Your new experience awaits. Try the new design now and help us make it even better

REVIEW article

Front. Oncol., 03 December 2025

Sec. Cancer Genetics

Volume 15 - 2025 | https://doi.org/10.3389/fonc.2025.1701249

This article is part of the Research TopicMetal-Induced Carcinogenesis: Exploring Genetic and Epigenetic Mechanisms Underpinning Cancer DevelopmentView all articles

Modulating Nrf2 to control lipid peroxidation and ferroptosis: implications for cancer management

  • 1Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Ankara University, Ankara, Türkiye
  • 2Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Ankara University, Ankara, Türkiye
  • 3Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, Campobasso, Italy
  • 4Department of Physiology and Pharmacology ‘‘Vittorio Erspamer”, Sapienza University of Rome, Rome, Italy

Nuclear factor erythroid 2–related factor 2 (Nrf2) is a key transcription factor that regulates the expression of genes involved in cellular protection against oxidative stress and the maintenance of redox homeostasis. In addition to modulating reactive oxygen species (ROS), Nrf2 influences lipid peroxidation (LP) and ferroptosis, an iron-dependent form of regulated cell death driven by lipid peroxide accumulation. Dysregulation of Nrf2 signaling has been implicated in the pathogenesis and progression of several malignancies, where it affects tumor cell survival, chemoresistance, and metabolic reprogramming. This review summarizes current knowledge on the complex relationship between Nrf2 activity, LP dynamics, and ferroptotic mechanisms in cancer biology. It also highlights how Nrf2-dependent transcriptional programs not only regulate antioxidant responses but also contribute to cellular detoxification, metabolism, autophagy, and proteostasis, processes closely linked to tumorigenesis and cancer cell adaptability. Given that Nrf2 may have a dual role in cancer, either promoting cytoprotection or supporting tumor progression and chemoresistance, we discuss emerging strategies to selectively modulate Nrf2 and ferroptosis-related pathways to increase cancer cell sensitivity to oxidative damage and reduce resistance.

Introduction

Lipid peroxidation (LP) represents a fundamental mechanism of oxidative damage of fatty acids, typically polyunsaturated fatty acids (PUFAs), in cellular membranes and occurs when reactive oxygen species (ROS) extract a hydrogen atom from bis-allylic methylene groups, generating lipid radicals. These radicals react with molecular oxygen to form lipid peroxyl radicals, which in turn propagate the chain reaction by attacking adjacent lipids. The process culminates in the accumulation of lipid hydroperoxides and reactive aldehydes, such as malondialdehyde (MDA) and 4-hydroxynonenal (4-HNE), which disrupt membrane architecture, impair protein function, and trigger cell death pathways (1).

Ferroptosis is critically dependent on this process. Unlike other forms of cell death, such as apoptosis or necrosis, ferroptosis is defined by the accumulation of lipid peroxides beyond the detoxification capacity of the cell. The failure of antioxidant systems, particularly glutathione peroxidase 4 (GPX4), which reduces lipid hydroperoxides to non-toxic lipid alcohols, leads to the collapse of membrane integrity and ferroptotic cell death (2, 3). As such, lipid peroxidation is not merely associated with ferroptosis but serves as both a trigger and a modulator, positioning LP as a mechanistic hallmark of this form of cell death. This process is particularly relevant in therapy-resistant tumors, in which ferroptosis activation has shown potential to overcome resistance mechanisms. Furthermore, ferroptosis is involved in the pathogenesis of several degenerative diseases associated with oxidative stress (OS), highlighting its therapeutic relevance (4).

A central regulator of ferroptosis and oxidative stress responses is nuclear factor erythroid 2-related factor 2 (Nrf2), a transcription factor that controls the expression of a wide array of antioxidant and cytoprotective genes. Nrf2 plays a pivotal role in redox homeostasis and is involved in key metabolic processes, including xenobiotic detoxification, iron and heme metabolism, lipid and carbohydrate metabolism, proteostasis, and the regulation of cell death pathways (5).

The activity of Nrf2 is primarily regulated by Kelch-like ECH-associated protein 1 (Keap1), a cytosolic repressor that acts as a substrate adaptor for a Cullin-3-based E3 ubiquitin ligase complex. Under basal conditions, Keap1 binds Nrf2 and promotes its proteasomal degradation, thereby limiting its transcriptional activity. Upon exposure to oxidative or electrophilic stress, critical cysteine residues on Keap1 are modified, resulting in the stabilization and nuclear accumulation of Nrf2. Once in the nucleus, Nrf2 binds to antioxidant response elements (AREs) and induces the expression of target genes involved in glutathione synthesis, iron sequestration, and detoxification of lipid peroxides (6).

In the cancer context, aberrant activation of the Nrf2 is frequently observed and contributes to tumor progression by counteracting lipid peroxidation and ferroptosis. Many tumor types, including lung, liver, and head and neck carcinomas, harbor mutations in KEAP1 or NRF2 that lead to sustained Nrf2 activity. This persistent activation enhances the antioxidant and iron-sequestering capacity of cancer cells, promoting glutathione synthesis, increasing the expression of ferroptosis-suppressor proteins, and reducing free iron availability (7). As a result, these cells acquire a metabolic advantage that allows them to resist high levels of ROS and evade ferroptotic cell death. While this mechanism protects normal cells from oxidative damage under stress, in cancer, it facilitates proliferation, chemoresistance, and immune evasion. However, Nrf2 does not play a uniformly oncogenic role. In the early stages of oncogenesis, transient activation of Nrf2 can exert tumor-suppressive effects by enhancing the antioxidant response, limiting genomic instability, and promoting the elimination of transformed or damaged cells. This suggests a context-dependent function, where Nrf2 initially acts as a guardian against malignant transformation but may later be co-opted by cancer cells to support their survival and resistance. This duality poses both challenges and opportunities: inhibiting Nrf2 may sensitize cancer cells to ferroptosis inducers, whereas activating Nrf2 may offer protection in non-neoplastic diseases driven by ferroptosis and oxidative stress (8).

Lipid peroxidation and its role in ferroptosis

Lipid peroxidation (LP) is a defining feature of oxidative damage and a central mediator of ferroptosis. It involves the oxidative degradation of polyunsaturated fatty acids (PUFAs) within cellular membranes. Triggered by reactive oxygen species (ROS), the process begins with hydrogen abstraction from bis-allylic methylene groups, producing lipid radicals that react with molecular oxygen to form lipid peroxyl radicals. These radicals propagate the chain reaction, leading to the accumulation of lipid hydroperoxides (LOOHs) and reactive aldehydes such as malondialdehyde (MDA) and 4-hydroxynonenal (4-HNE). These byproducts destabilize membranes, disrupt protein function, and activate pro-death signaling. Mitochondrial and plasma membranes, enriched in PUFAs, are particularly susceptible under conditions of iron overload and impaired antioxidant defense (9).

LP is tightly linked to the cellular redox state. Alongside lipid radicals, cells under oxidative stress generate diverse ROS and reactive nitrogen species (RNS) capable of damaging lipids, proteins, and nucleic acids. Chronic oxidative stress, driven by metabolic or environmental factors, amplifies LP and activates regulated cell death pathways, including ferroptosis (1012). Despite their high reactivity, ROS and RNS also function as signaling molecules in processes such as proliferation, immunity, inflammation, and autophagy. This dual nature, balancing signaling and damage, remains a subject of intense debate (1315).

ROS include radical species (O2-, •OH, RO•, RO2•, ¹O2, O3) and non-radicals such as hydrogen peroxide (H2O2). RNS encompass molecules like nitric oxide (•NO), nitrogen dioxide (•NO2), and peroxynitrite (ONOO-). These species are produced through mitochondrial respiration, particularly the tricarboxylic acid (TCA) cycle, and by enzymes including NADPH oxidase (NOX), xanthine oxidase, cytochrome P450s, and nitric oxide synthase (NOS). The reaction of NO with superoxide anion generates ONOO-, a potent oxidant capable of damaging proteins and lipids (16, 17).

The chemical course of LP proceeds through initiation, propagation, and termination. In the initiation phase, hydrogen abstraction generates a lipid radical (L•), which reacts with oxygen to form a lipid peroxyl radical (LOO•). This radical perpetuates the chain reaction by abstracting hydrogen from neighboring lipids. Termination occurs through radical–radical interactions or antioxidant-mediated quenching, forming more stable byproducts while leaving behind oxidative stress residues (18, 19).

LP proceeds via both non-enzymatic and enzymatic routes. Non-enzymatic LP is predominantly iron-driven: Fe²+ catalyzes the Fenton reaction (Equation 1), converting hydrogen peroxide into hydroxyl radicals (•OH) that rapidly oxidize PUFAs—central to ferroptosis (19). Enzymatic LP involves cyclooxygenases (COXs), lipoxygenases (LOXs), and cytochrome P450s (CYPs). COXs oxidize arachidonic acid into prostaglandins, modulating inflammation and apoptosis (20). LOXs catalyze PUFA peroxidation and play roles in immune responses (21). Cytochrome P450 oxidoreductase (POR), located on the endoplasmic reticulum, participates in lipid oxidation via CYP activation (22).

Fe3++ H2O2 Fe3+H2O2(1)
Fe3+H2O2 Fe2++ HO2+ H+(2)
Fe3++ HO2 Fe2++ O2+ H+(3)
Fe2++ H2O2 Fe3++ OH + OH(4)

Equation 1: Fenton reaction.

