Next Article in Journal
Staurosporine as an Antifungal Agent
Previous Article in Journal
Glia Cells Are Selectively Sensitive to Nanosized Titanium Dioxide Mineral Forms
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Metabolic Regulation of Ferroptosis in Breast Cancer

by
Natalija Glibetic
1,2,3 and
Michael Weichhaus
3,*
1
The IDeA Networks of Biomedical Research Excellence (INBRE) Program, School of Natural Sciences and Mathematics, Chaminade University, Honolulu, HI 96816, USA
2
Department of Cell and Molecular Biology, John A Burns School of Medicine, University of Hawaii, 685 Ilalo St, Honolulu, HI 96813, USA
3
Laboratory of Molecular Cancer Research, School of Natural Sciences and Mathematics, Chaminade University of Honolulu, Honolulu, HI 96816, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(19), 9686; https://doi.org/10.3390/ijms26199686
Submission received: 7 September 2025 / Revised: 29 September 2025 / Accepted: 3 October 2025 / Published: 4 October 2025

Abstract

Breast cancer, a leading global malignancy, exhibits extensive metabolic reprogramming that drives tumorigenesis, therapy resistance, and survival. Ferroptosis, an iron-dependent regulated cell death mechanism characterized by lipid peroxidation, emerges as a promising therapeutic vulnerability, particularly in aggressive subtypes like triple-negative breast cancer (TNBC). This literature review comprehensively explores the metabolic regulation of ferroptosis in breast cancer cells, focusing on how dysregulated pathways modulate sensitivity or resistance. The review will discuss iron homeostasis, including upregulated transferrin receptor 1 (TFR1), diminished ferroportin, mitochondrial dynamics, and ferritinophagy, which catalyze ROS via Fenton reactions. It will examine glutathione (GSH) metabolism through the GPX4-GSH axis, with subtype-specific reliance on cystine import via xCT or de novo cysteine synthesis. Lipid metabolism will be analyzed as the core battleground, highlighting polyunsaturated fatty acid (PUFA) incorporation by ACSL4 promoting peroxidation, contrasted with monounsaturated fatty acid (MUFA) protection via SCD1, alongside subtype adaptations. Further, the review will address tumor microenvironment influences, such as cysteine supply from cancer-associated fibroblasts and oleic acid from adipocytes. Oncogenic signaling (e.g., RAS, mTOR) and tumor suppressors (e.g., p53) will be evaluated for their roles in resistance or sensitivity. Intersections with glucose metabolism (Warburg effect) and selenium-dependent antioxidants will be explored. Therapeutically, the review will consider targeting these nodes with GPX4 inhibitors or iron overload, synergized with immunotherapy for immunogenic cell death. Future directions will emphasize multi-omics integration and patient-derived organoids to uncover subtype-specific strategies for precision medicine in breast cancer.

1. Introduction

Breast cancer remains a leading cause of cancer-related mortality worldwide, with rising incidence rates despite therapeutic advances [1]. Ferroptosis, an iron-dependent form of regulated cell death characterized by lipid peroxidation, was first described by Stockwell and colleagues in 2012 as a distinct mechanism from apoptosis, necrosis, or autophagy [2]. This cell death pathway has emerged as a promising therapeutic vulnerability in breast cancer, particularly in aggressive subtypes like triple-negative breast cancer (TNBC) that frequently develop resistance to conventional therapies.
While the core mechanisms of ferroptosis—iron accumulation, lipid peroxidation, and antioxidant system failure—are established, critical knowledge gaps remain regarding how breast cancer cells metabolically reprogram to evade ferroptotic death. Specifically, the field lacks comprehensive understanding of (1) how distinct breast cancer subtypes differentially regulate ferroptosis through metabolic adaptations, (2) the role of the tumor microenvironment in modulating ferroptosis sensitivity through metabolic crosstalk, (3) how oncogenic signaling pathways intersect with ferroptosis regulation to confer therapeutic resistance, and (4) which metabolic nodes represent actionable therapeutic targets for ferroptosis induction.
Recent evidence suggests that breast cancer cells exploit multiple metabolic pathways to maintain ferroptosis resistance, including altered iron homeostasis, enhanced glutathione synthesis, lipid metabolism reprogramming, and metabolic support from the tumor microenvironment (Figure 1). However, these adaptations vary significantly across breast cancer subtypes and remain incompletely characterized. For instance, while TNBC shows high baseline ferroptosis sensitivity, certain TNBC subtypes paradoxically upregulate ferroptosis-promoting factors like ACSL4, suggesting complex regulatory mechanisms that balance survival advantages against vulnerability to oxidative death.
This review addresses these knowledge gaps by providing a comprehensive analysis of the metabolic networks governing ferroptosis in breast cancer. We systematically examine iron metabolism, glutathione and amino acid regulation, lipid metabolism alterations, and the unique metabolic reprogramming of breast cancer subtypes. Furthermore, we explore how the breast tumor microenvironment creates ferroptosis-resistant niches through metabolic crosstalk and evaluate emerging therapeutic strategies that exploit these metabolic vulnerabilities. By integrating recent discoveries in ferroptosis biology with breast cancer metabolism, this review aims to identify actionable metabolic targets and inform precision medicine approaches for ferroptosis-based therapies.

2. Iron Metabolism: The Catalytic Foundation

2.1. Iron Uptake and Regulation

Iron metabolism drives ferroptosis through the labile iron pool (LIP), which catalyzes Fenton reactions generating hydroxyl radicals. Transferrin receptor 1 (TFR1), frequently overexpressed in breast cancer, shows a clear progression from benign to invasive carcinomas [3], with particularly high expression in high-grade and estrogen receptor (ER)-negative tumors [3,4]. This overexpression correlates with poor outcomes and tamoxifen resistance in ER+ patients [4], though the mechanistic link between TFR1 levels and therapy resistance remains incompletely understood.
Beyond TFR1, breast cancer cells employ multiple iron acquisition strategies. Divalent metal transporter 1 (DMT1) mediates non-transferrin-bound iron uptake, particularly in TNBC [5], while loss of ferroportin—the only iron exporter—creates intracellular accumulation [6]. The tumor microenvironment further modulates iron availability through interleukin-6 (IL-6) paracrine signaling, contributing to chemoresistance [7], though whether this is cause or consequence of treatment failure requires clarification. Alterations in endosomal pH can impair iron release from transferrin, disrupting homeostasis and favoring progression [8].
While TFR1 upregulation is consistently observed across breast cancer subtypes (strong evidence from >20 studies), the functional significance remains subject to debate. The paradox that cancer cells accumulate iron despite its pro-ferroptotic effects suggests that either (1) iron provides selective advantages outweighing ferroptosis risk, (2) concurrent antioxidant upregulation neutralizes iron toxicity, or (3) iron accumulation is a passenger alteration. The failure of iron chelators as monotherapy [6] supports the second hypothesis, indicating that targeting iron alone is insufficient without disrupting compensatory mechanisms.

2.2. Mitochondrial Iron Dynamics

Mitochondria play a crucial role in ferroptosis regulation as the primary site of iron utilization and ROS generation. Cancer cells accumulate iron in mitochondria, increasing their susceptibility to oxidative stress and ferroptotic death [9,10]. In TNBC specifically, dysregulated mitochondrial iron transport enhances ferroptosis vulnerability under metabolic stress [11]. CDGSH iron sulfur domain 1 (CISD1) emerges as a key protective factor—its knockdown significantly increases mitochondrial ferrous iron and promotes ferroptotic death [12].
While mitochondrial iron accumulation is documented across breast cancer types, the directionality remains unclear. Does increased mitochondrial iron drive transformation or result from it? The tissue-specific role of CISD1 is particularly puzzling, as its protective function varies dramatically between cell lines [12]. This variability suggests that mitochondrial iron’s contribution to ferroptosis may be context-dependent rather than universally applicable, limiting therapeutic targeting strategies.

2.3. Iron Storage and Release

The balance between iron storage and release critically determines ferroptosis susceptibility. Nuclear receptor coactivator 4 (NCOA4)-mediated ferritinophagy releases iron from ferritin storage, increasing the LIP and sensitizing cells to ferroptosis [13]. HERC2 downregulation enhances this process by stabilizing NCOA4 [14]. Breast cancer cells exploit this system—while maintaining iron storage for survival, they become vulnerable to forced ferritinophagy. Sorafenib-loaded nanoparticles effectively trigger this vulnerability, upregulating NCOA4 to induce ferroptosis while boosting immune responses [15].
Recent evidence shifts focus to lysosomal iron stores as the primary ferroptosis initiator, with lipid peroxidation beginning in lysosomes before propagating to other organelles [16]. This compartmentalized view of iron-induced death challenges the traditional cytoplasmic-centric model of ferroptosis.
The ferritinophagy pathway presents a therapeutic paradox in that cancer cells require NCOA4-mediated iron release for proliferation, yet this same pathway sensitizes them to ferroptosis. The recent lysosomal iron discovery [16] fundamentally challenges this understanding, suggesting that targeting lysosomal rather than cytoplasmic iron may be more effective. However, the clinical translation remains limited by our inability to selectively induce ferritinophagy in cancer versus normal cells, explaining why ferritinophagy-targeting agents remain in preclinical stages.

3. Glutathione Metabolism and Amino Acid Regulation

3.1. The Central GPX4-GSH Axis

Glutathione (GSH) serves as the cornerstone of ferroptosis regulation through its role as a cofactor for glutathione peroxidase 4 (GPX4), which converts lipid peroxides into non-toxic lipid alcohols [17,18]. When GSH is depleted, GPX4 becomes inactivated, leading to lethal lipid peroxide accumulation [19].
The xCT/GPX4 axis becomes upregulated in TNBC, with cells developing an “addiction” to this cystine import-glutathione defense system [20]. TNBC cells use the MUC1-C/xCT/CD44v complex to maintain glutathione levels and resist ferroptosis [21]. Mitochondria-targeted strategies show that iron overload combined with GSH depletion disrupts redox homeostasis and promotes ferroptosis in TNBC cells [11].
The GPX4-GSH axis represents the best-validated ferroptosis mechanism, with consistent evidence across >50 breast cancer studies. However, the clinical translation faces two major obstacles: (1) GPX4 inhibitors like RSL3 show poor bioavailability (t½ < 2 h) and hepatotoxicity at therapeutic doses, and (2) the “addiction” to xCT/GPX4 in TNBC paradoxically coexists with high baseline ferroptosis sensitivity. This suggests compensatory mechanisms we don’t fully understand, potentially explaining why GPX4-targeting monotherapies have yet to enter clinical trials for breast cancer.

3.2. Cysteine Transport and Synthesis

Cysteine availability rate-limits GSH synthesis through two distinct pathways. The system xc transporter (SLC7A11/xCT) imports cystine, which is reduced to cysteine for GSH synthesis [22]. In breast cancer, xCT upregulation reflects dependency on extracellular cysteine [23]. Inhibition with erastin or imidazole ketone erastin (IKE) depletes GSH and induces ferroptosis [22,24]. Tumor-associated macrophages (TAMs) in TNBC secrete TGFβ1, enhancing gamma-glutamyltransferase 1 (GGT1) transcription to increase cysteine availability and suppress ferroptosis [25]. Luminal A cancers specifically depend on xCT to protect against CDK4/6 inhibitor-induced ferroptosis [26].
Alternatively, the transsulfuration pathway synthesizes cysteine de novo through cystathionine beta-synthase (CBS) and cystathionine gamma-lyase (CTH) [27]. This pathway becomes critical when extracellular cysteine is limited [28]. Basal-like breast cancers exhibit high CBS activity, creating “cysteine addiction” that confers ferroptosis resistance—silencing CBS sensitizes these cells to death [29]. Low cytokine-like 1 (CYTL1) levels suppress CBS, increasing ferroptosis risk when cystine import is blocked [30].
Beyond import and synthesis, lysosomal cysteine storage provides a third ferroptosis resistance mechanism. Lysosomal cystine shortage specifically triggers ferroptosis through ATF4-mediated stress responses [31]. Breast cancer cells store cysteine via the MFSD12 transporter and release it through cystinosin (CTNS) to maintain GSH levels [32]. Enhancing lysosomal cystine with synthetic mRNA (CysRx) paradoxically increases ferroptosis and suppresses tumor growth in vivo [31], suggesting that disrupting lysosomal cysteine homeostasis represents a therapeutic vulnerability.
A clear subtype-specific dichotomy emerges: TNBC and drug-resistant subtypes primarily rely on CBS-driven transsulfuration, while luminal A and HER2+ subtypes depend on xCT-mediated import. This differential dependency provides precision targeting opportunities, yet clinical exploitation remains elusive. The paradox that both pathways can be simultaneously upregulated in some cancers suggests redundancy that may limit single-pathway targeting. Moreover, the TAM-mediated cysteine supply [22] indicates that tumor microenvironment contributions may override cancer cell-autonomous vulnerabilities. The lysosomal cysteine compartment adds yet another layer of redundancy [31,32], potentially explaining the limited success of targeting cytoplasmic cysteine pathways alone.

3.3. Alternative Cysteine Actions

Cysteine and homocysteine can act as alternative thiol donors for GPX4, providing GSH-independent ferroptosis protection [33]. Additionally, cysteine may inhibit ferroptosis through the ferroptosis suppressor protein 1 (FSP1) pathway [34]. FSP1 converts coenzyme Q10 (CoQ10) to ubiquinol using NADPH, creating a parallel antioxidant system at the plasma membrane [35,36].
Cysteine contributes to this FSP1-CoQ10 system through the pantothenate-coenzyme A synthesis pathway. Labeled cystine incorporates into both glutathione and coenzyme A synthesis [37], suggesting cysteine’s dual protective role against ferroptosis [34].
The discovery of GSH-independent ferroptosis protection mechanisms fundamentally challenges the GPX4-centric view of ferroptosis regulation. The FSP1-CoQ10 system’s existence [35,36] explains why GPX4 inhibition alone often fails to induce complete ferroptosis. However, the relative contribution of these alternative pathways in breast cancer remains unclear. The pantothenate connection [37], is particularly intriguing but lacks breast cancer-specific validation, representing a knowledge gap that could explain therapeutic resistance patterns.

3.4. Nucleotide Metabolism Connections

Nucleotide biosynthesis competes with ferroptosis regulation for GSH resources. Ribonucleotide reductase (RNR) consumes significant GSH for deoxyribonucleotide production, reducing GPX4-available GSH and heightening ferroptosis vulnerability [38]. The p53-p21 axis modulates this competition by suppressing RNR under stress [39]. Nucleotide supplementation can restore proliferation in cysteine-deprived breast cancer cells, confirming direct competition between these pathways [40]. mTORC1 further coordinates this balance by linking GPX4 synthesis to cysteine availability [41].
The GSH competition between nucleotide synthesis and ferroptosis defense represents an underexploited therapeutic vulnerability. While the mechanism is well-established [38,39,40,41], clinical translation remains absent. The key challenge lies in selectively disrupting this balance in cancer versus normal cells, as both require nucleotide synthesis. The finding that nucleotide supplementation rescues cysteine-deprived cells [40] suggests combination therapies blocking both cysteine import and nucleotide synthesis might overcome resistance, though toxicity concerns have prevented clinical advancement (Table 1).

4. Lipid Metabolism: The Primary Battleground

4.1. Polyunsaturated Fatty Acid (PUFA) Synthesis and Incorporation

Polyunsaturated fatty acids (PUFAs), particularly arachidonic acid (AA) and adrenic acid (AdA), serve as primary ferroptosis substrates when incorporated into membrane phospholipids. In breast cancer, PUFA metabolism shows marked dysregulation. Research reveals significant PUFA reduction in malignant phosphoglycerides, attributed to impaired fatty acid desaturases (FADS1/2) [47]. Estrogen further disrupts this balance by upregulating ELOVL2 through ERα, enhancing AA elongation to AdA and shifting membrane omega-6 profiles [48]. These alterations correlate with cancer progression and diminished survival [49].
Dietary omega-3 PUFAs offer potential protection by modulating membrane composition and countering omega-6 dominance [50,51]. However, the clinical translation of dietary interventions remains inconclusive.
The PUFA synthesis disruption in breast cancer presents a fundamental paradox—reduced PUFA content should theoretically protect against ferroptosis, yet these cells remain vulnerable. This suggests that either (1) the specific PUFA species matter more than total content, (2) subcellular PUFA distribution rather than total levels determines sensitivity, or (3) compensatory mechanisms override PUFA depletion effects. The estrogen-ELOVL2-AdA axis [48] provides a mechanistic link between hormonal signaling and ferroptosis susceptibility, potentially explaining ER+ tumors’ lower baseline sensitivity. However, targeting PUFA synthesis directly has shown limited success, likely due to dietary PUFA compensation.

