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.
Biliary tract cancer (BTC) includes a spectrum of invasive malignancies that arise from the bile duct epithelium. It is relatively uncommon but highly heterogeneous, comprising gallbladder cancer (GBC; which arises from the gallbladder or cystic duct) and cholangiocarcinoma (CCA; which arises from the intrahepatic, perihilar or distal biliary tree). BTC accounts for <1% of all human types of cancer and ~3% of digestive system tumors globally (1,2). In recent years, the incidence of BTC has been increasing globally, particularly in Asian countries, where it represents an important health problem (3,4). Globally, adenocarcinoma accounts for ~90% of all BTC cases (5,6). BTC is usually diagnosed at advanced stage. The majority of patients with symptomatic BTC, such as those presenting with biliary obstruction, have incurable tumors with invasion of the surrounding organs, lymph nodes and distant metastases, which results in poor prognoses (2,7). At present, surgery remains the gold standard for treating early-stage tumors; however, only a limited number of patients have this opportunity (8,9). The 5-year overall survival rate of BTC is <20% for patients from the United States of America and Europe (10). Furthermore, despite undergoing curative resection with a negative margin (R0), BTC still have a high recurrence rate of >50% (8,9,11). Gemcitabine-platinum chemotherapy is a recommended first-line treatment for advanced BTC; however, its efficacy is not satisfactory (12). The emergence of targeted therapies and immunotherapies, such as Durvalumab and Pembrolizumab, has revolutionized the treatment paradigm of malignant tumors. Immunotherapy plus chemotherapy has been verified to be effective in treating BTC (13-15). Investigating the key molecular mechanisms and identifying novel therapeutic targets for BTC is imperative.
Cell death can occur either due to unregulated causes or following a regulated process (Fig. 1) (16). Unregulated cell death is the instantaneous demise of cells due to severe physical, chemical or mechanical causes. Unlike unregulated cell death, regulated cell death (RCD) is characterized by organized signaling cascades and specific molecular mechanisms, suggesting that it can be controlled (17). The process of RCD can be influenced, either delayed or accelerated, through pharmacological or genetic interventions. A study by Dixon et al (18) first proposed the notion of 'ferroptosis' in 2012 (19). Ferroptosis is a new type of RCD, differing from necroptosis (Fig. 1A) (20), apoptosis (Fig. 1B) (21), pyroptosis (Fig. 1C) (22) and autophagy (Fig. 1D) (23) in morphology, genetics and biochemistry (24,25). As its name implies, ferroptosis is an iron-dependent mechanism of cell death (26). Ferroptosis is triggered by lipid peroxidation (LPO), which results from an imbalanced cellular metabolism and redox homeostasis. The morphological evidence of ferroptosis includes the presence of small mitochondria with increased mitochondrial membrane densities, reduced or missing mitochondrial crista, outer mitochondrial membrane rupture and a normal nucleus (Fig. 1E) (27,28). Ferroptosis is indicated to serve an important pathophysiological role in the development and occurrence of numerous diseases, such as ischemic heart disease and acute kidney injury, but particularly in cancer (19,29). Non-small cell lung cancer, hepatocellular carcinoma, pancreatic cancer, breast cancer and renal cell carcinoma are reported to be sensitive to ferroptosis (30-34). Additionally, various ferroptosis inducers, such as sorafenib, sulfasalazine and artemisinin, demonstrate a mitigation of tumor progression (35). Due to the issue of drug resistance, there is a growing interest regarding the potential of targeting ferroptosis as a therapeutic strategy for malignancies (36-38).
The present review aimed to provide a comprehensive overview of the signaling pathways and molecular mechanisms of ferroptosis, the research progress on ferroptosis in the occurrence and development of BTC, and the potential application of ferroptosis as a targeted therapy for BTC.
Following the first description of ferroptosis, a form of non-apoptosis and iron-dependent RCD, in 2012, there has been an increasing interest to investigate the process and regulation of ferroptosis (39). Previously, several studies investigated the mechanisms involved in ferroptosis (40-42). The present study aimed to review the core mechanisms of ferroptosis based on three aspects, namely iron metabolism, lipid metabolism and antioxidant defense systems.
Iron is one of the crucial minor elements for human health, and the correct intracellular level of iron is indispensable for a well-functioning metabolic balance (43). Iron is mainly obtained from dietary intake, existing in two distinct oxidation states. These are the divalent (Fe2+) and trivalent (Fe3+) ions (44). The continuous interconversion of Fe2+ and Fe3+ allows iron-dependent cofactors, such as cytochrome P450s, to effectively carry out their catalytic functions in various biological processes, including nucleic acid synthesis as well as repair, epigenetic regulation and cellular respiration (45). While in the Fe2+ state or combined with a transporter protein, iron is primarily absorbed in the duodenum and upper jejunum epithelial cells. The less soluble Fe3+ ion must first be reduced to absorbable ferrous Fe2+ and is then transported via divalent metal transporter 1 (DMT1) into epithelial cells (46). Absorbed iron then either enters the blood circulation via ferroportin (FPN) or is stored as ferritin. Fe3+ is the primary form of iron in blood circulation. FPN, encoded by the transporter solute carrier family 40 member 1 gene, is the only known iron-exporting protein in mammalian cells. It is responsible for exporting Fe3+ to the extracellular space (47). Subsequently, multi-copper ferroxidases on the epithelial cell membrane re-oxidize Fe2+ to Fe3+, which is then bound by transferrin (TF) and transported in the plasma (44). Lactotransferrin also contributes positively to the regulation of iron absorption by enhancing iron uptake, similar to TF (48). The majority of cells mainly obtain non-heme iron through two primary pathways that involve TF-bound iron uptake and non-TF-bound iron (NTBI) uptake. TF binds to transferrin receptor (TFR)1 and forms endosomes through clathrin-dependent endocytosis. Within cells, TF-bound iron separates, and six-transmembrane epithelial antigen of prostate 3 reduces Fe3+ to Fe2+, which then enters the cytoplasm via DMT1 for subsequent use or storage as ferritin (49,50). When there is an iron overload, such as in hemochromatosis, excess iron surpasses the capacity of TF, resulting in the increased circulation and uptake of NTBI (forming an unstable iron pool) through transporters such as solute carrier family 39 member 14 (51). The main repository for intracellular iron is the ferritin dimer, consisting of ferritin heavy and light chains. Nuclear receptor coactivator 4 (NCOA4) increases iron levels by facilitating ferritin degradation (52-54).
Ferroptosis relies on iron, as suggested by its name. An elevated iron level in the body has a direct association with the onset of ferroptosis (18). Excessive iron levels increase the production of reactive oxygen species (ROS), leading to cell dysfunction or death, tissue damage and diseases, such as human leukocyte antigen-linked hemochromatosis and Friedreich ataxia (44). An excessive accumulation of iron can result in ferroptosis either through Fenton reactions or by activating arachidonate lipoxygenases and cytochrome P450 oxidoreductase-mediated phospholipid peroxidation metabolism (55,56). As an important step in ferroptosis, the Fenton reaction involves the non-enzymatic oxidation of organic compounds, such as polyunsaturated fatty acids (PUFAs), into inorganic states by a mixture of hydrogen peroxide (H2O2) and Fe2+ (37). Glutathione peroxidase 4 (GPX4) is the major neutralizing enzyme of phospholipid (PL) hydroperoxides (OOHs; PLOOHs). GPX4 knockdown results in an accumulation of PLOOHs, which induces an increase in lipid peroxidation and ferroptosis (57).
Uncontrolled LPO represents a key hallmark of ferroptosis. A key initiation step of ferroptosis involves an excessive oxidation of PUFA-PLs (19). The susceptibility of PUFAs to peroxidation is attributed to their diallyl moieties (55). The magnitude of LPO is influenced by both the abundance and distribution of PUFAs within phospholipids (28). Arachidonic acid (AA) and adrenic acid (AdA) serve as the primary LPO substrates in ferroptosis. Acyl-CoA synthetase long-chain family member 4 (ACSL4) catalyzes the esterification of AA or AdA with CoA, generating oxidizable AA- or AdA-CoA. Subsequently, lysophosphatidylcholine acyltransferase 3 (LPCAT3) facilitates the PL remodeling of AA- or AdA-CoA and membrane phosphatidylethanolamine (PE) into AA- or AdA-PE, anchoring it to the cell or mitochondrial membranes (58). Conversely, acyl-CoA synthetase long-chain family member 3 (ACSL3) and stearoyl-CoA desaturase 1 (SCD1) confer cellular resistance to ferroptosis by catalyzing the synthesis of monounsaturated fatty acid (MUFA) and the displacement of PUFAs from plasma membrane phospholipids (55,59,60). Subsequently, 15-lipoxygenase (LOX) then oxidizes AA-PE or AdA-PE to PL-PUFA-OOHs, which acts as a ferroptosis signal (58). However, 12-LOX can mediate the ACSL4-independent pathway of LPO production via the p53/solute carrier family 7 member 11 (SLC7A11)/12-LOX axis (61).
The SLC7A11-GSH-GPX4 axis is the principal antioxidant defense system (62). The cystine/glutamate antiporter (system Xc−), located on the cell membrane, consists of solute carrier family 3 member 2 (SLC3A2) and SLC7A11 subunits. System Xc− facilitates the 1:1 exchange of extracellular cystine and intracellular glutamate. Intracellular cystine is reduced to cysteine, a precursor for GSH synthesis. GSH serves as an essential cofactor for GPX4. GPX4 uses GSH to reduce PL-PUFA-OOH to non-toxic alcohol phospholipid hydroxides, thereby halting the LPO chain reaction (63,64). The activity of SLC7A11 is downregulated by TP53, BRCA1-associated protein 1 and beclin-1, and upregulated by nuclear factor erythroid 2-related factor 2 (Nrf2) (65) (Fig. 2).
FSP1 is a GSH-independent ferroptosis suppressor, acting in parallel to GPX4. When GPX4 is knocked out, FSP1 (also known as apoptosis-inducing factor mitochondrial-associated protein 2) is activated as a compensatory antioxidant system. FSP1 is located on the plasma membrane and uses NAD(P)H to reduce CoQ to ubiquinol (CoQH2), which acts as an antioxidant by binding to radicals. This neutralizes lipid peroxide free radicals and blocks the propagation of LPO (66,67) (Fig. 2).