When examining the Fenton reaction (Equations 14) in vitro, the oxidation processes undergone by free radicals can be monitored using various model studies. One of the most well-known methods is that performed with hydrogen peroxide and iron salts (23). In many publications, EDTA has been selected as a reagent for both inhibiting and accelerating oxidation (24).

In vivo, Fe2+ ions have been found in the cell cytosol of organisms. Excessive levels of biogenic iron in the body are known to be associated with the pathology of certain diseases. These include inflammation, neurodegeneration, neuroinflammation, and even cancer (25, 26). Some sources have reported that excess iron in the body may also be associated with the development of liver disorders, cardiovascular diseases, and lung diseases (27, 28). The Fenton reaction is gaining attention in some in vivo applications. For example, nanotechnology methods involving polymer nanoparticles and light-sensitive nanoparticles have been used in many successful applications where the Fenton reaction has been adapted to the cancer microenvironment. It has been observed that the Fenton reaction, catalyzed by light-sensitive nanoparticles, leads to healing in cancer cells (29). The OH• radical released by the Fenton reaction is one of the most reactive molecules that causes oxidative damage in cells. This property is considered a potential therapeutic approach to prevent cancer development. The mechanisms of action of some known cancer drugs, such as doxorubicin (DOX) [13], β-lapachone [14], and cisplatin [15], are known to involve attacking tumor cells with the OH• radical via the Fenton reaction (30, 31).

To mitigate LOOH-induced toxicity, cells rely on glutathione peroxidase 4 (GPX4), a selenoenzyme that reduces lipid hydroperoxides to inert alcohols using glutathione (GSH) as an electron donor. Inhibition of GPX4—whether pharmacologically (e.g., by erastin or RSL3) or via GSH depletion—leads to lipid peroxide accumulation, membrane disruption, and ferroptotic death (32).

Ferroptosis is defined by three core biochemical events: (i) PUFA-phospholipid oxidation, (ii) the presence of redox-active iron, and (iii) compromised antioxidant defenses, particularly GSH and GPX4 (33, 34) (Figure 1). Mitochondrial TCA cycle activity, fatty acid β-oxidation, and amino acid metabolism contribute to ROS generation that, in concert with iron, amplify LP and drive ferroptotic signaling (35, 36).

Figure 1
Flowchart depicting the process of ferroptosis. GPX4 and GSH work together to inhibit lipid peroxidation caused by Fe2+. Lipid peroxidation leads to ferroptosis, which is cell death. The outcomes are cardiomyopathy, stroke, and ischemia/reperfusion, as well as heart failure, neurodegeneration, and reperfusion damage.

Figure 1. The role of ferroptosis and lipid peroxidation in the development of diseases.

Morphologically, ferroptosis diverges from classical apoptosis and necrosis. It lacks features such as apoptotic body formation or necrotic lysis. Instead, it is marked by shrunken mitochondria with dense membranes and ruptured outer membranes. Biochemical hallmarks include GSH depletion, GPX4 inactivation, and lipid peroxide accumulation (3739).

Ferroptosis plays a pathogenic role in ischemia–reperfusion injury (IRI). Upon reperfusion, elevated iron levels exacerbate ROS generation and LP, triggering ferroptotic cell death. This amplifies tissue injury and highlights ferroptosis as a therapeutic target for IRI-related pathologies (40, 41).

In cancer cells, chronic oxidative stress often creates selective vulnerability due to impaired GSH metabolism. Under these conditions, the GPX4–GSH axis becomes critical for survival. When GSH is depleted, GPX4 fails to detoxify lipid peroxides, culminating in ferroptosis. Conversely, tumor cells that upregulate GPX4 or maintain high GSH pools can evade ferroptosis, supporting proliferation and resistance to oxidative therapies (42, 43).

Nrf2 as a central regulator of lipid peroxidation and ferroptosis

Ferroptosis is primarily driven by excessive lipid peroxidation and intracellular iron accumulation. These processes are tightly controlled by the cellular redox environment, which is regulated by an integrated antioxidant network. Among the transcriptional regulators involved, Nrf2 plays a central role in maintaining redox balance, supporting mitochondrial function, and limiting the progression of ferroptosis (44, 45).

Nrf2 modulates several antioxidant and metabolic pathways that counteract the toxic effects of ROS and lipid peroxides. Its activity influences energy metabolism, mitochondrial dynamics, and the transcription of numerous cytoprotective genes. In particular, Nrf2 coordinates the expression of enzymes involved in the biosynthesis and recycling of glutathione (GSH), including glutamate-cysteine ligase catalytic and modifier subunits (GCLC and GCLM), glutathione synthetase (GSS), and the cystine/glutamate antiporter component SLC7A11. In parallel, it regulates the thioredoxin antioxidant system through targets such as thioredoxin-1 (TXN) and thioredoxin reductase-1 (TXNRD1), which collectively maintain the intracellular redox environment (6, 46).

The activity of Nrf2 is primarily controlled through its interaction with Kelch-like ECH-associated protein 1 (Keap1), a cytoplasmic repressor that facilitates the ubiquitin-dependent proteasomal degradation of Nrf2 (Figure 2).

Figure 2
Flowchart illustrating the pathway involving ROS, Keap1, Nrf2, and related proteins. PI3K activates Akt, leading to GSK-3β activation, which influences Fyn. ROS oxidizes Keap1, releasing Nrf2 for nuclear translocation. Nrf2/ARE complex results in antioxidant protein expression. Phosphorylation events by protein kinase C, AMPK, CK2, P38, MAPK, and ERK are indicated.

Figure 2. Nrf2/Keap1 pathway under oxidative stress.

Since the cloning of Keap1, tremendous progress has been achieved towards comprehending the mechanism of Keap1-mediated negative regulation of Nrf2. It has been postulated that Keap1 works as a molecular switch that is capable to turn the Nrf2-signaling pathway on or off according to intracellular redox circumstances. Serving as a molecular switch, Keap1 exhibits twin functions: it is able to (i) “sense” a disturbance in the redox balance and (ii) turn the Nrf2-mediated response on or off. Recent investigations have examined how these two roles are achieved by Keap1.

Under basal conditions, Keap1 switches the Nrf2-signaling pathway off and maintains low basal levels of Nrf2 by constantly targeting Nrf2 for ubiquitin-mediated protein degradation. When Keap1 “senses” a disturbance in the redox balance, it switches the Nrf2-signaling pathway on.

Under homeostatic conditions, Keap1 binds newly synthesized Nrf2 and targets it for continuous degradation via the proteasome, thereby maintaining low intracellular levels of Nrf2. Upon exposure to oxidative or electrophilic stress, cysteine residues on Keap1 undergo covalent modifications that impair its ability to recognize and ubiquitinate Nrf2. As a result, newly synthesized Nrf2 escapes degradation and translocates into the nucleus, where it binds to antioxidant response elements (AREs) and activates the transcription of a broad array of genes involved in redox regulation, detoxification, and metabolic adaptation (47, 48) (Figure 2). Several of the genes regulated by Nrf2 are directly involved in modulating ferroptosis. One of the most important is GPX4, a selenoenzyme that reduces phospholipid hydroperoxides to lipid alcohols. This activity prevents the accumulation of toxic lipid peroxides and preserves membrane integrity, as mentioned above. The function of GPX4 depends on the availability of GSH, whose synthesis and regeneration are under Nrf2 control. In addition, Nrf2 induces the expression of SLC7A11, which promotes cystine uptake and supports GSH production, thereby reinforcing the cellular defense against ferroptosis (49).

Nrf2 also plays a role in iron metabolism, which is closely linked to ferroptotic susceptibility. By promoting the expression of ferritin heavy and light chains (FTH1 and FTL) and the iron exporter ferroportin (FPN1), Nrf2 contributes to the reduction of the labile iron pool, thereby limiting the generation of ROS through the Fenton reaction. In the presence of excess iron, this reaction enhances lipid peroxidation and promotes ferroptotic cell death (50, 51).

When the complex structure of the Keap1-Nrf2 signaling pathway is examined, we see that it is modulated by a system that includes phosphoinositol 3-kinase (PI3K)/protein kinase B (Akt), protein kinase C and mitogen-activated protein kinase molecules. Research showed that several molecules show antioxidant effects through the PI3K/Akt/Nrf2 pathway. The PI3K/Akt signaling pathway regulates Nrf2 activity through GSK-3β, a serine/threonine kinase encoded by the alpha (α) and beta (β) genes. GSK-3β, a key Akt target, has multiple functions, including regulating cell proliferation, apoptosis, glycogen metabolism, and stem cell renewal (52).

The Nrf2-Keap1-ARE signaling axis plays a crucial role in antioxidant defense and is also effective in defense against tumor development. It is also known to play a role in chemotherapeutic drug resistance. Experimental data demonstrate a relationship between impaired autophagy and activation of the Nrf2 pathway. Specifically, p62 facilitates Keap1 degradation via selective autophagy, leading to Nrf2 translocation to the nucleus where it transcriptionally activates downstream antioxidant enzyme expression, protecting cells from OS. Since Nrf2 regulates p62 transcription, a positive feedback loop involving p62, Keap1, and Nrf2 is formed, which further enhances the cell’s protective effects (53).