4.2. ACSL4: The Critical Enzyme

Acyl-CoA synthetase long-chain family member 4 (ACSL4) emerges as the master regulator of ferroptosis sensitivity by preferentially activating AA and AdA for incorporation into phospholipids [52,53]. ACSL4’s role in breast cancer is paradoxically subtype-dependent: it inhibits progression in ER+ tumors while promoting proliferation and invasion in ER-negative cancers [54,55]. ACSL4 expression strongly correlates with ferroptosis sensitivity, positioning it as a predictive biomarker [52,56]. ACSL4 deficiency confers ferroptosis resistance, though targeted lipid delivery can bypass this protection [57].
ACSL4 presents the most striking paradox in breast cancer ferroptosis; it is preferentially expressed in basal-like breast cancer [56], predicting higher ferroptosis sensitivity, yet these tumors survive and proliferate aggressively. This suggests that ACSL4 provides selective advantages that outweigh ferroptosis vulnerability, possibly through enhanced membrane remodeling or signaling lipid production. The dual tumor-suppressive (ER+) versus oncogenic (ER−) roles [54,55] indicate context-dependent functions we don’t fully understand. While ACSL4 is the most validated ferroptosis biomarker, its therapeutic targeting remains challenging due to this functional duality and the lack of selective inhibitors that spare normal tissue function.

4.3. Lipid Peroxidation Mechanisms

Enzymatic lipid peroxidation involves arachidonate 15-lipoxygenase (ALOX15), which oxygenates AA-containing phospholipids at the 15th carbon, producing lipid hydroperoxides that disrupt membrane integrity [58]. In breast cancer, ALOX15 expression predicts ferroptosis sensitivity and is included in prognostic models [59,60]. TNBC cells exhibit unique iron and glutathione profiles that amplify both enzymatic and non-enzymatic peroxidation [61]. In addition to lipoxygenases, heme oxygenases contribute to lipid peroxidation in breast cancer cells [62].
While ALOX15 is consistently identified as a ferroptosis mediator, its expression varies dramatically across breast cancer subtypes without clear correlation to clinical outcomes [59,60]. The relative contribution of enzymatic versus non-enzymatic peroxidation remains undefined. Iron-catalyzed Fenton reactions may dominate in high-iron environments like TNBC, while enzymatic pathways prevail in iron-normal contexts. The context-dependent nature of ALOX15 function suggests it may serve as a prognostic marker rather than a therapeutic target. The lack of selective ALOX15 inhibitors that spare normal inflammatory resolution further limits clinical translation.

4.4. Monounsaturated vs. Polyunsaturated Fatty Acids

The SREBP1-SCD axis drives monounsaturated fatty acid (MUFA) production, converting saturated fatty acids to oleic acid, which protects against ferroptosis [63,64]. SCD1 upregulation correlates with worse prognosis in breast cancer, while its inhibition sensitizes cells to ferroptosis [64]. Conversely, ACSL3 activates MUFAs for incorporation via membrane-bound O-acyltransferases (MBOAT1/2), which transfer MUFA-CoAs into phospholipids, reducing peroxidizable substrates [65,66]. MBOAT1, regulated by ER signaling, is highly expressed in breast cancer [66].
Despite ACSL4’s high expression in basal-like breast cancer predicting ferroptosis sensitivity [67], these cells paradoxically survive. Similarly, a TNBC subset shows elevated FADS1/2 expression, rendering them susceptible to ferroptosis inducers [68].
The MUFA/PUFA balance represents a druggable metabolic switch, yet clinical exploitation remains elusive. The paradoxical survival of ACSL4-high/PUFA-rich TNBC cells [67] suggests unknown compensatory mechanisms, possibly through enhanced MUFA production or alternative antioxidant systems. The ER-MBOAT1 connection [66] provides rationale for combining ER antagonists with ferroptosis inducers in hormone-positive disease. However, the fundamental question remains: why do cancer cells maintain high ACSL4/PUFA levels despite ferroptosis risk? This likely reflects PUFAs’ essential roles in signaling and membrane dynamics that outweigh death susceptibility. SCD1 inhibitors show promise but face challenges with systemic toxicity due to MUFA requirements in normal tissues.

4.5. Lipid Availability and Trafficking

Extracellular lipid limitation paradoxically increases ferroptosis sensitivity despite reducing total PUFA levels [69]. Under lipid starvation, adipose triglyceride lipase (ATGL) mobilizes stored triglycerides, liberating PUFAs that are directly incorporated into membrane phospholipids rather than being stored in protective lipid droplets [70,71,72]. This creates highly peroxidizable phosphatidylethanolamine-AA/AdA species that serve as primary ferroptosis substrates.
The LPCAT enzyme family determines membrane PUFA incorporation: LPCAT3 enhances ferroptosis by selectively incorporating AA into phospholipids [73], while LPCAT1 and LPCAT4/MBOAT2 confer resistance through saturated fatty acid and MUFA incorporation [74,75]. The PLA2 family provides the hydrolytic component, with iPLA2β limiting ferroptosis by cleaving oxidized chains [76]. ACSL1 facilitates protective lipid droplet formation, sequestering PUFAs from vulnerable membranes [77,78].
The lipid availability paradox—where nutrient limitation increases death susceptibility—fundamentally challenges cancer biology dogma. This mechanism [69] explains why metabolically stressed tumor regions show enhanced ferroptosis sensitivity, offering therapeutic opportunities through dietary interventions or lipid uptake inhibition. However, the complexity of lipid trafficking networks (>20 enzymes with overlapping functions) makes targeted intervention challenging. The protective role of lipid droplets [77,78] suggests that combination strategies disrupting both storage and membrane incorporation might overcome resistance. Yet clinical translation remains limited by our inability to predict which tumors rely on stored versus dietary lipids, and normal tissues’ requirement for these same pathways.

5. Breast Cancer-Specific Metabolic Reprogramming

5.1. Breast Tumor Microenvironment Factors

Cancer-Associated Fibroblasts (CAFs). CAFs supply cysteine and glutathione while secreting IL-6 to enhance hepcidin expression and increase iron availability [79,80]. They produce cysteine de novo via CBS, directly supporting cancer cell GSH synthesis [81]. CAF-derived lactate induces histone lactylation, enhancing ZFP64 expression and promoting ferroptosis resistance in TNBC, contributing to doxorubicin resistance [82].
Cancer-Associated Adipocytes. Mammary adipocytes protect TNBC cells by secreting oleic acid, which is converted via ACSL3 into membrane phospholipids, displacing PUFAs and reducing peroxidation substrates [83,84]. Co-cultured breast cancer cells show elevated MUFA/PUFA ratios, creating oxidation-resistant membranes [83,85]. High-fat diet exposure downregulates ACSL4 expression, promoting ferroptosis resistance [86].
Immune Components. Tumor-associated macrophages (TAMs) show differential ferroptosis susceptibility—M1 macrophages resist through iNOS/nitric oxide, while M2 macrophages remain sensitive [87,88]. CD8+ T cells release IFNγ and arachidonic acid, sensitizing tumors to ferroptosis by inhibiting xCT and promoting ACSL4-mediated PUFA incorporation [89,90]. However, CAF-derived factors can override these pro-ferroptotic signals.
Matrix and Integration. α6β4 integrin protects against ferroptosis during matrix detachment by maintaining GPX4 expression through Src/STAT3 signaling [91,92].
The microenvironment creates a ferroptosis-resistant sanctuary that likely explains clinical treatment failures. The CAF-adipocyte metabolic support network provides redundant protection mechanisms—even if cancer cell-autonomous pathways are targeted, stromal supply of cysteine, glutathione, and protective lipids maintains resistance. The paradox that immune cells can both promote (CD8+ T cells) and protect against (M2 macrophages) ferroptosis [87,88,89,90] suggests that immunotherapy outcomes may depend on the ferroptosis status of both tumor and immune compartments. Most critically, no current therapies can selectively disrupt stromal–tumor metabolic coupling without affecting normal tissue homeostasis, representing a major barrier to clinical translation.

5.2. Breast Cancer Subtype Variations

TNBC exhibits the highest ferroptosis sensitivity, particularly the luminal androgen receptor (LAR) subtype, due to upregulated oxidized phosphatidylethanolamines (OxPE) and exhausted PUFA pools [93,94]. The LAR subtype’s hypersensitivity to GPX4 inhibitors correlates with distinct lipid profiles and glutathione metabolism alterations [95]. However, TNBC heterogeneity creates variable responses—mesenchymal subtypes show moderate sensitivity with enriched iron metabolism, while immunomodulatory subtypes exhibit minimal ferroptosis features (Table 2) [96].
ER-positive breast cancers (Luminal A/B) demonstrate low baseline ferroptosis sensitivity (low FERscores) [97]. Estrogen signaling upregulates MBOAT1, suppressing ferroptosis, but ER antagonists reverse this protection by downregulating MBOAT1 [98]. A ferroptosis-associated gene signature predicts treatment response across risk groups [99,100].
HER2-enriched tumors show context-dependent sensitivity—moderate baseline vulnerability increases substantially in trastuzumab-resistant states. Lapatinib-tolerant persister cells exhibit GPX4 dependency, creating opportunities for combination therapy [101,102].
The subtype-specific ferroptosis patterns reveal a fundamental inconsistency—TNBC’s high sensitivity should confer growth disadvantage, yet these tumors are the most aggressive. This paradox suggests that either (1) ferroptosis-sensitive cells are eliminated during tumor evolution, leaving resistant clones; (2) the microenvironment provides sufficient protection to override intrinsic sensitivity; or (3) we’re measuring sensitivity incorrectly in vitro. The LAR subtype’s extreme sensitivity [93,94,95] offers the clearest therapeutic window, yet no LAR-specific ferroptosis trials exist. The finding that therapy resistance (tamoxifen, trastuzumab) increases ferroptosis vulnerability [101,102] provides immediate translational opportunities for combination strategies in relapsed disease.

5.3. Oncogene and Tumor Suppressor Influences

RAS Pathway. RAS activation creates multilayered ferroptosis resistance. The RAS-RAF-MEK-ERK cascade upregulates xCT through ETS1 phosphorylation [103,104], while activating NRF2-dependent antioxidant programs [105,106]. The FASN-HIF1α axis provides additional protection—FASN binds HIF1α, enhancing xCT transcription while promoting MUFA synthesis [107,108]. FSP1 regulation through NRF2 establishes GPX4-independent protection via CoQ10 reduction [109,110].
mTOR Pathway. mTORC1 inhibits ferroptosis through the SREBP1-SCD1 axis, promoting MUFA synthesis that reduces membrane peroxidation [111,112]. Additionally, mTOR suppresses ferritinophagy by inhibiting NCOA4, limiting free iron [111,112,113], while activating the p62-KEAP1-NRF2 axis through p62 phosphorylation [114,115]. Paradoxically, mTOR inhibition can protect against ferroptosis by activating glutaminolysis, maintaining NADPH for antioxidant defense [116].
p53. p53 promotes ferroptosis through multiple mechanisms: direct xCT transcriptional repression [117], SAT1 upregulation leading to ALOX15-mediated lipid peroxidation [118], and GLS2 induction depleting glutamine pools [119]. The S47 p53 variant found in African populations shows impaired GLS2 transactivation, conferring ferroptosis resistance [120].
Other Suppressors. BRCA1 deficiency creates differential responses—erastin resistance but GPX4 inhibitor sensitivity [121]. PTEN loss activates AKT-GSK3β-NRF2-xCT, promoting resistance [122,123]. RB deficiency paradoxically increases sensitivity through E2F-mediated ACSL4 upregulation [124].
The oncogene-suppressor ferroptosis network reveals therapeutic complexities (Table 3). RAS pathway targeting faces redundancy—blocking one resistance mechanism (e.g., xCT) is compensated by others (FSP1, FASN). The mTOR paradox, where both activation and inhibition can prevent ferroptosis [112,116], suggests context-dependent effects we don’t fully understand. Most significantly, p53’s ferroptosis-promoting role [117,118,119] contradicts its loss in aggressive cancers. This likely reflects the dominance of oncogenic resistance pathways over tumor suppressor sensitization. The BRCA1 differential response [121] offers immediate clinical relevance for PARP inhibitor combinations, yet remains unexploited. These findings suggest single-pathway targeting will fail; successful strategies must simultaneously disrupt multiple resistance mechanisms.

6. Additional Metabolic Pathways

6.1. Glucose Metabolism Connections

Glucose metabolism critically modulates ferroptosis through NADPH generation and redox balance (Figure 2). The pentose phosphate pathway (PPP), via glucose-6-phosphate dehydrogenase (G6PD), produces NADPH essential for GSH regeneration and GPX4 function [138,139]. Paradoxically, NADPH also fuels NADPH oxidases that generate ROS, creating opposing ferroptosis effects [140]. In breast cancer, high xCT expression coupled with glucose deprivation triggers both ferroptosis and disulfidptosis—a novel death mechanism involving aberrant disulfide crosslinking when NADPH is depleted [141,142].
Glycolytic alterations affect ferroptosis sensitivity: PDK4 blocks pyruvate entry into the TCA cycle, conferring resistance [143], while ferroptosis inducers downregulate key glycolytic enzymes (HK2, PFKP, PKM2), increasing tumor vulnerability [144]. Our own research demonstrated that glucose deprivation in MCF-7 and T47D cells activates ferroptosis through NRF2 signaling [145]. The lactate/MCT1/AMPK axis shows context-dependent effects, promoting ferroptosis in normal environments but conferring resistance in acidic tumor microenvironments [146].
The glucose–ferroptosis relationship presents a therapeutic dilemma. While glucose restriction sensitizes cells to ferroptosis [141,145], tumors adapt through enhanced PPP flux or alternative NADPH sources (folate metabolism, IDH mutations). The discovery of disulfidptosis [141,142] reveals that targeting glucose metabolism can trigger multiple death pathways simultaneously, potentially overcoming resistance. However, the dual role of NADPH—protecting via GSH while promoting ROS via NOX—makes prediction of net effects challenging. The lactate paradox [146], where the same metabolite has opposite effects depending on pH, exemplifies why metabolic interventions fail clinically. GLUT inhibitors remain in preclinical development due to inability to selectively target cancer metabolism without affecting normal glucose homeostasis.

6.2. Selenium and Antioxidant Systems

Beyond glutathione, breast cancer cells employ multiple antioxidant systems. Selenium’s incorporation into GPX4 is essential for its function, with sodium selenite paradoxically inducing ferroptosis in TNBC through ATM kinase activation despite being an antioxidant cofactor [147,148]. The thioredoxin reductase system provides GSH-independent protection—its inhibition enhances radiation sensitivity in breast cancer stem cells [64].
The FSP1-CoQ10-NAD(P)H pathway operates independently of GPX4, reducing ubiquinone to ubiquinol at the plasma membrane [35,149]. However, the UBIAD1/CoQ10 system presents a striking paradox: UBIAD1 expression and CoQ10 levels, despite their antioxidant function, actually enhance ferroptosis sensitivity in breast cancer [150,151]. High UBIAD1/CoQ10 cells show decreased GPX4 synthesis and reduced FSP1 levels, with UBIAD1-low patients exhibiting significantly shorter survival, particularly in TNBC [150]. This counterintuitive finding suggests that elevated CoQ10 may disrupt the balance between different antioxidant systems, potentially through negative feedback on GPX4/FSP1 expression or by altering cellular redox setpoints that make cells more vulnerable to acute oxidative challenges.
Additional protective mechanisms include the GCH1–tetrahydrobiopterin (BH4) axis, which prevents ferroptosis through lipid remodeling [152], and vitamin E derivatives like D-α-tocopherol succinate that paradoxically induce rather than prevent ferroptosis in drug-resistant cells [153].
The non-glutathione antioxidant systems reveal unexpected complexities that challenge therapeutic targeting. The UBIAD1/CoQ10 paradox [150,151]—where antioxidant elevation increases death susceptibility—suggests that cellular redox homeostasis operates within narrow boundaries, and both deficiency and excess can trigger ferroptosis. This may explain why antioxidant supplementation trials often fail or worsen cancer outcomes. The selenium paradox [147,148], where the same element can prevent or promote ferroptosis depending on dose and form, further complicates therapeutic development. The existence of multiple parallel systems (FSP1, thioredoxin, BH4) explains why GPX4 inhibition alone rarely achieves complete ferroptosis. Clinical translation requires either simultaneous inhibition of all systems (likely toxic) or exploitation of paradoxical responses like UBIAD1, though mechanisms remain incompletely understood.

