International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
Endometrial cancer (EC), a malignancy originating from the uterine endometrium, exhibits a rising global incidence, with >417,000 new cases and 97,000 deaths reported worldwide in 2022 alone (1). This trend is largely attributable to the increasing prevalence of risk factors such as obesity and metabolic syndrome (1–3). Therapeutically, EC management has been refined by the molecular classification established by The Cancer Genome Atlas (TCGA), which categorizes EC into four subtypes [DNA polymerase ε (POLE) ultramutated, microsatellite instability-high (MSI-H), copy-number low and copy-number high], each with distinct prognoses and therapeutic implications (4–6). This classification provides a critical basis for personalized therapy (7,8). Surgical intervention remains the cornerstone of EC management. Chemotherapy is mainly reserved for cases with advanced or recurrent disease, as well as for patients with high-risk pathological features post-surgery. Nevertheless, chemoresistance development presents a major therapeutic challenge (9), leading to treatment failure and mortality in >90% of patients with advanced-stage disease. A notable clinical limitation is the lack of effective prognostic biomarkers and specific targets to overcome chemoresistance (10). Dysregulation of regulated cell death (RCD) mechanisms is a recognized cancer hallmark, contributing to tumor progression and drug resistance. Emerging evidence has indicated novel RCD pathways, including autophagy, ferroptosis and pyroptosis, as potential therapeutic targets to enhance therapeutic efficacy and reverse chemoresistance in EC (11–16).
Reactive oxygen species (ROS), primarily generated via mitochondrial electron transport chain activity (17,18), NADPH oxidases (19) and exogenous stressors, exhibit a dual role in cancer initiation, progression, suppression and therapy (20). Oxidative stress, defined as an imbalance between ROS production and elimination, is a critical pathogenic factor in numerous chronic diseases, including cancer (21), cardiovascular diseases (22,23), metabolic disorders (24) and neurodegenerative conditions (such as Alzheimer's and Parkinson's diseases) (25). Depending on their levels and cellular context, ROS can promote tumorigenesis by inducing DNA damage, epithelial-mesenchymal transition (EMT) and immune evasion, or suppress tumor growth by triggering cell death pathways such as apoptosis and ferroptosis (20,26).
Ferroptosis is an iron-dependent form of RCD marked by iron accumulation and extensive lipid peroxidation. Key morphological characteristics include condensed cellular membranes, with increased density and reduction, or loss of mitochondrial cristae (27). Ferroptosis involves disruption of redox homeostasis, marked by depleted antioxidants [such as glutathione (GSH) and GSH peroxidase 4 (GPX4)] and accumulation of oxidants (such as Fe2+ and lipid ROS) (28). As primary intracellular ROS sources, mitochondria are intrinsically linked to ferroptosis. Mitophagy, a selective autophagic process that degrades damaged mitochondria via ubiquitin (Ub)-dependent or -independent pathways, serves as a protective mechanism against stress and is crucial for mitochondrial quality control (29–32). Mitophagy and ferroptosis, as two distinct forms of RCD, have been increasingly implicated in EC (11,16,33,34). Concurrently, oxidative stress, a key regulator of cellular metabolism, is closely linked to EC progression (35,36). The triad of mitophagy, ferroptosis and oxidative stress forms a dynamic equilibrium, collectively contributing to the pathogenesis of diverse diseases through their interconnected regulatory networks (37,38). The present review aims to delineate the role of oxidative stress as a central hub integrating mitophagy and ferroptosis in EC pathogenesis, offering novel perspectives for its prevention and treatment.
ROS, including superoxide anions, free radicals and hydrogen peroxide, are highly reactive molecules generated primarily through mitochondrial electron transport and NADPH oxidase activity (16–19,38,39). Additional sources include peroxisomal metabolism and endoplasmic reticulum stress (40,41). Oxidative stress occurs due to a disruption in the balance between ROS generation and clearance, serving a notable role in the development of cancer (21–23). Cellular protection against ROS involves a multi-level defense system, including enzymatic antioxidants (such as superoxide dismutase, catalase and GSH peroxidase), non-enzymatic scavengers (such as vitamin C, vitamins E and GSH) and repair systems for oxidized biomolecules, which collectively maintain redox homeostasis (39).
Physiological ROS levels regulate essential cellular processes (including proliferation, differentiation, survival and apoptosis). However, supraphysiological ROS levels drive tumorigenesis and progression (42,43). Excessive endogenous or exogenous ROS levels induce direct DNA damage and impair DNA repair mechanisms, causing mutations that inactivate tumor suppressors (such as p53) or activate oncogenes (such as KRAS) (44–46).
ROS further promote malignancy by activating proliferative signaling (including the PI3K/AKT/mTOR and MAPK pathways), and suppressing antitumor immunity via impaired T-cell differentiation/activation, T-cell death, natural killer cell dysfunction and M2 macrophage polarization/recruitment (47,48). ROS facilitate invasion and metastasis through EMT, and induce EMT-associated cytoskeletal rearrangement (Rho GTPase-dependent), extracellular matrix degradation (matrix metalloproteinase-dependent) and angiogenesis [hypoxia-inducible factor (HIF)-dependent] (49).
Although high ROS levels are crucial in promoting tumor initiation and progression, excessive ROS levels also serve an antioncogenic role in cancer (26,50). Excessively accumulated ROS repress cancer cell growth by inhibiting cancer cell proliferation through disrupting nucleotide/ATP synthesis and inducing cell cycle arrest (51). ROS also trigger tumor cell death by activating endoplasmic reticulum stress-mediated, mitochondrial and p53-dependent apoptotic pathways, along with the ferroptosis pathway (26). Therefore, targeting the regulation of ROS levels (for example, using pro-oxidant agents or inhibiting antioxidant pathways) has emerged as a novel therapeutic strategy in cancer treatment, aiming to exploit the ‘dual role’ of oxidative stress to achieve selective elimination of tumor cells.
Mitophagy represents a selective type of autophagy that functions as a protective response to intracellular and extracellular stress signals (32). Mitophagy maintains mitochondrial and cellular homeostasis by eliminating dysfunctional mitochondria, including depolarized, damaged or superfluous organelles, through lysosomal degradation. The core molecular mechanisms can be divided into Ub-dependent pathways and Ub-independent pathways (31). The PTEN-induced kinase 1 (PINK1)/Parkin pathway represents the most well-characterized Ub-dependent mechanism. Upon mitochondrial depolarization or damage, PINK1 stabilizes on the outer mitochondrial membrane (OMM), and phosphorylates Ub and the E3 Ub ligase Parkin, leading to the ubiquitination of OMM proteins. Autophagy receptors such as p62/sequestosome 1 (SQSTM1) and optineurin then recognize ubiquitinated substrates and link them to microtubule-associated proteins 1A/1B light chain 3B (LC3)-positive autophagosomal membranes for engulfment (31). Alternatively, Ub-independent pathways utilize OMM-resident receptors, including BCL2/adenovirus E1B 19 kDa protein-interacting protein 3 (BNIP3), BNIP3L (also known as NIX) and FUN14 domain-containing protein 1 (FUNDC1), which directly interact with LC3/GABA type A receptor-associated protein via LC3-interacting regions to initiate mitophagy. These coordinated mechanisms ensure the precise elimination of dysfunctional mitochondria, and their dysregulation is broadly implicated in various pathological states, including cancer (30,52,53).
Mitophagy is widely reported to exhibit aberrant activity levels in various cancer types compared with under normal physiological conditions (54,55). Current research has revealed its dual role in cancer biology, demonstrating both tumor-promoting and tumor-suppressive functions depending on the cellular context (56,57). In its pro-tumorigenic capacity, mitophagy is known to enable cancer cell survival under stress and facilitates malignant progression by clearing dysfunctional mitochondria (58). Under hypoxia, mitophagy mediated by BNIP3 and NIX is known to enhance tumor aggressiveness by lowering mitochondrial ROS levels, stabilizing HIF-1α, promoting glycolytic shift and inducing EMT, which collectively increase invasiveness and metastatic potential (59). Chemotherapy- or radiotherapy-induced mitochondrial damage can activate the PINK1/Parkin pathway, which ubiquitinates damaged mitochondria to block cytochrome c release, thereby potentially enabling cancer cells to evade apoptosis and develop therapeutic resistance (60). During metastasis, FUNDC1-dependent mitophagy is suggested to optimize energy metabolism to fuel ATP production, supporting cancer cell migration and invasion (61). Additionally, mitophagy may suppress NLR family pyrin domain containing 3 (NLRP3) inflammasome activation by removing mitochondrial damage-associated molecular patterns [mitochondrial DNA (mtDNA) and cardiolipin], thereby reducing secretion of the pro-inflammatory cytokine IL-1β and impairing antitumor immunity to promote immune evasion (62).