A controversial issue in recent years is whether cancer cells are dependent on autophagy. One study suggests that while some cells exhibit direct autophagy-dependent development, these cells adapt to bypass the autophagy mechanism. These results demonstrate that resistance to autophagy inhibition is an adaptive mechanism that promotes new tumor formation (54). Studies have shown that ROS-induced autophagy is primarily mediated by O2- during long-term fasting. H2O2 is produced immediately after fasting and regulates autophagy. Furthermore, the observed relationship between autophagy and ROS varies depending on cell type. For example, autophagy is activated in renal tubular cells as a result of high ROS exposure. Thus, mitochondrial destruction and associated kidney damage can be observed. However, in tumor cells, ROS-induced autophagy activation can promote cell development and cancer progression (55).

The Nrf2-dependent adaptive response, which includes phase II detoxifying enzymes, antioxidants, and transporters that protect cells from further carcinogenic assaults, is how chemopreventive substances or synthetic chemicals carry out their chemopreventive action. As a result, Nrf2 has been considered a “good” protein that shields people from genotoxic harm brought on by carcinogens. Not only does Nrf2 prevent normal cells from becoming cancerous, but it also helps cancer cells survive in harmful environments. Examples of the microenvironmental conditions influencing the dual effects of Nrf2 in cancer include overexpression of the GSTP1 enzyme, Keap1 mutations, loss of heterozygosity, and hypoxia/reoxygenation.

The role of ferroptosis in cancer biology

Due to their altered redox status and increased iron dependency, cancer cells are particularly susceptible to ferroptosis. This regulated form of cell death, previously discussed in molecular terms, has now been recognized as a crucial determinant in tumor progression and therapy response. Recent studies have highlighted how perturbations in iron metabolism and oxidative stress responses contribute to both the development and suppression of malignancies. While iron accumulation and redox imbalance can promote genomic instability and tumor growth, they also create conditions that sensitize cancer cells to ferroptosis (56).

Inducing ferroptosis has emerged as a promising strategy to overcome resistance to chemotherapy and targeted therapies. Agents such as sorafenib, sulfasalazine, and artesunate have shown efficacy in promoting ferroptotic death in tumor models, particularly in those resistant to apoptosis-based treatments (57). At the same time, defects in ferroptotic signaling and antioxidant systems have been associated with poor prognosis in several cancer types. However, ferroptosis may exert a dual role in cancer, as persistent iron loading, metabolic regulation, and adaptation to oxidative stress may facilitate tumor survival (40, 58, 59).

Emerging evidence also suggests that ferroptosis is tightly coupled to lipid metabolism, membrane repair systems, and oxidative signaling. In tumors with high metabolic plasticity, ferroptosis-targeting strategies must account for the context-specific redox vulnerabilities that define their response to treatment (60, 61).

Nrf2 activation offers extensive and sustained cytoprotection, which is frequently taken over by cancer cells to enable their survival in adverse environments. Furthermore, resistance to chemotherapy, radiation, and immunotherapies is linked to Nrf2 activation in established human malignancies. Tumor-associated macrophages (TAMs) can also activate Nrf2, which promotes an immunosuppressive, anti-inflammatory tumor immune microenvironment (TIME) in addition to cancer cells. Numerous metabolites produced by cancer cells, including itaconate, L-kynurenine, lactic acid, and hyaluronic acid, have been demonstrated to activate Nrf2 and play a significant role in regulating the TIME and tumor-TAMs interaction. Pro-inflammatory cytokines are suppressed, programmed cell death ligand 1 (PD-L1), macrophage colony-stimulating factor (M-CSF), and kynureninase are expressed more frequently, cytotoxic labile heme is catabolized more quickly, and TAMs’ metabolic adaptation is aided. These effects of Nrf2 in TIME are context-dependent and involve several mechanisms. This knowledge offers potential as well as obstacles for the strategic targeting of Nrf2 in cancer (62).

Pancreatic cancer

Pancreatic ductal adenocarcinoma (PDAC), the most common form of pancreatic cancer, is a highly aggressive malignancy associated with poor prognosis and limited responsiveness to conventional therapies. It frequently originates from oncogenic mutations in the Kirsten rat sarcoma viral oncogene homolog (KRAS), which are present in over 90% of PDAC cases. Although ferroptosis was initially characterized in the context of RAS-driven cell death, it is now established as a distinct form of regulated necrosis initiated by lipid peroxidation. Dysregulation of ferroptosis contributes to both tumor initiation and progression in PDAC. On one hand, ferroptotic damage can promote chronic inflammation through the release of damage-associated molecular patterns (DAMPs), which enhance a tumor-promoting microenvironment. This inflammatory milieu can act synergistically with KRAS signaling to facilitate malignant transformation (63, 64).

On the other hand, ferroptosis represents a promising vulnerability in PDAC. The myokine irisin, secreted by skeletal muscle during physical activity, has been proposed as a potential adjunct to ferroptosis-inducing agents. Irisin appears to sensitize PDAC cells to ferroptosis, possibly by modulating redox balance and metabolic pathways (65).

Notably, the impact of ferroptosis in PDAC is modulated by genetic and microenvironmental factors. For example, the mutational status of tumor protein p53 (TP53) influences the cellular response to oxidative damage. Additionally, ferroptosis can promote autophagy and perturb intracellular iron storage, resulting in increased labile iron and further susceptibility to lipid peroxidation. These mechanisms underscore the dual nature of ferroptosis in PDAC, acting as both a suppressive and a tumor-promoting force depending on the context (66).

Hepatocellular carcinoma

Hepatocellular carcinoma (HCC), the most frequent primary liver malignancy, accounts for over 90% of all liver cancer cases. Its pathogenesis is strongly associated with chronic inflammation, metabolic stress, and iron accumulation—factors that closely intersect with ferroptotic signaling. Recent studies have highlighted the involvement of ferroptosis-related genes in HCC development and therapy response. In particular, resistance to sorafenib, a multi-kinase inhibitor widely used as first-line therapy in advanced HCC, has been linked to impaired ferroptotic mechanisms (67).

Histological analyses of HCC tumors frequently reveal intracellular iron accumulation and elevated lipid peroxide levels (68). Interestingly, some HCC cell lines that are resistant to apoptosis or necrosis remain highly sensitive to ferroptosis-inducing compounds. This suggests that ferroptosis could represent an alternative pathway for eradicating otherwise therapy-resistant tumor cells. Sorafenib has been shown to induce ferroptosis in HCC models by inhibiting the cystine/glutamate antiporter system Xc- and affecting key antioxidant enzymes (69, 70).

Breast cancer

Breast cancer remains the most commonly diagnosed malignancy worldwide, with its global burden continuing to rise. Accumulating evidence indicates that ferroptosis contributes to both tumor progression and therapeutic resistance in breast cancer, particularly in subtypes lacking effective targeted therapies. Pharmacological induction of ferroptosis has been shown to suppress tumor cell viability, highlighting its potential as a therapeutic avenue (71, 72).

In triple-negative breast cancer (TNBC), one of the most aggressive and treatment-resistant subtypes, ferroptosis appears to be tightly linked to cysteine availability. A study demonstrated that deprivation of cysteine induces ferroptotic death in TNBC cells, which was rescued by deferoxamine (DFO), an iron chelator, and ferrostatin-1, a ferroptosis inhibitor (73). These findings implicate cysteine metabolism as a critical regulator of ferroptosis susceptibility and suggest that metabolic vulnerabilities in TNBC could be exploited to enhance treatment efficacy (74).

Lung cancer

Lung carcinoma represents a major contributor to global cancer mortality. In experimental models, induction of ferroptosis in lung cancer cells has been associated with cell cycle arrest at the G2/M transition, along with a reduction in the G0/G1 and S phase cell populations. These changes suggest that ferroptotic stress disrupts mitotic progression, impairing the proliferative potential of lung cancer cells (75, 76).

Despite this vulnerability, lung tumors are frequently characterized by resistance to ferroptosis, partly due to their constant exposure to high oxygen levels. Chronic oxidative stress in pulmonary tissue imposes selective pressure for the upregulation of antioxidant systems, allowing tumor cells to counteract ferroptosis-inducing conditions (77). Studies have reported that ferroptosis-related pathways are suppressed in lung cancer, including reduced ROS accumulation and elevated expression of GPX4, a key enzyme involved in lipid peroxide detoxification (78, 79). This adaptive resistance underscores the complexity of ferroptosis regulation in lung tumors and highlights the need for context-specific approaches to restore ferroptotic vulnerability (80, 81).

Gastric cancer

Gastric cancer (GC) ranks among the most prevalent gastrointestinal malignancies and remains a leading cause of cancer-related deaths worldwide. Current therapeutic strategies rely predominantly on surgical intervention, yet outcomes remain suboptimal in advanced disease stages (82).

Iron metabolism has emerged as a crucial determinant of gastric tumor biology, given the stomach’s central role in systemic iron absorption and regulation. Helicobacter pylori infection, a key etiological factor in GC, is strongly associated with iron-deficiency disorders such as anemia (83, 84). Alterations in iron availability not only impact mucosal immunity but may also drive oxidative stress and lipid peroxidation, thereby facilitating malignant transformation. Disrupted iron homeostasis has been shown to influence multiple hallmarks of cancer, including proliferation, angiogenesis, and resistance to cell death, suggesting a mechanistic link between ferroptosis and gastric tumorigenesis (85).