7. Future Directions and Challenges

7.1. Multi-Omics Integration and Tumor Organoids

Multi-omics integration has revealed subtype-specific ferroptosis vulnerabilities in breast cancer. A lipid metabolism–ferroptosis signature successfully stratified HR+ patients into risk groups with distinct therapeutic responses [154]. Quantification of ferroptosis pathway status uncovered TNBC-specific fatty acid metabolism alterations predictive of treatment outcomes [155].
Patient-derived organoids (PDOs) provide functional validation of multi-omics findings [156]. HER2+ organoids revealed that combined anti-FGFR4/anti-HER2 therapy triggers synergistic ferroptosis [157]. Tamoxifen induces ferroptosis in MCF-7 organoids through lipid metabolism disruption [158], while minimal residual disease organoids exhibit dysregulated lipid metabolism with elevated oxidative stress, identifying a therapeutic window [159].
While multi-omics approaches generate comprehensive datasets, clinical translation remains limited. The identified signatures [154,155] lack prospective validation and standardized thresholds for patient stratification. PDO models [157,158,159], though promising, fail to recapitulate the tumor microenvironment’s metabolic contributions—a critical limitation given our findings that CAFs and adipocytes provide ferroptosis protection (Section 5.1). The correlation between multi-omics signatures and actual ferroptosis occurrence in patients remains unvalidated. Most critically, these approaches identify associations rather than causal relationships, limiting therapeutic targeting. The field requires functional validation studies linking specific metabolic signatures to ferroptosis sensitivity in clinical specimens, not just cell lines or organoids.

7.2. Immunotherapy Synergy

Ferroptosis induction synergizes with immune checkpoint inhibitors (ICIs) through multiple mechanisms (Figure 3). Ferroptotic cancer cells upregulate PD-L1 via IFNγ signaling, creating ideal ICI targets [160]. Cyclophosphamide, a ferroptosis-promoting chemotherapeutic, increases PD-L1 in TNBC immune infiltrates [160]. The LAR TNBC subtype shows high ferroptosis resistance correlating with poor ICI response, while targeting GPX4 with RSL3 sensitizes these cells and enhances T-cell infiltration [161].
PRMT5 emerges as a key resistance mechanism, methylating KEAP1 to stabilize NRF2 and suppress ferroptosis. PRMT5 inhibition combined with anti-PD-1 achieves 60–80% tumor growth inhibition versus monotherapy [162]. Natural compounds like toxicarioside H and statins deplete GSH to trigger ferroptosis in TNBC [163,164]. Targeted delivery strategies using PD-1-coated nanoparticles achieve 90% tumor inhibition with anti-PD-L1 combination [165].
While ferroptosis–immunotherapy synergy shows impressive preclinical results (60–90% tumor inhibition) [162,165], no clinical trials have validated these findings. The major paradox—that ferroptosis induces immunogenic cell death yet simultaneously impairs CD8+ T cells through CD36-mediated lipid uptake—remains unresolved. The PD-L1 upregulation mechanism [160] could theoretically enhance ICI response, but ferroptotic damage to effector T cells might negate benefits. PRMT5 inhibitors are in Phase I trials for other cancers, not specifically for ferroptosis enhancement. The field lacks biomarkers to identify patients who would benefit from combination therapy versus those at risk for immune dysfunction. Most critically, the optimal sequencing—simultaneous versus sequential ferroptosis induction and ICI—remains undefined, with opposing rationales for each approach.

7.3. Photodynamic-Ferroptosis Synergy Treatment

Photodynamic therapy (PDT) directly triggers ferroptosis through reactive oxygen species generation that overwhelms the GSH/GPX4 system [166] (Figure 4). PDT achieves tumor inhibition rates exceeding 95% when combined with ferroptosis inducers, addressing PDT’s oxygen dependence and ferroptosis’s requirement for specific cellular conditions [167,168].
Chlorin e6 (Ce6) and protoporphyrin IX serve as primary photosensitizers, generating singlet oxygen that depletes GSH and downregulates GPX4 [169,170]. Novel iridium-ferrocene conjugates (IrFc1) provide self-amplifying mechanisms where endogenous ROS enhance photosensitization specifically in TNBC [171]. The combination creates a feedback loop where PDT-generated ROS enhance ferroptosis while iron-dependent reactions improve PDT efficacy [166,172].
Despite impressive preclinical results (>95% tumor inhibition) [167,168], PDT-ferroptosis combination faces insurmountable clinical barriers. PDT’s fundamental limitation—light penetration restricted to <1 cm tissue depth—makes it unsuitable for most breast cancers except superficial recurrences. The oxygen dependence of both PDT and ferroptosis creates a therapeutic paradox in hypoxic tumor cores where treatment is most needed. While 217 publications appeared in 2023 alone [173], none report clinical trials. The photosensitizers showing best ferroptosis synergy (Ce6, IrFc1) lack FDA approval for any indication. Most critically, the simultaneous requirement for light delivery, adequate tissue oxygenation, and ferroptosis-permissive conditions may never align in clinical tumors. This approach likely remains limited to ex vivo treatment of surgical margins or palliative management of cutaneous metastases.

8. Conclusions

Ferroptosis represents both a promising therapeutic target and a cautionary tale of biological complexity in breast cancer. Our analysis reveals that breast cancer cells navigate a precarious balance, maintaining sufficient iron and PUFAs for proliferation while avoiding ferroptotic death through multilayered protective mechanisms involving glutathione systems, alternative antioxidants, and microenvironmental support.
The most striking finding is the prevalence of paradoxes that challenge conventional understanding. TNBC exhibits high ferroptosis sensitivity yet survives through microenvironmental protection from CAFs and adipocytes. The ACSL4 and UBIAD1/CoQ10 paradoxes suggest that cellular redox homeostasis operates within narrow boundaries that we don’t fully comprehend. These contradictions likely explain why ferroptosis-inducing monotherapies have failed to advance clinically despite compelling preclinical data.
Subtype-specific vulnerabilities offer the clearest path forward. The LAR TNBC subtype’s extreme sensitivity, luminal cancers’ dependence on xCT during CDK4/6 inhibitor treatment, and HER2+ tumors’ vulnerability in resistant states provide specific therapeutic windows. However, exploitation requires overcoming the redundancy problem—cancer cells employ at least six parallel protection systems (GPX4, FSP1, transsulfuration, lysosomal storage, microenvironmental supply, and MUFA incorporation), making single-target approaches futile.
Clinical Translation Barriers: Current strategies to exploit ferroptosis are summarized in Table 4. However, three fundamental obstacles prevent clinical advancement: (1) pharmacological—GPX4 inhibitors show poor bioavailability and hepatotoxicity; (2) biological—the tumor microenvironment provides metabolic sanctuary that cell-autonomous targeting cannot overcome; (3) selectivity—we cannot distinguish cancer from normal cell ferroptosis requirements. The promising synergies with immunotherapy and PDT face their own limitations, including immune cell toxicity and tissue penetration constraints.
Future Priorities: Rather than pursuing additional mechanistic studies, the field requires: (1) biomarker development to identify ferroptosis-primed tumors; (2) combination strategies that simultaneously target multiple resistance mechanisms while protecting normal tissues; (3) clinical trials in specific contexts where ferroptosis vulnerability is maximized (resistant disease, specific subtypes); (4) realistic assessment of which findings can translate versus those limited by fundamental biological constraints.
The metabolic regulation of ferroptosis in breast cancer ultimately demonstrates that targeting cell death pathways requires understanding not just the death mechanism itself, but the entire metabolic ecosystem that determines cellular fate. Success will come not from stronger ferroptosis inducers, but from strategic exploitation of context-specific vulnerabilities while managing the inevitable resistance mechanisms that evolution has provided cancer cells.

Author Contributions

Conceptualization, N.G. and M.W.; writing—original draft preparation, M.W.; writing—review and editing, N.G. and M.W.; visualization, N.G. All authors have read and agreed to the published version of the manuscript.

Funding

This project was supported by grants from the National Institutes of Health (NIH), National Institute of General Medical Sciences (NIGMS), IDeA Networks of Biomedical Research Excellence (INBRE), Award number: P20GM103466. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health. M.W. received funding from NSF INCLUDES Alliance: NSF HRD-2217242.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The original contributions presented in this study are included in the article/figures. Further inquiries may be directed to the corresponding author.

Acknowledgments

During the preparation of this manuscript, the authors used “Elicit: The AI Research Assistant” (elicit.com, last accessed on 14 July 2025); “Consensus” (consensus.app, last accessed on 14 July 2025) for the purposes of curating relevant publications. The authors used Anthropic’s Claude (version Claude Opus 4.1) and XAi (version Grok 3 and 4) to verify the scientific article content, create the outline for this article, and assist with the concision of the writing. The authors have reviewed and edited the output and take full responsibility for the content of this publication.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
AAArachidonic Acid
ACSL3Acyl-CoA Synthetase Long-Chain Family Member 3
ACSL4Acyl-CoA Synthetase Long-Chain Family Member 4
AdAAdrenic Acid
AGPSAlkylglycerone Phosphate Synthase
ALOX15Arachidonate 15-Lipoxygenase
ATGLAdipose Triglyceride Lipase
BH4Tetrahydrobiopterin
CAFCancer-Associated Fibroblast
CBSCystathionine Beta-Synthase
CISD1CDGSH Iron Sulfur Domain 1
CTHCystathionine Gamma-Lyase
CTNSCystinosin
DGLADihomo-Gamma-Linolenic Acid
DLDDihydrolipoamide Dehydrogenase
DMT1Divalent Metal Transporter 1
EREstrogen Receptor
FADS1Fatty Acid Desaturase 1
FADS2Fatty Acid Desaturase 2
FASNFatty Acid Synthase
FERscoreFerroptosis Score
FSP1Ferroptosis Suppressor Protein 1
FTH1Ferritin Heavy Chain 1
G6PDGlucose-6-Phosphate Dehydrogenase
GCH1GTP Cyclohydrolase 1
GGT1Gamma-Glutamyltransferase 1
GLAGamma-Linolenic Acid
GLS2Glutaminase 2
GPX4Glutathione Peroxidase 4
GSHGlutathione
HERC2HECT Domain and RCC1-Like Domain-Containing Protein 2
HIF1αHypoxia-Inducible Factor 1-Alpha
HK2Hexokinase 2
HSLHormone-Sensitive Lipase
ICDImmunogenic Cell Death
IFNγInterferon-Gamma
IKEImidazole Ketone Erastin
IL-6Interleukin-6
KGDHAlpha-Ketoglutarate Dehydrogenase
LALinoleic Acid
LARLuminal Androgen Receptor
LIPLabile Iron Pool
LPCATLysophosphatidylcholine Acyltransferase
MBOATMembrane-Bound O-Acyltransferase
MGLMonoglyceride Lipase
MFSD12Major Facilitator Superfamily Domain-Containing Protein 12
MUFAMonounsaturated Fatty Acid
NADPHNicotinamide Adenine Dinucleotide Phosphate
NCOA4Nuclear Receptor Coactivator 4
NRF2Nuclear Factor Erythroid 2-Related Factor 2
OxPEOxidized Phosphatidylethanolamine
PCPhosphatidylcholine
PDK4Pyruvate Dehydrogenase Kinase 4
PD-L1Programmed Death-Ligand 1
PEPhosphatidylethanolamine
PKAProtein Kinase A
PKM2Pyruvate Kinase M2
PLA2Phospholipase A2
PPPPentose Phosphate Pathway
PRMT5Protein Arginine Methyltransferase 5
PUFAPolyunsaturated Fatty Acid
RNRRibonucleotide Reductase
ROSReactive Oxygen Species
SAT1Spermidine/Spermine N1-Acetyltransferase 1
SCDStearoyl-CoA Desaturase
SREBP1Sterol Regulatory Element-Binding Protein 1
STEAP3Six-Transmembrane Epithelial Antigen of Prostate 3
TAMTumor-Associated Macrophage
TCATricarboxylic Acid
TFTransferrin
TFR1Transferrin Receptor 1
TNBCTriple-Negative Breast Cancer
TOSD-α-Tocopherol Succinate
TrxR1Thioredoxin Reductase 1
UBIAD1UbiA Prenyltransferase Domain-Containing Protein 1
VDAC3Voltage-Dependent Anion Channel 3
xCTCystine/Glutamate Antiporter (SLC7A11)

