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.
Malignant tumors, which are characterized by phenotypic plasticity, metabolic reprogramming and immune evasion, continuously adapt to conventional treatment strategies, highlighting the urgent need for the development of novel and more effective therapeutic approaches (1). Radiotherapy exerts its tumor-killing effects through two main mechanisms: i) Direct DNA damage induced by high-energy radiation; and ii) indirect DNA damage via reactive oxygen species (ROS). Ionizing radiation (IR) releases high-energy photons that disrupt the DNA structure of tumor cells, leading to base modifications and single- or double-strand breaks. These DNA lesions result in cell cycle arrest, apoptosis or necrosis, directly impacting tumor cell survival (2). In addition to DNA damage, IR induces the decomposition of intracellular water, generating substantial hydroxyl radicals (OH·) and other ROS species. IR causes a redox imbalance within cells, which leads to lipid peroxidation, protein denaturation and DNA damage, further disrupting normal cellular processes, particularly mitochondrial function (3). With advancements in computational modeling, image-guided technologies and biochemical agents, radiotherapy has been increasingly optimized and currently demonstrates distinct advantages in combination with other therapies. While previous studies have primarily focused on the mechanisms underlying DNA damage from radiotherapy (4,5), recent research on tumor hypoxia and metabolic reprogramming has shifted attention toward ROS-induced damage and mitochondrial dysfunction resulting from redox imbalance, which have become key areas of investigation (6,7).
Ferroptosis is a newly identified form of programmed cell death characterized by iron-dependent lipid peroxidation, accompanied by an immunogenic response. This form of cell death is intricately linked to the cellular metabolic state and redox balance, serving a critical role in the regulation of radiotherapy efficacy (8). IR promotes the generation of ROS, which directly damage nucleic acids and lipids in tumor cells, thereby triggering lipid peroxidation and ferroptosis (9). In the context of therapeutic resistance, particularly in chemotherapy, radiotherapy and immunotherapy, tumor cells (and especially those in a mesenchymal state with higher metastatic potential) become more susceptible to ferroptosis due to mitochondrial metabolic alterations dependent on the tumor microenvironment (TME) (10). Ferroptosis emerges as a promising pathway for enhancing radiation sensitivity, facilitated by the combined effects of mitochondrial-mediated redox reactions, lipid peroxidation and dysregulated iron metabolism. This offers notable clinical potential for overcoming resistance to conventional therapies (11,12).
Mitochondria are not only the powerhouse of the cell but also central hubs for redox reactions, iron metabolism and lipid peroxidation (13). Mitochondrial involvement in ferroptosis extends beyond regulating redox imbalance, as they are also crucial in ROS accumulation (14). As a result, mitochondria serve a pivotal role in maintaining intracellular redox equilibrium and are essential for executing ferroptotic cell death. In the context of radiotherapy, mitochondrial dysfunction and redox imbalance further drive ferroptosis, while the immunogenic response triggered by ferroptosis provides new opportunities for enhancing radioimmunotherapy. Ferroptosis is not merely a form of cell death; it elicits an immune response that activates immune cells within the TME, thereby improving the therapeutic efficacy of radiotherapy (15).
Despite these findings, the reciprocal regulatory mechanisms between mitochondria and ferroptosis, as well as the associated immune activation in the context of radiotherapy, remain incompletely understood. As research into the association between mitochondria and ferroptosis advances, targeting mitochondrial ferroptosis may become a key strategy for radiation immunosensitization. The present review explored the mechanisms underlying the interaction between ferroptosis and mitochondria under IR, and discussed the potential for targeting mitochondrial ferroptosis as a strategy for enhancing radioimmunotherapy, aiming to uncover the clinical importance of mitochondrial ferroptosis in radiation-based cancer treatment.
The incidence and mortality rates of cancer continue to rise, and radiotherapy, a cornerstone in cancer treatment, has been developed and use in the clinic for >100 years (16). Several common cancer types, including nasopharyngeal carcinoma and esophageal cancer, can be effectively treated with radiotherapy, either as a monotherapy or in combination with chemotherapy, immunotherapy and targeted-therapy (17). However, a subset of patients still experiences recurrence or metastasis, presenting a persistent challenge to the efficacy of clinical radiotherapy. Radiotherapy dosage is a crucial determinant of tumor control rates; however, high radiation doses may lead to significant damage to surrounding normal tissues and organs. Therefore, optimizing the radiation dose, enhancing tumor radiosensitivity and minimizing toxic side effects remain critical challenges yet unresolved in the field of radiotherapy (18,19).
Previous research has indicated that the primary mechanism of tumor cell death induced by radiotherapy is through IR-induced proliferative cell death, which includes apoptosis, autophagy and necrosis (20). However, increasing attention has been paid to the potential role of other forms of regulated cell death (RCD) in radiotherapy's therapeutic effects (21,22). Ferroptosis, a form of radiation-induced cell death, has emerged as an important contributor to tumor response, and exhibits characteristics of ICD, which is intricately linked with radioimmunotherapy (23). The hallmark of ferroptosis is iron-dependent lipid peroxidation, particularly of polyunsaturated fatty acid (PUFA)-phospholipids (PLs), accompanied by characteristic mitochondrial morphological changes such as shrinkage, loss or reduction of mitochondrial cristae and increased mitochondrial membrane density. In the context of radiotherapy, IR induces immune system recognition and activation by releasing specific immune stimulants, such as High Mobility Group Box 1 Protein (HMGB1, a nuclear protein promotes late-stage inflammation), ATP and exposed cell membrane antigens, thus triggering ICD and activating CD8+ T cells. IFN-γ produced by CD8+ T cells can alter the lipid metabolic profile of tumor cells, promoting ferroptosis through an ACSL4-dependent pathway (24).
Notably, ferroptosis exhibits crosstalk with various forms of RCD, particularly under oxidative stress conditions, a hallmark of radiation exposure. NADPH oxidase (NOX), a key regulator of lipid redox signaling, serves dual roles in both apoptotic pathways and ferroptosis initiation through lipid peroxidation induction (25). During bortezomib-induced apoptosis, ACSL4 inactivation may suppress the incorporation of PUFAs into cell membranes, consequently diminishing ferroptotic susceptibility. Furthermore, selective autophagy-mediated GPX4 degradation results in intracellular accumulation of free iron and lipid peroxides, ultimately driving ferroptosis progression (26,27).
Radiation-induced ferroptosis not only interacts with other RCD mechanisms, such as apoptosis and autophagy, to modulate tumor cell radiosensitivity, but also reshapes the TME by enhancing the activation and infiltration of immune cells (28). This immune activation, in turn, further sensitizes the tumor to radiotherapy, especially in radioimmunotherapy strategies (28). Specifically, radiotherapy-induced ferroptosis enhances the uptake of tumor antigens by dendritic cells (DCs), thus boosting tumor-specific T cell responses. Additionally, ferroptosis can modulate the activity of myeloid-derived suppressor cells (MDSCs) and regulatory T cells (Tregs), thus reshaping the immune landscape of the TME and preventing immune escape (29).
Research into the regulatory pathways of ferroptosis has identified several defense systems with distinct subcellular localizations, including Solute Carrier Family 7 Member 11 (SLC7A11, an antiporter that imports cystine for glutathione synthesis)-glutathione (GSH)-glutathione peroxidase 4 (GPX4), NAD(P)H-FSP1-CoQ, GCH1-BH4-DHFR, sex hormone-MBOAT1/2 and mitochondrial-localized dihydroorotate dehydrogenase (DHODH)-CoQH2 (30). These defense systems regulate key processes such as iron metabolism, redox balance and lipid peroxidation in different cellular compartments, collectively maintaining cellular homeostasis (Fig. 1). When these defense mechanisms are disrupted, cells become more vulnerable to ferroptosis, particularly metabolically active tumor cells (31). Inhibiting key antioxidant enzymes and iron homeostasis regulators can significantly enhance the efficacy of radiotherapy. Consequently, targeting the ferroptosis defense systems has emerged as an effective strategy for radiosensitization in malignant tumors.
Mitochondria are dynamic organelles with a double-membrane structure, serving a pivotal role in maintaining cellular homeostasis through multiple pathways, including the kallikrein system, the unfolded protein response, autophagy and various metabolic processes (32). However, common mitochondrial defects in tumor cells can significantly alter energy production, shifting from oxidative phosphorylation (OXPHOS) to a more simplified, oxygen-independent glycolysis pathway, in order to meet the high metabolic demands of rapid proliferation and the hypoxic, acidic microenvironment typical of tumors (33). These mitochondrial defects also downregulate the mitochondrial apoptosis pathway, a key factor that enables tumor cells to sustain their malignant phenotype and develop resistance to radiation. IR induces serious damage to the mitochondrial membrane, altering its permeability and leading to mitochondrial swelling, vacuolization and fragmentation of the cristae. The damage impairs the electron transport chain (ETC) and results in ROS accumulation (34). The dysfunction of mitochondria caused by IR can be further amplified through mitochondrial fusion and fission processes, which help dilute the damaged mitochondria. This leads to the genetic transmission of oxidative stress damage within the cell, causing side effects, exacerbating instability in both mitochondrial and nuclear DNA (35). In response to IR, tumor cells compensate by increasing the number of mitochondria and activating repair mechanisms to offset declines in mitochondrial respiratory efficiency, partially restoring mitochondrial function after IR-induced damage (36). However, this compensatory response leads to sustained ROS production, excessive depletion of mitochondrial antioxidants such as manganese superoxide dismutase and coenzyme Q, which further aggravates mitochondrial damage and generates more ROS, creating a vicious cycle that increases the risk of cell death (37).
In a microenvironment with persistently high ROS levels, tumor cells undergo long-term damage, leading to cellular senescence after repeated irradiation and exacerbating secondary radiation damage (38). Mitochondria play a critical role in cellular metabolic plasticity and regulate various forms of RCD, including ferroptosis, following radiation injury. Irreversible mitochondrial outer membrane permeabilization (MOMP) results in the release of pro-apoptotic factors such as cytochrome c from the intermembrane space into the cytoplasm, which activates the caspase-9-dependent intrinsic apoptosis pathway (39). Furthermore, mitochondrial DNA (mtDNA) released into the cytoplasm is recognized by cyclic GMP-AMP synthase (cGAS), which activates the cGAS-STING inflammatory pathway, triggering pyroptosis and inhibiting mitochondrial autophagy, thereby exacerbating mitochondrial damage (40). In addition to acting as a cytoplasmic DNA sensor that activates innate immune responses, cGAS also contains a mitochondrial targeting sequence, enabling its localization to the mitochondrial outer membrane (OMM), where it plays a protective role against ferroptosis (41). As IR intensifies its effects on mitochondrial function in tumor cells, structural and metabolic alterations in mitochondria significantly enhance ferroptosis, supporting the potential of ferroptosis in radioimmunotherapy strategies.
Mitophagy, a selective form of macroautophagy that induces the degradation of dysfunctional and damaged mitochondria via the autophagy pathway. Mitophagy serves a key role in maintaining cellular homeostasis (42). Protein disulfide isomerase (PDI), an enzyme primarily located in the endoplasmic reticulum (ER), mediates disulfide bond formation and is often highly expressed in tumor tissues, where its elevated levels correlate with increased pathological grading (43). In colorectal cancer (CRC) cells subjected to radiation-induced ER stress, PDI not only inhibits autophagy initiation by activating the GRP78-mTOR pathway but also competes with LC3Ⅱ for binding to the mitochondrial autophagy receptor PHB2, thereby regulating the mitophagy process and reducing radiation sensitivity (44). Specifically, PDI plays a critical role in balancing ferroptosis and mitophagy. During ferroptosis, PDI modulates cellular iron homeostasis and redox reactions by influencing antioxidant activity. Additionally, through its dual localization in both the ER and mitochondria, PDI regulates the ER stress response and impacts mitophagy initiation by interacting with PHB2 (44). Through these mechanisms, PDI not only suppresses autophagy initiation but also plays a pivotal role in regulating ferroptosis.
Functionally intact and structurally complete mitochondria are crucial in defending against ferroptosis through various metabolic and antioxidant pathways (45). On the OMM, voltage-dependent anion channels (VDAC) 2/3, iron transporter proteins Mfrn1/Mfrn2, the mitochondrial permeability transition pore and the mitochondrial Ca2+ uniporter facilitate the physiological uptake of Fe2+, maintaining a labile iron pool (46). Mitochondrial ferritin (FtMt) plays a pivotal role in regulating iron metabolism within the mitochondrial matrix. It is involved in the synthesis of iron-sulfur (Fe/S) cluster proteins, such as NADH dehydrogenase, cytochrome c and succinate dehydrogenase, thereby maintaining ATP production and preventing the accumulation of mitochondrial ROS (mtROS) (47). FtMt also regulates the availability of free iron in mitochondria, mitigating iron overload, oxidative stress and ferroptosis under conditions such as cerebral ischemia-reperfusion and IR injury. The Fe/S cluster proteins mitoNEET and frataxin on the OMM are similarly essential for maintaining mitochondrial iron metabolism balance and inhibiting ferroptosis. In addition to iron metabolism, cellular energy metabolic pathways, including the tricarboxylic acid cycle (TCA) and OXPHOS, as well as associated regulatory factors such as NADPH, GSH and AMPK, all play critical roles in ferroptosis regulation (48). For example, the mitochondrial transmembrane protein MTCH1 stabilizes the mitochondrial OXPHOS cycle and mtROS levels; its deficiency activates pro-ferroptotic retrograde signaling involving the FoxO1-GPX4 axis, which synergistically enhances the antitumor activity of ferroptosis-inducing agents such as sorafenib (49).
Mitochondrial cysteine metabolism also plays a central role in ferroptosis regulation. The cysteine desulfurase complex NFS1-ISD11 provides sulfur for Fe-S cluster formation and promotes GSH synthesis, thus upregulating the activity of the GSH-GPX4 antioxidant system (50). The hereditary inactivation of fumarase, a key enzyme in the TCA cycle, induces ferroptosis by downregulating GPX4 activity in conditions such as smooth muscle tumors and renal cell carcinoma (51). NADPH, which is critical for GSH conversion in the mitochondrial TCA cycle, is catalyzed by NADP+-dependent isocitrate dehydrogenase (IDH), and its cytoplasmic source can be provided by the reversible oxidative decarboxylation of malate to pyruvate catalyzed by malic enzyme 1 (ME1). NADPH not only drives ATP release through coupling with OXPHOS but also maintains the GSH cycle across various metabolic pathways, thus counteracting mitochondrial lipid peroxidation and reducing susceptibility to ferroptosis (51).
The ETC complexes (complexes I-IV) and ATP synthase (complex V) stimulate ATP synthesis through phosphocreatine and activate the AMPK signaling pathway. Inhibiting acetyl-CoA carboxylase phosphorylation and PUFA synthesis also negatively regulates mitochondrial ferroptosis, exerting similar effects to those of the LKB1-AMPK axis (52). Similarly, enzymes targeting mitochondrial CoQ reduction, such as DHODH and G3P dehydrogenase 2 (GPD2), inhibit mitochondrial lipid peroxidation and ferroptosis by reducing CoQ to CoQH2 (53). GPD2 further synergizes with mtGPX4, leading to the acetylation of mtGPX4 and contributing to cadmium-induced renal cell ferroptosis (54). The role of mitochondrial supercomplexes formation and its regulatory factor COX7A2L in ferroptosis sensitivity has also been confirmed (55). COX7A2L deficiency induces mitochondrial metabolic reprogramming, enhancing glutamine (GLN) metabolism, cell proliferation and oxidative defense against ferroptosis (56). Furthermore, several regulatory factors that are localized in or translocated to mitochondria after activation also serve a key role in mitochondrial ferroptosis. Mitochondrial inner membrane-localized PDK4 inhibits ferroptosis by blocking pyruvate dehydrogenase-dependent pyruvate oxidation and fatty acid synthesis. However, the roles of AMPK and PDK4 in ferroptosis are cell-type-dependent, with complex interactions between energy status and ferroptosis induced by different activators (57). The mitochondrial metabolic enzyme PCK2 phosphorylates ACSL4 and promotes ACSL4-associated PL remodeling in tumor cells. Cancer stem cells promote PCK2 ubiquitination and degradation, thus maintaining membrane PLs in a ferroptosis-tolerant state (58). Activated by PLC kinase, monocarboxylate transporter 1 (MCT1) translocates to the mitochondrial surface, mediating lactate uptake, promoting ATP production and inactivating AMPK, which increases the expression of sterol regulatory element-binding protein 1, stearoyl-CoA desaturase 1 and the end products monounsaturated fatty acids, thereby contributing to ferroptosis resistance (59). Furthermore, lithium carbonate-induced MCT1 translocation and Ca2+ release activate LA oxidation in mitochondria, reactivating LA-suppressive CD8+ T effector cells (60). Mitochondria serve a key role in cysteine deprivation-induced (CDI) ferroptosis, which is distinct from GPX4 inhibition-induced ferroptosis, with the TCA cycle and amino acid metabolism pathways closely linked to CDI ferroptosis (61). GLN, a key respiratory substrate for energy production and lipid synthesis in tumor cells, is degraded by mitochondrial glutaminases (GLS) 1/2. Due to their similar amino acid sequences encoded by unrelated genes, reduction of GLS2, rather than GLS1, through genetic or pharmacological mechanisms, inhibits CDI ferroptosis. Aminooxyacetate inhibits the conversion of GLN to α-ketoglutarate, rescuing CDI ferroptosis in tumor cells (61).
Mitochondria-associated ER membranes (MAMs) are highly dynamic and tightly interconnected structures between the mitochondria and the ER. MAMs serve as a platform for Ca2+ signaling between the two organelles and serve a vital role in regulating mitochondrial ferroptosis signaling (62). Mitochondrial microsomal glutathione S-transferase 1 limits lipid peroxidation and ferroptosis by binding to and downregulating ALOX5 activity (63). The calcium-sensitive receptor chaperone protein σ1R mediates the inhibitory effect of the kinase inhibitor CGI1746 on ferroptosis. CGI1746 also slows the Ca2+ transport rate within MAMs, disrupting the ferroptosis process (64). The IP3R-GRP75-VDAC1 axis is a key regulatory pathway for the dynamic changes in MAMs, and its inhibition leads to the accumulation of PUFA-containing triacylglycerols, conferring resistance to ferroptosis (65). Mitofusin-2 (Mfn-2), originating from both the mitochondrial membrane and the ER surface, mediates physical and biochemical interactions between the mitochondria and the ER. Mfn-2 serves a key role in mitochondrial fusion and stabilizing mitochondrial-ER contact sites (66). Mfn-2 interacts with the ER-resident protein inositol-requiring enzyme 1α (IRE1α) to participate in arsenic-induced ferroptosis, while IRE1α also influences ferroptosis sensitivity by regulating GSH synthesis (67). Deficiency in the autophagy regulator WIPI4 increases the localization of ATG2 to mitochondria and MAMs, which enhances the transfer of phosphatidylserine within MAMs and the ATG2-dependent synthesis of mitochondrial phosphatidylethanolamine (PE) by phosphatidylserine decarboxylase (PISD), ultimately triggering mitochondrial membrane lipid peroxidation and ferroptosis (68). Inhibition of PISD protects cells from ferroptosis induced by WIPI4 depletion and other inducers, suggesting that PE synthesis in mitochondria may represent a general early step in ferroptosis (69).
The regulatory role of mitochondrial metabolic pathways in ferroptosis varies across different tumor types (70). For instance, abnormal NADPH production and regulation may result in notable differences in ferroptotic effects across malignancies (71). Enzymes involved in NADPH production, such as IDH and ME1, exhibit mutations or deletions in different cancer types, leading to an imbalance in the antioxidant system (72). Specifically, in cholangiocarcinoma, IDH1 mutations promote ferroptosis by lowering GPX4 expression and accelerating GSH depletion (73). In synovial sarcoma, ME1 deficiency alters the mitochondrial NADPH-dependent antioxidant system, increasing susceptibility to ferroptosis induced by ACXT-3102 (74). MAMs also display different ferroptosis sensitivities under various treatments within the same tissue type. mtROS activate the PERK pathway, suppress Mfn-2 expression and cause MAM dysfunction, contributing to arsenic-induced ferroptosis in liver cells. Upregulation of PERK-c fragment expression in MAMs can trigger alcoholic liver ferroptosis, a process inhibited by quercetin (75). Thus, research into mitochondrial metabolic pathways and MAMs regulatory mechanisms across tumor types could offer novel insights into their susceptibility and resistance to ferroptosis, providing potential therapeutic targets for clinical treatment.
Overall, the regulatory role of mitochondria in ferroptosis encompasses iron metabolism, energy metabolism, antioxidant regulation and transmembrane signaling with the ER. These pathways and mechanisms exhibit marked variability across different tumor types. A deeper understanding of these differences could provide personalized therapeutic strategies for ferroptosis-related anticancer treatments.
IR induces ferroptosis in tumor cells through multiple mechanisms, which is closely tied to mitochondrial metabolic reprogramming (15). Upon exposure to IR, mitochondria in tumor cells undergo shrinkage and vacuolization, while key components of the cysteine/glutamate antiporter system (system XC-), including SLC3A2 and SLC7A11, are significantly downregulated. This downregulation reduces the synthesis of mtGPX4, thereby weakening the mitochondria's ability to counter lipid peroxidation (76). IR also enhances tumor cell glucose and GLN metabolism, dose-dependently increasing ROS production, which leads to mitochondrial energy stress. In addition, IR upregulates the expression of the iron transporter SLC39A14 and the key enzyme ACSL4, which is involved in PUFA-PLs metabolism, thus inducing lipid peroxidation (77). Furthermore, a reduction in mitochondrial membrane potential and the proteolytic cleavage of PTEN-induced kinase 1 (PINK1) by the inner membrane protease, presenilin-associated rhomboid-like protease, leads to the stabilization of full-length PINK1 on the OMM. This stabilization recruits the E3 ubiquitin ligase Parkin from the cytoplasm, promoting the ubiquitination of OMM proteins, including VDAC1 and mitochondrial fusion proteins, Mfn-1/2. p62 is subsequently recruited to the mitochondrial surface, initiating mitophagy (78). Lipid droplets (LDs), which serve as cellular organelles for fatty acid storage, are degraded by lysosomes via autophagy under radiation stress, increasing the release of free fatty acids (FFAs) and the risk of lipid peroxidation (79). IR also induces an increase in LD volume and their proximity to mitochondria, eventually forming mitochondrial-LD complexes that facilitate fatty acid transfer to mitochondria (80,81). Key components of this process, such as the LD-anchor protein PLIN5 and carnitine palmitoyltransferase 1, help mitigate lipid accumulation in LDs, thus enhancing mitochondrial fatty acid oxidation (FAO) and the risk of ferroptosis (82). Through the PINK1/Parkin-mediated mitophagy pathway, damaged mitochondrial-LD complexes are engulfed and degraded by lysosomes, and the released FFAs further accelerate the ferroptosis process (83). Additionally, the PINK1/Parkin pathway promotes the ubiquitin-mediated degradation of the mitochondrial iron transporters SLC25A37 and SLC25A28, which enhances mitochondrial sensitivity to ferroptosis. Inhibition of this pathway leads to disrupted mitochondrial iron-dependent immune metabolic function (84).