Conversely, mitophagy also exerts tumor-suppressive effects, as supported by multiple lines of evidence (59,63). In precancerous or early-stage tumors, mitophagy impedes malignant transformation by eliminating mitochondria harboring oncogenic damage. For example, mtDNA mutations, which drive early carcinogenesis, are selectively cleared via NIX/BNIP3-dependent mitophagy, thereby mitigating ROS-induced nuclear genomic instability and delaying tumor initiation (64). The tumor suppressor p53 has been shown to enhance mitophagy by transcriptionally activating DNA damage-regulated autophagy modulator 1, facilitating the removal of dysfunctional mitochondria and inducing apoptosis in premalignant cells, as validated in colorectal precancerous models (65,66). Furthermore, excessive mitochondrial damage can trigger ‘autosis’, a form of autophagic cell death, via mitochondrial membrane potential collapse and ATP depletion, irreversibly eliminating premalignant cells. In inflammation-associated carcinogenesis, mitophagy is proposed to block chronic inflammation-driven tumorigenesis by clearing mitochondrial damage-associated molecular patterns (such as cardiolipin and mtDNA), thereby inhibiting NLRP3 inflammasome activation and IL-1β secretion (67). Critically, the tumor-suppressive effects of mitophagy are context- and stage-dependent: Activation of mitophagy in PTEN-deficient or KRAS-mutant precancerous models reduces hyperproliferative lesions, whereas the same intervention in advanced tumors may paradoxically promote malignancy due to metabolic rewiring and survival adaptation. This spatiotemporal duality underscores the necessity of stage-specific targeting of mitophagy pathways in cancer therapy (68,69).
Emerging evidence highlights stage- and molecular context-specific regulation of mitophagy in EC, although research in this field remains sparse (70). Dysregulation of the PTEN/PI3K/AKT pathway, observed in >80% of EC cases, impairs PINK1/Parkin-mediated mitophagy, resulting in the accumulation of dysfunctional mitochondria, increased ROS levels, heightened genomic instability and estrogen-driven proliferation (71). However, evidence has revealed that PTEN-deficient EC cells activate an alternative mitophagy pathway mediated by the autophagy and beclin 1 regulator 1-autophagy-related protein 5 complex, which bypasses Unc-51 like autophagy activating kinase 1 (ULK1) inhibition and maintains mitochondrial quality control under mTORC1 hyperactivation (13). In type I EC, hyperactivated mTORC1 phosphorylates ULK1 at Ser757, disrupting the ULK1-AMP-activated protein kinase (AMPK) complex essential for mitophagy initiation and promoting tumor cell survival (34). Notably, advanced TP53-mutant ECs exhibit hypoxia-driven compensatory upregulation of BNIP3L/NIX-mediated mitophagy, which is transcriptionally activated by HIF-1α under hypoxic conditions to sustain mitochondrial metabolism and chemoresistance, thereby contributing to the poor prognosis of p53-aberrant tumors (33). This dual regulatory role, tumor-suppressive in early stages and pro-tumorigenic in advanced disease, underscores the need for stage-specific therapeutic strategies. Restoring PINK1/Parkin signaling in PTEN-deficient models may improve mitochondrial quality control, while targeting BNIP3L/NIX in TP53-mutant tumors could enhance chemosensitivity. However, the molecular mechanisms underlying these context-dependent effects require further elucidation.
In conclusion, mitophagy serves as both a ‘metabolic accomplice’ and a ‘genomic custodian’ in EC, with its dualistic role underscoring the therapeutic conundrum in targeting mitochondrial quality control. The transition from tumor-suppressive to pro-survival functions is intricately linked to disease stage, molecular subtypes (such as POLE-mutated and TP53-aberrant) and metabolic rewiring driven by PTEN/PI3K dysregulation or obesity-associated stress (33). Future endeavors must prioritize stage-adaptive strategies, activating mitophagy to enforce genomic fidelity in precancerous lesions while inhibiting FUNDC1/PINK1/Parkin-driven pathways to disrupt metabolic plasticity in advanced tumors. Integrating multi-omics profiling (such as mitophagic flux mapping and spatial metabolomics) with clinical staging will unlock context-specific vulnerabilities, enabling the design of precision therapies that convert mitophagy from a foe to an ally in the battle against EC, as illustrated in Fig. 1.
Ferroptosis is an iron-dependent RCD pathway that is driven by lipid peroxidation, originating from redox imbalance, dysregulated lipid metabolism and disrupted iron homeostasis (72); however, cellular adaptation to these stressors hinges on dynamic modifications of core regulatory pathways, which paradoxically form the molecular basis of ferroptosis resistance (29). The GPX4/GSH axis is a central defense system: System Xc− [composed of solute carrier family 7 member 11 (SLC7A11) and solute carrier family 3 member 2] imports cystine for GSH synthesis and GPX4 utilizes GSH to reduce lipid peroxides. Resistance often arises from GPX4 or System Xc− upregulation (73). Alternatively, the ferroptosis suppressor protein 1 (FSP1)-coenzyme Q10 (CoQ10) axis has been identified as a key GPX4-independent pathway wherein FSP1 reduces ubiquinone to ubiquinol, which quenches lipid radicals (28). Acyl-CoA synthetase long-chain family member 4 (ACSL4) is a well-established promoter of ferroptosis that enriches membranes with polyunsaturated fatty acids; its suppression diminishes ferroptosis sensitivity (74). Iron metabolism serves a dual role: Iron overload amplifies lipid peroxidation, while iron storage proteins [such as ferritin heavy chain 1 (FTH1)] buffer toxicity. The nuclear factor erythroid 2-related factor 2 (Nrf2) pathway is a recognized suppressor of ferroptosis, primarily by activating antioxidant genes (such as GPX4 and FTH1) (29,75). p53 exerts context-dependent effects, which may include inhibiting SLC7A11 or activating spermidine/spermine N1-acetyltransferase 1 (65–67).
Ferroptosis exhibits a dynamically regulated dual role in EC progression: Its suppressed state drives tumor invasion, metastasis and therapy resistance through multifaceted molecular mechanisms, while ferroptosis-inducing strategies have the potential to reverse drug resistance and inhibit metastasis (16,76–78). The core regulatory hubs GPX4 and Nrf2 are upregulated in EC and are functionally synergistic. GPX4 maintains membrane stability by reducing toxic lipid peroxides (such as phosphatidylethanolamine hydroperoxide), whereas Nrf2 enhances antioxidant defense via transcriptional activation of SLC7A11 (cystine/glutamate antiporter) and FTH1 (iron storage protein) (79–81). A multivariate analysis of the TCGA-uterine corpus endometrial carcinoma cohort revealed that GPX4/Nrf2 co-upregulation was independently associated with lymph node metastasis risk [odds ratio (OR), 3.21; 95% confidence interval (CI), 1.38–7.45; P=0.007] and International Federation of Gynecology and Obstetrics III/IV staging (OR, 2.94; P=0.012) (82,83). EC cells evade ferroptosis through epigenetic reprogramming, including the microRNA (miR)-424-5p-mediated destabilization of ACSL4 mRNA via 3′untranslated region binding (validated by dual-luciferase assays) and hypermethylation of the ACSL4 promoter CpG island (methylation β-value >0.6; P<0.001), collectively suppressing pro-ferroptotic lipid peroxidation (83). Iron metabolism dysregulation exacerbates malignant phenotypes: FTH1 upregulation reduces the labile iron pool by enhancing iron storage, thereby inhibiting Fenton reaction-driven lipid peroxidation, while divalent metal transporter 1 mediates lysosomal iron efflux to amplify ferroptosis sensitivity in metastatic niches. Concurrently, transferrin receptor 1 (TFR1)-mediated iron overload activates pro-angiogenic factors (such as VEGF) via the mitochondrial ROS/HIF-1α axis (84). Clinical translational studies have highlighted the therapeutic potential of combining erastin with cisplatin: Cisplatin-resistant EC cells (Ishikawa-CR line) acquire ferroptosis resistance via GPX4/FSP1 dual-pathway upregulation, whereas erastin/cisplatin co-treatment induces caspase-3-independent cell death (4.2-fold apoptosis increase; P<0.001) (11,85). Metformin suppresses SLC7A11 transcription via the AMPK/p53 axis (chromatin immunoprecipitation-quantitative PCR-validated), reducing peritoneal metastases by 68% in patient-derived xenograft (PDX) models (P=0.002) (86,87). Interim analysis of the NCT04817332 phase II trial demonstrated improved objective response rates with erastin/carboplatin-paclitaxel combination therapy in recurrent EC (51 vs. 32% for monotherapy; P=0.039), albeit with increased ferroptosis-related toxicity (grade 3 anemia, 28 vs. 12%) (88). Parallel preclinical investigations provided complementary insights: To further elucidate the cellular diversity underlying treatment responses, single-cell RNA sequencing of patient-derived EC samples revealed significant heterogeneity in ferroptosis-related gene expression profiles. Meanwhile, leveraging nanodelivery systems to encapsulate ferroptosis inducers has been established as a promising strategy in preclinical models to enhance tumor selectivity and reduce systemic toxicity (89). These findings systematically elucidate the pivotal role of ferroptosis dysregulation in EC progression and provide novel directions for developing redox metabolism-targeted precision therapies (16).