References

  1. Giaquinto, A.N.; Sung, H.; Miller, K.D.; Kramer, J.L.; Newman, L.A.; Minihan, A.; Jemal, A.; Siegel, R.L. Breast Cancer Statistics, 2022. CA Cancer J. Clin. 2022, 72, 524–541. [Google Scholar] [CrossRef]
  2. Dixon, S.J.; Lemberg, K.M.; Lamprecht, M.R.; Skouta, R.; Zaitsev, E.M.; Gleason, C.E.; Patel, D.N.; Bauer, A.J.; Cantley, A.M.; Yang, W.S.; et al. Ferroptosis: An Iron-Dependent Form of Nonapoptotic Cell Death. Cell 2012, 149, 1060–1072. [Google Scholar] [CrossRef]
  3. Singh, M.; Mugler, K.; Hailoo, D.W.; Burke, S.; Nemesure, B.; Torkko, K.; Shroyer, K.R. Differential expression of transferrin receptor (TfR) in a spectrum of normal to malignant breast tissues: Implications for in situ and invasive carcinoma. Appl. Immunohistochem. Mol. Morphol. 2011, 19, 417–423. [Google Scholar] [CrossRef]
  4. Habashy, H.O.; Powe, D.G.; Staka, C.M.; Rakha, E.A.; Ball, G.; Green, A.R.; Aleskandarany, M.; Paish, E.C.; Douglas Macmillan, R.; Nicholson, R.I.; et al. Transferrin Receptor (CD71) is a Marker of Poor Prognosis in Breast Cancer and can Predict Response to Tamoxifen. Breast Cancer Res. Treat. 2010, 119, 283–293. [Google Scholar] [CrossRef] [PubMed]
  5. Chen, C.; Liu, P.; Duan, X.; Cheng, M.; Xu, L.X. Deferoxamine-Induced High Expression of TfR1 and DMT1 Enhanced Iron Uptake in Triple-Negative Breast Cancer Cells by Activating IL-6/PI3K/AKT Pathway. Onco Targets Ther. 2019, 12, 4359–4377. [Google Scholar] [CrossRef] [PubMed]
  6. Jiang, X.P.; Elliott, R.L.; Head, J.F. Manipulation of Iron Transporter Genes Results in the Suppression of Human and Mouse Mammary Adenocarcinomas. Anticancer Res. 2010, 30, 759–765. [Google Scholar]
  7. Li, J.; He, K.; Liu, P.; Xu, L.X. Iron Participated in Breast Cancer Chemoresistance by Reinforcing IL-6 Paracrine Loop. Biochem. Biophys. Res. Commun. 2016, 475, 154–160. [Google Scholar] [CrossRef]
  8. Tubbesing, K.; Khoo, T.C.; Bahreini Jangjoo, S.; Sharikova, A.; Barroso, M.; Khmaladze, A. Iron-Binding Cellular Profile of Transferrin using Label-Free Raman Hyperspectral Imaging and Singular Value Decomposition (SVD). Free Radic. Biol. Med. 2021, 169, 416–424. [Google Scholar] [CrossRef]
  9. Dong, X.; Li, Y.; Sheng, X.; Zhou, W.; Sun, A.; Dai, H. Mitochondria-Related Signaling Pathways Involved in Breast Cancer Regulate Ferroptosis. Genes Dis. 2023, 11, 358–366. [Google Scholar] [CrossRef]
  10. Battaglia, A.M.; Chirillo, R.; Aversa, I.; Sacco, A.; Costanzo, F.; Biamonte, F. Ferroptosis and Cancer: Mitochondria Meet the “Iron Maiden” Cell Death. Cells 2020, 9, 1505. [Google Scholar] [CrossRef]
  11. Gan, H.; Huang, X.; Luo, X.; Li, J.; Mo, B.; Cheng, L.; Shu, Q.; Du, Z.; Tang, H.; Sun, W.; et al. A Mitochondria-Targeted Ferroptosis Inducer Activated by Glutathione-Responsive Imaging and Depletion for Triple Negative Breast Cancer Theranostics. Adv. Healthc. Mater. 2023, 12, 2300220. [Google Scholar] [CrossRef] [PubMed]
  12. Yuan, H.; Li, X.; Zhang, X.; Kang, R.; Tang, D. CISD1 Inhibits Ferroptosis by Protection Against Mitochondrial Lipid Peroxidation. Biochem. Biophys. Res. Commun. 2016, 478, 838–844. [Google Scholar] [CrossRef] [PubMed]
  13. Gao, M.; Monian, P.; Pan, Q.; Zhang, W.; Xiang, J.; Jiang, X. Ferroptosis is an Autophagic Cell Death Process. Cell Res. 2016, 26, 1021–1032. [Google Scholar] [CrossRef] [PubMed]
  14. Mancias, J.D.; Pontano Vaites, L.; Nissim, S.; Biancur, D.E.; Kim, A.J.; Wang, X.; Liu, Y.; Goessling, W.; Kimmelman, A.C.; Harper, J.W. Ferritinophagy Via NCOA4 is Required for Erythropoiesis and is Regulated by Iron Dependent HERC2-Mediated Proteolysis. eLife 2015, 4, e10308. [Google Scholar] [CrossRef]
  15. Zuo, T.; Zhang, J.; Deng, H.; Yang, J.; Xu, R.; Wang, Z.; Shen, Q.; Fang, T. pH-Sensitive Molecular-Switch-Containing Polymer Nanoparticle for Breast Cancer Therapy with Ferritinophagy-Cascade Ferroptosis and Tumor Immune Activation. Adv. Healthc. Mater. 2021, 10, 2100683. [Google Scholar] [CrossRef]
  16. Cañeque, T.; Baron, L.; Müller, S.; Carmona, A.; Colombeau, L.; Versini, A.; Solier, S.; Gaillet, C.; Sindikubwabo, F.; Sampaio, J.L.; et al. Activation of Lysosomal Iron Triggers Ferroptosis in Cancer. Nature 2025, 642, 492–500. [Google Scholar] [CrossRef]
  17. Forcina, G.C.; Dixon, S.J. GPX4 at the Crossroads of Lipid Homeostasis and Ferroptosis. Proteomics 2019, 19, 1800311. [Google Scholar] [CrossRef]
  18. Zhang, X.-D.; Liu, Z.-Y.; Wang, M.-S.; Guo, Y.-X.; Wang, X.-K.; Luo, K.; Huang, S.; Li, R.-F. Mechanisms and Regulations of Ferroptosis. Front. Immunol. 2023, 14, 1269451. [Google Scholar] [CrossRef]
  19. Yang, W.S.; SriRamaratnam, R.; Welsch, M.E.; Shimada, K.; Skouta, R.; Viswanathan, V.S.; Cheah, J.H.; Clemons, P.A.; Shamji, A.F.; Clish, C.B.; et al. Regulation of Ferroptotic Cancer Cell Death by GPX4. Cell 2014, 156, 317–331. [Google Scholar] [CrossRef]
  20. Lee, N.; Carlisle, A.E.; Peppers, A.; Park, S.J.; Doshi, M.B.; Spears, M.E.; Kim, D. xCT-Driven Expression of GPX4 Determines Sensitivity of Breast Cancer Cells to Ferroptosis Inducers. Antioxidants 2021, 10, 317. [Google Scholar] [CrossRef]
  21. Hasegawa, M.; Takahashi, H.; Rajabi, H.; Alam, M.; Suzuki, Y.; Yin, L.; Tagde, A.; Maeda, T.; Hiraki, M.; Sukhatme, V.P.; et al. Functional Interactions of the Cystine/Glutamate Antiporter, CD44v and MUC1-C Oncoprotein in Triple-Negative Breast Cancer Cells. Oncotarget 2016, 7, 11756–11769. [Google Scholar] [CrossRef]
  22. Li, F.; Long, H.; Zhou, Z.; Luo, H.; Xu, S.; Gao, L. System Xc −/GSH/GPX4 Axis: An Important Antioxidant System for the Ferroptosis in Drug-Resistant Solid Tumor Therapy. Front. Pharmacol. 2022, 13, 910292. [Google Scholar] [CrossRef] [PubMed]
  23. Jyotsana, N.; Ta, K.T.; DelGiorno, K.E. The role of cystine/glutamate antiporter SLC7A11/xCT in the pathophysiology of cancer. Front. Oncol. 2022, 12, 858462. [Google Scholar] [CrossRef] [PubMed]
  24. Azizi, R.; Ahmed, H.H.; Abdul Kareem, R.; Taher Waam, W.M.; Alwan, M.; Jawad, M.J.; Hamad, A.K.; Darzi, S. SLC7A11 Inhibitors Represent a Promising Therapeutic Target by Facilitating the Induction of Ferroptosis in Breast Cancer. Int. J. Mol. Cell. Med. 2025, 14, 496–516. [Google Scholar] [CrossRef] [PubMed]
  25. Li, H.; Yang, P.; Wang, J.; Zhang, J.; Ma, Q.; Jiang, Y.; Wu, Y.; Han, T.; Xiang, D. HLF Regulates Ferroptosis, Development and Chemoresistance of Triple-Negative Breast Cancer by Activating Tumor Cell-Macrophage Crosstalk. J. Hematol. Oncol. 2022, 15, 2. [Google Scholar] [CrossRef]
  26. Cui, Y.; Li, Y.; Xu, Y.; Liu, X.; Kang, X.; Zhu, J.; Long, S.; Han, Y.; Xue, C.; Sun, Z.; et al. SLC7A11 Protects Luminal A Breast Cancer Cells Against Ferroptosis Induced by CDK4/6 Inhibitors. Redox Biol. 2024, 76, 103304. [Google Scholar] [CrossRef]
  27. Koppula, P.; Zhuang, L.; Gan, B. Cystine Transporter SLC7A11/xCT in Cancer: Ferroptosis, Nutrient Dependency, and Cancer Therapy. Protein Cell 2021, 12, 599–620. [Google Scholar] [CrossRef]
  28. Chen, M.; Wang, S.; Hsu, C.; Yin, P.; Yeh, T.; Lee, H.; Tseng, L. 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, 114588–114602. [Google Scholar] [CrossRef]
  29. Erdélyi, K.; Ditrói, T.; Johansson, H.J.; Czikora, Á.; Balog, N.; Silwal-Pandit, L.; Ida, T.; Olasz, J.; Hajdú, D.; Mátrai, Z.; et al. Reprogrammed Transsulfuration Promotes Basal-Like Breast Tumor Progression Via Realigning Cellular Cysteine Persulfidation. Proc. Natl. Acad. Sci. USA 2021, 118, e2100050118. [Google Scholar] [CrossRef]
  30. Xue, W.; Yu, Y.; Yao, Y.; Zhou, L.; Huang, Y.; Wang, Y.; Chen, Z.; Wang, L.; Li, X.; Wang, X.; et al. Breast Cancer Cells have an Increased Ferroptosis Risk Induced by System X(C)(-) Blockade After Deliberately Downregulating CYTL1 to Mediate Malignancy. Redox Biol. 2024, 70, 103034. [Google Scholar] [CrossRef]
  31. Swanda, R.V.; Ji, Q.; Wu, X.; Yan, J.; Dong, L.; Mao, Y.; Uematsu, S.; Dong, Y.; Qian, S. Lysosomal Cystine Governs Ferroptosis Sensitivity in Cancer Via Cysteine Stress Response. Mol. Cell 2023, 83, 3347–3359.e9. [Google Scholar] [CrossRef]
  32. He, L.; Chen, J.; Deng, P.; Huang, S.; Liu, P.; Wang, C.; Huang, X.; Li, Y.; Chen, B.; Shi, D.; et al. Lysosomal Cyst(E)Ine Storage Potentiates Tolerance to Oxidative Stress in Cancer Cells. Mol. Cell 2023, 83, 3502–3519.e11. [Google Scholar] [CrossRef] [PubMed]
  33. Xia, C.; Xing, X.; Zhang, W.; Wang, Y.; Jin, X.; Tian, M.; Ba, X.; Hao, F. Cysteine and Homocysteine can be Exploited by GPX4 in Ferroptosis Inhibition Independent of GSH Synthesis. Redox Biol. 2023, 69, 102999. [Google Scholar] [CrossRef] [PubMed]
  34. Daher, B.; Vučetić, M.; Pouysségur, J. Cysteine Depletion, a Key Action to Challenge Cancer Cells to Ferroptotic Cell Death. Front. Oncol. 2020, 10, 723. [Google Scholar] [CrossRef] [PubMed]
  35. Doll, S.; Freitas, F.P.; Shah, R.; Aldrovandi, M.; da Silva, M.C.; Ingold, I.; Goya Grocin, A.; Xavier da Silva, T.N.; Panzilius, E.; Scheel, C.H.; et al. FSP1 is a Glutathione-Independent Ferroptosis Suppressor. Nature 2019, 575, 693–698. [Google Scholar] [CrossRef]
  36. Bersuker, K.; Hendricks, J.M.; Li, Z.; Magtanong, L.; Ford, B.; Tang, P.H.; Roberts, M.A.; Tong, B.; Maimone, T.J.; Zoncu, R.; et al. The CoQ Oxidoreductase FSP1 Acts Parallel to GPX4 to Inhibit Ferroptosis. Nature 2019, 575, 688–692. [Google Scholar] [CrossRef]
  37. Badgley, M.A.; Kremer, D.M.; Maurer, H.C.; DelGiorno, K.E.; Lee, H.; Purohit, V.; Sagalovskiy, I.R.; Ma, A.; Kapilian, J.; Firl, C.E.M.; et al. Cysteine Depletion Induces Pancreatic Tumor Ferroptosis in Mice. Science 2020, 368, 85–89. [Google Scholar] [CrossRef]
  38. Tarangelo, A.; Rodencal, J.; Kim, J.T.; Magtanong, L.; Long, J.Z.; Dixon, S.J. Nucleotide Biosynthesis Links Glutathione Metabolism to Ferroptosis Sensitivity. Life. Sci. Alliance 2022, 5, e202101157. [Google Scholar] [CrossRef]
  39. Tarangelo, A.; Magtanong, L.; Bieging-Rolett, K.T.; Li, Y.; Ye, J.; Attardi, L.D.; Dixon, S.J. p53 Suppresses Metabolic Stress-Induced Ferroptosis in Cancer Cells. Cell Rep. 2018, 22, 569–575. [Google Scholar] [CrossRef]
  40. Allen, A.E.; Sun, Y.; Wei, F.; Reid, M.A.; Locasale, J.W. Nucleotide Metabolism is Linked to Cysteine Availability. J. Biol. Chem. 2023, 299, 103039. [Google Scholar] [CrossRef]
  41. Zhang, Y.; Swanda, R.V.; Nie, L.; Liu, X.; Wang, C.; Lee, H.; Lei, G.; Mao, C.; Koppula, P.; Cheng, W.; et al. mTORC1 Couples Cyst(E)Ine Availability with GPX4 Protein Synthesis and Ferroptosis Regulation. Nat. Commun. 2021, 12, 1589. [Google Scholar] [CrossRef] [PubMed]
  42. Liu, Y.; Wan, Y.; Jiang, Y.; Zhang, L.; Cheng, W. GPX4: The Hub of Lipid Oxidation, Ferroptosis, Disease and Treatment. Biochim. Biophys. Acta-Rev. Cancer 2023, 1878, 188890. [Google Scholar] [CrossRef] [PubMed]
  43. Beatty, A.; Fink, L.; Singh, T.; Strigun, A.; Peter, E.; Ferrer, C.; Nicolas, E.; Cai, K.; Moran, T.; Reginato, M.; et al. Metabolite Profiling Reveals the Glutathione Biosynthetic Pathway as a Therapeutic Target in Triple-Negative Breast Cancer. Mol. Cancer Ther. 2017, 17, 264–275. [Google Scholar] [CrossRef] [PubMed]
  44. Combs, J.A.; DeNicola, G.M. The Non-Essential Amino Acid Cysteine Becomes Essential for Tumor Proliferation and Survival. Cancers 2019, 11, 678. [Google Scholar] [CrossRef]
  45. Chen, R.; Wen, L.; Guo, F.; He, J.; Wong, K.H.; Chen, M. Glutathione-Scavenging Natural-Derived Ferroptotic Nano-Amplifiers Strengthen Tumor Therapy through Aggravating Iron Overload and Lipid Peroxidation. J. Control. Release 2025, 379, 866–878. [Google Scholar] [CrossRef]
  46. Ryu, C.S.; Kwak, H.C.; Lee, K.S.; Kang, K.W.; Oh, S.J.; Lee, K.H.; Kim, H.M.; Ma, J.Y.; Kim, S.K. Sulfur Amino Acid Metabolism in Doxorubicin-Resistant Breast Cancer Cells. Toxicol. Appl. Pharmacol. 2011, 255, 94–102. [Google Scholar] [CrossRef]
  47. Grammatikos, S.I.; Subbaiah, P.V.; Victor, T.A.; Miller, W.M. N-3 and N-6 Fatty Acid Processing and Growth Effects in Neoplastic and Non-Cancerous Human Mammary Epithelial Cell Lines. Br. J. Cancer 1994, 70, 219–227. [Google Scholar] [CrossRef]
  48. González-Bengtsson, A.; Asadi, A.; Gao, H.; Dahlman-Wright, K.; Jacobsson, A. Estrogen Enhances the Expression of the Polyunsaturated Fatty Acid Elongase Elovl2 Via ERα in Breast Cancer Cells. PLoS ONE 2016, 11, e0164241. [Google Scholar] [CrossRef]
