Spandidos Publications Logo
  • About
    • About Spandidos
    • Aims and Scopes
    • Abstracting and Indexing
    • Editorial Policies
    • Reprints and Permissions
    • Job Opportunities
    • Terms and Conditions
    • Contact
  • Journals
    • All Journals
    • Oncology Letters
      • Oncology Letters
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Oncology
      • International Journal of Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular and Clinical Oncology
      • Molecular and Clinical Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Experimental and Therapeutic Medicine
      • Experimental and Therapeutic Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Molecular Medicine
      • International Journal of Molecular Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Biomedical Reports
      • Biomedical Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Reports
      • Oncology Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular Medicine Reports
      • Molecular Medicine Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • World Academy of Sciences Journal
      • World Academy of Sciences Journal
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Functional Nutrition
      • International Journal of Functional Nutrition
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Epigenetics
      • International Journal of Epigenetics
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Medicine International
      • Medicine International
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
  • Articles
  • Information
    • Information for Authors
    • Information for Reviewers
    • Information for Librarians
    • Information for Advertisers
    • Conferences
  • Language Editing
Spandidos Publications Logo
  • About
    • About Spandidos
    • Aims and Scopes
    • Abstracting and Indexing
    • Editorial Policies
    • Reprints and Permissions
    • Job Opportunities
    • Terms and Conditions
    • Contact
  • Journals
    • All Journals
    • Biomedical Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Experimental and Therapeutic Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Epigenetics
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Functional Nutrition
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Molecular Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Medicine International
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular and Clinical Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular Medicine Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Letters
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • World Academy of Sciences Journal
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
  • Articles
  • Information
    • For Authors
    • For Reviewers
    • For Librarians
    • For Advertisers
    • Conferences
  • Language Editing
Login Register Submit
  • This site uses cookies
  • You can change your cookie settings at any time by following the instructions in our Cookie Policy. To find out more, you may read our Privacy Policy.

    I agree
Search articles by DOI, keyword, author or affiliation
Search
Advanced Search
presentation
Oncology Reports
Join Editorial Board Propose a Special Issue
Print ISSN: 1021-335X Online ISSN: 1791-2431
Journal Cover
February-2026 Volume 55 Issue 2

Full Size Image

Sign up for eToc alerts
Recommend to Library

Journals

International Journal of Molecular Medicine

International Journal of Molecular Medicine

International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.

International Journal of Oncology

International Journal of Oncology

International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.

Molecular Medicine Reports

Molecular Medicine Reports

Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.

Oncology Reports

Oncology Reports

Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.

Oncology Letters

Oncology Letters

Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.

Biomedical Reports

Biomedical Reports

Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.

Molecular and Clinical Oncology

Molecular and Clinical Oncology

International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.

World Academy of Sciences Journal

World Academy of Sciences Journal

Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.

International Journal of Functional Nutrition

International Journal of Functional Nutrition

Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.

International Journal of Epigenetics

International Journal of Epigenetics

Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.

Medicine International

Medicine International

An International Open Access Journal Devoted to General Medicine.

Journal Cover
February-2026 Volume 55 Issue 2

Full Size Image

Sign up for eToc alerts
Recommend to Library

  • Article
  • Citations
    • Cite This Article
    • Download Citation
    • Create Citation Alert
    • Remove Citation Alert
    • Cited By
  • Similar Articles
    • Related Articles (in Spandidos Publications)
    • Similar Articles (Google Scholar)
    • Similar Articles (PubMed)
  • Download PDF
  • Download XML
  • View XML
Review Open Access

Research progress on the regulation of ferroptosis in NPC (Review)

  • Authors:
    • Shuai Bai
    • Yanjie Guo
    • Jingling Qiang
    • Qiangfang Dai
    • Yanling Yang
  • View Affiliations / Copyright

    Affiliations: Department of Neurobiology, School of Basic Medicine, Yan'an Medical College, Yan'an University, Yan'an, Shaanxi 716000, P.R. China, Department of Neurology, Affiliated Hospital of Yan'an University, Yan'an, Shaanxi 716000, P.R. China
    Copyright: © Bai et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
  • Article Number: 33
    |
    Published online on: December 18, 2025
       https://doi.org/10.3892/or.2025.9038
  • Expand metrics +
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Metrics: Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )
Cited By (CrossRef): 0 citations Loading Articles...

This article is mentioned in:



Abstract

Ferroptosis is a novel form of iron‑dependent programmed apoptosis, characterized by dysregulated iron metabolism, impaired antioxidant defense systems and accumulation of lipid peroxidation products. Nasopharyngeal carcinoma (NPC) cells exhibit marked susceptibility to ferroptosis, and its induction can effectively suppress tumor progression, offering a potential therapeutic strategy for NPC. At the molecular level, ferroptosis‑related genes [such as Solute Carrier Family 7 Member 11 (SLC7A11), Glutamate‑Cysteine Ligase Modifier Subunit (GCLM) and Glutamate‑Cysteine Ligase Catalytic Subunit (GCLC)] are notably upregulated in NPC tissues compared with normal tissues, and their overexpression associates with poor patient prognosis, suggesting their utility as diagnostic or prognostic biomarkers. The present review systematically summarizes the molecular mechanisms of ferroptosis, elucidates its role in NPC pathogenesis and discusses ferroptosis‑targeted therapeutic approaches for NPC.

Introduction

Nasopharyngeal carcinoma (NPC) is a malignant tumor originating from the mucosal epithelium of the nasopharynx, exhibiting distinct geographical distribution patterns. It is highly prevalent in regions such as South China (for example, Guangdong, Guangxi, Fujian and Hunan), Southeast Asia and North Africa (1). The pathogenesis of NPC involves genetic susceptibility, environmental factors and Epstein-Barr virus (EBV) infection, with EBV being a well-established oncogenic driver (1). Currently, the standard treatment for NPC primarily consists of radiotherapy combined with cisplatin-based concurrent chemoradiotherapy. However, chemotherapy resistance and severe side effects notably limit clinical efficacy (2). Consequently, there is a need to develop novel therapeutic strategies that are more effective, less toxic and capable of overcoming drug resistance.

In 2012, Dixon et al (3) first described ferroptosis as a novel iron-dependent form of programmed cell death, distinct from apoptosis, necrosis and other cell death modalities (4,5). In recent years, ferroptosis has garnered notable attention in cancer therapeutics, demonstrating particular promise in NPC research (6–13). Accumulating evidence indicates that ferroptosis participates in NPC progression regulation through multiple signaling pathways. Notably, NPC cells exhibiting resistance to conventional therapies or possessing highly metastatic characteristics often demonstrate increased susceptibility to ferroptosis (14). Consequently, targeting ferroptosis and its key regulatory proteins may represent a breakthrough therapeutic strategy for NPC.

Ferroptosis is a novel form of iron-dependent programmed cell death that is distinct from apoptosis and autophagy (15,16). Its core mechanism involves the catalysis of lipid peroxidation (LPO) in membrane polyunsaturated fatty acids (PUFAs) by ferrous iron (Fe2+) or lipoxygenases (LOXs), leading to membrane damage and subsequent cell death. Characteristic morphological features include mitochondrial shrinkage and increased membrane density, while nuclear structure typically remains intact (17–19). Currently identified regulatory pathways of ferroptosis include: The system cystine/glutamate transporter (Xc)-/glutathione (GSH)/GSH peroxidase 4 (GPX4) axis; guanosine triphosphate (GTP) cyclohydrolase 1 (GCH1)/tetrahydrobiopterin (BH4) pathway; dihydroorotate dehydrogenase (DHODH)-mediated pathway; membrane-bound O-acyltransferase 1/2 (MBOAT1/2)-monounsaturated fatty acid (MUFA) regulation; Nrf2 signaling; LPO mechanisms and apoptosis-inducing factor mitochondria-associated 2 (FSP1 otherwise known as AIFM2) pathway. The present review systematically examines the molecular mechanisms and key signaling pathways of ferroptosis. By integrating the epidemiological characteristics and risk factors of NPC, the present review elucidates the relationship between ferroptosis and NPC pathogenesis. Furthermore, the present review discusses the potential therapeutic value of targeting ferroptosis in NPC treatment, providing a theoretical foundation for developing novel anti-tumor strategies.

Molecular mechanisms and regulatory pathways of ferroptosis

Ferroptosis has emerged as a prominent research focus in cell biology as a distinct form of programmed cell death. A precise understanding of its definition and characteristics forms the essential foundation for in-depth investigation of this cell death mechanism. Defined as an iron-overload and reactive oxygen species (ROS)-dependent cell death process driven by lipid peroxide accumulation, ferroptosis reveals two key pathogenic factors: Iron ions and ROS (20–24). Morphologically, ferroptosis exhibits unique ultrastructural features: Markedly shrunken mitochondria with increased membrane density, reduced or vanished mitochondrial cristae, outer mitochondrial membrane rupture and loss of plasma membrane integrity (25–27). These characteristics distinctly differentiate ferroptosis from other cell death modalities such as apoptosis and necroptosis. Notably, nuclear morphology typically remains intact during ferroptosis, contrasting with classical apoptotic nuclear fragmentation.

These distinctive features suggest that ferroptosis is regulated through specific molecular mechanisms. In the following sections, the present study systematically elaborates on the key metabolic pathways and molecular mechanisms governing ferroptosis regulation.

Iron metabolism pathway

Iron is important for human survival, participating in physiological activities in the forms of Fe3+ and Fe2+: It is not only a key participant in the electron transport chain during oxidative phosphorylation but also the core of heme in hemoglobin, responsible for oxygen transport in the blood. The majority of iron in the body is bound to proteins or stored by ferritin, with only a small amount of free iron forming the labile iron pool (LIP) (28). The transport, metabolism and storage of iron are tightly regulated because free Fe2+, with its redox activity, can promote the generation of ROS through the Fenton reaction, exacerbating LPO (29–32). Extracellular Fe3+ first binds to transferrin and enters cells via endocytosis mediated by transferrin receptor 1. Within the endosome-lysosome system, as the pH decreases, Fe3+ dissociates and is reduced to Fe2+ by the metal reductase STEAP3, then transported to the cytoplasm by the divalent metal transporter 1 (33,34). In the cytoplasm, Fe2+ can be reduced and stored as Fe3+ by ferritin, exported out of the cell via ferroportin (FPN1/SLC40A1), involved in the synthesis of iron-containing proteins or exist as part of the transient LIP (35). Iron homeostasis is vital for cell survival and iron overload disrupts this balance, thereby inducing ferroptosis (Fig. 1).

Ferroptosis pathway. Schematic
depicts the global ferroptosis regulatory network, integrating
antioxidant systems (such as, GSH-GPX4 and FSP1-CoQ10), iron/lipid
metabolism molecules, MBOAT1/2, and antioxidant/membrane repair
factors to cover key pro- and anti-ferroptosis links. GSH,
glutathione; GSSG, GSH oxide; GPX4, GSH peroxidase 4; GSR, GSH
reductase; FSP1, ferroptosis inhibitory protein 1; CoQ10,
ubiquinone; CoQ10H2, ubiquinol; DHODH, dihydroorotate
dehydrogenase; GTP, phosphohydrolases; GCH1, cyclohydrolase-1; BH2,
dihydrobiopterin; BH4, tetrahydrobiopterin; ACSL4, acyl-CoA
synthetase long-chain family member 4; LOX, lipoxygenase; LPCAT3,
lyso-phosphatidylcholine acyltransferase 3; PUFA, polyunsaturated
fatty acids; CoA, coenzyme A; PL-PUFA, acids-polyunsaturated
polyunsaturated fatty; HO-1, heme oxygenase 1; TRF1, transferrin
receptor; DMT1, divalent metal transporter 1; Keap1, Kelch-like ECH
associated protein 1; MBOAT1/2, o-acyltransferase 1/2; ESCRT,
endosomal sorting complex, required for transport; CHAM5/6, charged
multivesicular proteins 5 and 6; Glu, glutamic acid; PE-PUFA,
unsaturated fat phospholipids; FPN, ferroportin; ARE, antioxidant
response element; Se, Selenium; PL-PUFA-OOH, lipid hydroperoxide;
VK, vitamin K; VKH2, hydroquinone; CoQ10, ubiquinone; CoQ10H2,
ubiquinol; LPLAT, Lys phospholipid acyltransferase.

Figure 1.

Ferroptosis pathway. Schematic depicts the global ferroptosis regulatory network, integrating antioxidant systems (such as, GSH-GPX4 and FSP1-CoQ10), iron/lipid metabolism molecules, MBOAT1/2, and antioxidant/membrane repair factors to cover key pro- and anti-ferroptosis links. GSH, glutathione; GSSG, GSH oxide; GPX4, GSH peroxidase 4; GSR, GSH reductase; FSP1, ferroptosis inhibitory protein 1; CoQ10, ubiquinone; CoQ10H2, ubiquinol; DHODH, dihydroorotate dehydrogenase; GTP, phosphohydrolases; GCH1, cyclohydrolase-1; BH2, dihydrobiopterin; BH4, tetrahydrobiopterin; ACSL4, acyl-CoA synthetase long-chain family member 4; LOX, lipoxygenase; LPCAT3, lyso-phosphatidylcholine acyltransferase 3; PUFA, polyunsaturated fatty acids; CoA, coenzyme A; PL-PUFA, acids-polyunsaturated polyunsaturated fatty; HO-1, heme oxygenase 1; TRF1, transferrin receptor; DMT1, divalent metal transporter 1; Keap1, Kelch-like ECH associated protein 1; MBOAT1/2, o-acyltransferase 1/2; ESCRT, endosomal sorting complex, required for transport; CHAM5/6, charged multivesicular proteins 5 and 6; Glu, glutamic acid; PE-PUFA, unsaturated fat phospholipids; FPN, ferroportin; ARE, antioxidant response element; Se, Selenium; PL-PUFA-OOH, lipid hydroperoxide; VK, vitamin K; VKH2, hydroquinone; CoQ10, ubiquinone; CoQ10H2, ubiquinol; LPLAT, Lys phospholipid acyltransferase.

LPO

Lipids are the core structural components of cell membranes and organelle membranes. Under normal physiological conditions, lipid oxidation and reduction maintain a dynamic balance, but cellular carcinogenesis or external stimuli can disrupt this equilibrium (36). Ferroptosis, a novel form of cell death, is characterized by oxidative damage to PUFA-containing phospholipids in cell membranes (37,38). PUFAs are the primary substrates of LPO and their oxidation severely disrupts membrane structure and function. By contrast, MUFAs can antagonize ferroptosis by inhibiting LPO (39–41). Thus, PUFA levels regulate both LPO and susceptibility to ferroptosis.

At the molecular level, acyl-CoA synthetase long-chain family member 4 (ACSL4) and lysophosphatidylcholine acyltransferase 3 (LPCAT3) are key enzymes regulating PUFA incorporation into phospholipids. Inhibition or loss of their activity confers resistance to ferroptosis (42–44). Specifically, ACSL4 and LPCAT3 work synergistically to esterify arachidonic acid (AA) or adrenic acid (AdA) into phosphatidylethanolamine (PE). Subsequently, LOXs catalyze the formation of lipid hydroperoxides. The breakdown products of these peroxides attack proteins, induce plasma membrane rupture and ultimately drive ferroptosis (45–47) (Fig. 1).

System Xc-/GSH/GPX4 pathway

System Xc− is composed of SLC7A11 (light chain) and SLC3A2 (heavy chain). As an important antioxidant defense mechanism in cells, it carries out a central role in regulating ferroptosis (48–51). Due to the limited ability of cells to synthesize cysteine intracellularly, they mainly rely on the Xc− system to uptake extracellular cystine, this transporter effluxes intracellular glutamate and uptakes cystine at a 1:1 ratio and the latter is reduced to cysteine after entering the cell (52–54).

As a key precursor, cysteine, together with glutamate and glycine, is catalyzed by glutamate-cysteine ligase and GSH synthetase in sequence to synthesize GSH. GSH is an essential cofactor for GPX4, a selenium-dependent enzyme that can reduce toxic lipid peroxides (PL-PUFA-OOH) to harmless lipid alcohols (PL-PUFA-OH), maintaining redox homeostasis (49,50,55).

In addition, GSH reductase can regenerate oxidized GSH into reduced GSH, thereby maintaining the activity of GPX4 and blocking ferroptosis triggered by LPO (50,51,56). Therefore, the targeted regulation of the three-level defense network of cystine uptake, GSH synthesis and GPX4 function is an effective strategy for intervening in ferroptosis (Fig. 1).

The GCH1/BH4 pathway

GCH1-BH4 pathway is central to the regulation of ferroptosis (57). BH4 is generated from GTP through three enzymatic steps: GTP GCH1 catalyzes the formation of an intermediate, which then undergoes a cascade reaction involving 6-pyruvoyltetrahydropterin synthase and dihydrobiopterin (BH2) reductase to produce the final product. BH4 exhibits notable antioxidant activity and can directly neutralize lipid peroxides. As a rate-limiting enzyme, the expression level of GCH1 determines the sensitivity of cells to ferroptosis, its inhibition leads to impaired BH4 synthesis, triggering ROS accumulation and ferroptosis (58); conversely, overexpression promotes BH4 production, reduces ROS and confers resistance to ferroptosis (59,60).

BH4 and BH2 form a redox cycle that synergistically scavenges oxygen-free radicals to inhibit ferroptosis (61). This cycle is regulated by dihydrofolate reductase (DHFR), which uses NAD(P)H as a cofactor to reduce BH2 to BH4. Elevated BH4 levels can induce cellular lipid remodeling, reducing the proportion of phospholipids containing di-polyunsaturated fatty acyl groups (diPUFA) and thereby blocking ferroptosis (62).

Furthermore, BH4 enhances the biosynthesis of coenzyme Q10 (CoQ10) by promoting the synthesis of 4-hydroxybenzoic acid, associating the GCH1-BH4-DHFR pathway to the FSP1-CoQ10 axis and forming a synergistic anti-ferroptosis network (Fig. 1).

DHODH pathway

DHODH is another pathway involved in the resistance to ferroptosis (63). As a flavin-dependent protein, DHODH is located in the inner mitochondrial membrane. It can convert dihydroorotate into orotate, simultaneously reduce ubiquinone to ubiquinol and generate lipophilic antioxidants, thereby inhibiting the accumulation of LPO and carrying out an inhibitory role against ferroptosis (64). It is worth noting that there is a considerable synergistic effect between DHODH and GPX4 in mitochondria. The two work together to effectively reduce the degree of LPO and prevent ferroptosis from occurring in the inner mitochondrial membrane (57,65).

In the biosynthetic pathway of pyrimidine nucleotides, DHODH occupies a key position and carries out an indispensable role in the synthesis of DNA and RNA. Once DHODH is inhibited, this biosynthetic pathway will be disrupted, resulting in abnormal or even terminated synthesis of pyrimidine nucleotides (66). This further leads to a decrease in the availability of pyrimidine nucleotides, making the purine-pyrimidine base pairing process unable to proceed normally and ultimately severely hindering the synthesis of RNA (67). Since RNA is a cofactor of GSH, a decrease in the amount of RNA will lead to an increase in the amount of GSH. At this time, GPX4 can reduce the peroxidized lipids back to their original state. As the level of LPO decreases, the process of ferroptosis is inhibited and the incidence of ferroptosis also decreases accordingly (68,69) (Fig. 1).

