang L, Yao C, Ma Y and Liu Y: Epithelial membrane protein 1 promotes sensitivity to RSL3-induced ferroptosis and intensifies gefitinib resistance in head and neck cancer. Oxid Med Cell Longev. 2022:47506712022.PubMed/NCBI

96 

van Zandwijk N: Tolerability of gefitinib in patients receiving treatment in everyday clinical practice. Br J Cancer. 89(Suppl 2): S9–S14. 2003. View Article : Google Scholar : PubMed/NCBI

97 

Liu Y, Hao J, Yuan G, Wei M, Bu Y, Jin T and Ma L: PER1 as a tumor suppressor attenuated in the malignant phenotypes of breast cancer cells. Int J Gen Med. 14:7077–7087. 2021. View Article : Google Scholar : PubMed/NCBI

98 

Chakrabarti S and Michor F: Circadian clock effects on cellular proliferation: Insights from theory and experiments. Curr Opin Cell Biol. 67:17–26. 2020. View Article : Google Scholar : PubMed/NCBI

99 

Zhao H, Zeng ZL, Yang J, Jin Y, Qiu MZ, Hu XY, Han J, Liu KY, Liao JW and Zou QF: Prognostic relevance of Period1 (Per1) and Period2 (Per2) expression in human gastric cancer. Int J Clin Exp Pathol. 7:619–630. 2014.PubMed/NCBI

100 

Krugluger W, Brandstaetter A, Kállay E, Schueller J, Krexner E, Kriwanek S, Bonner E and Cross HS: Regulation of genes of the circadian clock in human colon cancer: reduced period-1 and dihydropyrimidine dehydrogenase transcription correlates in high-grade tumors. Cancer Res. 67:7917–7922. 2007. View Article : Google Scholar : PubMed/NCBI

101 

Hsu CM, Lin SF, Lu CT, Lin PM and Yang MY: Altered expression of circadian clock genes in head and neck squamous cell carcinoma. Tumour Biol. 33:149–155. 2012. View Article : Google Scholar

102 

Liu B, Xu K, Jiang Y and Li X: Aberrant expression of Per1, Per2 and Per3 and their prognostic relevance in non-small cell lung cancer. Int J Clin Exp Pathol. 7:7863–7871. 2014.

103 

Gery S, Komatsu N, Baldjyan L, Yu A, Koo D and Koeffler HP: The circadian gene per1 plays an important role in cell growth and DNA damage control in human cancer cells. Mol Cell. 22:375–382. 2006. View Article : Google Scholar : PubMed/NCBI

104 

Yang G, Yang Y, Tang H and Yang K: Loss of the clock gene Per1 promotes oral squamous cell carcinoma progression via the AKT/mTOR pathway. Cancer Sci. 111:1542–1554. 2020. View Article : Google Scholar : PubMed/NCBI

105 

Yang Y, Tang H, Zheng J and Yang K: The PER1/HIF-1alpha negative feedback loop promotes ferroptosis and inhibits tumor progression in oral squamous cell carcinoma. Transl Oncol. 18:1013602022. View Article : Google Scholar : PubMed/NCBI

106 

Chen D and Che G: Value of caveolin-1 in cancer progression and prognosis: Emphasis on cancer-associated fibroblasts, human cancer cells and mechanism of caveolin-1 expression (Review). Oncol Lett. 8:1409–1421. 2014. View Article : Google Scholar : PubMed/NCBI

107 

Yang G, Goltsov AA, Ren C, Kurosaka S, Edamura K, Logothetis R, DeMayo FJ, Troncoso P, Blando J, DiGiovanni J and Thompson TC: Caveolin-1 upregulation contributes to c-Myc-induced high-grade prostatic intraepithelial neoplasia and prostate cancer. Mol Cancer Res. 10:218–229. 2012. View Article : Google Scholar

108 

Sun J, Lu Y, Yu C, Xu T, Nie G, Miao B and Zhang X: Involvement of the TGF-β1 pathway in caveolin-1-associated regulation of head and neck tumor cell metastasis. Oncol Lett. 19:1298–1304. 2020.PubMed/NCBI