  49. Hilvo, M.; Denkert, C.; Lehtinen, L.; Müller, B.; Brockmöller, S.; Seppänen-Laakso, T.; Budczies, J.; Bucher, E.; Yetukuri, L.; Castillo, S.; et al. Novel Theranostic Opportunities Offered by Characterization of Altered Membrane Lipid Metabolism in Breast Cancer Progression. Cancer Res. 2011, 71, 3236–3245. [Google Scholar] [CrossRef]
  50. Mukerjee, S.; Saeedan, A.S.; Ansari, M.N.; Singh, M. Polyunsaturated Fatty Acids Mediated Regulation of Membrane Biochemistry and Tumor Cell Membrane Integrity. Membranes 2021, 11, 479. [Google Scholar] [CrossRef]
  51. Azrad, M.; Turgeon, C.; Demark-Wahnefried, W. Current Evidence Linking Polyunsaturated Fatty Acids with Cancer Risk and Progression. Front. Oncol. 2013, 3, 224. [Google Scholar] [CrossRef]
  52. Doll, S.; Proneth, B.; Tyurina, Y.Y.; Panzilius, E.; Kobayashi, S.; Ingold, I.; Irmler, M.; Beckers, J.; Aichler, M.; Walch, A.; et al. ACSL4 Dictates Ferroptosis Sensitivity by Shaping Cellular Lipid Composition. Nat. Chem. Biol. 2017, 13, 91–98. [Google Scholar] [CrossRef]
  53. Kagan, V.E.; Mao, G.; Qu, F.; Angeli, J.P.F.; Doll, S.; Croix, C.S.; Dar, H.H.; Liu, B.; Tyurin, V.A.; Ritov, V.B.; et al. Oxidized Arachidonic and Adrenic PEs Navigate Cells to Ferroptosis. Nat. Chem. Biol. 2017, 13, 81–90. [Google Scholar] [CrossRef]
  54. Hou, J.; Jiang, C.; Wen, X.; Li, C.; Xiong, S.; Yue, T.; Long, P.; Shi, J.; Zhang, Z. ACSL4 as a Potential Target and Biomarker for Anticancer: From Molecular Mechanisms to Clinical Therapeutics. Front. Pharmacol. 2022, 13, 949863. [Google Scholar] [CrossRef] [PubMed]
  55. Quan, J.; Bode, A.M.; Luo, X. ACSL Family: The Regulatory Mechanisms and Therapeutic Implications in Cancer. Eur. J. Pharmacol. 2021, 909, 174397. [Google Scholar] [CrossRef] [PubMed]
  56. Yuan, H.; Li, X.; Zhang, X.; Kang, R.; Tang, D. Identification of ACSL4 as a Biomarker and Contributor of Ferroptosis. Biochem. Biophys. Res. Commun. 2016, 478, 1338–1343. [Google Scholar] [CrossRef] [PubMed]
  57. Luo, J.; Li, Y.; Li, Y.; Chen, X.; Du, P.; Wang, Z.; Tian, A.; Zhao, Y. Reversing Ferroptosis Resistance in Breast Cancer Via Tailored Lipid and Iron Presentation. ACS Nano 2023, 17, 25257–25268. [Google Scholar] [CrossRef]
  58. Shintoku, R.; Takigawa, Y.; Yamada, K.; Kubota, C.; Yoshimoto, Y.; Takeuchi, T.; Koshiishi, I.; Torii, S. Lipoxygenase-Mediated Generation of Lipid Peroxides Enhances Ferroptosis Induced by Erastin and RSL3. Cancer Sci. 2017, 108, 2187–2194. [Google Scholar] [CrossRef]
  59. Imam, M.; Ji, J.; Zhang, Z.; Yan, S. Targeting the Initiator to Activate both Ferroptosis and Cuproptosis for Breast Cancer Treatment: Progress and Possibility for Clinical Application. Front. Pharmacol. 2025, 15, 1493188. [Google Scholar] [CrossRef]
  60. Li, Z.; Chen, L.; Chen, C.; Zhou, Y.; Hu, D.; Yang, J.; Chen, Y.; Zhuo, W.; Mao, M.; Zhang, X.; et al. Targeting Ferroptosis in Breast Cancer. Biomark. Res. 2020, 8, 58. [Google Scholar] [CrossRef]
  61. Li, J.; He, D.; Li, S.; Xiao, J.; Zhu, Z. Ferroptosis: The Emerging Player in Remodeling Triple-Negative Breast Cancer. Front. Immunol. 2023, 14, 1284057. [Google Scholar] [CrossRef]
  62. Consoli, V.; Sorrenti, V.; Pittalà, V.; Greish, K.; D’Amico, A.G.; Romeo, G.; Intagliata, S.; Salerno, L.; Vanella, L. Heme Oxygenase Modulation Drives Ferroptosis in TNBC Cells. Int. J. Mol. Sci. 2022, 23, 5709. [Google Scholar] [CrossRef]
  63. Sen, U.; Coleman, C.; Sen, T. Stearoyl Coenzyme A Desaturase-1: Multitasker in Cancer, Metabolism, and Ferroptosis. Trends Cancer 2023, 9, 480–489. [Google Scholar] [CrossRef] [PubMed]
  64. Ackermann, T.; Shokry, E.; Deshmukh, R.; Anand, J.; Galbraith, L.C.A.; Mitchell, L.; Rodriguez-Blanco, G.; Villar, V.H.; Sterken, B.A.; Nixon, C.; et al. Breast Cancer Secretes Anti-Ferroptotic MUFAs and Depends on Selenoprotein Synthesis for Metastasis. EMBO Mol. Med. 2024, 16, 2749–2774. [Google Scholar] [CrossRef] [PubMed]
  65. Magtanong, L.; Ko, P.; To, M.; Cao, J.Y.; Forcina, G.C.; Tarangelo, A.; Ward, C.C.; Cho, K.; Patti, G.J.; Nomura, D.K.; et al. Exogenous Monounsaturated Fatty Acids Promote a Ferroptosis-Resistant Cell State. Cell Chem. Biol. 2019, 26, 420–432.e9. [Google Scholar] [CrossRef] [PubMed]
  66. Liang, D.; Feng, Y.; Zandkarimi, F.; Wang, H.; Zhang, Z.; Kim, J.; Cai, Y.; Gu, W.; Stockwell, B.R.; Jiang, X. Ferroptosis Surveillance Independent of GPX4 and Differentially Regulated by Sex Hormones. Cell 2023, 186, 2748–2764.e22. [Google Scholar] [CrossRef]
  67. Yang, Y.; Zhu, T.; Wang, X.; Xiong, F.; Hu, Z.; Qiao, X.; Yuan, X.; Wang, D. ACSL3 and ACSL4, Distinct Roles in Ferroptosis and Cancers. Cancers 2022, 14, 5896. [Google Scholar] [CrossRef]
  68. Lorito, N.; Subbiani, A.; Smiriglia, A.; Bacci, M.; Bonechi, F.; Tronci, L.; Romano, E.; Corrado, A.; Longo, D.L.; Iozzo, M.; et al. FADS1/2 Control Lipid Metabolism and Ferroptosis Susceptibility in Triple-Negative Breast Cancer. EMBO Mol. Med. 2024, 16, 1533–1559. [Google Scholar] [CrossRef]
  69. Danielli, M.; Perne, L.; Jarc Jovičić, E.; Petan, T. Lipid Droplets and Polyunsaturated Fatty Acid Trafficking: Balancing Life and Death. Front. Cell Dev. Biol. 2023, 11, 1104725, Erratum in Front. Cell Dev. Biol. 2023, 22, 1175493. [Google Scholar] [CrossRef]
  70. Xie, H.; Heier, C.; Kien, B.; Vesely, P.W.; Tang, Z.; Sexl, V.; Schoiswohl, G.; Strießnig-Bina, I.; Hoefler, G.; Zechner, R.; et al. Adipose Triglyceride Lipase Activity Regulates Cancer Cell Proliferation Via AMP-Kinase and mTOR Signaling. Biochim. Biophys. Acta-Mol. Cell Biol. Lipids 2020, 1865, 158737. [Google Scholar] [CrossRef]
  71. Lee, J.H.; Kong, J.; Jang, J.Y.; Han, J.S.; Ji, Y.; Lee, J.; Kim, J.B. Lipid Droplet Protein LID-1 Mediates ATGL-1-Dependent Lipolysis during Fasting in Caenorhabditis Elegans. Mol. Cell. Biol. 2014, 34, 4165–4176. [Google Scholar] [CrossRef] [PubMed]
  72. Dixon, S.J.; Olzmann, J.A. The Cell Biology of Ferroptosis. Nat. Rev. Mol. Cell Biol. 2024, 25, 424–442. [Google Scholar] [CrossRef] [PubMed]
  73. Kim, J.W.; Lee, J.; Oh, M.; Lee, E. An Integrated View of Lipid Metabolism in Ferroptosis Revisited Via Lipidomic Analysis. Exp. Mol. Med. 2023, 55, 1620–1631. [Google Scholar] [CrossRef] [PubMed]
  74. Li, Z.; Hu, Y.; Zheng, H.; Li, M.; Liu, Y.; Feng, R.; Li, X.; Zhang, S.; Tang, M.; Yang, M.; et al. LPCAT1-Mediated Membrane Phospholipid Remodelling Promotes Ferroptosis Evasion and Tumour Growth. Nat. Cell Biol. 2024, 26, 811–824. [Google Scholar] [CrossRef]
  75. Wan, M.; Pan, S.; Shan, B.; Diao, H.; Jin, H.; Wang, Z.; Wang, W.; Han, S.; Liu, W.; He, J.; et al. Lipid Metabolic Reprograming: The Unsung Hero in Breast Cancer Progression and Tumor Microenvironment. Mol. Cancer 2025, 24, 61. [Google Scholar] [CrossRef]
  76. Li, D.; Li, Y. The Interaction between Ferroptosis and Lipid Metabolism in Cancer. Signal Transduct. Target. Ther. 2020, 5, 108. [Google Scholar] [CrossRef]
  77. Lange, M.; Olzmann, J.A. Ending on a Sour Note: Lipids Orchestrate Ferroptosis in Cancer. Cell Metab. 2021, 33, 1507–1509. [Google Scholar] [CrossRef]
  78. Olzmann, J.A.; Carvalho, P. Dynamics and Functions of Lipid Droplets. Nat. Rev. Mol. Cell Biol. 2019, 20, 137–155. [Google Scholar] [CrossRef]
  79. Liu, Y.; Duan, C.; Dai, R.; Zeng, Y. Ferroptosis-Mediated Crosstalk in the Tumor Microenvironment Implicated in Cancer Progression and Therapy. Front. Cell Dev. Biol. 2021, 9, 739392. [Google Scholar] [CrossRef]
  80. Blanchette-Farra, N.; Kita, D.; Konstorum, A.; Tesfay, L.; Lemler, D.; Hegde, P.; Claffey, K.P.; Torti, F.M.; Torti, S.V. Contribution of Three-Dimensional Architecture and Tumor-Associated Fibroblasts to Hepcidin Regulation in Breast Cancer. Oncogene 2018, 37, 4013–4032. [Google Scholar] [CrossRef]
  81. Zhu, Y.; Fang, S.; Fan, B.; Xu, K.; Xu, L.; Wang, L.; Zhu, L.; Chen, C.; Wu, R.; Ni, J.; et al. Cancer-Associated Fibroblasts Reprogram Cysteine Metabolism to Increase Tumor Resistance to Ferroptosis in Pancreatic Cancer. Theranostics 2024, 14, 1683–1700. [Google Scholar] [CrossRef]
  82. Zhang, K.; Guo, L.; Li, X.; Hu, Y.; Luo, N. Cancer-Associated Fibroblasts Promote Doxorubicin Resistance in Triple-Negative Breast Cancer through Enhancing ZFP64 Histone Lactylation to Regulate Ferroptosis. J. Transl. Med. 2025, 23, 247. [Google Scholar] [CrossRef]
  83. Xie, Y.; Wang, B.; Zhao, Y.; Tao, Z.; Wang, Y.; Chen, G.; Hu, X. Mammary Adipocytes Protect Triple-Negative Breast Cancer Cells from Ferroptosis. J. Hematol. Oncol. 2022, 15, 72. [Google Scholar] [CrossRef]
  84. Balaban, S.; Shearer, R.F.; Lee, L.S.; van Geldermalsen, M.; Schreuder, M.; Shtein, H.C.; Cairns, R.; Thomas, K.C.; Fazakerley, D.J.; Grewal, T.; et al. Adipocyte Lipolysis Links Obesity to Breast Cancer Growth: Adipocyte-Derived Fatty Acids Drive Breast Cancer Cell Proliferation and Migration. Cancer Metab. 2017, 5, 1. [Google Scholar] [CrossRef]
  85. Urbanelli, L.; Buratta, S.; Logozzi, M.; Mitro, N.; Sagini, K.; Raimo, R.D.; Caruso, D.; Fais, S.; Emiliani, C. Lipidomic Analysis of Cancer Cells Cultivated at Acidic pH Reveals Phospholipid Fatty Acids Remodelling Associated with Transcriptional Reprogramming. J. Enzyme Inhib. Med. Chem. 2020, 35, 963–973. [Google Scholar] [CrossRef] [PubMed]
  86. Zhang, Q.; Deng, T.; Zhang, H.; Zuo, D.; Zhu, Q.; Bai, M.; Liu, R.; Ning, T.; Zhang, L.; Yu, Z.; et al. Adipocyte-Derived Exosomal MTTP Suppresses Ferroptosis and Promotes Chemoresistance in Colorectal Cancer. Adv. Sci. 2022, 9, e2203357. [Google Scholar] [CrossRef] [PubMed]
  87. Lu, P.; Bai, X.; Guo, L.; Tuoheti, K.; Zhan, S.; Liu, T. M1 Macrophage Inhibits Ferroptosis in Pseudomonas Aeruginosa-Induced Kidney Epithelial Cell Injury through the iNOS/ NO Pathway without Thiol. Front. Cell Dev. Biol. 2025, 13, 1597160. [Google Scholar] [CrossRef] [PubMed]
  88. Xu, X.; Zhang, Y.; Liao, C.; Zhou, H.; Wu, Y.; Zhang, W. Impact of Ferroptosis-Related Risk Genes on Macrophage M1/M2 Polarization and Prognosis in Glioblastoma. Front. Cell. Neurosci. 2024, 17, 1294029. [Google Scholar] [CrossRef]
  89. Gan, B. ACSL4, PUFA, and Ferroptosis: New Arsenal in Anti-Tumor Immunity. Signal Transduct. Target. Ther. 2022, 7, 128. [Google Scholar] [CrossRef]
  90. Liao, P.; Wang, W.; Wang, W.; Kryczek, I.; Li, X.; Bian, Y.; Sell, A.; Wei, S.; Grove, S.; Johnson, J.K.; et al. CD8(+) T Cells and Fatty Acids Orchestrate Tumor Ferroptosis and Immunity Via ACSL4. Cancer Cell 2022, 40, 365–378.e6. [Google Scholar] [CrossRef]
  91. Brown, C.W.; Amante, J.J.; Goel, H.L.; Mercurio, A.M. The α6β4 Integrin Promotes Resistance to Ferroptosis. J. Cell Biol. 2017, 216, 4287–4297. [Google Scholar] [CrossRef]
  92. Brown, C.W.; Amante, J.J.; Mercurio, A.M. Cell Clustering Mediated by the Adhesion Protein PVRL4 is Necessary for α6β4 Integrin-Promoted Ferroptosis Resistance in Matrix-Detached Cells. J. Biol. Chem. 2018, 293, 12741–12748. [Google Scholar] [CrossRef]
  93. Zhang, H.; Yang, F.; Xiao, Y.; Jiang, Y.; Shao, Z. Conceptualizing the Complexity of Ferroptosis to Treat Triple-Negative Breast Cancer: Theory-to-Practice. Cancer Biol. Med. 2023, 20, 98–103. [Google Scholar] [CrossRef]
  94. Tan, L.; Liu, J.; Ma, C.; Huang, S.; He, F.; Long, Y.; Zheng, Z.; Liang, J.; Xu, N.; Wang, G.; et al. Iron-Dependent Cell Death: Exploring Ferroptosis as a Unique Target in Triple-Negative Breast Cancer Management. Cancer Manag. Res. 2025, 17, 625–637. [Google Scholar] [CrossRef] [PubMed]
  95. Song, X.; Wang, X.; Liu, Z.; Yu, Z. Role of GPX4-Mediated Ferroptosis in the Sensitivity of Triple Negative Breast Cancer Cells to Gefitinib. Front. Oncol. 2020, 10, 597434. [Google Scholar] [CrossRef] [PubMed]
  96. Yang, F.; Xiao, Y.; Ding, J.; Jin, X.; Ma, D.; Li, D.; Shi, J.; Huang, W.; Wang, Y.; Jiang, Y.; et al. Ferroptosis Heterogeneity in Triple-Negative Breast Cancer Reveals an Innovative Immunotherapy Combination Strategy. Cell Metab. 2023, 35, 84–100.e8. [Google Scholar] [CrossRef] [PubMed]
  97. Hu, K.; Qiu, J.; Hu, Y.; Wang, Y.; Yu, C.; Wu, Y. Efficacy of FERscore in Predicting Sensitivity to Ferroptosis Inducers in Breast Cancer. NPJ Breast Cancer 2024, 10, 74. [Google Scholar] [CrossRef]