MBOAT1/2-MUFA pathway

Liang et al (70) revealed that membrane-bound proteins MBOAT1 and MBOAT2 are novel sex hormone-dependent inhibitors of ferroptosis. As members of the lysophospholipid acyltransferase (LPLAT) family, they can specifically select MUFAs as substrates, catalyze their binding to lysophosphatidylethanolamine, increase intracellular PE-MUFA levels and reduce PE-PUFA levels.

PE-PUFAs are the preferred substrates for LPO, and their content directly affects the degree of LPO and cellular sensitivity to ferroptosis. Thus, MBOAT1 and MBOAT2 regulate the composition of unsaturated fatty acids in membrane phospholipids, reducing oxidizable PE-PUFAs and increasing stable PE-MUFAs, to form a defense against ferroptosis, effectively inhibiting its occurrence (71). Further studies showed that their expression and function are specifically regulated by sex hormone receptors: MBOAT1 is regulated by estrogen receptors, with activity related to estrogen signaling; MBOAT2 is regulated by androgen receptors, with function dependent on androgen signal activation. This makes their ferroptosis-inhibiting effect sex hormone-dependent, providing a new perspective for analyzing the differential regulation of ferroptosis under different sex or hormonal microenvironments (70,72,73). This mechanism does not rely on classical pathways (70) such as GPX4 or AIFM2, but acts by directly remodeling the fatty acid composition of membrane phospholipids, adding new insights into the complexity and diversity of the ferroptosis regulatory network (Fig. 1).

FSP1-COQ10-NAD(P)H pathway

FSP1, a type II nicotinamide adenine dinucleotide-H (NADH): quinone oxidoreductase (NDH-2) with N-terminal hydrophobic/membrane (aa 1–27), NADH oxidoreductase (aa 81–285) and FAD (aa 286–308) domains (74), is a key anti-ferroptosis factor. Its GSH-independent antioxidant pathway, parallel to GPX4, regulates iron metabolism and protects against iron-dependent death (75).

Guo et al (76–78) showed FSP1 inhibits ferroptosis by generating antioxidant CoQ, reducing intracellular oxidized CoQ; exogenous CoQ fails to reverse ferroptosis in FSP1-deficient cells, indicating an endogenous role for CoQ. FSP1 mediates the NAD(P)H pathway, reducing membrane-bound NAD(P)H-dependent ubiquinone (oxidized CoQ) to ubiquinol (reduced CoQ) (79) (Fig. 1).

Vitamin K, a lipophilic molecule with 2-methyl-1,4-naphthoquinone and polyisoprene side chains (plant-derived phylloquinone K1; animal/bacterial menaquinone K2), converts to hydroquinone (VKH2) for the vitamin K cycle (79,80–82). FSP1 acts as a vitamin K reductase, consuming NAD(P)H to produce VKH2, which inhibits ferroptosis by blocking LPO (83,84) (Fig. 1).

Additionally, FSP1 enhances membrane repair via the ESCRT-III-dependent pathway (CoQ-independent) to inhibit ferroptosis (85). Ferroptosis inducers trigger FSP1 to suppress tumor cell ferroptosis; FSP1 knockout blocks RSL3-induced plasma membrane expression of CHMP5/6, while CHMP5 overexpression reverses inducer- and FSP1 knockout-induced cell death (86–89) (Fig. 1).

The Nrf2 pathway

Nrf2, the core of cellular antioxidant responses, binds to antioxidant response elements (AREs) to promote downstream gene transcription (90). It carries out a key role in regulating ferroptosis as an important transcription factor against it, involved in iron, lipid and amino acid metabolism (91). Its regulated antioxidant effectors [such as heme oxygenase-1 (HO-1) and GSH] which contain AREs (92). Elevated ROS activate the p62-Keap1-Nrf2 pathway: Nrf2 dissociates from Keap1, translocates to the nucleus, binds to AREs, initiates transcriptional cascades and upregulates downstream antioxidant genes, which is important for maintaining redox balance and inhibiting ferroptosis (77) (Fig. 1).

Additionally, cytoplasmic platelet-activating factor (PAF) acetylhydrolase (II) specifically inhibits short-chain fatty acid oxidation, blocks oxidized phospholipid (such as PAF) accumulation by interfering with cellular redox capacity, thus inhibiting ferroptosis (78). whereas LPLAT disrupts lipid bilayers, increases membrane permeability and triggers ferroptosis (93) (Fig. 1).

The incidence status and risk factors of NPC

NPC is a malignant tumor originating from the mucosal epithelium of the nasopharynx. Its incidence shows obvious regional characteristics and is particularly high in Southeast Asia and North Africa (79). The pathogenesis of this disease is multifactorial, involving the complex interaction of various risk factors such as genetic susceptibility, environmental exposure, EBV infection and lifestyle (79).

Genetic and environmental risk factors
Genetic factors

Genetic factors carry out an important role in the pathogenesis of NPC. Epidemiological studies have shown that NPC exhibits notable familial aggregation. The risk of disease in first-degree relatives is markedly compared with that in the general population, indicating the key role of genetic susceptibility in the occurrence of this disease (80–82). Currently, it is considered that specific gene polymorphisms (such as the HLA gene cluster), mutations in tumor susceptibility genes (such as TP53) and epigenetic changes may jointly affect the susceptibility of an individual to NPC. In addition, genetic factors may interact with environmental factors (such as EBV infection), further increasing the risk of developing the disease (Table I).

Table I.

Summary of possible risk factors for NPC.

Table I.

Summary of possible risk factors for NPC.

Factor classificationSpecific factorsAssociated with NPC morbidity
Hereditary factorFamilial aggregationNotably higher risk
Environmental factorAir pollutantDNA damage and mutation
Occupational exposureExposure to chemical carcinogensIncrease of risk
Living habitSmokeThe risk of mucosal injury and canceration increases
Excessive drinkingThe formation of DNA adducts intensifies the risk
Dietary factorPickled foodThe formation of carcinogens increases the risk
High-temperature cookingSmoke causes cancer and promotes tumor occurrence
EBV infectionCore pathogenic factorsOncogenic protein expression, genomic instability
Genomic instabilityKey mechanismGene mutation, deletion and tumor occurrence and development

[i] NPC, nasopharyngeal carcinoma.

Environmental factors

Environmental exposure is one of the important risk factors for NPC. Long-term exposure to air pollutants (such as PM2.5, sulfur dioxide and nitrogen oxides) can lead to chronic inflammation of the nasopharyngeal mucosa and DNA damage, thus promoting malignant transformation (94). In addition, occupational exposure to certain chemical carcinogens (such as formaldehyde or benzo(a)pyrene) may also increase the risk of NPC. Viral infection carries out a central role in the development of NPC, especially EBV infection. EBV can infect nasopharyngeal epithelial cells. By expressing latent membrane proteins (LMP) 1 and 2 and oncogenic proteins such as EBV nuclear antigen 1 (EBNA1), it can interfere with the regulation of the cell cycle and induce genomic instability (86,95) (Table I).

Lifestyle habits

Unhealthy lifestyles are modifiable risk factors for NPC. Smoking can notably increase the risk of developing the disease. Carcinogens in tobacco, such as nitrosamines and polycyclic aromatic hydrocarbons, can directly damage the nasopharyngeal mucosa and promote the oncogenic effect of EBV (87,88). Excessive alcohol consumption may induce the formation of DNA adducts through metabolites (such as acetaldehyde), exacerbating mucosal damage (89). Dietary factors also play a key role. Long-term consumption of pickled foods (such as salted fish and cured meat) may increase the risk of NPC because they are rich in nitrites, which can be converted into the potent carcinogen N-nitroso compounds in the body (85). In addition, heterocyclic amines and polycyclic aromatic hydrocarbons produced by high-temperature cooking (such as grilling and smoking) may also promote tumorigenesis (Table I).

The influence of EBV infection on NPC

EBV belongs to the gammaherpesvirus family and is widely prevalent in the human population, with >90% of adults having been infected. It can establish a lifelong latent state in epithelial cells and B cells. EBV is closely associated with a variety of lymphoid malignancies, such as Burkitt's lymphoma, Hodgkin's lymphoma, B-cell lymphoma, NK/T-cell lymphoma, as well as two types of epithelial cancer: NPC and gastric cancer (96). EBV exhibits two distinctly different life cycle states: lytic (productive) and latent (persistent). During primary infection, EBV first replicates in nasopharyngeal epithelial cells, then crosses the epithelial layer of the nasopharyngeal lining, infects naive B cells and continues to replicate, and finally establishes a stable latent state in host cells in the form of histone-associated episomes. To maintain latent viruses and serve as a stable reservoir for EBV-induced tumorigenesis, latent viruses need to be periodically reactivated. In NPC, the prevalence of type 2 and type 3 EBV reaches 100%, and high-level viral reactivation is a risk factor for EBV-associated NPC (97). The interaction between EBV infection, environmental factors, and genetic factors is the core of the pathogenesis of NPC. Recent studies have shown that abortive lytic infection promotes the establishment of the latent state during primary infection and the development of EBV-associated tumors. In-depth exploration of the mechanisms by which EBV viral products drive the development of NPC will help design more effective EBV-targeted therapies (98,99).

During the latent period, the virus only expresses a small number of genes key for genome maintenance and regulation. Based on late gene expression profiles, EBV latency can be divided into four types (100,101). In NPC, EBV infection is largely in type 2 latency, during which EBNA1, LMP1 and LMP2 protein are expressed. At the same time, some non-coding RNAs are also expressed, such as EBV-encoded RNA (EBER), BamHI A rightward transcript (BART), and BART miRNA. These expressed viral genes provide signals necessary for the maintenance of replication and survival of both EBV and host cells. However, EBV lytic phase proteins, such as BGLF5 (DNase) and BALF3, cause host genome instability and play an important role in the tumorigenesis of NPC (100).

Regulation of ferroptosis in NPC by EBV

Ferroptosis is a novel form of programmed cell death, characterized by iron-dependent accumulation of lipid peroxides, which ultimately leads to cell death (102,103). The occurrence of ferroptosis is associated with the balance of intracellular iron metabolism, lipid metabolism and antioxidant systems. In the field of tumor research, abnormal regulation of ferroptosis is associated with the occurrence, development and therapeutic sensitivity of tumors (104–106). The regulatory role of EBV infection on ferroptosis in NPC is an important content that urgently needs to be supplemented and improved in current research. In the type 2 latency of EBV infection in NPC, the expressed EBNA1, LMP1, LMP2 proteins and non-coding RNAs such as EBER, BART and BART miRNA may carry out key roles in regulating the ferroptosis process (107–109).

EBNA1, as an important nuclear antigen during EBV latency, not only participates in the replication and maintenance of the viral genome but also affects the physiological functions of host cells by regulating intracellular signaling pathways. Studies have shown that EBNA1 can affect the expression of intracellular antioxidant-related genes by activating the NF-κB signaling pathway. The activation of the NF-κB signaling pathway can promote the expression of GPX4 (110–113). As a key inhibitor of ferroptosis, GPX4 can inhibit the occurrence of ferroptosis by reducing LPOs to non-toxic alcohols. Therefore, EBNA1 may upregulate the expression of GPX4 by activating the NF-κB signaling pathway, thereby inhibiting ferroptosis in NPC cells and providing favorable conditions for the survival of cancer cells (114).

As a transmembrane protein, LMP1 can mimic the signal transduction function of members of the tumor necrosis factor receptor family and activate a variety of intracellular signaling pathways, such as MAPK and PI3K/Akt (115,116). Among them, the activation of the PI3K/Akt signaling pathway can promote the degradation of intracellular iron regulatory protein 2 (IRP2). IRP2 can bind to the mRNAs of ferritin heavy chain (FTH1) and ferritin light chain (FTL), inhibiting their translation and thus reducing the synthesis of intracellular ferritin (117,118). Ferritin is an important protein for intracellular iron storage; a decrease in ferritin content leads to an increase in intracellular free iron levels, which in turn promotes the occurrence of ferroptosis. However, after LMP1 activates the PI3K/Akt signaling pathway, it can increase the expression of FTH1 and FTL by promoting the degradation of IRP2, thereby increasing the intracellular ferritin content and reducing free iron levels, which inhibits ferroptosis in NPC cells (119,120). In addition, LMP1 can also upregulate the expression of SLC7A11 by activating the MAPK signaling pathway. SLC7A11 is an important component of the Xc-, which can promote the entry of cystine into cells and provide raw materials for the synthesis of GSH. GSH is an important coenzyme for GPX4 to exert its antioxidant effect; an increase in GSH content can enhance the activity of GPX4 and further inhibit the occurrence of ferroptosis (121–124).

LMP2 protein mainly includes two subtypes: LMP2A and LMP2B, among which LMP2A carries out an important role in the occurrence and development of NPC. LMP2A can activate signaling pathways such as PI3K/Akt and MAPK by mimicking the signal transduction of the B-cell receptor and its regulation of ferroptosis may be similar to that of LMP1. In addition, studies have found that LMP2A can affect the expression of intracellular iron metabolism-related genes, such as downregulating the expression of TFR1 (125–128). TFR1 is an important receptor for iron uptake by cells; a decrease in TFR1 expression reduces iron uptake by cells, lowers intracellular iron levels and thereby inhibits the occurrence of ferroptosis.

EBER is a small non-coding RNA encoded by EBV, mainly including two types: EBER1 and EBER2. Although EBER does not encode proteins, it can regulate the physiological functions and signaling pathways of cells by interacting with a variety of host cell proteins. Studies have shown that EBER can induce the production of IFNs by activating the toll-like receptor 3 signaling pathway and IFN can affect the balance of intracellular iron metabolism and antioxidant systems. In addition, EBER can also interact with RNA-activated protein (RIG-I) to activate downstream signaling pathways and regulate the expression of associated genes, which may further affect ferroptosis. For example, after EBER activates the RIG-I signaling pathway, it can promote the production of pro-inflammatory cytokines and some pro-inflammatory cytokines can upregulate the expression of SLC7A11 and inhibit the occurrence of ferroptosis.

Regulation of ferroptosis by EBV lytic phase-related proteins

EBV lytic phase proteins, such as BGLF5 and BALF3, not only cause host genome instability but also may regulate ferroptosis in NPC cells (103). BGLF5 is a DNase expressed during the EBV lytic phase, which can degrade the DNA of host cells and viruses, leading to genome instability. Genome instability causes an increase in intracellular oxidative stress levels and oxidative stress is one of the important inducers of ferroptosis (129–131). By degrading DNA, BGLF5 may increase intracellular ROS levels; ROS can attack intracellular lipid molecules, trigger LPO reactions and thereby promote the occurrence of ferroptosis. In addition, BGLF5 may also regulate the cellular stress response by affecting the expression of intracellular DNA damage repair-related genes, further influencing the process of ferroptosis. For example, BGLF5 can inhibit the expression of DNA damage repair proteins, preventing cells from effectively repairing DNA damage, leading cells to be in a continuous stress state and increasing their sensitivity to ferroptosis (132,133).

BALF3 is a DNA helicase expressed during the EBV lytic phase and is involved in the replication and packaging of viral DNA. In the process of exerting its biological functions, BALF3 may affect the intracellular redox balance and iron metabolism. Studies have shown that BALF3 can interact with certain intracellular antioxidant proteins, inhibiting their activity, reducing the antioxidant capacity of cells and thereby increasing the sensitivity of cells to ferroptosis (134–136). In addition, BALF3 may also regulate the expression of iron metabolism-related genes, such as upregulating the expression of TFR1, increasing iron uptake by cells, raising intracellular iron levels and promoting the occurrence of ferroptosis (137,138).

In addition to BGLF5 and BALF3, the EBV lytic phase also expresses a variety of other proteins, such as ZEBRA (BZLF1) and RTA (BRLF1). These proteins carry out key roles in initiating EBV lytic infection and may also regulate ferroptosis. For example, ZEBRA can regulate the expression of intracellular oxidative stress and iron metabolism-related genes by activating a variety of signaling pathways, thereby influencing the occurrence of ferroptosis (139,140). RTA can also interact with host cell proteins, affecting the physiological functions and signaling pathways of cells, which may indirectly affect ferroptosis (141,142).

In summary, EBV can regulate the ferroptosis process of NPC cells from multiple aspects, including iron metabolism, lipid metabolism and antioxidant systems, through its latency-related molecules and lytic phase-related proteins. In-depth study of the mechanism by which EBV regulates ferroptosis in NPC can not only improve the research on the pathogenesis of EBV and NPC but also provide new targets and strategies for the treatment of NPC (102,143,144). For example, designing corresponding inhibitors or antagonists targeting key molecules or signaling pathways involved in EBV-regulated ferroptosis may enhance the sensitivity of NPC cells to ferroptosis and improve the therapeutic effect of NPC (145,146) (Table I).

The influence of genomic instability on NPC

Genomic instability refers to an increased tendency for errors in DNA replication or repair, leading to changes such as mutations and deletions in the genome. Causes for genomic instability include endogenous abnormal cellular processes and exogenous environmental factors (such as radiation, chemicals and viruses) (147). There is evidence indicating that genomic instability is key in the development of NPC and several factors can promote the genomic instability of NPC. For example, EBV can cause DNA damage, inhibit the DNA repair mechanism of infected cells and increase the risk of mutations and chromosomal abnormalities. Exposure to environmental factors such as tobacco smoke, alcohol, formaldehyde and nitrosamines is associated with DNA damage and genomic instability, which can trigger NPC (148–150). Genetic factors (such as mutations in tumor suppressor genes or DNA repair genes) can also increase the susceptibility to NPC by impairing the ability of a cell to maintain genomic stability.

As a malignant tumor, NPC has complex and diverse clinical manifestations and early diagnosis is difficult. The clinical manifestations of NPC vary with the progression of the disease course. In the early stage, NPC may have no obvious symptoms. Some patients with NPC may have mild symptoms such as blood in nasal discharge or nasal bleeding. These symptoms are often ignored by patients, thus delaying the best treatment opportunity. As the disease progresses, the symptoms of NPC gradually become more obvious, including nasal bleeding, ulcers or cauliflower-like masses, tinnitus, hearing loss, nasal congestion, persistent unilateral headache or eye symptoms (151,152). Currently, radiotherapy alone or in combination with chemotherapy is the main treatment method for NPC; however, a large number of patients succumb to NPC due to recurrence and tumor metastasis. Distant metastasis is an important cause of treatment failure and mortality in patients with NPC. Previous studies have shown the key role of ferroptosis in tumor metastasis, emphasizing the importance of ferroptosis in tumor growth and metastasis (13,153–155). Determining new anti-cancer strategies and discovering new drugs that induce ferroptosis will be beneficial for improving the cure rate of advanced patients with NPC (Table I).

The role and mechanism of ferroptosis in the occurrence of NPC

Role of abnormal iron metabolism in NPC

NPC development is associated with abnormal iron metabolism. Cancer cells, with higher iron demand, accumulate intracellular iron by increasing Trf for enhanced uptake and regulating genes to reduce iron transport/storage (156). High iron levels trigger Fenton reactions, leading to LPO accumulation, membrane damage and exacerbated ferroptosis (possibly via antioxidant system inhibition). Iron imbalance also fuels proliferation, oxidative stress and DNA damage, accelerating progression (Fig. 2).