The GCH1-BH4 system is another important GPX4-independent ferroptosis inhibitor. GCH1 prevents ferroptosis via the synthesis of its metabolic derivatives, BH4 and dihydrobiopterin (BH2), and lipid remodeling. GCH1 overexpression selectively protects those PLs containing two PUFA chains from degradation, which potentially has two simultaneous mechanisms: i) Acting as an antioxidant that directly binds to radicals; and ii) contributing to the synthesis of CoQ (68,69) (Fig. 2).
The DHODH-CoQH2 system is a ferroptosis defense pathway, which is localized in the mitochondria and independent of cytosolic GPX4 or FSP1. Knocking out DHODH markedly induces mitochondrial LPO, inducing ferroptosis in cancer cells with low GPX4 expression levels. Furthermore, combining inhibitors of DHODH (such as brequinar) with inducers of ferroptosis (such as sulfasalazine) induces mitochondrial LPO and ferroptosis in malignancies with high GPX4 expression levels synergistically (70). Mechanistically, DHODH suppresses ferroptosis at the mitochondrial inner membrane by reducing CoQ to CoQH2, which is a parallel mechanism to the mitochondrial GPX4 pathway (70) (Fig. 2).
As PL-modifying enzymes, MBOAT1 and MBOAT2 function as GPX4/FSP1-independent ferroptosis suppressors. MBOAT1 and 2 inhibit ferroptosis by remodeling PLs by selectively incorporating MUFAs into lyso-PE. This increases the cellular MUFA-PE levels and decreases the PUFA-PE levels (71). However, MBOAT1 and 2 function as sex hormone-dependent regulators, and their transcription is upregulated by androgen and estrogen receptors (71) (Fig. 2).
The TME serves pivotal roles in the initiation, progression, metastasis and therapeutic resistance of tumors (72-75). It is a dynamic interacting network that is comprised of diverse cellular components, including malignant cells, immune cells, cancer-associated fibroblasts and endothelial cells, alongside acellular elements such as the extracellular matrix, cytokines and metabolites (76). Ferroptosis engages in a complex bidirectional crosstalk with the TME, particularly through metabolic reprogramming and immunomodulation, which regulates BTC malignancy and treatment responses (77,78).
Ferroptosis in malignant cells modulates antitumor immunity within the TME through immunogenic signaling (79,80). Previous evidence indicates that ferroptotic cancer cells release damage-associated molecular patterns (DAMPs), a hallmark of immunogenic cell death (ICD), which function as endogenous adjuvants to potentiate antitumor immunity (81,82). As well as the classic DAMPs, such as high mobility group protein 1 (83-85), adenosine triphosphate (86,87), calcium reticulum protein (84,86) and the recently identified proteoglycan decorin (88), ferroptotic cancer cells release immunomodulatory cytokines such as C-X-C motif chemokine ligand 1 (CXCL1), tumor necrosis factor (TNF) and interferon (IFN)-β, that contribute to the remodeling of the TME (89). However, whether ferroptosis is a classic form of ICD is still an ongoing scientific debate (89). In addition, the specific role of ferroptotic cancer cells in antitumor immunity is also contradictory. A previous study demonstrates that tumor cell competition for cystine compromises the function of CD8+ T cells within the TME. Cystine deprivation triggers glutamate accumulation, which potentiates CD36-mediated LPO (90). However, ferroptotic cells can activate antitumor immunity by expressing 1-steaoryl-2-15-HpETE-sn-glycero-3-PE. This serves as a signal that mediates phagocytosis by engaging Toll-like receptor 2 on macrophages (91) and enhancing the activities of natural killer cells (92).
Immune cells exhibit differential susceptibility to ferroptosis within the TME, and their functional heterogeneity enables distinct populations to either potentiate or suppress ferroptosis in malignant cells (80). CD8+ T cells exhibit an increased sensitivity to GPX4 inhibition compared with malignant cells, resulting in a premature impairment of antitumor immunity prior to notable tumor cell death (93). However, these effector lymphocytes demonstrate relative resistance to system Xc− inhibitors, such as sulfasalazine, as inhibition of system Xc− did not affect T cell proliferation and antitumor immune responses (94). Activated CD8+ T cells with high expression levels of fatty acid transporter CD36 increases the risk of LPO and ferroptosis (95). Compared with tumor-derived CD8+ T cells, tumor-infiltrating regulatory T cells (Tregs) typically have reduced levels of LPO, suggesting that they are less prone to ferroptosis in the TME (93). However, system Xc− inhibitors rarely impair the viability of Tregs (96). Tumor-associated macrophages (TAMs) are one of the most abundant types of immune cells in the TME and include the proinflammatory antitumor M1 and anti-inflammatory protumor M2 types (97,98). The sensitivity of these two types of cells to ferroptosis is different due to the high levels of inducible nitric oxide synthase and nitric oxide-free radicals in M1 type compared with M2 type TAMs (99). Another group of immune-suppressive cells, namely myeloid-derived suppressor cells (MDSCs), exhibit notable resistance to ferroptosis, which is associated with the upregulation of system Xc− and neutral ceramidase N-acylsphingosine amidohydrolase expression levels (100).
Metabolic reprogramming is a defining hallmark of malignancy, triggering cancer metastasis, aggressiveness and therapeutic resistance (101). Remodeled metabolic environments and immunosuppressive environments enable cancer cells to evade RCD (78). This metabolic reprogramming extends beyond canonical adaptations such as the Warburg effect (in which preferential glycolysis reduces mitochondrial ROS generation), and includes the suppression of pyruvate dehydrogenase, limiting the acetyl-CoA flux for PUFA biosynthesis, which is critical for ferroptosis (102-104). Within the TME, MDSCs increase immunosuppression through IL-6/JAK2/STAT3-mediated SLC7A11 upregulation, which depletes the extracellular cystine pools (105). This metabolic competition induces a GSH deprivation-induced ferroptosis in CD8+ T cells, further inactivating antitumor immunity (90).
To resist ferroptosis, cancer cells use lipid metabolic reprogramming as a defense strategy via two primary mechanisms: i) PL membrane restructuring via the activation of the sterol regulatory element-binding protein-1-SCD1 axis, replacing oxidation-vulnerable PUFAs with MUFAs (106); and ii) mevalonate pathway-derived antioxidant production, such as isopentenyl pyrophosphate from cholesterol metabolism, which neutralizes lipid peroxides (107,108). Iron homeostasis is also inhibited through TFR1-mediated uptake and ferritin heavy chain (FTH)-dependent storage, which reduces the levels of cytotoxic free Fe2+ (109,110). Furthermore, gut microbiota-derived metabolites, such as short-chain fatty acids (such as butyrate) and tryptophan metabolites (such as indole-acrylic acid), also affect the TME and influence the resistance of cancer cells to ferroptosis (111,112).
Epigenetic regulation is a heritable mechanism that regulates the expression of genes through chemical modifications without altering the DNA sequence. It primarily involves DNA methylation, histone modifications, non-coding RNA (ncRNA)-mediated regulation and RNA methylation (113).
Several key genes are dynamically controlled by DNA methylation. For example, in normal tissues, the core enzyme GPX4, which is essential for eliminating lipid peroxides, has notably reduced expression levels due to transcriptional repression caused by the hypermethylation of CpG islands within its promoter region compared with cancer tissues. This suppression increases cellular susceptibility to ferroptosis (114-117). By contrast, the promoter of the system Xc− light chain subunit SLC7A11 is frequently hypomethylated, leading to its elevated expression levels (118,119). This enhances cystine uptake and GSH synthesis, which inhibits ferroptosis. Furthermore, hypermethylation of the FSP1 promoter results in a reduction to its expression levels, which sensitizes cells to ferroptosis (120,121).
RNA methylation, the most abundant post-transcriptional modification in eukaryotic mRNA, also contributes to ferroptosis regulation. AlkB homolog 5 regulates glutamate-cysteine ligase modifier subunit (GCLM) mRNA levels though N6-methyladenosine (m6A) modification of GCLM and YTH m6A RNA binding protein-mediated decay of GCLM, which regulates ferroptosis (122). Additionally, methyltransferase-like protein 16 (METTL16) serves a role in ferroptosis and tumorigenesis by catalyzing the m6A modification of Sentrin/small ubiquitin-like modifier-specific protease 3 mRNA and regulating the stability of lactotransferrin (123).
Histone modifications serve an important role in shaping chromatin structure. It exerts precise transcriptional control over ferroptosis-associated genes, such as SLC7A11 and ACSL3, and impacts gene expression and cancer development (124-126). p53 promotes the nuclear translocation of a histone H2B monoubiquitylation (H2Bub1) deubiquitinase, ubiquitin-specific protease 7 (USP7), which negatively regulates H2Bub1 levels. The interaction between p53 and USP7 reduces the occupancy of H2Bub1 at the regulatory region of SLC7A11, leading to transcriptional repression of SLC7A11and enhancing the ferroptosis induction sensitivity of cells (125). Histone acetylation and methylation also demonstrate bidirectional regulatory effects on ferroptosis (126-129).
ncRNAs regulate the expression of ferroptosis-associated genes via post-transcriptional regulation, forming protein-RNA interaction networks. Specifically, microRNA (miR)-137/miR-9 and miR-15a-5p promote ferroptosis by targeting SLC7A11 and GPX4 mRNA, respectively (130,131), whereas miR-27a and miR-4717 exert anti-ferroptosis effects by targeting ACSL4 and NCOA4 (132,133). Long ncRNAs (lncRNAs) also participate in ferroptosis regulation by acting as protein decoys, chromatin modifiers or miRNA sponges (134,135).