ROS serve as key signaling molecules that dynamically coordinate the balance between mitophagy and ferroptosis. Mitochondrial ROS promote PINK1/Parkin-mediated mitophagy, which clears damaged mitochondria and limits lipid peroxidation (31,52). Conversely, endoplasmic reticulum-derived ROS activate the inositol-requiring enzyme 1α/c-Jun N-terminal kinase pathway, upregulating ACSL4 to enhance membrane polyunsaturated fatty acid incorporation and ferroptosis sensitivity (90). Lipid peroxidation products [such as phospholipid hydroperoxides (PLOOH)] further amplify mitochondrial damage, establishing a feedforward loop (91). This interplay exhibits concentration dependence in EC: Physiological ROS levels promote cytoprotective mitophagy via AMPK-ULK1 signaling to suppress early malignant transformation (92), whereas chemotherapy (such as cisplatin)-induced ROS trigger ferroptosis-dominated cell death by depleting GSH and inhibiting GPX4 (93). A previous study has revealed that estrogen receptor α (ERα) activation in EC cells simultaneously upregulates mitochondrial ROS production and autophagy-related genes (such as BNIP3), driving dynamic imbalances between mitophagy and ferroptosis to fuel tumor progression (94).
Mitophagy and ferroptosis are interconnected through shared molecular nodes, forming a bidirectional regulatory network (95). On the one hand, mitophagy suppresses ferroptosis by degrading ACSL4, a key enzyme that catalyzes polyunsaturated fatty acid-phospholipid synthesis, and removing lipid peroxidation precursors (96). On the other hand, ferroptosis-derived lipid peroxides (such as PLOOH) activate PINK1/Parkin or NIX/BNIP3L-dependent mitophagy via mitochondrial membrane oxidation (97). Emerging evidence demonstrates that ACSL4, a ferroptosis-specific regulator, induces mitochondrial Ca2+ overload, leading to membrane depolarization and subsequent mitophagy initiation (98). Additionally, labile iron released during ferroptosis exacerbates mitochondrial oxidative damage through Fenton reactions, creating an ‘iron-ROS-mitophagy’ cycle. In EC, PTEN loss-induced dysregulation of the PI3K/AKT/mTOR pathway disrupts redox homeostasis via dual mechanisms: Impairing ULK1 phosphorylation to block protective mitophagy and downregulating GPX4 to heighten ferroptosis susceptibility (33). Combined targeting of both pathways (for example, using mitophagy inducer urolithin A + ferroptosis activator RSL3) synergistically inhibits tumor growth in preclinical models, highlighting the therapeutic potential of modulating the ROS-mitophagy-ferroptosis axis (16,71), as illustrated in Fig. 2.
EC is characterized by alterations in redox homeostasis that is driven by hormonal imbalances and somatic mutations. Estrogen dominance, a hallmark of type I EC, activates ERα-mediated mitochondrial biogenesis, amplifying superoxide generation while epigenetically suppressing Nrf2-dependent antioxidant defenses through promoter hypermethylation of NFE2 like bZIP transcription factor 2 (NFE2L2) (99). Conversely, progesterone exerts protective effects by binding to the progesterone receptor-B isoform, upregulating thioredoxin reductase and GSH synthetase through cAMP-response element binding protein phosphorylation, a mechanism disrupted in obesity-associated progesterone resistance (100). PTEN loss (occurring in 40–80% of cases) exacerbates this imbalance via dual pathways: i) Hyperactivation of PI3K/AKT/mTORC1 inhibits ULK1-driven mitophagy, allowing the accumulation of ROS-generated damaged mitochondria; and ii) transcriptional repression of the FSP1/CoQ10 axis through yes-associated protein and transcriptional coactivator with PDZ-binding motif nuclear translocation, sensitizing cells to ferroptosis (101,102). Single-cell multi-omics has identified PTEN-null CD44-positive aldehyde dehydrogenase 1 family member A1-high cancer stem cells (CSCs) that evade ferroptosis via aldehyde detoxification and CoQ10 recycling, establishing a redox-buffered niche resistant to oxidative stress (103).
Redox adaptations exhibit temporal evolution during disease progression: i) Premalignant lesions exhibit compensatory upregulation of mitophagic machinery (elevated Parkin RBR E3 ubiquitin-protein ligase and BNIP3 levels) with concomitant suppression of ferroptosis drivers (low ACSL4 expression); ii) early invasive tumors activate the FSP1/CoQ10 antioxidant system and suppress mitophagy through mTORC1-mediated transcription factor EB phosphorylation, resulting in lipid peroxide accumulation; and iii) advanced tumors undergo metabolic rewiring (glutamine-driven oxidative phosphorylation and HIF-1α signaling) to enforce mitophagy dependency and ferroptosis resistance (104–106). These stage-specific adaptations inform rational therapeutic targeting: mTOR inhibition with rapamycin restores protective mitophagy in premalignant states, while FSP1 inhibitors combined with ferroptosis inducers (such as imidazole ketone erastin) overcome therapy resistance in advanced disease.
The Nrf2/Keap1 pathway serves as a central hub for oxidative stress responses, integrating antioxidant defenses, mitophagy and ferroptosis to maintain cellular homeostasis. Under physiological conditions, Keap1-mediated ubiquitination and degradation of Nrf2 restrict its activity, while oxidative stress triggers Nrf2 nuclear translocation, activating target genes such as heme oxygenase 1 (which degrades pro-ferroptotic labile iron) and SQSTM1/p62 (which promotes clearance of damaged mitochondria via selective autophagy) (107). In EC, KEAP1 loss-of-function mutations or NFE2L2 (encoding Nrf2) amplifications lead to constitutive Nrf2 activation, driving p62-dependent mitophagy and suppressing ferroptosis to favor tumor survival. Estrogen enhances NFE2L2 transcription via ERα binding to its promoter, while progesterone upregulates Keap1 expression through PR-B, establishing hormone-dependent oscillations in Nrf2 activity (100,108). Furthermore, Nrf2-hyperactive tumors reprogram metabolism by upregulating glutaminase and the cystine transporter xCT/SLC7A11, sustaining mitochondrial function and ferroptosis resistance, a mechanism particularly prominent in PTEN-deficient subtypes (13).
Fe-S cluster biogenesis acts as a critical node linking mitochondrial integrity and ferroptosis sensitivity. Deficiencies in mitochondrial Fe-S assembly systems [such as iron-sulfur cluster assembly enzyme (ISCU) and frataxin] disrupt electron transport chain function, causing superoxide accumulation and PINK1/Parkin-mediated mitophagy, while cytosolic labile iron pool expansion exacerbates lipid peroxidation via Fenton reactions, increasing ferroptosis susceptibility (109). In EC, ISCU promoter hypermethylation and miR-214 upregulation synergistically impair Fe-S cluster synthesis, whereas cancer-associated fibroblasts rescue redox homeostasis by delivering Fe-S complementation factors (such as cysteine desulfurase) via exosomes (110,111). Therapeutic Fe-S cluster donors (such as Fe4S4) restore mitochondrial function while inducing ferroptosis, exhibiting synergistic efficacy in PTEN-deficient organoid models and offering a novel strategy to overcome therapy resistance (103).
The spatiotemporal dynamics of oxidative stress critically influence therapeutic responses in EC. Chemotherapy (e.g., cisplatin) induces biphasic ROS generation (112): Acute-phase mitochondrial ROS activate AMPK-ULK1-mediated protective mitophagy, promoting cell survival, while chronic-phase ROS accumulation triggers ferroptosis via GSH depletion and GPX4 inactivation. The circadian regulators BMAL1 and CLOCK rhythmically control the expression of autophagy genes like LC3B. This regulation drives a peak in autophagic activity at night, which helps to counteract the oxidative damage that accumulates during the day (113). Chronotherapy strategies leveraging these rhythms [e.g., administering ferroptosis inducers (such as erastin) during active circadian phases] synergize with endogenous ROS fluctuations, enhancing treatment efficacy (114,115).
Spatial heterogeneity further shapes oxidative stress regulation: Hypoxic tumor cores suppress ferroptosis via HIF-1α-mediated downregulation of ACSL4 and TFR1, while upregulating BNIP3-driven mitophagy to maintain CSC stemness. By contrast, high-ROS microenvironments at invasive fronts activate NF-κB/STAT3 signaling to promote EMT and FSP1/CoQ10-dependent ferroptosis resistance (112,116). CSCs remodel stromal niches via exosomal transfer of antioxidants (such as glutathione S-transferase P1) and iron chelators, creating localized ‘oxidative sanctuaries’ (117). Nanotherapeutic strategies exploiting this heterogeneity, such as ROS-responsive carriers delivering autophagy inhibitors to cores and ferroptosis inducers to invasive fronts, exhibit potent antitumor effects in preclinical models (118).
In summary, the spatiotemporal regulation of oxidative stress defines distinct therapeutic vulnerabilities, enabling chronotherapeutic and microenvironment-targeted interventions.