Nrf2 as a therapeutic target in ferroptosis-driven cancer

Through coordinated regulation of antioxidant capacity, lipid peroxide detoxification, and iron metabolism, the transcription factor Nrf2 emerges as a key modulator of ferroptosis. Under physiological conditions, Nrf2 activation protects cells against oxidative insults by upregulating cytoprotective genes. However, in cancer, sustained activation of Nrf2, often driven by mutations that impair its repressor Keap1, can promote tumor progression, chemoresistance, and escape from ferroptosis. This dual behavior highlights the importance of context when targeting Nrf2 in therapies aimed at modulating ferroptosis (86).

While Nrf2 activation prevents ferroptosis under homeostatic conditions by sustaining antioxidant gene expression, in malignant contexts, it can paradoxically shield tumor cells from ferroptotic stress. Persistent Nrf2 signaling has been associated with enhanced proliferation, metastatic dissemination, and resistance to multiple chemotherapeutic agents. This has positioned the Nrf2 pathway as a major focus in the design of cancer therapies based on ferroptosis induction (8789).

Multiple pharmacological agents have been identified as modulators of Nrf2 activity. Among the Nrf2 inhibitors, metformin, isoorientin, wogonin, trigonelline, and sulfasalazine have shown antitumor activity by downregulating Nrf2 through diverse mechanisms (Figure 3). For example, sulfasalazine inhibit the Nrf2/xCT signaling axis. Metformin impairs Nrf2 nuclear translocation. Isoorientin acts via inhibition of the SIRT6/Nrf2/GPX4 pathway, while trigonelline suppresses Nrf2 through RNA interference (9094).

Figure 3
Chemical structures labeled (a) to (e). Structure (a) shows a polyamine compound. Structure (b) depicts a flavonoid glycoside with multiple hydroxyl groups. Structure (c) is a flavonoid with a methoxy group. Structure (d) illustrates a pyridine derivative with a carboxylate ion. Structure (e) features a sulfonamide azo compound with carboxylic and hydroxyl groups.

Figure 3. Chemical structures of some Nrf2 inhibitors molecules. metformin (a), isoorientin (homoorientin) (b), wogonin (c), trigonelline (d), sulfasalazine (e).

Conversely, several natural and synthetic molecules have demonstrated anticancer properties through activation of Nrf2. These include dexmedetomidine, dimethyl fumarate (DMF), tagitinin C, β-caryophyllene (BCP), and apigenin, as well as molecular regulators such as miR-24, miR-24-3p, Sirt1, and levistilide A. Their mechanisms include activation of the Nrf2/HO-1 pathway (levistilide A, dexmedetomidine), enhancement of Nrf2 nuclear translocation (BCP, Sirt1, tagitinin C, DMF), and stabilization of Nrf2 via inhibition of Keap1 expression (miR-24-3p) (95103) (Figure 4).

Figure 4
Chemical structures labeled (a) through (e): (a) a structure with a benzene and imidazole ring, (b) a diester with a conjugated double bond, (c) a cyclic compound with multiple functional groups including hydroxyl and carbonyl, (d) a bicyclic structure with methyl groups, (e) a flavonoid compound with hydroxyl groups and a benzopyran ring.

Figure 4. Chemical structures of some Nrf2 activator molecules. Dexmedetomidine (a) dimethyl fumarate (b), tagitinin C (c), caryophyllene (BCP) (d), apegenin (APG) (e).

This apparent contradiction, whereby both inhibition and activation of Nrf2 are explored as anticancer strategies, reflects the context-dependent nature of Nrf2 signaling. In some tumor types or stages, enhancing Nrf2 activity may restore redox balance, reduce chronic inflammation, and protect normal cells from malignant transformation. In contrast, in established tumors characterized by constitutive Nrf2 activation, further stimulation may exacerbate chemoresistance and tumor growth. This duality underlines the necessity of precise molecular characterization of the tumor microenvironment when considering Nrf2 as a therapeutic target (86).

Phase 1 clinical trials are ongoing for Nrf2 activators. Phase 3 clinical trials are also underway for vatiquinone, a 15-lipoxygenase inhibitor with both antioxidant and anti-inflammatory effects (104). Vatiquinone (Figure 5) target multiple metabolic pathways in the mitochondria. Omaveloxolone (Figure 5) is the first FDA-approved drug used against Friedreich’s ataxia (FA). Its mechanism of action is by targeting Nrf2 in the antioxidant pathway (105).

Figure 5
Chemical structures of two compounds: (a) a hexagonal ring with two hydroxyl groups, a long alkyl chain, and three carbonyl groups; (b) a complex polycyclic structure with multiple rings, nitrogen, and fluorine atoms.

Figure 5. Chemical structures of (a) Vatiquinone and (b) Omaveloxolone.

Given its broad regulatory functions, Nrf2 remains a compelling and complex target in ferroptosis-based cancer therapies (Figure 6). Its dual roles, protective in normal physiology and potentially oncogenic in malignancy, require precision strategies that take into account the tumor context, mutation profile, and redox environment.

Figure 6
Diagram illustrating the role of Nrf2 in lipid peroxidation and ferroptosis. Nrf2, interacting with ARE, influences GPX4 regulation. GPX4 mitigates lipid peroxidase activity. Factors like Fe and H2O2 contribute to ROS formation, impacting lipid peroxidase. This cascade is linked to ferroptosis, implicated in cancer cells formation and neurodegeneration. Elements like lipid alcohols and GSSG also interact with GPX4.

Figure 6. Formation of ferroptosis-related pathologies and the role of Nrf2.

Since Nrf2’s discovery, there has been an increasing amount of evidence demonstrating its beneficial effect in preventing cancer and how it is a crucial transcription factor in shielding people from illnesses linked to oxidative stress. Finding Nrf2 activators for chemoprevention has been the primary focus of research up to this point, but new discoveries indicate that Nrf2 has a “dark” side. According to in vitro research, Nrf2 overexpression can raise the expression of a number of intracellular redox-balancing proteins, phase II detoxifying enzymes, and transporters, which can provide cancer cells a growth advantage and result in chemotherapy resistance. It is important to verify the cancer promoting role of Nrf2 observed in cancer cell lines and tumor biopsies, using in vivo animal models (106).

Discovering Nrf2’s “dark” side offers a chance for therapeutic intervention against chemoresistance. Chemotherapy with a Nrf2 inhibitor can be utilized to make cancer cells more susceptible to chemical therapy. Nrf2 would be a more effective therapeutic target than a single downstream gene, such HMOX-1 or TrxR, because it is a transcription factor that controls the production of multiple downstream genes that shield cancer cells from apoptosis (106).

Conclusions

The discovery of ferroptosis as a distinct, iron-dependent form of regulated cell death has significantly advanced our understanding of redox biology and its relevance to cancer progression and therapy. Central to this process is lipid peroxidation, initiated by excessive ROS and altered iron metabolism, which disrupts membrane structure and leads to cell death. Although ROS are integral to physiological signaling, their uncontrolled accumulation shifts the redox equilibrium toward oxidative damage, emphasizing the delicate balance required for cellular homeostasis.

Ferroptosis represents a dual mechanism in oncology. It can suppress tumor growth by eliminating cells with impaired antioxidant capacity, particularly in contexts of chemoresistance or metastasis. However, ferroptotic damage may also promote chronic inflammation and create a tumor-permissive microenvironment, depending on the genetic and immune status of the tissue.

Nrf2 is a critical modulator of this process. Under normal conditions, it preserves redox stability through the regulation of antioxidant and metabolic genes. In cancer, however, its sustained activation, often linked to loss-of-function mutations in Keap1, confers resistance to ferroptosis and facilitates tumor progression. This dual function highlights both the potential and the complexity of targeting Nrf2 in anticancer strategies.

Research on NRF2 cancer still has a lot of unsolved questions. Whether constitutive activation and regulated activation of NRF2 increase the expression of the same target genes is still unknown (107). It wouldn’t be shocking if a particular threshold of NRF2 activation altered the transcriptome, as some research has demonstrated that some target genes are included in the basal but not the induced NRF2 transcriptome and vice versa. Additionally, many potential target genes need to be confirmed, and the NRF2 transcriptome still needs to be completely described. A thorough comprehension of the NRF2 transcriptome will enable us to investigate therapeutic tumor editing and improve our comprehension of the negative aspects of NRF2 in the characteristics of cancer. Furthermore, KEAP1-independent NRF2 control mechanisms should be investigated in relation to cancer treatment and prevention (108). Targeting this transcription factor may be a useful therapeutic strategy, as evidenced by the high frequency of NRF2 in cancer hallmarks. While NRF2 inhibitors may be used to treat cancer, NRF2 activators may be utilized to prevent chemical carcinogenesis (108).

Further investigations should aim to clarify how Nrf2 influences ferroptosis across different tumor contexts, and how its modulation interacts with oncogenic signaling, iron metabolism, and immune regulation. A context-aware approach will be essential to harness ferroptosis as a therapeutic mechanism while minimizing unintended effects on healthy tissues.