Ferroptosis induction has emerged as a novel strategy to overcome tumor therapy resistance (35,36). However, tumor cells establish complex ferroptosis defense networks by remodeling immune-metabolic environments, notably elevating the threshold for ferroptosis initiation (78). The development of this resistance limits the clinical application of ferroptosis-inducing therapies (78). However, a series of innovative strategies have been developed, providing a theoretical foundation for designing effective treatments to circumvent resistance. Recently identified, reversible palmitoylation modification of GPX4 is a key regulator of ferroptosis. The palmitoylation inhibitor 2-bromopalmitate notably enhances the antitumor efficacy of ferroptosis inducers by inhibiting GPX4 palmitoylation (136). Furthermore, targeted protein degradation represents an emerging therapeutic paradigm. The GPX4-targeted autophagy-targeting chimera (GPX4-AUTAC), based on selective autophagic degradation, induces ferroptosis and demonstrates antitumor activity in vitro, in vivo and in patient-derived organoids (137). In addition, proteolysis-targeting chimeras that target GPX4/DHODH have also been designed (138,139). The use of combination strategies that target immune checkpoints alongside ferroptosis inducers are also gaining attention (140). Furthermore, the development of novel nanodrug delivery systems offers additional approaches to overcome chemoresistance, such as high-density lipoprotein nanoparticles with dual metabolic disruption (selenium deprivation and LPO) (141) and antibody-targeted nanoplatforms (such as Fe-MOF@Erastin@ Herceptin), which deliver ferroptosis inducers (such as erastin) specifically to tumor cells (142).
Ferroptosis is demonstrated to have therapeutic potential in various types of cancer, including hepatocellular carcinoma, lung carcinoma, lymphoma, pancreatic ductal carcinoma and renal cell carcinoma (30-34). BTC is an uncommon type of gastrointestinal malignancy that has a poor prognosis. In the early stages of disease, surgical resection with negative margins can effectively treat BTC (8,9). For unresectable and metastatic disease, gemcitabine and cisplatin chemotherapies plus immunotherapy (such as with Durvalumab or Pembrolizumab) are the only preferred regimens approved by the National Comprehensive Cancer Network (143,144). With the increasing number of patients with BTC, investigating novel targets and alternative options is crucial. Ferroptosis, which is considered to be one of the most promising potential antitumor methods, can affect the occurrence and development of BTC by regulating intracellular iron and ROS levels, providing new treatment options for patients with BTC (70,124,145). The following section primarily concentrates on the advancements in ferroptosis-associated research in BTC, including potential therapeutic targets, treatment options and associated mechanisms (Table I; Fig. 3).
TP53 is acknowledged as a classic tumor suppressor gene, which serves important roles in controlling the cell cycle, cell proliferation and cell death (146,147). TP53 serves a role in controlling the susceptibility to ferroptosis via both transcription-dependent and transcription-independent mechanisms (148). Shank-associated RH domain interacting protein (SHARPIN) is a crucial part of the complex responsible for activating the linear ubiquitin chain, which inhibits ferroptosis through the p53/SLC7A11/GPX4 signaling pathway and promotes cell proliferation in CCA. Silencing the SHARPIN gene results in the suppression of p53 ubiquitination and degradation, as well as a decreased expression of SLC7A11, GPX4, superoxide dismutase (SOD)-1 and SOD-2. Targeting SHARPIN may serve as a potential strategy benefiting CCA treatment (149). Human hydroxysteroid dehydrogenase-like 2 (HSDL2) is also a regulator of cancer progression and lipid metabolism. Knocking down HSDL2 promotes CCA progression by inhibiting ferroptosis through the p53/SLC7A11 axis (150). As a member of Runt-domain family, Runt-related transcription factor 3 (RUNX3) has a reduced expression level in GBC. This downregulation, mediated by promoter DNA hypermethylation, is notably associated with adverse outcomes in patients with GBC. RUNX3 activates the transcription of inhibitor of growth protein 1, which leads to the suppression of SLC7A11 through a p53-dependent pathway and the induction of ferroptosis (151). Golgi phosphoprotein 3 also promotes CCA malignancy by inhibiting ferroptosis through SLC7A11 upregulation (152). Additionally, TP53-induced glycolysis and apoptosis regulator (TIGAR) is associated with adverse outcomes and ferroptosis resistance in patients with intrahepatic CCA (iCCA). TIGAR encodes an enzyme responsible for regulating glycolysis and scavenging ROS. Knockdown of TIGAR reduced the expression of GPX4, a key inhibitor of ferroptosis (153). Furthermore, combining TIGAR knockdown with cisplatin treatment synergistically induces a notable increase in ferroptosis (153).
The transcription factor Nrf2 controls the expression of genes involved in counteracting oxidative and electrophilic stresses, which serves a role in regulating the cellular antioxidant response (154,155). A study by Huang et al (156) reports that transcription factor activating enhancer-binding protein 2 α (TFAP2A) may serve a role as a regulator of ferroptosis in GBC via the Nrf2 signaling pathway. GBC cells exhibit elevated levels of TFAP2A, compared with non-tumorigenic human intrahepatic bile duct cells (H69), and knocking down TFAP2A lead to a decrease in GBC cell proliferation, migration and invasion, as well as a decreased expression of oxidative stress-associated genes, such as heme oxygenase 1 and Nrf2. A study by Zheng et al (157) demonstrates high methyltransferase-like 3 expression levels in cisplatin-resistant iCCA cells compared with parental cells. This upregulation enhances m6A modifications, leading to the inhibition of ferroptosis and a resistance to cisplatin through the stabilization of Nrf2 mRNA and an increase in the Nrf2 protein expression levels. Additionally, overexpression of METTL16 in patients with CCA is associated with poor prognosis. Mechanistically, METTL16 increases the m6A modifications of activating transcription factor 4 (ATF4) mRNA, which increases the expression of ATF4 and subsequently results in the suppression of ferroptosis (158).
At present, GPX4 is recognized as the only enzyme with the ability to directly reduce complex phospholipid hydroperoxide (159). Therefore, GPX4, which is responsible for the efficient removal of phospholipid hydroperoxides, is critical for cell survival (160). When GPX4 fails to effectively remove PLOOHs, there is an increase in LPO and ferroptosis (57,161). GPX4 is also one of the important adverse prognostic indicators in iCCA (162). A study by Lei et al (163) demonstrates that JUND enhances the transcription of linc00976, which promotes the development of CCA and prevents ferroptosis by regulating the miR-3202/GPX4 axis. F-box protein 31 (FBXO31) stimulates the ubiquitination process of GPX4 and consequently promotes the degradation of the GPX4 proteasome, which enhances the occurrence of ferroptosis. Additionally, FBXO31-upregulated cancer stem cell-like cells present enhanced sensitivity to cisplatin compared with control cells (164). In addition, lncRNA paired box 8-antisense RNA 1 binds to p62 and activates Nrf2, which promotes GPX4 transcription and stabilizes GPX4 mRNA by interacting with insulin-like growth factor 2 mRNA binding protein 3. This inhibits ferroptosis and promotes resistance to gemcitabine and cisplatin. Treatment with JKE-1674 (1 μM), a GPX4 inhibitor, combined with gemcitabine (5 μM) and cisplatin (10 μM) exhibits an improved antitumor potential in preclinical models of both subcutaneous tumor and orthotopic mice models, compared with gemcitabine and cisplatin therapy (165).
Previous studies reveal ACSL4 promotes ferroptosis by catalyzing the esterification of long-chain PUFAs into PUFA-CoA, which serve as substrates for LPO (166,167). Data from the Gene Expression Omnibus and The Cancer Genome Atlas databases demonstrates that there is a higher level of ACSL4 in CCA compared with normal adjacent tissues. Additionally, ACSL4 levels are associated with the prognosis of patients with CCA as well as the immune infiltration in CCA (168). Targeting the ACSL4 pathway may be an anti-cancer approach. A study by Liao et al (169) reveals that, via ACSL4-dependent lipid reprogramming, IFNγ from cytotoxic T lymphocytes and AA from the TME induce tumor cell ferroptosis. Furthermore, cancer cells that derive from migration exhibit a higher ferroptosis sensitivity and PUFA lipid contents compared with primary-tumor-derived cells. This supports the possibility of targeting ferroptosis in the treatment of cancer (170). Specific and targeted inhibitors of ACSL4 with anti-ferroptosis function such as AS-252424 have been screened and identified (171). In GBC, sirtuin 3 has tumor suppressive effects through the induction of the expression of ACSL4 and AKT-dependent ferroptosis, and inhibition of the epithelial-mesenchymal transition (172). Another member of the acyl-CoA synthetase long chain family, ACSL3, synthesizes MUFAs that suppress PUFAs peroxidation, which confers ferroptosis protection. Furthermore, ACSL3 is an unfavorable prognostic biomarker in patients with CCA and a mediator of ferroptosis resistance in CCA cells. Therefore, ACSL3 may be a potential therapeutic target (173).
Furthermore, several other factors potentially regulate ferroptosis sensitivity. For example, circular (circ)forkhead box protein P1 and Homo sapiens_circ_0050900 affect ferroptosis by interacting with OTU domain-containing protein 4 and regulating SLC3A2, respectively (174,175). TFR modulates the ferroptosis sensitivity of cells by regulating intracellular iron levels. Additionally, TFR is also an adverse prognostic factor in patients with iCCA (176). The regulatory importance of the E26 transformation-specific variant 4-ALY/REF export factor-pyruvate kinase M2 and Aldo-keto reductase family 1 member C1-cytochrome P450 family 1 subfamily B member 1-cAMP axes also warrant attention (177,178). Additionally, gut microbiotas inhibit ferroptosis in iCCA by altering glutamine metabolism through the regulation of the activin receptor-like kinase 5/NADPH oxidase 1 axis (179).
Ferroptosis serves a role in the pathogenesis, progression and treatment resistance of BTC. Furthermore, accumulating evidence demonstrates that ferroptosis-associated indicators are notably associated with patient prognosis and represent potential prognostic biomarkers (Table II). Techniques such as RNA sequencing and immunohistochemistry reveal elevated expression of ferroptosis-associated markers, including ACSL3/4, SLC7A11 and GPX4, in BTC tumor tissues compared with normal adjacent tissues, which is notably associated with poorer clinical outcomes (168,173,176,180-183). By contrast, isocitrate dehydrogenase 1 (IDH1) mutations are associated with a more favorable prognosis in BTC (183,184). In addition, other key ferroptosis-associated markers, such as LPCAT3 and FSP1, are also associated with prognosis in various solid tumors including ovarian cancer and lung adenocarcinoma; however, their clinical validation in BTC requires further investigation (185-192). Taken together, these ferroptosis-associated indicators may serve as novel prognostic markers for BTC, potentially offering a new perspective for precise prognosis assessment and individualized treatment strategies. Furthermore, integrating these indicators to establish multi-parameter prognostic models may enhance the accuracy of existing BTC prognostic assessment systems.
In addition to conventional GPX4 inhibitors and system Xc− inhibitors, numerous other compounds have the potential to induce and notably contribute to the ferroptosis process in BTC. A number of these compounds can function independently, while others require co-administration with traditional chemotherapies, inducers and technologies.
Erastin, an inducer of ferroptosis, specifically inhibits system Xc− (18). A study by Su et al (193) demonstrates that IDH1 mutations suppress CCA progression through erastin-induced ferroptosis. Erastin-treated IDH1 mutation cells have increased numbers of propidium iodide-positive cells, increased levels of lipid ROS and decreased cell viability compared with that in erastin-treated wild type cells. However, IDH1 mutation-targeted inhibitors, such as ivosidenib (AG-120), demonstrates notable clinical efficacy in CCA and are, at present, incorporated into second-line therapy. Therefore, the feasibility of combining ferroptosis inducers with IDH1-targeted agents requires further investigation (12,194). Furthermore, erastin and lenvatinib combined with photodynamic therapy (PDT) can increase ROS levels and inhibit antioxidant systems intracellularly, thereby enhancing the induction of ferroptosis in CCA cells (195).
Traditional Chinese medicine (TCM) is currently recognized as a viable complementary treatment in malignancies (196). Accumulating evidence demonstrates that TCM can notably enhance the chemosensitivity of a patient, potentiate the antitumor efficacy of therapeutic agents and mitigate treatment-associated adverse effects in cancer management (197,198). Quercetin (QE), a flavonoid in flowers, stems and leaves of various plants, possesses the ability to impede the progression of breast and colon cancer by interrupting the cell cycle (199,200). Previous studies demonstrate that QE induces ferroptosis in iCCA cells by inhibiting the NF-κB signaling pathway, which inhibits iCCA cell invasion (201,202). Isoliquiritigenin (ISL), also known as 2',4',4-trihydroxychalcone, is also a type of flavonoid and is extracted from the root of the liquorice plant. ISL induces apoptosis and inhibits proliferation in tumors (203,204). A study by Wang et al (205) reveals that ISL triggers ferroptosis in GBC by activating the p62-Kelch-like ECH-associated protein 1-Nrf2-haem oxygenase-1 (HMOX1) signaling pathway and reducing the expression of GPX4. Silencing HMOX1 or increasing GPX4 expression levels decreases the susceptibility of GBC cells to ISL-induced ferroptosis and enhances the survival of GBC cells (205). Another naturally occurring triterpenoid compound named liquidambaric acid (LCD) also exhibits notable therapeutic potential. LCD can bind to and inhibit signal transducing adaptor molecule binding protein like 1, which stabilizes Nrf2 through de-ubiquitination, and notably enhances CCA cell proliferation and migration while impeding ferroptosis (206). Wu-Mei-Wan, a long-utilized TCM formula, demonstrates potential efficacy as a second-line GBC therapy. It enhances gemcitabine chemosensitivity and induces ferroptosis through phosphorylated (p)-STAT3-mediated transcriptional regulation of ferroptosis-associated targets, downregulating GPX4, HIF-1α and FTH1, while upregulating ACSL4 (207).
At present, chemoimmunotherapy is the first-line treatment option for BTC due to the durvalumab plus gemcitabine and cisplatin in advanced BTC and KEYNOTE-966 studies (143,144). A study by Zhu et al (208) reveals that, in AKT-hyperactivated iCCA, the p-AKT-p-phosphoenolpyruvate carboxykinase 1 (PCK1)-p-lactate dehydrogenase A-SPRINGlac axis is a driver of ferroptosis resistance. This combines p-PCK1-mediated glycolytic activation and mevalonate flux reprogramming. However, simvastatin treatment effectively reverses this resistance, underscoring its therapeutic potential for improving the chemoimmunotherapy efficacy through ferroptosis sensitivity (208).
In addition, metabolic products such as lithocholic acid (LCA) are reagents involved in inhibiting tumorigenesis. A study by Li et al (209) reveals that LCA induces ferroptosis in GBC by suppressing glutaminase-mediated glutamine metabolism, which may be a tumor-suppressive mechanism with therapeutic potential.
PDT is a treatment modality that selectively destroys tumor tissue through the light activation of photosensitizers and release of ROS (210). The tumor-selective destruction capacity of PDT minimizes damage to healthy tissues, which suggests it may be a potential antitumor therapeutic strategy (211,212). The first-generation photosensitizer-HiPorfin-mediated PDT promotes the apoptosis and ferroptosis of CCA cells through the activation of the p53/SLC7A11/GPX4 signaling pathway (213). Furthermore, the second-generation photosensitizer-Hypericin-mediated PDT induces ferroptosis in CCA by inhibiting the AKT/mTORC1/GPX4 signaling pathway (214). These findings provide possible insights into individualized precision therapy for CCA. Furthermore, combination strategies represent novel therapeutic paradigms for BTC. Surufatinib (SUR), a multi-targeted kinase inhibitor, has comparable clinical efficacy in patients with advanced CCA compared with regorafenib, which is one of the recommend regimens used for subsequent-line therapy of BTC (12,215). Mechanistically, both SUR and PDT induce ferroptosis through the upregulation of ACSL4 and suppression of GPX4, and their effect is synergistically enhanced during combination treatment (215). In addition, an integrated nanotherapeutic platform known as CMArg@ Lip has been successfully developed for combined PDT and gas therapy. This platform encapsulates the photosensitizer, NO gas-generating agent and Nrf2 inhibitor with ROS-responsive liposomes, which enables it to effectively address the challenges of tumor hypoxia and the antioxidant microenvironment (216).
Ferroptosis, an iron-dependent, LPO-driven form of RCD, is a research focus in BTC therapeutics. Accumulating evidence demonstrates marked vulnerability of BTC cells to ferroptosis, unveiling novel avenues for targeted treatment. Despite promising advances, challenges persist. While core ferroptosis pathways, such as the GPX4-GSH axis and system Xc− regulation, are delineated, the integrated regulatory network, particularly crosstalk with metabolic reprogramming and TME interactions, is yet to be fully elucidated. At present, clinically applicable biomarkers for predicting ferroptosis sensitivity are lacking, which hampers patient stratification. Furthermore, current ferroptosis inducers, such as erastin analogs and RSL3 derivatives, have limitations in tumor specificity, systemic toxicity and pharmacokinetic profiles.
Future studies should prioritize the following: i) Systematic investigations of ferroptosis synergism with immune checkpoint inhibitors and molecular-targeted agents, using multiomics approaches to decipher resistance mechanisms; ii) development of integrated biomarker panels incorporating genetic, epigenetic and microenvironmental features for precision patient stratification; iii) therapeutic validation through physiologically relevant platforms, including patient derived organoids, orthotopic BTC models and humanized mouse systems simulating tumor-stroma crosstalk; and iv) engineering of tumor-targeted nano-formulations, such as the CMArg@Lip platform, to enhance the tumor-targeted delivery of ferroptosis inducers while mitigating off-target effects.
In summary, bridging mechanistic insights from laboratory studies with rationally designed clinical trials incorporating biomarker-driven enrollment may highlight ferroptosis modulation as a potnetial therapeutic paradigm for BTC.
Not applicable.
RZ and YD contributed to literature searching and screening, manuscript drafting and revision of the manuscript. SY and HH contributed to the revision of the manuscript. FLi and FLiu contributed to the study design and revision of the manuscript. Data authentication is not applicable. All authors read and approved the final version of the manuscript.
Not applicable.
Not applicable.
The authors declare that they have no competing interests.
Not applicable.
The present study was supported by 1.3.5 project for disciplines of excellence (West China Hospital, Sichuan University; grant no. ZYJC21046), 1.3.5 project for disciplines of excellence-Clinical Research Incubation Project (West China Hospital, Sichuan University; grant no. 2021HXFH001), China Telecom Sichuan Company Biliary Tract Tumor Big Data Platform and Application Phase I R&D Project (grant no. 312230752), National Natural Science Foundation of China for Young Scientists Fund (grant no. 82303669), Sichuan University-Sui Ning School-local Cooperation project (grant no. 2022CDSN-18), and The Post-doctor Research Fund of West China Hospital, Sichuan University (grant nos. ZYJC21046, 2021HXFH001 and 2024HXBH083).
|
Valle JW, Kelley RK, Nervi B, Oh DY and Zhu AX: Biliary tract cancer. Lancet. 397:428–444. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Brindley PJ, Bachini M, Ilyas SI, Khan SA, Loukas A, Sirica AE, The BT, Wongkham S and Gores GJ: Cholangiocarcinoma. Nat Rev Dis Primers. 7:652021. View Article : Google Scholar : PubMed/NCBI | |
|
Siegel RL, Miller KD, Wagle NS and Jemal A: Cancer statistics, 2023. CA Cancer J Clin. 73:17–48. 2023.PubMed/NCBI | |
|
Torre LA, Siegel RL, Islami F, Bray F and Jemal A: Worldwide burden of and trends in mortality from gallbladder and other biliary tract cancers. Clin Gastroenterol Hepatol. 16:427–437. 2018. View Article : Google Scholar | |
|
Nagtegaal ID, Odze RD, Klimstra D, Paradis V, Rugge M, Schirmacher P, Washington KM, Carneiro F and Cree IA; WHO classification of tumours editorial board: The 2019 WHO classification of tumours of the digestive system. Histopathology. 76:182–188. 2020. View Article : Google Scholar : | |
|
Nakanuma Y and Kakuda Y: Pathologic classification of cholangiocarcinoma: New concepts. Best Pract Res Clin Gastroenterol. 29:277–293. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Roa JC, García P, Kapoor VK, Maithel SK, Javle M and Koshiol J: Gallbladder cancer. Nat Rev Dis Primers. 8:692022. View Article : Google Scholar : PubMed/NCBI | |
|
Vogel A, Bridgewater J, Edeline J, Kelley RK, Klümpen HJ, Malka D, Primrose JN, Rimassa L, Stenzinger A, Valle JW, et al: Biliary tract cancer: ESMO Clinical Practice Guideline for diagnosis, treatment and follow-up. Ann Oncol. 34:127–140. 2023. View Article : Google Scholar | |
|
Ruff SM, Cloyd JM and Pawlik TM: Annals of surgical oncology practice guidelines series: Management of primary liver and biliary tract cancers. Ann Surg Oncol. 30:7935–7949. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Baria K, De Toni EN, Yu B, Jiang Z, Kabadi SM and Malvezzi M: Worldwide incidence and mortality of biliary tract cancer. Gastro Hep Adv. 1:618–626. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Lamarca A, Edeline J and Goyal L: How I treat biliary tract cancer. ESMO Open. 7:1003782022. View Article : Google Scholar : PubMed/NCBI | |
|
Benson AB, D'Angelica MI, Abrams T, Abbott DE, Ahmed A, Anaya DA, Anders R, Are C, Bachini M, Binder D, et al: NCCN Guidelines® insights: Biliary tract cancers, version 2.2023. J Natl Compr Canc Netw. 21:694–704. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Harding JJ, Khalil DN, Fabris L and Abou-Alfa GK: Rational development of combination therapies for biliary tract cancers. J Hepatol. 78:217–228. 2023. View Article : Google Scholar | |
|
Palmieri LJ, Lavolé J, Dermine S, Brezault C, Dhooge M, Barré A, Chaussade S and Coriat R: The choice for the optimal therapy in advanced biliary tract cancers: Chemotherapy, targeted therapies or immunotherapy. Pharmacol Ther. 210:1075172020. View Article : Google Scholar : PubMed/NCBI | |
|
Lamarca A, Barriuso J, McNamara MG and Valle JW: Molecular targeted therapies: Ready for 'prime time' in biliary tract cancer. J Hepatol. 73:170–185. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Galluzzi L, Vitale I, Aaronson SA, Abrams JM, Adam D, Agostinis P, Alnemri ES, Altucci L, Amelio I, Andrews DW, et al: Molecular mechanisms of cell death: Recommendations of the nomenclature committee on cell death 2018. Cell Death Differ. 25:486–541. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Koren E and Fuchs Y: Modes of regulated cell death in cancer. Cancer Discov. 11:245–265. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Dixon SJ, Lemberg KM, Lamprecht MR, Skouta R, Zaitsev EM, Gleason CE, Patel DN, Bauer AJ, Cantley AM, Yang WS, et al: Ferroptosis: An Iron-dependent form of nonapoptotic cell death. Cell. 149:1060–1072. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Jiang X, Stockwell BR and Conrad M: Ferroptosis: Mechanisms, biology and role in disease. Nat Rev Mol Cell Biol. 22:266–282. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Weinlich R, Oberst A, Beere HM and Green DR: Necroptosis in development, inflammation and disease. Nat Rev Mol Cell Biol. 18:127–136. 2017. View Article : Google Scholar | |
|
Wyllie AH: Glucocorticoid-induced thymocyte apoptosis is associated with endogenous endonuclease activation. Nature. 284:555–556. 1980. View Article : Google Scholar : PubMed/NCBI | |
|
Bergsbaken T and Cookson BT: Macrophage activation redirects Yersinia-infected host cell death from apoptosis to caspase-1-dependent pyroptosis. PLoS Pathog. 3:e1612007. View Article : Google Scholar : PubMed/NCBI | |
|
Mizushima N and Komatsu M: Autophagy: Renovation of cells and tissues. Cell. 147:728–741. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Tang D, Kang R, Berghe TV, Vandenabeele P and Kroemer G: The molecular machinery of regulated cell death. Cell Res. 29:347–364. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Stockwell BR, Friedmann Angeli JP, Bayir H, Bush AI, Conrad M, Dixon SJ, Fulda S, Gascón S, Hatzios SK, Kagan VE, et al: Ferroptosis: A regulated cell death nexus linking metabolism, redox biology, and disease. Cell. 171:273–285. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Dixon SJ and Stockwell BR: The role of iron and reactive oxygen species in cell death. Nat Chem Biol. 10:9–17. 2014. View Article : Google Scholar | |
|
Yang WS and Stockwell BR: Ferroptosis: Death by lipid peroxidation. Trends Cell Biol. 26:165–176. 2016. View Article : Google Scholar : | |
|
Liang D, Minikes AM and Jiang X: Ferroptosis at the intersection of lipid metabolism and cellular signaling. Mol Cell. 82:2215–2227. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Chen X, Kang R, Kroemer G and Tang D: Broadening horizons: The role of ferroptosis in cancer. Nat Rev Clin Oncol. 18:280–296. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Gao GB, Chen L, Pan JF, Lei T, Cai X, Hao Z, Wang Q, Shan G and Li J: LncRNA RGMB-AS1 inhibits HMOX1 ubiquitination and NAA10 activation to induce ferroptosis in non-small cell lung cancer. Cancer Lett. 590:2168262024. View Article : Google Scholar : PubMed/NCBI | |
|
Sun X, Ou Z, Chen R, Niu X, Chen D, Kang R and Tang D: Activation of the p62-Keap1-NRF2 pathway protects against ferroptosis in hepatocellular carcinoma cells. Hepatology. 63:173–184. 2016. View Article : Google Scholar | |
|
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 | |
|
Ding Y, Chen X, Liu C, Ge W, Wang Q, Hao X, Wang M, Chen Y and Zhang Q: Identification of a small molecule as inducer of ferroptosis and apoptosis through ubiquitination of GPX4 in triple negative breast cancer cells. J Hematol Oncol. 14:192021. View Article : Google Scholar | |
|
Zou Y, Palte MJ, Deik AA, Li H, Eaton JK, Wang W, Tseng YY, Deasy R, Kost-Alimova M, Dančík V, et al: A GPX4-dependent cancer cell state underlies the clear-cell morphology and confers sensitivity to ferroptosis. Nat Commun. 10:16172019. View Article : Google Scholar : PubMed/NCBI | |
|
Shen Z, Song J, Yung BC, Zhou Z, Wu A and Chen X: Emerging strategies of cancer therapy based on ferroptosis. Adv Mater. 30:e17040072018. View Article : Google Scholar : PubMed/NCBI | |
|
Friedmann Angeli JP, Krysko DV and Conrad M: Ferroptosis at the crossroads of cancer-acquired drug resistance and immune evasion. Nat Rev Cancer. 19:405–414. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Hassannia B, Vandenabeele P and Vanden Berghe T: Targeting ferroptosis to iron out cancer. Cancer Cell. 35:830–849. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Angeli JPF, Shah R, Pratt DA and Conrad M: Ferroptosis inhibition: Mechanisms and opportunities. Trends Pharmacol Sci. 38:489–498. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Mou Y, Wang J, Wu J, He D, Zhang C, Duan C and Li B: Ferroptosis, a new form of cell death: Opportunities and challenges in cancer. J Hematol Oncol. 12:342019. View Article : Google Scholar : PubMed/NCBI | |
|
Tang D, Chen X, Kang R and Kroemer G: Ferroptosis: Molecular mechanisms and health implications. Cell Res. 31:107–125. 2021. View Article : Google Scholar : | |
|
Chen X, Li J, Kang R, Klionsky DJ and Tang D: Ferroptosis: Machinery and regulation. Autophagy. 17:2054–2081. 2021. View Article : Google Scholar : | |
|
Zeng F, Nijiati S, Tang L, Ye J, Zhou Z and Chen X: Ferroptosis detection: From approaches to applications. Angew Chem Int Ed Engl. 62:e2023003792023. View Article : Google Scholar : PubMed/NCBI | |
|
Frey PA and Reed GH: The ubiquity of iron. ACS Chem Biol. 7:1477–1481. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Hentze MW, Muckenthaler MU and Andrews NC: Balancing acts: Molecular control of mammalian iron metabolism. Cell. 117:285–297. 2004. View Article : Google Scholar : PubMed/NCBI | |
|
Gonciarz RL, Collisson EA and Renslo AR: Ferrous Iron-dependent pharmacology. Trends Pharmacol Sci. 42:7–18. 2021. View Article : Google Scholar | |
|
Gao M, Monian P, Pan Q, Zhang W, Xiang J and Jiang X: Ferroptosis is an autophagic cell death process. Cell Res. 26:1021–1032. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Geng N, Shi BJ, Li SL, Zhong ZY, Li YC, Xua WL, Zhou H and Cai JH: Knockdown of ferroportin accelerates erastin-induced ferroptosis in neuroblastoma cells. Eur Rev Med Pharmacol Sci. 22:3826–3836. 2018.PubMed/NCBI | |
|