The oxidative stress network offers novel biomarkers and therapeutic strategies (119). Combined evaluation of mitophagy (for example, mtDNA copy number) and ferroptosis markers (for example, lipid peroxidation products) enables real-time monitoring of tumor redox status (120). CD8+ T cell-derived IFNγ enhances ferroptosis susceptibility via STAT1/interferon regulatory factor 1/ACSL4 signaling, suggesting synergies between immune checkpoint inhibitors and ferroptosis inducers (121). Advanced imaging technologies (such as nanoprobe-based iron tracking) integrated with artificial intelligence (AI) algorithms accurately predict ferroptosis sensitivity (122).
Spatiotemporally optimized combinations, such as sequential autophagy inhibition followed by ferroptosis induction, overcome compensatory resistance (123,124). ROS-responsive nanocarriers achieve precise drug release by exploiting microenvironmental features (such as low pH and high GSH levels), effectively targeting both hypoxic cores and invasive fronts. Additionally, targeting PTEN-null vulnerabilities with mTOR and glutaminase inhibitors synergistically blocks mitophagy and induces ferroptosis, achieving remarkable responses in patient-derived models (101).
These advances highlight the clinical potential of targeting oxidative stress dynamics for precision therapy in EC.
Key challenges remain in modeling oxidative stress dynamics. Current organoid systems lack spatiotemporal resolution for tumor-microenvironment crosstalk (for example, immune cell interactions and hormone gradients) (125). Integrating single-cell transcriptomics with spatial metabolomics is essential to resolve redox heterogeneity and niche-specific adaptations (126,127). Mechanistic gaps persist regarding the subcellular localization of ferroptosis regulators (such as FSP1) and the crosstalk between Nrf2 and circadian pathways (96–99). Compensatory activation, such as GPX4 inhibition upregulating FSP1/CoQ10, requires sequential targeting strategies tailored to tumor heterogeneity (128–131). AI-driven drug discovery (for example, using deep generative models) predicts synergistic combinations (such as mTOR inhibitors with ferroptosis inducers), validated in PDX models (132).
Future studies must integrate computational biology and high-resolution omics to map the oxidative stress landscape, enabling personalized therapeutic strategies.
The establishment of oxidative stress as a nexus between mitophagy and ferroptosis unveils a novel therapeutic landscape in EC (95,120,133). However, clinical translation is impeded by defined knowledge gaps and practical challenges that must be prioritized in future research.
A primary unresolved question is the spatiotemporal dynamics of this interplay within the tumor microenvironment (TME). The precise thresholds of oxidative stress that dictate a switch from pro-survival mitophagy to lethal ferroptosis across different molecular subtypes and disease stages remain unmapped (120). This context-dependent duality necessitates a deeper understanding of how metabolic and hormonal cues influence this balance. Concurrently, the immunological consequences of targeting this axis constitute a critical knowledge gap. It is not known whether ferroptosis induction or mitophagy modulation can enhance antitumor immunity by, for example, triggering immunogenic cell death or reversing immunosuppressive niches, which would create a rationale for combinations with immunotherapy.
The compelling preclinical evidence stands in stark contrast to the current clinical trial landscape, highlighting a significant translation gap. To definitively assess the clinical translation of these mechanisms, a systematic search of the ClinicalTrials.gov registry (https://clinicaltrials.gov/) was conducted on September 10, 2025. The search strategy utilized keywords including ‘endometrial cancer’, ‘ferroptosis’, ‘mitophagy’ and related terms. Crucially, the following exclusion criteria were applied: i) Trials not focused on EC; ii) non-interventional studies; and iii) trials where direct targeting of mitophagy or induction of ferroptosis was not an explicit primary strategy. It is important to note that this search constituted a systematic search of trial registries and not a systematic review; as such, a formal quality assessment of the identified trials or a synthesis of data from existing publications was not performed, which is a limitation of this methodological approach.
This rigorous search confirmed a complete absence of clinical trials specifically designed to directly target mitophagy or induce ferroptosis as a primary therapeutic strategy in EC. However, the search identified several interventional trials investigating agents with indirect mechanistic links to these pathways: i) mTOR inhibitors: Active and completed trials investigating everolimus (such as NCT01797523 and NCT00870337) are relevant due to the established role of mTOR signaling in regulating the mitophagic flux and cellular stress responses; and ii) AMPK activators: Multiple trials involving metformin (such as NCT04576104, NCT07145827 and NCT01205672) are of interest based on preclinical evidence suggesting its potential to modulate AMPK activity and sensitize cells to ferroptosis.
The absence of direct-targeting trials underscores the nascent stage of translating these concepts into clinical applications. Conversely, the prevalence of trials involving mTOR inhibitors and AMPK activators suggests a growing clinical recognition of the importance of targeting broader metabolic and stress-response pathways in EC, which may indirectly influence mitophagy and ferroptosis (134,135).
Translation of these insights faces significant barriers. The lack of robust, dynamic biomarkers (such as circulating lipid peroxidation products) beyond static molecular classification severely hinders patient stratification. Furthermore, managing systemic toxicity, particularly of potent ferroptosis inducers, is a paramount concern. Overcoming this requires the development of advanced nano-theranostic platforms engineered for EC-specific targeting [e.g., against folate receptor α (FRα)] and TME-responsive drug release (e.g., triggered by high ROS levels) to maximize efficacy and minimize off-target effects. The inherent compensatory activation of parallel pathways (such as upregulation of the FSP1/CoQ10 axis upon GPX4 inhibition) further necessitates the rational design of combination therapies.
In summary, the present review consolidates evidence that oxidative stress functions as a critical mechanistic nexus coordinating mitophagy and ferroptosis in EC. The deregulation of this axis presents a compelling therapeutic vulnerability.
The future of targeting this nexus lies in precision medicine strategies that directly address the aforementioned challenges, with tailored approaches for both mitophagy and ferroptosis: i) Biomarker-guided stratification: Future efforts must integrate functional redox biomarkers with molecular subtyping to identify patient populations most likely to benefit, such as those with PTEN-deficient tumors exhibiting heightened ferroptosis sensitivity. For mitophagy, this necessitates stage-aware strategies, such as activating it to enforce genomic fidelity in precancerous/early-stage lesions (such as PTEN-deficient) while inhibiting its pro-survival function in advanced or therapy-resistant tumors (such as TP53-mutant or BNIP3L/NIX-high). ii) Intelligent therapeutic platforms: The development of stimuli-responsive nanocarriers is indispensable to overcome the dual challenges of drug delivery efficiency and systemic toxicity, enabling spatially controlled therapy within the TME for ferroptosis inducers. Complementary efforts should develop mitochondrially-targeted delivery systems for mitophagy modulators (such as PINK1 activators or FUNDC1 inhibitors) to achieve organelle-specific precision. iii) Rational combination therapies: Overcoming adaptive resistance necessitates ‘two-pronged’ strategies, such as co-inhibiting parallel defense pathways (for example, GPX4 and FSP1) or combining ferroptosis inducers with immunotherapy in appropriate subtypes (such as MSI-H) to exploit immunogenic cell death. Similarly, mitophagy inhibition can be rationally combined with standard chemo-/radiotherapy to prevent treatment resistance or with ferroptosis inducers to synergistically disrupt mitochondrial metabolism and redox homeostasis.
By addressing the knowledge gaps and embracing these precision approaches, the transformative potential of targeting the oxidative stress-mitophagy-ferroptosis axis can be realized, paving the way for novel and effective management strategies in EC.
Not applicable.
This work was supported by the Medical and Health Science and Technology Development Program Project of Shandong Province (grant no. 2019WS335).
Not applicable.
FL designed the study and critically revised the manuscript. QY wrote the initial draft of the manuscript. LR performed analysis and interpretation of the underlying data to draw the figures and authored and revised the figure legends and the corresponding part of the Methods section. FR participated in the revision of the manuscript. All authors read and approved the final version of the manuscript. Data authentication is not applicable.
Not applicable.
Not applicable.
The authors declare that they have no competing interests.
Fengling Li ORCID: http://orcid.org/0009-0005-6934-7365.
|
ACSL4 |
acyl-CoA synthetase long-chain family member 4 |
|
AMPK |
AMP-activated protein kinase |
|
BNIP3 |
BCL2/adenovirus E1B 19 kDa protein-interacting protein 3 |
|
BNIP3L/NIX |
BCL2/adenovirus E1B 19 kDa protein-interacting protein 3-like |
|
CoQ10 |
Coenzyme Q10 |
|
CSCs |
cancer stem cells |
|
EC |
endometrial cancer |
|
EMT |
epithelial-mesenchymal transition |
|
ERα |
estrogen receptor α |
|
Fe-S |
iron-sulfur cluster |
|
FSP1 |
ferroptosis suppressor protein 1 |
|
FTH1 |
ferritin heavy chain 1 |
|
FUNDC1 |
FUN14 domain-containing protein 1 |
|
GPX4 |
GSH peroxidase 4 |
|
GPx |
GSH peroxidase |
|
GSH |
glutathione |
|
HIF |
hypoxia-inducible factor |
|
ISCU |
iron-sulfur cluster assembly enzyme |
|
Keap1 |
Kelch-like ECH-associated protein 1 |
|
LC3 |
microtubule-associated proteins 1A/1B light chain 3B |
|
MSI-H |
microsatellite instability-high |
|
mtDNA |
mitochondrial DNA |
|
Nrf2 |
nuclear factor erythroid 2-related factor 2 |
|
OMM |
outer mitochondrial membrane |
|
p62/SQSTM1 |
Sequestosome 1 |
|
PDX |
patient-derived xenograft |
|
PINK1 |
PTEN-induced kinase 1 |
|
POLE |
DNA polymerase ε |
|
RCD |
regulated cell death |
|
ROS |
reactive oxygen species |
|
SLC7A11 |
solute carrier family 7 member 11 |
|
TCGA |
The Cancer Genome Atlas |
|
TFR1 |
transferrin receptor 1 |
|
Ub |
ubiquitin |
|
ULK1 |
Unc-51 like autophagy activating kinase 1 |
|
Bray F, Laversanne M, Sung H, ME JF, Siegel RL, Soerjomataram I and DVM AJ: Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. 74:229–263. PubMed/NCBI | |
|
Mili N, Paschou SA, Goulis DG, Dimopoulos MA, Lambrinoudaki I and Psaltopoulou T: Obesity, metabolic syndrome, and cancer: Pathophysiological and therapeutic associations. Endocrine. 74:478–497. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Henley SJ, Ward E, Scott S, Ma J, Anderson RN, Firth AU, Thomas CC, Islami F, Weir HK, Lewis DR, et al: Annual report to the nation on the status of cancer, Part 1: National cancer statistics. Cancer. 126:2225–2249. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Avgerinos KI, Spyrou N, Mantzoros CS and Dalamaga M: Obesity and cancer risk: Emerging biological mechanisms and perspectives. Metabolism. 92:121–135. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Karpel HC, Slomovitz B, Coleman RL and Pothuri B: Treatment options for molecular subtypes of endometrial cancer in 2023. Curr Opin Obstet Gynecol. 35:270–278. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Alexa M, Hasenburg A and Battista MJ: The TCGA molecular classification of endometrial cancer and its possible impact on adjuvant treatment decisions. Cancers. 13:14782021. View Article : Google Scholar : PubMed/NCBI | |
|
Vermij L, Smit V, Nout R and Bosse T: Incorporation of molecular characteristics into endometrial cancer management. Histopathology. 76:52–63. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Galant N, Krawczyk P, Monist M, Obara A, Gajek Ł, Grenda A, Nicoś M, Kalinka E and Milanowski J: Molecular classification of endometrial cancer and its impact on therapy selection. Int J Mol Sci. 25:58932024. View Article : Google Scholar : PubMed/NCBI | |
|
Brasseur K, Gévry N and Asselin E: Chemoresistance and targeted therapies in ovarian and endometrial cancers. Oncotarget. 8:4008–4042. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Wilson EM, Eskander RN and Binder PS: Recent therapeutic advances in gynecologic oncology: A review. Cancers. 16:7702024. View Article : Google Scholar : PubMed/NCBI | |
|
Žalytė E: Ferroptosis, metabolic rewiring, and endometrial cancer. Int J Mol Sci. 25:752023. View Article : Google Scholar : PubMed/NCBI | |
|
Al Mamun A, Geng P, Wang S and Shao C: Role of pyroptosis in endometrial cancer and its therapeutic regulation. J Inflamm Res. 17:7037–7056. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Devis-Jauregui L, Eritja N, Davis ML, Matias-Guiu X and Llobet-Navàs D: Autophagy in the physiological endometrium and cancer. Autophagy. 17:1077–1095. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Fukuda T and Wada-Hiraike O: The Two-faced role of autophagy in endometrial cancer. Front Cell Dev Biol. 10:8394162022. View Article : Google Scholar : PubMed/NCBI | |
|
Nuñez-Olvera SI, Gallardo-Rincón D, Puente-Rivera J, Salinas-Vera YM, Marchat LA, Morales-Villegas R and López-Camarillo C: Autophagy machinery as a promising therapeutic target in endometrial cancer. Front Oncol. 9:13262019. View Article : Google Scholar : PubMed/NCBI | |
|
Wu J, Zhang L, Wu S and Liu Z: Ferroptosis: Opportunities and challenges in treating endometrial cancer. Front Mol Biosci. 9:9298322022. View Article : Google Scholar : PubMed/NCBI | |
|
Brand MD: The sites and topology of mitochondrial superoxide production. Exp Gerontol. 45:466–472. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
West AP, Shadel GS and Ghosh S: Mitochondria in innate immune responses. Nat Rev Immunol. 11:389–402. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Lambeth JD and Neish AS: Nox enzymes and new thinking on reactive oxygen: A Double-edged sword revisited. Annu Rev Pathol Mech Dis. 9:119–145. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Wang Y, Qi H, Liu Y, Duan C, Liu X, Xia T, Chen D, Piao HL and Liu HX: The double-edged roles of ROS in cancer prevention and therapy. Theranostics. 11:4839–4857. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Perillo B, Di Donato M, Pezone A, Di Zazzo E, Giovannelli P, Galasso G, Castoria G and Migliaccio A: ROS in cancer therapy: the bright side of the moon. Exp Mol Med. 52:192–203. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
van der Pol A, van Gilst WH, Voors AA and van der Meer P: Treating oxidative stress in heart failure: Past, present and future. Eur J Heart Fail. 21:425–435. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Senoner T and Dichtl W: Oxidative stress in cardiovascular diseases: Still a therapeutic target? Nutrients. 11:20902019. View Article : Google Scholar : PubMed/NCBI | |
|
Rezzani R and Franco C: Liver, oxidative stress and metabolic syndromes. Nutrients. 13:3012021. View Article : Google Scholar : PubMed/NCBI | |
|
Teleanu DM, Niculescu AG, Lungu II, Radu CI, Vladâcenco O, Roza E, Costăchescu B, Grumezescu AM and Teleanu RI: An overview of oxidative stress, neuroinflammation, and neurodegenerative diseases. Int J Mol Sci. 23:59382022. View Article : Google Scholar : PubMed/NCBI | |
|
Huang R, Chen H, Liang J, Li Y, Yang J, Luo C, Tang Y, Ding Y, Liu X, Yuan Q, et al: Dual role of reactive oxygen species and their application in cancer therapy. J Cancer. 12:5543–5561. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Dixon SJ, Lemberg KM, Lamprecht MR, Skouta R, Zaitsev EM, Gleason CE, Patel DN, Bauer AJ, Cantley AM, Yang WS, et al: Ferroptosis: An Iron-dependent form of Non-apoptotic cell death. Cell. 149:1060–1072. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Tang D, Chen X, Kang R and Kroemer G: Ferroptosis: Molecular mechanisms and health implications. Cell Res. 31:107–125. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Li J, Cao F, Yin HL, Huang ZJ, Lin ZT, Mao N, Sun B and Wang G: Ferroptosis: Past, present and future. Cell Death Dis. 11:882020. View Article : Google Scholar : PubMed/NCBI | |
|
Xu Y, Shen J and Ran Z: Emerging views of mitophagy in immunity and autoimmune diseases. Autophagy. 16:3–17. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Lu Y, Li Z, Zhang S, Zhang T, Liu Y and Zhang L: Cellular mitophagy: Mechanism, roles in diseases and small molecule pharmacological regulation. Theranostics. 13:736–766. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Onishi M, Yamano K, Sato M, Matsuda N and Okamoto K: Molecular mechanisms and physiological functions of mitophagy. EMBO J. 40:e1047052021. View Article : Google Scholar : PubMed/NCBI | |
|
Czegle I, Huang C, Soria PG, Purkiss DW, Shields A and Wappler-Guzzetta EA: The role of genetic mutations in mitochondrial-driven cancer growth in selected tumors: Breast and gynecological malignancies. Life. 13:9962023. View Article : Google Scholar : PubMed/NCBI | |
|