Author contributions

RT: Writing – original draft, Resources. IA: Conceptualization, Methodology, Project administration, Visualization, Writing – original draft, Writing – review & editing. BY: Data curation, Resources, Writing – original draft. AM: Project administration, Software, Supervision, Writing – original draft, Writing – review & editing. SD: Investigation, Project administration, Supervision, Validation, Writing – original draft, Writing – review & editing. SS: Project administration, Writing – original draft, Writing – review & editing. LS: Funding acquisition, Project administration, Writing – original draft, Writing – review & editing.

Funding

The author(s) declared financial support was received for this work and/or its publication. This work was partially supported by the Italian Ministry of Research, under the complementary actions to the NRRP “D34Health - Digital Driven Diagnostics, prognostics and therapeutics for sustainable Health care”.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

The author(s) declared that they were an editorial board member of Frontiers, at the time of submission. This had no impact on the peer review process and the final decision.

Generative AI statement

The author(s) declare that no Generative AI was used in the creation of this manuscript.

Any alternative text (alt text) provided alongside figures in this article has been generated by Frontiers with the support of artificial intelligence and reasonable efforts have been made to ensure accuracy, including review by the authors wherever possible. If you identify any issues, please contact us.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Ayala A, Muñoz MF, and Argüelles S. Lipid peroxidation: production, metabolism, and signaling mechanisms of malondialdehyde and 4-hydroxy-2-nonenal. Oxid Med Cell Longev. (2014) 2014:360438. doi: 10.1155/2014/360438

PubMed Abstract | Crossref Full Text | Google Scholar

2. Stockwell BR. Ferroptosis turns 10: Emerging mechanisms, physiological functions, and therapeutic applications. Cell. (2022) 185:2401–21. doi: 10.1016/j.cell.2022.06.003

PubMed Abstract | Crossref Full Text | Google Scholar

3. Dai E, Chen X, Linkermann A, Jiang X, Kang R, Kagan VE, et al. A guideline on the molecular ecosystem regulating ferroptosis. Nat Cell Biol. (2024) 26:1447–57. doi: 10.1038/s41556-024-01360-8

PubMed Abstract | Crossref Full Text | Google Scholar

4. von Krusenstiern AN, Robson RN, Qian N, Qiu B, Hu F, Reznik E, et al. Identification of essential sites of lipid peroxidation in ferroptosis. Nat Chem Biol. (2023) 19:719–30. doi: 10.1038/s41589-022-01249-3

PubMed Abstract | Crossref Full Text | Google Scholar

5. Davinelli S, Medoro A, Intrieri M, Saso L, Scapagnini G, and Kang JX. Targeting NRF2-KEAP1 axis by Omega-3 fatty acids and their derivatives: Emerging opportunities against aging and diseases. Free Radic Biol Med. (2022) 193:736–50. doi: 10.1016/j.freeradbiomed.2022.11.017

PubMed Abstract | Crossref Full Text | Google Scholar

6. Davinelli S, Medoro A, Savino R, and Scapagnini G. Sleep and oxidative stress: current perspectives on the role of NRF2. Cell Mol Neurobiol. (2024) 44:52. doi: 10.1007/s10571-024-01487-0

PubMed Abstract | Crossref Full Text | Google Scholar

7. Xu K, Ma J, Hall SRR, Peng RW, Yang H, and Yao F. Battles against aberrant KEAP1-NRF2 signaling in lung cancer: intertwined metabolic and immune networks. Theranostics. (2023) 13:704–23. doi: 10.7150/thno.80184

PubMed Abstract | Crossref Full Text | Google Scholar

8. Panieri E, Buha A, Telkoparan-Akillilar P, Cevik D, Kouretas D, Veskoukis A, et al. Potential applications of NRF2 modulators in cancer therapy. Antioxidants (Basel). (2020) 9:193. doi: 10.3390/antiox9030193

PubMed Abstract | Crossref Full Text | Google Scholar

9. Lin Z, Liu J, Kang R, Yang M, and Tang D. Lipid metabolism in ferroptosis. Adv Biol (Weinh). (2021) 8):e2100396. doi: 10.1002/adbi.202100396

PubMed Abstract | Crossref Full Text | Google Scholar

10. Liu C, Miyajima T, Melangath G, Miyai T, Vasanth S, Deshpande N, et al. Ultraviolet A light induces DNA damage and estrogen-DNA adducts in Fuchs endothelial corneal dystrophy causing females to be more affected. Proc Natl Acad Sci U.S.A. (2020) 117:573–83. doi: 10.1073/pnas.1912546116

PubMed Abstract | Crossref Full Text | Google Scholar

11. Chen X, Li J, Kang R, Klionsky DJ, and Tang D. Ferroptosis: machinery and regulation. Autophagy. (2021) 17:2054–81. doi: 10.1080/15548627.2020.1810918

PubMed Abstract | Crossref Full Text | Google Scholar

12. Singhal R, Mitta SR, Das NK, Kerk SA, Sajjakulnukit P, Solanki S, et al. HIF-2α activation potentiates oxidative cell death in colorectal cancers by increasing cellular iron. J Clin Invest. (2021) 131:e143691. doi: 10.1172/JCI143691

PubMed Abstract | Crossref Full Text | Google Scholar

13. D’Autréaux B and Toledano MB. ROS as signalling molecules: mechanisms that generate specificity in ROS homeostasis. Nat Rev Mol Cell Biol. (2007) 8:813–24. doi: 10.1038/nrm2256

PubMed Abstract | Crossref Full Text | Google Scholar

14. Hameister R, Kaur C, Dheen ST, Lohmann CH, and Singh G. Reactive oxygen/nitrogen species (ROS/RNS) and oxidative stress in arthroplasty. J BioMed Mater Res B Appl Biomater. (2020) 108:2073–87. doi: 10.1002/jbm.b.34546

PubMed Abstract | Crossref Full Text | Google Scholar

15. Puglia GD. Reactive oxygen and nitrogen species (RONS) signalling in seed dormancy release, perception of environmental cues, and heat stress response. Plant Growth Regul. (2024) 103:9–32. doi: 10.1007/s10725-023-01094-x

Crossref Full Text | Google Scholar

16. Aranda-Rivera AK, Cruz-Gregorio A, Arancibia-Hernández YL, Hernández-Cruz EY, and Pedraza-Chaverri J. RONS and oxidative stress: an overview of basic concepts. Oxygen. (2022) 2:437–78. doi: 10.3390/oxygen2040030

Crossref Full Text | Google Scholar

17. Jomova K, Raptova R, Alomar SY, Alwasel SH, Nepovimova E, Kuca K, et al. Reactive oxygen species, toxicity, oxidative stress, and antioxidants: chronic diseases and aging. Arch Toxicol. (2023) 97:2499–574. doi: 10.1007/s00204-023-03562-9

PubMed Abstract | Crossref Full Text | Google Scholar

18. Greene LE, Lincoln R, and Cosa G. Rate of Lipid Peroxyl Radical Production during Cellular Homeostasis Unraveled via Fluorescence Imaging. J Am Chem Soc. (2017) 139:15801–11. doi: 10.1021/jacs.7b08036

PubMed Abstract | Crossref Full Text | Google Scholar

19. Zheng Y, Sun J, Luo Z, Li Y, and Huang Y. Emerging mechanisms of lipid peroxidation in regulated cell death and its physiological implications. Cell Death Dis. (2024) 15:859. doi: 10.1038/s41419-024-07244-x

PubMed Abstract | Crossref Full Text | Google Scholar

20. Ucar BI, Erikci A, Kosemehmetoglu K, Ozkul C, Iskit AB, Ucar G, et al. Effects of endothelin receptor blockade and COX inhibition on intestinal I/R injury in a rat model: Experimental research. Int J Surg. (2020) 83:89–97. doi: 10.1016/j.ijsu.2020.08.061

PubMed Abstract | Crossref Full Text | Google Scholar

21. Yang WS, Kim KJ, Gaschler MM, Patel M, Shchepinov MS, and Stockwell BR. Peroxidation of polyunsaturated fatty acids by lipoxygenases drives ferroptosis. Proc Natl Acad Sci U.S.A. (2016) 113:E4966–75. doi: 10.1073/pnas.1603244113

PubMed Abstract | Crossref Full Text | Google Scholar

22. Su LJ, Zhang JH, Gomez H, Murugan R, Hong X, Xu D, et al. Reactive oxygen species-induced lipid peroxidation in apoptosis, autophagy, and ferroptosis. Oxid Med Cell Longev. (2019) 2019:5080843. doi: 10.1155/2019/5080843

PubMed Abstract | Crossref Full Text | Google Scholar

23. Zamenhof S, Griboff G, and Marullo N. Studies on the resistance of desoxyribonucleic acids to physical and chemical factors. Biochim Biophys Acta. (1954) 13:459–70. doi: 10.1016/0006-3002(54)90362-5

PubMed Abstract | Crossref Full Text | Google Scholar

24. Sreenivasan S and Rathore AS. Combined presence of ferrous ions and hydrogen peroxide in normal saline and in vitro models induces enhanced aggregation of therapeutic igG due to hydroxyl radicals. Mol Pharm. (2023) 20:3033–48. doi: 10.1021/acs.molpharmaceut.3c00051

PubMed Abstract | Crossref Full Text | Google Scholar

25. Hagemeier J, Geurts JJ, and Zivadinov R. Brain iron accumulation in aging and neurodegenerative disorders. Expert Rev Neurother. (2012) 12:1467–80. doi: 10.1586/ern.12.128