The ferroptosis pathway associated
with NPC. The schematic illustrates the core ferroptosis regulatory
pathway in NPC, involving the GSH-GPX4 axis,
SLC7A11/SLC3A2-mediated cystine uptake and ROS balance, as well as
the regulatory role of FGF5 secreted by CAFs in the tumor
microenvironment. NPC, nasopharyngeal carcinoma; GSH, glutathione;
GSS, GSH oxide; GPX4, GSH peroxidase 4; SLC7A11, solute carrier
family 7 member 11; SLC3A2, solute carrier family 3 member 2; ROS,
reactive oxygen species; CAFs, cancer-associated fibroblasts; FGF5,
fibroblast growth factor.

Figure 2.

The ferroptosis pathway associated with NPC. The schematic illustrates the core ferroptosis regulatory pathway in NPC, involving the GSH-GPX4 axis, SLC7A11/SLC3A2-mediated cystine uptake and ROS balance, as well as the regulatory role of FGF5 secreted by CAFs in the tumor microenvironment. NPC, nasopharyngeal carcinoma; GSH, glutathione; GSS, GSH oxide; GPX4, GSH peroxidase 4; SLC7A11, solute carrier family 7 member 11; SLC3A2, solute carrier family 3 member 2; ROS, reactive oxygen species; CAFs, cancer-associated fibroblasts; FGF5, fibroblast growth factor.

Role of SLC7A11 in NPC

As a key Xc− component, SLC7A11 mediates cystine uptake and glutamate efflux; cystine is reduced to GSH synthesis, resisting ROS. In NPC, overexpressed EGFR stabilizes SLC7A11 (kinase-independently) to inhibit ferroptosis. SLC7A11 knockdown induces ROS accumulation, ferroptosis-related protein upregulation and subcellular changes (157). Sorafenib can activate the ATF4-CHOP signaling axis to upregulate the expression of CHOP protein; CHOP directly inhibits the expression of SLC7A11 (a key subunit of the cystine/glutamate antiporter system), ultimately enhancing cellular sensitivity to ferroptosis (158) (Fig. 2).

Role of the Nrf2 signaling pathway in NPC

Ferroptosis interacts with other pathways in regulating NPC, such as the FGF5/FGFR2/Nrf2 pathway inhibits ferroptosis and reduces cisplatin sensitivity. Cancer-associated fibroblasts (CAFs)-secreted FGF5 binds FGFR2, activating Nrf2 to inhibit ferroptosis (159), indicating a role for CAFs and potential targets (Fig. 2).

Role of GPX4 in NPC

GPX4, a key antioxidant enzyme converting lipid peroxides to inactive forms, has higher positivity in nasopharyngeal carcinoma (NPC) tissues than normal nasopharyngeal tissues (102). Its expression associates with clinical features of NPC: Higher in stage III–IV vs. I–II NPC, poorly vs. well-differentiated tumors, patients with vs. without lymph node metastasis, and those with poor vs. good radiotherapy response. Moreover, GPX4-positive NPC cases have lower 5-year survival rates than GPX4-negative cases (160), indicating high GPX4 as a poor prognosis marker for NPC (Fig. 2).

The application of ferroptosis in the treatment of NPC

PRMT4 reduces erastin-induced ferroptosis in cisplatin-resistant NPC cells via the Nrf2/GPX4 pathway

The protein arginine methyltransferase (Prmt) family catalyzes the methylation of arginine residues in both histones and non-histone proteins. As a key post-translational modification, arginine methylation is widely involved in regulating various cellular processes. Prmt4, also known as coactivator-associated arginine methyltransferase 1, is the first identified member of the PRMT family. It can catalyze the asymmetric dimethylation of arginine residues in protein substrates, carries out a key role in the regulation of gene transcription and participates in the modulation of multiple cellular processes (161).

Studies have shown that Prmt4 is overexpressed in a variety of tumors, including breast cancer, prostate cancer and colorectal cancer (162–164). Its overexpression activates key factors of multiple oncogenic signaling pathways, such as FOS, E2F1, Wnt/β-catenin and nuclear receptor coactivator 3 (NCOA3/AIB1), thereby creating a favorable microenvironment for tumor growth, invasion and metastasis. A study by Pu et al (161) further revealed that the upregulation of PRMT4 reduces the sensitivity of cisplatin-resistant NPC cells to erastin-induced ferroptosis through mitochondrial damage; additionally, the interaction between PRMT4 and Nrf2 promotes the enzymatic methylation activity of PRMT4. These findings indicate that m6A methylation enhances the stability of PRMT4 in cisplatin-resistant NPC cells, thereby affecting the erastin-induced ferroptosis process.

Currently, research on PRMT4 is mainly in the preclinical stage. Although notable achievements have been made in cellular and animal experiments, progress in clinical trials is relatively slow. The application of PRMT4 as a therapeutic target for tumors faces numerous challenges in clinical practice. Firstly, due to the complexity of the human physiological environment, PRMT4-targeted drugs are difficult to act precisely and tend to interfere with the normal physiological functions of healthy cells, leading to adverse reactions such as myelosuppression and liver injury. Secondly, tumor cells exhibit high heterogeneity, and there are key differences in PRMT4-related characteristics (including expression level, activity and associated signaling pathways) among tumor cells from patients with different types of cancer or even within the same cancer type, making it difficult to develop a unified and effective treatment regimen. Thirdly, long-term use of PRMT4 inhibitors may lead to tumor drug resistance; tumor cells can evade the effects of drugs through alternative pathways or gene mutations, thereby impairing therapeutic efficacy (161). Therefore, to promote the translational application of PRMT4-targeted tumor therapy in the future, it is necessary to optimize the specificity of drugs and reduce their toxic and side effects by leveraging structural biology and targeted delivery technologies; establish personalized diagnosis and treatment regimens through multi-omics stratification and predictive models; and clarify the mechanisms of drug resistance while exploring the combined use of PRMT4 inhibitors with ferroptosis inducers (such as erastin) and chemotherapeutic drugs (such as cisplatin) to reverse drug resistance (Table II).

Table II.

Analysis table of therapeutic-related elements and experimental effects.

Table II.

Analysis table of therapeutic-related elements and experimental effects.

Therapeutic factorSignaling pathwayKey moleculeIn vitro resultsIn vivo resultsTherapeutic effect(Refs.)
PRMT4The Nrf2/GPX4 pathwayUpregulates PRMT4, Nrf2 and GPX4Reduces ferroptosis induced by erastinReduce the susceptibility of cisplatin-resistant cells in NPC to ferroptosisPrevents the development of NPC(161)
α-SolanineThe HSP90α/p53 axisNo effect: HSP90α Upregulates: p53 Downregulates: GPX4, SLC7A11Inhibits cell proliferationInhibits the growth of tumor volume and weight and is non-toxic to miceInduces ferroptosis in nasopharyngeal carcinoma cells(165)
GSTM3Regulates GPX4Overexpression of GSTM3, upregulates: GPX4 and 4-HNEPromotes radiation-induced ferroptosis in cellsOverexpression of GSTM3 combined with IR treatment markedly reduced the size and weight of the tumorEnhances the radio-sensitivity of NPC(160)
BBRThe Xc-/GSH/GPX4 axisDownregulates: GPX4, SLC7A11 and SLC3A2Inhibits cell metastasisThe metastatic lesions decreased, and the protein expression of GPX4, SLC7A11 and SLC3A2 increasedInhibits the metastasis of NPC(187)
IsoquercitrinAMPK/NF-κB pathwayDownregulates: p-p65/p65, p-IκB/IκB, IL-1β and p-AMPK/AMPKInduces ferroptosisReduces tumor weight and decreases the expression of ferroptosis-related markersInhibits the occurrence of NPC(204,209)
CuBDirectly targets ferroptosis-related moleculesDownregulates: GPX4The contents of iron and lipid peroxides in cells increaseThe tumor volume shrank and no toxic or pathological changes were observed in the major organsInduces ferroptosis in NPC cells(227)
Disulfiram/copperROS/MAPK and ferroptosis pathwayUpregulates: p53, p21, BAX and ROSCell deathTumor growth was inhibited, but there was no change in the body weight of the miceInduces the death of NPC cells(228)
P4HA1Activates HMGCS1Upregulates: HMGCS1Enhances ferroptosis resistancePromotes the proliferation and survival of NPC cellsPromotes the progression of NPC(248)
IcaritinRegulates ferroptosisUpregulates: ACSL4 Downregulates: GPX4Promotes ROSaccumulation, leading to DNA damage and G2 phase arrest of NPC cells Increases the sensitivity of NPC cells to radiotherapy(255)

[i] PRMT4, protein arginine methyltransferase 4; Nrf2, nuclear factor erythroid 2-related factor 2; GPX4, glutathione peroxidase 4; HSP90α, heat shock protein 90 αfamily class A member 1; p53, tumour suppressor gene TP53; SLC7A11, recombinant solute carrier family 7, member 11; GSTM3, glutathione-S-transferase mu 3; 4-HNE, 4-hydroxynonenal; BBR, berberine; SLC3A2, recombinant solute carrier family 3, member 2; AMPK/NF-κB, AMP-dependent protein kinase/nuclear factor-κB; p-p65/p65, phosphorylated p65/NF-κB p65; p-IκB/IκB, phosphorylated IκB/NF-κB kinase; IL-1β, interleukin-1β; p-AMPK/AMPK, phosphorylated AMP-activated protein kinase/AMP-activated protein kinase; CuB, cucurbitacin B; p21,cyclin-dependent kinase inhibitor 1; BAX, Bcl-2-associated X protein; ROS, reactive oxygen species; P4HA1, prolyl 4-hydroxylase subunit α1; HMGCS1,3, -hydroxy-3-methylglutaryl coenzyme A synthetase 1; ACSL4, Acyl-CoA synthetase long-chain family member 4.

α-Solanine induces ferroptosis in NPC by targeting the HSP90α/p53 axis

Cui et al (165) subcutaneously injected HK-1 cells into BALB/c nude mice, and continuously administered α-solanine for 15 days after tumor formation (administration group). Analysis revealed that compared with the tumor-bearing non-drug-administered group, α-solanine was effective in inhibiting the growth rates of tumor volume and tumor weight. To determine whether the administered dose of α-solanine was toxic to the mice, the researchers also evaluated its effects on the body weight and internal organs of the mice. Treatment with α-solanine had no effect on the body weight of the mice and the detection of relevant indicators of the important organs (heart, liver and kidney) of the mice also showed no notable differences between the model group and the administration group, indicating that the drug was non-toxic. At the same time, the researchers further measured ferroptosis-related indicators of the mice after the administration of α-solanine. Consistent with the results of in vitro experiments (specifically, the in vitro experiments on HK-1 nasopharyngeal carcinoma cells treated with α-solanine, which were conducted as part of the same study to verify the regulatory effect of α-solanine on ferroptosis-related proteins), compared with the model group (tumor-bearing non-drug-administered group), GPX4 and SLC7A11 in the administration group were markedly downregulated. Moreover, α-solanine had no effect on the expression level of HSP90α in the xenograft tumors, while increasing the level of p53. This implies that α-solanine stimulates LPO in vivo, further confirming that α-solanine mainly induces the death of NPC cells through ferroptosis. In terms of clinical research progress, the anti-cancer potential of α-solanine has gradually attracted attention. Currently, studies have explored the potential value of belladonna extract combined with chemotherapy for advanced solid tumors, and preliminary results have shown that this combination regimen exhibits good tolerability and anti-angiogenic activity (165–167). In addition, α-solanine has demonstrated adjuvant therapeutic effects in chemotherapy experiments on breast cancer animal models (168). Meanwhile, research has also found that α-solanine exerts certain positive effects in the combined treatment studies of various types of cancer such as liver cancer and nasopharyngeal carcinoma, but the relevant applications have not yet entered the clinical trial phase (169–173) (Table II).

GSTM3 promotes radiation-induced ferroptosis by regulating GPX4 and enhances the radiosensitivity of NPC

GSH S-transferase mu 3 (GSTM3) is a member of the GSH S-transferase family and has multiple effects on the progression of various malignant tumors (160). To investigate the effect of GSTM3 on ionizing radiation (IR; induced ferroptosis in vivo, researchers subcutaneously injected GSTM3-overexpressing stable 5–8F cells into nude mice, leading to the formation of palpable tumors (174,175). Subsequently, the mice bearing xenograft tumors received conventional IR treatment. The results showed that the body weight of the mice remained stable throughout the treatment period (176,177). Compared with the control group, in the xenograft model, GSTM3 overexpression alone did not exhibit any effect on tumor growth, while IR effectively inhibited tumor growth. Notably, compared with the group treated with IR alone, the combination of GSTM3 overexpression and IR treatment led to a notable reduction in tumor size and weight. 4-Hydroxy-2-nonenal (4-HNE) serves as a ferroptosis marker reflecting the level of LPO. Immunohistochemical staining showed that IR moderately increased the contents of GSTM3 and 4-HNE (178–180). In addition, GSTM3 overexpression combined with IR treatment resulted in a marked increase in the signal level of 4-HNE (181,182). Overall, these results indicate that GSTM3 enhances IR-mediated ferroptosis and improves the radiosensitivity of NPC.

To the best of our knowledge, currently, clinical research on GSTM3 is relatively limited. Expression of GSTM3 is associated with the inhibition of breast cancer stem cell phenotypes and favorable clinical prognosis (183,184). Additionally, chemotherapeutic drugs can inhibit GSTM3 expression, which promotes the enrichment of breast cancer stem cells and ultimately leads to tumor recurrence and metastasis (182,185). In clinical application, due to the functional redundancy of the GSTM3 family and high heterogeneity of tumor cells, single-target intervention on GSTM3 shows poor efficacy, and it is difficult to develop a unified targeted treatment plan. In the future, it is necessary to analyze the functional cooperation mechanism between GSTM3 and its family members, and construct a patient stratification model using multi-omics technology to promote the translation of GSTM3-related targeted strategies from basic research to clinical practice, thereby providing a new direction for improving tumor treatment efficacy (186) (Table II).

Berberine (BBR) inhibits the metastasis of NPC through ferroptosis mediated by the Xc-/GSH/GPX4 axis system

Ferroptosis is triggered by LPO and is strictly regulated by SLC7A11 and SLC3A2, which are key components of the cystine-glutamate antiporter (187). Due to the inhibition of LPO, the downregulation of GPX4 can directly or indirectly trigger ferroptosis. To determine the specific molecular mechanism of BBR-induced ferroptosis, Wu et al (187) treated NPC cells with different concentrations of BBR and detected the mRNA and protein levels of GPX4, SLC7A11 and SLC3A2. The results showed that the protein levels of GPX4, SLC7A11 and SLC3A2 in S18 and 5–8F NPC cells decreased in a dose-dependent manner. Meanwhile, the mRNA levels of GPX4, SLC7A11 and SLC3A2 also decreased (188–190). Moreover, the use of deferoxamine (DFO) and Fer-1 reversed the expression levels of proteins and mRNAs associated with BBR-induced ferroptosis. In addition, the results of the study on the in vivo anti-metastatic effect of berberine (BBR) on nasopharyngeal carcinoma (NPC) cells (using a nude mouse xenograft model) showed that the number of metastatic lesions in the BBR treatment group was reduced compared with that in the control group (191). Moreover, compared with the control group, the proteins of GPX4, SLC7A11 and SLC3A2 were elevated, which was consistent with the in vitro results of the downregulation of the expression of GPX4, SLC7A11 and SLC3A2 in the BBR treatment group (190,192). These findings indicate that BBR considerably inhibits NPC metastasis both in vitro and in vivo. In conclusion, GPX4 is a major molecule and the Xc-/GPX4 axis system carries out an important role in BBR-induced ferroptosis of NPC cells (189,193).

In terms of clinical trial progress, studies have shown that BBR can inhibit high glucose-induced ferroptosis in cells associated with diabetic retinopathy by activating the Nrf2/HO-1/GPX4 pathway (194–196), providing a theoretical basis for its application in the treatment of associated diseases. Although in vitro and animal experiments have confirmed that BBR can inhibit tumor cell growth and induce ferroptosis (197–200), its clinical application in NPC still faces challenges. Specifically, the complex human physiological environment leads to differences in the pharmacokinetics of BBR between in vivo and in vitro settings, making it difficult to ensure that BBR acts precisely on NPC cells to induce ferroptosis while avoiding damage to normal cells. Additionally, NPC cells exhibit high heterogeneity, patients vary in their sensitivity to BBR and the expression of molecules related to the Xc−/GPX4 axis, which hinders the development of a unified and effective clinical treatment regimen (201–203). In the future, more large-scale, multi-center clinical trials are needed to investigate the safety and efficacy of BBR in humans, explore personalized treatment strategies and promote the translation of BBR from basic research to clinical application in NPC treatment (Table II).

Isoquercitrin promotes ferroptosis and oxidative stress in NPC through the AMPK/NF-κB pathway

Numerous studies have shown that the NF-κB pathway carries out an important role in the regulation of oxidative stress and ferroptosis (204–208). Based on this, Luo et al (209) speculated that isoquercitrin might play a promoting role in the oxidative stress and ferroptosis processes of NPC cells by regulating the NF-κB pathway (210,211). By detecting the expression levels of proteins associated with the NF-κB pathway, it was found that isoquercitrin markedly reduced the ratios of phosphorylated (p)-p65/p65 and p-IκB/IκB and simultaneously inhibited the expression of IL-1β. These results indicate that isoquercitrin can inhibit the activation of the NF-κB pathway (212,213).

In addition, as a key molecule regulating various metabolic processes (including oxidative stress), the activity of AMPK is also affected by isoquercitrin. Zhang et al (214) showed that isoquercitrin markedly reduced the ratio of p-AMPK/AMPK, indicating that it has an inhibitory effect on the activity of AMPK. These results suggest that isoquercitrin may carry out a role in NPC by inhibiting the AMPK/NF-κB p65 signaling axis. To further verify this mechanism, Luo et al (209) established a xenograft tumor model. Analysis revealed that isoquercitrin markedly reduced tumor weight, however, had no obvious effect on the body weight of the mice. At the same time, the level of LPO in the isoquercitrin treatment group was notably increased, while the expressions of ferroptosis-related markers (such as ATF4, xCT, GPX4 and HO-1) were considerably decreased, indicating that isoquercitrin can induce ferroptosis in vivo (215,216). In conclusion, isoquercitrin may inhibit tumorigenesis of NPC in vivo, enhance oxidative stress and promote ferroptosis by inhibiting the AMPK/NF-κB p65 signaling pathway (209,217).

To the best of our knowledge, to date, there are no publicly available dedicated clinical studies on isoquercitrin for NPC. However, research has explored its potential in other diseases and tumor types: for instance, in early clinical observations of colorectal cancer, isoquercitrin combined with chemotherapy showed a synergistic effect in inhibiting tumor growth without notably increasing adverse reactions, initially demonstrating its safety and potential for combined therapy (218). In small-sample trials for ulcerative colitis (a chronic inflammation-related disease), isoquercitrin alleviated intestinal inflammation by regulating the NF-κB pathway (219,220), this mechanism shares commonality with the ‘NF-κB pathway inhibition’ observed in NPC research (221), providing an indirect reference value. Nevertheless, to promote the application of isoquercitrin in NPC treatment, targeted Phase I and II clinical trials are still needed to verify its pharmacokinetic characteristics, the efficacy of monotherapy or combined therapy and long-term safety. Additionally, it is necessary to clarify the differences in efficacy across patients with NPC with different molecular subtypes, so as to provide a basis for formulating subsequent clinical protocols (222–226) (Table II).

The molecular mechanism and therapeutic potential of cucurbitacin B (CuB) in inducing ferroptosis of human NPC cells

As a key element in the mitochondrial respiratory chain, iron carries out an important role in the process of ferroptosis. Lipid hydroperoxides are considered to be the main driving force and marker of ferroptosis. To explore the mechanism of action of CuB, Huang et al (227) detected the concentrations of intracellular iron and lipid peroxides. The results showed that after treatment with CuB, the contents of intracellular iron and lipid peroxides increased considerably in a dose-dependent manner, and this effect could be effectively reversed by inhibitors such as DFO, CPX and Fer-1. In addition, GPX4, as a key regulatory factor of ferroptosis, its expression level decreased markedly after CuB treatment, further supporting the role of CuB in inducing ferroptosis.

To further verify the anti-tumor effect of CuB, Huang et al (227) established a human nasopharyngeal carcinoma (NPC) xenograft model using BALB/c nude mice. The experimental results showed that, compared with the control group, the tumor volume of the mice in the CuB treatment group was notably reduced, and there was no obvious change in the body weight of the mice, indicating that CuB has good tolerance. Through H&E staining observation of the main organs such as the heart, liver, spleen, lung and kidney, and analysis of blood biochemical indexes, it was found that all the mice treated with CuB did not show visceral organ toxicity or obvious pathological morphological changes. These results indicate that CuB has potential application value in the treatment of NPC and is a promising ferroptosis inducer.

To the best of our knowledge, there are currently no publicly available clinical trial data on CuB for NPC, and it has not yet entered the formal clinical trial phase, with only some preliminary explorations conducted. To promote the clinical translation of CuB in the future, targeted Phase II and III clinical trials are still needed to evaluate the objective response rate, progression-free survival, and long-term safety (especially the effects on the hematopoietic system and liver/kidney function) of its monotherapy or combined therapy regimens, thereby providing sufficient clinical evidence (Table II).

Disulfiram (DSF)/copper (Cu) induces antitumor activity against NPC cells and CAFs through ROS/MAPK and ferroptosis pathways

DSF is a drug clinically used for the treatment of alcoholism, which exerts its effect by inhibiting the activity of aldehyde dehydrogenase (228). Studies have shown that DSF has potential application value in cancer treatment. The combined action of DSF and Cu can induce the aggregation of NPL4, leading to complex cellular phenotypes and ultimately triggering cell death (229–231). Experiments have shown that DSF/Cu treatment upregulates the expression of p53 protein and its downstream targets p21 and BAX, and this effect can be reversed by the p53 inhibitor Pifithrin-α (232). In addition, DSF/Cu markedly increases the level of lipid ROS in 5–8F cells, and the ROS scavenger N-acetylcysteine can partially reverse this phenomenon. In the 5–8F xenograft model, DSF/Cu markedly inhibits tumor growth without causing changes in the body weight of the mice (233–235). These results indicate that DSF/Cu carries out an important role in the treatment of NPC by inducing ROS-mediated ferroptosis and has the potential to be used as an adjuvant therapeutic drug in clinical practice (236,237).

Although DSF/Cu shows promising prospects in NPC treatment research, its clinical application faces challenges: DSF has a short half-life and is easily metabolized into inactive substances after oral administration, making it difficult to maintain effective concentrations (238,239); the complex human physiological environment and pronounced individual differences in pharmacokinetics complicate the determination of precise dosages; additionally, tumor cell heterogeneity increases the difficulty of personalized treatment (240,241). In the future, it is necessary to deepen basic research to clarify the molecular mechanism of DSF/Cu-induced ferroptosis and differences across NPC subtypes, conduct Phase I–III clinical trials to verify its pharmacokinetics, efficacy and safety, screen patients sensitive to DSF/Cu with precision medicine technologies (such as genetic testing and liquid biopsy), and explore innovative drug delivery systems to improve pharmacokinetic properties of DSF, all to promote its development as a clinical adjuvant treatment for NPC (242,243) (Table II).

P4HA1 activates HMGCS1 to promote ferroptosis resistance and progression of NPC

P4H is a heterotetramer composed of P4HA subtypes (P4HA1, P4HA2 and P4HA3) and P4HB, forming P4H1, P4H2 and P4H3 holoenzymes respectively. It carries out a key role in the proline hydroxylation of procollagen, catalyzing the formation of hydroxyproline from the proline residues in the Xaa-Pro-Gly triplet, which is essential for the folding of procollagen into a stable triple-helix structure and its secretion out of the cell (244,245). In addition, P4H also regulates the hydroxylation modification of proteins containing collagen-like sequences (246,247) (such as AGO2).

Previous studies have found that P4HA1 is a novel regulator of ferroptosis. Its overexpression enhances the ferroptosis resistance of NPC cells by activating HMGCS1 (248,249). Meanwhile, the P4HA1/HMGCS1 regulatory axis also promotes the proliferation of NPC cells, as well as the survival and ferroptosis resistance of NPC cells that are separated from the extracellular matrix (248,250,251).

Currently, relevant research on P4HA1 is mostly in the stages of basic research and model validation, but preliminary attempts have been made in other cancer fields: A retrospective study on non-small cell lung cancer revealed that high expression of P4HA1 is associated with poor prognosis in patients, providing indirect evidence for its potential as a prognostic marker and therapeutic target (252,253); In early preclinical trials for pancreatic cancer, small-molecule inhibitors of P4HA1 were able to inhibit tumor growth without obvious toxicity, laying a foundation for research on P4HA1 in NPC (254). In the future, it is first necessary to conduct large-scale retrospective studies to clarify the association between P4HA1 expression and the prognosis, staging and treatment response of patients with NPC. On this basis, Phase I/II clinical trials of P4HA1 inhibitors should be advanced to evaluate their safety, pharmacokinetic characteristics and efficacy, thereby providing evidence for clinical translation (Table II).

Icaritin increases the radiosensitivity of NPC cells by regulating ferroptosis

Icaritin has a variety of pharmacological effects, including antioxidant effects, prevention and treatment of osteoporosis, improvement of cardiovascular function and protection against neurodegenerative damage (255,256). In terms of anti-tumor effects, icaritin has been proven to inhibit the proliferation and induce apoptosis of a variety of tumor cells. These effects make icaritin a potential anti-tumor drug.

Studies have shown that icaritin can regulate the expression of proteins associated with ferroptosis. For example, In NPC cells, the expression of ACSL4, a marker protein of ferroptosis, is upregulated, while the expression of GPX4 is downregulated. Similarly, when icaritin is combined with radiotherapy, it can markedly promote the accumulation of ROS in NPC cells (255,257,258). The accumulation of ROS will further lead to DNA damage, such as the upregulation of the expression of γ-H2AX (259,260). These damages will exacerbate the death of NPC cells, thereby improving the effect of radiotherapy. In addition, icaritin can also cause NPC cells to arrest in the G2 phase. This arrest will further affect the proliferation and division ability of cells, thus enhancing the killing effect of radiotherapy on NPC cells. The aforementioned studies indicate that icaritin may enhance the radiosensitivity of NPC cells by promoting the occurrence of ferroptosis (261,262).

However, there are still challenges in applying icariin to the clinical treatment of NPC: It remains unclear whether the mechanism by which icariin regulates ferroptosis and enhances radiosensitivity in experiments is fully applicable in the complex human environment (263,264). Additionally, there is a lack of clinical data to support balancing efficacy and adverse reactions (such as mucosal damage and myelosuppression) when combined with radiotherapy (256,266). To address these issues in the future, breakthroughs can be made in three aspects: Conducting clinical sample studies on NPC to verify the effect of icariin on ferroptosis-related pathways and the impact of molecular subtypes; developing local formulations for the nasopharynx (such as nano-sprays) to increase drug concentration in tumor tissues; and advancing small-sample Phase I/II clinical trials to explore the safe dosage and administration timing of icariin combined with radiotherapy (267) (Table II).

Summary and future prospects

Nasopharyngeal carcinoma (NPC) is a head and neck malignant tumor, with >70% of new cases occurring in East Asia and Southeast Asia (268–270), and its treatment has been a major focus of clinical and research attention (271). Traditional treatment methods, such as radiotherapy and chemotherapy, have achieved curative effects, but present side effects and drugs that need to be addressed. In recent years, with the in-depth study of the mechanism of cell death, ferroptosis, as a new type of programmed cell death, has provided a new perspective and strategy for the treatment of NPC (13,272–274).

Ferroptosis, also referred to as iron-dependent LPO-mediated cell death, is a unique form of cell death driven by iron ions and LPO. Its core characteristic is the intracellular accumulation of iron ions coupled with an uncontrolled LPO reaction, which ultimately results in cell membrane rupture and subsequent cell death. Compared with traditional cell death methods such as apoptosis and necrosis, ferroptosis has its own uniqueness in morphological, biochemical and genetic characteristics.

Numerous studies have shown that ferroptosis carries out an important role in the occurrence, development and treatment of NPC (10,275–278). Conversely, the abnormal iron metabolism and increased LPO levels in NPC cells make them more sensitive to ferroptosis. On the other hand, some chemotherapeutic drugs, such as cisplatin, have been shown to be able to induce ferroptosis in NPCnuo cells (279–281), thereby exerting an anti-tumor effect. Based on the important role of ferroptosis in NPC, researchers have begun to explore the application of ferroptosis inducers in the treatment of NPC. For example, Erastin, as a small molecule compound that can induce ferroptosis in a variety of cancer cells, has been proven to enhance the sensitivity of traditional anti-cancer drugs to NPC cells (161,282). In addition, some new chemotherapeutic drugs and targeted therapeutic drugs are also being developed. They induce ferroptosis in NPC cells by regulating ferroptosis-related signaling pathways, such as GPX4 and SLC7A11. Although ferroptosis inducers have shown great potential in the treatment of NPC, ferroptosis inhibitors also carry out a role that cannot be ignored. A number of studies have shown that ferroptosis inhibitors can protect normal cells from ferroptosis, thereby reducing the side effects of chemotherapeutic drugs (283–285). In addition, ferroptosis inhibitors can also be used in combination with ferroptosis inducers. By regulating the degree and speed of ferroptosis, a more precise therapeutic effect can be achieved.

Although the application of ferroptosis in the treatment of NPC has made progress, there are still several issues that remain to be solved. For example, how to precisely regulate the degree and speed of ferroptosis to achieve the best therapeutic effect; how to overcome the problem of drug resistance and improve the sensitivity of ferroptosis inducers; how to develop safer and more effective ferroptosis inducers and inhibitors and new treatment methods all need to go through clinical trials and evaluations to ensure their safety and effectiveness. In the future, with the in-depth study of the mechanism of ferroptosis and the development of new drugs, it is considered that ferroptosis will carry out a greater role in the treatment of NPC, and it is expected to achieve more precise and effective therapeutic effects.

Acknowledgements

Not applicable.

Funding

The present review was supported by the grants from Yan'an science and technology bureau (grant no. 2024SF-YBXM-037 Y.Y.L).

Availability of data and materials

Not applicable.

Authors' contributions

SB was responsible for writing and revising the manuscript. YG was responsible for the preparation of figures and revisions of the present review. QQ was responsible for the revisions of the present review. JQ was responsible for the revisions of the present review. YY was responsible for revising the article. All authors contributed to the article and all authors read and approved the final manuscript. Data authentication not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Lim CY, Ng GWY, Goh CK, Lee MKC, Cheong I, Ooi EE, Liu J, West RB, Loh KS and Tay JK: Impact of high-risk EBV strains on nasopharyngeal carcinoma gene expression. Oral Oncol. 157:1069412024. View Article : Google Scholar : PubMed/NCBI

2 

Yip PL, Lee AWM and Chua MLK: Adjuvant chemotherapy in nasopharyngeal carcinoma. Lancet Oncol. 24:713–715. 2023. View Article : Google Scholar : PubMed/NCBI

3 

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

4 

Lee S, Hwang N, Seok BG, Lee S, Lee SJ and Chung SW: Autophagy mediates an amplification loop during ferroptosis. Cell Death Dis. 14:4642023. View Article : Google Scholar : PubMed/NCBI

5 

Kinowaki Y, Taguchi T, Onishi I, Kirimura S, Kitagawa M and Yamamoto K: Overview of ferroptosis and synthetic lethality strategies. Int J Mol Sci. 22:92712021. View Article : Google Scholar : PubMed/NCBI

6 

Alborzinia H, Chen Z, Yildiz U, Freitas FP, Vogel FCE, Varga JP, Batani J, Bartenhagen C, Schmitz W, Büchel G, et al: LRP8-mediated selenocysteine uptake is a targetable vulnerability in MYCN-amplified neuroblastoma. EMBO Mol Med. 15:e180142023. View Article : Google Scholar : PubMed/NCBI

7 

Floros KV, Cai J, Jacob S, Kurupi R, Fairchild CK, Shende M, Coon CM, Powell KM, Belvin BR, Hu B, et al: MYCN-amplified neuroblastoma is addicted to iron and vulnerable to inhibition of the system Xc-/glutathione axis. Cancer Res. 81:1896–1908. 2021. View Article : Google Scholar : PubMed/NCBI

8 

Lu Y, Yang Q, Su Y, Ji Y, Li G, Yang X, Xu L, Lu Z, Dong J, Wu Y, et al: MYCN mediates TFRC-dependent ferroptosis and reveals vulnerabilities in neuroblastoma. Cell Death Dis. 12:5112021. 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 

Zhou J, Guo T, Zhou L, Bao M, Wang L, Zhou W, Tan S, Li G, He B and Guo Z: The ferroptosis signature predicts the prognosis and immune microenvironment of nasopharyngeal carcinoma. Sci Rep. 13:18612023. View Article : Google Scholar : PubMed/NCBI

11 

Teng Y, Gao L, Mäkitie AA, Florek E, Czarnywojtek A, Saba NF and Ferlito A: Iron, ferroptosis, and head and neck cancer. Int J Mol Sci. 24:151272023. View Article : Google Scholar : PubMed/NCBI

12 

Mao C, Wang M, Zhuang L and Gan B: Metabolic cell death in cancer: Ferroptosis, cuproptosis, disulfidptosis, and beyond. Protein Cell. 15:642–660. 2024. View Article : Google Scholar : PubMed/NCBI

13 

Luo T, Wang Y and Wang J: Ferroptosis assassinates tumor. J Nanobiotechnology. 20:4672022. View Article : Google Scholar : PubMed/NCBI

14 

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

15 

Li Y, Du Y, Zhou Y, Chen Q, Luo Z, Ren Y, Chen X and Chen G: Iron and copper: critical executioners of ferroptosis, cuproptosis and other forms of cell death. Cell Commun Signal. 21:3272023. View Article : Google Scholar : PubMed/NCBI

16 

Hadian K and Stockwell BR: SnapShot: Ferroptosis. Cell. 181:1188–1188.e1. 2020. View Article : Google Scholar : PubMed/NCBI

17 

Ma TL, Chen JX, Zhu P, Zhang CB, Zhou Y and Duan JX: Focus on ferroptosis regulation: Exploring novel mechanisms and applications of ferroptosis regulator. Life Sci. 307:1208682022. View Article : Google Scholar : PubMed/NCBI

18 

Gan B: Mitochondrial regulation of ferroptosis. J Cell Biol. 220:e2021050432021. View Article : Google Scholar : PubMed/NCBI

19 

Chen Y, Guo X, Zeng Y, Mo X, Hong S, He H, Li J, Fatima S and Liu Q: Oxidative stress induces mitochondrial iron overload and ferroptotic cell death. Sci Rep. 13:155152023. View Article : Google Scholar : PubMed/NCBI

20 

Wahida A and Conrad M: Ferroptosis: Under pressure! Curr Biol. 33:R269–R272. 2023. View Article : Google Scholar : PubMed/NCBI

21 

Lai L, Tan M, Hu M, Yue X, Tao L, Zhai Y and Li Y: Important molecular mechanisms in ferroptosis. Mol Cell Biochem. 480:639–658. 2025. View Article : Google Scholar : PubMed/NCBI

22 

Zhang XD, Liu ZY, Wang MS, Guo YX, Wang XK, Luo K, Huang S and Li RF: Mechanisms and regulations of ferroptosis. Front Immunol. 14:12694512023. View Article : Google Scholar : PubMed/NCBI

23 

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

24 

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

25 

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

26 

Chen X, Li J, Kang R, Klionsky DJ and Tang D: Ferroptosis: Machinery and regulation. Autophagy. 17:2054–2081. 2021. View Article : Google Scholar : PubMed/NCBI

27 

Seibt T, Wahida A, Hoeft K, Kemmner S, Linkermann A, Mishima E and Conrad M: The biology of ferroptosis in kidney disease. Nephrol Dial Transplant. 39:1754–1761. 2024. View Article : Google Scholar : PubMed/NCBI

28 

Dutt S, Hamza I and Bartnikas TB: Molecular mechanisms of iron and heme metabolism. Annu Rev Nutr. 42:311–335. 2022. View Article : Google Scholar : PubMed/NCBI

29 

Zhao Y, Yang M and Liang X: The role of mitochondria in iron overload-induced damage. J Transl Med. 22:10572024. View Article : Google Scholar : PubMed/NCBI

30 

Wang H, Zhang Z, Ruan S, Yan Q, Chen Y, Cui J, Wang X, Huang S and Hou B: Regulation of iron metabolism and ferroptosis in cancer stem cells. Front Oncol. 13:12515612023. View Article : Google Scholar : PubMed/NCBI

31 

Li S, Zhang G, Hu J, Tian Y and Fu X: Ferroptosis at the nexus of metabolism and metabolic diseases. Theranostics. 14:5826–5852. 2024. View Article : Google Scholar : PubMed/NCBI

32 

Venkataramani V: Iron homeostasis and metabolism: Two sides of a coin. Adv Exp Med Biol. 1301:25–40. 2021. View Article : Google Scholar : PubMed/NCBI

33 

Huang L, Bian M, Zhang J and Jiang L: Iron metabolism and ferroptosis in peripheral nerve injury. Oxid Med Cell Longev. 2022:59182182022. View Article : Google Scholar : PubMed/NCBI

34 

Capelletti MM, Manceau H, Puy H and Peoc'h K: Ferroptosis in liver diseases: An Overview. Int J Mol Sci. 21:49082020. View Article : Google Scholar : PubMed/NCBI

35 

Wu L, Xian X, Tan Z, Dong F, Xu G, Zhang M and Zhang F: The role of iron metabolism, lipid metabolism, and redox homeostasis in Alzheimer's disease: From the perspective of ferroptosis. Mol Neurobiol. 60:2832–2850. 2023. View Article : Google Scholar : PubMed/NCBI

36 

Lyamzaev KG, Panteleeva AA, Simonyan RA, Avetisyan AV and Chernyak BV: Mitochondrial lipid peroxidation is responsible for ferroptosis. Cells. 12:6112023. View Article : Google Scholar : PubMed/NCBI

37 

Do Q and Xu L: How do different lipid peroxidation mechanisms contribute to ferroptosis? Cell Rep Phys Sci. 4:1016832023. View Article : Google Scholar : PubMed/NCBI

38 

Naowarojna N, Wu TW, Pan Z, Li M, Han JR and Zou Y: Dynamic regulation of ferroptosis by lipid metabolism. Antioxid Redox Signal. 39:59–78. 2023. View Article : Google Scholar : PubMed/NCBI

39 

Xin S and Schick JA: PUFAs dictate the balance of power in ferroptosis. Cell Calcium. 110:1027032023. View Article : Google Scholar : PubMed/NCBI

40 

Cui S, Simmons G Jr, Vale G, Deng Y, Kim J, Kim H, Zhang R, McDonald JG and Ye J: FAF1 blocks ferroptosis by inhibiting peroxidation of polyunsaturated fatty acids. Proc Natl Acad Sci USA. 119:e21071891192022. View Article : Google Scholar : PubMed/NCBI

41 

Qiu B, Zandkarimi F, Bezjian CT, Reznik E, Soni RK, Gu W, Jiang X and Stockwell BR: Phospholipids with two polyunsaturated fatty acyl tails promote ferroptosis. Cell. 187:1177–1190.e18. 2024. View Article : Google Scholar : PubMed/NCBI

42 

Ma XH, Liu JH, Liu CY, Sun WY, Duan WJ, Wang G, Kurihara H, He RR, Li YF, Chen Y and Shang H: ALOX15-launched PUFA-phospholipids peroxidation increases the susceptibility of ferroptosis in ischemia-induced myocardial damage. Signal Transduct Target Ther. 7:2882022. View Article : Google Scholar : PubMed/NCBI

43 

Do Q, Zhang R, Hooper G and Xu L: Differential contributions of distinct free radical peroxidation mechanisms to the induction of ferroptosis. JACS Au. 3:1100–1117. 2023. View Article : Google Scholar : PubMed/NCBI

44 

Li Y, Li Z, Ran Q and Wang P: Sterols in ferroptosis: From molecular mechanisms to therapeutic strategies. Trends Mol Med. 31:36–49. 2025. View Article : Google Scholar : PubMed/NCBI

45 

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

46 

Sokol KH, Lee CJ, Rogers TJ, Waldhart A, Ellis AE, Madireddy S, Daniels SR, House RRJ, Ye X, Olesnavich M, et al: Lipid availability influences ferroptosis sensitivity in cancer cells by regulating polyunsaturated fatty acid trafficking. Cell Chem Biol. 32:408–422.e6. 2025. View Article : Google Scholar : PubMed/NCBI

47 

Hirata Y, Ferreri C, Yamada Y, Inoue A, Sansone A, Vetica F, Suzuki W, Takano S, Noguchi T, Matsuzawa A and Chatgilialoglu C: Geometrical isomerization of arachidonic acid during lipid peroxidation interferes with ferroptosis. Free Radic Biol Med. 204:374–384. 2023. View Article : Google Scholar : PubMed/NCBI

48 

Liu S, Zhang HL, Li J, Ye ZP, Du T, Li LC, Guo YQ, Yang D, Li ZL, Cao JH, et al: Tubastatin A potently inhibits GPX4 activity to potentiate cancer radiotherapy through boosting ferroptosis. Redox Biol. 62:1026772023. View Article : Google Scholar : PubMed/NCBI

49 

Nishida Xavier da Silva T, Friedmann Angeli JP and Ingold I: GPX4: Old lessons, new features. Biochem Soc Trans. 50:1205–1213. 2022. View Article : Google Scholar : PubMed/NCBI

50 

Chen T, Leng J, Tan J, Zhao Y, Xie S, Zhao S, Yan X, Zhu L, Luo J, Kong L and Yin Y: Discovery of novel potent covalent glutathione peroxidase 4 inhibitors as highly selective ferroptosis inducers for the treatment of triple-negative breast cancer. J Med Chem. 66:10036–10059. 2023. View Article : Google Scholar : PubMed/NCBI

51 

Rochette L, Dogon G, Rigal E, Zeller M, Cottin Y and Vergely C: Lipid peroxidation and iron metabolism: Two corner stones in the homeostasis control of ferroptosis. Int J Mol Sci. 24:4492022. View Article : Google Scholar : PubMed/NCBI

52 

Guo W, Li K, Sun B, Xu D, Tong L, Yin H, Liao Y, Song H, Wang T, Jing B, et al: Dysregulated glutamate transporter SLC1A1 propels cystine uptake via Xc− for glutathione synthesis in lung cancer. Cancer Res. 81:552–566. 2021. View Article : Google Scholar : PubMed/NCBI

53 

Li Z, Li Y, Yang Y, Gong Z, Zhu H and Qian Y: In vivo tracking cystine/glutamate antiporter-mediated cysteine/cystine pool under ferroptosis. Anal Chim Acta. 1125:66–75. 2020. View Article : Google Scholar : PubMed/NCBI

54 

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

55 

Wang Y, Zheng L, Shang W, Yang Z, Li T, Liu F, Shao W, Lv L, Chai L, Qu L, et al: Wnt/beta-catenin signaling confers ferroptosis resistance by targeting GPX4 in gastric cancer. Cell Death Differ. 29:2190–2202. 2022. View Article : Google Scholar : PubMed/NCBI

56 

Xia C, Xing X, Zhang W, Wang Y, Jin X, Wang Y, Tian M, Ba X and Hao F: Cysteine and homocysteine can be exploited by GPX4 in ferroptosis inhibition independent of GSH synthesis. Redox Biol. 69:1029992024. View Article : Google Scholar : PubMed/NCBI

57 

Wang F and Min J: DHODH tangoing with GPX4 on the ferroptotic stage. Signal Transduct Target Ther. 6:2442021. View Article : Google Scholar : PubMed/NCBI

58 

Hu Q, Wei W, Wu D, Huang F, Li M, Li W, Yin J, Peng Y, Lu Y, Zhao Q and Liu L: Blockade of GCH1/BH4 axis activates ferritinophagy to mitigate the resistance of colorectal cancer to erastin-induced ferroptosis. Front Cell Dev Biol. 10:8103272022. View Article : Google Scholar : PubMed/NCBI

59 

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 : PubMed/NCBI

60 

Feng Y, Feng Y, Gu L, Mo W, Wang X, Song B, Hong M, Geng F, Huang P, Yang H, et al: Tetrahydrobiopterin metabolism attenuates ROS generation and radiosensitivity through LDHA S-nitrosylation: Novel insight into radiogenic lung injury. Exp Mol Med. 56:1107–1122. 2024. View Article : Google Scholar : PubMed/NCBI

61 

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 : PubMed/NCBI

62 

Lv Y, Wu M, Wang Z and Wang J: Ferroptosis: From regulation of lipid peroxidation to the treatment of diseases. Cell Biol Toxicol. 39:827–851. 2023. View Article : Google Scholar : PubMed/NCBI

63 

Zhang S, Kang L, Dai X, Chen J, Chen Z, Wang M, Jiang H, Wang X, Bu S, Liu X, et al: Manganese induces tumor cell ferroptosis through type-I IFN dependent inhibition of mitochondrial dihydroorotate dehydrogenase. Free Radic Biol Med. 193:202–212. 2022. View Article : Google Scholar : PubMed/NCBI

64 

Amos A, Amos A, Wu L and Xia H: The Warburg effect modulates DHODH role in ferroptosis: A review. Cell Commun Signal. 21:1002023. View Article : Google Scholar : PubMed/NCBI

65 

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

66 

Desler C, Durhuus JA, Hansen TLL, Anugula S, Zelander NT, Bøggild S and Rasmussen LJ: Partial inhibition of mitochondrial-linked pyrimidine synthesis increases tumorigenic potential and lysosome accumulation. Mitochondrion. 64:73–81. 2022. View Article : Google Scholar : PubMed/NCBI

67 

Tarangelo A, Rodencal J, Kim JT, Magtanong L, Long JZ and Dixon SJ: Nucleotide biosynthesis links glutathione metabolism to ferroptosis sensitivity. Life Sci Alliance. 5:e2021011572022. View Article : Google Scholar : PubMed/NCBI

68 

Yang C, Zhao Y, Wang L, Guo Z, Ma L, Yang R, Wu Y, Li X, Niu J, Chu Q, et al: De novo pyrimidine biosynthetic complexes support cancer cell proliferation and ferroptosis defence. Nat Cell Biol. 25:836–847. 2023. View Article : Google Scholar : PubMed/NCBI

69 

Liu Y, Lu S, Wu LL, Yang L, Yang L and Wang J: The diversified role of mitochondria in ferroptosis in cancer. Cell Death Dis. 14:5192023. View Article : Google Scholar : PubMed/NCBI

70 

Liang D, Feng Y, Zandkarimi F, Wang H, Zhang Z, Kim J, Cai Y, Gu W, Stockwell BR and Jiang X: Ferroptosis surveillance independent of GPX4 and differentially regulated by sex hormones. Cell. 186:2748–64.e22. 2023. View Article : Google Scholar : PubMed/NCBI

71 

Sun S, Shen J, Jiang J, Wang F and Min J: Targeting ferroptosis opens new avenues for the development of novel therapeutics. Signal Transduct Target Ther. 8:3722023. View Article : Google Scholar : PubMed/NCBI

72 

Li P, Xu J, Xu B, Hu X, Xiong Y, Wang Y and Liu P: NR5A2 (located on chromosome 1q32) inhibits ferroptosis and promotes drug resistance by regulating phospholipid remodeling in multiple myeloma. Int J Biol Sci. 21:5789–5801. 2025. View Article : Google Scholar : PubMed/NCBI

73 

Phat NK, Tien NTN, Anh NK, Yen NTH, Lee YA, Trinh HKT, Le KM, Ahn S, Cho YS, Park S, et al: Alterations of lipid-related genes during anti-tuberculosis treatment: insights into host immune responses and potential transcriptional biomarkers. Front Immunol. 14:12103722023. View Article : Google Scholar : PubMed/NCBI

74 

Nguyen HP, Yi D, Lin F, Viscarra JA, Tabuchi C, Ngo K, Shin G, Lee AY, Wang Y and Sul HS: Aifm2, a NADH oxidase, supports robust glycolysis and is required for cold- and diet-induced thermogenesis. Mol Cell. 77:600–617.e4. 2020. View Article : Google Scholar : PubMed/NCBI

75 

Lv Y, Liang C, Sun Q, Zhu J, Xu H, Li X, Li YY, Wang Q, Yuan H, Chu B and Zhu D: Structural insights into FSP1 catalysis and ferroptosis inhibition. Nat Commun. 14:59332023. View Article : Google Scholar : PubMed/NCBI

76 

Guo J, Chen L and Ma M: Ginsenoside Rg1 suppresses ferroptosis of renal tubular epithelial cells in sepsis-induced acute kidney injury via the FSP1-CoQ10- NAD(P)H pathway. Curr Med Chem. 31:2119–2132. 2024. View Article : Google Scholar : PubMed/NCBI

77 

Mikac S, Dziadosz A, Padariya M, Kalathiya U, Fahraeus R, Marek-Trzonkowska N, Chruściel E, Urban-Wójciuk Z, Papak I, Arcimowicz Ł, et al: Keap1-resistant ΔN-Nrf2 isoform does not translocate to the nucleus upon electrophilic stress. bioRxiv. 2022.06. 10.495609. 2022.

78 

Zhang Q, Sun T, Yu F, Liu W, Gao J, Chen J, Zheng H, Liu J, Miao C, Guo H, et al: PAFAH2 suppresses synchronized ferroptosis to ameliorate acute kidney injury. Nat Chem Biol. 20:835–846. 2024. View Article : Google Scholar : PubMed/NCBI

79 

Su ZY, Siak PY, Lwin YY and Cheah SC: Epidemiology of nasopharyngeal carcinoma: Current insights and future outlook. Cancer Metastasis Rev. 43:919–939. 2024. View Article : Google Scholar : PubMed/NCBI

80 

Siak PY, Khoo AS, Leong CO, Hoh BP and Cheah SC: Current status and future perspectives about molecular biomarkers of nasopharyngeal carcinoma. Cancers (Basel). 13:34902021. View Article : Google Scholar : PubMed/NCBI

81 

Zheng MQ, Wang TM, Liao Y, Xue WQ, He YQ, Wu ZY, Yang DW, Li DH, Deng CM, Jia YJ, et al: Nasopharyngeal Epstein-Barr virus DNA loads in high-risk nasopharyngeal carcinoma families: Familial aggregation and host heritability. J Med Virol. 92:3717–3725. 2020. View Article : Google Scholar : PubMed/NCBI

82 

Liao Y, Tong XT, Zhou T, Xue WQ, Wang TM, He YQ, Zheng MQ, Jia YJ, Yang DW, Wu YX, et al: Unveiling familial aggregation of nasopharyngeal carcinoma: Insights from oral microbiome dysbiosis. Cell Rep Med. 6:1019792025. View Article : Google Scholar : PubMed/NCBI

83 

Li W, Liang L, Liu S, Yi H and Zhou Y: FSP1: A key regulator of ferroptosis. Trends Mol Med. 29:753–764. 2023. View Article : Google Scholar : PubMed/NCBI

84 

Ward NP and DeNicola GM: Long-sought mediator of vitamin K recycling discovered. Nature. 608:673–674. 2022. View Article : Google Scholar : PubMed/NCBI

85 

Feng H, Zhou Y, Wang L, Wang Y, Zhou S and Tian F: Consumption of processed food and risk of nasopharyngeal carcinoma: A systematic review and meta-analysis. Transl Cancer Res. 11:872–879. 2022. View Article : Google Scholar : PubMed/NCBI

86 

Zhong L, Krummenacher C, Zhang W, Hong J, Feng Q, Chen Y, Zhao Q, Zeng MS, Zeng YX, Xu M, Zhang X, et al: Urgency and necessity of Epstein-Barr virus prophylactic vaccines. NPJ Vaccines. 7:1592022. View Article : Google Scholar : PubMed/NCBI

87 

Lin JH, Wen CP, Jiang CQ, Yuan JM, Chen CJ, Ho SY, Gao W, Zhang W, Wang R, Chien YC, et al: Smoking and nasopharyngeal cancer: Individual data meta-analysis of six prospective studies on 334 935 men. Int J Epidemiol. 50:975–986. 2021. View Article : Google Scholar : PubMed/NCBI

88 

Arfaeinia H, Ghaemi M, Jahantigh A, Soleimani F and Hashemi H: Secondhand and thirdhand smoke: A review on chemical contents, exposure routes, and protective strategies. Environ Sci Pollut Res Int. 30:78017–78029. 2023. View Article : Google Scholar : PubMed/NCBI

89 

Feng R, Chang ET, Liu Q, Cai Y, Zhang Z, Chen G, Huang QH, Xie SH, Cao SM, Zhang Y, et al: Intake of alcohol and tea and risk of nasopharyngeal carcinoma: A population-based case-control study in Southern China. Cancer Epidemiol Biomarkers Prev. 30:545–553. 2021. View Article : Google Scholar : PubMed/NCBI

90 

George M, Reddy AP, Reddy PH and Kshirsagar S: Unraveling the NRF2 confusion: Distinguishing nuclear respiratory factor 2 from nuclear erythroid factor 2. Ageing Res Rev. 98:1023532024. View Article : Google Scholar : PubMed/NCBI

91 

Yan R, Lin B, Jin W, Tang L, Hu S and Cai R: NRF2, a superstar of ferroptosis. Antioxidants (Basel). 12:17392023. View Article : Google Scholar : PubMed/NCBI

92 

Yuan L, Wang Y, Li N, Yang X, Sun X, Tian He and Zhang Y: Mechanism of action and therapeutic implications of Nrf2/HO-1 in inflammatory bowel disease. Antioxidants (Basel). 13:10122024. View Article : Google Scholar : PubMed/NCBI

93 

Wang D, Tang L, Zhang Y, Ge G, Jiang X, Mo Y, Wu P, Deng X, Li L, Zuo S, et al: Regulatory pathways and drugs associated with ferroptosis in tumors. Cell Death Dis. 13:5442022. View Article : Google Scholar : PubMed/NCBI

94 

Huang HC, Tantoh DM, Hsu SY, Nfor ON, Frank CL, Lung CC, Ho CC, Chen CY and Liaw YP: Association between coarse particulate matter (PM10-2.5) and nasopharyngeal carcinoma among Taiwanese men. J Investig Med. 68:419–424. 2020. View Article : Google Scholar : PubMed/NCBI

95 

Xue WQ, Wang TM, Huang JW, Zhang JB, He YQ, Wu ZY, Liao Y, Yuan LL, Mu J and Jia WH: A comprehensive analysis of genetic diversity of EBV reveals potential high-risk subtypes associated with nasopharyngeal carcinoma in China. Virus Evol. 7:veab0102021. View Article : Google Scholar : PubMed/NCBI

96 

Damania B, Kenney SC and Raab-Traub N: Epstein-Barr virus: Biology and clinical disease. Cell. 185:3652–3670. 2022. View Article : Google Scholar : PubMed/NCBI

97 

Manners O, Murphy JC, Coleman A, Hughes DJ and Whitehouse A: Contribution of the KSHV and EBV lytic cycles to tumourigenesis. Curr Opin Virol. 32:60–70. 2018. View Article : Google Scholar : PubMed/NCBI

98 

Phan TG: Epstein-Barr virus and multiple sclerosis: The dawn of a new age. Clin Transl Immunology. 12:e14572023. View Article : Google Scholar : PubMed/NCBI

99 

Tyler KL: The enigmatic links between Epstein-Barr virus infection and multiple sclerosis. J Clin Invest. 132:e1604682022. View Article : Google Scholar : PubMed/NCBI

100 

Lyu L, Li Q and Wang C: EBV latency programs: Molecular and epigenetic regulation and its role in disease pathogenesis. J Med Virol. 97:e705012025. View Article : Google Scholar : PubMed/NCBI

101 

Murray-Nerger LA, Lozano C, Burton EM, Liao Y, Ungerleider NA, Guo R and Gewurz BE: The nucleic acid binding protein SFPQ represses EBV lytic reactivation by promoting histone H1 expression. Nat Commun. 15:41562024. View Article : Google Scholar : PubMed/NCBI

102 

Yuan L, Li S, Chen Q, Xia T, Luo D, Li L, Liu S, Guo S, Liu L, Du C, et al: EBV infection-induced GPX4 promotes chemoresistance and tumor progression in nasopharyngeal carcinoma. Cell Death Differ. 29:1513–1527. 2022. View Article : Google Scholar : PubMed/NCBI

103 

He S, Luo C, Shi F, Zhou J and Shang L: The emerging role of ferroptosis in EBV-associated cancer: Implications for cancer therapy. Biology (Basel). 13:5432024.PubMed/NCBI

104 

Ge A, Xiang W, Li Y, Zhao D, Chen J, Daga P, Dai CC, Yang K, Yan Y, Hao M, et al: Broadening horizons: The multifaceted role of ferroptosis in breast cancer. Front Immunol. 15:14557412024. View Article : Google Scholar : PubMed/NCBI

105 

Li X, Zhang R, Zhao X and Zhuang X: Advances in the role of FOXM1 and ferroptosis in the diagnosis, treatment, and prognosis of hepatocellular carcinoma. Curr Protein Pept Sci. Oct 28–2025.(Epub ahead of print). View Article : Google Scholar

106 

Wu J, Zhu S, Wang P, Wang J, Huang J, Wang T, Guo L, Liang D, Meng Q and Pan H: Regulators of epigenetic change in ferroptosis-associated cancer (Review). Oncol Rep. 48:2152022. View Article : Google Scholar : PubMed/NCBI

107 

Li J, Zhang XS, Xie D, Deng HX, Gao YF, Chen QY, Huang WL, Masucci MG and Zeng YX: Expression of immune-related molecules in primary EBV-positive Chinese nasopharyngeal carcinoma: Associated with latent membrane protein 1 (LMP1) expression. Cancer Biol Ther. 6:1997–2004. 2007. View Article : Google Scholar : PubMed/NCBI