  98. Cao, J.; Zhou, T.; Wu, T.; Lin, R.; Huang, J.; Shi, D.; Yu, J.; Ren, Y.; Qian, C.; He, L.; et al. Targeting Estrogen-Regulated System X(C)(-) Promotes Ferroptosis and Endocrine Sensitivity of ER+ Breast Cancer. Cell Death Dis. 2025, 16, 30. [Google Scholar] [CrossRef]
  99. Herrera-Abreu, M.; Guan, J.; Khalid, U.; Ning, J.; Costa, M.R.; Chan, J.; Li, Q.; Fortin, J.; Wong, W.R.; Perampalam, P.; et al. Inhibition of GPX4 Enhances CDK4/6 Inhibitor and Endocrine Therapy Activity in Breast Cancer. Nat. Commun. 2024, 15, 9550. [Google Scholar] [CrossRef]
  100. Peng, Y.; Yu, H.; Zhang, Y.; Qu, F.; Tang, Z.; Qu, C.; Tian, J.; Zong, B.; Wang, Y.; Ren, H.; et al. A Ferroptosis-Associated Gene Signature for the Prediction of Prognosis and Therapeutic Response in Luminal-Type Breast Carcinoma. Sci. Rep. 2021, 11, 17610. [Google Scholar] [CrossRef]
  101. Hangauer, M.J.; Viswanathan, V.S.; Ryan, M.J.; Bole, D.; Eaton, J.K.; Matov, A.; Galeas, J.; Dhruv, H.D.; Berens, M.E.; Schreiber, S.L.; et al. Drug-Tolerant Persister Cancer Cells are Vulnerable to GPX4 Inhibition. Nature 2017, 551, 247–250. [Google Scholar] [CrossRef]
  102. Park, S.Y.; Jeong, K.J.; Poire, A.; Zhang, D.; Tsang, Y.H.; Blucher, A.S.; Mills, G.B. Irreversible HER2 Inhibitors Overcome Resistance to the RSL3 Ferroptosis Inducer in Non-HER2 Amplified Luminal Breast Cancer. Cell Death Dis. 2023, 14, 532. [Google Scholar] [CrossRef] [PubMed]
  103. Su, Z.; Liu, Y.; Wang, L.; Gu, W. Regulation of SLC7A11 as an Unconventional Checkpoint in Tumorigenesis through Ferroptosis. Genes Dis. 2024, 12, 101254. [Google Scholar] [CrossRef] [PubMed]
  104. Akiyama, H.; Carter, B.Z.; Andreeff, M.; Ishizawa, J. Molecular Mechanisms of Ferroptosis and Updates of Ferroptosis Studies in Cancers and Leukemia. Cells 2023, 12, 1128. [Google Scholar] [CrossRef]
  105. Yan, R.; Lin, B.; Jin, W.; Tang, L.; Hu, S.; Cai, R. NRF2, a Superstar of Ferroptosis. Antioxidants 2023, 12, 1739. [Google Scholar] [CrossRef]
  106. Sun, X.; Ou, Z.; Chen, R.; Niu, X.; Chen, D.; Kang, R.; Tang, D. Activation of the p62-Keap1-NRF2 Pathway Protects Against Ferroptosis in Hepatocellular Carcinoma Cells. Hepatology 2016, 63, 173–184. [Google Scholar] [CrossRef]
  107. Li, Y.; Yang, W.; Zheng, Y.; Dai, W.; Ji, J.; Wu, L.; Cheng, Z.; Zhang, J.; Li, J.; Xu, X.; et al. Targeting Fatty Acid Synthase Modulates Sensitivity of Hepatocellular Carcinoma to Sorafenib Via Ferroptosis. J. Exp. Clin. Cancer Res. 2023, 42, 6. [Google Scholar] [CrossRef]
  108. Fan, Y.; Zhang, R.; Wang, C.; Pan, M.; Geng, F.; Zhong, Y.; Su, H.; Kou, Y.; Mo, X.; Lefai, E.; et al. STAT3 Activation of SCAP-SREBP-1 Signaling Upregulates Fatty Acid Synthesis to Promote Tumor Growth. J. Biol. Chem. 2024, 300, 107351. [Google Scholar] [CrossRef]
  109. Chen, X.; Tsvetkov, A.S.; Shen, H.; Isidoro, C.; Ktistakis, N.T.; Linkermann, A.; Koopman, W.J.H.; Simon, H.; Galluzzi, L.; Luo, S.; et al. International Consensus Guidelines for the Definition, Detection, and Interpretation of Autophagy-Dependent Ferroptosis. Autophagy 2024, 20, 1213–1246. [Google Scholar] [CrossRef]
  110. Hadian, K. Ferroptosis Suppressor Protein 1 (FSP1) and Coenzyme Q10 Cooperatively Suppress Ferroptosis. Biochemistry 2020, 59, 637–638. [Google Scholar] [CrossRef]
  111. Porstmann, T.; Santos, C.R.; Griffiths, B.; Cully, M.; Wu, M.; Leevers, S.; Griffiths, J.R.; Chung, Y.; Schulze, A. SREBP Activity is Regulated by mTORC1 and Contributes to Akt-Dependent Cell Growth. Cell Metab. 2008, 8, 224–236. [Google Scholar] [CrossRef]
  112. Yi, J.; Zhu, J.; Wu, J.; Thompson, C.B.; Jiang, X. Oncogenic Activation of PI3K-AKT-mTOR Signaling Suppresses Ferroptosis Via SREBP-Mediated Lipogenesis. Proc. Natl. Acad. Sci. USA 2020, 117, 31189–31197. [Google Scholar] [CrossRef]
  113. Su, H.; Peng, C.; Liu, Y. Regulation of Ferroptosis by PI3K/Akt Signaling Pathway: A Promising Therapeutic Axis in Cancer. Front. Cell Dev. Biol. 2024, 12, 1372330. [Google Scholar] [CrossRef]
  114. Mitsuishi, Y.; Motohashi, H.; Yamamoto, M. The Keap1-Nrf2 System in Cancers: Stress Response and Anabolic Metabolism. Front. Oncol. 2012, 2, 200. [Google Scholar] [CrossRef] [PubMed]
  115. Ichimura, Y.; Waguri, S.; Sou, Y.; Kageyama, S.; Hasegawa, J.; Ishimura, R.; Saito, T.; Yang, Y.; Kouno, T.; Fukutomi, T.; et al. Phosphorylation of p62 Activates the Keap1-Nrf2 Pathway during Selective Autophagy. Mol. Cell 2013, 51, 618–631. [Google Scholar] [CrossRef] [PubMed]
  116. Cosialls, E.; Pacreau, E.; Duruel, C.; Ceccacci, S.; Elhage, R.; Desterke, C.; Roger, K.; Guerrera, C.; Ducloux, R.; Souquere, S.; et al. mTOR Inhibition Suppresses Salinomycin-Induced Ferroptosis in Breast Cancer Stem Cells by Ironing Out Mitochondrial Dysfunctions. Cell Death Dis. 2023, 14, 744. [Google Scholar] [CrossRef] [PubMed]
  117. Jiang, L.; Kon, N.; Li, T.; Wang, S.; Su, T.; Hibshoosh, H.; Baer, R.; Gu, W. Ferroptosis as a p53-Mediated Activity during Tumour Suppression. Nature 2015, 520, 57–62. [Google Scholar] [CrossRef]
  118. Ou, Y.; Wang, S.; Li, D.; Chu, B.; Gu, W. Activation of SAT1 Engages Polyamine Metabolism with p53-Mediated Ferroptotic Responses. Proc. Natl. Acad. Sci. USA 2016, 113, E6806–E6812. [Google Scholar] [CrossRef]
  119. Hu, W.; Zhang, C.; Wu, R.; Sun, Y.; Levine, A.; Feng, Z. Glutaminase 2, a Novel p53 Target Gene Regulating Energy Metabolism and Antioxidant Function. Proc. Natl. Acad. Sci. USA 2010, 107, 7455–7460. [Google Scholar] [CrossRef]
  120. Zhou, Q.; Meng, Y.; Li, D.; Yao, L.; Le, J.; Liu, Y.; Sun, Y.; Zeng, F.; Chen, X.; Deng, G. Ferroptosis in Cancer: From Molecular Mechanisms to Therapeutic Strategies. Signal Transduct. Target. Ther. 2024, 9, 55. [Google Scholar] [CrossRef]
  121. Lei, G.; Mao, C.; Horbath, A.D.; Yan, Y.; Cai, S.; Yao, J.; Jiang, Y.; Sun, M.; Liu, X.; Cheng, J.; et al. BRCA1-Mediated Dual Regulation of Ferroptosis Exposes a Vulnerability to GPX4 and PARP Co-Inhibition in BRCA1-Deficient Cancers. Cancer Discov. 2024, 14, 1476–1495. [Google Scholar] [CrossRef]
  122. Cahuzac, K.M.; Lubin, A.; Bosch, K.; Stokes, N.; Shoenfeld, S.M.; Zhou, R.; Lemon, H.; Asara, J.; Parsons, R.E. AKT Activation because of PTEN Loss Upregulates xCT Via GSK3β/NRF2, Leading to Inhibition of Ferroptosis in PTEN-Mutant Tumor Cells. Cell Rep. 2023, 42, 112536. [Google Scholar] [CrossRef] [PubMed]
  123. Lin, Y.; Dong, Y.; Liu, W.; Fan, X.; Sun, Y. Pan-Cancer Analyses Confirmed the Ferroptosis-Related Gene SLC7A11 as a Prognostic Biomarker for Cancer. Int. J. Gen. Med. 2022, 15, 2501–2513. [Google Scholar] [CrossRef] [PubMed]
  124. Wang, M.; Chen, J.; Lu, Y.; Bawcom, A.R.; Wu, J.; Ou, J.; Asara, J.M.; Armstrong, A.J.; Wang, Q.; Li, L.; et al. RB1-Deficient Prostate Tumor Growth and Metastasis are Vulnerable to Ferroptosis Induction Via the E2F/ACSL4 Axis. J. Clin. Investig. 2023, 133, e166647. [Google Scholar] [CrossRef] [PubMed]
  125. Lim, J.K.M.; Leprivier, G. The Impact of Oncogenic RAS on Redox Balance and Implications for Cancer Development. Cell Death Dis. 2019, 10, 955. [Google Scholar] [CrossRef]
  126. Fan, Z.; Wirth, A.; Chen, D.; Wruck, C.J.; Rauh, M.; Buchfelder, M.; Savaskan, N. Nrf2-Keap1 Pathway Promotes Cell Proliferation and Diminishes Ferroptosis. Oncogenesis 2017, 6, e371. [Google Scholar] [CrossRef]
  127. Xie, Y.; Lei, X.; Zhao, G.; Guo, R.; Cui, N. mTOR in Programmed Cell Death and its Therapeutic Implications. Cytokine Growth Factor Rev. 2023, 71–72, 66–81. [Google Scholar] [CrossRef]
  128. Drake, L.E.; Springer, M.Z.; Poole, L.P.; Kim, C.J.; Macleod, K.F. Expanding Perspectives on the Significance of Mitophagy in Cancer. Semin. Cancer Biol. 2017, 47, 110–124. [Google Scholar] [CrossRef]
  129. Woo, Y.; Lee, H.; Jung, Y.M.; Jung, Y. mTOR-Mediated Antioxidant Activation in Solid Tumor Radioresistance. J. Oncol. 2019, 2019, 5956867. [Google Scholar] [CrossRef]
  130. Su, F.; Koeberle, A. Regulation and Targeting of SREBP-1 in Hepatocellular Carcinoma. Cancer Metastasis Rev. 2024, 43, 673–708. [Google Scholar] [CrossRef]
  131. Liu, Y.; Gu, W. P53 in Ferroptosis Regulation: The New Weapon for the Old Guardian. Cell Death Differ. 2022, 29, 895–910. [Google Scholar] [CrossRef]
  132. Wang, S.; Li, D.; Ou, Y.; Jiang, L.; Chen, Y.; Zhao, Y.; Gu, W. Acetylation is Crucial for p53-Mediated Ferroptosis and Tumor Suppression. Cell Rep. 2016, 17, 366–373. [Google Scholar] [CrossRef]
  133. Murphy, M.E. Ironing Out how p53 Regulates Ferroptosis. Proc. Natl. Acad. Sci. USA 2016, 113, 12350–12352. [Google Scholar] [CrossRef] [PubMed]
  134. Suzuki, S.; Tanaka, T.; Poyurovsky, M.V.; Nagano, H.; Mayama, T.; Ohkubo, S.; Lokshin, M.; Hosokawa, H.; Nakayama, T.; Suzuki, Y.; et al. Phosphate-Activated Glutaminase (GLS2), a p53-Inducible Regulator of Glutamine Metabolism and Reactive Oxygen Species. Proc. Natl. Acad. Sci. USA 2010, 107, 7461–7466. [Google Scholar] [CrossRef] [PubMed]
  135. Mao, Y.; Jiang, P. The Crisscross between p53 and Metabolism in Cancer. Acta Biochim. Biophys. Sin. 2023, 55, 914–922. [Google Scholar] [CrossRef] [PubMed]
  136. Xu, R.; Wang, W.; Zhang, W. Ferroptosis and the Bidirectional Regulatory Factor p53. Cell Death Discov. 2023, 9, 197. [Google Scholar] [CrossRef]
  137. Sen, U.; Coleman, C.; Gandhi, N.; Jethalia, V.; Demircioglu, D.; Elliott, A.; Vanderwalde, A.M.; Hayatt, O.; de Stanchina, E.; Halmos, B.; et al. SCD1 Inhibition Blocks the AKT-NRF2-SLC7A11 Pathway to Induce Lipid Metabolism Remodeling and Ferroptosis Priming in Lung Adenocarcinoma. Cancer Res. 2025, 85, 2485–2503. [Google Scholar] [CrossRef]
  138. Patra, K.C.; Hay, N. The Pentose Phosphate Pathway and Cancer. Trends Biochem. Sci. 2014, 39, 347–354. [Google Scholar] [CrossRef]
  139. Choi, J.; Kim, E.; Koo, J.S. Expression of Pentose Phosphate Pathway-Related Proteins in Breast Cancer. Dis. Markers 2018, 2018, 9369358. [Google Scholar] [CrossRef]
  140. Mi, T.; Kong, X.; Chen, M.; Guo, P.; He, D. Inducing Disulfidptosis in Tumors:Potential Pathways and Significance. MedComm 2024, 5, e791. [Google Scholar] [CrossRef]
  141. Zhu, W.; Liu, Y.; Yu, Z.; Wang, H. SLC7A11-Mediated Cell Death Mechanism in Cancer: A Comparative Study of Disulfidptosis and Ferroptosis. Front. Cell Dev. Biol. 2025, 13, 1559423. [Google Scholar] [CrossRef] [PubMed]
  142. Li, X.; Zhu, D. Role of Disulfide Death in Cancer (Review). Oncol. Lett. 2024, 29, 55. [Google Scholar] [CrossRef] [PubMed]
  143. Song, X.; Liu, J.; Kuang, F.; Chen, X.; Zeh, H.J.; Kang, R.; Kroemer, G.; Xie, Y.; Tang, D. PDK4 Dictates Metabolic Resistance to Ferroptosis by Suppressing Pyruvate Oxidation and Fatty Acid Synthesis. Cell Rep. 2021, 34, 108767. [Google Scholar] [CrossRef] [PubMed]
  144. Li, M.; Chen, X.; Wang, X.; Wei, X.; Wang, D.; Liu, X.; Xu, L.; Batu, W.; Li, Y.; Guo, B.; et al. RSL3 Enhances the Antitumor Effect of Cisplatin on Prostate Cancer Cells Via Causing Glycolysis Dysfunction. Biochem. Pharmacol. 2021, 192, 114741. [Google Scholar] [CrossRef]
  145. Maldonado, R.; Talana, C.A.; Song, C.; Dixon, A.; Uehara, K.; Weichhaus, M. β-hydroxybutyrate does Not Alter the Effects of Glucose Deprivation on Breast Cancer Cells. Oncol. Lett. 2020, 21, 65. [Google Scholar] [CrossRef]
  146. Zhao, Y.; Li, M.; Yao, X.; Fei, Y.; Lin, Z.; Li, Z.; Cai, K.; Zhao, Y.; Luo, Z. HCAR1/MCT1 Regulates Tumor Ferroptosis through the Lactate-Mediated AMPK-SCD1 Activity and its Therapeutic Implications. Cell Rep. 2020, 33, 108487. [Google Scholar] [CrossRef]
  147. Subburayan, K.; Thayyullathil, F.; Pallichankandy, S.; Cheratta, A.R.; Galadari, S. Superoxide-Mediated Ferroptosis in Human Cancer Cells Induced by Sodium Selenite. Transl. Oncol. 2020, 13, 100843. [Google Scholar] [CrossRef]
  148. Xu, M.; Gao, X.; Yue, L.; Li, J.; Feng, X.; Huang, D.; Cai, H.; Qi, Y. Sensitivity of Triple Negative Breast Cancer Cells to ATM-Dependent Ferroptosis Induced by Sodium Selenite. Exp. Cell Res. 2024, 442, 114222. [Google Scholar] [CrossRef]
  149. Gao, J.; Ma, N.; Ni, S.; Han, X. Intersection of Ferroptosis and Nanomaterials Brings Benefits to Breast Cancer. Cell Biol. Toxicol. 2025, 41, 119. [Google Scholar] [CrossRef]
  150. Tosi, G.; Paoli, A.; Zuccolotto, G.; Turco, E.; Simonato, M.; Tosoni, D.; Tucci, F.; Lugato, P.; Giomo, M.; Elvassore, N.; et al. Cancer Cell Stiffening Via CoQ(10) and UBIAD1 Regulates ECM Signaling and Ferroptosis in Breast Cancer. Nat. Commun. 2024, 15, 8214. [Google Scholar] [CrossRef]