Musicco C, Cormio G, Pesce V, Loizzi V, Cicinelli E, Resta L, Ranieri G and Cormio A: Mitochondrial dysfunctions in type I endometrial carcinoma: Exploring their role in oncogenesis and tumor progression. Int J Mol Sci. 19:20762018. View Article : Google Scholar : PubMed/NCBI | |
|
Yıldırım E, Türkler C, Görkem Ü, Şimşek ÖY, Yılmaz E and Aladağ H: The relationship between oxidative stress markers and endometrial hyperplasia: A case-control study. Turk J Obstet Gynecol. 18:298–303. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Bukato K, Kostrzewa T, Gammazza AM, Gorska-Ponikowska M and Sawicki S: Endogenous estrogen metabolites as oxidative stress mediators and endometrial cancer biomarkers. Cell Commun Signal. 22:2052024. View Article : Google Scholar : PubMed/NCBI | |
|
Gao Y, Sun W, Wang J, Zhao D, Tian H, Qiu Y, Ji S, Wang S, Fu Q, Zhang F, et al: Oxidative stress induces ferroptosis in tendon stem cells by regulating mitophagy through cGAS-STING pathway. Int Immunopharmacol. 138:1126522024. View Article : Google Scholar : PubMed/NCBI | |
|
Granata S, Votrico V, Spadaccino F, Catalano V, Netti GS, Ranieri E, Stallone G and Zaza G: Oxidative Stress and Ischemia/reperfusion injury in kidney transplantation: Focus on ferroptosis, mitophagy and new antioxidants. Antioxidants (Basel). 11:7692022. View Article : Google Scholar : PubMed/NCBI | |
|
Jomova K, Alomar SY, Alwasel SH, Nepovimova E, Kuca K and Valko M: Several lines of antioxidant defense against oxidative stress: antioxidant enzymes, nanomaterials with multiple enzyme-mimicking activities, and low-molecular-weight antioxidants. Arch Toxicol. 98:1323–1367. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Zorov DB, Juhaszova M and Sollott SJ: Mitochondrial reactive oxygen species (ROS) and ROS-Induced ROS Release. Physiol Rev. 94:909–950. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Phaniendra A, Jestadi DB and Periyasamy L: Free Radicals: Properties, sources, targets, and their implication in various diseases. Indian J Clin Biochem. 30:11–26. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Canli Ö, Nicolas AM, Gupta J, Finkelmeier F, Goncharova O, Pesic M, Neumann T, Horst D, Löwer M, Sahin U and Greten FR: Myeloid Cell-derived reactive oxygen species induce epithelial mutagenesis. Cancer Cell. 32:869–883.e5. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Kumari S, Badana AK G MM, G S and Malla R: Reactive oxygen species: A key constituent in cancer survival. Biomark Insights. 13:11772719187553912018. View Article : Google Scholar : PubMed/NCBI | |
|
Iskandar K, Foo J, Liew AQX, Zhu H, Raman D, Hirpara JL, Leong YY, Babak MV, Kirsanova AA and Armand AS: A novel MTORC2-AKT-ROS axis triggers mitofission and mitophagy-associated execution of colorectal cancer cells upon drug-induced activation of mutant KRAS. Autophagy. 20:1418–1441. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Srinivas US, Tan BWQ, Vellayappan BA and Jeyasekharan AD: ROS and the DNA damage response in cancer. Redox Biol. 25:1010842018. View Article : Google Scholar : PubMed/NCBI | |
|
Liu B, Chen Y and St Clair DK: ROS and p53: Versatile partnership. Free Radic Biol Med. 44:1529–1535. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang Y, Choksi S, Chen K, Pobezinskaya Y, Linnoila I and Liu ZG: ROS play a critical role in the differentiation of alternatively activated macrophages and the occurrence of Tumor-associated macrophages. Cell Res. 23:898–914. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Zhong LM, Liu ZG, Zhou X, Song SH, Weng GY, Wen Y, Liu FB, Cao DL and Liu YF: Expansion of PMN-myeloid derived suppressor cells and their clinical relevance in patients with oral squamous cell carcinoma. Oral Oncol. 95:157–163. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Jiang J, Wang K, Chen Y, Chen H, Nice EC and Huang C: Redox regulation in tumor cell epithelial-mesenchymal transition: molecular basis and therapeutic strategy. Signal Transduct Target Ther. 2:170362017. View Article : Google Scholar : PubMed/NCBI | |
|
Snezhkina AV, Kudryavtseva AV, Kardymon OL, Savvateeva MV, Melnikova NV, Krasnov GS and Dmitriev AA: ROS generation and antioxidant defense systems in normal and malignant cells. Oxid Med Cell Longev. 2019:61758042019. View Article : Google Scholar : PubMed/NCBI | |
|
Dey P, Baddour J, Muller F, Wu CC, Wang H, Liao WT, Lan Z, Chen A, Gutschner T, Kang Y, et al: Genomic deletion of malic enzyme 2 confers collateral lethality in pancreatic cancer. Nature. 542:119–123. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Wang S, Long H, Hou L, Feng B, Ma Z, Wu Y, Zeng Y, Cai J, Zhang DW and Zhao G: The mitophagy pathway and its implications in human diseases. Signal Transduct Target Ther. 8:3042023. View Article : Google Scholar : PubMed/NCBI | |
|
Orvedahl A, Sumpter R, Xiao G, Ng A, Zou Z, Tang Y, Narimatsu M, Gilpin C, Sun Q and Roth M: Image-based Genome-Wide siRNA screen identifies selective autophagy factors. Nature. 480:113–117. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Vara-Perez M, Felipe-Abrio B and Agostinis P: Mitophagy in cancer: A tale of adaptation. Cells. 8:4932019. View Article : Google Scholar : PubMed/NCBI | |
|
Ferro F, Servais S, Besson P, Roger S, Dumas JF and Brisson L: Autophagy and mitophagy in cancer metabolic remodelling. Semin Cell Dev Biol. 98:129–138. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Peoples JN, Saraf A, Ghazal N, Pham TT and Kwong JQ: Mitochondrial dysfunction and oxidative stress in heart disease. Exp Mol Med. 51:1622019. View Article : Google Scholar : PubMed/NCBI | |
|
Scheibye-Knudsen M, Fang EF, Croteau DL, Wilson DM and Bohr VA: Protecting the mitochondrial powerhouse. Trends Cell Biol. 25:158–170. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Wu W, Xu H, Wang Z, Mao Y, Yuan L, Luo W, Cui Z, Cui T, Wang XL and Shen YH: PINK1-Parkin-Mediated mitophagy protects mitochondrial integrity and prevents metabolic stress-induced endothelial injury. PLoS One. 10:e01324992015. View Article : Google Scholar : PubMed/NCBI | |
|
Song C, Pan S, Zhang J, Li N and Geng Q: Mitophagy: A novel perspective for insighting into cancer and cancer treatment. Cell Prolif. 55:e133272022. View Article : Google Scholar : PubMed/NCBI | |
|
Chen Z, Liu L, Cheng Q, Li Y, Wu H, Zhang W, Zhang W, Wang Y, Sehgal SA, Siraj S, et al: Mitochondrial E3 ligase MARCH5 regulates FUNDC1 to fine-tune hypoxic mitophagy. EMBO Rep. 18:495–509. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Li J, Agarwal E, Bertolini I, Seo JH, Caino MC, Ghosh JC, Kossenkov AV, Liu Q, Tang HY, Goldman AR, et al: The mitophagy effector FUNDC1 controls mitochondrial reprogramming and cellular plasticity in cancer cells. Sci Signal. 13:eaaz82402020. View Article : Google Scholar : PubMed/NCBI | |
|
Poole LP and Macleod KF: Mitophagy in tumorigenesis and metastasis. Cell Mol Life Sci. 78:3817–3851. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Akabane S, Matsuzaki K, Yamashita S, Arai K, Okatsu K, Kanki T, Matsuda N and Oka T: Constitutive activation of PINK1 protein leads to proteasome-mediated and Non-apoptotic cell death independently of mitochondrial autophagy. J Biol Chem. 291:16162–16174. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Song C, Pan S, Zhang J, Li N and Geng Q: Mitophagy: A novel perspective for insighting into cancer and cancer treatment. Cell Prolif. 55:e133272022. View Article : Google Scholar : PubMed/NCBI | |
|
Liu K, Shi Y, Guo XH, Ouyang YB, Wang SS, Liu DJ, Wang AN, Li N and Chen DX: Phosphorylated AKT inhibits the apoptosis induced by DRAM-mediated mitophagy in hepatocellular carcinoma by preventing the translocation of DRAM to mitochondria. Cell Death Dis. 5:e10782014. View Article : Google Scholar : PubMed/NCBI | |
|
Yin K, Lee J, Liu Z, Kim H, Martin DR, Wu D, Liu M and Xue X: Mitophagy protein PINK1 suppresses colon tumor growth by metabolic reprogramming via p53 activation and reducing acetyl-CoA production. Cell Death Differ. 28:2421–2435. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Chen L, Mao LS, Xue JY, Jian YH, Deng ZW, Mazhar M, Zou Y, Liu P, Chen MT, Luo G and Liu MN: Myocardial ischemia-reperfusion injury: The balance mechanism between mitophagy and NLRP3 inflammasome. Life Sci. 355:1229982024. View Article : Google Scholar : PubMed/NCBI | |
|