PubMed Abstract | Crossref Full Text | Google Scholar

26. Jung M, Mertens C, Tomat E, and Brüne B. Iron as a central player and promising target in cancer progression. Int J Mol Sci. (2019) 20:273. doi: 10.3390/ijms20020273

PubMed Abstract | Crossref Full Text | Google Scholar

27. Kobayashi M, Suhara T, Baba Y, Kawasaki NK, Higa JK, and Matsui T. Pathological roles of iron in cardiovascular disease. Curr Drug Targets. (2018) 19:1068–76. doi: 10.2174/1389450119666180605112235

PubMed Abstract | Crossref Full Text | Google Scholar

28. Ganz T. Does pathological iron overload impair the function of human lungs? EBioMedicine. (2017) 20:13–4. doi: 10.1016/j.ebiom.2017.05.023

PubMed Abstract | Crossref Full Text | Google Scholar

29. Ou R, Aodeng G, and Ai J. Advancements in the application of the fenton reaction in the cancer microenvironment. Pharmaceutics. (2023) 15:2337. doi: 10.3390/pharmaceutics15092337

PubMed Abstract | Crossref Full Text | Google Scholar

30. Abe C, Miyazawa T, and Miyazawa T. Current use of fenton reaction in drugs and food. Molecules. (2022) 27:5451. doi: 10.3390/molecules27175451

PubMed Abstract | Crossref Full Text | Google Scholar

31. Yang N, Zhang T, Cao C, Mao G, Shao J, Song X, et al. BSA stabilized photothermal-fenton reactor with cisplatin for chemo/chemodynamic cascade oncotherapy. Nano Res. (2022) 15:2235–43. doi: 10.1007/s12274-021-3758-4

Crossref Full Text | Google Scholar

32. Jiang X, Stockwell BR, and Conrad M. Ferroptosis: mechanisms, biology and role in disease. Nat Rev Mol Cell Biol. (2021) 22:266–82. doi: 10.1038/s41580-020-00324-8

PubMed Abstract | Crossref Full Text | Google Scholar

33. Kagan VE, Mao G, Qu F, Angeli JP, Doll S, Croix CS, et al. Oxidized arachidonic and adrenic PEs navigate cells to ferroptosis. Nat Chem Biol. (2017) 13:81–90. doi: 10.1038/nchembio.2238

PubMed Abstract | Crossref Full Text | Google Scholar

34. Gaschler MM, Andia AA, Liu H, Csuka JM, Hurlocker B, Vaiana CA, et al. FINO2 initiates ferroptosis through GPX4 inactivation and iron oxidation. Nat Chem Biol. (2018) 14:507–15. doi: 10.1038/s41589-018-0031-6

PubMed Abstract | Crossref Full Text | Google Scholar

35. Stockwell BR, Friedmann Angeli JP, Bayir H, Bush AI, Conrad M, Dixon SJ, et al. Ferroptosis: A regulated cell death nexus linking metabolism, redox biology, and disease. Cell. (2017) 171:273–85. doi: 10.1016/j.cell.2017.09.021

PubMed Abstract | Crossref Full Text | Google Scholar

36. Gao M, Yi J, Zhu J, Minikes AM, Monian P, Thompson CB, et al. Role of mitochondria in ferroptosis. Mol Cell. (2019) 73:354–363.e3. doi: 10.1016/j.molcel.2018.10.042

PubMed Abstract | Crossref Full Text | Google Scholar

37. Dixon SJ, Lemberg KM, Lamprecht MR, Skouta R, Zaitsev EM, Gleason CE, et al. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell. (2012) 149:1060–72. doi: 10.1016/j.cell.2012.03.042

PubMed Abstract | Crossref Full Text | Google Scholar

38. Ingold I, Berndt C, Schmitt S, Doll S, Poschmann G, Buday K, et al. Selenium utilization by GPX4 is required to prevent hydroperoxide-induced ferroptosis. Cell. (2018) 172:409–422.e21. doi: 10.1016/j.cell.2017.11.048

PubMed Abstract | Crossref Full Text | Google Scholar

39. Maiorino M, Conrad M, and Ursini F. GPx4, lipid peroxidation, and cell death: discoveries, rediscoveries, and open issues. Antioxid Redox Signal. (2018) 29:61–74. doi: 10.1089/ars.2017.7115

PubMed Abstract | Crossref Full Text | Google Scholar

40. Fang X, Wang H, Han D, Xie E, Yang X, Wei J, et al. Ferroptosis as a target for protection against cardiomyopathy. Proc Natl Acad Sci U.S.A. (2019) 116:2672–80. doi: 10.1073/pnas.1821022116

PubMed Abstract | Crossref Full Text | Google Scholar

41. Yu Y, Yan Y, Niu F, Wang Y, Chen X, Su G, et al. Ferroptosis: a cell death connecting oxidative stress, inflammation and cardiovascular diseases. Cell Death Discov. (2021) 7:193. doi: 10.1038/s41420-021-00579-w

PubMed Abstract | Crossref Full Text | Google Scholar

42. Lv H, Zhen C, Liu J, Yang P, Hu L, and Shang P. Unraveling the potential role of glutathione in multiple forms of cell death in cancer therapy. Oxid Med Cell Longev. (2019) 2019:3150145. doi: 10.1155/2019/3150145

PubMed Abstract | Crossref Full Text | Google Scholar

43. Bai L, Yan F, Deng R, Gu R, Zhang X, and Bai J. Thioredoxin-1 rescues MPP+/MPTP-induced ferroptosis by increasing glutathione peroxidase 4. Mol Neurobiol. (2021) 58:3187–97. doi: 10.1007/s12035-021-02320-1

PubMed Abstract | Crossref Full Text | Google Scholar

44. Feng H and Stockwell BR. Unsolved mysteries: How does lipid peroxidation cause ferroptosis? PloS Biol. (2018) 16:e2006203. doi: 10.1371/journal.pbio.2006203

PubMed Abstract | Crossref Full Text | Google Scholar

45. Song X and Long D. Nrf2 and ferroptosis: A new research direction for neurodegenerative diseases. Front Neurosci. (2020) 14:267. doi: 10.3389/fnins.2020.00267

PubMed Abstract | Crossref Full Text | Google Scholar

46. Medoro A, Saso L, Scapagnini G, and Davinelli S. NRF2 signaling pathway and telomere length in aging and age-related diseases. Mol Cell Biochem. (2024) 479:2597–613. doi: 10.1007/s11010-023-04878-x

PubMed Abstract | Crossref Full Text | Google Scholar

47. Medoro A, Jafar TH, Sallustio F, Scapagnini G, Saso L, and Davinelli S. Computational assessment of carotenoids as keap1-nrf2 protein-protein interaction inhibitors: implications for antioxidant strategies. Biochem (Mosc). (2024) 89:1681–91. doi: 10.1134/S0006297924100031

PubMed Abstract | Crossref Full Text | Google Scholar

48. Davinelli S, Medoro A, Siracusano M, Savino R, Saso L, Scapagnini G, et al. Oxidative stress response and NRF2 signaling pathway in autism spectrum disorder. Redox Biol. (2025) 83:103661. doi: 10.1016/j.redox.2025.103661

PubMed Abstract | Crossref Full Text | Google Scholar

49. Kang YP, Mockabee-Macias A, Jiang C, Falzone A, Prieto-Farigua N, Stone E, et al. Non-canonical glutamate-cysteine ligase activity protects against ferroptosis. Cell Metab. (2021) 33:174–189.e7. doi: 10.1016/j.cmet.2020.12.007

PubMed Abstract | Crossref Full Text | Google Scholar

50. Toyokuni S, Ito F, Yamashita K, Okazaki Y, and Akatsuka S. Iron and thiol redox signaling in cancer: An exquisite balance to escape ferroptosis. Free Radic Biol Med. (2017) 108:610–26. doi: 10.1016/j.freeradbiomed.2017.04.024

PubMed Abstract | Crossref Full Text | Google Scholar

51. Lei P, Bai T, and Sun Y. Mechanisms of ferroptosis and relations with regulated cell death: A review. Front Physiol. (2019) 10:139. doi: 10.3389/fphys.2019.00139

PubMed Abstract | Crossref Full Text | Google Scholar

52. Yu C and Xiao JH. The keap1-nrf2 system: A mediator between oxidative stress and aging. Oxid Med Cell Longev. (2021) 2021:6635460. doi: 10.1155/2021/6635460

PubMed Abstract | Crossref Full Text | Google Scholar

53. Shan C, Wang Y, and Wang Y. The crosstalk between autophagy and nrf2 signaling in cancer: from biology to clinical applications. Int J Biol Sci. (2024) 20:6181–206. doi: 10.7150/ijbs.103187

PubMed Abstract | Crossref Full Text | Google Scholar

54. Towers CG, Fitzwalter BE, Regan D, Goodspeed A, Morgan MJ, Liu CW, et al. Cancer cells upregulate NRF2 signaling to adapt to autophagy inhibition. Dev Cell. (2019) 50:690–703.e6. doi: 10.1016/j.devcel.2019.07.010

PubMed Abstract | Crossref Full Text | Google Scholar

55. Ning B, Hang S, Zhang W, Mao C, and Li D. An update on the bridging factors connecting autophagy and Nrf2 antioxidant pathway. Front Cell Dev Biol. (2023) 11:1232241. doi: 10.3389/fcell.2023.1232241