  151. Woldeselassie, M.; Tamene, A. Therapeutic Controversies Over use of Antioxidant Supplements during Cancer Treatment: A Scoping Review. Front. Nutr. 2024, 11, 1480780. [Google Scholar] [CrossRef] [PubMed]
  152. Kraft, V.A.N.; Bezjian, C.T.; 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. 2020, 6, 41–53. [Google Scholar] [CrossRef] [PubMed]
  153. Gao, Q.; Liu, T.; Sun, L.; Yao, Y.; Li, F.; Mao, L. Triggered Ferroptotic Albumin-Tocopherol Nanocarriers for Treating Drug-Resistant Breast Cancer. Front. Oncol. 2024, 14, 1464909. [Google Scholar] [CrossRef] [PubMed]
  154. Zeng, C.; Wang, J.; Zhao, S.; Wei, Y.; Qi, Y.; Liu, S.; Wang, Y.; Ge, H.; Yang, X.; Tan, Y.; et al. Multi-Cohort Validation of a Lipid Metabolism and Ferroptosis-Associated Index for Prognosis and Immunotherapy Response Prediction in Hormone Receptor-Positive Breast Cancer. Int. J. Biol. Sci. 2025, 21, 3968–3992. [Google Scholar] [CrossRef]
  155. Li, Y.; Li, T.; Zhai, D.; Xie, C.; Kuang, X.; Lin, Y.; Shao, N. Quantification of Ferroptosis Pathway Status Revealed Heterogeneity in Breast Cancer Patients with Distinct Immune Microenvironment. Front. Oncol. 2022, 12, 956999. [Google Scholar] [CrossRef]
  156. Glibetic, N.; Bowman, S.; Skaggs, T.; Weichhaus, M. The use of Patient-Derived Organoids in the Study of Molecular Metabolic Adaptation in Breast Cancer. Int. J. Mol. Sci. 2024, 25, 10503. [Google Scholar] [CrossRef]
  157. Lu, D.; Xia, B.; Feng, T.; Qi, G.; Ma, Z. The Role of Cancer Organoids in Ferroptosis, Pyroptosis, and Necroptosis: Functions and Clinical Implications. Biomolecules 2025, 15, 659. [Google Scholar] [CrossRef]
  158. Ye, L.; Zhong, F.; Sun, S.; Ou, X.; Yuan, J.; Zhu, J.; Zeng, Z. Tamoxifen Induces Ferroptosis in MCF-7 Organoid. J. Cancer Res. Ther. 2023, 19, 1627–1635. [Google Scholar] [CrossRef]
  159. Havas, K.M.; Milchevskaya, V.; Radic, K.; Alladin, A.; Kafkia, E.; Garcia, M.; Stolte, J.; Klaus, B.; Rotmensz, N.; Gibson, T.J.; et al. Metabolic Shifts in Residual Breast Cancer Drive Tumor Recurrence. J. Clin. Investig. 2017, 127, 2091–2105. [Google Scholar] [CrossRef]
  160. Desterke, C.; Xiang, Y.; Elhage, R.; Duruel, C.; Chang, Y.; Hamaï, A. Ferroptosis Inducers Upregulate PD-L1 in Recurrent Triple-Negative Breast Cancer. Cancers 2024, 16, 155. [Google Scholar] [CrossRef]
  161. Yu, L.; Huang, K.; Liao, Y.; Wang, L.; Sethi, G.; Ma, Z. Targeting Novel Regulated Cell Death: Ferroptosis, Pyroptosis and Necroptosis in Anti-PD-1/PD-L1 Cancer Immunotherapy. Cell Prolif. 2024, 57, e13644. [Google Scholar] [CrossRef]
  162. Wang, Z.; Li, R.; Hou, N.; Zhang, J.; Wang, T.; Fan, P.; Ji, C.; Zhang, B.; Liu, L.; Wang, Y.; et al. PRMT5 Reduces Immunotherapy Efficacy in Triple-Negative Breast Cancer by Methylating KEAP1 and Inhibiting Ferroptosis. J. Immunother. Cancer. 2023, 11, e006890. [Google Scholar] [CrossRef] [PubMed]
  163. Lin, S.; Huang, F.; Wu, R.; Xie, W.; Chen, M.; Dai, S.; Xu, W.; Zheng, W.; Tan, G. Toxicarioside H Induces Ferroptosis in Triple-Negative Breast Cancer Cells through Nrf2/HO-1 Pathway. Discov. Oncol. 2025, 16, 772. [Google Scholar] [CrossRef] [PubMed]
  164. Yao, X.; Xie, R.; Cao, Y.; Tang, J.; Men, Y.; Peng, H.; Yang, W. Simvastatin Induced Ferroptosis for Triple-Negative Breast Cancer Therapy. J. Nanobiotechnol. 2021, 19, 311. [Google Scholar] [CrossRef] [PubMed]
  165. Mu, Y.; Fan, Y.; He, L.; Hu, N.; Xue, H.; Guan, X.; Zheng, Z. Enhanced Cancer Immunotherapy through Synergistic Ferroptosis and Immune Checkpoint Blockade using Cell Membrane-Coated Nanoparticles. Cancer Nanotechnol. 2023, 14, 83. [Google Scholar] [CrossRef]
  166. Han, N.; Li, L.; Peng, X.; Ma, Q.; Yang, Z.; Wang, X.; Li, J.; Li, Q.; Yu, T.; Xu, H.; et al. Ferroptosis Triggered by Dihydroartemisinin Facilitates Chlorin E6 Induced Photodynamic Therapy Against Lung Cancer through Inhibiting GPX4 and Enhancing ROS. Eur. J. Pharmacol. 2022, 919, 174797. [Google Scholar] [CrossRef]
  167. Wang, H.; Qiao, C.; Guan, Q.; Wei, M.; Li, Z. Nanoparticle-Mediated Synergistic Anticancer Effect of Ferroptosis and Photodynamic Therapy: Novel Insights and Perspectives. Asian J. Pharm. Sci. 2023, 18, 100829. [Google Scholar] [CrossRef]
  168. Kojima, Y.; Tanaka, M.; Sasaki, M.; Ozeki, K.; Shimura, T.; Kubota, E.; Kataoka, H. Induction of Ferroptosis by Photodynamic Therapy and Enhancement of Antitumor Effect with Ferroptosis Inducers. J. Gastroenterol. 2024, 59, 81–94. [Google Scholar] [CrossRef]
  169. Luo, J.; Xu, L.; Feng, J.; Xu, K.; Tian, P.; Bai, X.; Xu, S.; Wen, L.; Lu, C.; Song, J. Tumor Microenvironment-Activated and ROS-Augmented Nanoplatform Amplified PDT Against Colorectal Cancer through Impairing GPX4 to Induce Ferroptosis. ACS Appl. Mater. Interfaces 2025, 17, 41586–41596. [Google Scholar] [CrossRef]
  170. Zhang, J.; Liu, L.; Li, X.; Shen, X.; Yang, G.; Deng, Y.; Hu, Z.; Zhang, J.; Lu, Y. 5-ALA-PDT Induced Ferroptosis in Keloid Fibroblasts Via ROS, Accompanied by Downregulation of xCT, GPX4. Photodiagn. Photodyn. Ther. 2023, 42, 103612. [Google Scholar] [CrossRef]
  171. Le, Y.; Yunjie, X.; Kim, J.S. Photo-Activated Ferroptosis for Cancer Therapy: Advances, Challenges, and Prospects. CCS Chem. 2023, 5, 1718–1736. [Google Scholar] [CrossRef]
  172. Mishchenko, T.A.; Balalaeva, I.V.; Vedunova, M.V.; Krysko, D.V. Ferroptosis and Photodynamic Therapy Synergism: Enhancing Anticancer Treatment. Trends Cancer. 2021, 7, 484–487. [Google Scholar] [CrossRef]
  173. Luo, J.; Deng, Y.; Lu, S.; Chen, S.; He, R.; Qin, D.; Chi, B.; Chen, G.; Yang, X.; Peng, W. Current Status and Future Directions of Ferroptosis Research in Breast Cancer: Bibliometric Analysis. Interact. J. Med. Res. 2025, 14, e66286. [Google Scholar] [CrossRef]
  174. Yang, X.; Wu, L.; Xu, S. An Overview of GPX4-Targeting TPDs for Cancer Therapy. Bioorg. Med. Chem. 2025, 118, 118046. [Google Scholar] [CrossRef]
  175. Guo, J.; Yan, Y.; Zhang, L.; Chen, H.; Zhang, W.; Yuan, H.; Lin, J.; Sun, Q.; Yan, L.; Wang, B.; et al. Dual Ferroptosis Induction in N2-TANs and TNBC Cells Via FTH1 Targeting: A Therapeutic Strategy for Triple-Negative Breast Cancer. Cell Rep. Med. 2025, 6, 101915. [Google Scholar] [CrossRef]
  176. Santana-Codina, N.; Mancias, J. The Role of NCOA4-Mediated Ferritinophagy in Health and Disease. Pharmaceuticals 2018, 11, 114. [Google Scholar] [CrossRef] [PubMed]
  177. Lei, G.; Zhuang, L.; Gan, B. The Roles of Ferroptosis in Cancer: Tumor Suppression, Tumor Microenvironment, and Therapeutic Interventions. Cancer Cell 2024, 42, 513–534. [Google Scholar] [CrossRef] [PubMed]
  178. Tang, Y.; Zhou, J.; Hooi, S.C.; Jiang, Y.; Lu, G. Fatty Acid Activation in Carcinogenesis and Cancer Development: Essential Roles of Long-Chain Acyl-CoA Synthetases. Oncol. Lett. 2018, 16, 1390–1396. [Google Scholar] [CrossRef]
  179. Ingold, I.; Berndt, C.; Schmitt, S.; Doll, S.; Poschmann, G.; Buday, K.; Roveri, A.; Peng, X.; Porto Freitas, F.; Seibt, T.; et al. Selenium Utilization by GPX4 is Required to Prevent Hydroperoxide-Induced Ferroptosis. Cell 2018, 172, 409–422.e21. [Google Scholar] [CrossRef] [PubMed]
  180. Ito, J.; Nakamura, T.; Toyama, T.; Chen, D.; Berndt, C.; Poschmann, G.; Mourão, A.S.D.; Doll, S.; Suzuki, M.; Zhang, W.; et al. PRDX6 Dictates Ferroptosis Sensitivity by Directing Cellular Selenium Utilization. Mol. Cell 2024, 84, 4629–4644.e9. [Google Scholar] [CrossRef]
  181. Conrad, M.; Proneth, B. Selenium: Tracing another Essential Element of Ferroptotic Cell Death. Cell Chem. Biol. 2020, 27, 409–419. [Google Scholar] [CrossRef]
  182. Dai, Q.; Wei, X.; Zhao, J.; Zhang, D.; Luo, Y.; Yang, Y.; Xiang, Y.; Liu, X. Inhibition of FSP1: A New Strategy for the Treatment of Tumors (Review). Oncol. Rep. 2024, 52, 105. [Google Scholar] [CrossRef]
  183. Koppula, P.; Lei, G.; Zhang, Y.; Yan, Y.; Mao, C.; Kondiparthi, L.; Shi, J.; Liu, X.; Horbath, A.; Das, M.; et al. A Targetable CoQ-FSP1 Axis Drives Ferroptosis- and Radiation-Resistance in KEAP1 Inactive Lung Cancers. Nat. Commun. 2022, 13, 2206. [Google Scholar] [CrossRef]
  184. Li, W.; Liang, L.; Liu, S.; Yi, H.; Zhou, Y. FSP1: A Key Regulator of Ferroptosis. Trends Mol. Med. 2023, 29, 753–764. [Google Scholar] [CrossRef]
  185. Martinez, S.; Sentis, S.; Poulard, C.; Trédan, O.; Le Romancer, M. Role of PRMT1 and PRMT5 in Breast Cancer. Int. J. Mol. Sci. 2024, 25, 8854. [Google Scholar] [CrossRef] [PubMed]
  186. Chen, D.; Liang, C.; Qu, X.; Zhang, T.; Mou, X.; Cai, Y.; Wang, W.; Shao, J.; Dong, X. Metal-Free Polymer Nano-Photosensitizer Actuates Ferroptosis in Starved Cancer. Biomaterials 2023, 292, 121944. [Google Scholar] [CrossRef] [PubMed]
  187. Szurko, A.; Rams-Baron, M.; Montforts, F.; Bauer, D.; Kozub, P.; Gubernator, J.; Altmann, S.; Stanek, A.; Sieroń, A.; Ratuszna, A. Photodynamic Performance of Amphiphilic Chlorin E6 Derivatives with Appropriate Properties: A Comparison between Different-Type Liposomes as Delivery Systems. Photodiagn. Photodyn. Ther. 2020, 30, 101799. [Google Scholar] [CrossRef] [PubMed]
  188. Sokol, K.H.; Lee, C.J.; Rogers, T.J.; Waldhart, A.; Ellis, A.E.; Madireddy, S.; Daniels, S.R.; House, R.J.; Ye, X.; Olesnavich, M.; et al. Lipid Availability Influences Ferroptosis Sensitivity in Cancer Cells by Regulating Polyunsaturated Fatty Acid Trafficking. Cell Chem. Biol. 2025, 32, 408–422.e6. [Google Scholar] [CrossRef]
  189. Wei, X.; Li, Y.; Chen, H.; Gao, R.; Ning, P.; Wang, Y.; Huang, W.; Chen, E.; Fang, L.; Guo, X.; et al. A Lysosome-Targeted Magnetic Nanotorquer Mechanically Triggers Ferroptosis for Breast Cancer Treatment. Adv. Sci. 2024, 11, e2302093. [Google Scholar] [CrossRef]
  190. Lorito, N.; Bacci, M.; Smiriglia, A.; Mannelli, M.; Parri, M.; Comito, G.; Ippolito, L.; Giannoni, E.; Bonechi, M.; Benelli, M.; et al. Glucose Metabolic Reprogramming of ER Breast Cancer in Acquired Resistance to the CDK4/6 Inhibitor Palbociclib. Cells 2020, 9, 668. [Google Scholar] [CrossRef]
Figure 1. Schematic overview of metabolic pathways regulating ferroptosis in breast cancer. The diagram illustrates key alterations in iron metabolism (top left), glucose metabolism (top right), amino acid metabolism (bottom left), and lipid metabolism (bottom right) that influence ferroptosis susceptibility. Upregulated (↑) and downregulated (↓) genes/proteins are indicated, contributing to increased iron accumulation, enhanced antioxidant defenses, altered glucose utilization, and promoted lipid peroxidation, collectively modulating cell death resistance in breast cancer. Abbreviations: ACSL4, acyl-CoA synthetase long-chain family member 4; ALOX15, arachidonate 15-lipoxygenase; CBS, cystathionine beta-synthase; CD44v, CD44 variant; DMT1, divalent metal transporter 1; FSP1, ferroptosis suppressor protein 1; G6PD, glucose-6-phosphate dehydrogenase; GPX4, glutathione peroxidase 4; GSH, glutathione; HERC2, HECT and RLD domain containing E3 ubiquitin protein ligase 2; LPCAT3, lysophosphatidylcholine acyltransferase 3; MUC1-C, mucin 1 C-terminal subunit; NRF2, nuclear factor erythroid 2-related factor 2; PKM2, pyruvate kinase M2; SCD1, stearoyl-CoA desaturase 1; SLC11A2, solute carrier family 11 member 2; SLC7A11, solute carrier family 7 member 11; TF, transferrin; TFR1, transferrin receptor 1; xCT, cystine/glutamate antiporter.
Figure 1. Schematic overview of metabolic pathways regulating ferroptosis in breast cancer. The diagram illustrates key alterations in iron metabolism (top left), glucose metabolism (top right), amino acid metabolism (bottom left), and lipid metabolism (bottom right) that influence ferroptosis susceptibility. Upregulated (↑) and downregulated (↓) genes/proteins are indicated, contributing to increased iron accumulation, enhanced antioxidant defenses, altered glucose utilization, and promoted lipid peroxidation, collectively modulating cell death resistance in breast cancer. Abbreviations: ACSL4, acyl-CoA synthetase long-chain family member 4; ALOX15, arachidonate 15-lipoxygenase; CBS, cystathionine beta-synthase; CD44v, CD44 variant; DMT1, divalent metal transporter 1; FSP1, ferroptosis suppressor protein 1; G6PD, glucose-6-phosphate dehydrogenase; GPX4, glutathione peroxidase 4; GSH, glutathione; HERC2, HECT and RLD domain containing E3 ubiquitin protein ligase 2; LPCAT3, lysophosphatidylcholine acyltransferase 3; MUC1-C, mucin 1 C-terminal subunit; NRF2, nuclear factor erythroid 2-related factor 2; PKM2, pyruvate kinase M2; SCD1, stearoyl-CoA desaturase 1; SLC11A2, solute carrier family 11 member 2; SLC7A11, solute carrier family 7 member 11; TF, transferrin; TFR1, transferrin receptor 1; xCT, cystine/glutamate antiporter.