109 

Nwosu ZC, Ebert MP, Dooley S and Meyer C: Caveolin-1 in the regulation of cell metabolism: A cancer perspective. Mol Cancer. 15:712016. View Article : Google Scholar : PubMed/NCBI

110 

Vered M, Lehtonen M, Hotakainen L, Pirilä E, Teppo S, Nyberg P, Sormunen R, Zlotogorski-Hurvitz A, Salo T and Dayan D: Caveolin-1 accumulation in the tongue cancer tumor microenvironment is significantly associated with poor prognosis: An in-vivo and in-vitro study. BMC Cancer. 15:252015. View Article : Google Scholar : PubMed/NCBI

111 

Deng G, Li Y, Ma S, Gao Z, Zeng T, Chen L, Ye H, Yang M, Shi H, Yao X, et al: Caveolin-1 dictates ferroptosis in the execution of acute immune-mediated hepatic damage by attenuating nitrogen stress. Free Radic Biol Med. 148:151–161. 2020. View Article : Google Scholar

112 

Lu T, Zhang Z, Pan X, Zhang J, Wang X, Wang M, Li H, Yan M and Chen W: Caveolin-1 promotes cancer progression via inhibiting ferroptosis in head and neck squamous cell carcinoma. J Oral Pathol Med. 51:52–62. 2022. View Article : Google Scholar

113 

Hu ZW, Chen L, Ma RQ, Wei FQ, Wen YH, Zeng XL, Sun W and Wen WP: Comprehensive analysis of ferritin subunits expression and positive correlations with tumor-associated macrophages and T regulatory cells infiltration in most solid tumors. Aging (Albany NY). 13:11491–11506. 2021. View Article : Google Scholar : PubMed/NCBI

114 

Salatino A, Aversa I, Battaglia AM, Sacco A, Di Vito A, Santamaria G, Chirillo R, Veltri P, Tradigo G, Di Cello A, et al: H-ferritin affects cisplatin-induced cytotoxicity in ovarian cancer cells through the modulation of ROS. Oxid Med Cell Longev. 2019:34612512019. View Article : Google Scholar : PubMed/NCBI

115 

Black W, Chen Y, Matsumoto A, Thompson DC, Lassen N, Pappa A and Vasiliou V: Molecular mechanisms of ALDH3A1-mediated cellular protection against 4-hydroxy-2-nonenal. Free Radic Biol Med. 52:1937–1944. 2012. View Article : Google Scholar : PubMed/NCBI

116 

Okazaki S, Shintani S, Hirata Y, Suina K, Semba T, Yamasaki J, Umene K, Ishikawa M, Saya H and Nagano O: Synthetic lethality of the ALDH3A1 inhibitor dyclonine and xCT inhibitors in glutathione deficiency-resistant cancer cells. Oncotarget. 9:33832–33843. 2018. View Article : Google Scholar : PubMed/NCBI

117 

Yardley DA: Taxanes in the elderly patient with metastatic breast cancer. Breast Cancer (Dove Med Press). 7:293–301. 2015.PubMed/NCBI

118 

Choi YH and Yoo YH: Taxol-induced growth arrest and apoptosis is associated with the upregulation of the Cdk inhibitor, p21WAF1/CIP1, in human breast cancer cells. Oncol Rep. 28:2163–2169. 2012. View Article : Google Scholar : PubMed/NCBI

119 

Lv C, Qu H, Zhu W, Xu K, Xu A, Jia B, Qing Y, Li H, Wei HJ and Zhao HY: Low-dose paclitaxel inhibits tumor cell growth by regulating glutaminolysis in colorectal carcinoma cells. Front Pharmacol. 8:2442017. View Article : Google Scholar : PubMed/NCBI

120 

Shen YA, Li WH, Chen PH, He CL, Chang YH and Chuang CM: Intraperitoneal delivery of a novel liposome-encapsulated paclitaxel redirects metabolic reprogramming and effectively inhibits cancer stem cells in Taxol(®)-resistant ovarian cancer. Am J Transl Res. 7:841–855. 2015.

121 

Li J, Cao F, Yin HL, Huang ZJ, Lin ZT, Mao N, Sun B and Wang G: Ferroptosis: Past, present and future. Cell Death Dis. 11:882020. View Article : Google Scholar : PubMed/NCBI

122 

Lin L, Song C, Wei Z, Zou H, Han S, Cao Z, Zhang X, Zhang G, Ran J, Cai Y and Han W: Multifunctional photodynamic/photothermal nano-agents for the treatment of oral leukoplakia. J Nanobiotechnology. 20:1062022. View Article : Google Scholar : PubMed/NCBI

123 

Gurudath S, Ganapathy K, D S, Pai A, Ballal S and Ml A: Estimation of superoxide dismutase and glutathione peroxidase in oral submucous fibrosis, oral leukoplakia and oral cancer-a comparative study. Asian Pac J Cancer Prev. 13:4409–4412. 2012. View Article : Google Scholar

124 

Zhu T, Shi L, Yu C, Dong Y, Qiu F, Shen L, Qian Q, Zhou G and Zhu X: Ferroptosis promotes photodynamic therapy: Supramolecular photosensitizer-inducer nanodrug for enhanced cancer treatment. Theranostics. 9:3293–3307. 2019. View Article : Google Scholar : PubMed/NCBI

125 

Liang F, Wang R, Du Q and Zhu S: An epithelial-mesenchymal transition hallmark gene-based risk score system in head and neck squamous-cell carcinoma. Int J Gen Med. 14:4219–4227. 2021. View Article : Google Scholar : PubMed/NCBI

126 

Jiang X, Stockwell BR and Conrad M: Ferroptosis: Mechanisms, biology and role in disease. Nat Rev Mol Cell Biol. 22:266–282. 2021. View Article : Google Scholar : PubMed/NCBI

127 

Li FJ, Long HZ, Zhou ZW, Luo HY, Xu SG and Gao LC: System Xc /GSH/GPX4 axis: An important antioxidant system for the ferroptosis in drug-resistant solid tumor therapy. Front Pharmacol. 13:9102922022. View Article : Google Scholar

128 

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

129 

Tang Y, Li C, Zhang YJ and Wu ZH: Ferroptosis-related long non-coding RNA signature predicts the prognosis of head and neck squamous cell carcinoma. Int J Biol Sci. 17:702–711. 2021. View Article : Google Scholar : PubMed/NCBI

130 

Mao C, Liu X, Zhang Y, Lei G, Yan Y, Lee H, Koppula P, Wu S, Zhuang L, Fang B, et al: DHODH-mediated ferroptosis defence is a targetable vulnerability in cancer. Nature. 593:586–590. 2021. View Article : Google Scholar : PubMed/NCBI

131 

Hassannia B, Vandenabeele P and Vanden Berghe T: Targeting ferroptosis to iron out cancer. Cancer Cell. 35:830–849. 2019. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2023
Volume 51 Issue 6

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yang M, Guo R, Chen X, Song G and Zhang F: Advances in the study of regulators of ferroptosis in head and neck squamous cell carcinoma (Review). Int J Mol Med 51: 45, 2023
APA
Yang, M., Guo, R., Chen, X., Song, G., & Zhang, F. (2023). Advances in the study of regulators of ferroptosis in head and neck squamous cell carcinoma (Review). International Journal of Molecular Medicine, 51, 45. https://doi.org/10.3892/ijmm.2023.5248
MLA
Yang, M., Guo, R., Chen, X., Song, G., Zhang, F."Advances in the study of regulators of ferroptosis in head and neck squamous cell carcinoma (Review)". International Journal of Molecular Medicine 51.6 (2023): 45.
Chicago
Yang, M., Guo, R., Chen, X., Song, G., Zhang, F."Advances in the study of regulators of ferroptosis in head and neck squamous cell carcinoma (Review)". International Journal of Molecular Medicine 51, no. 6 (2023): 45. https://doi.org/10.3892/ijmm.2023.5248