Wang Y, Liu Y, Liu J, Kang R and Tang D: NEDD4L-mediated LTF protein degradation limits ferroptosis. Biochem Biophys Res Commun. 531:581–587. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Alvarez SW, Sviderskiy VO, Terzi EM, Papagiannakopoulos T, Moreira AL, Adams S, Sabatini DM, Birsoy K and Possemato R: NFS1 undergoes positive selection in lung tumours and protects cells from ferroptosis. Nature. 551:639–643. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Du J, Wang T, Li Y, Zhou Y, Wang X, Yu X, Ren X, An Y, Wu Y, Sun W, et al: DHA inhibits proliferation and induces ferroptosis of leukemia cells through autophagy dependent degradation of ferritin. Free Radic Biol Med. 131:356–369. 2019. View Article : Google Scholar | |
|
Knutson MD: Non-transferrin-bound iron transporters. Free Radic Biol Med. 133:101–111. 2019. View Article : Google Scholar | |
|
Song N, Zhang J, Zhai J, Hong J, Yuan C and Liang M: Ferritin: A multifunctional nanoplatform for biological detection, imaging diagnosis, and drug delivery. Acc Chem Res. 54:3313–3325. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Hou W, Xie Y, Song X, Sun X, Lotze MT, Zeh HJ III, Kang R and Tang D: Autophagy promotes ferroptosis by degradation of ferritin. Autophagy. 12:1425–1428. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Mancias JD, Wang X, Gygi SP, Harper JW and Kimmelman AC: Quantitative proteomics identifies NCOA4 as the cargo receptor mediating ferritinophagy. Nature. 509:105–109. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Yang WS, Kim KJ, Gaschler MM, Patel M, Shchepinov MS and Stockwell BR: Peroxidation of polyunsaturated fatty acids by lipoxygenases drives ferroptosis. Proc Natl Acad Sci USA. 113:E4966–E4975. 2016. 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 | |
|
Conrad M and Pratt DA: The chemical basis of ferroptosis. Nat Chem Biol. 15:1137–1147. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Kagan VE, Mao G, Qu F, Angeli JP, Doll S, Croix CS, Dar HH, Liu B, Tyurin VA, Ritov VB, et al: Oxidized arachidonic and adrenic PEs navigate cells to ferroptosis. Nat Chem Biol. 13:81–90. 2017. View Article : Google Scholar | |
|
Magtanong L, Ko PJ, To M, Cao JY, Forcina GC, Tarangelo A, Ward CC, Cho K, Patti GJ, Nomura DK, et al: Exogenous monounsaturated fatty acids promote a Ferroptosis-resistant cell state. Cell Chem Biol. 26:420–432.e9. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Tesfay L, Paul BT, Konstorum A, Deng Z, Cox AO, Lee J, Furdui CM, Hegde P, Torti FM and Torti SV: Stearoyl-CoA desaturase 1 protects ovarian cancer cells from ferroptotic cell death. Cancer Res. 79:5355–5366. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Chu B, Kon N, Chen D, Li T, Liu T, Jiang L, Song S, Tavana O and Gu W: ALOX12 is required for p53-mediated tumour suppression through a distinct ferroptosis pathway. Nat Cell Biol. 21:579–591. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Yang WS, SriRamaratnam R, Welsch ME, Shimada K, Skouta R, Viswanathan VS, Cheah JH, Clemons PA, Shamji AF, Clish CB, et al: Regulation of ferroptotic cancer cell death by GPX4. Cell. 156:317–331. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Ingold I, Berndt C, Schmitt S, Doll S, Poschmann G, Buday K, Roveri A, Peng X, Porto Freitas F, Seibt T, et al: Selenium utilization by GPX4 is required to prevent hydroperoxide-induced ferroptosis. Cell. 172:409–422.e421. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Ursini F and Maiorino M: Lipid peroxidation and ferroptosis: The role of GSH and GPx4. Free Radic Biol Med. 152:175–185. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Dai C, Chen X, Li J, Comish P, Kang R and Tang D: Transcription factors in ferroptotic cell death. Cancer Gene Ther. 27:645–656. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Doll S, Freitas FP, Shah R, Aldrovandi M, da Silva MC, Ingold I, Goya Grocin A, Xavier da Silva TN, Panzilius E, Scheel CH, et al: FSP1 is a glutathione-independent ferroptosis suppressor. Nature. 575:693–698. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Bersuker K, Hendricks JM, Li Z, Magtanong L, Ford B, Tang PH, Roberts MA, Tong B, Maimone TJ, Zoncu R, et al: The CoQ oxidoreductase FSP1 acts parallel to GPX4 to inhibit ferroptosis. Nature. 575:688–692. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Kraft VAN, Bezjian CT, Pfeiffer S, Ringelstetter L, Müller C, Zandkarimi F, Merl-Pham J, Bao X, Anastasov N, Kössl J, et al: GTP Cyclohydrolase 1/tetrahydrobiopterin counteract ferroptosis through lipid remodeling. ACS Cent Sci. 6:41–53. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Soula M, Weber RA, Zilka O, Alwaseem H, La K, Yen F, Molina H, Garcia-Bermudez J, Pratt DA, Birsoy K, et al: Metabolic determinants of cancer cell sensitivity to canonical ferroptosis inducers. Nat Chem Biol. 16:1351–1360. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Mao C, Liu X, Zhang Y, Lei G, Yan Y, Lee H, Koppula P, Wu S, Zhuang L, Fang B, et al: DHODH-mediated ferroptosis defence is a targetable vulnerability in cancer. Nature. 593:586–590. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Liang D, Feng Y, Zandkarimi F, Wang H, Zhang Z, Kim J, Cai Y, Gu W, Stockwell BR and Jiang X: Ferroptosis surveillance independent of GPX4 and differentially regulated by sex hormones. Cell. 186:2748–2764.e2722. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Lei G, Zhuang L and Gan B: The roles of ferroptosis in cancer: Tumor suppression, tumor microenvironment, and therapeutic interventions. Cancer Cell. 42:513–534. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
de Visser KE and Joyce JA: The evolving tumor microenvironment: From cancer initiation to metastatic outgrowth. Cancer Cell. 41:374–403. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Binnewies M, Roberts EW, Kersten K, Chan V, Fearon DF, Merad M, Coussens LM, Gabrilovich DI, Ostrand-Rosenberg S, Hedrick CC, et al: Understanding the tumor immune microenvironment (TIME) for effective therapy. Nat Med. 24:541–550. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Quail DF and Joyce JA: Microenvironmental regulation of tumor progression and metastasis. Nat Med. 19:1423–1437. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Elhanani O, Ben-Uri R and Keren L: Spatial profiling technologies illuminate the tumor microenvironment. Cancer Cell. 41:404–420. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Cui K, Wang K and Huang Z: Ferroptosis and the tumor microenvironment. J Exp Clin Cancer Res. 43:3152024. View Article : Google Scholar : PubMed/NCBI | |
|
Bhowmick S, Banerjee S, Shridhar V and Mondal S: Reprogrammed immuno-metabolic environment of cancer: The driving force of ferroptosis resistance. Mol Cancer. 24:1612025. View Article : Google Scholar : PubMed/NCBI | |
|
Kim R, Taylor D, Vonderheide RH and Gabrilovich DI: Ferroptosis of immune cells in the tumor microenvironment. Trends Pharmacol Sci. 44:542–552. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Zheng Y, Sun L, Guo J and Ma J: The crosstalk between ferroptosis and anti-tumor immunity in the tumor microenvironment: Molecular mechanisms and therapeutic controversy. Cancer Commun (Lond). 43:1071–1096. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Demuynck R, Efimova I, Naessens F and Krysko DV: Immunogenic ferroptosis and where to find it? J Immunother Cancer. 9:e0034302021. View Article : Google Scholar : PubMed/NCBI | |
|
Fucikova J, Kepp O, Kasikova L, Petroni G, Yamazaki T, Liu P, Zhao L, Spisek R, Kroemer G and Galluzzi L: Detection of immunogenic cell death and its relevance for cancer therapy. Cell Death Dis. 11:10132020. View Article : Google Scholar : PubMed/NCBI | |
|
Wen Q, Liu J, Kang R, Zhou B and Tang D: The release and activity of HMGB1 in ferroptosis. Biochem Biophys Res Commun. 510:278–283. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao YY, Lian JX, Lan Z, Zou KL, Wang WM and Yu GT: Ferroptosis promotes anti-tumor immune response by inducing immunogenic exposure in HNSCC. Oral Dis. 29:933–941. 2023. View Article : Google Scholar | |
|
Han W, Duan X, Ni K, Li Y, Chan C and Lin W: Co-delivery of dihydroartemisinin and pyropheophorbide-iron elicits ferroptosis to potentiate cancer immunotherapy. Biomaterials. 280:1213152022. View Article : Google Scholar : | |
|
Wan C, Sun Y, Tian Y, Lu L, Dai X, Meng J, Huang J, He Q, Wu B, Zhang Z, et al: Irradiated tumor cell-derived microparticles mediate tumor eradication via cell killing and immune reprogramming. Sci Adv. 6:eaay97892020. View Article : Google Scholar : PubMed/NCBI | |
|
Efimova I, Catanzaro E, Van der Meeren L, Turubanova VD, Hammad H, Mishchenko TA, Vedunova MV, Fimognari C, Bachert C, Coppieters F, et al: Vaccination with early ferroptotic cancer cells induces efficient antitumor immunity. J Immunother Cancer. 8:e0013692020. View Article : Google Scholar : PubMed/NCBI | |
|
Liu J, Zhu S, Zeng L, Li J, Klionsky DJ, Kroemer G, Jiang J, Tang D and Kang R: DCN released from ferroptotic cells ignites AGER-dependent immune responses. Autophagy. 18:2036–2049. 2022. View Article : Google Scholar : | |
|
Wiernicki B, Maschalidi S, Pinney J, Adjemian S, Vanden Berghe T, Ravichandran KS and Vandenabeele P: Cancer cells dying from ferroptosis impede dendritic Cell-mediated Anti-tumor immunity. Nat Commun. 13:36762022. View Article : Google Scholar : PubMed/NCBI | |
|
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 : | |
|