Hypoxic microenvironments markedly diminish the radiosensitivity of tumor cells and are a major factor contributing to radiation resistance (85). Hypoxia induces the expression of BCL2-interacting protein 3 (BNIP3), which integrates into and anchors to the OMM. By competitively binding to the BH3 domain and activating the PI3K pathway, BNIP3 promotes the initiation of mitophagy (86). Under hypoxic conditions, ROS production increases, promoting the formation of PUFA-PLs through the hypoxia inducible factors (HIFs)-hypoxia inducible lipid droplet associated (HILPDA) axis. The LD-associated protein HILPDA remodels the cellular lipidome, particularly altering the levels of mitochondrial cardiolipin (CL). The HILPDA-CLS1 axis further enhances the accumulation of CL, which promotes mitophagy induced by IR (87). Therefore, mitophagy-dependent ferroptosis may represent a key pathway for enhancing radiosensitivity in hypoxic solid tumors. Furthermore, IR activates multiple signaling pathways that regulate mitochondrial metabolic reprogramming and ferroptosis. These pathways, including AMPK/NF-κB, JAK2/STAT3 and PI3K/Akt/mTOR, activate the reprogramming of mitochondrial glycolysis and lipid metabolism, thereby enhancing ferroptosis susceptibility (3).
Mitochondrial activity-driven innate and adaptive immune responses also serve a key role in the IR-mediated enhancement of ferroptosis sensitivity in tumor cells (15). Upon IR exposure, the mitochondrial metabolism of immune cells, such as T cells, monocytes, macrophages and B cells, becomes increasingly reliant on FAO as a substrate. This metabolic shift serves as a critical endogenous trigger for T cell exhaustion (88). Elevation of FAO leads to mitochondrial depolarization, impaired biosynthesis and excessive ROS production, thereby inducing oxidative stress. Furthermore, FAO inhibits the proteasomal degradation of HIF1α, mediating the glycolytic reprogramming of precursor exhausted T cells. This reprogramming drives their transcriptional and metabolic transformation into terminally exhausted T cells (89).
The newly identified metabolic reprogramming and autophagy pathways offer valuable insights into how modulation of mitochondrial function and ferroptosis mechanisms can enhance the efficacy of clinical radiotherapy for radiation-resistant tumors. These findings potentially provide novel therapeutic targets and sensitization strategies for radioimmunotherapy (Fig. 2).
Mitochondrial dysfunction in various cell types, including tumor cells and immune cells within the TME, serves a pivotal role in tumor progression and immune evasion (90). Specifically, during T cell activation, naïve T cells undergo a metabolic shift from OXPHOS and FAO to aerobic glycolysis and fatty acid synthesis. During this mitochondrial reprogramming, mitochondria accumulate beneath the activated T cell receptor clusters, promoting mitochondrial fission and concurrent relaxation of the inner membrane cristae. This process stimulates mitochondrial activity, including increased production of mtROS and ATP synthesis (91). This metabolic transition is essential for T cell activation and supports their short-term immune responses. However, as T cells differentiate into memory T cells or Tregs, mitochondrial metabolism gradually shifts back to OXPHOS and FAO to support cell survival, maintain cellular phenotype and ensure immune tolerance or immunosuppressive functions (76). In the TME, tumor-infiltrating T cells exposed to chronic hypoxia environments sustained mitochondrial dysfunction due to aberrant activation of the Akt1-PGC1α signaling pathway, leading to persistent mitochondrial damage. This metabolic dysfunction weakens T cell anti-tumor immunity, inhibits cytokine secretion and ultimately facilitates tumor immune evasion (92). Similarly, the immune response of natural killer (NK) cells is significantly influenced by mitochondrial metabolic alterations. During NK cell activation, glycolytic capacity and basal OXPHOS rates increase remarkably. However, in the transition to memory NK cells, damaged mitochondria are removed via mitophagy-related proteins such as BNIP3(L), thereby reducing mtROS production and supporting the formation of memory NK cells (93). By contrast, within a hypoxic TME, tumor-infiltrating NK cells exhibit severe mitochondrial fragmentation, which impairs their tumor-killing capacity and immune surveillance function, thus promoting tumor immune evasion (94). Furthermore, mitochondrial metabolic states profoundly impact macrophage polarization and function within the TME. Upon M1 polarization, macrophages shift their mitochondrial metabolism from OXPHOS to aerobic glycolysis, accompanied by increased mitochondrial fission and elevated mtROS production. By contrast, M2 polarization is characterized by enhanced mitochondrial fusion, increased OXPHOS and FAO (95). These metabolic reprogramming processes not only govern macrophage polarization but also directly regulate their immune functions and responses to tumor progression. Therefore, dynamic alterations in mitochondrial metabolic states and morphology have a direct impact on immune cell polarization and immune efficacy within the TME, serving a crucial role in tumor immune evasion (Fig. 3).
IR-induced mitochondrial stress triggers the release of mtDNA into the cytoplasm and extracellular space, where it functions as a damage-associated molecular pattern (DAMP) that activates pattern recognition receptors such as Toll-like receptors (TLRs) and NOD-like receptors (NLRs). This activation sets off a cascade of innate immune signaling (27). Notably, NLRP3 recognizes cytosolic mtDNA, initiating caspase-1 activation, and promoting the cleavage and maturation of the inflammatory cytokines IL-1 and IL-18. These cytokines recruit T lymphocytes, macrophages and neutrophils, thereby amplifying the immune response (96). In normal cells, caspase-dependent mitophagy serves to mitigate immune signaling induced by mtDNA by clearing damaged mitochondria. However, under pathological conditions, impaired mitophagy or caspase-dependent mtDNA release may convert immunologically silent cell death into ICD. In the absence of caspases, MOMP activates the mtDNA-STING pathway, initiating antitumor immune responses (97). IR also upregulates the expression of mitochondrial fission protein 1 Drp1, contributing to mitochondrial dysfunction and subsequent mtDNA release. This extracellular mtDNA activates immune cells via the cGAS-STING and TLR9-NF-κB pathways, modulating the polarization and function of immune cells, including T lymphocytes, macrophages and DCs (98). Thus, IR-induced ICD and released mtDNA serves a dual role in tumor immune evasion and activation. On one hand, mtDNA enhances the host's antitumor immune response by stimulating immune reactions. On the other hand, IR-induced ICD may also foster tumor immune evasion. Therefore, IR not only activates immune responses through mtDNA release but also modulates immune evasion via mitophagy regulation (99).
ROS serve a pivotal role in tumorigenesis, particularly in the inflammatory TME, where they contribute to the formation of immune-suppressive environments (100). Elevated ROS levels inhibit the formation of TCR-antigen peptide-MHC complexes on the surface of infiltrating T cells, thereby impairing T cell activation and facilitating tumor immune escape (101). mtROS regulate immune cell phenotypes and functions, thus supporting the establishment of an immune-suppressive TME (102). For instance, HIF-1α-induced mitochondrial Lon protease promotes mtROS production through interaction with pyrroline-5-carboxylate reductase (P5CRs), which in turn activates the NF-κB pathway, leading to the secretion of immune-suppressive cytokines such as IFN-γ and TGF-β by tumor cells (103). In addition, mtROS can further enhance Lon protease expression, activate the mitochondrial Na+/Ca2+ exchanger and regulate mitochondrial calcium efflux, thus improving tumor cell resistance to cisplatin (24). The central role of mtROS in immune evasion underscores the importance of understanding the complex regulatory mechanisms between mtROS and immune cell activation in the TME. It is crucial for enhancing radioimmunotherapy through mitochondrial ferroptosis sensitization. Tumor-associated fibroblasts amplify oxidative stress in surrounding monocytes, promoting their differentiation into MDSCs, which in turn inhibit CD8+ T cell proliferation (104). Elevated ROS levels, through pathways involving HIF-1α and Lon protease, contribute to the generation of immune-suppressive cells such as MDSCs, tumor-associated macrophages (TAMs) and Tregs, thereby further promoting immune suppression (105). ROS also alter the immunogenicity of antigen peptides in antigen-presenting cells, affecting the interaction between damage-associated molecular patterns (DAMPs) and DC receptors, thus inhibiting antitumor immunity triggered by ICD. Conversely, in the antitumor immune response, activated T lymphocytes and NK cells utilize ROS to recruit neutrophils and macrophages, thereby enhancing tumor cell killing. In reduced GSH-deficient Tregs, abnormal serine metabolism leads to oxidative stress, which downregulates Foxp3 expression and impairs the immune-suppressive function of Tregs (106). Therefore, the levels and sustained production of ROS in both tumor cells and immune cell subsets are critical factors in determining the occurrence of ICD and its capacity to induce effective antitumor immunity.
Macrophages are key immune cells within the TME, serving a crucial role in maintaining immune homeostasis during tumor progression. Tumor cells remodel the surrounding and distal microenvironments by secreting tumor-derived factors that activate monocytes and macrophages in circulation or local tissues, thereby accelerating tumor progression (107,108). While mtROS were once regarded as harmful byproducts of mitochondrial metabolism, recent research underscores their signaling roles in preventing excessive immune responses, particularly in regulating macrophage immune functions (109). A previous study has shown that mitochondrial Lon protease expression is elevated in M2-type macrophages, suggesting that, during tumorigenesis, macrophages regulate Lon expression through multiple signals to induce mtROS generation, thereby serving a critical role in the differentiation of TAMs (110).
Radiotherapy enhances antitumor immunity through multiple mechanisms, notably inducing tumor ICD, which activates adaptive anti-tumor immune responses. IR promotes the release of double-stranded DNA (dsDNA) from the nucleus, leading to its accumulation in tumor-derived exosomes and increasing OMM permeability, thereby exposing mtDNA in the cytoplasm (111). As potent mediators, dsDNA and mtDNA initiate the cGAS-STING pathway, upregulating IFN I transcription, which activates the innate immune system, promotes T-cell activation and suppresses tumor growth. Additionally, IR induces the expression of MHC class I molecules and tumor-associated antigens on tumor cell surfaces, while increasing the exposure of calreticulin, HMGB1 and ATP, expanding the antigen pool for presentation. This further enhances tumor immunogenicity and promotes immune responses within the TME (112). Furthermore, IR stimulates the release of inflammatory mediators and chemokines, such as CXCL9, CXCL10 and CXCL11, from tumor and stromal cells. These factors increase the infiltration of DCs, macrophages and T cells, triggering both local and systemic antitumor immune responses (113). These immune-enhancing mechanisms suggest that radiotherapy not only exerts direct cytotoxic effects on tumor cells but also significantly activates the host immune system by reshaping the TME. When combined with anti-PD-L1 immunotherapy, IR further reduces the accumulation of MDSCs in the TME, creating a more inflammatory environment and effectively reversing immunosuppression (114).
However, under certain dosing schedules and time gradients, radiotherapy may also induce immunosuppressive effects (115). These effects, driven by alterations in the tumor hypoxic microenvironment, particularly metabolic reprogramming and expansion of immune-suppressive cells such as Tregs, may impair therapeutic efficacy and promote tumor immune escape. IR can activate the GPR81/mTOR/HIF-1α/STAT3 pathway by enhancing tumor cell glycolysis, which in turn promotes MDSC activation in the TME (116). Additionally, tumor metabolic reprogramming increases mitochondrial FAO, providing ATP through mitochondrial catabolism, thus facilitating tumor cell radioresistance. FAO upregulates CD47 transcription via the citrate-acetyl-CoA-RelA pathway, further contributing to immunosuppression by protecting radioresistant tumor cells from macrophage-mediated attack (116). The sustained activation of HIF-1α by IR not only maintains the hypoxic microenvironment but also drives macrophage subtypes (such as Macro-HK2, Macro-HSP and Macro-MT) toward a pro-angiogenic phenotype. These macrophages secrete additional chemokines, which recruit immunosuppressive neutrophils after radiotherapy (117).
Under conventional fractionated radiotherapy, sufficient radiotoxicity depletes circulating peripheral blood monocytes and induces tumor cells to secrete Gal-1, which promotes T-cell apoptosis (118). Tumor cells subjected to repeated irradiation induce chronic expression of FN I and IFN-γ, upregulating PD-L1 and IDO, leading to T-cell exhaustion. This process is further enhanced by the IR-activated cGAS-STING signaling pathway, which mobilizes Tregs and MDSCs while inhibiting effector T-cell function (119). In local radiotherapy, IR upregulates the secretion of CCL2 and CCL5, recruiting monocytes and activating Tregs in a TNF-α-dependent manner, further diminishing the therapeutic effect of radiotherapy (120). In later stages of radiotherapy, the proportion of terminally exhausted CD8+ T cells increases, along with upregulated exhaustion markers such as Pdcd1, Lag3 and Tigit. This is accompanied by a limited maturation of DCs, impairing their ability to activate CD4+/CD8+ T cells and exacerbating T-cell dysfunction (121).
Although radiotherapy combined with anti-PD-1 antibodies enhances tumor control via a CD8+ T-cell-dependent mechanism in certain therapeutic regimens, low-dose fractionated radiotherapy (such as 2 Gy x10) may inhibit IFN-γ expression in tumor-specific CD8+ T cells within draining LNs. This effect is closely linked to radiation-induced lymphopenia and the immunosuppressive TME (122). In localized radiotherapy, selective lymph node irradiation (which is effective in addressing subclinical lymph node micrometastases) may contribute to an immunosuppressive TME when combined with anti-CTLA-4 antibodies. This combination upregulates PD-L1 expression, leading to T-cell exhaustion and radioresistance (123).
Among the immunomodulatory mechanisms of radiotherapy, ferroptosis has emerged as a critical effector pathway. Ferroptosis is not only a form of cell death but also a potential mechanism through which radiotherapy induces ICD, thereby activating antitumor immunity (124). IR induces ICD, prompting activated CD8+ T cells in the TME to release IFN-γ. This process inhibits system XC-(SLC3A2 and SLC7A11) expression, reduces cysteine uptake by tumor cells and impairs the chelation of GSH-cisplatin complexes, thus diminishing the ferroptotic defense system and potentially reversing cisplatin-based chemoresistance (125). Furthermore, IR remodels the lipid profile of tumor cells, increasing the incorporation of linoleic acid into C16- and C18-acyl PLs in an ACSL4-dependent manner, thereby inducing ferroptosis (126). IFN-γ secreted by Th1, NK and NKT cells can further promote ferroptosis by interacting with specific fatty acids in the TME, thereby enhancing antitumor immunity (127). This suggests a complex interplay between ferroptosis, radiotherapy and immune responses, presenting novel therapeutic targets for radioimmunotherapy strategies. Within tumor cells, DNA damage sensors (including ATRIP, Rad24p, γH2AX, NBS1, BRCA1/2, Ku70/80 and RNA polymerases) recognize radiation-induced damage signals and recruit core DNA damage response kinases ATM, ATR and DNA-dependent protein kinase (DNA-PK) to DNA break sites, initiating repair mechanisms (128). Previous research has shown that IR-activated ATM kinase, together with IFN-γ, synergistically suppresses SLC7A11 expression and cysteine uptake, promoting lipid peroxidation and ferroptosis, thus enhancing CD8+ T-cell-mediated antitumor immunity (129,130). This highlights the dual role of ATM in radiation biology. Inhibition of ferroptosis diminishes the efficacy of immune checkpoint inhibitors (ICIs) in combination with radiotherapy, while inactivation of SLC7A11 enhances the synergistic effects of ICIs and IR (131). Additionally, resistance to ferroptosis mediated by lysosomal exocytosis pathways has been negatively correlated with the efficacy of tumor radioimmunotherapy. Although previous studies suggest that tumor-derived CD8+ T cells exhibit elevated levels of lipid peroxidation within the TME, thereby increasing their susceptibility to ferroptosis (an important factor influencing radiotherapy efficacy), the exact mechanism by which DC function and antitumor immunity are influenced by radiotherapy-induced ferroptosis warrants further investigation (132-134). Ferroptosis in immune cell subsets can also modulate immune responses within the TME. Tumor cells damaged by radiation typically exhibit dysregulated lipid metabolism, while tumor-infiltrating T cells accumulate lipids in a CD36 receptor-dependent manner, increasing intracellular oxidized low-density lipoprotein levels, which induces lipid peroxidation and ferroptosis. Inhibition of CD36 reduces ferroptosis in CD8+ T cells, preserves antitumor immunity while maintaining radiation-induced damage and enhances the efficacy of immune checkpoint blockade (135). Activation of the ER stress response factor X-box binding protein 1 (XBP1) in DCs inhibits T cell-DC interactions by driving abnormal lipid accumulation. Silencing XBP1 improves the antitumor and immune-stimulatory functions of DCs, suggesting that targeting the ER stress response could enhance antitumor immunity and promote ferroptosis in tumor cells (136).
The role of granulocytes in ferroptosis within the TME is still under extensive investigation. Granulocytes isolated from mouse glioma models have been shown to transfer myeloperoxidase to tumor cells via cytoplasmic vesicles, thereby inducing lipid peroxidation products and triggering ferroptosis in tumor cells (137). Granulocytic myeloid-derived suppressor cells (G-MDSCs) in the TME are highly sensitive to ferroptosis. They release oxidized lipids that suppress T-cell activity, thereby contributing to immune evasion. Beyond G-MDSCs, T-cell subsets may also undergo ferroptosis under different environmental conditions (138,139). In Treg cells, the deletion of GPX4 following TCR signaling leads to lipid peroxidation and ferroptosis, which inhibits tumor growth and enhances antitumor immunity (140). By contrast, the deletion of GPX4 in CD8+ T cells results in the failure of antigen-stimulated T-cell proliferation, attributed to the rapid accumulation of membrane lipid peroxides and ferroptosis within the cells. Furthermore, the suppression of GPX4 expression in naïve CD4+ T cells prevents the generation of follicular helper T (Tfh) cells and impedes germinal center responses, limiting antibody production (141). Although T-cell proliferation in vitro depends on system XC-, this transporter is not an essential factor for T-cell proliferation or primary/memory immune responses in vivo, with cysteine's role potentially compensated by other mechanisms. Ferroptotic cells release surface-exposed oxidized lipid species, such as SAPE-OOH, which deliver key phagocytic signals, enhancing macrophage-mediated phagocytosis via TLR2 (142). However, the regulatory mechanisms of SAPE-OOH and the immunogenicity of the TLR pathway remain incompletely understood. Therefore, the composition of the TME serves a decisive role in determining whether ferroptosis induces immune suppression or activation. Before utilizing ferroptosis induction as a strategy to sensitize radioimmunotherapy, targeted therapies for specific immune cell populations may be required.
Inducing mitochondrial ferroptosis has emerged as a promising approach to enhance tumor radiosensitivity in hypoxic microenvironments (143). IR-induced incomplete MOMP is a critical factor contributing to radiation resistance. This process leads to the upregulation of phosphorylayed-eIF2α and ATF4, which exacerbates tumor radioresistance (144). Liposomal formulations containing metformin and 2-deoxyglucose, by targeting ATF4, effectively reverse radioresistance in CRC, preventing T lymphocyte exhaustion and enhancing the efficacy of fractionated radiotherapy through ferroptosis sensitization (145). PGC1α, a key regulator of mitochondrial metabolism, is recognized by DNA-PK under IR conditions, promoting its ubiquitination and degradation. This contributes to radioresistance in glioma subtypes. Therefore, the use of the PGC1α activator ZLM005 can reactivate mitochondrial ROS (mtROS) production and upregulate various forms of cell death, including ferroptosis, thereby enhancing radiosensitivity (146). Additionally, subcellular-targeted nanostructures, including DHODH inhibitors such as QSSP, can enhance radiosensitivity by disrupting the ferroptotic defense system. The mitochondria-targeted ferroptosis inducer IR780-SPhF, which exhibits specific GSH-responsive properties, induces mitochondrial redox imbalance, offering a novel treatment strategy for breast cancer (147). Tubastatin A inhibits GPX4 enzyme activity in a concentration- and time-dependent manner, synergistically enhancing ferroptosis and sensitizing breast cancer cells to radiotherapy (148).
Apart from targeting the ferroptosis defense system, regulating the concentration of metal ions such as Fe2+ within mitochondria serves a key role in enhancing ferroptosis-mediated radiosensitization (48). In nasopharyngeal carcinoma, previous research has demonstrated that nanocarriers targeting circular (circ) RNA circADARB1 can elevate intracellular iron ion concentrations, thereby promoting radiotherapy-induced ferroptosis and increasing radiosensitivity. Self-assembled pH-sensitive superparamagnetic iron oxide nanoclusters enhance mitochondrial-localized ferroptosis and apoptosis, synergistically sensitizing tumors to radiotherapy (149). Cationic nanometal-organic frameworks such as Th-Ir-DBB/digoxin, developed for ICD-induced ferroptosis, utilize mitochondrial targeting and cholesterol depletion to enhance the antitumor efficacy of radiotherapy-radiodynamic therapy (150).
In the era of radioimmunotherapy, radiation resistance and immune suppression within the TME often limit the effectiveness of radiation-induced antitumor immune responses. Ferroptosis has emerged as an effective strategy to enhance antitumor immunity in this context (Fig. 4) (151). Targeting macrophage membrane-coated biomimetic nanoparticles enables immune camouflage to evade phagocytosis by the mononuclear phagocyte system, damaging mitochondria and inducing ferroptosis in glioblastoma cells (152). The mitochondrial-targeting synthetic cluster Au25(S-TPP)18 inhibits the thioredoxin reductase system, enhancing mtROS production. This cluster exhibits potent radiosensitizing effects, induces a bystander effect and activates the immune response (153). Tri-metallic nanoparticles (AuBiCu-PEG NPs) with dual enzymatic activities not only promote ICD induced by radiotherapy and enhance antitumor immunity, but also effectively reverse radiation-induced PD-L1 upregulation. These NPs suppress tumor immune evasion, reshape the immune microenvironment and synergistically sensitize radiotherapy through X-ray deposition, hypoxia alleviation and the induction of ferroptosis and cuproptosis (154). Fe3O4-αPD-L1 nanoprobes, when combined with radiotherapy, specifically target MDSCs, modulating lipid and amino acid metabolism, particularly unsaturated fatty acids and antioxidant metabolites associated with ferroptosis. This approach reprograms the tumor immune-suppressive microenvironment, promoting both local and systemic antitumor immune responses (155). Equally important, iron-containing hyaluronic acid (HA) NPs, which internalize by high-affinity interactions with overexpressed CD44 receptors through HA, generate lipid ROS. When combined with radiotherapy, these nanoparticles induce both mitochondrial apoptosis and ferroptosis (156). Additionally, CSM nanoparticles loaded with single-atom nanocatalysts (SAzyme), and manganese carbon monoxide enhance FLASH radioimmunotherapy by inducing ferroptosis through multiple pathways, including GSH depletion and CD8+ T-cell infiltration (157). Other ferroptosis-inducing NPs, such as BZAMH NPs, 8HSA8 NPs, Au/CuNDs and Cu2WS4-PEG, also serve crucial roles in sensitization of radioimmunotherapy (158).
Radiopharmaceutical therapy (RPT), or targeted radionuclide therapy, directly treats malignant tumors by delivering radioactive atoms (β- or α-particles) to the tumor site (159). Compared with X-ray-mediated IR, RPT allows for precise delivery of radiation to cancer cells or the TME, maximizing tumor cell killing while sparing surrounding healthy tissues (160). For instance, 3-nm monocrystalline iron NPs (iRGD-bcc-USINP), when combined with a PD-L1 blocking agent, target ferroptosis and synergistically exert antitumor immune effects. However, ultra-small NPs (<6 nm) demonstrate poor tumor accumulation (161). Particle size-optimized NPs, assembled with radiopharmaceuticals such as USINP-131I-aPD-L1, promote ROS production and enhance ferroptosis induced by USINP, activating immune pathways and synergizing with PD-L1 blockade to enhance antitumor immune responses. Furthermore, the 124I-P/C@ CMLvs system, combined with PET imaging tracer, can synergistically enhance ferroptosis and necroptosis, inducing lung cancer regression and improving the efficacy of anti-PD-L1 immunotherapy (162).
In addition to enhancing radiotherapy, mitochondrial ferroptosis can also sensitize emerging tumor treatments, such as photothermal therapy (PTT), photodynamic therapy (PDT), and sonodynamic therapy (SDT) (163). The IR780/Ce@ EGCG/APT nanoreactor, by combining ferroptosis with PTT, improves the immune-suppressive microenvironment in breast cancer and activates systemic immune responses, leading to long-term immune memory. Mitochondria-specific type I PDT mediated by organic NIR-II photosensitizers (TPEQM-DMA) and two-photon nanosensitizers (IR-g-C3N4) disrupt the lipid repair system, resulting in mitochondrial dysfunction and induction of both apoptosis and ferroptosis, providing an effective phototherapy strategy for hypoxic tumors (164). Furthermore, the mitochondria-specific organic-metal ultrasound sensitizer IRCur-Pt enhances the synergistic effect of ferroptosis and SDT, further activating immune pathways. Pancreatic cancer-specific antibody NRP2-guided MOFs@ COF nanocarriers target and disrupt mitochondrial and ER homeostasis, inducing autophagy-dependent ferroptosis while sensitizing chemotherapy and SDT (165). In summary, inducing mitochondrial ferroptosis through the disruption of iron ion homeostasis and peroxidation intervention has emerged as an effective strategy to improve the radiosensitivity of malignant tumors and enhance the efficacy of radioimmunotherapy.
The present review summarized the research advancements in novel radioimmunotherapy strategies, focusing on the transition from 'ferroptosis' to 'mitochondrial dysfunction'. Ferroptosis, a form of RCD, has recently been implicated in tumor resistance to radiotherapy. Inducing ferroptosis by modulating iron metabolism and redox imbalance has emerged as a promising strategy to enhance the efficacy of radiotherapy [Table SI (166-276)]. Concurrently, radiation-induced mitochondrial dysfunction serves as a critical pathway for ICD in tumor cells. Radiotherapy-ICIs has shown considerable therapeutic benefits in clinical studies across various cancer types. These studies underscore the complex interplay between ferroptosis and mitochondrial dysfunction in tumor treatment responses, highlighting the potential of combination strategies that integrate radiotherapy, immunotherapy and ferroptosis modulation.
Although the potential of ferroptosis and mitochondrial dysfunction in enhancing radioimmunotherapy is becoming increasingly apparent, the research presently described demonstrates several unresolved paradoxes in the regulation of ferroptosis. Specifically, i) in hepatocytes, while mtROS-activated PERK signaling promotes arsenite-induced ferroptosis by downregulating Mfn-2 and impairing MAMs function, its cleaved C-terminal fragment paradoxically exerts protective effects against alcoholic hepatocyte ferroptosis (75); ii) the complex role of cGAS-STING in either promoting cell death (via pyroptosis) (40) or supporting cell survival (by suppressing ferroptosis) (41); iii) the TME presents even more complex dichotomies: Although hypoxia generally diminishes radiosensitivity, hypoxia-induced BNIP3 protein can stimulate mitophagy and elevate ROS production, mechanisms known to potentiate ferroptosis and consequently improve radiation response (86); and iv) radiation-enhanced FAO exhibits dual effects: While increasing tumor cell vulnerability to ferroptosis, it simultaneously induces T cell exhaustion, ultimately attenuating anti-tumor immunity and compromising radioimmunotherapy efficacy (88).
The current research still faces other challenges. First, the immunogenicity of ferroptosis in different contexts remains unclear. Second, whether DAMPs are released during ferroptosis, and how these DAMPs mediate immune responses within the TME, require further investigation. Additionally, the interactions between various cell death pathways and the optimization of drugs that either promote or inhibit ferroptosis are critical issues that need to be addressed. Resolving these questions will help clarify the precise role of ferroptosis in the TME and provide new insights for cancer treatment.
These challenges collectively indicate that the current understanding remains incomplete regarding the molecular mechanisms of ferroptosis are not fully understood, particularly in terms of its heterogeneous effects across different tumor types and subcellular organelles, which warrants further exploration [Table SII (277-430)] such as the impact of TME factors (oxidative stress, hypoxia and lipid metabolism) on ferroptosis susceptibility and the precise mechanisms by which ferroptosis synergizes with radioimmunotherapy, including the efficacy of radioimmunotherapy combined with ferroptosis modulation may vary significantly depending on the clinical context. Therefore, accurately assessing patient responses and resistance mechanisms remains a key area of research. Furthermore, while the combination of ICIs and radiotherapy has shown promise, the interactions between immune-related side effects and radiotherapy-induced toxicity have not been thoroughly evaluated. Balancing therapeutic efficacy with safety and optimizing treatment regimens remains a major challenge in clinical applications.
Future research should focus on several pivotal areas. First, a deeper exploration of the molecular mechanisms underpinning ferroptosis and mitochondrial dysfunction, particularly their roles within different TMEs, is essential. By utilizing molecular targeting technologies, the development of novel drugs or small molecule modulators that specifically induce ferroptosis or restore mitochondrial function could offer new strategies to enhance radiotherapy efficacy. Second, there is a need for the advancement of immune monitoring techniques, particularly those aimed at precisely assessing the association between immune system responses and tumor cell death. This will provide the foundation for personalized treatment strategies. Furthermore, combining modern gene-editing technologies with immunotherapeutic approaches to overcome the current resistance to radiotherapy will be a major focus of future research.
In conclusion, the combination of radiotherapy and immunotherapy holds significant promise in cancer treatment. However, to achieve widespread clinical application, several mechanistic and clinical challenges must be overcome. Future research will deepen the current understanding of the interactions between ferroptosis, mitochondrial dysfunction and immunotherapy. Based on these insights, more precise and safer treatment regimens will be developed, offering patients with cancer a broader range of therapeutic options.
Not applicable.
TW, XZ and XY conceived the study and wrote the manuscript. AZ, YF, KC, HT, ZT and PZ conducted literature search/selection and data extraction. XH and LY revised and edited the manuscript. All authors have read and approved the final manuscript. Data authentication is not applicable.
Not applicable.
Not applicable.
The authors declare that they have no competing interests.
Not applicable.
The present work was supported by grants from the National Natural Science Foundation of China (grant no. 82172804), the Yishan Research Project of Jiangsu Cancer Hospital (grant no. YSPY202407) and Clinical Research Foundation of collaborative innovation center for cancer personalized medicine (grant no. JZ21449020210616).
|
Hanahan D: Hallmarks of cancer: New dimensions. Cancer Discov. 12:31–46. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Petroni G, Cantley LC, Santambrogio L, Formenti SC and Galluzzi L: Radiotherapy as a tool to elicit clinically actionable signalling pathways in cancer. Nat Rev Clin Oncol. 19:114–131. 2022. View Article : Google Scholar : | |
|
Azzam EI, Jay-Gerin JP and Pain D: Ionizing radiation-induced metabolic oxidative stress and prolonged cell injury. Cancer Lett. 327:48–60. 2012. View Article : Google Scholar | |
|
Su W, Wang H, Zhao S, Ji X, Jiang H, Li K, Zuo C, Zheng J, Ni D and Hu J: Lysosome-escaping and nucleus-targeting nanomedicine for enhanced cancer catalytic internal radiotherapy. Eur J Nucl Med Mol Imaging. July 1–2025.Epub ahead of print. View Article : Google Scholar | |
|
Yuan P, Wang Y, Wang A, Lin J, Chen X, Liu X, Zhang Y, Gao W, Wang P, Zhang G and Liu L: Synergistic cancer treatment with PCuLu nanocatalysts by inducing cuproptosis and enhancing radiotherapy. J Colloid Interface Sci. 699:1382602025. View Article : Google Scholar : PubMed/NCBI | |
|
Liu H, Tang Y, Singh A, Vong J, Cordero J, Mathes A, Gao R, Jia Y, Garvalov BK, Acker T, et al: RNF20 links the DNA damage response and metabolic rewiring in lung cancer through HIF1α. Nat Commun. 16:49292025. View Article : Google Scholar | |
|
Shankar V, Wilhelmy J, Curtis EJ, Michael B, Cervantes L, Mallajosyula V, Davis RW, Snyder M, Younis S, Robinson WH, et al: Oxidative stress is a shared characteristic of ME/CFS and Long COVID. Proc Natl Acad Sci USA. 122:e24265641222025. View Article : Google Scholar : PubMed/NCBI | |
|
Zheng J and Conrad M: The metabolic underpinnings of ferroptosis. Cell Metab. 32:920–937. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Rochette L, Dogon G, Rigal E, Zeller M, Cottin Y and Vergely C: Lipid peroxidation and iron metabolism: Two corner stones in the homeostasis control of ferroptosis. Int J Mol Sci. 24:4492022. View Article : Google Scholar | |
|
Wang M, Li M, Jiang Y, Wang S, Yang X, Naseem A, Algradi AM, Hao Z, Guan W, Chen Q, et al: Saponins from Astragalus membranaceus (Fisch.) bge alleviated neuronal ferroptosis in Alzheimer's disease by regulating the NOX4/Nrf2 signaling pathway. J Agric Food Chem. 73:7725–7740. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Wang Q, Liu J, Zhang Y, Li Z, Zhao Z, Jiang W, Zhao J, Hou L and Wang Q: Microglial CR3 promotes neuron ferroptosis via NOX2-mediated iron deposition in rotenone-induced experimental models of Parkinson's disease. Redox Biol. 77:1033692024. View Article : Google Scholar : PubMed/NCBI | |
|
Martínez-Reyes I and Chandel NS: Cancer metabolism: Looking forward. Nat Rev Cancer. 21:669–680. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Wang YP, Sharda A, Xu SN, van Gastel N, Man CH, Choi U, Leong WZ, Li X and Scadden DT: Malic enzyme 2 connects the Krebs cycle intermediate fumarate to mitochondrial biogenesis. Cell Metab. 33:1027–1041.e8. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Miwa S, Kashyap S, Chini E and von Zglinicki T: Mitochondrial dysfunction in cell senescence and aging. J Clin Invest. 132:e1584472022. View Article : Google Scholar : PubMed/NCBI | |
|
Lei G, Mao C, Yan Y, Zhuang L and Gan B: Ferroptosis, radiotherapy, and combination therapeutic strategies. Protein Cell. 12:836–857. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Bray F, Laversanne M, Weiderpass E and Soerjomataram I: The ever-increasing importance of cancer as a leading cause of premature death worldwide. Cancer. 127:3029–3030. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Chen W, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, Jemal A, Yu XQ and He J: Cancer statistics in China, 2015. CA Cancer J Clin. 66:115–132. 2016.PubMed/NCBI | |
|
Liermann-Wooldrik KT, Kosmacek EA, McDowell JA, Takkar S, Murthy D, Singh PK, Schott MB, Ponnusamy MP and Oberley-Deegan RE: Radiation promotes acute and chronic damage to adipose tissue. Int J Mol Sci. 26:56262025. View Article : Google Scholar : PubMed/NCBI | |
|
Wungcharoen P, Prayongrat A and Tangjaturonrasme N: Hearing loss and middle ear effusion in nasopharyngeal carcinoma following radiotherapy: Dose-response relationship and normal tissue complication probability modeling. Int Arch Otorhinolaryngol. 29:1–9. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Ning J, Chen L, Zeng Y, Xiao G, Tian W, Wu Q, Tang J, He S, Tanzhu G and Zhou R: The scheme, and regulative mechanism of pyroptosis, ferroptosis, and necroptosis in radiation injury. Int J Biol Sci. 20:1871–1883. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Gao S, Huang J, Zhao R, He H, Zhang J and Wen X: Comprehensive analysis of multiple regulated cell death risk signatures in lung adenocarcinoma. Heliyon. 10:e386412024. View Article : Google Scholar : PubMed/NCBI | |
|
Kuwahara Y, Tomita K, Urushihara Y, Sato T, Kurimasa A and Fukumoto M: Association between radiation-induced cell death and clinically relevant radioresistance. Histochem Cell Biol. 150:649–659. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang S, Zhang J, Fan X, Liu H, Zhu M, Yang M, Zhang X, Zhang H and Yu F: Ionizing radiation-induced ferroptosis based on nanomaterials. Int J Nanomedicine. 17:3497–3507. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Yan B, Ai Y, Sun Q, Ma Y, Cao Y, Wang J, Zhang Z and Wang X: Membrane damage during ferroptosis is caused by oxidation of phospholipids catalyzed by the oxidoreductases POR and CYB5R1. Mol Cell. 81:355–369.e10. 2021. View Article : Google Scholar | |
|
Kim JH, Rho JR and Choi JH: Marine sponge-derived gukulenin A sensitizes ovarian cancer cells to PARP inhibition via ferroptosis induction. Mar Drugs. 23:1382025. View Article : Google Scholar : PubMed/NCBI | |
|
Wu Y, Ji Y, Jin X, Xu G, Wang X, Song S, Li R, Wang Y, Liu R and Li Z: YLKTT, a potent biopeptide that ameliorates oxygen-glucose deprivation-induced neuronal cell ferroptosis by ACSL4/GPX4 and SLC7A11/GPX4 axis. J Pharm Pharmacol. rgaf0372025. View Article : Google Scholar : PubMed/NCBI | |
|
Dong J, Qi F, Qie H, Du S, Li L, Zhang Y, Xu K, Li D and Xu Y: Oleic acid inhibits SDC4 and promotes ferroptosis in lung cancer through GPX4/ACSL4. Clin Respir J. 18:e700142024. View Article : Google Scholar : PubMed/NCBI | |
|
Lei G, Zhang Y, Koppula P, Liu X, Zhang J, Lin SH, Ajani JA, Xiao Q, Liao Z, Wang H and Gan B: The role of ferroptosis in ionizing radiation-induced cell death and tumor suppression. Cell Res. 30:146–162. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Liu T, Zhu C, Chen X, Guan G, Zou C, Shen S, Wu J, Wang Y, Lin Z, Chen L, et al: Ferroptosis, as the most enriched programmed cell death process in glioma, induces immunosuppression and immunotherapy resistance. Neuro Oncol. 24:1113–1125. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Wang Y, Hu J, Wu S, Fleishman JS, Li Y, Xu Y, Zou W, Wang J, Feng Y, Chen J and Wang H: Targeting epigenetic and posttranslational modifications regulating ferroptosis for the treatment of diseases. Signal Transduct Target Ther. 8:4492023. View Article : Google Scholar : PubMed/NCBI | |
|
Galy B, Conrad M and Muckenthaler M: Mechanisms controlling cellular and systemic iron homeostasis. Nat Rev Mol Cell Biol. 25:133–155. 2024. View Article : Google Scholar | |
|
Zhang H, Tsui CK, Garcia G, Joe LK, Wu H, Maruichi A, Fan W, Pandovski S, Yoon PH, Webster BM, et al: The extracellular matrix integrates mitochondrial homeostasis. Cell. 187:4289–4304.e26. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Zong WX, Rabinowitz JD and White E: Mitochondria and cancer. Mol Cell. 61:667–676. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Kim S, Piao S, Lee I, Nagar H, Choi SJ, Shin N, Kim DW, Shong M, Jeon BH and Kim CS: CR6 interacting factor 1 deficiency induces premature senescence via SIRT3 inhibition in endothelial cells. Free Radic Biol Med. 150:161–171. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Rucheton B, Jardel C, Filaut S, Amador MDM, Maisonobe T, Serre I, Romero NB, Leonard-Louis S, Haraux F and Lombès A: Homoplasmic deleterious MT-ATP6/8 mutations in adult patients. Mitochondrion. 55:64–77. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Bo T, Yamamori T, Yamamoto K, Fujimoto M, Yasui H and Inanami O: Mitochondrial fission promotes radiation-induced increase in intracellular Ca2+ level leading to mitotic catastrophe in mouse breast cancer EMT6 cells. Biochem Biophys Res Commun. 522:144–150. 2020. View Article : Google Scholar | |
|
Okon IS and Zou MH: Mitochondrial ROS and cancer drug resistance: Implications for therapy. Pharmacol Res. 100:170–174. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Kam WW and Banati RB: Effects of ionizing radiation on mitochondria. Free Radic Biol Med. 65:607–619. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Saunders TL, Windley SP, Gervinskas G, Balka KR, Rowe C, Lane R, Tailler M, Nguyen TN, Ramm G, Lazarou M, et al: Exposure of the inner mitochondrial membrane triggers apoptotic mitophagy. Cell Death Differ. 31:335–347. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Xia L, Yan X and Zhang H: Mitochondrial DNA-activated cGAS-STING pathway in cancer: Mechanisms and therapeutic implications. Biochim Biophys Acta Rev Cancer. 1880:1892492025. View Article : Google Scholar | |