Ijms 26 09686 g001
Figure 2. Schematic representation of ferroptosis inhibitor RSL3 actions on glycose metabolism pathways modulating ferroptosis sensitivity in breast cancer cells. The diagram highlights glycolysis (pink), TCA cycle (blue), pentose phosphate pathway (PPP, purple), glutaminolysis (orange), and the GSH-GPX4 antioxidant pathway (red), illustrating their interplay in ferroptosis regulation. Key enzymes (e.g., HK2, PFK, PKM2, LDH1/2, IDH1/2, G6PD, GR, GPX4) and metabolites (e.g., NADPH, GSH, GSSG, lipid ROS) are shown, with arrows indicating activation or inhibition. RSL3 inhibits ferroptosis by targeting the xCT-Glutathione Axis (purple), reducing cysteine uptake and glutathione synthesis under low glucose and high xCT (SLC7A11) conditions. Abbreviations: α-KG, alpha-ketoglutarate; CDC25A, cell division cycle 25A; GDH, glutamate dehydrogenase; GLS1, glutaminase 1; MCT1, monocarboxylate transporter 1; PDK4, pyruvate dehydrogenase kinase 4.
Figure 2. Schematic representation of ferroptosis inhibitor RSL3 actions on glycose metabolism pathways modulating ferroptosis sensitivity in breast cancer cells. The diagram highlights glycolysis (pink), TCA cycle (blue), pentose phosphate pathway (PPP, purple), glutaminolysis (orange), and the GSH-GPX4 antioxidant pathway (red), illustrating their interplay in ferroptosis regulation. Key enzymes (e.g., HK2, PFK, PKM2, LDH1/2, IDH1/2, G6PD, GR, GPX4) and metabolites (e.g., NADPH, GSH, GSSG, lipid ROS) are shown, with arrows indicating activation or inhibition. RSL3 inhibits ferroptosis by targeting the xCT-Glutathione Axis (purple), reducing cysteine uptake and glutathione synthesis under low glucose and high xCT (SLC7A11) conditions. Abbreviations: α-KG, alpha-ketoglutarate; CDC25A, cell division cycle 25A; GDH, glutamate dehydrogenase; GLS1, glutaminase 1; MCT1, monocarboxylate transporter 1; PDK4, pyruvate dehydrogenase kinase 4.
Ijms 26 09686 g002
Figure 3. Schematic representation of the interplay between ferroptosis and immunotherapy in luminal androgen receptor (LAR) triple-negative breast cancer (TNBC). The diagram illustrates how ferroptosis inducers (Erastin, FTH1 inhibitors, PRMT5 inhibitors) and GSH depletion trigger ROS accumulation and lipid peroxidation, mediated by GSH/GPX4, KEAP, and NRF2 pathways. Immune checkpoint inhibitors (ICIs) sensitize ferroptosis resistance in LAR TNBC, enhancing tumor-associated macrophage (TAM) activity and immunogenic cell death (ICD) through IDO1, TNFAIP3, IFN1, and HMOX1 signaling. This activates CD8+ T cells and PD-1/PD-L1 targeting (Atezolizumab), promoting immune therapy efficacy via PD-L1 targeting, ICD, and T cell infiltration. Abbreviations: FTH1, ferritin heavy chain 1; GSH, glutathione; GPX4, glutathione peroxidase 4; ICD, immunogenic cell death; IDO1, indoleamine 2,3-dioxygenase 1; KEAP, Kelch-like ECH-associated protein; NRF2, nuclear factor erythroid 2-related factor 2; PRMT5, protein arginine methyltransferase 5; ROS, reactive oxygen species; TAM, tumor-associated macrophage; TNFAIP3, tumor necrosis factor alpha-induced protein 3.
Figure 3. Schematic representation of the interplay between ferroptosis and immunotherapy in luminal androgen receptor (LAR) triple-negative breast cancer (TNBC). The diagram illustrates how ferroptosis inducers (Erastin, FTH1 inhibitors, PRMT5 inhibitors) and GSH depletion trigger ROS accumulation and lipid peroxidation, mediated by GSH/GPX4, KEAP, and NRF2 pathways. Immune checkpoint inhibitors (ICIs) sensitize ferroptosis resistance in LAR TNBC, enhancing tumor-associated macrophage (TAM) activity and immunogenic cell death (ICD) through IDO1, TNFAIP3, IFN1, and HMOX1 signaling. This activates CD8+ T cells and PD-1/PD-L1 targeting (Atezolizumab), promoting immune therapy efficacy via PD-L1 targeting, ICD, and T cell infiltration. Abbreviations: FTH1, ferritin heavy chain 1; GSH, glutathione; GPX4, glutathione peroxidase 4; ICD, immunogenic cell death; IDO1, indoleamine 2,3-dioxygenase 1; KEAP, Kelch-like ECH-associated protein; NRF2, nuclear factor erythroid 2-related factor 2; PRMT5, protein arginine methyltransferase 5; ROS, reactive oxygen species; TAM, tumor-associated macrophage; TNFAIP3, tumor necrosis factor alpha-induced protein 3.
Ijms 26 09686 g003
Figure 4. Schematic illustration of the interplay between photodynamic therapy (PDT) and ferroptosis in cancer cells. The diagram depicts how PDT, utilizing Ce6 or IrFc1 photosensitizers (blue), generates reactive oxygen species (ROS) that enhance ferroptosis (purple arrows). Key processes include the conversion of Fe3+ to Fe2+, production of hydroxyl radicals (•OH), and depletion of GSH/GPX4, leading to lipid peroxidation and ROS accumulation (red). Iron-dependent reactions amplify PDT efficacy, creating a feedback loop that boosts ferroptosis-mediated cell death. Abbreviations: GSH, glutathione; GPX4, glutathione peroxidase 4; ROS, reactive oxygen species.
Figure 4. Schematic illustration of the interplay between photodynamic therapy (PDT) and ferroptosis in cancer cells. The diagram depicts how PDT, utilizing Ce6 or IrFc1 photosensitizers (blue), generates reactive oxygen species (ROS) that enhance ferroptosis (purple arrows). Key processes include the conversion of Fe3+ to Fe2+, production of hydroxyl radicals (•OH), and depletion of GSH/GPX4, leading to lipid peroxidation and ROS accumulation (red). Iron-dependent reactions amplify PDT efficacy, creating a feedback loop that boosts ferroptosis-mediated cell death. Abbreviations: GSH, glutathione; GPX4, glutathione peroxidase 4; ROS, reactive oxygen species.
Ijms 26 09686 g004
Table 1. Summary of Glutathione and Cysteine Metabolism in Breast Cancer Ferroptosis Regulation.
Table 1. Summary of Glutathione and Cysteine Metabolism in Breast Cancer Ferroptosis Regulation.
Pathway/MechanismClinical StatusCritical Gaps/ParadoxesKey References
GPX4-GSH AxisRSL3: preclinical only;
hepatotoxicity at therapeutic doses
TNBC shows both xCT/GPX4 addiction AND high ferroptosis sensitivity;
mechanism unclear
[11,17,18,19,20,21,42]
Cystine ImportErastin: preclinical;
poor bioavailability
TAM-supplied cysteine may override cell-autonomous targeting[22,23,24,25,26,43,44,45]
TranssulfurationNo CBS inhibitors in developmentBoth import and synthesis can be simultaneously upregulated[27,28,29,30,46]
Lysosomal StorageCysRx: proof-of-concept onlyEnhancing lysosomal cystine paradoxically increases ferroptosis[31,32]
Alternative GPX4-IndependentNo FSP1 inhibitors in trialsRelative contribution vs. GPX4 pathway undefined in breast cancer[33,34,35,36,37]
Nucleotide CompetitionNo selective RNR modulatorsCannot target without affecting normal cell proliferation[38,39,40,41]
Table 2. TNBC is heterogeneous, with subtypes showing varying ferroptosis responses [96].
Table 2. TNBC is heterogeneous, with subtypes showing varying ferroptosis responses [96].
TNBC SubtypeFerroptosis SensitivityKey Regulatory Features
Luminal Androgen Receptor (LAR)HighUpregulated GPX4, OxPE, glutathione metabolism
Mesenchymal (MES)ModerateEnriched iron metabolism, low FA/ROS activity
Immunomodulatory (IM) and Basal-Like Immune-Suppressed (BLIS)LowMinimal ferroptosis features, other cell death pathways dominant
Table 3. Oncogene and Tumor Suppressor Regulation of Ferroptosis in Breast Cancer.
Table 3. Oncogene and Tumor Suppressor Regulation of Ferroptosis in Breast Cancer.
Gene/
Pathway
CategoryEffect on
Ferroptosis
Primary MechanismsClinical RelevanceKey References
RASOncogeneStrong ResistanceETS1→xCT upregulation
NRF2 activation; FASN-HIF1α→MUFA
FSP1 induction
Common in aggressive BC[103,104,105,106,107,108,109,110,125,126]
mTORC1OncogeneContext-dependentSREBP1-SCD1→MUFA
Suppresses ferritinophagy
p62-KEAP1-NRF2
Glutaminolysis paradox
Activated in most BC[111,112,113,114,115,116,127,128,129,130]
p53Tumor SuppressorPromotesDirect xCT repression
SAT1→ALOX15
GLS2→glutamine depletion
Acetylation-dependent
Lost in 30% BC[39,117,118,119,131,132,133,134,135,136]
BRCA1Tumor SuppressorDifferentialErastin resistance
GPX4 inhibitor sensitive
VDAC3-dependent
5–10% hereditary BC[121]
PTENTumor SuppressorLoss→ResistanceAKT→GSK3β→NRF2
xCT upregulation
Pan-cancer mechanism
Lost in 30–40% BC[122,123,137]
RBTumor SuppressorLoss→SensitivityE2F→ACSL4 upregulation
Increased PUFA incorporation
Lost in TNBC[124]
HER2OncogeneContext-dependentBaseline resistance
Sensitivity when inhibited
Amplified 15–20% BC[101,102]
ERNuclear ReceptorResistanceMBOAT1 upregulation
ELOVL2→AdA synthesis
Positive in 70% BC[48,66,98]
Table 4. Ferroptosis-Targeting Therapeutic Strategies in Breast Cancer.
Table 4. Ferroptosis-Targeting Therapeutic Strategies in Breast Cancer.
StrategyTarget/
Mechanism
Representative AgentsDevelopment StatusEfficacyMajor LimitationsKey References
GPX4 Direct InhibitionGPX4 enzymatic activityRSL3
ML162
FIN56
Preclinical only60–90% growth inhibition in vitroPoor bioavailability (t½ < 2 h), hepatotoxicity[19,42,95,101,174]
Cystine Import BlockadeSystem xc (SLC7A11)Erastin
IKE
Sulfasalazine
Preclinical;
Sulfasalazine FDA-approved
Variable (30–70% inhibition)Poor solubility, compensatory CBS upregulation[22,24,26,28,43]
Iron Overload/FerritinophagyNCOA4
FTH1
iron import
Sorafenib-NPs
CT-1
Salinomycin
Preclinical70–80% tumor reductionSystemic iron toxicity[15,16,175,176]
ACSL4 ModulationPUFA incorporationNo specific inhibitorsTarget identifiedContext-dependentDual role, no selective agents[52,54,56,67,177,178]
SCD1 InhibitionMUFA synthesis blockadeA939572
CAY10566
PreclinicalSensitizes to ferroptosisNormal tissue toxicity[63,64,137]
Selenium ManipulationGPX4 cofactorSodium selenitePreclinicalTNBC-specific toxicityNarrow therapeutic window[147,148,179,180,181]
FSP1 InhibitionCoQ10-ubiquinol systemiFSP1Early preclinicalLimited single-agentRedundancy with GPX4[35,36,149,182,183,184]
Immunotherapy CombinationPD-1/PD-L1 + ferroptosisRSL3/Erastin + ICIsPreclinical60–90% tumor inhibitionT-cell toxicity[160,161,162,165]
PRMT5 InhibitionKEAP1 methylationGSK3326595Phase I (not BC)60–80% with anti-PD-1Not ferroptosis-specific[162,185]
PDT CombinationROS + ferroptosisCe6/PpIX + lightPreclinical>95% local controlLimited penetration[166,167,168,171,186,187]
Lipid/Metabolic InterventionLipid availability, glucoseDietary
GLUT inhibitors
Observational/PreclinicalContext-dependentPatient compliance[69,141,145,188]
Nanoparticle DeliveryTargeted deliveryFe3+-NPs
glutathione-scavenging
Preclinical90% tumor inhibitionManufacturing complexity[45,149,165,189]
Hormone Therapy CombinationER/HER2 + ferroptosisTamoxifen/Lapatinib + GPX4iProof of conceptEnhanced in resistantRequires resistance[98,99,101,102,190]
UBIAD1/CoQ10 ExploitationAntioxidant paradoxNo specific agentsConcept onlyUnknownMechanism unclear[150,151]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Glibetic, N.; Weichhaus, M. Metabolic Regulation of Ferroptosis in Breast Cancer. Int. J. Mol. Sci. 2025, 26, 9686. https://doi.org/10.3390/ijms26199686

AMA Style

Glibetic N, Weichhaus M. Metabolic Regulation of Ferroptosis in Breast Cancer. International Journal of Molecular Sciences. 2025; 26(19):9686. https://doi.org/10.3390/ijms26199686

Chicago/Turabian Style

Glibetic, Natalija, and Michael Weichhaus. 2025. "Metabolic Regulation of Ferroptosis in Breast Cancer" International Journal of Molecular Sciences 26, no. 19: 9686. https://doi.org/10.3390/ijms26199686

APA Style

Glibetic, N., & Weichhaus, M. (2025). Metabolic Regulation of Ferroptosis in Breast Cancer. International Journal of Molecular Sciences, 26(19), 9686. https://doi.org/10.3390/ijms26199686

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop