Open Access

The role of ferroptosis in radiotherapy and combination therapy for head and neck squamous cell carcinoma (Review)

  • Authors:
    • Yu Feng
    • Xiulei Li
    • Bingwu Yang
    • Maocai Li
    • Yongya Du
    • Jing Wang
    • Siyu Liu
    • Lili Gong
    • Lianqing Li
    • Lei Gao
  • View Affiliations

  • Published online on: April 18, 2024     https://doi.org/10.3892/or.2024.8738
  • Article Number: 79
  • Copyright: © Feng 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) is a highly aggressive, heterogeneous tumour usually caused by alcohol and tobacco consumption, making it one of the most common malignancies worldwide. Despite the fact that various therapeutic approaches such as surgery, radiation therapy (RT), chemotherapy (CT) and targeted therapy have been widely used for HNSCC in recent years, its recurrence rate and mortality rate remain high. RT is the standard treatment choice for HNSCC, which induces reactive oxygen species production and causes oxidative stress, ultimately leading to tumour cell death. CT is a widely recognized form of cancer treatment that treats a variety of cancers by eliminating cancer cells and preventing them from reproducing. Immune checkpoint inhibitor and epidermal growth factor receptor are important in the treatment of recurrent or metastatic HNSCC. Iron death, a type of cell death regulated by peroxidative damage to phospholipids containing polyunsaturated fatty acids in cell membranes, has been found to be a relevant death response triggered by tumour RT in recent years. In the present review, an overview of the current knowledge on RT and combination therapy and iron death in HNSCC was provided, the mechanisms by which RT induces iron death in tumour cells were summarized, and therapeutic strategies to target iron death in HNSCC were explored. The current review provided important information for future studies of iron death in the treatment of HNSCC.

Introduction

Head and neck squamous cell carcinoma (HNSCC) is a general term for squamous epithelial malignant tumors originating from the nasal cavity, oral cavity, pharynx, or larynx. HNSCC accounts for ~90% of all head and neck cancers and is characterized by high invasiveness and a poor prognosis (1). P53, as a transcription factor, can play its role in tumor suppression by activating the expression of numerous target genes (2). However, p53 is one of the most commonly mutated genes, which frequently harbors missense mutations. These missense mutations are nucleotide substitutions that result in the substitution of an amino acid in the DNA binding domain. Most p53 mutations in HNSCC are missense mutations and the mutation rate of p53 reaches 65–85% (3,4). Mutant p53 in HNSCC can interact with proteins and have effects on HNSCC proliferation, migration, invasion, immunosuppression and metabolism. Studies have shown that mutant p53 can alter metabolic pathways, including reactive oxygen species (ROS), autophagy and lipid metabolism pathways (5,6). Current treatments for HNSCC include more precise surgical treatments, such as radiation therapy (RT) and chemotherapy (CT) (7). Early-stage HNSCC can be treated using radical RT or surgery, whereas advanced HNSCC should be treated with RT, surgery and CT, as well as multidisciplinary management of toxicity and side effects and follow-up (8). However, despite the extensive use of these treatments for the management of HNSCC, the five-year overall survival of patients with HNSCC has not changed significantly, and the recurrence rate for advanced HNSCC has remained high at ~50% (9). RT involves the use of α, β, γ, and X rays [ionizing radiation (IR)] to eliminate tumor cells and inhibit tumor cell proliferation, with the ultimate goal of achieving radical cure of tumors and controlling tumor progression (10). Direct transmission of radiation through the surface of superficial tumors may lead to irradiation of the normal tissues behind the tumor, leading to severe damage of the normal tissues, particularly those that are sensitive to radiation. Therefore, limiting the irradiation of normal tissues is an important challenge in the development of tumor RT. Currently, the developmental direction of clinical RT technology is to improve RT technology, change the local RT mode, maximize the accuracy of irradiation of tumor tissues, and avoid damage to normal tissues. Notably, patients with advanced HNSCC are radioresistant; thus, increasing their radiation dose to therapeutic levels increases the risk of damage to the surrounding vital organs and causes severe side effects. Therefore, protecting organ function is important in improving the curative effects of RT and reducing its side effects in patients with HNSCC.

Ferroptosis is a regulated form of cell death. Unlike the traditional mode of cell death, ferroptosis is caused by the accumulation of iron ions and ROS-induced lipid peroxidation (11). It is closely related to the occurrence and development of numerous human diseases, such as cancer, viral infections and degenerative diseases. Previous studies have shown that ferroptosis plays an important role in RT-induced cell death and tumor suppression, and that promoting ferroptosis in tumor cells enhances the sensitivity of the cells to radiation and CT drugs (12). Clinically, RT usually needs to be combined with CT, targeted therapy, or immunotherapy to eliminate tumor cells (1315). In the present study, the mechanisms of ferroptosis and its regulatory factors in HNSCC were reviewed, and the mechanism underlying RT-induced tumor cell death in HNSCC was discussed to provide a theoretical basis for further improving the radiosensitivity of HNSCC.

Data collection methods

The PubMed (https://pubmed.ncbi.nlm.nih.gov/) and CNKI (https://oversea.cnki.net/index/) databases were searched for original research articles and reviews on the progression of ferroptosis after RT for HNSCC published until November 2023. The search terms included ferroptosis, ferroptosis and HNSCC, regulatory mechanisms of ferroptosis, ferroptosis and glutathione peroxidase 4 (GPX4), ferroptosis and solute carrier family 7 member 11 (SLC7A11). Information on ferroptosis was retrieved from the FerrDb database (http://www.zhounan.org/ferrdb/current/). FerrDb is the world's first database on ferroptosis regulatory factors and ferroptosis-associated diseases (16).

Cancer therapies

RT for head and neck cancer

RT is a common method of cancer therapy that involves the use of IR to eliminate tumor cells and inhibit tumor cell growth and metastasis (17). The clinical applications of radiation technology include palliative RT, conventional RT in vitro, stereotactic RT surgery, radionuclide therapy and intensity-modulated RT (IMRT) (18,19). IMRT is one of the most advanced and commonly used RT techniques. IMRT can minimize the amount of radiation normal tissues are exposed to and meet the treatment requirements for irregularly shaped tumor targets (20). Therefore, utilization of IMRT techniques for the treatment of patients with HNSCC can be adopted as an organ protection strategy, especially in patients with locally advanced disease (21).

RT-induced tumor cell death can be divided into accidental cell death (ACD) and regulated cell death (RCD) (22). ACD is an uncontrolled passive cell death process, whereas RCD is a controlled cell death process. RCD includes apoptosis, autophagic cell death, necrosis, cornification, atypical cell death (including mitotic catastrophe), anoikis, paraptosis, pyroptosis, entosis, excitotoxicity and ferroptosis (23,24). IR emitted by RT can cause a variety of DNA damage, including base damage, DNA single-strand breaks and DNA double-strand breaks (DSBs), which can affect the integrity of DNA or alter its chemical properties (25). Among them, DNA DSBs have been reported to be the most deleterious effect triggering genome stability, eliminating cancer cells, leading to genome instability, apoptosis, altered cell cycle checkpoints or post-mitotic death, and are the main cause of RCD in tumour cells (26).

Combination therapies

RT and surgery can achieve similar curative effects in patients with early-stage HNSCC. However, due to the lack of effective biomarkers for early diagnosis of HSNCC, most patients are diagnosed at the terminal stage of the disease. For patients with recurrent or metastatic HNSCC (R/M HNSCC) who have lost the opportunity for surgery and RT, long-term control of tumor growth, distant metastasis and clinical symptoms are more important than aiming for a cure. Systemic CT, immunotherapy and targeted therapy can be administered as the primary treatments.

Concurrent CT for head and neck cancer

With the development and advancement of CT, various platinum-based drugs have been revealed to exert certain therapeutic effects on HNSCC. Drugs used for the treatment of HNSCC include cisplatin, bleomycin, fluorouracil and methotrexate. These drugs act mainly through a cytotoxic mechanism to control the proliferation of tumor cells. The most desirable drugs from the drug screen are platinum-based drugs, which constitute the highest proportion of cancer drugs evaluated in clinical trials. In the TAX 323 (EORTC 24971) trials, patients with distant metastasis that occurred during TPF therapy (docetaxel + cisplatin + 5-fluorouracil) showed significantly prolonged progression-free survival and median overall survival (27).

Several clinical trials have shown that concurrent chemo-RT (CRT) for advanced HNSCC to increase local tumor control is relatively simple and can improve survival. Brizel et al (28) compared patients with advanced HNSCC who underwent surgery and received CRT, targeted therapy combined with CRT, and other treatments, and found that CRT is improved compared with RT alone for the treatment of locally advanced metastatic HNSCC.

Immunotherapy for head and neck cancer

Immune checkpoint inhibitors (ICIs) restore the antitumor immune response by blocking immune checkpoints. Inhibitory immune checkpoints, including programmed death receptor-1 (PD-1) and cytotoxic T lymphocyte-associated protein-4 (CTLA-4), are primarily expressed in T cells. Tumor-induced immune responses mediate tumor immune escape by upregulating the expression of immune checkpoints. ICIs can restore the antitumor immunity function of T cells through competitive inhibition of inhibitory receptors on T cells (29).

PD-1/PD-L1 and CTLA-4 inhibitors are the main immunotherapy agents for the treatment of R/M HNSCC. PD-1 inhibitors include pembrolizumab, nivolumab, camrelizumab, toripalimab, tislelizumab, avelumab and duvalumab, whereas CTLA-4 inhibitors include tremelimumab and ipilimumab. The PD-1 inhibitor pembrolizumab is currently used as a first-line treatment for HNSCC. The findings of the KEYNOTE-048 trial indicated that PD-L1 biomarkers are useful for selection of appropriate treatments for HNSCC (30). In 2016, the United States Food and Drug Administration approved the use of the ICIs (PD-1) pembrolizumab and nivolumab for the treatment of R/M HNSCC based on the results of the KEYNOTE-012 and CheckMate 141 trials (3134), which indicated that anti-PD-1 monoclonal antibodies significantly prolong and improve overall survival in patients who show disease progression within 6 months of receiving platinum-based therapy (3537). In a study on preoperative induction therapy plus postoperative adjuvant immunization, patients in the intermediate-risk group continued pembrolizumab monotherapy depending on whether positive surgical margins or lymphatic tissue metastasis was present. The patients who received pembrolizumab throughout the course of treatment demonstrated significantly improved one-year disease-free survival and overall.

Targeted therapy for head and neck cancer

Basic biochemical pathways and mutant proteins play important roles in targeted antitumor therapies by blocking tumor cell growth and survival (38). The epidermal growth factor receptor (EGFR) is an important member of the complex receptor tyrosine kinase family and plays a major role in cell signaling. EGFR is involved in cell and organism growth regulation and is highly expressed in most HNSCCs (39). Drugs currently used in the targeted therapy for HNSCC include the monoclonal antibodies cetuximab, panitumumab and zalutumumab; the small-molecule tyrosine kinase inhibitors gefitinib and erlotinib; and the dual-target tyrosine kinase inhibitor lapatinib. In 2016, the FDA approved the use of cetuximab in combination with RT for the treatment of patients with locally advanced HNSCC and R/M HNSCC that do not to respond to platinum-based CT (40). Cetuximab is a clinically effective monoclonal antibody against EGFR (41). These monoclonal antibodies enhance tumor antigen presentation by forming immune complexes that enhance the induction of tumor-specific T cells (42). In addition, cetuximab promotes natural killer cell antibody-dependent cytotoxicity and enhance the tumor cell-killing ability of complement-dependent cytotoxicity (4345).

Ferroptosis

Ferroptosis is a form of iron-dependent cell death that was proposed in 2012 by Dixon et al (46). From a morphological and molecular biological perspective, ferroptosis is characterized by the presence of malformed small mitochondria, reduced mitochondrial crista, increased mitochondrial membrane density, rupture of the outer mitochondrial membrane, normal nuclear size, and a lack of condensed chromatin caused by overwhelming lipid peroxidation and oxidative disturbances in the intracellular microenvironment (13). Ferroptosis is mainly characterized by the presence of iron in the cell death execution and regulatory defense systems (47). Generally, the ferroptosis defense system can eliminate lipid peroxides and maintain a non-toxic state; however, if the amount of iron in the cell death execution system is higher than that in the ferroptosis defense system, the accumulation of lipid peroxides in the cell membrane increases to toxic levels, leading to ferroptosis (48). Ferroptosis gene regulators include drivers, repressors, markers and unclassified regulators (FerrDb; Fig. 1). Of these, only regulators play an important role in the ferroptosis regulatory network. The regulation network in the SLC7A11, GPX4 and acyl coenzyme A synthetase long-chain family member 4 (ACSL4) is highly expressed in the progression of the HNSCC cell line and reversible ferroptosis (Fig. 2A).

Iron metabolism

Ferroptosis is characterized by accumulation of iron ions and lipid peroxides. Iron is an essential trace element involved in redox activity in the human body. Increased levels of iron and/or iron-binding proteins and the dysregulation of iron metabolism contribute to the risk for cancer and promote tumor growth (49). Tumor cells are more dependent on iron than normal cells, and some tumor cells exhibit iron-ion aggregation. By iron, therefore, the steady state adjusts iron death by increasing iron intake; reduced iron can be stored and limit loss to promote iron death, as well as through the iron chelating agent and antioxidant to prevent death, effectively eliminating tumor cells (50). A recent study revealed that when iron metabolism disorder leads to increase in the amount of free iron in cells, the iron produced by Fenton's reaction catalyzes the production of ROS to further promote lipid peroxidation and induce ferroptosis (51).

Fatty acid oxidation

The underlying mechanism for ferroptosis is the iron-dependent accumulation of lipid peroxides. The ferroptosis defense system can inhibit lipid peroxidation under normal conditions. However, if the amount of iron in the cell death execution system is higher than that in the ferroptosis defense system, lipid peroxide rapidly accumulates in the cell membrane to toxic levels, triggering ferroptosis (48,52). Dysregulation of ferroptosis has been associated with numerous tumors, including HNSCC (5356) (Fig. 2B). In the iron metabolism execution system, polyunsaturated fatty acids (PUFA) are produced in cells through the catalytic formation of PUFA-phospholipid-peroxide (PUFA-PL-OOH), causing the accumulation of the lipid peroxide in the cell membrane (47,57,58). This accumulation of lipid peroxides disrupts the integrity of the membrane, thereby inducing ferroptosis.

SLC7A11/GPX4 axon-dependent system

SLC7A11/reduced glutathione (GSH)/GPX4 signaling axis is considered to be the cell death defense system. The theory of ferroptosis was initially based on the findings of research on this pathway (59,60). SLC7A11, also known as the system Xc-, is a cystine/glutamate transporter that reverses transport of proteins by mediating the exchange of intracellular glutamate with extracellular cystine (61). Cystine is exchanged with glutamate in the cell in a 1:1 ratio and is rapidly reduced to cysteine, which is involved in the synthesis of GSH and GPX4 within the cell (62). GSH is a key cofactor in GPX4 function, and its depletion disrupts cellular redox homeostasis, leading to accumulation of ROS and ultimately inducing the onset of ferroptosis. Therefore, inhibiting the expression of SLC7A11 can induce ferroptosis. Moreover, GPX4 is a key regulator of ferroptosis. The basic function of the enzymes in the GPX family members is to reduce H2O2 at the expense of GSH. GPX4 is the only enzyme that reduces cholesterol, hydrogen peroxide and oxidized fatty acids. GPX4 can convert reduced GSH to oxidized glutathione and lipid hydrogen peroxide (L-H2O2, toxic) to lipid alcohols (L-OH, non-toxic), thereby promoting the decomposition of hydrogen peroxide and inhibiting ferroptosis (63,64). This indicated that reduced GPX4 activity favors ferroptosis (65,66). The SLC7A11/GPX4 axon-dependent system constitutes a ferroptosis defense mechanism that maintains non-toxic lipid peroxides levels, thus sustaining cellular viability.

Non-SLC7A11/GPX4 axon-dependent systems: The non-SLC7A11/GPX4 axon-dependent system consists of three regulatory pathways: (i) NAD(P)H-FSP1-CoQ is a newly discovered ferroptosis defense system (67). Ferroptosis suppressor protein 1 (FSP1), also known as apoptosis-inducing factor mitochondria-associated 2, is the oxidoreductase of ubiquinone (CoQ) and is mainly located in the plasma membrane (6870). FSP1 acts mainly by catalyzing the reduction of CoQ into coenzyme Q (CoQH2) using NAD(P)H. Thereafter, CoQH2 exerts lipotropic and antioxidant effects, thereby inhibiting ferroptosis (71,72).

(ii) The GCH1-BH4-DHFR axis inhibits ferroptosis. Guanosine triphosphate cyclization hydrolase (GCH1) and guanosine-5′-triphosphate are the rate-limiting enzymes involved in the synthesis of tetrahydrobiopterin (BH4) (73). BH4, which is produced by dihydrofolate reductase through the reduction of dihydrogen biopterin (dihydrobiopterin, BH2), reduces the oxidation of endogenous free radicals and protects the lipid membrane from ferroptosis (74). The inhibition of GCH1 expression reduces BH4, thereby oxidizing iron and increasing GCH1 expression and BH4 synthesis, which inhibits ferroptosis (75).

(iii) The dihydrogen orotic acid dehydrogenase (DHODH)-CoQH 2 system inhibits ferroptosis by blocking mitochondrial lipid peroxidation (76). DHODH is mainly located in the mitochondrial membrane and inhibits ferroptosis through the reduction of CoQ into CoQH2, thus reducing the production of ROS.

Relationship between HNSCC treatment and ferroptosis

Relationship between ferroptosis and RT

DNA damage is one of the most important effects of IR in cells. IR-induced damage may directly affect cell proliferation and reduce the water content inside cells (~80% of the cell is water) to produce ROS and indirectly cause DNA damage (~60–70%) (77,78). This is because IR causes cytoplasm damage and generates highly active OH free radicals and other ROS, including O2 and H2O2, which subsequently attack nucleic acids, lipids and proteins (79,80). Tumor cells are more susceptible to RT than normal cells owing to the high replication rate of tumor cells and defects in the DNA damage response (DDR) pathway (81,82). DNA DSB is the most serious type of DNA damage. Cell death may occur if a DSB is not repaired in a timely manner. In RT, the absorption of IR by water leads to the generation of ROS, which subsequently act on PUFA, leading to lipid peroxidation, peroxidation of membrane phospholipid lipids, and ultimately, ferroptosis (83). Therefore, RT can inhibit tumour progression by inducing iron death in tumour cells. It has been found that a variety of morphological features associated with iron death, such as shrunken mitochondria, increased mitochondrial membrane density and reduced mitochondrial cristae, were observed in tumour cells eliminated by radiation, including lung, breast, esophageal and ovarian cancers (84). Herrera et al (85) discussed the existing treatment of ovarian tumours with RT and the mechanisms by which RT mobilizes anticancer immunity. Lang et al (86) described iron death as a previously unappreciated mechanism of action of RT. Finally, the study named SLC7A11, a key regulator of iron mutations, as a mechanistic determinant of the synergistic effect of RT and immunotherapy (86). Furthermore, IR-induced ACSL4 expression increases PUFA-PL biosynthesis, which together with ROS, drives PUFA-PL peroxidation (PUFA-PL-OOH) and ferroptosis (87). SLC7A11 reduces ferroptosis by promoting the synthesis of GSH and reducing the production of L-OOH (88). RT increases the production of ROS, which can induce the activation the nuclear factor erythrocyte 2 related factor 2 (Nrf2)-heme oxygenase 1 (HO-1) pathway. The role of the Nrf2/HO-1 pathway in ferroptosis is bidirectional. A previous study showed that Nrf2 can activate SLC7A11, inhibit ferroptosis, and reduce the radiosensitivity of esophageal squamous cell carcinomas (89). Moreover, Wei et al (90) found that activation of the Nrf2/HO-1 pathway can increase Fe2+ levels in colorectal cancer cells and induce ferroptosis. Induction of ferroptosis in tumor cells is one way to enhance radiosensitivity. For example, pancreatic and renal cell carcinomas are sensitive to RT (91), which may be related to their dependence on cystine uptake (92). Inhibition of SLC7A11 and promotion of ferroptosis can increase the radiosensitivity of esophageal squamous cell carcinomas (93). A previous study found that IR may induce ferroptosis in tumor cells. This is because RT induces an increase in siderophiles, which can increase the sensitivity of tumor cells to RT.

RT for DSB is the most effective method of damaging and eliminating cancer cells; however, the intrinsic efficiency of tumor cells in DNA damage repair may lead to cellular resistance and impair therapeutic outcomes. Genes and proteins involved in DSB repair are targets of cancer therapy because their alteration, interaction, translocation and regulation can affect the repair process and render tumor cells more sensitive to RT. Therefore, targeting DNA damage repair as a means of sensitizing cancer cells to RT is a promising strategy for precise and effective treatment of patients with cancer.

Ferroptosis and radiosensitization

RT can eliminate tumor cells to a certain extent and remains one of the most effective non-surgical treatments for numerous tumors. However, reduced effects of RT on tumor cells is usually unavoidable because of RT resistance (RR), which is the reduction in the effectiveness of antitumor treatment (94). Tumor cells may show increased expression of antioxidant defense system-related proteins and ferroptosis to control the RT-induced abnormal increase in lipid peroxides, which leads to ferroptosis and RR (95,96). RR can lead to tumor recurrence, poor treatment response, poor prognosis, decreased quality of life and an increased treatment burden. Therefore, increased radiation sensitivity helps to reduce the incidence of adverse reactions through RT-induced tumor cell death.

IR-induced radiobiological effects are closely associated with ferroptosis. IR can induce ferroptosis in tumor cells, and the ROS produced by ferroptosis are involved in the regulation of tumor cell radiosensitivity and the tumor microenvironment (TME) (97). Local hypoxia and inherent or adaptive RR of tumor cells may lead to decreased radiosensitivity. Some KEAP1 mutant tumors rely on ferroptosis defense mechanisms, such as adaptive upregulation of FSP1/CoQ and inhibition of PUFA-PL synthesis, to avoid IR-induced ferroptosis, and thus develop RR (98). Hypoxia has long been recognized as a key regulator of RR. Due to impaired ribonucleotide reductase activity at low oxygen concentrations, reduced nucleotide levels lead to accumulation of single-stranded DNA at stalled replication forks and under replication stress. At the same time, hypoxic environments can also lead to a DDR that activates ATR-mediated and ATM-mediated downstream targets such as p53, H2AX and CHK-1/2 (99,100). Subsequently, this downstream signalling can shift cells to a less radiosensitive phase by inducing cell cycle arrest (101,102). Besides, RT-induced expression of SLC7A11 and GPX4 contributes to RR as an adaptive response that protects cells from ferroptosis. Therefore, depletion of SCL7A11 and GPX4 can induce ferroptosis and achieve radiosensitization (103,104). Chen et al (105) reported that suppressor of cytokine signaling 2 (SOCS2), a potential prognostic predictor of RT, promotes ferroptosis and increases the radiosensitivity of tumor cells by increasing the ubiquitination and degradation of SLC7A11. Thus, SOCS2 can promote the radiosensitization of tumor cells in vivo and in vitro (105).

Inactivation of ACSL4 impairs the biosynthesis of PUFA-PL, which in turn causes RR. Ferroptosis inducers (FINs), such as erastin, FIN and sulfasalazine, block the activation of the ferroptosis defense system, increase total intracellular iron content, promote ROS production, reduce glutathione concentration, and increase lipid peroxidation in radioresistant tumor cells, thereby enhancing the radiosensitivity of the cells by inducing ferroptosis (106108). A previous study demonstrated that FINs have a synergistic effect in tumor treatment (109). Class I FINs targeting SLC7A11, such as erastin and sulfasalazine (SAS); class II FINs targeting GPX 4, such as RSL3 and ML162; and class III FINs depleting CoQ and GPX 4, such as FIN 56, can induce tumor sensitivity to RT in vitro (110).

In radioresistant tumor cells, activation of the ferroptosis execution system or inhibition of the ferroptosis defense system can further enhance ferroptosis and inhibit the development of RR. Therefore, further research on the mechanisms related to ferroptosis are needed to clarify how to maximize the antitumor effects of targeted ferroptosis combined with radiation while minimizing damage to normal tissues.

Relationship between ferroptosis and other treatments

RT can eliminate tumor cells and activate antitumor immunity. The antitumor immune system activated by RT can further induce ferroptosis in tumor cells and inhibit tumor development. Jhunjhunwala et al found that dendritic cells are the most important antigen-presenting cells that can ingest, process and present antigens and activate CD8+ T cells (111). Activation of CD4+ T cells releases interferon gamma (IFN gamma) and activates the system Xc-, thereby promoting tumor lipid peroxidation and cell death (111,112). The combination of RT and some drugs can significantly induce ferroptosis in tumor cells compared with single drug therapy, leading to a considerable increase in the number of immune cells in the tumor tissue (113).

Cisplatin is a platinum-based drug commonly used as a first-line chemotherapeutic agent for the treatment of solid tumors. Owing to its wide antitumor spectrum and high curative effect, cisplatin is recommended by the World Health Organization for the treatment of cancer. Cisplatin can bind with guanine residues induced between multiple chain and chain stated, a crosslinking that leads to rapid cell death (114). In addition, Ma et al (115) found that iron-oxide nanocarriers enhance the anticancer efficacy of cisplatin and simultaneously reduce toxicity caused by generation of ROS. Cisplatin can produce H2O2 through a cascade in the cytoplasm of the cells in the TME. H2O2 can be further catalyzed by ferric iron ions into toxic hydroxyl free radicals generated by Fenton's reaction, leading to tumor cell apoptosis and ferroptosis (115).

Ferroptosis induced by the tumor suppressor p53 inhibits tumor development. The induction of p53 in the presence of lipid peroxidation may eliminate cells stressed by ferroptosis. Ferroptosis can induce cell death by releasing PUFA into the extracellular environment or by driving the expression of enzymes that stimulate PUFA-PL synthesis, such as LPCAT3 and ACSL4 (116). Sulfasalazine can inhibit the expression of glutathione by downregulating SLC7A11, thereby inactivating GPX4, causing ROS accumulation, and inducing ferroptosis to play a role as a tumor suppressor (117).

Ferroptosis is a newly discovered mode of programmed cell death that is closely related to RT and combined immunotherapy. Drugs can play a role in tumor suppression by inducing ferroptosis in tumor cells. The underlying mechanism involves inducing ferroptosis in tumor cells in a variety of ways and eventually inhibiting tumor progression.

Conclusion and perspective

HNSCC is the most common malignant head and neck tumor, with recurrence and metastasis rates of >65% and a five-year survival rate of <50% (118). The combination of surgery and RT has increased the survival rate for HNSCC over the past 20 years. However, the first-line therapeutic agents used for HNSCC, such as platinum-based agents, 5-fluorouracil, polyene paclitaxel, and cetuximab, have little effect on most patients. Therefore, effective treatment of patients with HNSCC to improve their quality of life and prognosis remains a challenge.

RT currently plays an important role in the treatment of HNSCC. Continuous improvement in RT technology will allow for more accurate dosing and mapping of the radiation area in patients with HNSCC. However, there are still certain limitations: How to make the radiation dose received by HNSCC more precise and reduce the damage to the surrounding normal tissues needs further exploration. In addition, RT resistance is a major barrier to improving the survival benefit of HNSCC treatment; therefore, methods to reduce RT resistance need to be explored. Ferroptosis plays a key role in radiation-induced cell death. Induction of ferroptosis can enhance the radiosensitivity of tumor cells by inducing iron overload and lipid peroxidation, thereby maintaining the efficacy of RT.

Research on ferroptosis will help solve the major problems associated with HNSCC treatment and identify novel therapeutic targets and strategies for the diagnosis and clinical treatment of HNSCC. In the future, continuous research shall be conducted by the authors and improved experimental protocols will be utilized to further explore the effects of RT and other combined immunotherapies to improve the prognosis of patients with HNSCC and minimize treatment-related side effects.

Acknowledgements

Not applicable.

Funding

Funding: No funding was received.

Availability of data and materials

Not applicable.

Authors' contributions

YF and XL wrote the manuscript. BY and ML were responsible for gathering the associated research and designing the review. YD, JW and SL collected and analyzed data. LGa, LGo and LL contributed to the study design, interpretation of the research articles, editing and critical revision of the manuscript. Data authentication is not applicable. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare they have no competing interests.

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 

Vousden KH and Lane DP: p53 in health and disease. Nat Rev Mol Cell Biol. 8:275–283. 2007. View Article : Google Scholar

3 

Gleber-Netto FO, Zhao M, Trivedi S, Wang J, Jasser S, McDowell C, Kadara H, Zhang J, Wang J, William WN Jr, et al: Distinct pattern of TP53 mutations in human immunodeficiency virus-related head and neck squamous cell carcinoma. Cancer. 124:84–94. 2018. View Article : Google Scholar : PubMed/NCBI

4 

Kandoth C, McLellan MD, Vandin F, Ye K, Niu B, Lu C, Xie M, Zhang Q, McMichael JF, Wyczalkowski MA, et al: Mutational landscape and significance across 12 major cancer types. Nature. 502:333–339. 2013. View Article : Google Scholar : PubMed/NCBI

5 

Berkers CR, Maddocks OD, Cheung EC, Mor I and Vousden KH: Metabolic regulation by p53 family members. Cell Metab. 18:617–633. 2013. View Article : Google Scholar : PubMed/NCBI

6 

Goldstein I and Rotter V: Regulation of lipid metabolism by p53-fighting two villains with one sword. Trends Endocrinol Metab. 23:567–575. 2012. View Article : Google Scholar : PubMed/NCBI

7 

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

8 

Saraniti C, Speciale R, Santangelo M, Massaro N, Maniaci A, Gallina S, Serra A and Cocuzza S: Functional outcomes after supracricoid modified partial laryngectomy. J Biol Regul Homeost Agents. 33:1903–1907. 2019.PubMed/NCBI

9 

Nissi L, Suilamo S, Kytö E, Vaittinen S, Irjala H and Minn H: Recurrence of head and neck squamous cell carcinoma in relation to high-risk treatment volume. Clin Transl Radiat Oncol. 27:139–146. 2021.

10 

Ma L, Men Y, Feng L, Kang J, Sun X, Yuan M, Jiang W and Hui Z: A current review of dose-escalated radiotherapy in locally advanced non-small cell lung cancer. Radiol Oncol. 53:6–14. 2019. View Article : Google Scholar

11 

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

12 

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

13 

Ma S, Henson ES, Chen Y and Gibson SB: Ferroptosis is induced following siramesine and lapatinib treatment of breast cancer cells. Cell Death Dis. 7:e23072016. View Article : Google Scholar : PubMed/NCBI

14 

Sun X, Niu X, Chen R, He W, Chen D, Kang R and Tang D: Metallothionein-1G facilitates sorafenib resistance through inhibition of ferroptosis. Hepatology. 64:488–500. 2016. View Article : Google Scholar : PubMed/NCBI

15 

Guo J, Xu B, Han Q, Zhou H, Xia Y, Gong C, Dai X, Li Z and Wu G: Ferroptosis: A novel anti-tumor action for cisplatin. Cancer Res Treat. 50:445–460. 2018. View Article : Google Scholar

16 

Shi Y, Wei W, Li L, Wei Q, Jiang F, Xia G and Yu H: The global status of research in breast cancer liver metastasis: A bibliometric and visualized analysis. Bioengineered. 12:12246–12262. 2021. View Article : Google Scholar : PubMed/NCBI

17 

Schaue D and McBride WH: Opportunities and challenges of radiotherapy for treating cancer. Nat Rev Clin Oncol. 12:527–540. 2015. View Article : Google Scholar

18 

Sierko E, Hempel D, Zuzda K and Wojtukiewicz MZ: Personalized radiation therapy in cancer pain management. Cancers (Basel). 11:3902019. View Article : Google Scholar

19 

Stevens S, Moloney S, Blackmore A, Hart C, Rixham P, Bangiri A, Pooler A and Doolan P: IPEM topical report: Guidance for the clinical implementation of online treatment monitoring solutions for IMRT/VMAT. Phys Med Biol. 68:10.1088/1361–6560/acecd0. 2023. View Article : Google Scholar

20 

Gao S, Xu Q, Lan Y and He L: Recurrent trichilemmal carcinoma of the periorbital region treated with IMRT radiotherapy: A case report and a review of literature. Medicine (Baltimore). 102:e340382023. View Article : Google Scholar : PubMed/NCBI

21 

Machiels JP, René Leemans C, Golusinski W, Grau C, Licitra L and Gregoire V; EHNS Executive Board: ESMO Guidelines Committee, : ESTRO Executive Board: Reprint of ‘Squamous cell carcinoma of the oral cavity, larynx, oropharynx and hypopharynx: EHNS-ESMO-ESTRO Clinical Practice Guidelines for diagnosis, treatment and follow-up’. Oral Oncol. 113:1050422021. View Article : Google Scholar

22 

Galluzzi L, Vitale I, Aaronson SA, Abrams JM, Adam D, Agostinis P, Alnemri ES, Altucci L, Amelio I, Andrews DW, et al: Molecular mechanisms of cell death: Recommendations of the Nomenclature Committee on Cell Death 2018. Cell Death Differ. 25:486–541. 2018. View Article : Google Scholar : PubMed/NCBI

23 

Tang D, Kang R, Berghe TV, Vandenabeele P and Kroemer G: The molecular machinery of regulated cell death. Cell Res. 29:347–364. 2019. View Article : Google Scholar : PubMed/NCBI

24 

Kroemer G, Galluzzi L, Vandenabeele P, Abrams J, Alnemri ES, Baehrecke EH, Blagosklonny MV, El-Deiry WS, Golstein P, Green DR, et al: Classification of cell death: Recommendations of the Nomenclature committee on cell death 2009. Cell Death Differ. 16:3–11. 2009. View Article : Google Scholar : PubMed/NCBI

25 

Povirk LF: Biochemical mechanisms of chromosomal translocations resulting from DNA double-strand breaks. DNA Repair (Amst). 5:1199–1212. 2006. View Article : Google Scholar : PubMed/NCBI

26 

Mladenov E, Magin S, Soni A and Iliakis G: DNA double-strand break repair as determinant of cellular radiosensitivity to killing and target in radiation therapy. Front Oncol. 3:1132013. View Article : Google Scholar

27 

Vermorken JB, Remenar E, van Herpen C, Gorlia T, Mesia R, Degardin M, Stewart JS, Jelic S, Betka J, Preiss JH, et al: Cisplatin, fluorouracil, and docetaxel in unresectable head and neck cancer. N Engl J Med. 357:1695–1704. 2007. View Article : Google Scholar : PubMed/NCBI

28 

Brizel DM and Esclamado R: Concurrent chemoradiotherapy for locally advanced, nonmetastatic, squamous carcinoma of the head and neck: Consensus, controversy, and conundrum. J Clin Oncol. 24:2612–2617. 2006. View Article : Google Scholar

29 

Ghosh C, Luong G and Sun Y: A snapshot of the PD-1/PD-L1 pathway. J Cancer. 12:2735–2746. 2021. View Article : Google Scholar : PubMed/NCBI

30 

Ferris RL and Licitra L: PD-1 immunotherapy for recurrent or metastatic HNSCC. Lancet. 394:1882–1884. 2019. View Article : Google Scholar

31 

Seiwert TY, Burtness B, Mehra R, Weiss J, Berger R, Eder JP, Heath K, McClanahan T, Lunceford J, Gause C, et al: Safety and clinical activity of pembrolizumab for treatment of recurrent or metastatic squamous cell carcinoma of the head and neck (KEYNOTE-012): An open-label, multicentre, phase 1b trial. Lancet Oncol. 17:956–965. 2016. View Article : Google Scholar

32 

Cohen EEW, Soulières D, Le Tourneau C, Dinis J, Licitra L, Ahn MJ, Soria A, Machiels JP, Mach N, Mehra R, et al: Pembrolizumab versus methotrexate, docetaxel, or cetuximab for recurrent or metastatic head-and-neck squamous cell carcinoma (KEYNOTE-040): A randomised, open-label, phase 3 study. Lancet. 393:156–167. 2019. View Article : Google Scholar

33 

Burtness B, Harrington KJ, Greil R, Soulières D, Tahara M, de Castro G Jr, Psyrri A, Basté N, Neupane P, Bratland Å, et al: Pembrolizumab alone or with chemotherapy versus cetuximab with chemotherapy for recurrent or metastatic squamous cell carcinoma of the head and neck (KEYNOTE-048): A randomised, open-label, phase 3 study. Lancet. 394:1915–1928. 2019. View Article : Google Scholar

34 

Haddad RI, Harrington K, Tahara M, Ferris RL, Gillison M, Fayette J, Daste A, Koralewski P, Zurawski B, Taberna M, et al: Nivolumab plus ipilimumab versus EXTREME Regimen as First-Line treatment for Recurrent/Metastatic squamous cell carcinoma of the head and neck: The final results of CheckMate 651. J Clin Oncol. 41:2166–2180. 2023. View Article : Google Scholar

35 

Ferris RL, Blumenschein G Jr, Fayette J, Guigay J, Colevas AD, Licitra L, Harrington K, Kasper S, Vokes EE, Even C, et al: Nivolumab for recurrent squamous-cell carcinoma of the head and neck. N Engl J Med. 375:1856–1867. 2016. View Article : Google Scholar : PubMed/NCBI

36 

Ferris RL, Blumenschein G Jr, Fayette J, Guigay J, Colevas AD, Licitra L, Harrington KJ, Kasper S, Vokes EE, Even C, et al: Nivolumab vs investigator's choice in recurrent or metastatic squamous cell carcinoma of the head and neck: 2-year long-term survival update of CheckMate 141 with analyses by tumor PD-L1 expression. Oral Oncol. 81:45–51. 2018. View Article : Google Scholar

37 

Wise-Draper TM, Gulati S, Palackdharry S, Hinrichs BH, Worden FP, Old MO, Dunlap NE, Kaczmar JM, Patil Y, Riaz MK, et al: Phase II clinical trial of neoadjuvant and adjuvant pembrolizumab in resectable local-regionally advanced head and neck squamous cell carcinoma. Clin Cancer Res. 28:1345–1352. 2022. View Article : Google Scholar : PubMed/NCBI

38 

Druker BJ: David A. Karnofsky Award lecture. Imatinib as a paradigm of targeted therapies. J Clin Oncol. 21 (23 Suppl):239S–245S. 2003. View Article : Google Scholar

39 

Xu MJ, Johnson DE and Grandis JR: EGFR-targeted therapies in the post-genomic era. Cancer Metastasis Rev. 36:463–473. 2017. View Article : Google Scholar : PubMed/NCBI

40 

Parmar K, Mohamed A, Vaish E, Thawani R, Cetnar J and Thein KZ: Immunotherapy in head and neck squamous cell carcinoma: An updated review. Cancer Treat Res Commun. 33:1006492022. View Article : Google Scholar : PubMed/NCBI

41 

Dougan M and Dranoff G: Immune therapy for cancer. Annu Rev Immunol. 27:83–117. 2009. View Article : Google Scholar

42 

Correale P, Botta C, Cusi MG, Del Vecchio MT, De Santi MM, Gori Savellini G, Bestoso E, Apollinari S, Mannucci S, Marra M, et al: Cetuximab ± chemotherapy enhances dendritic cell-mediated phagocytosis of colon cancer cells and ignites a highly efficient colon cancer antigen-specific cytotoxic T-cell response in vitro. Int J Cancer. 130:1577–1589. 2012. View Article : Google Scholar : PubMed/NCBI

43 

Maréchal R, De Schutter J, Nagy N, Demetter P, Lemmers A, Devière J, Salmon I, Tejpar S and Van Laethem JL: Putative contribution of CD56 positive cells in cetuximab treatment efficacy in first-line metastatic colorectal cancer patients. BMC Cancer. 10:3402010. View Article : Google Scholar

44 

Dechant M, Weisner W, Berger S, Peipp M, Beyer T, Schneider-Merck T, Lammerts van Bueren JJ, Bleeker WK, Parren PW, van de Winkel JG and Valerius T: Complement-dependent tumor cell lysis triggered by combinations of epidermal growth factor receptor antibodies. Cancer Res. 68:4998–5003. 2008. View Article : Google Scholar : PubMed/NCBI

45 

Hsu YF, Ajona D, Corrales L, Lopez-Picazo JM, Gurpide A, Montuenga LM and Pio R: Complement activation mediates cetuximab inhibition of non-small cell lung cancer tumor growth in vivo. Mol Cancer. 9:1392010. View Article : Google Scholar : PubMed/NCBI

46 

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

47 

Wang Y, Wang Y, Pan J, Gan L and Xue J: Ferroptosis, necroptosis, and pyroptosis in cancer: Crucial cell death types in radiotherapy and post-radiotherapy immune activation. Radiother Oncol. 184:1096892023. View Article : Google Scholar

48 

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

49 

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

50 

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

51 

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

52 

Zheng J and Conrad M: The metabolic underpinnings of ferroptosis. Cell Metab. 32:920–937. 2020. View Article : Google Scholar : PubMed/NCBI

53 

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

54 

Roh JL, Kim EH, Jang H and Shin D: Nrf2 inhibition reverses the resistance of cisplatin-resistant head and neck cancer cells to artesunate-induced ferroptosis. Redox Biol. 11:254–262. 2017. View Article : Google Scholar

55 

Kim EH, Shin D, Lee J, Jung AR and Roh JL: CISD2 inhibition overcomes resistance to sulfasalazine-induced ferroptotic cell death in head and neck cancer. Cancer Lett. 432:180–190. 2018. View Article : Google Scholar

56 

Lin R, Zhang Z, Chen L, Zhou Y, Zou P, Feng C, Wang L and Liang G: Dihydroartemisinin (DHA) induces ferroptosis and causes cell cycle arrest in head and neck carcinoma cells. Cancer Lett. 381:165–175. 2016. View Article : Google Scholar

57 

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

58 

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

59 

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

60 

Dixon SJ, Patel DN, Welsch M, Skouta R, Lee ED, Hayano M, Thomas AG, Gleason CE, Tatonetti NP, Slusher BS and Stockwell BR: Pharmacological inhibition of cystine-glutamate exchange induces endoplasmic reticulum stress and ferroptosis. Elife. 3:e025232014. View Article : Google Scholar : PubMed/NCBI

61 

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

62 

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

63 

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

64 

Shi JF, Liu Y, Wang Y, Gao R, Wang Y and Liu J: Targeting ferroptosis, a novel programmed cell death, for the potential of alcohol-related liver disease therapy. Front Pharmacol. 14:11943432023. View Article : Google Scholar

65 

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 : PubMed/NCBI

66 

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

67 

Bersuker K, Hendricks JM, Li Z, Magtanong L, Ford B, Tang PH, Roberts MA, Tong B, Maimone TJ, Zoncu R, et al: The CoQ oxidoreductase FSP1 acts parallel to GPX4 to inhibit ferroptosis. Nature. 575:688–692. 2019. View Article : Google Scholar : PubMed/NCBI

68 

Wu M, Xu LG, Li X, Zhai Z and Shu HB: AMID, an apoptosis-inducing factor-homologous mitochondrion-associated protein, induces caspase-independent apoptosis. J Biol Chem. 277:25617–25623. 2002. View Article : Google Scholar : PubMed/NCBI

69 

Marshall KR, Gong M, Wodke L, Lamb JH, Jones DJ, Farmer PB, Scrutton NS and Munro AW: The human apoptosis-inducing protein AMID is an oxidoreductase with a modified flavin cofactor and DNA binding activity. J Biol Chem. 280:30735–30740. 2005. View Article : Google Scholar : PubMed/NCBI

70 

Elguindy MM and Nakamaru-Ogiso E: Apoptosis-inducing factor (AIF) and its family member protein, AMID, are rotenone-sensitive NADH:Ubiquinone oxidoreductases (NDH-2). J Biol Chem. 290:20815–20826. 2015. View Article : Google Scholar : PubMed/NCBI

71 

Doll S, Freitas FP, Shah R, Aldrovandi M, da Silva MC, Ingold I, Goya Grocin A, Xavier da Silva TN, Panzilius E, Scheel CH, et al: FSP1 is a glutathione-independent ferroptosis suppressor. Nature. 575:693–698. 2019. View Article : Google Scholar : PubMed/NCBI

72 

Shimada K, Skouta R, Kaplan A, Yang WS, Hayano M, Dixon SJ, Brown LM, Valenzuela CA, Wolpaw AJ and Stockwell BR: Global survey of cell death mechanisms reveals metabolic regulation of ferroptosis. Nat Chem Biol. 12:497–503. 2016. View Article : Google Scholar

73 

Xu J, Wu Y, Song P, Zhang M, Wang S and Zou MH: Proteasome-dependent degradation of guanosine 5′-triphosphate cyclohydrolase I causes tetrahydrobiopterin deficiency in diabetes mellitus. Circulation. 116:944–953. 2007. View Article : Google Scholar : PubMed/NCBI

74 

Soula M, Weber RA, Zilka O, Alwaseem H, La K, Yen F, Molina H, Garcia-Bermudez J, Pratt DA and Birsoy K: Metabolic determinants of cancer cell sensitivity to canonical ferroptosis inducers. Nat Chem Biol. 16:1351–1360. 2020. View Article : Google Scholar

75 

Kraft VAN, Bezjian CT, Pfeiffer S, Ringelstetter L, Müller C, Zandkarimi F, Merl-Pham J, Bao X, Anastasov N, Kössl J, et al: GTP Cyclohydrolase 1/Tetrahydrobiopterin counteract ferroptosis through lipid remodeling. ACS Cent Sci. 6:41–53. 2020. View Article : Google Scholar

76 

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

77 

Ward JF: DNA damage produced by ionizing radiation in mammalian cells: Identities, mechanisms of formation, and reparability. Prog Nucleic Acid Res Mol Biol. 35:95–125. 1988. View Article : Google Scholar

78 

Santivasi WL and Xia F: Ionizing radiation-induced DNA damage, response, and repair. Antioxid Redox Signal. 21:251–259. 2014. View Article : Google Scholar

79 

Azzam EI, Jay-Gerin JP and Pain D: Ionizing radiation-induced metabolic oxidative stress and prolonged cell injury. Cancer Lett. 327:48–60. 2012. View Article : Google Scholar

80 

Reisz JA, Bansal N, Qian J, Zhao W and Furdui CM: Effects of ionizing radiation on biological molecules-mechanisms of damage and emerging methods of detection. Antioxid Redox Signal. 21:260–292. 2014. View Article : Google Scholar

81 

Adjemian S, Oltean T, Martens S, Wiernicki B, Goossens V, Van den Berghe T, Cappe B, Ladik M, Riquet FB, Heyndrickx L, et al: Ionizing radiation results in a mixture of cellular outcomes including mitotic catastrophe, senescence, methuosis, and iron-dependent cell death. Cell Death Dis. 11:10032020. View Article : Google Scholar : PubMed/NCBI

82 

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

83 

Yang WS, Kim KJ, Gaschler MM, Patel M, Shchepinov MS and Stockwell BR: Peroidation of polyunsaturated fatty acids by lipoxygenases drives ferroptosis. Proc Natl Acad Sci USA. 113:E4966–E4975. 2016. View Article : Google Scholar : PubMed/NCBI

84 

Lei G, Mao C, Yan Y, Zhuang L and Gan B: Ferroptosis, radiotherapy, and combination therapeutic strategies. Protein Cell. 12:836–857. 2021. View Article : Google Scholar

85 

Herrera FG, Irving M, Kandalaft LE and Coukos G: Rational combinations of immunotherapy with radiotherapy in ovarian cancer. Lancet Oncol. 20:e417–e433. 2019. View Article : Google Scholar

86 

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

87 

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

88 

Wang H, An P, Xie E, Wu Q, Fang X, Gao H, Zhang Z, Li Y, Wang X, Zhang J, et al: Characterization of ferroptosis in murine models of hemochromatosis. Hepatology. 66:449–465. 2017. View Article : Google Scholar : PubMed/NCBI

89 

Dong H, Qiang Z, Chai D, Peng J, Xia Y, Hu R and Jiang H: Nrf2 inhibits ferroptosis and protects against acute lung injury due to intestinal ischemia reperfusion via regulating SLC7A11 and HO-1. Aging (Albany NY). 12:12943–12959. 2020. View Article : Google Scholar : PubMed/NCBI

90 

Wei R, Zhao Y, Wang J, Yang X, Li S, Wang Y, Yang X, Fei J, Hao X, Zhao Y, et al: Tagitinin C induces ferroptosis through PERK-Nrf2-HO-1 signaling pathway in colorectal cancer cells. Int J Biol Sci. 17:2703–2717. 2021. View Article : Google Scholar