|
Qiu S, Zhong X, Meng X, Li S, Qian X, Lu H, Cai J, Zhang Y, Wang M, Ye Z, et al: Mitochondria-localized cGAS suppresses ferroptosis to promote cancer progression. Cell Res. 33:299–311. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Qiu YH, Zhang TS, Wang XW, Wang MY, Zhao WX, Zhou HM, Zhang CH, Cai ML, Chen XF, Zhao WL and Shao RG: Mitochondria autophagy: A potential target for cancer therapy. J Drug Target. 29:576–591. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Xu S, Sankar S and Neamati N: Protein disulfide isomerase: A promising target for cancer therapy. Drug Discov Today. 19:222–240. 2014. View Article : Google Scholar | |
|
Wang R, Shang Y, Chen B, Xu F, Zhang J, Zhang Z, Zhao X, Wan X, Xu A, Wu L and Zhao G: Protein disulfide isomerase blocks the interaction of LC3II-PHB2 and promotes mTOR signaling to regulate autophagy and radio/chemo-sensitivity. Cell Death Dis. 13:8512022. View Article : Google Scholar : PubMed/NCBI | |
|
Gao M, Yi J, Zhu J, Minikes AM, Monian P, Thompson CB and Jiang X: Role of mitochondria in ferroptosis. Mol Cell. 73:354–363.e353.e3. 2019. View Article : Google Scholar : | |
|
Richardson DR, Lane DJ, Becker EM, Huang ML, Whitnall M, Suryo Rahmanto Y, Sheftel AD and Ponka P: Mitochondrial iron trafficking and the integration of iron metabolism between the mitochondrion and cytosol. Proc Natl Acad Sci USA. 107:10775–10782. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Wang P, Cui Y, Liu Y, Li Z, Bai H, Zhao Y and Chang YZ: Mitochondrial ferritin alleviates apoptosis by enhancing mitochondrial bioenergetics and stimulating glucose metabolism in cerebral ischemia reperfusion. Redox Biol. 57:1024752022. View Article : Google Scholar : PubMed/NCBI | |
|
Sun K, Zhi Y, Ren W, Li S, Zhou X, Gao L and Zhi K: The mitochondrial regulation in ferroptosis signaling pathway and its potential strategies for cancer. Biomed Pharmacother. 169:1158922023. View Article : Google Scholar : PubMed/NCBI | |
|
Wang X, Ji Y, Qi J, Zhou S, Wan S, Fan C, Gu Z, An P, Luo Y and Luo J: Mitochondrial carrier 1 (MTCH1) governs ferroptosis by triggering the FoxO1-GPX4 axis-mediated retrograde signaling in cervical cancer cells. Cell Death Dis. 14:5082023. View Article : Google Scholar : PubMed/NCBI | |
|
Adam AC, Bornhövd C, Prokisch H, Neupert W and Hell K: The Nfs1 interacting protein Isd11 has an essential role in Fe/S cluster biogenesis in mitochondria. EMBO J. 25:174–183. 2006. View Article : Google Scholar | |
|
Kerins MJ, Milligan J, Wohlschlegel JA and Ooi A: Fumarate hydratase inactivation in hereditary leiomyomatosis and renal cell cancer is synthetic lethal with ferroptosis induction. Cancer Sci. 109:2757–2766. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Yang J, Lee Y and Hwang CS: The ubiquitin-proteasome system links NADPH metabolism to ferroptosis. Trends Cell Biol. 33:1088–1103. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Jonckheere AI, Smeitink JA and Rodenburg RJ: Mitochondrial ATP synthase: Architecture, function and pathology. J Inherit Metab Dis. 35:211–225. 2012. View Article : Google Scholar : | |
|
Gawargi FI and Mishra PK: MMP9 drives ferroptosis by regulating GPX4 and iron signaling. iScience. 27:1106222024. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang K, Chen L, Wang B, Chen D, Ye X, Han X, Fang Q, Yu C, Wu J, Guo S, et al: Mitochondrial supercomplex assembly regulates metabolic features and glutamine dependency in mammalian cells. Theranostics. 13:3165–3187. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Lobo-Jarne T, Nývltová E, Pérez-Pérez R, Timón-Gómez A, Molinié T, Choi A, Mourier A, Fontanesi F, Ugalde C and Barrientos A: Human COX7A2L regulates complex III biogenesis and promotes supercomplex organization remodeling without affecting mitochondrial bioenergetics. Cell Rep. 25:1786–1799.e4. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Song X, Liu J, Kuang F, Chen X, Zeh HJ III, Kang R, Kroemer G, Xie Y and Tang D: PDK4 dictates metabolic resistance to ferroptosis by suppressing pyruvate oxidation and fatty acid synthesis. Cell Rep. 34:1087672021. View Article : Google Scholar : PubMed/NCBI | |
|
Li Z, Xu ZM, Chen WP, Du XJ, Ou CX, Luo ZK, Wang R, Zhang CQ, Ge CD, Han M, et al: Tumor-repopulating cells evade ferroptosis via PCK2-dependent phospholipid remodeling. Nat Chem Biol. 20:1341–1352. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao Y, Li M, Yao X, Fei Y, Lin Z, Li Z, Cai K, Zhao Y and Luo Z: HCAR1/MCT1 regulates tumor ferroptosis through the lactate-mediated AMPK-SCD1 activity and its therapeutic implications. Cell Rep. 33:1084872020. View Article : Google Scholar : PubMed/NCBI | |
|
Ma J, Tang L, Tan Y, Xiao J, Wei K, Zhang X, Ma Y, Tong S, Chen J, Zhou N, et al: Lithium carbonate revitalizes tumor-reactive CD8+ T cells by shunting lactic acid into mitochondria. Nat Immunol. 25:552–561. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Gao M, Monian P, Quadri N, Ramasamy R and Jiang X: Glutaminolysis and transferrin regulate ferroptosis. Mol Cell. 59:298–308. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Missiroli S, Patergnani S, Caroccia N, Pedriali G, Perrone M, Previati M, Wieckowski MR and Giorgi C: Mitochondria-associated membranes (MAMs) and inflammation. Cell Death Dis. 9:3292018. View Article : Google Scholar : PubMed/NCBI | |
|
Kuang F, Liu J, Xie Y, Tang D and Kang R: MGST1 is a redox-sensitive repressor of ferroptosis in pancreatic cancer cells. Cell Chem Biol. 28:765–775.e5. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Pontisso I and Combettes L: Role of sigma-1 receptor in calcium modulation: Possible involvement in cancer. Genes (Basel). 12:1392021. View Article : Google Scholar : PubMed/NCBI | |
|
Liu Y, Ma X, Fujioka H, Liu J, Chen S and Zhu X: DJ-1 regulates the integrity and function of ER-mitochondria association through interaction with IP3R3-Grp75-VDAC1. Proc Natl Acad Sci USA. 116:25322–25328. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Rieusset J: The role of endoplasmic reticulum-mitochondria contact sites in the control of glucose homeostasis: An update. Cell Death Dis. 9:3882018. View Article : Google Scholar : PubMed/NCBI | |
|
Wei S, Qiu T, Wang N, Yao X, Jiang L, Jia X, Tao Y, Zhang J, Zhu Y, Yang G, et al: Ferroptosis mediated by the interaction between Mfn2 and IREα promotes arsenic-induced nonalcoholic steatohepatitis. Environ Res. 188:1098242020. View Article : Google Scholar | |
|
Zhu Y, Fujimaki M, Snape L, Lopez A, Fleming A and Rubinsztein DC: Loss of WIPI4 in neurodegeneration causes autophagy-independent ferroptosis. Nat Cell Boil. 26:542–551. 2024. View Article : Google Scholar | |
|
Liu Y and Yang H: WIPI4 loss linked to ferroptosis. Nat Cell Biol. 26:506–507. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Zheng J and Conrad M: Ferroptosis: When metabolism meets cell death. Physiol Rev. 105:651–706. 2025. View Article : Google Scholar | |
|
Ju HQ, Lin JF, Tian T, Xie D and Xu RH: NADPH homeostasis in cancer: Functions, mechanisms and therapeutic implications. Signal Transduct Target Ther. 5:2312020. View Article : Google Scholar : PubMed/NCBI | |
|
Pirozzi CJ and Yan H: The implications of IDH mutations for cancer development and therapy. Nat Rev Clin Oncol. 18:645–661. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Sarcognato S, Sacchi D, Fabris L, Zanus G, Gringeri E, Niero M, Gallina G and Guido M: Ferroptosis in intrahepatic cholangiocarcinoma: IDH1105GGT single nucleotide polymorphism is associated with its activation and better prognosis. Front Med (Lausanne). 9:8862292022. View Article : Google Scholar | |
|
Brashears CB, Prudner BC, Rathore R, Caldwell KE, Dehner CA, Buchanan JL, Lange SES, Poulin N, Sehn JK, Roszik J, et al: Malic enzyme 1 absence in synovial sarcoma shifts antioxidant system dependence and increases sensitivity to ferroptosis induction with ACXT-3102. Clin Cancer Res. 28:3573–3589. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Chen J, Li X, Ge C, Min J and Wang F: The multifaceted role of ferroptosis in liver disease. Cell Death Differ. 29:467–480. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang X, Li X, Zheng C, Yang C, Zhang R, Wang A, Feng J, Hu X, Chang S and Zhang H: Ferroptosis, a new form of cell death defined after radiation exposure. Int J Radiat Biol. 98:1201–1209. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Ye LF, Chaudhary KR, Zandkarimi F, Harken AD, Kinslow CJ, Upadhyayula PS, Dovas A, Higgins DM, Tan H, Zhang Y, et al: Radiation-induced lipid peroxidation triggers ferroptosis and synergizes with ferroptosis inducers. ACS Chem Biol. 15:469–484. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Yao N, Wang C, Hu N, Li Y, Liu M, Lei Y, Chen M, Chen L, Chen C, Lan P, et al: Inhibition of PINK1/Parkin-dependent mitophagy sensitizes multidrug-resistant cancer cells to B5G1, a new betulinic acid analog. Cell Death Dis. 10:2322019. View Article : Google Scholar : PubMed/NCBI | |
|
Singh R, Kaushik S, Wang Y, Xiang Y, Novak I, Komatsu M, Tanaka K, Cuervo AM and Czaja MJ: Autophagy regulates lipid metabolism. Nature. 458:1131–1135. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Xiao Y, Hu X, Rudolphi CF, Nollet EE, Nederlof R, Wang Q, Bakker D, Nikolaou PE, Knol JC, Haas RRG, et al: The innate immune receptor NLRX1 is a novel required modulator for mPTP opening: Implications for cardioprotection. Basic Res Cardiol. June 19–2025.Epub ahead of print. View Article : Google Scholar : | |
|
Zhong Z, Hou Y, Zhou C, Wang J, Gao L, Wu X, Zhou G, Liu S, Xu Y and Yang W: FL3 mitigates cardiac ischemia-reperfusion injury by promoting mitochondrial fusion to restore calcium homeostasis. Cell Death Discov. 11:3042025. View Article : Google Scholar : PubMed/NCBI | |
|
Fan H and Tan Y: Lipid droplet-mitochondria contacts in health and disease. Int J Mol Sci. 25:68782024. View Article : Google Scholar : PubMed/NCBI | |
|
Li J, Yang D, Li Z, Zhao M, Wang D, Sun Z, Wen P, Dai Y, Gou F, Ji Y, et al: PINK1/Parkin-mediated mitophagy in neurodegenerative diseases. Ageing Res Rev. 84:1018172023. View Article : Google Scholar | |
|
Springer W and Kahle PJ: Regulation of PINK1-Parkin-mediated mitophagy. Autophagy. 7:266–278. 2011. View Article : Google Scholar | |
|
Telarovic I, Wenger RH and Pruschy M: Interfering with tumor hypoxia for radiotherapy optimization. J Exp Clin Cancer Res. 40:1972021. View Article : Google Scholar : PubMed/NCBI | |
|
Li Y, Zheng W, Lu Y, Zheng Y, Pan L, Wu X, Yuan Y, Shen Z, Ma S, Zhang X, et al: BNIP3L/NIX-mediated mitophagy: Molecular mechanisms and implications for human disease. Cell Death Dis. 13:142021. View Article : Google Scholar : PubMed/NCBI | |
|
Bensaad K, Favaro E, Lewis CA, Peck B, Lord S, Collins JM, Pinnick KE, Wigfield S, Buffa FM, Li JL, et al: Fatty acid uptake and lipid storage induced by HIF-1α contribute to cell growth and survival after hypoxia-reoxygenation. Cell Rep. 9:349–365. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Tapio S, Little MP, Kaiser JC, Impens N, Hamada N, Georgakilas AG, Simar D and Salomaa S: Ionizing radiation-induced circulatory and metabolic diseases. Environ Int. 146:1062352021. View Article : Google Scholar | |
|
Rao D, Verburg F, Renner K, Peeper DS, Lacroix R and Blank CU: Metabolic profiles of regulatory T cells in the tumour microenvironment. Cancer Immunol Immunother. 70:2417–2427. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Biswas SK: Metabolic reprogramming of immune cells in cancer progression. Immunity. 43:435–449. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Buck MD, O'Sullivan D, Klein Geltink RI, Curtis JD, Chang CH, Sanin DE, Qiu J, Kretz O, Braas D, van der Windt GJ, et al: Mitochondrial dynamics controls T cell fate through metabolic programming. Cell. 166:63–76. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Dumauthioz N, Tschumi B, Wenes M, Marti B, Wang H, Franco F, Li W, Lopez-Mejia IC, Fajas L, Ho PC, et al: Enforced PGC-1α expression promotes CD8 T cell fitness, memory formation and anti-tumor immunity. Cell Mol Immunol. 18:1761–1771. 2021. View Article : Google Scholar | |
|
Miao L, Lu C, Zhang B, Li H, Zhao X, Chen H, Liu Y and Cui X: Advances in metabolic reprogramming of NK cells in the tumor microenvironment on the impact of NK therapy. J Transl Med. 22:2292024. View Article : Google Scholar : PubMed/NCBI | |
|
Meybodi SM, Ejlalidiz M, Manshadi MR, Raeisi M, Zarin M, Kalhor Z, Saberiyan M and Hamblin MR: Crosstalk between hypoxia-induced pyroptosis and immune escape in cancer: From mechanisms to therapy. Crit Rev Oncol Hematol. 197:1043402024. View Article : Google Scholar : PubMed/NCBI | |
|
Galván-Peña S and O'Neill LA: Metabolic reprograming in macrophage polarization. Front Immunol. 5:4202014.PubMed/NCBI | |
|
Qiu Y, Huang Y, Chen M, Yang Y, Li X and Zhang W: Mitochondrial DNA in NLRP3 inflammasome activation. Int Immunopharmacol. 108:1087192022. View Article : Google Scholar : PubMed/NCBI | |
|
Yan C, Liu X, Xu H and Wang L: Cytoplasmic mtDNA clearance suppresses inflammatory immune responses. Trends Cell Biol. 34:897–900. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Bai R and Cui J: Mitochondrial immune regulation and anti-tumor immunotherapy strategies targeting mitochondria. Cancer Lett. 564:2162232023. View Article : Google Scholar : PubMed/NCBI | |
|
Vinay DS, Ryan EP, Pawelec G, Talib WH, Stagg J, Elkord E, Lichtor T, Decker WK, Whelan RL, Kumara HMCS, et al: Immune evasion in cancer: Mechanistic basis and therapeutic strategies. Semin Cancer Biol. 35(Suppl): S185–S198. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Kuo CL, Ponneri Babuharisankar A, Lin YC, Lien HW, Lo YK, Chou HY, Tangeda V, Cheng LC, Cheng AN and Lee AY: Mitochondrial oxidative stress in the tumor microenvironment and cancer immunoescape: foe or friend? J Biomed Sci. 29:742022. View Article : Google Scholar : PubMed/NCBI | |
|
Chen X, Song M, Zhang B and Zhang Y: Reactive oxygen species regulate T cell immune response in the tumor microenvironment. Oxid Med Cell Longev. 2016:15809672016. View Article : Google Scholar : PubMed/NCBI | |
|
Hopfner KP and Hornung V: Molecular mechanisms and cellular functions of cGAS-STING signalling. Nat Rev Mol Cell Biol. 21:501–521. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Taneja N, Chauhan A, Kulshreshtha R and Singh S: HIF-1 mediated metabolic reprogramming in cancer: Mechanisms and therapeutic implications. Life Sci. 352:1228902024. View Article : Google Scholar : PubMed/NCBI | |
|
Yang D, Liu J, Qian H and Zhuang Q: Cancer-associated fibroblasts: from basic science to anticancer therapy. Exp Mol Med. 55:1322–1332. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Mortezaee K and Majidpoor J: The impact of hypoxia on immune state in cancer. Life Sci. 286:1200572021. View Article : Google Scholar : PubMed/NCBI | |
|
Kim J, Kim J and Bae JS: ROS homeostasis and metabolism: A critical liaison for cancer therapy. Exp Mol Med. 48:e2692016. View Article : Google Scholar : PubMed/NCBI | |
|
He L, Tam PK and Deng CX: Orchestration of tumor-associated macrophages in the tumor cell-macrophage-CD8+ T cell loop for cancer immunotherapy. Int J Biol Sci. 21:4098–4116. 2025. View Article : Google Scholar : | |
|
Li Y, You J, Zou Z, Sun G, Shi Y, Sun Y, Xu S and Zhang X: Decoding the tumor microenvironment: Exosome-mediated macrophage polarization and therapeutic frontiers. Int J Biol Sci. 21:4187–4214. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Ricchi F, Kenno S, Pedretti N, Brenna G, De Seta F, Ardizzoni A and Pericolini E: Cutibacterium acnes lysate improves cellular response against Candida albicans, Escherichia coli and Gardnerella vaginalis in an in vitro model of vaginal infection. Front Cell Infect Microbiol. 15:15788312025. View Article : Google Scholar : PubMed/NCBI | |
|
Matsushima Y, Takahashi K, Yue S, Fujiyoshi Y, Yoshioka H, Aihara M, Setoyama D, Uchiumi T, Fukuchi S and Kang D: Mitochondrial Lon protease is a gatekeeper for proteins newly imported into the matrix. Commun Biol. 4:9742021. View Article : Google Scholar : PubMed/NCBI | |
|
Li JY, Zhao Y, Gong S, Wang MM, Liu X, He QM, Li YQ, Huang SY, Qiao H, Tan XR, et al: TRIM21 inhibits irradiation-induced mitochondrial DNA release and impairs antitumour immunity in nasopharyngeal carcinoma tumour models. Nat Commun. 14:8652023. View Article : Google Scholar : PubMed/NCBI | |
|
Liu S, Wang W, Hu S, Jia B, Tuo B, Sun H, Wang Q, Liu Y and Sun Z: Radiotherapy remodels the tumor microenvironment for enhancing immunotherapeutic sensitivity. Cell Death Dis. 14:6792023. View Article : Google Scholar : PubMed/NCBI | |
|
Seo YN, Baik JS, Lee SM, Lee JE, Ahn HR, Lim MS, Park MT and Kim SD: Ionizing radiation selectively increases CXC ligand 10 level via the DNA-damage-induced p38 MAPK-STAT1 pathway in murine J774A.1 macrophages. Cells. 12:10092023. View Article : Google Scholar : PubMed/NCBI | |
|
Gong J, Le TQ, Massarelli E, Hendifar AE and Tuli R: Radiation therapy and PD-1/PD-L1 blockade: The clinical development of an evolving anticancer combination. J Immunother Cancer. 6:462018. View Article : Google Scholar : PubMed/NCBI | |
|
Donlon NE, Power R, Hayes C, Reynolds JV and Lysaght J: Radiotherapy, immunotherapy, and the tumour microenvironment: Turning an immunosuppressive milieu into a therapeutic opportunity. Cancer Lett. 502:84–96. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang H, Li S, Wang D, Liu S, Xiao T, Gu W, Yang H, Wang H, Yang M and Chen P: Metabolic reprogramming and immune evasion: The interplay in the tumor microenvironment. Biomark Res. 12:962024. View Article : Google Scholar : PubMed/NCBI | |
|
Feng M, Jiang W, Kim BYS, Zhang CC, Fu YX and Weissman IL: Phagocytosis checkpoints as new targets for cancer immunotherapy. Nat Rev Cancer. 19:568–586. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Cagnoni AJ, Giribaldi ML, Blidner AG, Cutine AM, Gatto SG, Morales RM, Salatino M, Abba MC, Croci DO, Mariño KV and Rabinovich GA: Galectin-1 fosters an immunosuppressive microenvironment in colorectal cancer by reprogramming CD8+ regulatory T cells. Proc Natl Acad Sci USA. 118:e21029501182021. View Article : Google Scholar | |
|
Guo S, Yao Y, Tang Y, Xin Z, Wu D, Ni C, Huang J, Wei Q and Zhang T: Radiation-induced tumor immune microenvironments and potential targets for combination therapy. Signal Transduct Target Ther. 8:2052023. View Article : Google Scholar : PubMed/NCBI | |
|
Spiotto M, Fu YX and Weichselbaum RR: The intersection of radiotherapy and immunotherapy: Mechanisms and clinical implications. Sci Immunol. 1:EAAG12662016. View Article : Google Scholar : PubMed/NCBI | |
|
Kang K, Lin X, Chen P, Liu H, Liu F, Xiong W, Li G, Yi M, Li X, Wang H and Xiang B: T cell exhaustion in human cancers. Biochim Biophys Acta Rev Cancer. 1879:1891622024. View Article : Google Scholar : PubMed/NCBI | |
|
Zhou S, Zhu M, Wei X, Mu P, Shen L, Wang Y, Wan J, Zhang H, Xia F and Zhang Z: Low-dose radiotherapy synergizes with iRGD-antiCD3-modified T cells by facilitating T cell infiltration. Radiother Oncol. 194:1102132024. View Article : Google Scholar : PubMed/NCBI | |
|
Lumniczky K and Sáfrány G: The impact of radiation therapy on the antitumor immunity: Local effects and systemic consequences. Cancer Lett. 356:114–125. 2015. View Article : Google Scholar | |
|
Lynch C, Pitroda SP and Weichselbaum RR: Radiotherapy, immunity, and immune checkpoint inhibitors. Lancet Oncol. 25:e352–e362. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Liu T, Pei P, Shen W, Hu L and Yang K: Radiation-induced immunogenic cell death for cancer radioimmunotherapy. Small Methods. 7:e22014012023. View Article : Google Scholar : PubMed/NCBI | |
|
Liao P, Wang W, Wang W, Kryczek I, Li X, Bian Y, Sell A, Wei S, Grove S, Johnson JK, et al: CD8+ T cells and fatty acids orchestrate tumor ferroptosis and immunity via ACSL4. Cancer Cell. 40:365–378.e6. 2022. View Article : Google Scholar | |
|