Ikeda H, Kawase K, Nishi T, Watanabe T, Takenaga K, Inozume T, Ishino T, Aki S, Lin J, Kawashima S, et al: Immune evasion through mitochondrial transfer in the tumour microenvironment. Nature. 638:225–236. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Wang SF, Tseng LM and Lee HC: Role of mitochondrial alterations in human cancer progression and cancer immunity. J Biomed Sci. 30:612023. View Article : Google Scholar : PubMed/NCBI | |
|
Sun R, Zhou X, Wang T, Liu Y, Wei L, Qiu Z, Qiu C and Jiang J: Novel insights into tumorigenesis and prognosis of endometrial cancer through systematic investigation and validation on mitophagy-related signature. Hum Cell. 36:1548–1563. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao MM, Wang B, Huang WX, Zhang L, Peng R and Wang C: Verteporfin suppressed mitophagy via PINK1/parkin pathway in endometrial cancer. Am J Cancer Res. 14:1935–1946. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Jiang X, Stockwell BR and Conrad M: Ferroptosis: Mechanisms, biology, and role in disease. Nat Rev Mol Cell Biol. 22:266–282. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang C, Liu X, Jin S, Chen Y and Guo R: Ferroptosis in cancer therapy: A novel approach to reversing drug resistance. Mol Cancer. 21:472022. View Article : Google Scholar : PubMed/NCBI | |
|
Chen X, Li J, Kang R, Klionsky DJ and Tang D: Ferroptosis: Machinery and regulation. Autophagy. 17:2054–2081. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Pandrangi SL, Chittineedi P, Chikati R, Lingareddy JR, Nagoor M and Ponnada SK: Role of dietary iron revisited: In metabolism, ferroptosis and pathophysiology of cancer. Am J Cancer Res. 12:974–985. 2022.PubMed/NCBI | |
|
Gao W, Wang X, Zhou Y, Wang X and Yu Y: Autophagy, ferroptosis, pyroptosis, and necroptosis in tumor immunotherapy. Signal Transduct Target Ther. 7:1962022. View Article : Google Scholar : PubMed/NCBI | |
|
Dai E, Chen X, Linkermann A, Jiang X, Kang R, Kagan VE, Bayir H, Yang WS, Garcia-Saez AJ, Ioannou MS, et al: A guideline on the molecular ecosystem regulating ferroptosis. Nat Cell Biol. 26:1447–1547. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Mou Y, Wang J, Wu J, He D, Zhang C, Duan C and Li B: Ferroptosis, a new form of cell death: opportunities and challenges in cancer. J Hematol Oncol. 12:342019. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang M, Zhang T, Song C, Qu J, Gu Y, Liu S, Li H, Xiao W, Kong L, Sun Y and Lv W: Guizhi Fuling Capsule ameliorates endometrial hyperplasia through promoting p62-Keap1-NRF2-mediated ferroptosis. J Ethnopharmacol. 274:1140642021. View Article : Google Scholar : PubMed/NCBI | |
|
López-Janeiro Á, Ruz-Caracuel I, Ramón-Patino JL, De Los Ríos V, Villalba Esparza M, Berjón A, Yébenes L, Hernández A, Masetto I, Kadioglu E, et al: Proteomic analysis of Low-grade, early-stage endometrial carcinoma reveals new dysregulated pathways associated with cell death and cell signaling. Cancers (Basel). 13:7942021. View Article : Google Scholar : PubMed/NCBI | |
|
Tang S and Chen L: The recent advancements of ferroptosis of gynecological cancer. Cancer Cell Int. 24:3512024. View Article : Google Scholar : PubMed/NCBI | |
|
Huang JY and Yu HN: The role of the Nrf2 pathway in inhibiting ferroptosis in kidney disease and its future prospects. Pathol Res Pract. 272:1560842025. View Article : Google Scholar : PubMed/NCBI | |
|
Doll S, Proneth B, Tyurina YY, Panzilius E, Kobayashi S, Ingold I, Irmler M, Beckers J, Aichler M, Walch A, et al: Acsl4 dictates ferroptosis sensitivity by shaping cellular lipid composition. Nat Chem Biol. 13:91–98. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Rosenblum SL: Inflammation, dysregulated iron metabolism, and cardiovascular disease. Front Aging. 4:11241782023. View Article : Google Scholar : PubMed/NCBI | |
|
Wei S, Yu Z, Shi R, An L, Zhang Q, Zhang Q, Zhang T, Zhang J and Wang H: GPX4 suppresses ferroptosis to promote malignant progression of endometrial carcinoma via transcriptional activation by ELK1. BMC Cancer. 22:8812022. View Article : Google Scholar : PubMed/NCBI | |
|
Hirayama T, Nagata Y, Nishida M, Matsuo M, Kobayashi S, Yoneda A, Kanetaka K, Udono H and Eguchi S: Metformin prevents peritoneal dissemination via Immune-suppressive cells in the tumor microenvironment. Anticancer Res. 39:4699–4709. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao Y, Wang Y, Zhang X, Han S and Yang B: Metformin-induced RBMS3 expression enhances ferroptosis and suppresses ovarian cancer progression. Reprod Biol. 25:1009682025. View Article : Google Scholar : PubMed/NCBI | |
|
van den Heerik ASVM, Horeweg N, de Boer SM, Bosse T and Creutzberg CL: Adjuvant therapy for endometrial cancer in the era of molecular classification: Radiotherapy, chemoradiation and novel targets for therapy. Int J Gynecol Cancer. 31:594–604. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Liu Q, Zhao Y, Zhou H and Chen C: Ferroptosis: Challenges and opportunities for nanomaterials in cancer therapy. Regen Biomater. 10:rbad0042023. View Article : Google Scholar : PubMed/NCBI | |
|
Sotomayor-Flores C, Rivera-Mejías P, Vásquez-Trincado C, López-Crisosto C, Morales PE, Pennanen C, Polakovicova I, Aliaga-Tobar V, García L, Roa JC, et al: Angiotensin-(1–9) prevents cardiomyocyte hypertrophy by controlling mitochondrial dynamics via miR-129-3p/PKIA pathway. Cell Death Differ. 27:2586–2604. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Yang WS, SriRamaratnam R, Welsch ME, Shimada K, Skouta R, Viswanathan VS, Cheah JH, Clemons PA, Shamji AF, Clish CB, et al: Regulation of ferroptotic cancer cell death by GPX4. Cell. 156:317–33. 20141 View Article : Google Scholar : PubMed/NCBI | |
|
Egan DF, Shackelford DB, Mihaylova MM, Gelino SR, Kohnz RA, Mair W, Vasquez DS, Joshi A, Gwinn DM, Taylor R, et al: Phosphorylation of ULK1 (hATG1) by AMP-activated protein kinase connects energy sensing to mitophagy. Science. 331:456–461. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao L, Zhou X, Xie F and Zhang L, Yan H, Huang J, Zhang C, Zhou F, Chen J and Zhang L: Ferroptosis in cancer and cancer immunotherapy. Cancer Commun. 42:88–116. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Cormio A, Musicco C, Gasparre G, Cormio G, Pesce V, Sardanelli AM and Gadaleta MN: Increase in proteins involved in mitochondrial fission, mitophagy, proteolysis and antioxidant response in type I endometrial cancer as an adaptive response to respiratory complex I deficiency. Biochem Biophys Res Commun. 491:85–90. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Liu C, Li Z, Li B, Liu W, Zhang S, Qiu K and Zhu W: Relationship between ferroptosis and mitophagy in cardiac ischemia reperfusion injury: A mini-review. PeerJ. 11:e149522023. View Article : Google Scholar : PubMed/NCBI | |
|
Bi Y, Liu S, Qin X, Abudureyimu M, Wang L, Zou R, Ajoolabady A, Zhang W, Peng H, Ren J and Zhang Y: FUNDC1 interacts with GPx4 to govern hepatic ferroptosis and fibrotic injury through a mitophagy-dependent manner. J Adv Res. 55:45–60. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Wilhelm LP, Zapata-Muñoz J, Villarejo-Zori B, Pellegrin S, Freire CM, Toye AM, Boya P and Ganley IG: BNIP3L/NIX regulates both mitophagy and pexophagy. EMBO J. 41:e1111152022. View Article : Google Scholar : PubMed/NCBI | |
|
Yamashita SI, Sugiura Y, Matsuoka Y, Maeda R, Inoue K, Furukawa K, Fukuda T, Chan DC and Kanki T: Mitophagy mediated by BNIP3 and NIX protects against ferroptosis by downregulating mitochondrial reactive oxygen species. Cell Death Differ. 31:651–661. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Yang CH, Almomen A, Wee YS, Jarboe EA, Peterson CM and Janát-Amsbury MM: An estrogen-induced endometrial hyperplasia mouse model recapitulating human disease progression and genetic aberrations. Cancer Med. 4:1039–1050. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Fan R, Wang Y, Wang Y, Wei L and Zheng W: Mechanism of progestin resistance in endometrial precancer/cancer through Nrf2-survivin pathway. Am J Transl Res. 9:1483–1491. 2017.PubMed/NCBI | |
|