PubMed Abstract | Crossref Full Text | Google Scholar

56. Manz DH, Blanchette NL, Paul BT, Torti FM, and Torti SV. Iron and cancer: recent insights. Ann N Y Acad Sci. (2016) 1368:149–61. doi: 10.1111/nyas.13008

PubMed Abstract | Crossref Full Text | Google Scholar

57. Hassannia B, Vandenabeele P, and Vanden Berghe T. Targeting ferroptosis to iron out cancer. Cancer Cell. (2019) 35:830–49. doi: 10.1016/j.ccell.2019.04.002

PubMed Abstract | Crossref Full Text | Google Scholar

58. Galluzzi L, Vitale I, Aaronson SA, Abrams JM, Adam D, Agostinis P, et al. Molecular mechanisms of cell death: recommendations of the Nomenclature Committee on Cell Death 2018. Cell Death Differ. (2018) 25:486–541. doi: 10.1038/s41418-017-0012-4

PubMed Abstract | Crossref Full Text | Google Scholar

59. Zhao X, Li X, and Xu Y. Ferroptosis: a dual-edged sword in tumour growth. Front Pharmacol. (2024) 14:1330910. doi: 10.3389/fphar.2023.1330910

PubMed Abstract | Crossref Full Text | Google Scholar

60. Li Z, Chen L, Chen C, Zhou Y, Hu D, Yang J, et al. Targeting ferroptosis in breast cancer. biomark Res. (2020) 8:58. doi: 10.1186/s40364-020-00230-3

PubMed Abstract | Crossref Full Text | Google Scholar

61. Xing N, Du Q, Guo S, Xiang G, Zhang Y, Meng X, et al. Ferroptosis in lung cancer: a novel pathway regulating cell death and a promising target for drug therapy. Cell Death Discov. (2023) 9:110. doi: 10.1038/s41420-023-01407-z

PubMed Abstract | Crossref Full Text | Google Scholar

62. Feng J, Read OJ, and Dinkova-Kostova AT. Nrf2 in TIME: the emerging role of nuclear factor erythroid 2-related factor 2 in the tumor immune microenvironment. Mol Cells. (2023) 46:142–52. doi: 10.14348/molcells.2023.2183

PubMed Abstract | Crossref Full Text | Google Scholar

63. Bianchi ME. DAMPs, PAMPs and alarmins: all we need to know about danger. J Leukoc Biol. (2007) 81:1–5. doi: 10.1189/jlb.0306164

PubMed Abstract | Crossref Full Text | Google Scholar

64. Liu J, Kang R, and Tang D. The art of war: ferroptosis and pancreatic cancer. Front Pharmacol. (2021) 12:773909. doi: 10.3389/fphar.2021.773909

PubMed Abstract | Crossref Full Text | Google Scholar

65. Yang BC and Leung PS. Irisin is a positive regulator for ferroptosis in pancreatic cancer. Mol Ther Oncolytics. (2020) 18:457–66. doi: 10.1016/j.omto.2020.08.002

PubMed Abstract | Crossref Full Text | Google Scholar

66. Chen X, Kang R, Kroemer G, and Tang D. Targeting ferroptosis in pancreatic cancer: a double-edged sword. Trends Cancer. (2021) 7:891–901. doi: 10.1016/j.trecan.2021.04.005

PubMed Abstract | Crossref Full Text | Google Scholar

67. Jiang Y, Yu Y, Pan Z, Glandorff C, and Sun M. Ferroptosis: a new hunter of hepatocellular carcinoma. Cell Death Discov. (2024) 10:136. doi: 10.1038/s41420-024-01863-1

PubMed Abstract | Crossref Full Text | Google Scholar

68. Pan F, Lin X, Hao L, Wang T, Song H, and Wang R. The critical role of ferroptosis in hepatocellular carcinoma. Front Cell Dev Biol. (2022) 10:882571. doi: 10.3389/fcell.2022.882571

PubMed Abstract | Crossref Full Text | Google Scholar

69. Xu T, Ding W, Ji X, Ao X, Liu Y, Yu W, et al. Molecular mechanisms of ferroptosis and its role in cancer therapy. J Cell Mol Med. (2019) 23:4900–12. doi: 10.1111/jcmm.14511

PubMed Abstract | Crossref Full Text | Google Scholar

70. Guo L, Hu C, Yao M, and Han G. Mechanism of sorafenib resistance associated with ferroptosis in HCC. Front Pharmacol. (2023) 14:1207496. doi: 10.3389/fphar.2023.1207496

PubMed Abstract | Crossref Full Text | Google Scholar

71. Li J, Cao F, Yin HL, Huang ZJ, Lin ZT, Mao N, et al. Ferroptosis: past, present and future. Cell Death Dis. (2020) 11:88. doi: 10.1038/s41419-020-2298-2

PubMed Abstract | Crossref Full Text | Google Scholar

72. Sui S, Xu S, and Pang D. Emerging role of ferroptosis in breast cancer: New dawn for overcoming tumor progression. Pharmacol Ther. (2022) 232:107992. doi: 10.1016/j.pharmthera.2021.107992

PubMed Abstract | Crossref Full Text | Google Scholar

73. Chen MS, Wang SF, Hsu CY, Yin PH, Yeh TS, Lee HC, et al. CHAC1 degradation of glutathione enhances cystine-starvation-induced necroptosis and ferroptosis in human triple negative breast cancer cells via the GCN2-eIF2α-ATF4 pathway. Oncotarget. (2017) 8:114588–602. doi: 10.18632/oncotarget.23055

PubMed Abstract | Crossref Full Text | Google Scholar

74. Liu Y, Hu Y, Jiang Y, Bu J, and Gu X. Targeting ferroptosis, the achilles’ heel of breast cancer: A review. Front Pharmacol. (2022) 13:1036140. doi: 10.3389/fphar.2022.1036140

PubMed Abstract | Crossref Full Text | Google Scholar

75. Luo L and Xu G. Fascaplysin induces apoptosis and ferroptosis, and enhances anti-PD-1 immunotherapy in non-small cell lung cancer (NSCLC) by promoting PD-L1 expression. Int J Mol Sci. (2022) 23:13774. doi: 10.3390/ijms232213774

PubMed Abstract | Crossref Full Text | Google Scholar

76. Zhou J, Zhang L, Yan J, Hou A, Sui W, and Sun M. Curcumin induces ferroptosis in A549 CD133+ Cells through the GSH-GPX4 and FSP1-coQ10-NAPH pathways. Discov Med. (2023) 35:251–63. doi: 10.24976/Discov.Med.202335176.26

PubMed Abstract | Crossref Full Text | Google Scholar

77. Schneider JL, Rowe JH, Garcia-de-Alba C, Kim CF, Sharpe AH, and Haigis MC. The aging lung: Physiology, disease, and immunity. Cell. (2021) 184:1990–2019. doi: 10.1016/j.cell.2021.03.005

PubMed Abstract | Crossref Full Text | Google Scholar

78. Bersuker K, Hendricks JM, Li Z, Magtanong L, Ford B, Tang PH, et al. The CoQ oxidoreductase FSP1 acts parallel to GPX4 to inhibit ferroptosis. Nature. (2019) 575:688–92. doi: 10.1038/s41586-019-1705-2

PubMed Abstract | Crossref Full Text | Google Scholar

79. Wang H, Cheng Y, Mao C, Liu S, Xiao D, Huang J, et al. Emerging mechanisms and targeted therapy of ferroptosis in cancer. Mol Ther. (2021) 29:2185–208. doi: 10.1016/j.ymthe.2021.03.022

PubMed Abstract | Crossref Full Text | Google Scholar

80. Hecker L. Mechanisms and consequences of oxidative stress in lung disease: therapeutic implications for an aging populace. Am J Physiol Lung Cell Mol Physiol. (2018) 314:L642–53. doi: 10.1152/ajplung.00275.2017

PubMed Abstract | Crossref Full Text | Google Scholar

81. Wu Z, Zhang Y, Zhong W, Wu K, Zhong T, and Jiang T. Targeting ferroptosis: a promising approach for treating lung carcinoma. Cell Death Discov. (2025) 11:33. doi: 10.1038/s41420-025-02308-z

PubMed Abstract | Crossref Full Text | Google Scholar

82. Gu R, Xia Y, Li P, Zou D, Lu K, Ren L, et al. Ferroptosis and its role in gastric cancer. Front Cell Dev Biol. (2022) 10:860344. doi: 10.3389/fcell.2022.860344

PubMed Abstract | Crossref Full Text | Google Scholar

83. Hagymási K and Tulassay Z. Helicobacter pylori infection: new pathogenetic and clinical aspects. World J Gastroenterol. (2014) 20:6386–99. doi: 10.3748/wjg.v20.i21.6386

PubMed Abstract | Crossref Full Text | Google Scholar

84. Durazzo M, Adriani A, Fagoonee S, Saracco GM, and Pellicano R. Helicobacter pylori and respiratory diseases: 2021 update. Microorganisms. (2021) 9:2033. doi: 10.3390/microorganisms9102033

PubMed Abstract | Crossref Full Text | Google Scholar

85. Janssen HJB, Fransen LFC, Ponten JEH, Nieuwenhuijzen GAP, and Luyer MDP. Micronutrient deficiencies following minimally invasive esophagectomy for cancer. Nutrients. (2020) 12:778. doi: 10.3390/nu12030778