Xu H, Ye D, Ren M, Zhang H and Bi F: Ferroptosis in the tumor microenvironment: Perspectives for immunotherapy. Trends Mol Med. 27:856–867. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Goldstein M and Kastan MB: The DNA damage response: implications for tumor responses to radiation and chemotherapy. Annu Rev Med. 66:129–143. 2015. View Article : Google Scholar | |
|
Yu X, Zhu D, Luo B, Kou W, Cheng Y and Zhu Y: IFNγ enhances ferroptosis by increasing JAK-STAT pathway activation to suppress SLCA711 expression in adrenocortical carcinoma. Oncol Rep. 47:972022. View Article : Google Scholar | |
|
Liang B, Khan M, Storts H, Zhang EH, Zheng X, Xing X, Claybon H, Wilson J, Li C, Jin N, et al: Riluzole enhancing anti-PD-1 efficacy by activating cGAS/STING signaling in colorectal cancer. Mol Cancer Ther. 24:131–140. 2025. View Article : Google Scholar : | |
|
Li H, Li X, Kong Y and Sun W: Ubiquitin-specific protease 34 in macrophages limits CD8 T cell-mediated onset of vitiligo in mice. Immunobiology. 228:1523832023. View Article : Google Scholar : PubMed/NCBI | |
|
Machado E, White-Gilbertson S, van de Vlekkert D, Janke L, Moshiach S, Campos Y, Finkelstein D, Gomero E, Mosca R, Qiu X, et al: Regulated lysosomal exocytosis mediates cancer progression. Sci Adv. 1:e15006032015. View Article : Google Scholar | |
|
Han C, Ge M, Xing P, Xia T, Zhang C, Ma K, Ma Y, Li S, Li W, Liu X, et al: Cystine deprivation triggers CD36-mediated ferroptosis and dysfunction of tumor infiltrating CD8+ T cells. Cell Death Dis. 15:1452024. View Article : Google Scholar | |
|
Mai LT, Swaminathan S, Nguyen TH, Collette E, Charpentier T, Carmona-Pérez L, Loucif H, Lamarre A, Heinonen KM, Langlais D, et al: Transcription factor IRF-5 regulates lipid metabolism and mitochondrial function in murine CD8+ T-cells during viral infection. EMBO J. June 10–2025.Epub ahead of print. | |
|
Deschler S, Pohl-Topcu J, Ramsauer L, Meiser P, Erlacher S, Schenk RP, Maurer HC, Shen P, Kager J, Zink J, et al: Polyunsaturated fatty acid-induced metabolic exhaustion and ferroptosis impair the anti-tumour function of MAIT cells in MASLD. J Hepatol. June 18–2025.Epub ahead of print. View Article : Google Scholar : PubMed/NCBI | |
|
Anderson DA III, Murphy KM and Briseño CG: Development, diversity, and function of dendritic cells in mouse and human. Cold Spring Harb Perspect Biol. 10:a0286132018. View Article : Google Scholar | |
|
Powell DR and Huttenlocher A: Neutrophils in the tumor micro-environment. Trends Immunol. 37:41–52. 2016. View Article : Google Scholar | |
|
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 | |
|
Lou X, Gao D, Yang L, Wang Y and Hou Y: Endoplasmic reticulum stress mediates the myeloid-derived immune suppression associated with cancer and infectious disease. J Transl Med. 21:12023. View Article : Google Scholar : PubMed/NCBI | |
|
Shi H, Dong G, Yan F, Zhang H, Li C, Ma Q, Zhang J, Ning Z, Li Z, Dai J, et al: Arctigenin ameliorates inflammation by regulating accumulation and functional activity of MDSCs in endotoxin shock. Inflammation. 41:2090–2100. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Li J, Liu J, Zhou Z, Wu R, Chen X, Yu C, Stockwell B, Kroemer G, Kang R and Tang D: Tumor-specific GPX4 degradation enhances ferroptosis-initiated antitumor immune response in mouse models of pancreatic cancer. Sci Transl Med. 15:eadg30492023. View Article : Google Scholar : PubMed/NCBI | |
|
Yang Y, Wang Y, Guo L, Gao W, Tang TL and Yan M: Interaction between macrophages and ferroptosis. Cell Death Dis. 13:3552022. View Article : Google Scholar : PubMed/NCBI | |
|
Luo L, Wang H, Tian W, Zeng J, Huang Y and Luo H: Targeting ferroptosis for cancer therapy: Iron metabolism and anticancer immunity. Am J Cancer Res. 11:5508–5525. 2021.PubMed/NCBI | |
|
Ma S, Fu X, Liu L, Liu Y, Feng H, Jiang H, Liu X, Liu R, Liang Z, Li M, et al: Iron-dependent autophagic cell death induced by radiation in MDA-MB-231 breast cancer cells. Front Cell Dev Biol. 9:7238012021. View Article : Google Scholar : PubMed/NCBI | |
|
Shukla SK, Kulkarni NS, Chan A, Parvathaneni V, Farrales P, Muth A and Gupta V: Metformin-encapsulated liposome delivery system: An effective treatment approach against breast cancer. Pharmaceutics. 11:5592019. View Article : Google Scholar : PubMed/NCBI | |
|
Abu Shelbayeh O, Arroum T, Morris S and Busch KB: PGC-1α is a master regulator of mitochondrial lifecycle and ros stress response. Antioxidants (Basel). 12:10752023. View Article : Google Scholar | |
|
Mao C, Liu X, Zhang Y, Lei G, Yan Y, Lee H, Koppula P, Wu S, Zhuang L, Fang B, et al: DHODH-mediated ferroptosis defence is a targetable vulnerability in cancer. Nature. 593:586–590. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Liu S, Zhang HL, Li J, Ye ZP, Du T, Li LC, Guo YQ, Yang D, Li ZL, Cao JH, et al: Tubastatin A potently inhibits GPX4 activity to potentiate cancer radiotherapy through boosting ferroptosis. Redox Biol. 62:1026772023. View Article : Google Scholar : PubMed/NCBI | |
|
Wang D, Tang L, Chen M, Gong Z, Fan C, Qu H, Liu Y, Shi L, Mo Y, Wang Y, et al: Nanocarriers targeting circular RNA ADARB1 boost radiosensitivity of nasopharyngeal carcinoma through synergically promoting ferroptosis. ACS Nano. 18:31055–31075. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Zhong XF and Sun X: Nanomedicines based on nanoscale metal-organic frameworks for cancer immunotherapy. Acta Pharmacol Sin. 41:928–935. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Lin J, Lai Y, Lu F and Wang W: Targeting ACSLs to modulate ferroptosis and cancer immunity. Trends Endocrinol Metab. 36:677–690. 2025. View Article : Google Scholar | |
|
Zhang Y, Cai K, Li C, Guo Q, Chen Q, He X, Liu L, Zhang Y, Lu Y, Chen X, et al: Macrophage-membrane-coated nanoparticles for tumor-targeted chemotherapy. Nano Lett. 18:1908–1915. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Liu N, Lin Q, Huang Z, Liu C, Qin J, Yu Y, Chen W, Zhang J, Jiang M, Gao X, et al: Mitochondria-targeted prodrug nanoassemblies for efficient ferroptosis-based therapy via devastating ferroptosis defense systems. ACS Nano. 18:7945–7958. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Zeng L, Gowda BHJ, Ahmed MG, Abourehab MAS, Chen ZS, Zhang C, Li J and Kesharwani P: Advancements in nanoparticle-based treatment approaches for skin cancer therapy. Mol Cancer. 22:102023. View Article : Google Scholar : PubMed/NCBI | |
|
Jiang Q, Wang K, Zhang X, Ouyang B, Liu H, Pang Z and Yang W: Platelet membrane-camouflaged magnetic nanoparticles for ferroptosis-enhanced cancer immunotherapy. Small. 16:e20017042020. View Article : Google Scholar : PubMed/NCBI | |
|
Yang M, Zhang Y, Hu Z, Xie H, Tian W and Liu Z: Application of hyaluronic acid-based nanoparticles for cancer combination therapy. Int J Pharm. 646:1234592023. 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 | |
|
Shao C, Li Z, Zhang C, Zhang W, He R, Xu J and Cai Y: Optical diagnostic imaging and therapy for thyroid cancer. Mater Today Bio. 17:1004412022. View Article : Google Scholar : PubMed/NCBI | |
|
Mulford DA, Scheinberg DA and Jurcic JG: The promise of targeted {alpha}-particle therapy. J Nucl Med. 46(Suppl 1): 199S–204S. 2005.PubMed/NCBI | |
|
Seidl C: Radioimmunotherapy with α-particle-emitting radionuclides. Immunotherapy. 6:431–458. 2014. View Article : Google Scholar | |
|
Xu J, Song M, Fang Z, Zheng L, Huang X and Liu K: Applications and challenges of ultra-small particle size nanoparticles in tumor therapy. J Control Release. 353:699–712. 2023. View Article : Google Scholar | |
|
Vangijzegem T, Stanicki D and Laurent S: Magnetic iron oxide nanoparticles for drug delivery: Applications and characteristics. Expert Opin Drug Deliv. 16:69–78. 2019. View Article : Google Scholar | |
|
Wang H, Qiao C, Guan Q, Wei M and Li Z: Nanoparticle-mediated synergistic anticancer effect of ferroptosis and photodynamic therapy: Novel insights and perspectives. Asian J Pharm Sci. 18:1008292023.PubMed/NCBI | |
|
Chang Q, Wang P, Zeng Q and Wang X: A review on ferroptosis and photodynamic therapy synergism: Enhancing anticancer treatment. Heliyon. 10:e289422024. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang L, Yi H, Song J, Huang J, Yang K, Tan B, Wang D, Yang N, Wang Z and Li X: Mitochondria-targeted and ultrasound-activated nanodroplets for enhanced deep-penetration sonodynamic cancer therapy. ACS Appl Mater Interfaces. 11:9355–9366. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Ding Y, Liu Z, Dai X, Ruan R, Zhong H, Wu Z, Yao Y, Chen J, Deng J and Xiong J: Ubiquitin-specific peptidase 49 promotes adenocarcinoma of the esophagogastric junction malignant progression via activating SHCBP1-β-catenin-GPX4 axis. Carcinogenesis. 46:bgae0602025. View Article : Google Scholar | |
|
Rittase WB, Slaven JE, Suzuki YJ, Muir JM, Lee SH, Rusnak M, Brehm GV, Bradfield DT, Symes AJ and Day RM: Iron deposition and ferroptosis in the spleen in a murine model of acute radiation syndrome. Int J Mol Sci. 23:110292022. View Article : Google Scholar : PubMed/NCBI | |
|
Wu Z, Chen T, Qian Y, Luo G, Liao F, He X, Xu W, Pu J and Ding S: High-dose ionizing radiation accelerates atherosclerotic plaque progression by regulating P38/NCOA4-mediated ferritinophagy/ferroptosis of endothelial cells. Int J Radiat Oncol Biol Phys. 117:223–236. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Fu S, Li Y, Shen L, Chen Y, Lu J, Ran Y, Zhao Y, Tang H, Tan L, Lin Q and Hao Y: Cu2WS4-PEG nanozyme as multifunctional sensitizers for enhancing immuno-radiotherapy by inducing ferroptosis. Small. 20:e23095372024. View Article : Google Scholar | |
|
Duan Q, Cui Z, Wang M, Li R, Han F and Ma J: Ginkgetin enhances breast cancer radiotherapy sensitization by suppressing NRF2-HO-1 axis activity. Toxicol Appl Pharmacol. 495:1171992025. View Article : Google Scholar | |
|
Shi J, Cui G, Jin Y, Mi B, Liu K, Zhao L, Bao K, Lu Z, Liu J, Wang Y, et al: Glutathione-depleted photodynamic nanoadjuvant for triggering nonferrous ferroptosis to amplify radiotherapy of breast cancer. Adv Healthc Mater. 13:e24024742024. View Article : Google Scholar : PubMed/NCBI | |
|
Hu H, Zheng S, He C, Zheng Y, Wei Q, Chen S, Wu Z, Xu Y, Zhao B and Yan C: Radiotherapy-sensitized cancer immunotherapy via cGAS-STING immune pathway by activatable nanocascade reaction. J Nanobiotechnology. 22:2342024. View Article : Google Scholar : PubMed/NCBI | |
|
Yu J, Zhang Y, Li L, Xiang Y, Yao X, Zhao Y, Cai K, Li M, Li Z and Luo Z: Coordination-driven FBXW7 DNAzyme-Fe nanoassembly enables a binary switch of breast cancer cell cycle checkpoint responses for enhanced ferroptosis-radiotherapy. Acta Biomater. 169:434–450. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Zheng S, Hu H, Hou M, Zhu K, Wu Z, Qi L, Xia H, Liu G, Ren Y, Xu Y, et al: Proton pump inhibitor-enhanced nanocatalytic ferroptosis induction for stimuli-responsive dual-modal molecular imaging guided cancer radiosensitization. Acta Biomater. 162:72–84. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Wei M, Bai J, Shen X, Lou K, Gao Y, Lv R, Wang P, Liu X and Zhang G: Glutathione-exhausting nanoprobes for NIR-II fluorescence imaging-guided surgery and boosting radiation therapy efficacy via ferroptosis in breast cancer. ACS Nano. 17:11345–11361. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang Z, Lo H, Zhao X, Li W, Wu K, Zeng F, Li S and Sun H: Mild photothermal/radiation therapy potentiates ferroptosis effect for ablation of breast cancer via MRI/PA imaging guided all-in-one strategy. J Nanobiotechnology. 21:1502023. View Article : Google Scholar : PubMed/NCBI | |
|
El-Benhawy SA, Abdelrhman IG, Sadek NA, Fahmy EI, AboGabal AA, Elmasry H, Saleh SAM, Sakr OA, Elwany MN and Rabie MAF: Studying ferroptosis and iron metabolism pre- and post-radiotherapy treatment in breast cancer patients. J Egypt Natl Canc Inst. 35:42023. View Article : Google Scholar : PubMed/NCBI | |
|
Hou YK, Zhang ZJ, Li RT, Peng J, Chen SY, Yue YR, Zhang WH, Sun B, Chen JX and Zhou Q: Remodeling the tumor microenvironment with core-shell nanosensitizer featuring dual-modal imaging and multimodal therapy for breast cancer. ACS Appl Mater Interfaces. 15:2602–2616. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Liu L, Zhang C, Qu S, Liu R, Chen H, Liang Z, Tian Z, Li L, Ma S and Liu X: ESR1 inhibits ionizing radiation-induced ferroptosis in breast cancer cells via the NEDD4L/CD71 pathway. Arch Biochem Biophys. 725:1092992022. View Article : Google Scholar : PubMed/NCBI | |
|
Liu R, Liu L, Bian Y, Zhang S, Wang Y, Chen H, Jiang X, Li G, Chen Q, Xue C, et al: The dual regulation effects of ESR1/NEDD4L on SLC7A11 in breast cancer under ionizing radiation. Front Cell Dev Biol. 9:7723802022. View Article : Google Scholar : PubMed/NCBI | |
|
Long Q, Tao H, Wang P, Wu B, Zhu Q, Chen H, Lao G, Yang Y, Liu G, Liu S and Wu Y: Fludarabine enhances radiosensitivity by promoting ferroptosis in B-cell lymphoma. Radiat Res. 201:224–239. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Chen H, Zhu P, Zhu D, Jin J, Yang Q and Han X: Role and mechanism of KIAA1429 in regulating cellular ferroptosis and radioresistance in colorectal cancer. Biomol Biomed. 24:1669–1681. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang Z, Ye B, Lin Y, Liu W, Deng J and Ji W: LncRNA OTUD6B-AS1 overexpression promoted GPX4-mediated ferroptosis to suppress radioresistance in colorectal cancer. Clin Transl Oncol. 25:3217–3229. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Shen D, Luo J, Chen L, Ma W, Mao X, Zhang Y, Zheng J, Wang Y, Wan J, Wang S, et al: PARPi treatment enhances radiotherapy-induced ferroptosis and antitumor immune responses via the cGAS signaling pathway in colorectal cancer. Cancer Lett. 550:2159192022. View Article : Google Scholar : PubMed/NCBI | |
|
Singhal R, Mitta SR, Das NK, Kerk SA, Sajjakulnukit P, Solanki S, Andren A, Kumar R, Olive KP, Banerjee R, et al: HIF-2α activation potentiates oxidative cell death in colorectal cancers by increasing cellular iron. J Clin Invest. 131:e1436912021. View Article : Google Scholar | |
|
Chen C, Gu W, Shao Y, Zheng P and Jiang J: Expression of glutathione peroxidase4 in patients with esophageal squamous carcinoma and its impact on the radiosensitivity of esophageal squamous carcinoma. Ann Clin Lab Sci. 54:160–169. 2024.PubMed/NCBI | |
|
Jin Y, Pan S, Wang M, Huang S, Ke Y, Li D, Luo H, Kou Z, Shi D, Kou W, et al: The m6A modification of ACSL4 mRNA sensitized esophageal squamous cell carcinoma to irradiation via accelerating ferroptosis. Cell Biol Int. 48:1877–1890. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Yan L, Hu H, Feng L, Li Z, Zheng C, Zhang J, Yin X and Li B: ML385 promotes ferroptosis and radiotherapy sensitivity by inhibiting the NRF2-SLC7A11 pathway in esophageal squamous cell carcinoma. Med Oncol. 41:3092024. View Article : Google Scholar : PubMed/NCBI | |
|
Chen J, Ying K, Sun J, Wang Y, Ji M and Sun Y: NEDD4L affects KLF5 stability through ubiquitination to control ferroptosis and radiotherapy resistance in oesophageal squamous cell carcinoma. J Cell Mol Med. 28:e700622024. View Article : Google Scholar : PubMed/NCBI | |
|
Heng J, Li Z, Liu L, Zheng Z, Zheng Y, Xu X, Liao L, Xu H, Huang H, Li E and Xu L: Acetyl-CoA acetyltransferase 2 confers radioresistance by inhibiting ferroptosis in esophageal squamous cell carcinoma. Int J Radiat Oncol Biol Phys. 117:966–978. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao M, Lu T, Bi G, Hu Z, Liang J, Bian Y, Feng M and Zhan C: PLK1 regulating chemoradiotherapy sensitivity of esophageal squamous cell carcinoma through pentose phosphate pathway/ferroptosis. Biomed Pharmacother. 168:1157112023. View Article : Google Scholar : PubMed/NCBI | |
|
Jiang K, Yin X, Zhang Q, Yin J, Tang Q, Xu M, Wu L, Shen Y, Zhou Z, Yu H and Yan S: STC2 activates PRMT5 to induce radioresistance through DNA damage repair and ferroptosis pathways in esophageal squamous cell carcinoma. Redox Biol. 60:1026262023. View Article : Google Scholar : PubMed/NCBI | |
|
Luo H, Wang X, Song S, Wang Y, Dan Q and Ge H: Targeting stearoyl-coa desaturase enhances radiation induced ferroptosis and immunogenic cell death in esophageal squamous cell carcinoma. Oncoimmunology. 11:21017692022. View Article : Google Scholar : PubMed/NCBI | |
|
Feng L, Zhao K, Sun L, Yin X, Zhang J, Liu C and Li B: SLC7A11 regulated by NRF2 modulates esophageal squamous cell carcinoma radiosensitivity by inhibiting ferroptosis. J Transl Med. 19:3672021. View Article : Google Scholar : PubMed/NCBI | |
|
Xue Y, Lu F, Chang Z, Li J, Gao Y, Zhou J, Luo Y, Lai Y, Cao S, Li X, et al: Intermittent dietary methionine deprivation facilitates tumoral ferroptosis and synergizes with checkpoint blockade. Nat Commun. 14:47582023. View Article : Google Scholar : PubMed/NCBI | |
|
Wang Z, Wang Y, Shen N, Liu Y, Xu X, Zhu R, Jiang H, Wu X, Wei Y and Tang J: AMPKα1-mediated ZDHHC8 phosphorylation promotes the palmitoylation of SLC7A11 to facilitate ferroptosis resistance in glioblastoma. Cancer Lett. 584:2166192024. View Article : Google Scholar | |
|
Li H, Qi X, He L, Yang H and Ju H: PRMT1 promotes radiotherapy resistance in glioma stem cells by inhibiting ferroptosis. Jpn J Radiol. 43:129–137. 2025. View Article : Google Scholar | |
|
Liu X, Cao Z, Wang W, Zou C, Wang Y, Pan L, Jia B, Zhang K, Zhang W, Li W, et al: Engineered extracellular vesicle-delivered CRISPR/Cas9 for radiotherapy sensitization of glioblastoma. ACS Nano. 17:16432–16447. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Cirotti C, Taddei I, Contadini C, Di Girolamo C, Pepe G, De Bardi M, Borsellino G, Helmer-Citterich M and Barilà D: NRF2 connects Src tyrosine kinase to ferroptosis resistance in glioblastoma. Life Sci Alliance. 7:e2023022052023. View Article : Google Scholar : PubMed/NCBI | |
|
Koike N, Kota R, Naito Y, Hayakawa N, Matsuura T, Hishiki T, Onishi N, Fukada J, Suematsu M, Shigematsu N, et al: 2-Nitroimidazoles induce mitochondrial stress and ferroptosis in glioma stem cells residing in a hypoxic niche. Commun Biol. 3:4502020. View Article : Google Scholar : PubMed/NCBI | |
|
He J, Li M, Bao J, Peng Y, Xue W, Chen J and Zhao J: β-Elemene promotes ferroptosis and reverses radioresistance in gastric cancer by inhibiting the OTUB1-GPX4 interaction. Front Pharmacol. 15:14691802024. View Article : Google Scholar | |
|
Wang H, Niu H, Luo X, Zhu N, Xiang J, He Y, Chen Z, Li G and Hu Y: Radiosensitizing effects of pyrogallol-loaded mesoporous or-ganosilica nanoparticles on gastric cancer by amplified ferroptosis. Front Bioeng Biotechnol. 11:11714502023. View Article : Google Scholar : PubMed/NCBI | |
|
Huang W, He Y, Yang S, Xue X, Qin H, Sun T and Yang W: CA9 knockdown enhanced ionizing radiation-induced ferroptosis and radiosensitivity of hypoxic glioma cells. Int J Radiat Biol. 99:1908–1924. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Sun S, Qi G, Chen H, He D, Ma D, Bie Y, Xu L, Feng B, Pang Q, Guo H and Zhang R: Ferroptosis sensitization in glioma: Exploring the regulatory mechanism of SOAT1 and its therapeutic implications. Cell Death Dis. 14:7542023. View Article : Google Scholar : PubMed/NCBI | |
|
Qiu L, Ji H, Wang K, Liu W, Huang Q, Pan X, Ye H, Li Z, Chen G, Xing X, et al: TLR3 activation enhances abscopal effect of radiotherapy in HCC by promoting tumor ferroptosis. EMBO Mol Med. 16:1193–1219. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Chen Q, Zheng W, Guan J, Liu H, Dan Y, Zhu L, Song Y, Zhou Y, Zhao X, Zhang Y, et al: SOCS2-enhanced ubiquitination of SLC7A11 promotes ferroptosis and radiosensitization in hepatocellular carcinoma. Cell Death Differ. 30:137–151. 2023. View Article : Google Scholar : | |
|
Feng H, Liu Y, Gan Y, Li M, Liu R, Liang Z, Liu L, Li L, Chen H, Li G, et al: AdipoR1 regulates ionizing radiation-induced ferroptosis in HCC cells through Nrf2/xCT pathway. Oxid Med Cell Longev. 2022:80914642022. View Article : Google Scholar : PubMed/NCBI | |
|