Luo X, Gong HB, Gao HY, Wu YP, Sun WY, Li ZQ, Wang G, Liu B, Liang L, Kurihara H, et al: Oxygenated phosphatidylethanolamine navigates phagocytosis of ferroptotic cells by interacting with TLR2. Cell Death Differ. 28:1971–1989. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Kim KS, Choi B, Choi H, Ko MJ and Kim DH and Kim DH: Enhanced natural killer cell anti-tumor activity with nanoparticles mediated ferroptosis and potential therapeutic application in prostate cancer. J Nanobiotechnology. 20:4282022. View Article : Google Scholar : PubMed/NCBI | |
|
Drijvers JM, Gillis JE, Muijlwijk T, Nguyen TH, Gaudiano EF, Harris IS, LaFleur MW, Ringel AE, Yao CH, Kurmi K, et al: Pharmacologic screening identifies metabolic vulnerabilities of CD8+ T cells. Cancer Immunol Res. 9:184–199. 2021. View Article : Google Scholar : | |
|
Arensman MD, Yang XS, Leahy DM, Toral-Barza L, Mileski M, Rosfjord EC, Wang F, Deng S, Myers JS, Abraham RT and Eng CH: Cystine-glutamate antiporter xCT deficiency suppresses tumor growth while preserving antitumor immunity. Proc Natl Acad Sci USA. 116:9533–9542. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Xu S, Chaudhary O, Rodríguez-Morales P, Sun X, Chen D, Zappasodi R, Xu Z, Pinto AFM, Williams A, Schulze I, et al: Uptake of oxidized lipids by the scavenger receptor CD36 promotes lipid peroxidation and dysfunction in CD8+ T cells in tumors. Immunity. 54:1561–1577.e7. 2021. View Article : Google Scholar | |
|
Xu C, Sun S, Johnson T, Qi R, Zhang S, Zhang J and Yang K: The glutathione peroxidase Gpx4 prevents lipid peroxidation and ferroptosis to sustain Treg cell activation and suppression of antitumor immunity. Cell Rep. 35:1092352021. View Article : Google Scholar : PubMed/NCBI | |
|
Sica A and Mantovani A: Macrophage plasticity and polarization: In vivo veritas. J Clin Invest. 122:787–795. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Biswas SK and Mantovani A: Macrophage plasticity and interaction with lymphocyte subsets: Cancer as a paradigm. Nat Immunol. 11:889–896. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Kapralov AA, Yang Q, Dar HH, Tyurina YY, Anthonymuthu TS, Kim R, St Croix CM, Mikulska-Ruminska K, Liu B, Shrivastava IH, et al: Redox lipid reprogramming commands susceptibility of macrophages and microglia to ferroptotic death. Nat Chem Biol. 16:278–290. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Zhu H, Klement JD, Lu C, Redd PS, Yang D, Smith AD, Poschel DB, Zou J, Liu D, Wang PG, et al: Asah2 represses the p53-Hmox1 axis to protect Myeloid-derived suppressor cells from ferroptosis. J Immunol. 206:1395–1404. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Hanahan D and Weinberg RA: Hallmarks of cancer: The next generation. Cell. 144:646–674. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Ždralević M, Vučetić M, Daher B, Marchiq I, Parks SK and Pouysségur J: Disrupting the 'Warburg effect' re-routes cancer cells to OXPHOS offering a vulnerability point via 'ferroptosis'-induced cell death. Adv Biol Regul. 68:55–63. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Amos A, Amos A, Wu L and Xia H: The Warburg effect modulates DHODH role in ferroptosis: A review. Cell Commun Signal. 21:1002023. View Article : Google Scholar : PubMed/NCBI | |
|
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 | |
|
Dai Y, Cui C, Jiao D and Zhu X: JAK/STAT signaling as a key regulator of ferroptosis: Mechanisms and therapeutic potentials in cancer and diseases. Cancer Cell Int. 25:832025. View Article : Google Scholar : PubMed/NCBI | |
|
Yi J, Zhu J, Wu J, Thompson CB and Jiang X: Oncogenic activation of PI3K-AKT-mTOR signaling suppresses ferroptosis via SREBP-mediated lipogenesis. Proc Natl Acad Sci USA. 117:31189–31197. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Garcia-Bermudez J, Baudrier L, Bayraktar EC, Shen Y, La K, Guarecuco R, Yucel B, Fiore D, Tavora B, Freinkman E, et al: Squalene accumulation in cholesterol auxotrophic lymphomas prevents oxidative cell death. Nature. 567:118–122. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Warner GJ, Berry MJ, Moustafa ME, Carlson BA, Hatfield DL and Faust JR: Inhibition of selenoprotein synthesis by selenocysteine tRNA[Ser]Sec lacking isopentenyladenosine. J Biol Chem. 275:28110–28119. 2000. View Article : Google Scholar : PubMed/NCBI | |
|
Qu L, He X, Tang Q, Fan X, Liu J and Lin A: Iron metabolism, ferroptosis, and lncRNA in cancer: Knowns and unknowns. J Zhejiang Univ Sci B. 23:844–862. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang S, Xin W, Anderson GJ, Li R, Gao L, Chen S, Zhao J and Liu S: Double-edge sword roles of iron in driving energy production versus instigating ferroptosis. Cell Death Dis. 13:402022. View Article : Google Scholar : PubMed/NCBI | |
|
He Y, Ling Y, Zhang Z, Mertens RT, Cao Q, Xu X, Guo K, Shi Q, Zhang X, Huo L, et al: Butyrate reverses ferroptosis resistance in colorectal cancer by inducing c-Fos-dependent xCT suppression. Redox Biol. 65:1028222023. View Article : Google Scholar : PubMed/NCBI | |
|
Cui W, Guo M, Liu D, Xiao P, Yang C, Huang H, Liang C, Yang Y, Fu X, Zhang Y, et al: Gut microbial metabolite facilitates colorectal cancer development via ferroptosis inhibition. Nat Cell Biol. 26:124–137. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Cavalli G and Heard E: Advances in epigenetics link genetics to the environment and disease. Nature. 571:489–499. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang X, Sui S, Wang L, Li H, Zhang L, Xu S and Zheng X: Inhibition of tumor propellant glutathione peroxidase 4 induces ferroptosis in cancer cells and enhances anticancer effect of cisplatin. J Cell Physiol. 235:3425–3437. 2020. View Article : Google Scholar | |
|
Zhang X, Huang Z, Xie Z, Chen Y, Zheng Z, Wei X, Huang B, Shan Z, Liu J, Fan S, et al: Homocysteine induces oxidative stress and ferroptosis of nucleus pulposus via enhancing methylation of GPX4. Free Radic Biol Med. 160:552–565. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Kim JW, Min DW, Kim D, Kim J, Kim MJ, Lim H and Lee JY: GPX4 overexpressed non-small cell lung cancer cells are sensitive to RSL3-induced ferroptosis. Sci Rep. 13:88722023. View Article : Google Scholar : PubMed/NCBI | |
|
Lee J, You JH, Kim MS and Roh JL: Epigenetic reprogramming of epithelial-mesenchymal transition promotes ferroptosis of head and neck cancer. Redox Biol. 37:1016972020. View Article : Google Scholar : PubMed/NCBI | |
|
Koppula P, Zhuang L and Gan B: Cystine transporter SLC7A11/xCT in cancer: Ferroptosis, nutrient dependency, and cancer therapy. Protein Cell. 12:599–620. 2021. View Article : Google Scholar : | |
|
Liu J, Xia X and Huang P: xCT: A critical molecule that links cancer metabolism to redox signaling. Mol Ther. 28:2358–2366. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Pontel LB, Bueno-Costa A, Morellato AE, Carvalho Santos J, Roué G and Esteller M: Acute lymphoblastic leukemia necessitates GSH-dependent ferroptosis defenses to overcome FSP1-epigenetic silencing. Redox Biol. 55:1024082022. View Article : Google Scholar : PubMed/NCBI | |
|
Wang X, Kong X, Feng X and Jiang DS: Effects of DNA, RNA, and protein methylation on the regulation of ferroptosis. Int J Biol Sci. 19:3558–3575. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Lv D, Zhong C, Dixit D, Yang K, Wu Q, Godugu B, Prager BC, Zhao G, Wang X, Xie Q, et al: EGFR promotes ALKBH5 nuclear retention to attenuate N6-methyladenosine and protect against ferroptosis in glioblastoma. Mol Cell. 83:4334–4351.e4337. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Wang J, Xiu M, Wang J, Gao Y and Li Y: METTL16-SENP3-LTF axis confers ferroptosis resistance and facilitates tumorigenesis in hepatocellular carcinoma. J Hematol Oncol. 17:782024. 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 | |
|
Wang Y, Yang L, Zhang X, Cui W, Liu Y, Sun QR, He Q, Zhao S, Zhang GA, Wang Y and Chen S: Epigenetic regulation of ferroptosis by H2B monoubiquitination and p53. EMBO Rep. 20:e475632019. View Article : Google Scholar : PubMed/NCBI | |
|
Ma M, Kong P, Huang Y, Wang J, Liu X, Hu Y, Chen X, Du C and Yang H: Activation of MAT2A-ACSL3 pathway protects cells from ferroptosis in gastric cancer. Free Radic Biol Med. 181:288–299. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Li H, Liu W, Zhang X, Wu F, Sun D and Wang Z: Ketamine suppresses proliferation and induces ferroptosis and apoptosis of breast cancer cells by targeting KAT5/GPX4 axis. Biochem Biophys Res Commun. 585:111–116. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang X, Du L, Qiao Y, Zhang X, Zheng W, Wu Q, Chen Y, Zhu G, Liu Y, Bian Z, et al: Ferroptosis is governed by differential regulation of transcription in liver cancer. Redox Biol. 24:1012112019. View Article : Google Scholar : PubMed/NCBI | |
|
Zille M, Kumar A, Kundu N, Bourassa MW, Wong VSC, Willis D, Karuppagounder SS and Ratan RR: Ferroptosis in neurons and cancer cells is similar but differentially regulated by histone deacetylase inhibitors. eNeuro. 6:ENEURO.0263-18.20192019. View Article : Google Scholar : PubMed/NCBI | |
|