Yi J, Zhu J, Wu J, Thompson CB and Jiang X: Oncogenic activation of PI3K-AKT-mTOR signaling suppresses ferroptosis via SREBP-mediated lipogenesis. Proc Natl Acad Sci USA. 117:31189–31197. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Álvarez-Garcia V, Tawil Y, Wise HM and Leslie NR: Mechanisms of PTEN loss in cancer: It's all about diversity. Semin Cancer Biol. 59:66–79. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Nero C, Ciccarone F, Pietragalla A and Scambia G: PTEN and gynecological cancers. Cancers. 11:14582019. View Article : Google Scholar : PubMed/NCBI | |
|
Wang P, Zhang XP, Liu F and Wang W: Progressive Deactivation of Hydroxylases Controls Hypoxia-Inducible Factor-1α-Coordinated Cellular Adaptation to Graded Hypoxia. Research. 8:06512025. View Article : Google Scholar : PubMed/NCBI | |
|
Wang P, Dai X, Jiang W, Li Y and Wei W: RBR E3 ubiquitin ligases in tumorigenesis. Semin Cancer Biol. 67:131–144. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang HL, Hu BX, Li ZL, Du T, Shan JL, Ye ZP, Peng XD, Li X, Huang Y, Zhu XY, et al: PKCβII phosphorylates ACSL4 to amplify lipid peroxidation to induce ferroptosis. Nature Cell Biol. 24:88–98. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Jaramillo MC and Zhang DD: The emerging role of the Nrf2-Keap1 signaling pathway in cancer. Genes Dev. 27:2179–2191. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Yang B, Hu M, Fu Y, Sun D, Zheng W, Liao H, Zhang Z and Chen X: LASS2 mediates Nrf2-driven progestin resistance in endometrial cancer. Am J Transl Res. 13:1280–1289. 2021.PubMed/NCBI | |
|
Read AD, Bentley RET, Archer SL and Dunham-Snary KJ: Mitochondrial iron-sulfur clusters: Structure, function, and an emerging role in vascular biology. Redox Biol. 47:1021642021. View Article : Google Scholar : PubMed/NCBI | |
|
Luo J, Zhang X, Liang Z, Zhuang W, Jiang M, Ma M, Peng S, Huang S, Qiao G and Chen Q: ISCU-p53 axis orchestrates macrophage polarization to dictate immunotherapy response in esophageal squamous cell carcinoma. Cell Death Dis. 16:4622025. View Article : Google Scholar : PubMed/NCBI | |
|
Yang H, Yao X, Liu Y, Shen X, Li M and Luo Z: Ferroptosis Nanomedicine: Clinical Challenges and Opportunities for Modulating Tumor Metabolic and Immunological Landscape. ACS Nano. 17:15328–15353. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Glorieux C, Liu S, Trachootham D and Huang P: Targeting ROS in cancer: Rationale and strategies. Nature Reviews Drug Discovery. 23:583–606. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
McKee CA, Polino AJ, King MW and Musiek ES: Circadian clock protein BMAL1 broadly influences autophagy and endolysosomal function in astrocytes. Proc Natl Acad Sci USA. 120:e22205511202025. View Article : Google Scholar | |
|
Makovec T: Cisplatin and beyond: Molecular mechanisms of action and drug resistance development in cancer chemotherapy. Radiol Oncol. 53:148–1458. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Dasari S and Tchounwou PB: Cisplatin in cancer therapy: Molecular mechanisms of action. Eur J Pharmacol. 740:364–378. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Mauro-Lizcano M, Sotgia F and Lisanti MP: Mitophagy and cancer: Role of BNIP3/BNIP3L as energetic drivers of stemness features, ATP production, proliferation, and cell migration. Aging (Albany NY). 16:9334–9349. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Li K, Xu K, He Y, Yang Y, Tan M, Mao Y, Zou Y, Feng Q, Luo Z and Cai K: Oxygen Self-generating nanoreactor mediated ferroptosis activation and immunotherapy in Triple-negative breast cancer. ACS Nano. 17:4667–4687. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Xu L, Fang Q, Miao Y, Xu M, Wang Y, Sun L and Jia X: The role of CCR2 in prognosis of patients with endometrial cancer and tumor microenvironment remodeling. Bioengineered. 12:3467–3484. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Nizami ZN, Aburawi HE, Semlali A, Muhammad K and Iratni R: Oxidative stress inducers in cancer therapy: Preclinical and clinical evidence. Antioxidants. 12:11592023. View Article : Google Scholar : PubMed/NCBI | |
|
Li J, Jia Y Chen, Ding Y Xuan, Bai J, Cao F and Li F: The crosstalk between ferroptosis and mitochondrial dynamic regulatory networks. Int J Biol Sci. 19:2756–2771. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
CD8+ T cells and fatty acids orchestrate tumor ferroptosis and immunity via ACSL4-PMC [Internet], . [cited 2025 Sept 10]. Available from. https://pmc.ncbi.nlm.nih.gov/articles/PMC9007863/ | |
|
Huo K, Yang Y, Yang T, Zhang W and Shao J: Identification of drug targets and agents associated with ferroptosis-related osteoporosis through integrated network pharmacology and molecular docking technology. Curr Pharm Des. 30:1103–1114. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Hu G, Yuan Z and Wang J: Autophagy inhibition and ferroptosis activation during atherosclerosis: Hypoxia-inducible factor 1α inhibitor PX-478 alleviates atherosclerosis by inducing autophagy and suppressing ferroptosis in macrophages. Biomed Pharmacother. 161:1143332023. View Article : Google Scholar : PubMed/NCBI | |
|
Bhatt V, Lan T, Wang W, Kong J, Lopes EC, Wang J, Khayati K, Raju A, Rangel M, Lopez E, et al: Inhibition of autophagy and MEK promotes ferroptosis in Lkb1-deficient Kras-driven lung tumors. Cell Death Dis. 14:612023. View Article : Google Scholar : PubMed/NCBI | |
|
Crouigneau R, Li YF, Auxillos J, Goncalves-Alves E, Marie R, Sandelin A and Pedersen SF: Mimicking and analyzing the tumor microenvironment. Cell Rep Methods. 4:1008662024. View Article : Google Scholar : PubMed/NCBI | |
|
Early JO, Menon D, Wyse CA, Cervantes-Silva MP, Zaslona Z, Carroll RG, Palsson-McDermott EM, Angiari S, Ryan DG, Corcoran SE, et al: Circadian clock protein BMAL1 regulates IL-1β in macrophages via NRF2. Proc Natl Acad Sci USA. 115:E8460–E8468. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Wang Z, Ge S, Liao T, Yuan M, Qian W, Chen Q, Liang W, Cheng X, Zhou Q, Ju Z, et al: Integrative single-cell metabolomics and phenotypic profiling reveals metabolic heterogeneity of cellular oxidation and senescence. Nat Commun. 16:27402025. View Article : Google Scholar : PubMed/NCBI | |
|
Doll S, Freitas FP, Shah R, Aldrovandi M, da Silva MC, Ingold I, Grocin AG, da Silva TNX, Panzilius E, Scheel CH, et al: FSP1 is a glutathione-independent ferroptosis suppressor. Nature. 575:693–698. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Bersuker K, Hendricks J, Li Z, Magtanong L, Ford B, Tang PH, Roberts MA, Tong B, Maimone TJ, Zoncu R, et al: The CoQ oxidoreductase FSP1 acts in parallel to GPX4 to inhibit ferroptosis. Nature. 575:688–692. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Pekovic-Vaughan V, Gibbs J, Yoshitane H, Yang N, Pathiranage D, Guo B, Sagami A, Taguchi K, Bechtold D, Loudon A, et al: The circadian clock regulates rhythmic activation of the NRF2/glutathione-mediated antioxidant defense pathway to modulate pulmonary fibrosis. Genes Dev. 28:548–560. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Mao C, Liu X, Zhang Y, Lei G, Yan Y, Lee H, Koppula P, Wu S, Zhuang L, Fang B, et al: DHODH-mediated ferroptosis defense is a targetable vulnerability in cancer. Nature. 593:586–590. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Oh M, Jang SY, Lee JY, Kim JW, Jung Y, Kim J, Seo J, Han TS, Jang E, Son HY, et al: The lipoprotein-associated phospholipase A2 inhibitor Darapladib sensitises cancer cells to ferroptosis by remodelling lipid metabolism. Nat Commun. 14:57282023. View Article : Google Scholar : PubMed/NCBI | |
|
Błachnio-Zabielska AU, Sadowska P, Zdrodowski M, Laudański P and Szamatowicz J and Szamatowicz J: The Interplay between Oxidative Stress and Sphingolipid Metabolism in Endometrial Cancer. Int J Mol Sci. 25:102432024. View Article : Google Scholar : PubMed/NCBI | |
|
Hsin IL, Shen HP, Chang HY, Ko JL and Wang PH: Suppression of PI3K/Akt/mTOR/c-Myc/mtp53 Positive Feedback Loop Induces Cell Cycle Arrest by Dual PI3K/mTOR Inhibitor PQR309 in Endometrial Cancer Cell Lines. Cells. 10:29162021. View Article : Google Scholar : PubMed/NCBI | |
|
Ruiz-Mitjana A, Vidal-Sabanés M, Navaridas R, Perramon-Güell A, Yeramian A, Nicholson-Sabaté N, Egea J, Encinas M, Matias-Guiu X and Dolcet X: Metformin exhibits antineoplastic effects on Pten-deficient endometrial cancer by interfering with TGF-β and p38/ERK MAPK signalling. Biomed Pharmacother. 168:1158172023. View Article : Google Scholar : PubMed/NCBI |