Dang W, Chen WC, Ju E, Xu Y, Li K, Wang H, Wang K, Lv S, Shao D, Tao Y and Li M: 3D printed hydrogel scaffolds combining glutathione depletion-induced ferroptosis and photothermia-augmented chemodynamic therapy for efficiently inhibiting postoperative tumor recurrence. J Nanobiotechnology. 20:2662022. View Article : Google Scholar : PubMed/NCBI | |
|
Yang M, Wu X, Hu J, Wang Y, Wang Y, Zhang L, Huang W, Wang X, Li N, Liao L, et al: COMMD10 inhibits HIF1α/CP loop to enhance ferroptosis and radiosensitivity by disrupting Cu-Fe balance in hepatocellular carcinoma. J Hepatol. 76:1138–1150. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Yuan Z, Liu T, Huo X, Wang H, Wang J and Xue L: Glutamine transporter SLC1A5 regulates ionizing radiation-derived oxidative damage and ferroptosis. Oxid Med Cell Longev. 2022:34030092022. View Article : Google Scholar : PubMed/NCBI | |
|
Yuan Y, Cao W, Zhou H, Qian H and Wang H: CLTRN, regulated by NRF1/RAN/DLD protein complex, enhances radiation sensitivity of hepatocellular carcinoma cells through ferroptosis pathway. Int J Radiat Oncol Biol Phys. 110:859–871. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Huang P, Ning X, Kang M and Wang R: Ferroptosis-related genes are associated with radioresistance and immune suppression in head and neck cancer. Genet Test Mol Biomarkers. 28:100–113. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Song A, Wu L, Zhang BX, Yang QC, Liu YT, Li H, Mao L, Xiong D, Yu HJ and Sun ZJ: Glutamine inhibition combined with CD47 blockade enhances radiotherapy-induced ferroptosis in head and neck squamous cell carcinoma. Cancer Lett. 588:2167272024. View Article : Google Scholar : PubMed/NCBI | |
|
Reinema FV, Hudson N, Adema GJ, Peeters WJM, Neuzil J, Stursa J, Werner L, Sweep FCGJ, Bussink J and Span PN: MitoTam induces ferroptosis and increases radiosensitivity in head and neck cancer cells. Radiother Oncol. 200:1105032024. View Article : Google Scholar : PubMed/NCBI | |
|
Chen L, Lin W, Zhang H, Geng S, Le Z, Wan F, Huang Q, Chen H, Liu X, Lu JJ and Kong L: TRIB3 promotes malignancy of head and neck squamous cell carcinoma via inhibiting ferroptosis. Cell Death Dis. 15:1782024. View Article : Google Scholar : PubMed/NCBI | |
|
Noh JK, Lee MK, Lee Y, Bae M, Min S, Kong M, Lee JW, Kim SI, Lee YC, Ko SG, et al: Targeting ferroptosis for improved radiotherapy outcomes in HPV-negative head and neck squamous cell carcinoma. Mol Oncol. 19:540–557. 2025. View Article : Google Scholar | |
|
Liao W, Chen X, Zhang S, Chen J, Liu C, Yu K, Zhang Y, Chen M, Chen F, Shen M, et al: Megakaryocytic IGF1 coordinates activation and ferroptosis to safeguard hematopoietic stem cell regeneration after radiation injury. Cell Commun Signal. 22:2922024. View Article : Google Scholar : PubMed/NCBI | |
|
Song H, Sun H, He N, Xu C, Du L, Ji K, Wang J, Zhang M, Gu Y, Wang Y and Liu Q: Glutathione depletion-induced versatile nanomedicine for potentiating the ferroptosis to overcome solid tumor radioresistance and enhance immunotherapy. Adv Healthc Mater. 13:e23034122021. View Article : Google Scholar | |
|
Almahi WA, Yu KN, Mohammed F, Kong P and Han W: Hemin enhances radiosensitivity of lung cancer cells through ferroptosis. Exp Cell Res. 410:1129462022. View Article : Google Scholar | |
|
Li Y, Yang J, Gu G, Guo X, He C, Sun J, Zou H, Wang H, Liu S, Li X, et al: Pulmonary delivery of theranostic nanoclusters for lung cancer ferroptosis with enhanced chemodynamic/radiation synergistic therapy. Nano Lett. 22:963–972. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang W, Sun Y, Bai L, Zhi L, Yang Y, Zhao Q, Chen C, Qi Y, Gao W, He W, et al: RBMS1 regulates lung cancer ferroptosis through translational control of SLC7A11. J Clin Invest. 131:e1520672021. View Article : Google Scholar : PubMed/NCBI | |
|
Rai A, Patwardhan RS, Jayakumar S, Pachpatil P, Das D, Panigrahi GC, Gota V, Patwardhan S and Sandur SK: Clobetasol propionate, a Nrf-2 inhibitor, sensitizes human lung cancer cells to radiation-induced killing via mitochondrial ROS-dependent ferroptosis. Acta Pharmacol Sin. 45:1506–1519. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Liang J, Bi G, Huang Y, Zhao G, Sui Q, Zhang H, Bian Y, Yin J, Wang Q, Chen Z and Zhan C: MAFF confers vulnerability to cisplatin-based and ionizing radiation treatments by modulating ferroptosis and cell cycle progression in lung adenocarcinoma. Drug Resist Updat. 73:1010572024. View Article : Google Scholar : PubMed/NCBI | |
|
Gotorbe C, Segui F, Echavidre W, Durivault J, Blanchard T, Vial V, Pagnuzzi-Boncompagni M, Villeneuve R, Amblard R, Garnier N, et al: Exploiting integrin-αVβ3 to enhance radiotherapy efficacy in medulloblastoma via ferroptosis. Curr Oncol. 31:7390–7402. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Zhou H, Wang YX, Wu M, Lan X, Xiang D, Cai R, Ma Q, Miao J, Fang X, Wang J, et al: FANCD2 deficiency sensitizes SHH medulloblastoma to radiotherapy via ferroptosis. J Pathol. 262:427–440. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Lin Y, Chen X, Yu C, Xu G, Nie X, Cheng Y, Luan Y and Song Q: Radiotherapy-mediated redox homeostasis-controllable nanomedicine for enhanced ferroptosis sensitivity in tumor therapy. Acta Biomater. 159:300–311. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Yang F, Gong H, Chen S, Li J, Huang N and Wang M: Depletion of SLC7A11 sensitizes nasopharyngeal carcinoma cells to ionizing radiation. Protein Pept Lett. 31:323–331. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Amos A, Jiang N, Zong D, Gu J, Zhou J, Yin L, He X, Xu Y and Wu L: Depletion of SOD2 enhances nasopharyngeal carcinoma cell radiosensitivity via ferroptosis induction modulated by DHODH inhibition. BMC Cancer. 23:1172023. View Article : Google Scholar : PubMed/NCBI | |
|
Huang WM, Li ZX, Wu YH, Shi ZL, Mi JL, Hu K and Wang RS: m6A demethylase FTO renders radioresistance of nasopharyngeal carcinoma via promoting OTUB1-mediated anti-ferroptosis. Transl Oncol. 27:1015762023. View Article : Google Scholar | |
|
Han F, Chen S, Zhang K, Zhang K, Wang M and Wang P: Targeting Nrf2/PHKG2 axis to enhance radiosensitivity in NSCLC. NPJ Precis Oncol. 8:1832024. View Article : Google Scholar : PubMed/NCBI | |
|
Ma Y, Peng Y, Cheng S and Jin L: Targeting MGST1 makes non-small cell lung cancer cells sensitive to radiotherapy by epigenetically enhancing ALOX15-mediated ferroptosis. Curr Cancer Drug Targets. September 27–2024.Epub ahead of print. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang Y, Liu X, Zeng L, Zhao X, Chen Q, Pan Y, Bai Y, Shao C and Zhang J: Exosomal protein angiopoietin-like 4 mediated radioresistance of lung cancer by inhibiting ferroptosis under hypoxic microenvironment. Br J Cancer. 127:1760–1772. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Bao Y, Pan Z, Zhao L, Qiu J, Cheng J, Liu L and Qian D: BIBR1532 combined with radiotherapy induces ferroptosis in NSCLC cells and activates cGAS-STING pathway to promote anti-tumor immunity. J Transl Med. 22:5192024. View Article : Google Scholar : PubMed/NCBI | |
|
Koppula P, Lei G, Zhang Y, Yan Y, Mao C, Kondiparthi L, Shi J, Liu X, Horbath A, Das M, et al: A targetable CoQ-FSP1 axis drives ferroptosis- and radiation-resistance in KEAP1 inactive lung cancers. Nat Commun. 13:22062022. View Article : Google Scholar : PubMed/NCBI | |
|
Xiao H, Jiang N, Zhang H, Wang S, Pi Q, Chen H, He X, Luo W, Lu Y, Deng Y and Zhong Z: Inhibitors of APE1 redox and ATM synergistically sensitize osteosarcoma cells to ionizing radiation by inducing ferroptosis. Int Immunopharmacol. 139:1126722024. View Article : Google Scholar : PubMed/NCBI | |
|
Lee HM, Kuo PC, Chen WH, Chen PJ, Lam SH, Su YC and Chen CH: Diterpenoid from croton tonkinensis as a potential radiation sensitizer in oral squamous cell carcinoma: An in vitro study. Int J Mol Sci. 25:118392024. View Article : Google Scholar : PubMed/NCBI | |
|
Liu J, An W, Zhao Q, Liu Z, Jiang Y, Li H and Wang D: Hyperbaric oxygen enhances X-ray induced ferroptosis in oral squamous cell carcinoma cells. Oral Dis. 30:116–127. 2024. View Article : Google Scholar | |
|
Guo SS, Chen MM, Yang YH, Zhang YY, Pang X, Shi YP, Zhuang YC, Fan DD, Bao JF and Ji ZY: Magnetic-vortex nanodonuts enhance ferroptosis effect of tumor ablation through an imaging-guided hyperthermia/radiosensitization strategy. iScience. 27:1105332024. View Article : Google Scholar : PubMed/NCBI | |
|
Cai J, Xu X and Saw PE: Nanomedicine targeting ferroptosis to overcome anticancer therapeutic resistance. Sci China Life Sci. 67:19–40. 2024. View Article : Google Scholar | |
|
Mao C, Lei G, Horbath A, Wang M, Lu Z, Yan Y, Liu X, Kondiparthi L, Chen X, Cheng J, et al: Unraveling ETC complex I function in ferroptosis reveals a potential ferroptosis-inducing therapeutic strategy for LKB1-deficient cancers. Mol Cell. 84:1964–1979.e6. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang HL, Hu BX, Ye ZP, Li ZL, Liu S, Zhong WQ, Du T, Yang D, Mai J, Li LC, et al: TRPML1 triggers ferroptosis defense and is a potential therapeutic target in AKT-hyperactivated cancer. Sci Transl Med. 16:eadk03302024. View Article : Google Scholar : PubMed/NCBI | |
|
Wu A, Han M, Ni Z, Li H, Chen Y, Yang Z, Feng Y, He Z, Zhen H and Wang X: Multifunctional Sr/Se co-doped ZIF-8 nanozyme for chemo/chemodynamic synergistic tumor therapy via apoptosis and ferroptosis. Theranostics. 14:1939–1955. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Deng Z, Li B, Yang M, Lu L, Shi X, Lovell JF, Zeng X, Hu W and Jin H: Irradiated microparticles suppress prostate cancer by tumor microenvironment reprogramming and ferroptosis. J Nanobiotechnology. 22:2252024. View Article : Google Scholar : PubMed/NCBI | |
|
Bi G, Liang J, Bian Y, Shan G, Huang Y, Lu T, Zhang H, Jin X, Chen Z, Zhao M, et al: Polyamine-mediated ferroptosis amplification acts as a targetable vulnerability in cancer. Nat Commun. 15:24612024. View Article : Google Scholar : PubMed/NCBI | |
|
Bae C, Hernández Millares R, Ryu S, Moon H, Kim D, Lee G, Jiang Z, Park MH, Kim KH, Koom WS, et al: Synergistic effect of ferroptosis-inducing nanoparticles and X-ray irradiation combination therapy. Small. 20:e23108732024. View Article : Google Scholar : PubMed/NCBI | |
|
Da Silva J, Bienassis C, Schmitt P, Berjaud C, Guedj M and Paris S: Radiotherapy-activated NBTXR3 nanoparticles promote ferroptosis through induction of lysosomal membrane permeabilization. J Exp Clin Cancer Res. 43:112024. View Article : Google Scholar : PubMed/NCBI | |
|
Zhu W, Cheng X, Xu P, Gu Y, Xu H, Xu J and Wang Y, Zhang LW and Wang Y: Radiotherapy-driven nanoprobes targeting for visualizing tumor infiltration dynamics and inducing ferroptosis in myeloid-derived suppressor cells. J Am Chem Soc. 146:22455–22468. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao J, Chen Y, Xiong T, Han S, Li C, He Y, He Y, Zhao G, Wang T, Wang L, et al: Clustered cobalt nanodots initiate ferroptosis by upregulating heme oxygenase 1 for radiotherapy sensitization. Small. 19:e22064152023. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang Q, Wang X, Zhao Y, Cheng Z, Fang D, Liu Y, Tian G, Li M and Luo Z: Nanointegrative in situ reprogramming of tumor-intrinsic lipid droplet biogenesis for low-dose radiation-activated ferroptosis immunotherapy. ACS Nano. 17:25419–25438. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Yang P, Li J, Zhang T, Ren Y, Zhang Q, Liu R, Li H, Hua J, Wang WA, Wang J and Zhou H: Ionizing radiation-induced mitophagy promotes ferroptosis by increasing intracellular free fatty acids. Cell Death Differ. 30:2432–2445. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Jiang W, Wei L, Chen B, Luo X, Xu P, Cai J and Hu Y: Platinum prodrug nanoparticles inhibiting tumor recurrence and metastasis by concurrent chemoradiotherapy. J Nanobiotechnology. 20:1292022. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao C, Yu D, He Z, Bao L, Feng L, Chen L, Liu Z, Hu X, Zhang N, Wang T and Fu Y: Endoplasmic reticulum stress-mediated autophagy activation is involved in cadmium-induced ferroptosis of renal tubular epithelial cells. Free Radic Biol Med. 175:236–248. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Tomita K, Nagasawa T, Kuwahara Y, Torii S, Igarashi K, Roudkenar MH, Roushandeh AM, Kurimasa A and Sato T: MiR-7-5p is involved in ferroptosis signaling and radioresistance Thru the generation of ROS in radioresistant HeLa and SAS cell lines. Int J Mol Sci. 22:83002021. View Article : Google Scholar : PubMed/NCBI | |
|
Takashi Y, Tomita K, Kuwahara Y, Roudkenar MH, Roushandeh AM, Igarashi K, Nagasawa T, Nishitani Y and Sato T: Mitochondrial dysfunction promotes aquaporin expression that controls hydrogen peroxide permeability and ferroptosis. Free Radic Biol Med. 161:60–70. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Chen M, Li X, Du B, Chen S and Li Y: Upstream stimulatory factor 2 inhibits erastin-induced ferroptosis in pancreatic cancer through transcriptional regulation of pyruvate kinase M2. Biochem Pharmacol. 205:1152552022. View Article : Google Scholar : PubMed/NCBI | |
|
Zhu Y, Fang S, Fan B, Xu K, Xu L, Wang L, Zhu L, Chen C, Wu R, Ni J and Wang J: Cancer-associated fibroblasts reprogram cysteine metabolism to increase tumor resistance to ferroptosis in pancreatic cancer. Theranostics. 14:1683–1700. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Feng Y, Luo X, Li Z, Fan X, Wang Y, He RR and Liu M: A ferroptosis-targeting ceria anchored halloysite as orally drug delivery system for radiation colitis therapy. Nat Commun. 14:50832023. View Article : Google Scholar : PubMed/NCBI | |
|
Wang X, Li W, Dong Y, Zhang Y, Huo Q, Lu L, Zhang J, Zhao Y, Fan S, Dong H and Li D: Ferrostatin-1 mitigates ionizing radiation-induced intestinal injuries by inhibiting apoptosis and ferroptosis: An in vitro and in vivo study. Int J Radiat Biol. 99:1607–1618. 2023. View Article : Google Scholar | |
|
Tang LF, Ma X, Xie LW, Zhou H, Yu J, Wang ZX and Li M: Perillaldehyde mitigates ionizing radiation-induced intestinal injury by inhibiting ferroptosis via the Nrf2 signaling pathway. Mol Nutr Food Res. 67:e23002322023. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang F, Liu T, Huang HC, Zhao YY, He M, Yuan W, Li L, Li J, Wu DM and Xu Y: Activation of pyroptosis and ferroptosis is involved in radiation-induced intestinal injury in mice. Biochem Biophys Res Commun. 631:102–109. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Zhou H, Zhou YL, Mao JA, Tang LF, Xu J, Wang ZX, He Y and Li M: NCOA4-mediated ferritinophagy is involved in ionizing radiation-induced ferroptosis of intestinal epithelial cells. Redox Biol. 55:1024132022. View Article : Google Scholar : PubMed/NCBI | |
|
Deng S, Wu D, Li L, Li J and Xu Y: TBHQ attenuates ferroptosis against 5-fluorouracil-induced intestinal epithelial cell injury and intestinal mucositis via activation of Nrf2. Cell Mol Biol Lett. 26:482021. View Article : Google Scholar : PubMed/NCBI | |
|
Xie LW, Cai S, Zhao TS, Li M and Tian Y: Green tea derivative (−)-epigallocatechin-3-gallate (EGCG) confers protection against ionizing radiation-induced intestinal epithelial cell death both in vitro and in vivo. Free Radic Biol Med. 161:175–186. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Ning X, Zhao W, Wu Q, Wang C and Liang S: Therapeutic potential of dihydroartemisinin in mitigating radiation-induced lung injury: Inhibition of ferroptosis through Nrf2/HO-1 pathways in mice. Immun Inflamm Dis. 12:e11752024. View Article : Google Scholar : PubMed/NCBI | |
|
Li X, Chen J, Yuan S, Zhuang X and Qiao T: Activation of the P62-Keap1-NRF2 pathway protects against ferroptosis in radiation-induced lung injury. Oxid Med Cell Longev. 2022:89735092022. View Article : Google Scholar : PubMed/NCBI | |
|
Li L, Wu D, Deng S, Li J, Zhang F, Zou Y, Zhang T and Xu Y: NVP-AUY922 alleviates radiation-induced lung injury via inhibition of autophagy-dependent ferroptosis. Cell Death Discov. 8:862022. View Article : Google Scholar : PubMed/NCBI | |
|
Guo XW, Zhang H, Huang JQ, Wang SN, Lu Y, Cheng B, Dong SH, Wang YY, Li FS and Li YW: PIEZO1 ion channel mediates ionizing radiation-induced pulmonary endothelial cell ferroptosis via Ca2+/calpain/VE-cadherin signaling. Front Mol Biosci. 8:7252742021. View Article : Google Scholar | |
|
Cao J, Wu M, Mo W, Zhao M, Gu L, Wang X, Zhang B and Cao J: Upregulation of PRRX2 by silencing Marveld3 as a protective mechanism against radiation-induced ferroptosis in skin cells. Mol Med. 30:1822024. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao J, Tang M, Tang H, Wang M, Guan H, Tang L and Zhang H: Sphingosine 1-phosphate alleviates radiation-induced ferroptosis in ovarian granulosa cells by upregulating glutathione peroxidase 4. Reprod Toxicol. 115:49–55. 2023. View Article : Google Scholar | |
|
Zhang L, Xu Y, Cheng Z, Zhao J, Wang M, Sun Y, Mi Z, Yuan Z and Wu Z: The EGR1/miR-139/NRF2 axis orchestrates radiosensitivity of non-small-cell lung cancer via ferroptosis. Cancer Lett. 595:2170002024. View Article : Google Scholar : PubMed/NCBI | |
|
Yang X, Zhang M, Xia W, Mai Z, Ye Y, Zhao B and Song Y: CHAC1 promotes cell ferroptosis and enhances radiation sensitivity in thyroid carcinoma. Neoplasma. 70:777–786. 2023. View Article : Google Scholar | |
|
Guo Y, Wang H, Wang X, Chen K and Feng L: Enhancing radiotherapy in triple-negative breast cancer with hesperetin-induced ferroptosis via AURKA targeting nanocomposites. J Nanobiotechnology. 22:7442024. View Article : Google Scholar : PubMed/NCBI | |
|
Chen R, Jiang Z, Cheng Y, Ye J, Li S, Xu Y, Ye Z, Shi Y, Ding J, Zhao Y, et al: Multifunctional iron-apigenin nanocomplex conducting photothermal therapy and triggering augmented immune response for triple negative breast cancer. Int J Pharm. 655:1240162024. View Article : Google Scholar : PubMed/NCBI | |
|
Zeng L, Ding S, Cao Y, Li C, Zhao B, Ma Z, Zhou J, Hu Y, Zhang X, Yang Y, et al: A MOF-based potent ferroptosis inducer for enhanced radiotherapy of triple negative breast cancer. ACS Nano. 17:13195–13210. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Sun H, Cai H, Xu C, Zhai H, Lux F, Xie Y, Feng L, Du L, Liu Y, Sun X, et al: AGuIX nanoparticles enhance ionizing radiation-induced ferroptosis on tumor cells by targeting the NRF2-GPX4 signaling pathway. J Nanobiotechnology. 20:4492022. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang Z, Lu M, Chen C, Tong X, Li Y, Yang K, Lv H, Xu J and Qin L: Holo-lactoferrin: The link between ferroptosis and radiotherapy in triple-negative breast cancer. Theranostics. 11:3167–3182. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Tan H, Shen Z, Wang X, Shu S, Deng J, Lu L, Fan Z, Hu D, Cheng P, Cao X and Huang Q: Endoplasmic reticulum-targeted biomimetic nanoparticles induce apoptosis and ferroptosis by regulating endoplasmic reticulum function in colon cancer. J Control Release. 375:422–437. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
de la Harpe A, Beukes N and Frost C: Mitochondrial calcium overload contributes to cannabinoid-induced paraptosis in hormone-responsive breast cancer cells. Cell Prolif. 57:e136502024. View Article : Google Scholar : PubMed/NCBI | |
|
Qian Y, Ma S, Qiu R, Sun Z, Liu W, Wu F, Lam SM, Xia Z, Wang K, Fang L, et al: Golgi protein ACBD3 downregulation sensitizes cells to ferroptosis. Cell Biol Int. 48:1559–1572. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Bai Y, Meng L, Han L, Jia Y, Zhao Y, Gao H, Kang R, Wang X, Tang D and Dai E: Lipid storage and lipophagy regulates ferroptosis. Biochem Biophys Res Commun. 508:997–1003. 2019. View Article : Google Scholar | |
|
Lee H, Horbath A, Kondiparthi L, Meena JK, Lei G, Dasgupta S, Liu X, Zhuang L, Koppula P, Li M, et al: Cell cycle arrest induces lipid droplet formation and confers ferroptosis resistance. Nat Commun. 15:792024. View Article : Google Scholar : PubMed/NCBI | |
|
Wang Y, Song Y, Xu L, Zhou W, Wang W, Jin Q, Xie Y, Zhang J, Liu J, Wu W, et al: A membrane-targeting aggregation-induced emission probe for monitoring lipid droplet dynamics in ischemia/reperfusion-induced cardiomyocyte ferroptosis. Adv Sci (Weinh). 11:e23099072024. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao J, Wang Q, Liu Z, Zhang M, Li J, Fu ZF, Zhao L and Zhou M: Neuroinvasive virus facilitates viral replication by employing lipid droplets to reduce arachidonic acid-induced ferroptosis. J Biol Chem. 300:1071682024. View Article : Google Scholar : PubMed/NCBI | |