108 

Abstracts from the 22nd International Symposium on Signal Transduction at the Blood-Brain Barriers. Würzburg, Germany. September 11–13, 2019. Fluids Barriers CNS. 16 (Suppl 2):292019.PubMed/NCBI

109 

Verhoeven RJA, Tong S, Mok BWY, Liu J, He S, Zong J, Chen Y, Tsao SW, Lung ML and Chen H: Epstein-Barr virus BART long non-coding RNAs function as epigenetic modulators in nasopharyngeal carcinoma. Front Oncol. 9:11202019. View Article : Google Scholar : PubMed/NCBI

110 

Xu H, Koganti S, Li C, McIntosh MT and Bhaduri-McIntosh S: STAT3, MYC, and EBNA1 cooperate through a ZC3H18 transcriptional network to regulate survival and proliferation of EBV-positive lymphomas. PLoS Pathog. 21:e10131662025. View Article : Google Scholar : PubMed/NCBI

111 

Ji HZ, Chen L, Ren M, Li S, Liu TY, Chen HJ, Yu HH and Sun Y: CXCL8 promotes endothelial-to-mesenchymal transition of endothelial cells and protects cells from erastin-induced ferroptosis via CXCR2-mediated activation of the NF-κB signaling pathway. Pharmaceuticals (Basel). 16:12102023. View Article : Google Scholar : PubMed/NCBI

112 

Verhoeven RJA, Tong S, Zong J, Chen Y, Tsao SW, Pan J and Chen H: NF-κB signaling regulates epstein-barr virus BamHI-Q-driven EBNA1 expression. Cancers (Basel). 10:1192018. View Article : Google Scholar : PubMed/NCBI

113 

Yuan L, Zhang L, Yao N, Wu L, Liu J, Liu F, Zhang H, Hu X, Xiong Y and Xia C: Upregulation of UGT1A1 expression by ursolic acid and oleanolic acid via the inhibition of the PKC/NF-κB signaling pathway. Phytomedicine. 92:1537262021. View Article : Google Scholar : PubMed/NCBI

114 

Iglesias-Matesanz P, Lacalle-Gonzalez C, Lopez-Blazquez C, Ochieng' Otieno M, Garcia-Foncillas J and Martinez-Useros J: Glutathione peroxidases: An emerging and promising therapeutic target for pancreatic cancer treatment. Antioxidants (Basel). 13:14052024. View Article : Google Scholar : PubMed/NCBI

115 

Morgos DT, Stefani C, Miricescu D, Greabu M, Stanciu S, Nica S, Stanescu-Spinu II, Balan DG, Balcangiu-Stroescu AE, Coculescu EC, et al: Targeting PI3K/AKT/mTOR and MAPK signaling pathways in gastric cancer. Int J Mol Sci. 25:18482024. View Article : Google Scholar : PubMed/NCBI

116 

Tóthová Z, Šemeláková M, Solárová Z, Tomc J, Debeljak N and Solár P: The role of PI3K/AKT and MAPK signaling pathways in erythropoietin signalization. Int J Mol Sci. 22:76822021. View Article : Google Scholar : PubMed/NCBI

117 

Chen K, Wang J, Guo L, Wang J, Yang L, Hu T, Zhao Y, Wang X and Zhu Y: Lactobacillus johnsonii L531 ameliorates salmonella enterica serovar typhimurium diarrhea by modulating iron homeostasis and oxidative stress via the IRP2 pathway. Nutrients. 15:11272023. View Article : Google Scholar : PubMed/NCBI

118 

Jia F, Li H, Jiao Q, Li C, Fu L, Cui C, Jiang H and Zhang L: Deubiquitylase OTUD3 prevents Parkinson's disease through stabilizing iron regulatory protein 2. Cell Death Dis. 13:4182022. View Article : Google Scholar : PubMed/NCBI

119 

Jiao Q, Du X, Wei J, Li Y and Jiang H: Oxidative stress regulated iron regulatory protein IRP2 through FBXL5-mediated ubiquitination-proteasome way in SH-SY5Y cells. Front Neurosci. 13:202019. View Article : Google Scholar : PubMed/NCBI

120 

Terzi EM, Sviderskiy VO, Alvarez SW, Whiten GC and Possemato R: Iron-sulfur cluster deficiency can be sensed by IRP2 and regulates iron homeostasis and sensitivity to ferroptosis independent of IRP1 and FBXL5. Sci Adv. 7:eabg43022021. View Article : Google Scholar : PubMed/NCBI

121 

Dasgupta S and Gan B: Ferroptosis vulnerability in MYCN-driven neuroblastomas. Clin Transl Med. 12:e9632022. View Article : Google Scholar : PubMed/NCBI

122 

Hill RA and Liu YY: N6-methyladenosine-RNA methylation promotes expression of solute carrier family 7 member 11, an uptake transporter of cystine for lipid reactive oxygen species scavenger glutathione synthesis, leading to hepatoblastoma ferroptosis resistance. Clin Transl Med. 12:e8892022. View Article : Google Scholar : PubMed/NCBI

123 

Li X, Qi H, Zhang X, Liang H and Zeng N: Jing-Fang n-butanol extract and its isolated JFNE-C inhibit ferroptosis and inflammation in LPS induced RAW264.7 macrophages via STAT3/p53/SLC7A11 signaling pathway. J Ethnopharmacol. 316:1166892023. View Article : Google Scholar : PubMed/NCBI

124 

Li X, Zou Y, Fu YY, Xing J, Wang KY, Wan PZ and Zhai XY: A-Lipoic acid alleviates folic acid-induced renal damage through inhibition of ferroptosis. Front Physiol. 12:6805442021. View Article : Google Scholar : PubMed/NCBI

125 

Zhao X, Shi D, Sun L, Gong Z, Liu W, Zhang Y and Luo B: Epstein-Barr virus modulates iron metabolism and ferritin expression to promote tumorigenesis in gastric cancer. J Mol Histol. 56:2312025. View Article : Google Scholar : PubMed/NCBI

126 

Altamura S, Colucci S, Schmidt J, Muedder K, Neves J, Hentze M and Muckenthaler M: Hepatocyte iron content controls BMP6-dependent hepcidin regulation. Blood. 132 (Suppl 1):S3626. 2018. View Article : Google Scholar

127 

Cabrera C, Frisk C, Löfström U, Lyngå P, Linde C, Hage C, Persson H, Eriksson MJ, Wallén H, Persson B and Ekström M: Relationship between iron deficiency and expression of genes involved in iron metabolism in human myocardium and skeletal muscle. Int J Cardiol. 379:82–88. 2023. View Article : Google Scholar : PubMed/NCBI

128 

Huang H, Mei L, Wang L, Bai Y, Gao K, Song J, Jiang M, Chen Y, Zhang S, Pang B, et al: Ferroptosis contributes to lead-induced cochlear spiral ganglion neurons injury. Toxicology. 509:1539382024. View Article : Google Scholar : PubMed/NCBI

129 

Ameziane El Hassani R, Buffet C, Leboulleux S and Dupuy C: Oxidative stress in thyroid carcinomas: Biological and clinical significance. Endocr Relat Cancer. 26:R131–R143. 2019. View Article : Google Scholar : PubMed/NCBI

130 

Backert S, Linz B and Tegtmeyer N: Helicobacter pylori-induced host cell DNA damage and genetics of gastric cancer development. Curr Top Microbiol Immunol. 444:185–206. 2023.PubMed/NCBI

131 

da Silva MS, Segatto M, Pavani RS, Gutierrez-Rodrigues F, Bispo VD, de Medeiros MH, Calado RT, Elias MC and Cano MI: Consequences of acute oxidative stress in Leishmania amazonensis: From telomere shortening to the selection of the fittest parasites. Biochim Biophys Acta Mol Cell Res. 1864:138–150. 2017. View Article : Google Scholar : PubMed/NCBI

132 

Lee J, Lim JW and Kim H: Astaxanthin inhibits oxidative stress-induced Ku protein degradation and apoptosis in gastric epithelial cells. Nutrients. 14:39392022. View Article : Google Scholar : PubMed/NCBI

133 

Sekhar KR, Hanna DN, Cyr S, Baechle JJ, Kuravi S, Balusu R, Rathmell K and Baregamian N: Glutathione peroxidase 4 inhibition induces ferroptosis and mTOR pathway suppression in thyroid cancer. Sci Rep. 12:193962022. View Article : Google Scholar : PubMed/NCBI

134 

Dong Q, Shen X, Fang G, Shi J, Zhu X, Du J, Zhang H and Ge C: Theory-screened Prussian blue analogues-based nanozymes for promoting diabetic wound healing via ferroptosis inhibition. Mater Today Bio. 32:1018392025. View Article : Google Scholar : PubMed/NCBI

135 

Yang R, Zhou Y, Zhang T, Wang S, Wang J, Cheng Y, Li H, Jiang W, Yang Z and Zhang X: The transcription factor HBP1 promotes ferroptosis in tumor cells by regulating the UHRF1-CDO1 axis. PLoS Biol. 21:e30018622023. View Article : Google Scholar : PubMed/NCBI

136 

Zhu JF, Liu Y, Li WT, Li MH, Zhen CH, Sun PW, Chen JX, Wu WH and Zeng W: Ibrutinib facilitates the sensitivity of colorectal cancer cells to ferroptosis through BTK/NRF2 pathway. Cell Death Dis. 14:1512023. View Article : Google Scholar : PubMed/NCBI

137 

Hitt MM, Allday MJ, Hara T, Karran L, Jones MD, Busson P, Tursz T, Ernberg I and Griffin BE: EBV gene expression in an NPC-related tumour. EMBO J. 8:2639–2651. 1989. View Article : Google Scholar : PubMed/NCBI

138 

Chiu SH, Wu MC, Wu CC, Chen YC, Lin SF, Hsu JTA, ang CS, Tsai CH, Takada K, Chen MR and Chen JY: Epstein-Barr virus BALF3 has nuclease activity and mediates mature virion production during the lytic cycle. J Virol. 88:4962–4975. 2014. View Article : Google Scholar : PubMed/NCBI

139 

Ragoczy T, Heston L and Miller G: The Epstein-Barr virus Rta protein activates lytic cycle genes and can disrupt latency in B lymphocytes. J Virol. 72:7978–7984. 1998. View Article : Google Scholar : PubMed/NCBI

140 

Germini D, Sall FB, Shmakova A, Wiels J, Dokudovskaya S, Drouet E and Vassetzky Y: Oncogenic properties of the EBV ZEBRA protein. Cancers (Basel). 12:14792020. View Article : Google Scholar : PubMed/NCBI

141 

Meng C, Dai X, Sun L, Huang D, Xu X and Cheng Y: Meeting abstracts from the 10th international conference on cGMP: Generators, effectors and therapeutic implications. J Transl Med. Jan 31–2023.(Epub ahead of print).

142 

Meng C, Dai X, Sun L, Huang D, Xu X, Cheng Y and Zhang W: ZDHHC21-driven S-palmitoylation of Themis regulates the function of T cells and maintains homeostatic balance. Cell Commun Signal. 23:4012025. View Article : Google Scholar : PubMed/NCBI

143 

Png YT, Yang AZY, Lee MY, Chua MJM and Lim CM: The role of NK cells in EBV infection and EBV-associated NPC. Viruses. 13:3002021. View Article : Google Scholar : PubMed/NCBI

144 

Hsu WL, Tao J, Fu S, Yu KJ, Simon J, Chen TC, Chen CJ, Goldstein AM, Yu K, Hildesheim A, et al: Kinetics of EBV antibody-based NPC risk scores in Taiwan NPC multiplex families. Int J Cancer. 155:1400–1408. 2024. View Article : Google Scholar : PubMed/NCBI

145 

Wang J and Cao X: Dietary long-chain fatty acid metabolism boosts antitumor immune response. Cancer Commun (Lond). 44:580–583. 2024. View Article : Google Scholar : PubMed/NCBI

146 

Peng WX and Mo YY: Connecting N6-methyladenosine modification to ferroptosis resistance in hepatoblastoma. Clin Transl Med. 12:e8202022. View Article : Google Scholar : PubMed/NCBI

147 

Chiu SH, Wu CC, Fang CY, Yu SL, Hsu HY, Chow YH and Chen JY: Epstein-Barr virus BALF3 mediates genomic instability and progressive malignancy in nasopharyngeal carcinoma. Oncotarget. 5:8583–8601. 2014. View Article : Google Scholar : PubMed/NCBI

148 

Benedetti F, Curreli S, Gallo RC and Zella D: Tampering of viruses and bacteria with host DNA repair: Implications for cellular transformation. Cancers (Basel). 13:2412021. View Article : Google Scholar : PubMed/NCBI

149 

Dylawerska A, Barczak W, Wegner A, Golusinski W and Suchorska WM: Association of DNA repair genes polymorphisms and mutations with increased risk of head and neck cancer: A review. Med Oncol. 34:1972017. View Article : Google Scholar : PubMed/NCBI

150 

Mekonnen N, Yang H and Shin YK: Homologous recombination deficiency in ovarian, breast, colorectal, pancreatic, non-small cell lung and prostate cancers, and the mechanisms of resistance to PARP inhibitors. Front Oncol. 12:8806432022. View Article : Google Scholar : PubMed/NCBI

151 

No authors listed. The international headache congress-IHS and EHF joint congress 2021: Late breaking abstracts: Virtual. 8–12 September 2021. J Headache Pain. 22 (Suppl 2):S1302021. View Article : Google Scholar

152 

Abstracts from the 17th European headache congress (EHC). J Headache Pain. 25 (Suppl 1):1232024. View Article : Google Scholar

153 

Li F, Xu T, Chen P, Sun R, Li C, Zhao X, Ou J, Li J, Liu T, Zeng M, et al: Platelet-derived extracellular vesicles inhibit ferroptosis and promote distant metastasis of nasopharyngeal carcinoma by upregulating ITGB3. Int J Biol Sci. 18:5858–5872. 2022. View Article : Google Scholar : PubMed/NCBI

154 

Jin N, Qian YY, Jiao XF, Wang Z, Li X, Pan W, Jiang JK, Huang P, Wang SY, Jin P, et al: Niraparib restricts intraperitoneal metastases of ovarian cancer by eliciting CD36-dependent ferroptosis. Redox Biol. 80:1035282025. View Article : Google Scholar : PubMed/NCBI

155 

Zhang B, Hou Q, Zhang X, Ma Y, Yuan J, Li S, Zhao X, Sun L, Wang H and Zheng H: Anesthetic propofol inhibits ferroptosis and aggravates distant cancer metastasis via Nrf2 upregulation. Free Radic Biol Med. 195:298–308. 2023. View Article : Google Scholar : PubMed/NCBI

156 

Zhang R, Shen Y, Zhou X, Li J, Zhao H, Zhang Z, Zhao J, Jin H, Guo S, Ding H, et al: Hypoxia-tropic delivery of nanozymes targeting transferrin receptor 1 for nasopharyngeal carcinoma radiotherapy sensitization. Nat Commun. 16:8902025. View Article : Google Scholar : PubMed/NCBI

157 

Wang HH, Fan SQ, Zhan YT, Peng SP and Wang WY: Suppression of the SLC7A11/glutathione axis causes ferroptosis and apoptosis and alters the mitogen-activated protein kinase pathway in nasopharyngeal carcinoma. Int J Biol Macromol. 254:1279762024. View Article : Google Scholar : PubMed/NCBI

158 

Nhung DT, Yousif OEA and Kwon B: Sorafenib induces ferroptosis in human renal cell carcinoma cells through CCAT/enhancer-binding protein homologous protein. Biochem Biophys Rep. 43:1021432025.PubMed/NCBI

159 

Liu F, Tang L, Liu H, Chen Y, Xiao T, Gu W, Yang H, Wang H and Chen P: Cancer-associated fibroblasts secrete FGF5 to inhibit ferroptosis to decrease cisplatin sensitivity in nasopharyngeal carcinoma through binding to FGFR2. Cell Death Dis. 15:2792024. View Article : Google Scholar : PubMed/NCBI

160 

Chen Y, Feng Y, Lin Y, Zhou X, Wang L, Zhou Y, Lin K and Cai L: GSTM3 enhances radiosensitivity of nasopharyngeal carcinoma by promoting radiation-induced ferroptosis through USP14/FASN axis and GPX4. Br J Cancer. 130:755–768. 2024. View Article : Google Scholar : PubMed/NCBI

161 

Pu X, Wu H, Liu X and Yang F: PRMT4 reduced erastin-induced ferroptosis in nasopharyngeal carcinoma cisplatin-resistant cells by Nrf2/GPX4 pathway. J Environ Pathol Toxicol Oncol. 44:57–71. 2025. View Article : Google Scholar : PubMed/NCBI

162 

Du P, Luo K, Li G, Zhu J, Xiao Q, Li Y and Zhang X: PRMT4 promotes hepatocellular carcinoma progression by activating AKT/mTOR signaling and indicates poor prognosis. Int J Med Sci. 18:3588–3598. 2021. View Article : Google Scholar : PubMed/NCBI

163 

Berman AY, Manna S, Schwartz NS, Katz YE, Sun Y, Behrmann CA, Yu JJ, Plas DR, Alayev A and Holz MK: ERRα regulates the growth of triple-negative breast cancer cells via S6K1-dependent mechanism. Signal Transduct Target Ther. 2:e170352017. View Article : Google Scholar : PubMed/NCBI

164 

Xiao H, Zhang F, Zou Y, Li J, Liu Y and Huang W: The function and mechanism of long non-coding RNA-ATB in cancers. Front Physiol. 9:3212018. View Article : Google Scholar : PubMed/NCBI

165 

Cui X, Gong Y, Ge J, Feng X, Xiong X, Shi Z, Zheng Q, Li D and Bi S: α-Solanine induces ferroptosis in nasopharyngeal carcinoma via targeting HSP90α/p53 axis. J Funct Foods. 104:1055172023. View Article : Google Scholar

166 

Yap TA, Bessudo A, Hamilton E, Sachdev J, Patel MR, Rodon J, Evilevitch L, Duncan M, Guo W, Kumar S, et al: IOLite: Phase 1b trial of doublet/triplet combinations of dostarlimab with niraparib, carboplatin-paclitaxel, with or without bevacizumab in patients with advanced cancer. J Immunother Cancer. 10:e0039242022. View Article : Google Scholar : PubMed/NCBI

167 

Wang Y, Xu H, Sa Q, Zhou Y, Cheng H, Gao R, Xu B and Wang J: Efficacy and safety of vascular-targeting agents in advanced soft tissue sarcoma: A systematic review and network meta-analysis. Ther Adv Med Oncol. 17:175883592513789342025. View Article : Google Scholar : PubMed/NCBI

168 

Ochsenreither S, Fiedler WM, Conte GD, Macchini M, Matos I, Habel B, Ahrens-Fath I, Raspagliesi F, Lorusso D, Keilholz U, et al: Safety and preliminary activity results of the GATTO study, a phase Ib study combining the anti-TA-MUC1 antibody gatipotuzumab with the anti-EGFR tomuzotuximab in patients with refractory solid tumors. ESMO Open. 7:1004472022. View Article : Google Scholar : PubMed/NCBI

169 

Zargarani N, Kavousi M and Aliasgari E: A potential new strategy for BC treatment: NPs containing solanine and evaluation of its anticancer and antimetastatic properties. BMC Cancer. 25:8602025. View Article : Google Scholar : PubMed/NCBI