91 

Bump EA and Brown JM: Role of glutathione in the radiation response of mammalian cells in vitro and in vivo. Pharmacol Ther. 47:117–136. 1990. View Article : Google Scholar

92 

Zou Y and Schreiber SL: Progress in understanding ferroptosis and challenges in its targeting for therapeutic benefit. Cell Chem Biol. 27:463–471. 2020. View Article : Google Scholar

93 

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

94 

Steinbichler TB, Dudás J, Skvortsov S, Ganswindt U, Riechelmann H and Skvortsova II: Therapy resistance mediated by exosomes. Mol Cancer. 18:582019. View Article : Google Scholar : PubMed/NCBI

95 

Zhang W, Sun Y, Bai L, Zhi L, Yang Y, Zhao Q, Chen C, Qi Y, Gao W, He W, et al: RBMS1 regulates lung cancer ferroptosis through translational control of SLC7A11. J Clin Invest. 131:e1520672021. View Article : Google Scholar

96 

Yang M, Wu X, Hu J, Wang Y, Wang Y, Zhang L, Huang W, Wang X, Li N, Liao L, et al: COMMD10 inhibits HIF1α/CP loop to enhance ferroptosis and radiosensitivity by disrupting Cu-Fe balance in hepatocellular carcinoma. J Hepatol. 76:1138–1150. 2022. View Article : Google Scholar

97 

Zheng Z, Su J, Bao X, Wang H, Bian C, Zhao Q and Jiang X: Mechanisms and applications of radiation-induced oxidative stress in regulating cancer immunotherapy. Front Immunol. 14:12472682023. View Article : Google Scholar

98 

Koppula P, Lei G, Zhang Y, Yan Y, Mao C, Kondiparthi L, Shi J, Liu X, Horbath A, Das M, et al: A targetable CoQ-FSP1 axis drives ferroptosis- and radiation-resistance in KEAP1 inactive lung cancers. Nat Commun. 13:22062022. View Article : Google Scholar : PubMed/NCBI

99 

Olcina MM, Grand RJ and Hammond EM: ATM activation in hypoxia-causes and consequences. Mol Cell Oncol. 1:e299032014. View Article : Google Scholar

100 

Fallone F, Britton S, Nieto L, Salles B and Muller C: ATR controls cellular adaptation to hypoxia through positive regulation of hypoxia-inducible factor 1 (HIF-1) expression. Oncogene. 32:4387–4396. 2013. View Article : Google Scholar : PubMed/NCBI

101 

Olcina M, Lecane PS and Hammond EM: Targeting hypoxic cells through the DNA damage response. Clin Cancer Res. 16:5624–5629. 2010. View Article : Google Scholar : PubMed/NCBI

102 

Hammond EM, Asselin MC, Forster D, O'Connor JP, Senra JM and Williams KJ: The meaning, measurement and modification of hypoxia in the laboratory and the clinic. Clin Oncol (R Coll Radiol). 26:277–288. 2014. View Article : Google Scholar : PubMed/NCBI

103 

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

104 

Pan X, Lin Z, Jiang D, Yu Y, Yang D, Zhou H, Zhan D, Liu S, Peng G, Chen Z and Yu Z: Erastin decreases radioresistance of NSCLC cells partially by inducing GPX4-mediated ferroptosis. Oncol Lett. 17:3001–3008. 2019.

105 

Chen Q, Zheng W, Guan J, Liu H, Dan Y, Zhu L, Song Y, Zhou Y, Zhao X, Zhang Y, et al: SOCS2-enhanced ubiquitination of SLC7A11 promotes ferroptosis and radiosensitization in hepatocellular carcinoma. Cell Death Differ. 30:137–151. 2023. View Article : Google Scholar : PubMed/NCBI

106 

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

107 

Zhang Z, Lu M, Chen C, Tong X, Li Y, Yang K, Lv H, Xu J and Qin L: Holo-lactoferrin: The link between ferroptosis and radiotherapy in triple-negative breast cancer. Theranostics. 11:3167–3182. 2021. View Article : Google Scholar : PubMed/NCBI

108 

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

109 

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. 15L:469–484. 2020. View Article : Google Scholar

110 

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

111 

Jhunjhunwala S, Hammer C and Delamarre L: Antigen presentation in cancer: Insights into tumour immunogenicity and immune evasion. Nat Rev Cancer. 21:298–312. 2021. View Article : Google Scholar : PubMed/NCBI

112 

Wang W, Green M, Choi JE, Gijón M, Kennedy PD, Johnson JK, Liao P, Lang X, Kryczek I, Sell A, et al: CD8+T cells regulate tumour ferroptosis during cancer immunotherapy. Nature. 569:270–274. 2019. View Article : Google Scholar : PubMed/NCBI

113 

Xu L and Fan W: Efficacy of sorafenib combined with radiofrequency ablation in renal cancer and its effects on immunity and inflammation in patients. J BUON. 25:514–519. 2020.PubMed/NCBI

114 

Li S, Liu Y, Li J, Zhao X and Yu D: Mechanisms of Ferroptosis and application to head and neck squamous cell carcinoma treatments. DNA Cell Biol. 40:720–732. 2021. View Article : Google Scholar

115 

Ma P, Xiao H, Yu C, Liu J, Cheng Z, Song H, Zhang X, Li C, Wang J, Gu Z and Lin J: Enhanced cisplatin chemotherapy by iron oxide nanocarrier-mediated generation of highly toxic reactive oxygen species. Nano Lett. 17:928–937. 2017. View Article : Google Scholar

116 

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

117 

Gout PW, Buckley AR, Simms CR and Bruchovsky N: Sulfasalazine, a potent suppressor of lymphoma growth by inhibition of the x(c)-cystine transporter: A new action for an old drug. Leukemia. 15:1633–1640. 2001. View Article : Google Scholar

118 

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

Related Articles

Journal Cover

June-2024
Volume 51 Issue 6

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Feng Y, Li X, Yang B, Li M, Du Y, Wang J, Liu S, Gong L, Li L, Gao L, Gao L, et al: The role of ferroptosis in radiotherapy and combination therapy for head and neck squamous cell carcinoma (Review). Oncol Rep 51: 79, 2024
APA
Feng, Y., Li, X., Yang, B., Li, M., Du, Y., Wang, J. ... Gao, L. (2024). The role of ferroptosis in radiotherapy and combination therapy for head and neck squamous cell carcinoma (Review). Oncology Reports, 51, 79. https://doi.org/10.3892/or.2024.8738
MLA
Feng, Y., Li, X., Yang, B., Li, M., Du, Y., Wang, J., Liu, S., Gong, L., Li, L., Gao, L."The role of ferroptosis in radiotherapy and combination therapy for head and neck squamous cell carcinoma (Review)". Oncology Reports 51.6 (2024): 79.
Chicago
Feng, Y., Li, X., Yang, B., Li, M., Du, Y., Wang, J., Liu, S., Gong, L., Li, L., Gao, L."The role of ferroptosis in radiotherapy and combination therapy for head and neck squamous cell carcinoma (Review)". Oncology Reports 51, no. 6 (2024): 79. https://doi.org/10.3892/or.2024.8738