|
Ferrada L, Barahona MJ, Vera M, Stockwell BR and Nualart F: Dehydroascorbic acid sensitizes cancer cells to system xc− inhibition-induced ferroptosis by promoting lipid droplet peroxidation. Cell Death Dis. 14:6372023. View Article : Google Scholar | |
|
Wei D, Dai Y, Cao J and Fu N: A novel fluorescent probe for visualizing viscosity changes in lipid droplets during chemotherapy-induced ferroptosis. Anal Chim Acta. 1299:3424222024. View Article : Google Scholar : PubMed/NCBI | |
|
Silva MJSA, Zhang Y, Vinck R, Santos FMF, António JPM, Gourdon-Grünewaldt L, Zaouter C, Castonguay A, Patten SA, Cariou K, et al: BASHY dyes are highly efficient lipid droplet-targeting photosensitizers that induce ferroptosis through lipid peroxidation. Bioconjug Chem. 34:2337–2344. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Swanda RV, Ji Q, Wu X, Yan J, Dong L, Mao Y, Uematsu S, Dong Y and Qian SB: Lysosomal cystine governs ferroptosis sensitivity in cancer via cysteine stress response. Mol Cell. 83:3347–3359.e9. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Armenta DA, Laqtom NN, Alchemy G, Dong W, Morrow D, Poltorack CD, Nathanson DA, Abu-Remalieh M and Dixon SJ: Ferroptosis inhibition by lysosome-dependent catabolism of extracellular protein. Cell Chem Biol. 29:1588–1600.e7. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Bhardwaj M, Lee JJ, Versace AM, Harper SL, Goldman AR, Crissey MAS, Jain V, Singh MP, Vernon M, Aplin AE, et al: Lysosomal lipid peroxidation regulates tumor immunity. J Clin Invest. 133:e1645962023. View Article : Google Scholar : PubMed/NCBI | |
|
Mai TT, Hamaï A, Hienzsch A, Cañeque T, Müller S, Wicinski J, Cabaud O, Leroy C, David A, Acevedo V, et al: Salinomycin kills cancer stem cells by sequestering iron in lysosomes. Nat Chem. 9:1025–1033. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Wang ZX, Ma J, Li XY, Wu Y, Shi H, Chen Y, Lu G, Shen HM, Lu GD and Zhou J: Quercetin induces p53-independent cancer cell death through lysosome activation by the transcription factor EB and reactive oxygen species-dependent ferroptosis. Br J Pharmacol. 178:1133–1148. 2021. View Article : Google Scholar | |
|
Feng Y, Wei H, Lyu M, Yu Z, Chen J, Lyu X and Zhuang F: Iron retardation in lysosomes protects senescent cells from ferroptosis. Aging (Albany NY). 16:7683–7703. 2024.PubMed/NCBI | |
|
Feng J, Wang ZX, Bin JL, Chen YX, Ma J, Deng JH, Huang XW, Zhou J and Lu GD: Pharmacological approaches for targeting lysosomes to induce ferroptotic cell death in cancer. Cancer Lett. 587:2167282024. View Article : Google Scholar : PubMed/NCBI | |
|
Ralhan I, Chang J, Moulton MJ, Goodman LD, Lee NYJ, Plummer G, Pasolli HA, Matthies D, Bellen HJ and Ioannou MS: Autolysosomal exocytosis of lipids protect neurons from ferroptosis. J Cell Biol. 222:e2022071302023. View Article : Google Scholar : PubMed/NCBI | |
|
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 (Weinh). 11:e23020932024. View Article : Google Scholar | |
|
Peng S, Chen G, Yu KN, Feng Y, Zhao L, Yang M, Cao W, Almahi WAA, Sun M, Xu Y, Zhao Y, et al: Synergism of non-thermal plasma and low concentration RSL3 triggers ferroptosis via promoting xCT lysosomal degradation through ROS/AMPK/mTOR axis in lung cancer cells. Cell Commun Signal. 22:1122024. View Article : Google Scholar : PubMed/NCBI | |
|
Liu Y, Pang Z, Wang Y, Liu J, Wang G and Du J: Targeting PKD2 aggravates ferritinophagy-mediated ferroptosis via promoting autophagosome-lysosome fusion and enhances efficacy of carboplatin in lung adenocarcinoma. Chem Biol Interact. 387:1107942024. View Article : Google Scholar | |
|
Yang YH, Li W, Ren LW, Yang H, Zhang YZ, Zhang S, Hao Y, Yu DK, Tong RS, Du GH, et al: S670, an amide derivative of 3-O-acetyl-11-keto-β-boswellic acid, induces ferroptosis in human glioblastoma cells by generating ROS and inhibiting STX17-mediated fusion of autophagosome and lysosome. Acta Pharmacol Sin. 45:209–222. 2024. View Article : Google Scholar | |
|
Li C, Sun S, Zhuang Y, Luo Z, Ji G and Liu Z: CTSB nuclear translocation facilitates DNA damage and lysosomal stress to promote retinoblastoma cell death. Mol Biotechnol. 66:2583–2594. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Zhu X, Wang L, Wang K, Yao Y and Zhou F: Erdafitinib promotes ferroptosis in human uveal melanoma by inducing ferritinophagy and lysosome biogenesis via modulating the FGFR1/mTORC1/TFEB signaling axis. Free Radic Biol Med. 222:552–568. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Chen Y, Yang Z, Wang S, Ma Q, Li L, Wu X, Guo Q, Tao L and Shen X: Boosting ROS-mediated lysosomal membrane permeabilization for cancer ferroptosis therapy. Adv Healthc Mater. 12:e22021502023. View Article : Google Scholar | |
|
An S and Hu M: Quercetin promotes TFEB nuclear translocation and activates lysosomal degradation of ferritin to induce ferroptosis in breast cancer cells. Comput Intell Neurosci. 2022:52992182022. View Article : Google Scholar : PubMed/NCBI | |
|
You P, Lu F, Ouyang C, Yu J, González-García J, Song J, Ni W, Wang J, Yin C and Zhou CQ: Acidic lysosome-anchoring croconium-based nanoplatform for enhanced triple-mode bioimaging and Fe3+-triggered tumor synergistic therapy. ACS Appl Mater Interfaces. 16:46066–46078. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang Z, Xiang J, Guan L, Chen P, Li C, Guo C, Hu Y, Huang S, Cai L and Gong P: Inducing tumor ferroptosis via a pH-responsive NIR-II photothermal agent initiating lysosomal dysfunction. Nanoscale. 15:19074–19078. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Liang Q, Yu F, Cai H, Wu X, Ma M, Li Z, Tedesco AC, Zhu J, Xu Q and Bi H: Photo-activated autophagy-associated tumour cell death by lysosome impairment based on manganese-doped graphene quantum dots. J Mater Chem B. 11:2466–2477. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Turcu AL, Versini A, Khene N, Gaillet C, Cañeque T, Müller S and Rodriguez R: DMT1 inhibitors kill cancer stem cells by blocking lysosomal iron translocation. Chemistry. 26:7369–7373. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Torii S, Shintoku R, Kubota C, Yaegashi M, Torii R, Sasaki M, Suzuki T, Mori M, Yoshimoto Y, Takeuchi T and Yamada K: An essential role for functional lysosomes in ferroptosis of cancer cells. Biochem J. 473:769–777. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Li W, Yin S, Shen Y, Li H, Yuan L and Zhang XB: Molecular engineering of pH-responsive NIR oxazine assemblies for evoking tumor ferroptosis via triggering lysosomal dysfunction. J Am Chem Soc. 145:3736–3747. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Jiang L, Zheng H, Lyu Q, Hayashi S, Sato K, Sekido Y, Nakamura K, Tanaka H, Ishikawa K, Kajiyama H, et al: Lysosomal nitric oxide determines transition from autophagy to ferroptosis after exposure to plasma-activated Ringer's lactate. Redox Biol. 43:1019892021. View Article : Google Scholar : PubMed/NCBI | |
|
Lan Y, Zhang K, Wang F, Zhang Y, Yan M and Zuo Y: Polysiloxane-based hyperbranched fluorescent probe for dynamic visualization of HClO in lysosomes and vivo. Spectrochim Acta A Mol Biomol Spectrosc. 294:1225272023. View Article : Google Scholar : PubMed/NCBI | |
|
Sun J, Cheng X, Pan S, Wang L, Dou W, Liu J and Shi X: Dichloroacetate attenuates the stemness of colorectal cancer cells via trigerring ferroptosis through sequestering iron in lysosomes. Environ Toxicol. 36:520–529. 2021. View Article : Google Scholar | |
|
Hirata Y, Oka K, Yamamoto S, Watanabe H, Oh-Hashi K, Hirayama T, Nagasawa H, Takemori H and Furuta K: Haloperidol prevents oxytosis/ferroptosis by targeting lysosomal ferrous ions in a manner independent of dopamine D2 and sigma-1 receptors. ACS Chem Neurosci. 13:2719–2727. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Fang X, Wang H, Han D, Xie E, Yang X, Wei J, Gu S, Gao F, Zhu N, Yin X, et al: Ferroptosis as a target for protection against cardiomyopathy. Proc Natl Acad Sci USA. 116:2672–2680. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Kalkavan H, Chen MJ, Crawford JC, Quarato G, Fitzgerald P, Tait SWG, Goding CR and Green DR: Sublethal cytochrome c release generates drug-tolerant persister cells. Cell. 185:3356–3374.e22. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Wei S, Qiu T, Yao X, Wang N, Jiang L, Jia X, Tao Y, Wang Z, Pei P, Zhang J, et al: Arsenic induces pancreatic dysfunction and ferroptosis via mitochondrial ROS-autophagy-lysosomal pathway. J Hazard Mater. 384:1213902020. View Article : Google Scholar | |
|
Ahola S, Rivera Mejías P, Hermans S, Chandragiri S, Giavalisco P, Nolte H and Langer T: OMA1-mediated integrated stress response protects against ferroptosis in mitochondrial cardiomyopathy. Cell Metab. 34:1875–1891.e7. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Sun L, Wang H, Yu S, Zhang L, Jiang J and Zhou Q: Herceptin induces ferroptosis and mitochondrial dysfunction in H9c2 cells. Int J Mol Med. 49:172022. View Article : Google Scholar : | |
|
Yao S, Pang M, Wang Y, Wang X, Lin Y, Lv Y, Xie Z, Hou J, Du C, Qiu Y, et al: Mesenchymal stem cell attenuates spinal cord injury by inhibiting mitochondrial quality control-associated neuronal ferroptosis. Redox Biol. 67:1028712023. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang Z, Zhou H, Gu W, Wei Y, Mou S, Wang Y, Zhang J and Zhong Q: CGI1746 targets σ1R to modulate ferroptosis through mitochondria-associated membranes. Nat Chem Biol. 20:699–709. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Park MW, Cha HW, Kim J, Kim JH, Yang H, Yoon S, Boonpraman N, Yi SS, Yoo ID and Moon JS: NOX4 promotes ferroptosis of astrocytes by oxidative stress-induced lipid peroxidation via the impairment of mitochondrial metabolism in Alzheimer's diseases. Redox Biol. 41:1019472021. View Article : Google Scholar : PubMed/NCBI | |
|
Liang FG, Zandkarimi F, Lee J, Axelrod JL, Pekson R, Yoon Y, Stockwell BR and Kitsis RN: OPA1 promotes ferroptosis by augmenting mitochondrial ROS and suppressing an integrated stress response. Mol Cell. 84:3098–3114.e6. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Akiyama H, Zhao R, Ostermann LB, Li Z, Tcheng M, Yazdani SJ, Moayed A, Pryor ML II, Slngh S, Baran N, et al: Mitochondrial regulation of GPX4 inhibition-mediated ferroptosis in acute myeloid leukemia. Leukemia. 38:729–740. 2024. View Article : Google Scholar | |
|
Deshwal S, Onishi M, Tatsuta T, Bartsch T, Cors E, Ried K, Lemke K, Nolte H, Giavalisco P and Langer T: Mitochondria regulate intracellular coenzyme Q transport and ferroptotic resistance via STARD7. Nat Cell Biol. 25:246–257. 2023.PubMed/NCBI | |
|
Jang S, Chapa-Dubocq XR, Tyurina YY, St Croix CM, Kapralov AA, Tyurin VA, Bayır H, Kagan VE and Javadov S: Elucidating the contribution of mitochondrial glutathione to ferroptosis in cardiomyocytes. Redox Biol. 45:1020212021. View Article : Google Scholar : PubMed/NCBI | |
|
Wang P, Cui Y, Ren Q, Yan B, Zhao Y, Yu P, Gao G, Shi H, Chang S and Chang YZ: Mitochondrial ferritin attenuates cerebral ischaemia/reperfusion injury by inhibiting ferroptosis. Cell Death Dis. 12:4472021. View Article : Google Scholar : PubMed/NCBI | |
|
Chen T, Majerníková N, Marmolejo-Garza A, Trombetta-Lima M, Sabogal-Guáqueta AM, Zhang Y, Ten Kate R, Zuidema M, Mulder PPMFA, den Dunnen W, et al: Mitochondrial transplantation rescues neuronal cells from ferroptosis. Free Radic Biol Med. 208:62–72. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Wu S, Mao C, Kondiparthi L, Poyurovsky MV, Olszewski K and Gan B: A ferroptosis defense mechanism mediated by glycerol-3-phosphate dehydrogenase 2 in mitochondria. Proc Natl Acad Sci USA. 119:e21219871192022. View Article : Google Scholar : PubMed/NCBI | |
|
Chen Y, Li S, Yin M, Li Y, Chen C, Zhang J, Sun K, Kong X, Chen Z and Qian J: Isorhapontigenin attenuates cardiac microvascular injury in diabetes via the inhibition of mitochondria-associated ferroptosis through PRDX2-MFN2-ACSL4 pathways. Diabetes. 72:389–404. 2023. View Article : Google Scholar | |
|
Li MD, Fu L, Lv BB, Xiang Y, Xiang HX, Xu DX and Zhao H: Arsenic induces ferroptosis and acute lung injury through mtROS-mediated mitochondria-associated endoplasmic reticulum membrane dysfunction. Ecotoxicol Environ Saf. 238:1135952022. View Article : Google Scholar : PubMed/NCBI | |
|
Chen J, Fu CY, Shen G, Wang J, Xu L, Li H, Cao X, Zheng MZ, Shen YL, Zhong J, et al: Macrophages induce cardiomyocyte ferroptosis via mitochondrial transfer. Free Radic Biol Med. 190:1–14. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Guo J, Wang S, Wan X, Liu X, Wang Z, Liang C, Zhang Z, Wang Y, Yan M, Wu P, et al: Mitochondria-derived methylmalonic acid aggravates ischemia-reperfusion injury by activating reactive oxygen species-dependent ferroptosis. Cell Commun Signal. 22:532024. View Article : Google Scholar : PubMed/NCBI | |
|
Li Z, Zhao B, Zhang Y, Fan W, Xue Q, Chen X, Wang J and Qi X: Mitochondria-mediated ferroptosis contributes to the inflammatory responses of bovine viral diarrhea virus (BVDV) in vitro. J Virol. 98:e01880232024. View Article : Google Scholar : PubMed/NCBI | |
|
Tan Q, Zhang X, Li S, Liu W, Yan J, Wang S, Cui F, Li D and Li J: DMT1 differentially regulates mitochondrial complex activities to reduce glutathione loss and mitigate ferroptosis. Free Radic Biol Med. 207:32–44. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Li Y, Zhou Y, Liu D, Wang Z, Qiu J, Zhang J, Chen P, Zeng G, Guo Y, Wang X, et al: Glutathione Peroxidase 3 induced mitochondria-mediated apoptosis via AMPK/ERK1/2 pathway and resisted autophagy-related ferroptosis via AMPK/mTOR pathway in hyperplastic prostate. J Transl Med. 21:5752023. View Article : Google Scholar | |
|
Xiao Q, Yan L, Han J, Yang S, Tang Y, Li Q, Lao X, Chen Z, Xiao J, Zhao H, et al: Metabolism-dependent ferroptosis promotes mitochondrial dysfunction and inflammation in CD4+ T lymphocytes in HIV-infected immune non-responders. EBioMedicine. 86:1043822022. View Article : Google Scholar | |
|
Wu T, Liang X, Liu X, Li Y, Wang Y, Kong L and Tang M: Induction of ferroptosis in response to graphene quantum dots through mitochondrial oxidative stress in microglia. Part Fibre Toxicol. 17:302020. View Article : Google Scholar : PubMed/NCBI | |
|
Sun L, Wang H, Xu D, Yu S, Zhang L and Li X: Lapatinib induces mitochondrial dysfunction to enhance oxidative stress and ferroptosis in doxorubicin-induced cardiomyocytes via inhibition of PI3K/AKT signaling pathway. Bioengineered. 13:48–60. 2022. View Article : Google Scholar : | |
|
Qian B, Che L, Du ZB, Guo NJ, Wu XM, Yang L, Zheng ZX, Gao YL, Wang MZ, Chen XX, et al: Protein phosphatase 2A-B55β mediated mitochondrial p-GPX4 dephosphorylation promoted sorafenib-induced ferroptosis in hepatocellular carcinoma via regulating p53 retrograde signaling. Theranostics. 13:4288–4302. 2023. View Article : Google Scholar : | |
|
Zeng Y, He Y and Wang L, Xu H, Zhang Q, Wang Y, Zhang J and Wang L: Dihydroquercetin improves experimental acute liver failure by targeting ferroptosis and mitochondria-mediated apoptosis through the SIRT1/p53 axis. Phytomedicine. 128:1555332024. View Article : Google Scholar : PubMed/NCBI | |
|
Yang H, Li Q, Chen X, Weng M, Huang Y, Chen Q, Liu X, Huang H, Feng Y, Zhou H, et al: Targeting SOX13 inhibits assembly of respiratory chain supercomplexes to overcome ferroptosis resistance in gastric cancer. Nat Commun. 15:42962024. View Article : Google Scholar : PubMed/NCBI | |
|
Du C, Liu C, Yu K, Zhang S, Fu Z, Chen X, Liao W, Chen J, Zhang Y, Wang X, et al: Mitochondrial serine catabolism safeguards maintenance of the hematopoietic stem cell pool in homeostasis and injury. Cell Stem Cell. 31:1484–1500.e9. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Ding Q, Tang W, Li X, Ding Y, Chen X, Cao W, Wang X, Mo W, Su Z, Zhang Q and Guo H: Mitochondrial-targeted brequinar liposome boosted mitochondrial-related ferroptosis for promoting checkpoint blockade immunotherapy in bladder cancer. J Control Release. 363:221–234. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Reichardt C, Brandt S, Bernhardt A, Krause A, Lindquist JA, Weinert S, Geffers R, Franz T, Kahlfuss S, Dudeck A, et al: DNA-binding protein-A promotes kidney ischemia/reperfusion injury and participates in mitochondrial function. Kidney Int. 106:241–257. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Wu X, Pan J, Yu JJ, Kang J, Hou S, Cheng M, Xu L, Gong L and Li Y: DiDang decoction improves mitochondrial function and lipid metabolism via the HIF-1 signaling pathway to treat atherosclerosis and hyperlipidemia. J Ethnopharmacol. 308:1162892023. View Article : Google Scholar : PubMed/NCBI | |
|
Dong S, Zhang M, Cheng Z, Zhang X, Liang W, Li S, Li L, Xu Q, Song S, Liu Z, et al: Redistribution of defective mitochondria-mediated dihydroorotate dehydrogenase imparts 5-fluorouracil resistance in colorectal cancer. Redox Biol. 73:1032072024. View Article : Google Scholar : PubMed/NCBI | |
|
Guo YY, Liang NN, Zhang XY, Ren YH, Wu WZ, Liu ZB, He YZ, Zhang YH, Huang YC, Zhang T, et al: Mitochondrial GPX4 acetylation is involved in cadmium-induced renal cell ferroptosis. Redox Biol. 73:1031792024. View Article : Google Scholar : PubMed/NCBI | |
|
Yang M, Chen X, Cheng C, Yan W, Guo R, Wang Y, Zhang H, Chai J, Cheng Y and Zhang F: Cucurbitacin B induces ferroptosis in oral leukoplakia via the SLC7A11/mitochondrial oxidative stress pathway. Phytomedicine. 129:1555482024. View Article : Google Scholar : PubMed/NCBI | |
|
Lu C, Zhang Z, Fan Y, Wang X, Qian J and Bian Z: Shikonin induces ferroptosis in osteosarcomas through the mitochondrial ROS-regulated HIF-1α/HO-1 axis. Phytomedicine. 135:1561392024. View Article : Google Scholar | |
|
Hai Y, Fan R, Zhao T, Lin R, Zhuang J, Deng A, Meng S, Hou Z and Wei G: A novel mitochondria-targeting DHODH inhibitor induces robust ferroptosis and alleviates immune suppression. Pharmacol Res. 202:1071152024. View Article : Google Scholar : PubMed/NCBI | |
|
Yang B, Xu Y, Yu J, Wang Q, Fan Q, Zhao X, Qiao Y, Zhang Z, Zhou Q, Yin D, et al: Salidroside pretreatment alleviates ferroptosis induced by myocardial ischemia/reperfusion through mitochondrial superoxide-dependent AMPKα2 activation. Phytomedicine. 128:1553652024. View Article : Google Scholar | |
|
Lv T, Xiong X, Yan W, Liu M, Xu H and He Q: Mitochondrial general control of amino acid synthesis 5 like 1 promotes nonalcoholic steatohepatitis development through ferroptosis-induced formation of neutrophil extracellular traps. Clin Transl Med. 13:e13252023. View Article : Google Scholar : PubMed/NCBI | |
|
Qi Y, Chen L, Ding S, Shen X, Wang Z, Qi H and Yang S: Neutrophil extracellular trap-induced ferroptosis promotes abdominal aortic aneurysm formation via SLC25A11-mediated depletion of mitochondrial glutathione. Free Radic Biol Med. 221:215–224. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Zhou Q, Dian Y, He Y, Yao L, Su H, Meng Y, Sun Y, Li D, Xiong Y, Zeng F, et al: Propafenone facilitates mitochondrial-associated ferroptosis and synergizes with immunotherapy in melanoma. J Immunother Cancer. 12:e0098052024. View Article : Google Scholar : PubMed/NCBI | |
|
Tang J, Zhu J, Xie H, Song L, Xu G, Li W, Cai L and Han XX: Mitochondria-specific molecular crosstalk between ferroptosis and apoptosis revealed by in situ raman spectroscopy. Nano Lett. 24:2384–2391. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang X, Zhang M, Zhang Z and Zhou S: Salidroside induces mitochondrial dysfunction and ferroptosis to inhibit melanoma progression through reactive oxygen species production. Exp Cell Res. 438:1140342024. View Article : Google Scholar : PubMed/NCBI | |
|
Yang C, Yang X, Harrington A, Potts C, Kaija A, Ryzhova L and Liaw L: Notch signaling regulates mouse perivascular adipose tissue function via mitochondrial pathways. Genes (Basel). 14:19642023. View Article : Google Scholar : PubMed/NCBI | |