170 

Chowański S, Winkiel M, Szymczak-Cendlak M, Marciniak P, Mańczak D, Walkowiak-Nowicka K, Spochacz M, Bufo SA, Scrano L and Adamski Z: Solanaceae glycoalkaloids: α-solanine and α-chaconine modify the cardioinhibitory activity of verapamil. Pharm Biol. 60:1317–1330. 2022. View Article : Google Scholar : PubMed/NCBI

171 

Li W, Liu Y, Wei M, Yang Z, Tang H and Huang W: Chondrocyte-targeted α-Solanine through HIF-1α regulating glycolysis to reduce the ferroptosis of chondrocyte in osteoarthritis. Int Immunopharmacol. 159:1148412025. View Article : Google Scholar : PubMed/NCBI

172 

Wei Z, Wang H, Zhong R, Chen L, Vigors S and Zhang H: Biodegradation of α-solanine and α-chaconine: Insights into microbial detoxification and enzymatic deglycosylation pathways. Food Chem X. 31:1029682025. View Article : Google Scholar : PubMed/NCBI

173 

Zhou J, Wu J, Fu F, Yao S, Zheng W, Du W, Luo H, Jin H, Tong P, Wu C and Ruan H: α-Solanine attenuates chondrocyte pyroptosis to improve osteoarthritis via suppressing NF-κB pathway. J Cell Mol Med. 28:e181322024. View Article : Google Scholar : PubMed/NCBI

174 

Seven D, Dalan AB and Bayrak ÖF: Targeting GSTM3 for therapeutic potential in advanced prostate cancer. BMC Cancer. 25:14932025. View Article : Google Scholar : PubMed/NCBI

175 

Llavanera M, Ribas-Maynou J, Delgado-Bermúdez A, Recuero S, Salas-Huetos A, Benet J and Yeste M: P-049 sperm GSTM3: A potential molecular biomarker for sperm quality and male (in)fertility. Hum Reprod. 37 (Suppl 1):deac104.111. 2022. View Article : Google Scholar

176 

Wang B, Gu Q and Li J: DOC-2/DAB2 interactive protein regulates proliferation and mobility of nasopharyngeal carcinoma cells by targeting PI3K/Akt pathway. Oncol Rep. 38:317–324. 2017. View Article : Google Scholar : PubMed/NCBI

177 

Niu Z, Liu H, Zhou M, Wang H, Liu Y, Li X, Xiong W, Ma J, Li X and Li G: Knockdown of c-Myc inhibits cell proliferation by negatively regulating the Cdk/Rb/E2F pathway in nasopharyngeal carcinoma cells. Acta Biochim Biophys Sin (Shanghai). 47:183–191. 2015. View Article : Google Scholar : PubMed/NCBI

178 

Soulage CO, Pelletier CC, Florens N, Lemoine S, Dubourg L, Juillard L and Guebre-Egziabher F: Two toxic lipid aldehydes, 4-hydroxy-2-hexenal (4-HHE) and 4-hydroxy-2-nonenal (4-HNE), accumulate in patients with chronic kidney disease. Toxins (Basel). 12:5672020. View Article : Google Scholar : PubMed/NCBI

179 

Li Y, Zhao T, Li J, Xia M, Li Y, Wang X, Liu C, Zheng T, Chen R, Kan D, et al: Oxidative stress and 4-hydroxy-2-nonenal (4-HNE): Implications in the pathogenesis and treatment of aging-related diseases. J Immunol Res. 2022:22339062022.PubMed/NCBI

180 

Globisch M, Kaden D and Henle T: 4-Hydroxy-2-nonenal (4-HNE) and Its lipation product 2-pentylpyrrole lysine (2-PPL) in peanuts. J Agric Food Chem. 63:5273–81. 2015. View Article : Google Scholar : PubMed/NCBI

181 

Llavanera M, Delgado-Bermúdez A, Fernandez-Fuertes B, Recuero S, Mateo Y, Bonet S, Barranco I and Yeste M: GSTM3, but not IZUMO1, is a cryotolerance marker of boar sperm. J Anim Sci Biotechnol. 10:612019. View Article : Google Scholar : PubMed/NCBI

182 

Li B, Zhou J, Zhang G, Wang Y, Kang L, Wu J, Chen J and Guan H: Relationship between the altered expression and epigenetics of GSTM3 and age-related cataract. Invest Ophthalmol Vis Sci. 57:4721–4732. 2016. View Article : Google Scholar : PubMed/NCBI

183 

Mandic-Maravic V, Mitkovic-Voncina M, Pljesa-Ercegovac M, Savic-Radojevic A, Djordjevic M, Ercegovac M, Pekmezovic T, Simic T and Pejovic-Milovancevic M: Glutathione S-transferase polymorphisms and clinical characteristics in autism spectrum disorders. Front Psychiatry. 12:6723892021. View Article : Google Scholar : PubMed/NCBI

184 

Li X, Gou J, Li H and Yang X: Bioinformatic analysis of the expression and prognostic value of chromobox family proteins in human breast cancer. Sci Rep. 10:177392020. View Article : Google Scholar : PubMed/NCBI

185 

Li G, Cai Y, Wang C, Huang M and Chen J: LncRNA GAS5 regulates the proliferation, migration, invasion and apoptosis of brain glioma cells through targeting GSTM3 expression. The effect of LncRNA GAS5 on glioma cells. J Neurooncol. 143:525–536. 2019. View Article : Google Scholar : PubMed/NCBI

186 

Chen T, Jinlin D, Wang F, Yuan Z, Xue J, Lu T, Huang W, Liu Y and Zhang Y: GSTM3 deficiency impedes DNA mismatch repair to promote gastric tumorigenesis via CAND1/NRF2-KEAP1 signaling. Cancer Lett. 538:2156922022. View Article : Google Scholar : PubMed/NCBI

187 

Wu Y, Jia Q, Tang Q, Deng H, He Y and Tang F: Berberine-mediated ferroptosis through system Xc−/GSH/GPX4 axis inhibits metastasis of nasopharyngeal carcinoma. J Cancer. 15:685–698. 2024. View Article : Google Scholar : PubMed/NCBI

188 

Hou J, Zi L, Shi M, Wang Y, Gao F and Chen W: The contribution of SLC7A11-mediated ferroptosis to cardiac injury in iron overload cardiomyopathy: An in vitro study. Eur Heart J. 45 (Suppl 1):ehae666.3648. 2024. View Article : Google Scholar

189 

Wang F, Sun Z, Zhang Q, Yang H, Yang G, Yang Q, Zhu Y, Wu W, Xu W and Wu X: Curdione induces ferroptosis mediated by m6A methylation via METTL14 and YTHDF2 in colorectal cancer. Chin Med. 18:1222023. View Article : Google Scholar : PubMed/NCBI

190 

Zhao YY, Yang YQ, Sheng HH, Tang Q, Han L, Wang SM and Wu WY: GPX4 plays a crucial role in Fuzheng Kang'ai decoction-induced non-small cell lung cancer cell ferroptosis. Front Pharmacol. 13:8516802022. View Article : Google Scholar : PubMed/NCBI

191 

Gao X, Guo N, Xu H, Pan T, lei H, Yan A, Mi Y and Xu L: Ibuprofen induces ferroptosis of glioblastoma cells via downregulation of nuclear factor erythroid 2-related factor 2 signaling pathway. Anticancer Drugs. 31:27–34. 2020. View Article : Google Scholar : PubMed/NCBI

192 

Zheng J, Fang Y, Zhang M, Gao Q, Li J, Yuan H, Jin W, Lin Z and Lin W: Mechanisms of ferroptosis in hypoxic-ischemic brain damage in neonatal rats. Exp Neurol. 372:1146412024. View Article : Google Scholar : PubMed/NCBI

193 

Yujiao C, Meng Z, Shanshan L, Wei W, Yipeng W and Chenghong Y: Exposure to bisphenol A induces abnormal fetal heart development by promoting ferroptosis. Ecotoxicol Environ Saf. 255:1147532023. View Article : Google Scholar : PubMed/NCBI

194 

Wang S, Xu Z, Cai B and Chen Q: Berberine as a potential multi-target agent for metabolic diseases: A review of investigations for berberine. Endocr Metab Immune Disord Drug Targets. 21:971–979. 2021. View Article : Google Scholar : PubMed/NCBI

195 

Chen J, Peng W, Yang W, You Z and Zou Y: Berberine inhibits high glucose-induced ferroptosis in retinal vascular endothelial cells: Mechanism and implications. Exp Eye Res. 259:1105312025. View Article : Google Scholar : PubMed/NCBI

196 

Bao L, Jin Y, Han J, Wang W, Qian L and Wu W: Berberine regulates GPX4 to inhibit ferroptosis of islet β cells. Planta Med. 89:254–261. 2023. View Article : Google Scholar : PubMed/NCBI

197 

Shah D, Challagundla N, Dave V, Patidar A, Saha B, Nivsarkar M, Trivedi VB and Agrawal-Rajput R: Berberine mediates tumor cell death by skewing tumor-associated immunosuppressive macrophages to inflammatory macrophages. Phytomedicine. 99:1539042022. View Article : Google Scholar : PubMed/NCBI

198 

Sun Q, Tu K, Xu Q, Yan L, Yang S, Wang J, Lv L, Liu H and Cai L: Berberine suppresses colorectal cancer progression by inducing ferroptosis-mediated energy metabolism disorders. J Adv Res. Oct 24–2025.(Epub ahead of print). View Article : Google Scholar

199 

Xie Z, Zhou Y, Lin M and Huang C: Binding of berberine to PEBP1 synergizes with sorafenib to induce the ferroptosis of hepatic stellate cells. Amino Acids. 55:1867–1878. 2023. View Article : Google Scholar : PubMed/NCBI

200 

Zhang Y, Liu X, Yu M, Xu M, Xiao Y, Ma W, Huang L, Li X and Ye X: Berberine inhibits proliferation and induces G0/G1 phase arrest in colorectal cancer cells by downregulating IGF2BP3. Life Sci. 260:1184132020. View Article : Google Scholar : PubMed/NCBI

201 

Eke I, Makinde AY, Aryankalayil MJ, Sandfort V, Palayoor ST, Rath BH, Liotta L, Pierobon M, Petricoin EF, Brown MF, et al: Exploiting radiation-induced signaling to increase the susceptibility of resistant cancer cells to targeted drugs: AKT and mTOR inhibitors as an example. Mol Cancer Ther. 17:355–367. 2018. View Article : Google Scholar : PubMed/NCBI

202 

Bagnulo M and García-Martínez A: When less is more: BBR versus LEDBAT++. Comput Netw. 219:1094602022. View Article : Google Scholar

203 

Atxutegi E, Liberal F, Haile HK, Grinnemo KJ, Brunstrom A and Arvidsson Å: On the use of TCP BBR in cellular networks. IEEE Commun Mag. 56:172–179. 2018. View Article : Google Scholar

204 

Abdelmawgood IA, Kotb MA, Hassan HS, Mahana NA, Rochdi AM, Sayed NH, Elsafoury RH, Saber AM, Youssef MN, Waheeb NG, et al: Gentisic acid attenuates ovalbumin-induced airway inflammation, oxidative stress, and ferroptosis through the modulation of Nrf2/HO-1 and NF-κB signaling pathways. Int Immunopharmacol. 146:1137642025. View Article : Google Scholar : PubMed/NCBI

205 

Zhong X, Zhang Z, Shen H, Xiong Y, Shah YM, Liu Y, Liu Y, Fan XG and Rui L: Hepatic NF-κB-inducing kinase and inhibitor of NF-κB kinase subunit α promote liver oxidative stress, ferroptosis, and liver injury. Hepatol Commun. 5:1704–1720. 2021. View Article : Google Scholar : PubMed/NCBI

206 

Zhou JC, Wu B, Zhang JJ and Zhang W: Lupeol triggers oxidative stress, ferroptosis, apoptosis and restrains inflammation in nasopharyngeal carcinoma via AMPK/NF-κB pathway. Immunopharmacol Immunotoxicol. 44:621–631. 2022. View Article : Google Scholar : PubMed/NCBI

207 

Wang Z, Di Y, Ye L, Fang W, Wen X, Zhang X, Qin J, Wang Y, Hu K, Zhu Z, et al: NANS suppresses NF-κB signaling to promote ferroptosis by perturbing iron homeostasis. Cell Rep. 44:1157012025. View Article : Google Scholar : PubMed/NCBI

208 

Wu S, Zhou Y, Liang J, Ying P, Situ Q, Tan X and Zhu J: Upregulation of NF-κB by USP24 aggravates ferroptosis in diabetic cardiomyopathy. Free Radic Biol Med. 210:352–366. 2024. View Article : Google Scholar : PubMed/NCBI

209 

Luo X, Gong Y, Jiang Q, Wang Q, Li S and Liu L: Isoquercitrin promotes ferroptosis and oxidative stress in nasopharyngeal carcinoma via the AMPK/NF-κB pathway. J Biochem Mol Toxicol. 38:e235422024. View Article : Google Scholar : PubMed/NCBI

210 

Song X, Zhu H, Chen Z, Wang Y, Zhang J, Wang Y, Rong P and Wang J: Transcutaneous auricular vagus nerve stimulation alleviates inflammation-induced depression by modulating peripheral-central inflammatory cytokines and the NF-κB pathway in rats. Front Immunol. 16:15360562025. View Article : Google Scholar : PubMed/NCBI

211 

Gong X, Yang Y, Huang L, Zhang Q, Wan RZ, Zhang P and Zhang X: Antioxidation, anti-inflammation and anti-apoptosis by paeonol in LPS/d-GalN-induced acute liver failure in mice. Int Immunopharmacol. 46:124–132. 2017. View Article : Google Scholar : PubMed/NCBI

212 

Zeng MY and Tong QY: Anti-inflammation effects of sinomenine on macrophages through suppressing activated TLR4/NF-κB signaling pathway. Curr Med Sci. 40:130–137. 2020. View Article : Google Scholar : PubMed/NCBI

213 

Zhao S, Zuo W, Chen H, Bao T, Liu X, Sun T and Wang S: Effects of pilose antler peptide on bleomycin-induced pulmonary fibrosis in mice. Biomed Pharmacother. 109:2078–2083. 2018. View Article : Google Scholar : PubMed/NCBI

214 

Zhang J, Qi S, Du Y, Dai H and Lu N: Effect of quercetin on inhibiting gefitinib-activated non-small cell lung cancer-induced cell pyroptosis in cardiomyocytes via modulating mitochondrial autophagy mediated by the SHP2/ROS/AMPK/XBP-1/DJ-1 signaling pathway. Oncol Rep. 53:572025. View Article : Google Scholar : PubMed/NCBI

215 

Wang N, Chen HQ, Zeng Y, Shi Y, Zhang Z, Li JY, Zhou SM, Li YW, Deng SW, Han X, et al: Benzo(a)pyrene promotes the malignant progression of malignant-transformed BEAS-2B cells by regulating YTH N6-methyladenosine RNA binding protein 1 to inhibit ferroptosis. Toxicology. 507:1538862024. View Article : Google Scholar : PubMed/NCBI

216 

Shao C, Luo T, Wang S, Li Z, Yu X, Wu Y, Jiang S, Zhou B, Song Q, Song S, et al: Selenium nanoparticles alleviates cadmium induced hepatotoxicity by inhibiting ferroptosis and oxidative stress in vivo and in vitro. Chemosphere. 364:1430042024. View Article : Google Scholar : PubMed/NCBI

217 

Meng J, Hu C, Qian Z, Yue J, Zhang S, Jiang W, Su R, Jiang G and Huang G: Isoquercitrin inhibits ferroptosis and ameliorates insulin resistance: Evidence from network pharmacology and in vitro studies. Biochem Biophys Res Commun. 781:1525002025. View Article : Google Scholar : PubMed/NCBI

218 

Lee J, Jang CH, Kim Y, Oh J and Kim JS: Quercetin-Induced glutathione depletion sensitizes colorectal cancer cells to oxaliplatin. Foods. 12:17332023. View Article : Google Scholar : PubMed/NCBI

219 

Kottakis G, Kambouri K, Giatromanolaki A, Valsami G, Kostomitsopoulos N, Tsaroucha A and Pitiakoudis M: Effects of the antioxidant quercetin in an experimental model of ulcerative colitis in mice. Medicina (Kaunas). 59:872022. View Article : Google Scholar : PubMed/NCBI

220 

Li Y, Xu Z, Zhao S, Huang T, Xu J, Wang S, Sang Y, Yu W and Wang X: Oral administration of curcumin and quercetin nanoparticles can improve ulcerative colitis by regulating intestinal microorganisms. Front Nutr. 12:16966992025. View Article : Google Scholar : PubMed/NCBI

221 

Wei Q, Jiang H, Zeng J, Xu J, Zhang H, Xiao E, Lu Q and Huang G: Quercetin protected the gut barrier in ulcerative colitis by activating aryl hydrocarbon receptor. Phytomedicine. 140:1566332025. View Article : Google Scholar : PubMed/NCBI

222 

Li J, Yan Y and Chen F: Clinical trial landscape for histone deacetylation inhibitors in breast cancer: A dawn in the darkness? J Transl Med. 22:10812024. View Article : Google Scholar : PubMed/NCBI

223 

Tan LLY, Le QT, Lee NYY and Chua MLK: JUPITER-02 trial: Advancing survival for recurrent metastatic nasopharyngeal carcinoma and next steps. Cancer Commun (Lond). 42:56–59. 2022. View Article : Google Scholar : PubMed/NCBI

224 

Zhu D, Wang Z, Zhang G, Ma C, Qiu X, Wang Y, Liu M, Guo X, Chen H, Deng Q and Kang X: Periostin promotes nucleus pulposus cells apoptosis by activating the Wnt/β-catenin signaling pathway. FASEB J. 36:e223692022. View Article : Google Scholar : PubMed/NCBI

225 

Takashima K, Okano H, Ojiro R, Tang Q, Takahashi Y, Ozawa S, Zou X, Koyanagi M, Maronpot RR, Yoshida T and Shibutani M: Continuous exposure to alpha-glycosyl isoquercitrin from gestation ameliorates disrupted hippocampal neurogenesis in rats induced by gestational injection of valproic acid. Neurotox Res. 40:2278–2296. 2022. View Article : Google Scholar : PubMed/NCBI

226 

Takashima K, Okano H, Ojiro R, Tang Q, Takahashi Y, Ozawa S, Zou X, Koyanagi M, Maronpot RR, Yoshida T and Shibutani M: Continuous exposure to alpha-glycosyl isoquercitrin from mid-gestation ameliorates polyinosinic-polycytidylic acid-disrupted hippocampal neurogenesis in rats. J Chem Neuroanat. 128:1022192023. View Article : Google Scholar : PubMed/NCBI

227 

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

228 

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

229 

Chen C, Nie D, Huang Y, Yu X, Chen Z, Zhong M, Liu X, Wang X, Sui S, Liu Z, et al: Anticancer effects of disulfiram in T-cell malignancies through NPL4-mediated ubiquitin-proteasome pathway. J Leukoc Biol. 112:919–929. 2022. View Article : Google Scholar : PubMed/NCBI

230 

Serra R, Zhao T, Huq S, Gorelick NL, Casaos J, Cecia A, Mangraviti A, Bai R, Olivi A, Brem H, et al: Disulfiram and copper combination therapy targets NPL4, cancer stem cells and prolongs survival in group 3 medulloblastoma. Neurosurgery. 87 (Suppl 1):S9082020.