PubMed Abstract | Crossref Full Text | Google Scholar

86. Menegon S, Columbano A, and Giordano S. The dual roles of NRF2 in cancer. Trends Mol Med. (2016) 22:578–93. doi: 10.1016/j.molmed.2016.05.002

PubMed Abstract | Crossref Full Text | Google Scholar

87. Padmanabhan B, Tong KI, Ohta T, Nakamura Y, Scharlock M, Ohtsuji M, et al. Structural basis for defects of Keap1 activity provoked by its point mutations in lung cancer. Mol Cell. (2006) 21:689–700. doi: 10.1016/j.molcel.2006.01.013

PubMed Abstract | Crossref Full Text | Google Scholar

88. Singh A, Misra V, Thimmulappa RK, Lee H, Ames S, Hoque MO, et al. Dysfunctional KEAP1-NRF2 interaction in non-small-cell lung cancer. PloS Med. (2006) 3:e420. doi: 10.1371/journal.pmed.0030420

PubMed Abstract | Crossref Full Text | Google Scholar

89. Tao S, Rojo de la Vega M, Chapman E, Ooi A, and Zhang DD. The effects of NRF2 modulation on the initiation and progression of chemically and genetically induced lung cancer. Mol Carcinog. (2018) 57:182–92. doi: 10.1002/mc.22745

PubMed Abstract | Crossref Full Text | Google Scholar

90. Fouzder C, Mukhuty A, Mukherjee S, Malick C, and Kundu R. Trigonelline inhibits Nrf2 via EGFR signalling pathway and augments efficacy of Cisplatin and Etoposide in NSCLC cells. Toxicol In Vitro. (2021) 70:105038. doi: 10.1016/j.tiv.2020.105038

PubMed Abstract | Crossref Full Text | Google Scholar

91. Zhuang J, Liu X, Yang Y, Zhang Y, and Guan G. Sulfasalazine, a potent suppressor of gastric cancer proliferation and metastasis by inhibition of xCT: Conventional drug in new use. J Cell Mol Med. (2021) 25:5372–80. doi: 10.1111/jcmm.16548

PubMed Abstract | Crossref Full Text | Google Scholar

92. Deng C, Xiong L, Chen Y, Wu K, and Wu J. Metformin induces ferroptosis through the Nrf2/HO-1 signaling in lung cancer. BMC Pulm Med. (2023) 23:360. doi: 10.1186/s12890-023-02655-6

PubMed Abstract | Crossref Full Text | Google Scholar

93. Feng S, Li Y, Huang H, Huang H, Duan Y, Yuan Z, et al. Isoorientin reverses lung cancer drug resistance by promoting ferroptosis via the SIRT6/Nrf2/GPX4 signaling pathway. Eur J Pharmacol. (2023) 954:175853. doi: 10.1016/j.ejphar.2023.175853

PubMed Abstract | Crossref Full Text | Google Scholar

94. Liu X, Peng X, Cen S, Yang C, Ma Z, and Shi X. Wogonin induces ferroptosis in pancreatic cancer cells by inhibiting the Nrf2/GPX4 axis. Front Pharmacol. (2023) 14:1129662. doi: 10.3389/fphar.2023.1129662

PubMed Abstract | Crossref Full Text | Google Scholar

95. Gao AM, Zhang XY, and Ke ZP. Apigenin sensitizes BEL-7402/ADM cells to doxorubicin through inhibiting miR-101/Nrf2 pathway. Oncotarget. (2017) 8:82085–91. doi: 10.18632/oncotarget.18294

PubMed Abstract | Crossref Full Text | Google Scholar

96. Xiao X, Lu Z, Lin V, May A, Shaw DH, Wang Z, et al. MicroRNA miR-24-3p reduces apoptosis and regulates keap1-nrf2 pathway in mouse cardiomyocytes responding to ischemia/reperfusion injury. Oxid Med Cell Longev. (2018) 2018:7042105. doi: 10.1155/2018/7042105

PubMed Abstract | Crossref Full Text | Google Scholar

97. Li Y, Jia Y, Xu Y, and Li K. DMF activates NRF2 to inhibit the pro-invasion ability of TAMs in breast cancer. Front Oncol. (2021) 11:706448. doi: 10.3389/fonc.2021.706448

PubMed Abstract | Crossref Full Text | Google Scholar

98. Wei R, Zhao Y, Wang J, Yang X, Li S, Wang Y, et al. Tagitinin C induces ferroptosis through PERK-Nrf2-HO-1 signaling pathway in colorectal cancer cells. Int J Biol Sci. (2021) 17:2703–17. doi: 10.7150/ijbs.59404

PubMed Abstract | Crossref Full Text | Google Scholar

99. Hu Q, Zuo T, Deng L, Chen S, Yu W, Liu S, et al. β-Caryophyllene suppresses ferroptosis induced by cerebral ischemia reperfusion via activation of the NRF2/HO-1 signaling pathway in MCAO/R rats. Phytomedicine. (2022) 102:154112. doi: 10.1016/j.phymed.2022.154112

PubMed Abstract | Crossref Full Text | Google Scholar

100. Jing S, Lu Y, Zhang J, Ren Y, Mo Y, Liu D, et al. Levistilide a induces ferroptosis by activating the nrf2/HO-1 signaling pathway in breast cancer cells. Drug Des Devel Ther. (2022) 16:2981–93. doi: 10.2147/DDDT.S374328

PubMed Abstract | Crossref Full Text | Google Scholar

101. Mukherjee S, Shelar B, and Krishna S. Versatile role of miR-24/24-1*/24-2* expression in cancer and other human diseases. Am J Transl Res. (2022) 14:20–54.

PubMed Abstract | Google Scholar

102. Dong J, Che J, Wu Y, Deng Y, Jiang X, He Z, et al. Dexmedetomidine promotes colorectal cancer progression via Piwil2 signaling. Cell Oncol (Dordr). (2024) 47:1459–74. doi: 10.1007/s13402-024-00944-8

PubMed Abstract | Crossref Full Text | Google Scholar

103. Mohammad SIS, Vasudevan A, Enwa FO, Bansal J, Chahar M, Eldesoqui M, et al. The Sirt1/Nrf2 pathway is a key factor for drug therapy in chemotherapy-induced cardiotoxicity: a Mini-Review. Med Oncol. (2024) 41:244. doi: 10.1007/s12032-024-02494-3

PubMed Abstract | Crossref Full Text | Google Scholar

104. Pane C, Marra AM, Aliberti L, Campanile M, Coscetta F, Crisci G, et al. Rationale and protocol of a double-blind, randomized, placebo-controlled trial to test the efficacy, safety, and tolerability of dimethyl fumarate in Friedreich Ataxia (DMF-FA-201). Front Neurosci. (2023) 17:1260977. doi: 10.3389/fnins.2023.1260977

PubMed Abstract | Crossref Full Text | Google Scholar

105. Pilotto F, Chellapandi DM, and Puccio H. Omaveloxolone: a groundbreaking milestone as the first FDA-approved drug for Friedreich ataxia. Trends Mol Med. (2024) 30:117–25. doi: 10.1016/j.molmed.2023.12.002

PubMed Abstract | Crossref Full Text | Google Scholar

106. Rojo de la Vega M, Dodson M, Chapman E, and Zhang DD. NRF2-targeted therapeutics: New targets and modes of NRF2 regulation. Curr Opin Toxicol. (2016) 1:62–70. doi: 10.1016/j.cotox.2016.10.005

PubMed Abstract | Crossref Full Text | Google Scholar

107. Tebay LE, Robertson H, Durant ST, Vitale SR, Penning TM, Dinkova-Kostova AT, et al. Mechanisms of activation of the transcription factor Nrf2 by redox stressors, nutrient cues, and energy status and the pathways through which it attenuates degenerative disease. Free Radic Biol Med. (2015) 88:108–46. doi: 10.1016/j.freeradbiomed.2015.06.021

PubMed Abstract | Crossref Full Text | Google Scholar

108. Rojo de la Vega M, Chapman E, and Zhang DD. NRF2 and the hallmarks of cancer. Cancer Cell. (2018) 34:21–43. doi: 10.1016/j.ccell.2018.03.022

PubMed Abstract | Crossref Full Text | Google Scholar

Keywords: Nrf2, Nrf2 modulators, lipid peroxidation, ferroptosis, oxidative stress, cancer

Citation: Tunc R, Ates I, Yılmaz B, Medoro A, Davinelli S, Suzen S and Saso L (2025) Modulating Nrf2 to control lipid peroxidation and ferroptosis: implications for cancer management. Front. Oncol. 15:1701249. doi: 10.3389/fonc.2025.1701249

Received: 08 September 2025; Accepted: 20 November 2025; Revised: 13 November 2025;
Published: 03 December 2025.

Edited by:

Ashish Tyagi, Texas A and M University, United States

Reviewed by:

Matteo Borgini, Wellstar-Medical College of Georgia Health, United States
Xiangfei Xue, Shanghai Jiao Tong University, China

Copyright © 2025 Tunc, Ates, Yılmaz, Medoro, Davinelli, Suzen and Saso. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Ilker Ates, aWxrZXJhdGVzOTc2QGdtYWlsLmNvbQ==

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.