|
Hirata Y, Yamada Y, Taguchi S, Kojima R, Masumoto H, Kimura S, Niijima T, Toyama T, Kise R, Sato E, et al: Conjugated fatty acids drive ferroptosis through chaperone-mediated autophagic degradation of GPX4 by targeting mitochondria. Cell Death Dis. 15:8842024. View Article : Google Scholar : PubMed/NCBI | |
|
Chen H, Nie P, Li J, Wu Y, Yao B, Yang Y, Lash GE and Li P: Cyclophosphamide induces ovarian granulosa cell ferroptosis via a mechanism associated with HO-1 and ROS-mediated mitochondrial dysfunction. J Ovarian Res. 17:1072024. View Article : Google Scholar : PubMed/NCBI | |
|
Jiang J, Zhou X, Chen H, Wang X, Ruan Y, Liu X and Ma J: 18β-Glycyrrhetinic acid protects against deoxynivalenol-induced liver injury via modulating ferritinophagy and mitochondrial quality control. J Hazard Mater. 471:1343192024. View Article : Google Scholar | |
|
Wang X, Wei T, Luo J, Lang K, Song Y, Ning X, Chao Y, Gu Z, Wang L, Chen C, et al: Iron overload-dependent ferroptosis aggravates LPS-induced acute lung injury by impairing mitochondrial function. Inflammation. 47:2013–2026. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Liang NN, Guo YY, Zhang XY, Ren YH, He YZ, Liu ZB, Xu DX and Xu S: Mitochondrial dysfunction-evoked DHODH acetylation is involved in renal cell ferroptosis during cisplatin-induced acute kidney injury. Adv Sci (Weinh). 11:e24047532024. View Article : Google Scholar : PubMed/NCBI | |
|
Tang S, Fuß A, Fattahi Z and Culmsee C: Drp1 depletion protects against ferroptotic cell death by preserving mitochondrial integrity and redox homeostasis. Cell Death Dis. 15:6262024. View Article : Google Scholar : PubMed/NCBI | |
|
Hu H, Zhang F, Sheng Z, Tian S, Li G, Tang S, Niu Y, Yang J and Liu Y: Synthesis and mitochondria-localized iridium (III) complexes induce cell death through pyroptosis and ferroptosis pathways. Eur J Med Chem. 268:1162952024. View Article : Google Scholar : PubMed/NCBI | |
|
Song X, Hao X and Zhu BT: Role of mitochondrial reactive oxygen species in chemically-induced ferroptosis. Free Radic Biol Med. 223:473–492. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Lin YS, Tsai YC, Li CJ, Wei TT, Wang JL, Lin BW, Wu YN, Wu SR and Lin SC and Lin SC: Overexpression of NUDT16L1 sustains proper function of mitochondria and leads to ferroptosis insensitivity in colorectal cancer. Redox Biol. 77:1033582024. View Article : Google Scholar : PubMed/NCBI | |
|
Deng J, Zhuang H, Shao S, Zeng X, Xue P, Bai T, Wang X, Shangguan S, Chen Y, Yan S and Huang W: Mitochondrial-targeted copper delivery for cuproptosis-based synergistic cancer therapy. Adv Healthc Mater. 13:e23045222024. View Article : Google Scholar : PubMed/NCBI | |
|
Lyamzaev KG, Panteleeva AA, Simonyan RA, Avetisyan AV and Chernyak BV: Mitochondrial lipid peroxidation is responsible for ferroptosis. Cells. 12:6112023. View Article : Google Scholar : PubMed/NCBI | |
|
Zhong S, Chen W, Wang B, Gao C, Liu X, Song Y, Qi H, Liu H, Wu T, Wang R and Chen B: Energy stress modulation of AMPK/FoxO3 signaling inhibits mitochondria-associated ferroptosis. Redox Biol. 63:1027602023. View Article : Google Scholar : PubMed/NCBI | |
|
Li C, Liu J, Hou W, Kang R and Tang D: STING1 promotes ferroptosis through MFN1/2-dependent mitochondrial fusion. Front Cell Dev Biol. 9:6986792021. View Article : Google Scholar : PubMed/NCBI | |
|
Wang X, Chen J, Tie H, Tian W, Zhao Y, Qin L, Guo S, Li Q and Bao C: Eriodictyol regulated ferroptosis, mitochondrial dysfunction, and cell viability via Nrf2/HO-1/NQO1 signaling pathway in ovarian cancer cells. J Biochem Mol Toxicol. 37:e233682023. 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 | |
|
Ward NP, Yoon SJ, Flynn T, Sherwood AM, Olley MA, Madej J and DeNicola GM: Mitochondrial respiratory function is preserved under cysteine starvation via glutathione catabolism in NSCLC. Nat Commun. 15:42442024. View Article : Google Scholar : PubMed/NCBI | |
|
Zheng J, Wang Q, Chen J, Cai G, Zhang Z, Zou H, Zou JX, Liu Q, Ji S, Shao G, et al: Tumor mitochondrial oxidative phosphorylation stimulated by the nuclear receptor RORγ represents an effective therapeutic opportunity in osteosarcoma. Cell Rep Med. 5:1015192024. View Article : Google Scholar | |
|
Luo Y, Zhang Y, Pang S, Min J, Wang T, Wu D, Lin C, Xiao Z, Xiang Q, Li Q and Ma L: PCBP1 protects bladder cancer cells from mitochondria injury and ferroptosis by inducing LACTB mRNA degradation. Mol Carcinog. 62:907–919. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Wang Z, Tang S, Cai L, Wang Q, Pan D, Dong Y, Zhou H, Li J, Ji N, Zeng X, et al: DRP1 inhibition-mediated mitochondrial elongation abolishes cancer stemness, enhances glutaminolysis, and drives ferroptosis in oral squamous cell carcinoma. Br J Cancer. 130:1744–1757. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Kumar A, Ye C, Nkansah A, Decoville T, Fogo GM, Sajjakulnukit P, Reynolds MB, Zhang L, Quaye O, Seo YA, et al: Iron regulates the quiescence of naive CD4 T cells by controlling mitochondria and cellular metabolism. Proc Natl Acad Sci USA. 121:e23184201212024. View Article : Google Scholar : PubMed/NCBI | |
|
Wang W, Xu X, Zhao L, Ye K, Wang S and Lin C: 3,5-diCQA suppresses colorectal cancer cell growth through ROS/AMPK/mTOR mediated mitochondrial dysfunction and ferroptosis. Cell Cycle. 22:1951–1968. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Feng Y, Xu J, Shi M, Liu R, Zhao L, Chen X, Li M, Zhao Y, Chen J, Du W and Liu P: COX7A1 enhances the sensitivity of human NSCLC cells to cystine deprivation-induced ferroptosis via regulating mitochondrial metabolism. Cell Death Dis. 13:9882022. View Article : Google Scholar : PubMed/NCBI | |
|
Wang J, Zhuang H, Yang X, Guo Z, Zhou K, Liu N, An Y, Chen Y, Zhang Z, Wang M, et al: Exploring the mechanism of ferroptosis induction by sappanone A in cancer: Insights into the mitochondrial dysfunction mediated by NRF2/xCT/GPX4 axis. Int J Biol Sci. 20:5145–5161. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Li H, Guo Y, Su W, Zhang H, Wei X, Ma X, Gong S, Qu G, Zhang L, Xu H, et al: The mitochondria-targeted antioxidant MitoQ ameliorates inorganic arsenic-induced DCs/Th1/Th2/Th17/Treg differentiation partially by activating PINK1-mediated mitophagy in murine liver. Ecotoxicol Environ Saf. 277:1163502024. View Article : Google Scholar : PubMed/NCBI | |
|
Lin J, Ou H, Luo B, Ling M, Lin F, Cen L, Hu Z, Ye L and Pan L: Capsaicin mitigates ventilator-induced lung injury by suppressing ferroptosis and maintaining mitochondrial redox homeostasis through SIRT3-dependent mechanisms. Mol Med. 30:1482024. View Article : Google Scholar : PubMed/NCBI | |
|
Vaishampayan P and Lee Y: Redox-active vitamin C suppresses human osteosarcoma growth by triggering intracellular ROS-iron-calcium signaling crosstalk and mitochondrial dysfunction. Redox Biol. 75:1032882024. View Article : Google Scholar : PubMed/NCBI | |
|
Fu X, Qu L, Xu H and Xie J: Ndfip1 protected dopaminergic neurons via regulating mitochondrial function and ferroptosis in Parkinson's disease. Exp Neurol. 375:1147242024. View Article : Google Scholar : PubMed/NCBI | |
|
He M, Cai Y, Yuan Z, Zhang L and Lu H: Upregulation of SOCS2 causes mitochondrial dysfunction and promotes ferroptosis in pancreatic cancer cells. Acta Biochim Pol. 70:163–168. 2023.PubMed/NCBI | |
|
Zhu ZH, Xu XT, Shen CJ, Yuan JT, Lou SY, Ma XL, Chen X, Yang B and Zhao HJ: A novel sesquiterpene lactone fraction from Eupatorium chinense L. suppresses hepatocellular carcinoma growth by triggering ferritinophagy and mitochondrial damage. Phytomedicine. 112:1546712023. View Article : Google Scholar : PubMed/NCBI | |
|
Neitemeier S, Jelinek A, Laino V, Hoffmann L, Eisenbach I, Eying R, Ganjam GK, Dolga AM, Oppermann S and Culmsee C: BID links ferroptosis to mitochondrial cell death pathways. Redox Biol. 12:558–570. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Mu M, Chen B, Li H, Fan R, Yang Y, Zhou L, Han B, Zou B, Chen N and Guo G: Augmented the sensitivity of photothermal-ferroptosis therapy in triple-negative breast cancer through mitochondria-targeted nanoreactor. J Control Release. 375:733–744. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Geroyska S, Mejia I, Chan AA, Navarrete M, Pandey V, Kharpatin S, Noguti J, Wang F, Srole D, Chou TF, et al: N-Myristoytransferase inhibition causes mitochondrial iron overload and parthanatos in TIM17A-dependent aggressive lung carcinoma. Cancer Res Commun. 4:1815–1833. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Li LG, Zhang D, Huang Q, Yan M, Chen NN, Yang Y, Xiao RC, Liu H, Han N, Qureshi AM, et al: Mitochondrial disruption resulting from Cepharanthine-mediated TOM inhibition triggers ferroptosis in colorectal cancer cells. J Cancer Res Clin Oncol. 150:4602024. View Article : Google Scholar : PubMed/NCBI | |
|
Vu NT, Kim M, Stephenson DJ, MacKnight HP and Chalfant CE: Ceramide kinase inhibition drives ferroptosis and sensitivity to cisplatin in mutant KRAS lung cancer by dysregulating VDAC-mediated mitochondria function. Mol Cancer Res. 20:1429–1442. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Shen J, He Y, Zhou B, Qin H, Zhang S, Huang Z and Zhang X: TFAP2C/FLT3 axis reduces ferroptosis in breast cancer cells by inhibiting mitochondrial autophagy. Int J Biochem Cell Biol. 177:1066912024. View Article : Google Scholar : PubMed/NCBI | |
|
He LW, Lin CJ, Zhuang LJ, Sun YH, Li YC and Ye ZY: Targeting hepatocellular carcinoma: Schisandrin A triggers mitochondrial disruption and ferroptosis. Chem Biol Drug Des. 104:e700102024. View Article : Google Scholar : PubMed/NCBI | |
|
Sun X, Sun P, Zhen D, Xu X, Yang L, Fu D, Wei C, Niu X, Tian J and Li H: Melatonin alleviates doxorubicin-induced mitochondrial oxidative damage and ferroptosis in cardiomyocytes by regulating YAP expression. Toxicol Appl Pharmacol. 437:1159022022. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao B, Xu X, Wen X, Liu Q, Dong C, Yang Q, Fan C, Yoon J and Lu Z: Ratiometric near-infrared fluorescent probe monitors ferroptosis in HCC Cells by imaging HClO in mitochondria. Anal Chem. 96:5992–6000. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Lei L, Dong Z, Xu L, Yang F, Yin B, Wang Y, Yue R, Guan G, Xu J, Song G and Zhang XB: Metal-fluorouracil networks with disruption of mitochondrion enhanced ferroptosis for synergistic immune activation. Theranostics. 12:6207–6222. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Yu H, Li JM, Deng K, Zhou W, Li KH, Wang CX, Wang Q, Wu M and Huang SW: GPX4 inhibition synergistically boosts mitochondria targeting nanoartemisinin-induced apoptosis/ferroptosis combination cancer therapy. Biomater Sci. 11:5831–5845. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Fang H, Chen Y, Geng S, Yao S, Guo Z and He W: Super-resolution imaging of mitochondrial HClO during cell ferroptosis using a near-infrared fluorescent probe. Anal Chem. 94:17904–17912. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Negi M, Kaushik N, Lamichhane P, Patel P, Jaiswal A, Choi EH and Kaushik NK: Nitric oxide water-driven immunogenic cell death: Unfolding mitochondrial dysfunction's role in sensitizing lung adenocarcinoma to ferroptosis and autophagic cell death. Free Radic Biol Med. 222:1–15. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Ye RR, Chen BC, Lu JJ, Ma XR and Li RT: Phosphorescent rhenium(I) complexes conjugated with artesunate: Mitochondrial targeting and apoptosis-ferroptosis dual induction. J Inorg Biochem. 223:1115372021. View Article : Google Scholar : PubMed/NCBI | |
|
Guo L: Mitochondrial permeability transition mediated by MTCH2 and F-ATP synthase contributes to ferroptosis defense. FEBS Lett. 599:352–366. 2025. View Article : Google Scholar | |
|
Liang Z, Zhang N, Wang X, Zhang J, Li K and Lei T: Epothilone B inactivation of Sirtuin1 promotes mitochondrial reactive oxygen species to induce dysfunction and ferroptosis of Schwann cells. Eur J Pharm Sci. 181:1063502023. View Article : Google Scholar | |
|
Yang F, Yu W, Yu Q, Liu X, Liu C, Lu C, Liao X, Liu Y and Peng N: Mitochondria-targeted nanosystem with reactive oxygen species-controlled release of CO to enhance photodynamic therapy of PCN-224 by sensitizing ferroptosis. Small. 19:e22061242023. View Article : Google Scholar : PubMed/NCBI | |
|
Huang Y, Liu G, Zheng F, Chen J, Lin Y, Wang J, Huang Y and Peng Y: Asymmetric silicon phthalocyanine based nanoparticle with spatiotemporally targeting of mitochondria for synergistic apoptosis-ferroptosis antitumor treatment. Colloids Surf B Biointerfaces. 238:1138902024. View Article : Google Scholar : PubMed/NCBI | |
|
Wang J, Zhao Z, Liu Y, Cao X, Li F, Ran H, Cao Y and Wu C: 'Mito-Bomb': A novel mitochondria-targeting nanosystem for ferroptosis-boosted sonodynamic antitumor therapy. Drug Deliv. 29:3111–3122. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Yin Y, Chen S, Li H, Pang X, Wang C, Wang L, Liu P, Xu S and Luo X: Exogenous and endogenous dual-activated nanoladder for precise imaging of mitochondrial ferroptosis-related inhibition miRNA with tumor cell specificity. Anal Chem. 96:7550–7557. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Lee YS, Kalimuthu K, Park YS, Luo X, Choudry MHA, Bartlett DL and Lee YJ: BAX-dependent mitochondrial pathway mediates the crosstalk between ferroptosis and apoptosis. Apoptosis. 25:625–631. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Nikoo A, Roudkenar MH, Sato T, Kuwahara Y, Tomita K, Pourmohammadi-Bejarpasi Z, Najafi-Ghalehlou N and Roushandeh AM: Mitochondrial transfer in PC-3 cells fingerprinted in ferroptosis sensitivity: A brand new approach targeting cancer metabolism. Hum Cell. 36:1441–1450. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Tian L, Ji H, Wang W, Han X, Zhang X, Li X, Guo L, Huang L and Gao W: Mitochondria-targeted pentacyclic triterpenoid carbon dots for selective cancer cell destruction via inducing autophagy, apoptosis, as well as ferroptosis. Bioorg Chem. 130:1062592023. View Article : Google Scholar | |
|
Xia T, Xia Z, Tang P, Fan J and Peng X: Light-driven mitochondrion-to-nucleus DNA cascade fluorescence imaging and enhanced cancer cell photoablation. J Am Chem Soc. 146:12941–12949. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Asperti M, Bellini S, Grillo E, Gryzik M, Cantamessa L, Ronca R, Maccarinelli F, Salvi A, De Petro G, Arosio P, et al: H-ferritin suppression and pronounced mitochondrial respiration make Hepatocellular carcinoma cells sensitive to RSL3-induced ferroptosis. Free Radic Biol Med. 169:294–303. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Tan M, Duan J, Chen S, Chen Y, Wang J, Xu X and Ke F: Construction of a mitochondria-targeted probe to monitor cysteine levels in cancer cells and zebrafish. Photochem Photobiol Sci. 23:1425–1434. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Xie Y, Zhou X, Li J, Yao XC, Liu WL, Kang FH, Zou ZX, Xu KP, Xu PS and Tan GS: Identification of a new natural biflavonoids against breast cancer cells induced ferroptosis via the mitochondrial pathway. Bioorg Chem. 109:1047442021. View Article : Google Scholar : PubMed/NCBI | |
|
Karmi O, Sohn YS, Zandalinas SI, Rowland L, King SD, Nechushtai R and Mittler R: Disrupting CISD2 function in cancer cells primarily impacts mitochondrial labile iron levels and triggers TXNIP expression. Free Radic Biol Med. 176:92–104. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Fedotcheva TA and Fedotcheva NI: Protectors of the mitochondrial permeability transition pore activated by iron and doxorubicin. Curr Cancer Drug Targets. 21:514–525. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Ogor P, Yoshida T, Koike M and Kakizuka A: VCP relocalization limits mitochondrial activity, GSH depletion and ferroptosis during starvation in PC3 prostate cancer cells. Genes Cells. 26:570–582. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Wei G, Sun J, Hou Z, Luan W, Wang S, Cui S, Cheng M and Liu Y: Novel antitumor compound optimized from natural saponin Albiziabioside A induced caspase-dependent apoptosis and ferroptosis as a p53 activator through the mitochondrial pathway. Eur J Med Chem. 157:759–772. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
DeHart DN, Fang D, Heslop K, Li L, Lemasters JJ and Maldonado EN: Opening of voltage dependent anion channels promotes reactive oxygen species generation, mitochondrial dysfunction and cell death in cancer cells. Biochem Pharmacol. 148:155–162. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Li Y, Xia J, Shao F, Zhou Y, Yu J, Wu H, Du J and Ren X: Sorafenib induces mitochondrial dysfunction and exhibits synergistic effect with cysteine depletion by promoting HCC cells ferroptosis. Biochem Biophys Res Commun. 534:877–884. 2021. View Article : Google Scholar | |
|
Zheng Z, Zeng Y, Bao X, Huang C, Guo F, Xu F and Luo Z: OTULIN confers cisplatin resistance in osteosarcoma by mediating GPX4 protein homeostasis to evade the mitochondrial apoptotic pathway. J Exp Clin Cancer Res. 43:3302024. View Article : Google Scholar : PubMed/NCBI | |
|
Sun Y, Deng R and Zhang C: Erastin induces apoptotic and ferroptotic cell death by inducing ROS accumulation by causing mitochondrial dysfunction in gastric cancer cell HGC-27. Mol Med Rep. 22:2826–2832. 2020.PubMed/NCBI | |
|
Ren J, Zhou J, Liu H, Jiao X, Cao Y, Xu Z, Kang Y and Xue P: Ultrasound (US)-activated redox dyshomeostasis therapy reinforced by immunogenic cell death (ICD) through a mitochondrial targeting liposomal nanosystem. Theranostics. 11:9470–9491. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Liu S, Shang M, Gong J, Sun H and Hu B: WTAP regulates mitochondrial damage and Lipid oxidation in HCC by NOA1 mediated m6A modification. J Cancer. 16:315–330. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Yagi M, Do Y, Hirai H, Miki K, Toshima T, Fukahori Y, Setoyama D, Abe C, Nabeshima YI, Kang D and Uchiumi T: Improving lysosomal ferroptosis with NMN administration protects against heart failure. Life Sci Alliance. 6:e2023021162023. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao Z, Wu Y, Liang X, Liu J, Luo Y, Zhang Y, Li T, Liu C, Luo X, Chen J, et al: Sonodynamic therapy of NRP2 monoclonal antibody-guided MOFs@COF targeted disruption of mitochondrial and endoplasmic reticulum homeostasis to induce autophagy-dependent ferroptosis. Adv Sci (Weinh). 10:e23038722023. View Article : Google Scholar : PubMed/NCBI | |
|
Rykowski S, Gurda-Woźna D, Fedoruk-Wyszomirska A, Orlicka-Płocka M, Kowalczyk A, Stączek P, Denel-Bobrowska M, Biniek-Antosiak K, Rypniewski W, Wyszko E and Olejniczak AB: Carboranyl-1,8-naphthalimide intercalators induce lysosomal membrane permeabilization and ferroptosis in cancer cell lines. J Enzyme Inhib Med Chem. 38:21710282023. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang G, Wang Y, Lin J, Wang B, Mohsin A, Cheng Z, Hao W, Gao WQ, Xu H and Guo M: Biological activity reduction and mitochondrial and lysosomal dysfunction of mesenchymal stem cells aging in vitro. Stem Cell Res Ther. 13:4112022. View Article : Google Scholar : PubMed/NCBI | |
|
Zou Y, Henry WS, Ricq EL, Graham ET, Phadnis VV, Maretich P, Paradkar S, Boehnke N, Deik AA, Reinhardt F, et al: Plasticity of ether lipids promotes ferroptosis susceptibility and evasion. Nature. 585:603–608. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Tang D and Kroemer G: Peroxisome: The new player in ferroptosis. Signal Transduct Target Ther. 5:2732020. View Article : Google Scholar : PubMed/NCBI | |
|
Gan Y, Deng J, Hao Q, Huang Y, Han T, Xu JG, Zhao M, Yao L, Xu Y, Xiong J, et al: UTP11 deficiency suppresses cancer development via nucleolar stress and ferroptosis. Redox Biol. 62:1027052023. View Article : Google Scholar : PubMed/NCBI | |
|
Li Z, Ferguson L, Deol KK, Roberts MA, Magtanong L, Hendricks JM, Mousa GA, Kilinc S, Schaefer K, Wells JA, et al: Ribosome stalling during selenoprotein translation exposes a ferroptosis vulnerability. Nat Chem Biol. 18:751–761. 2022. View Article : Google Scholar : PubMed/NCBI |