231 

Serra R, Zhao T, Huq S, Gorelick NL, Casaos J, Cecia A, Mangraviti A, Eberhart C, Bai R, Olivi A, et al: Disulfiram and copper combination therapy targets NPL4, cancer stem cells and extends survival in a medulloblastoma model. PLoS One. 16:e02519572021. View Article : Google Scholar : PubMed/NCBI

232 

Lian Q, Chen F, Sha Z, Zhao H, Li J, Chen T, Liu C, Wang B, Wang Z and Qiao S: Disulfiram upgrades the radiosensitivity of osteosarcoma by enhancing apoptosis and P53-induced cell cycle arrest. Radiat Res. 202:752–764. 2024. View Article : Google Scholar : PubMed/NCBI

233 

Wu X, Xue X, Wang L, Wang W, Han J, Sun X, Zhang H, Liu Y, Che X, Yang J and Wu C: Suppressing autophagy enhances disulfiram/copper-induced apoptosis in non-small cell lung cancer. Eur J Pharmacol. 827:1–12. 2018. View Article : Google Scholar : PubMed/NCBI

234 

Wang K, Michelakos T, Wang B, Shang Z, DeLeo AB, Duan Z, Hornicek FJ, Schwab JH and Wang X: Targeting cancer stem cells by disulfiram and copper sensitizes radioresistant chondrosarcoma to radiation. Cancer Lett. 505:37–48. 2021. View Article : Google Scholar : PubMed/NCBI

235 

Park YM, Kim DH, Kang MS, Koh YW, Choi EC and Kim SH: Abstract B25: Anticancer effects of disulfiram in head and neck squamous cell carcinoma via autophagic cell death. Clin Cancer Res. 26 (12 Suppl 2):B252020. View Article : Google Scholar

236 

Li Z, Cao S, Sun Y, Niu Z, Liu X, Niu J and Zhou Y: TIPE3 is a candidate prognostic biomarker promoting tumor progression via elevating RAC1 in pancreatic cancer. Mol Cancer. 21:1602022. View Article : Google Scholar : PubMed/NCBI

237 

Li Z, Cao S, Sun Y, Niu Z, Liu X, Niu J, PavelM A, Sridhar A, Maienschein-Cline M, Ong SG, et al: Late-breaking basic science abstracts from the american heart association's scientific sessions 2021. Circ Res. Dec 2–2021.(Epub ahead of print).

238 

Guo J, Ma Y, Tang T, Bian Z, Li Q, Tang L, Li Z, Li M, Wang L, Zeng A, et al: Modulation of immune-responses by DSF/Cu enhances the anti-tumor effects of DTX for metastasis breast cancer. J Cancer. 15:1523–1535. 2024. View Article : Google Scholar : PubMed/NCBI

239 

Ghaffarianhoseini A, Ghaffarianhoseini A, Berardi U, Tookey J, Li DHW and Kariminia S: Exploring the advantages and challenges of double-skin façades (DSFs). Renew Sustain Energy Rev. 60:1052–1065. 2016. View Article : Google Scholar

240 

Zhu Y, Lei C, Jiang Q, Yu Q and Qiu L: DSF/Cu induces antitumor effect against diffuse large B-cell lymphoma through suppressing NF-κB/BCL6 pathways. Cancer Cell Int. 22:2362022. View Article : Google Scholar : PubMed/NCBI

241 

Waldron EJ, Snyder D, Fernandez NL, Sileo E, Inoyama D, Freundlich JS, Waters CM, Cooper VS and Neiditch MB: Structural basis of DSF recognition by its receptor RpfR and its regulatory interaction with the DSF synthase RpfF. PLoS Biol. 17:e30001232019. View Article : Google Scholar : PubMed/NCBI

242 

Gennaro G, Catto Lucchino E, Goia F and Favoino F: Modelling double skin façades (DSFs) in whole-building energy simulation tools: Validation and inter-software comparison of naturally ventilated single-story DSFs. Build Environ. 231:1100022023. View Article : Google Scholar

243 

Catto Lucchino E, Gelesz A, Skeie K, Gennaro G, Reith A, Serra V and Goia F: Modelling double skin façades (DSFs) in whole-building energy simulation tools: Validation and inter-software comparison of a mechanically ventilated single-story DSF. Build Environ. 199:1079062021. View Article : Google Scholar

244 

Rempfer C, Hoernstein SNW, van Gessel N, Graf AW, Spiegelhalder RP, Bertolini A, Bohlender LL, Parsons J, Decker EL and Reski R: Differential prolyl hydroxylation by six Physcomitrella prolyl-4 hydroxylases. bioRxiv. 2024.

245 

Rempfer C, Hoernstein SNW, van Gessel N, Graf AW, Spiegelhalder RP, Bertolini A, Bohlender LL, Parsons J, Decker EL and Reski R: Differential prolyl hydroxylation by six Physcomitrella prolyl-4 hydroxylases. Comput Struct Biotechnol J. 23:2580–2594. 2024. View Article : Google Scholar : PubMed/NCBI

246 

McKenzie AJ, Hoshino D, Hong NH, Cha DJ, Franklin JL, Coffey RJ, Patton JG and Weaver AM: KRAS-MEK signaling controls Ago2 sorting into exosomes. Cell Rep. 15:978–987. 2016. View Article : Google Scholar : PubMed/NCBI

247 

Hu P, Zhao H, Zhu P, Xiao Y, Miao W, Wang Y and Jin H: Dual regulation of Arabidopsis AGO2 by arginine methylation. Nat Commun. 10:8442019. View Article : Google Scholar : PubMed/NCBI

248 

Zhou R, Qiu L, Zhou L, Geng R, Yang S and Wu J: P4HA1 activates HMGCS1 to promote nasopharyngeal carcinoma ferroptosis resistance and progression. Cell Signal. 105:1106092023. View Article : Google Scholar : PubMed/NCBI

249 

Meng Y, Sun HY, He Y, Zhou Q, Liu YH, Su H, Yin MZ, Zeng FR, Chen X and Deng GT: BET inhibitors potentiate melanoma ferroptosis and immunotherapy through AKR1C2 inhibition. Mil Med Res. 10:612023.PubMed/NCBI

250 

Li Q and Gan B: Uncovering the IL-1β-PCAF-NNT axis: A new player in ferroptosis and tumor immune evasion. Cancer Commun (Lond). 43:1048–1050. 2023. View Article : Google Scholar : PubMed/NCBI

251 

Qiu L, Zhou R, Zhou L, Yang S and Wu J: CAPRIN2 upregulation by LINC00941 promotes nasopharyngeal carcinoma ferroptosis resistance and metastatic colonization through HMGCR. Front Oncol 6. 12:9317492022. View Article : Google Scholar : PubMed/NCBI

252 

Ning Y, Zheng H, Zhan Y, Liu S, Yang Y, Zang H, Wen Q, Zhang Y and Fan S: Overexpression of P4HA1 associates with poor prognosis and promotes cell proliferation and metastasis of lung adenocarcinoma. J Cancer. 12:6685–6694. 2021. View Article : Google Scholar : PubMed/NCBI

253 

Robinson AD, Chakravarthi BVSK, Agarwal S, Chandrashekar DS, Davenport ML, Chen G, Manne U, Beer DG, Edmonds MD and Varambally S: Collagen modifying enzyme P4HA1 is overexpressed and plays a role in lung adenocarcinoma. Transl Oncol. 14:1011282021. View Article : Google Scholar : PubMed/NCBI

254 

Navarro-Serer B, Wissler MF, Glover BK, Lerner MG, Oza HH, Wang V, Knutsdottir H, Shojaeian F, Noller K, Baskaran SG, et al: P4HA1 mediates hypoxia-induced invasion in human pancreatic cancer organoids. Cancer Res Commun. 5:881–895. 2025. View Article : Google Scholar : PubMed/NCBI

255 

Hu T, Gou W, Ren Z, Liu G, Li Y, Zuo D and Hou W: Icaritin increases radiosensitivity of nasopharyngeal carcinoma cells by regulating iron death. Nan Fang Yi Ke Da Xue Xue Bao. 43:1665–1673. 2023.(In Chinese). PubMed/NCBI

256 

Zhang D, Wu X, Xue X, Li W, Zhou P, Lv Z, Zhao K and Zhu F: Ancient dormant virus remnant ERVW-1 drives ferroptosis via degradation of GPX4 and SLC3A2 in schizophrenia. Virol Sin. 39:31–43. 2024. View Article : Google Scholar : PubMed/NCBI

257 

Chen H, Li Y, Deng C, Liang X and Liu G: O-254 ACTRT2 deficiency increases spermatogonia vulnerability to ferroptosis. Hum Reprod. 38:dead093.308. 2023. View Article : Google Scholar

258 

Qin J, Chen Z, Ye M, Liang L and Ding X: High glucose promotes O-GlcNAcylation of ACSL4 to induce ferroptosis of renal tubular epithelial cell. Autoimmunity. 58:25768812025. View Article : Google Scholar : PubMed/NCBI

259 

Zhou R, Wang X, Jin Y, Chen B, Liu H, Zhao X, Jing D and Zhao B: Mechanism of lidocaine-induced ROS generation triggering DNA double-strand breaks and promoting intervertebral disc cell senescence via the MYC-DUSP1-P53 axis. Free Radic Biol Med. 240:457–471. 2025. View Article : Google Scholar : PubMed/NCBI

260 

Yue C, Qian Y, Wang C, Chen J, Wang J, Wang Z, Wan X, Cao S, Zhu J, Tao Q, et al: TRIM29 acts as a potential senescence suppressor with epigenetic activation in nasopharyngeal carcinoma. Cancer Sci. 114:3176–3189. 2023. View Article : Google Scholar : PubMed/NCBI

261 

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

262 

Ding Y, Xiu H, Zhang Y, Ke M, Lin L, Yan H, Hu P, Xiao M, He X and Zhang T: Learning and investigation of the role of angiotensin-converting enzyme in radiotherapy for nasopharyngeal carcinoma. Biomedicines. 11:15812023. View Article : Google Scholar : PubMed/NCBI

263 

Wang SY, Xu XW, Yao JJ, Peng PJ, Zhou B, Liu QD, Huang XP and Lin Z: Dose escalation of lobaplatin concurrent with IMRT for the treatment of stage III–IVb NPC: A phase I clinical trial. Transl Oncol. 11:1007–1011. 2018. View Article : Google Scholar : PubMed/NCBI

264 

Tang Q, Mei C, Huang B, Huang R, Kang L, Chen A, Lei N, Deng P, Ying S, Zhang P and Qin Y: Risk stratification of LA-NPC during chemoradiotherapy based on clinical classification and TVRR. Cancer Med. 13:e70292024. View Article : Google Scholar : PubMed/NCBI

265 

Kim KY, Le QT, Yom SS, Ng RHW, Chan KCA, Bratman SV, Welch JJ, Divi RL, Petryshyn RA and Conley BA: Clinical utility of Epstein-Barr virus DNA testing in the treatment of nasopharyngeal carcinoma patients. Int J Radiat Oncol Biol Phys. 98:996–1001. 2017. View Article : Google Scholar : PubMed/NCBI

266 

He Q, Tuo Y, Zhou Y, Yan Y, Liu X, Zhao D, Wang Q, Luo H, Zhang Z, Meng F, et al: MB based RT-qPCR increase the clinical application of cfEBV DNA for NPC in non-endemic area of China. Sci Rep. 15:91862025. View Article : Google Scholar : PubMed/NCBI

267 

Burton BK, Ellis AG, Orr B, Chatlani S, Yoon K, Shoaff JR and Gallo D: Estimating the prevalence of Niemann-Pick disease type C (NPC) in the United States. Mol Genet Metab. 134:182–187. 2021. View Article : Google Scholar : PubMed/NCBI

268 

Wang MD, Li HT, Peng LX, Mei Y, Zheng LS, Li CZ, Meng DF, Lang YH, Xu L, Peng XS, et al: TSPAN1 inhibits metastasis of nasopharyngeal carcinoma via suppressing NF-kB signaling. Cancer Gene Ther. 31:454–463. 2024. View Article : Google Scholar : PubMed/NCBI

269 

Zhang X, Qian S, Wu P, Yu B, Yin D, Peng X, Li S, Xiao Z and Xie Z: Tumor-associated macrophage-derived itaconic acid contributes to nasopharyngeal carcinoma progression by promoting immune escape via TET2. Cell Commun Signal. 22:4132024. View Article : Google Scholar : PubMed/NCBI

270 

Zhang X, Song X, Lai Y, Zhu B, Luo J, Yu H and Yu Y: Identification of key pseudogenes in nasopharyngeal carcinoma based on RNA-Seq analysis. BMC Cancer. 21:4832021. View Article : Google Scholar : PubMed/NCBI

271 

Renaud S, Lefebvre A, Mordon S, Moralès O and Delhem N: Novel therapies boosting T cell immunity in epstein barr virus-associated nasopharyngeal carcinoma. Int J Mol Sci. 21:42922020. View Article : Google Scholar : PubMed/NCBI

272 

Cheu JWS, Lee D, Li Q, Goh CC, Bao MHR, Yuen VWH, Zhang MS, Yang C, Chan CY, Tse AP, et al: Ferroptosis suppressor protein 1 inhibition promotes tumor ferroptosis and anti-tumor immune responses in liver cancer. Cell Mol Gastroenterol Hepatol. 16:133–159. 2023. View Article : Google Scholar : PubMed/NCBI

273 

Cui K, Wang K and Huang Z: Ferroptosis and the tumor microenvironment. J Exp Clin Cancer Res. 43:3152024. View Article : Google Scholar : PubMed/NCBI

274 

Kojima Y, Tanaka M, Sasaki M, Ozeki K, Shimura T, Kubota E and Kataoka H: Induction of ferroptosis by photodynamic therapy and enhancement of antitumor effect with ferroptosis inducers. J Gastroenterol. 59:81–94. 2024. View Article : Google Scholar : PubMed/NCBI

275 

Cai W, Wu S, Lin Z, Ming X, Yang X, Yang M and Chen X: Hypoxia-induced BAP1 enhances erastin-induced ferroptosis in nasopharyngeal carcinoma by stabilizing H2A. Cancer Cell Int. 24:3072024. View Article : Google Scholar : PubMed/NCBI

276 

Chen W, Zuo F, Zhang K, Xia T, Lei W, Zhang Z, Bao L and You Y: Exosomal MIF derived from nasopharyngeal carcinoma promotes metastasis by repressing ferroptosis of macrophages. Front Cell Dev Biol. 9:7911872021. View Article : Google Scholar : PubMed/NCBI

277 

Chen X, Wang X, Zou Y, Wang Y, Duan T, Zhou Z, Huang Y and Ye Q: EMC2 suppresses ferroptosis via regulating TFRC in nasopharyngeal carcinoma. Transl Oncol. 52:1022512025. View Article : Google Scholar : PubMed/NCBI

278 

Zheng D, Chu T, Yang D, Liang S, Yang L, Yang Y, Zhang K and Ma W: Targeting ferroptosis in nasopharyngeal carcinoma: Mechanisms of therapy resistance and therapeutic opportunities. Adv Biol (Weinh). Oct 30–2025.(Epub ahead of print). View Article : Google Scholar

279 

Liang Z, Zhao W, Li X, Wang L, Meng L and Yu R: Cisplatin synergizes with PRLX93936 to induce ferroptosis in non-small cell lung cancer cells. Biochem Biophys Res Commun. 569:79–85. 2021. View Article : Google Scholar : PubMed/NCBI

280 

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 : PubMed/NCBI

281 

Takahashi R, Kamizaki K, Yamanaka K, Terai Y and Minami Y: Expression of Ferredoxin1 in cisplatin-resistant ovarian cancer cells confers their resistance against ferroptosis induced by cisplatin. Oncol Rep. 49:1242023. View Article : Google Scholar : PubMed/NCBI

282 

Perera L, Brown SM, Silver BB, Tokar EJ and Sinha BK: Ferroptosis inducers erastin and RSL3 enhance adriamycin and topotecan sensitivity in ABCB1/ABCG2-expressing tumor cells. Int J Mol Sci. 26:6352025. View Article : Google Scholar : PubMed/NCBI

283 

Bao C, Liu C, Liu Q, Hua L, Hu J, Li Z and Xu S: Liproxstatin-1 alleviates LPS/IL-13-induced bronchial epithelial cell injury and neutrophilic asthma in mice by inhibiting ferroptosis. Int Immunopharmacol. 109:1087702022. View Article : Google Scholar : PubMed/NCBI

284 

Zhang B, Chen X, Ru F, Gan Y, Li B, Xia W, Dai G, He Y and Chen Z: Liproxstatin-1 attenuates unilateral ureteral obstruction-induced renal fibrosis by inhibiting renal tubular epithelial cells ferroptosis. Cell Death Dis. 12:8432021. View Article : Google Scholar : PubMed/NCBI

285 

Cui J, Chen Y, Yang Q, Zhao P, Yang M, Wang X, Mang G, Yan X, Wang D, Tong Z, et al: Protosappanin A protects DOX-induced myocardial injury and cardiac dysfunction by targeting ACSL4/FTH1 axis-dependent ferroptosis. Adv Sci (Weinh). 11:e23102272024. View Article : Google Scholar : PubMed/NCBI

Related Articles

  • Abstract
  • View
  • Download
  • Twitter
Copy and paste a formatted citation
Spandidos Publications style
Bai S, Guo Y, Qiang J, Dai Q and Yang Y: Research progress on the regulation of ferroptosis in NPC (Review). Oncol Rep 55: 33, 2026.
APA
Bai, S., Guo, Y., Qiang, J., Dai, Q., & Yang, Y. (2026). Research progress on the regulation of ferroptosis in NPC (Review). Oncology Reports, 55, 33. https://doi.org/10.3892/or.2025.9038
MLA
Bai, S., Guo, Y., Qiang, J., Dai, Q., Yang, Y."Research progress on the regulation of ferroptosis in NPC (Review)". Oncology Reports 55.2 (2026): 33.
Chicago
Bai, S., Guo, Y., Qiang, J., Dai, Q., Yang, Y."Research progress on the regulation of ferroptosis in NPC (Review)". Oncology Reports 55, no. 2 (2026): 33. https://doi.org/10.3892/or.2025.9038
Copy and paste a formatted citation
x
Spandidos Publications style
Bai S, Guo Y, Qiang J, Dai Q and Yang Y: Research progress on the regulation of ferroptosis in NPC (Review). Oncol Rep 55: 33, 2026.
APA
Bai, S., Guo, Y., Qiang, J., Dai, Q., & Yang, Y. (2026). Research progress on the regulation of ferroptosis in NPC (Review). Oncology Reports, 55, 33. https://doi.org/10.3892/or.2025.9038
MLA
Bai, S., Guo, Y., Qiang, J., Dai, Q., Yang, Y."Research progress on the regulation of ferroptosis in NPC (Review)". Oncology Reports 55.2 (2026): 33.
Chicago
Bai, S., Guo, Y., Qiang, J., Dai, Q., Yang, Y."Research progress on the regulation of ferroptosis in NPC (Review)". Oncology Reports 55, no. 2 (2026): 33. https://doi.org/10.3892/or.2025.9038
Follow us
  • Twitter
  • LinkedIn
  • Facebook
About
  • Spandidos Publications
  • Careers
  • Cookie Policy
  • Privacy Policy
How can we help?
  • Help
  • Live Chat
  • Contact
  • Email to our Support Team