Yadav P, Sharma P, Sundaram S, Venkatraman G, Bera AK and Karunagaran D: SLC7A11/xCT is a target of miR-5096 and its restoration partially rescues miR-5096-mediated ferroptosis and anti-tumor effects in human breast cancer cells. Cancer Lett. 522:211–224. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Deng SH, Wu DM, Li L, Liu T, Zhang T, Li J, Yu Y, He M, Zhao YY, Han R and Xu Y: miR-324-3p reverses cisplatin resistance by inducing GPX4-mediated ferroptosis in lung adenocarcinoma cell line A549. Biochem Biophys Res Commun. 549:54–60. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Bao C, Zhang J, Xian SY and Chen F: MicroRNA-670-3p suppresses ferroptosis of human glioblastoma cells through targeting ACSL4. Free Radic Res. 55:853–864. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Ma LL, Liang L, Zhou D and Wang SW: Tumor suppressor miR-424-5p abrogates ferroptosis in ovarian cancer through targeting ACSL4. Neoplasma. 68:165–173. 2021. View Article : Google Scholar | |
|
Qi W, Li Z, Xia L, Dai J, Zhang Q, Wu C and Xu S: LncRNA GABPB1-AS1 and GABPB1 regulate oxidative stress during erastin-induced ferroptosis in HepG2 hepatocellular carcinoma cells. Sci Rep. 9:161852019. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang Y, Guo S, Wang S, Li X, Hou D, Li H, Wang L, Xu Y, Ma B, Wang H and Jiang X: LncRNA OIP5-AS1 inhibits ferroptosis in prostate cancer with long-term cadmium exposure through miR-128-3p/SLC7A11 signaling. Ecotoxicol Environ Saf. 220:1123762021. View Article : Google Scholar : PubMed/NCBI | |
|
Huang B, Wang H, Liu S, Hao M, Luo D, Zhou Y, Huang Y, Nian Y, Zhang L, Chu B and Yin C: Palmitoylation-dependent regulation of GPX4 suppresses ferroptosis. Nat Commun. 16:8672025. View Article : Google Scholar : PubMed/NCBI | |
|
Gong R, Wan X, Jiang S, Guan Y, Li Y, Jiang T, Chen Z, Zhong C, He L, Xiang Z, et al: GPX4-AUTAC induces ferroptosis in breast cancer by promoting the selective autophagic degradation of GPX4 mediated by TRAF6-p62. Cell Death Differ. May 20–2025. View Article : Google Scholar : Epub ahead of print. | |
|
Yao L, Yang N, Zhou W, Akhtar MH, Zhou W, Liu C, Song S, Li Y, Han W and Yu C: Exploiting cancer vulnerabilities by blocking of the DHODH and GPX4 pathways: A multifunctional Bodipy/PROTAC nanoplatform for the efficient synergistic ferroptosis therapy. Adv Healthc Mater. 12:e23008712023. View Article : Google Scholar : PubMed/NCBI | |
|
Dong J, Ma F, Cai M, Cao F, Li H, Liang H, Li Y, Ding G, Li J, Cheng X and Qin JJ: Heat shock protein 90 interactome-mediated proteolysis targeting chimera (HIM-PROTAC) degrading glutathione peroxidase 4 to trigger ferroptosis. J Med Chem. 67:16712–16736. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Wang S, Zhu L, Li T, Lin X, Zheng Y, Xu D, Guo Y, Zhang Z, Fu Y, Wang H, et al: Disruption of MerTK increases the efficacy of checkpoint inhibitor by enhancing ferroptosis and immune response in hepatocellular carcinoma. Cell Rep Med. 5:1014152024. View Article : Google Scholar : PubMed/NCBI | |
|
Lamperis SM, McMahon KM, Calvert AE, Rink JS, Vasan K, Pandkar MR, Crentsil EU, Chalmers ZR, McDonald NR, Kosmala CJ, et al: CRISPR screen reveals a simultaneous targeted mechanism to reduce cancer cell selenium and increase lipid oxidation to induce ferroptosis. Proc Natl Acad Sci USA. 122:e25028761222025. View Article : Google Scholar : PubMed/NCBI | |
|
Gao J, Ye T, Miao H, Liu M, Wen L, Tian Y, Fu Z, Sun L, Wang L and Wang Y: Antibody-functionalized iron-based nanoplatform for ferroptosis-augmented targeted therapy of HER2-positive breast cancer. Bioact Mater. 52:702–718. 2025.PubMed/NCBI | |
|
Burris HA III, Okusaka T, Vogel A, Lee MA, Takahashi H, Breder V, Blanc JF, Li J, Bachini M, Żotkiewicz M, et al: Durvalumab plus gemcitabine and cisplatin in advanced biliary tract cancer (TOPAZ-1): Patient-reported outcomes from a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 25:626–635. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Kelley RK, Ueno M, Yoo C, Finn RS, Furuse J, Ren Z, Yau T, Klümpen HJ, Chan SL, Ozaka M, et al: Pembrolizumab in combination with gemcitabine and cisplatin compared with gemcitabine and cisplatin alone for patients with advanced biliary tract cancer (KEYNOTE-966): A randomised, double-blind, placebo-controlled, phase 3 trial. Lancet. 401:1853–1865. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang C, Liu X, Jin S, Chen Y and Guo R: Ferroptosis in cancer therapy: A novel approach to reversing drug resistance. Mol Cancer. 21:472022. View Article : Google Scholar : PubMed/NCBI | |
|
Cheok CF, Verma CS, Baselga J and Lane DP: Translating p53 into the clinic. Nat Rev Clin Oncol. 8:25–37. 2011. View Article : Google Scholar | |
|
Bykov VJN, Eriksson SE, Bianchi J and Wiman KG: Targeting mutant p53 for efficient cancer therapy. Nat Rev Cancer. 18:89–102. 2018. View Article : Google Scholar | |
|
Jiang L, Kon N, Li T, Wang SJ, Su T, Hibshoosh H, Baer R and Gu W: Ferroptosis as a p53-mediated activity during tumour suppression. Nature. 520:57–62. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Zeng C, Lin J, Zhang K, Ou H, Shen K, Liu Q, Wei Z, Dong X, Zeng X, Zeng L, et al: SHARPIN promotes cell proliferation of cholangiocarcinoma and inhibits ferroptosis via p53/SLC7A11/GPX4 signaling. Cancer Sci. 113:3766–3775. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Ma S, Ma Y, Qi F, Lei J, Chen F, Sun W, Wang D, Zhou S, Liu Z, Lu Z, et al: HSDL2 knockdown promotes the progression of cholangiocarcinoma by inhibiting ferroptosis through the P53/SLC7A11 axis. World J Surg Oncol. 21:2932023. View Article : Google Scholar : PubMed/NCBI | |
|
Cai C, Zhu Y, Mu J, Liu S, Yang Z, Wu Z, Zhao C, Song X, Ye Y, Gu J, et al: DNA methylation of RUNX3 promotes the progression of gallbladder cancer through repressing SLC7A11-mediated ferroptosis. Cell Signal. 108:1107102023. View Article : Google Scholar : PubMed/NCBI | |
|
Yin Z, Liu Q, Gao Y, Wang R, Qi Y, Wang D, Chen L, Yin X, He M and Li W: GOLPH3 promotes tumor malignancy via inhibition of ferroptosis by upregulating SLC7A11 in cholangiocarcinoma. Mol Carcinog. 63:912–925. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Toshida K, Itoh S, Iseda N, Izumi T, Yoshiya S, Toshima T, Ninomiya M, Iwasaki T, Oda Y and Yoshizumi T: Impact of TP53-induced glycolysis and apoptosis regulator on malignant activity and resistance to ferroptosis in intrahepatic cholangiocarcinoma. Cancer science. 115:170–183. 2024. View Article : Google Scholar | |
|
Dodson M, Castro-Portuguez R and Zhang DD: NRF2 plays a critical role in mitigating lipid peroxidation and ferroptosis. Redox Biol. 23:1011072019. View Article : Google Scholar : PubMed/NCBI | |
|
Kerins MJ and Ooi A: The roles of NRF2 in modulating cellular iron homeostasis. Antioxid Redox Signal. 29:1756–1773. 2018. View Article : Google Scholar : | |
|
Huang HX, Yang G, Yang Y, Yan J, Tang XY and Pan Q: TFAP2A is a novel regulator that modulates ferroptosis in gallbladder carcinoma cells via the Nrf2 signalling axis. Eur Rev Med Pharmacol Sci. 24:4745–4755. 2020.PubMed/NCBI | |
|
Zheng X, Li H, Lin J, Li P, Yang X, Luo Z and Jin L: METTL3-mediated m6A modification promotes chemoresistance of intrahepatic cholangiocarcinoma by up-regulating NRF2 to inhibit ferroptosis in cisplatin-resistant cells. J Chemother. 37:596–606. 2025. View Article : Google Scholar | |
|
Zhao S, Cao J, Liang R, Peng T, Wu S, Liu Z, Wu Y, Song L, Sun C, Liu Y, et al: METTL16 suppresses ferroptosis in cholangiocarcinoma by promoting ATF4 via m6A modification. Int J Biol Sci. 21:189–203. 2025. View Article : Google Scholar : | |
|
Ursini F, Maiorino M, Valente M, Ferri L and Gregolin C: Purification from pig liver of a protein which protects liposomes and biomembranes from peroxidative degradation and exhibits glutathione peroxidase activity on phosphatidylcholine hydroperoxides. Biochim Biophys Acta. 710:197–211. 1982. View Article : Google Scholar : PubMed/NCBI | |
|
Yant LJ, Ran Q, Rao L, Van Remmen H, Shibatani T, Belter JG, Motta L, Richardson A and Prolla TA: The selenoprotein GPX4 is essential for mouse development and protects from radiation and oxidative damage insults. Free Radic Biol Med. 34:496–502. 2003. View Article : Google Scholar : PubMed/NCBI | |
|
Shah R, Shchepinov MS and Pratt DA: Resolving the role of lipoxygenases in the initiation and execution of ferroptosis. ACS Cent Sci. 4:387–396. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Hori Y, Yoh T, Nishino H, Okura K, Kurimoto M, Takamatsu Y, Satoh M, Nishio T, Koyama Y, Ishii T, et al: Ferroptosis-related gene glutathione peroxidase 4 promotes reprogramming of glucose metabolism via Akt-mTOR axis in intrahepatic cholangiocarcinoma. Carcinogenesis. 45:119–130. 2024. View Article : Google Scholar | |
|
Lei S, Cao W, Zeng Z, Zhang Z, Jin B, Tian Q, Wu Y, Zhang T, Li D, Hu C, et al: JUND/linc00976 promotes cholangiocarcinoma progression and metastasis, inhibits ferroptosis by regulating the miR-3202/GPX4 axis. Cell Death Dis. 13:9672022. View Article : Google Scholar : PubMed/NCBI | |
|
Zhu Z, Zheng Y, He H, Yang L, Yang J, Li M, Dai W and Huang H: FBXO31 sensitizes cancer stem cells-like cells to cisplatin by promoting ferroptosis and facilitating proteasomal degradation of GPX4 in cholangiocarcinoma. Liver Int. 42:2871–2888. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Chen ZW, Shan JJ, Chen M, Wu Z, Zhao YM, Zhu HX, Jin X, Wang YX, Wu YB, Xiang Z, et al: Targeting GPX4 to induce ferroptosis overcomes chemoresistance mediated by the PAX8-AS1/GPX4 axis in intrahepatic cholangiocarcinoma. Adv Sci (Weinh). 12:e010422025. View Article : Google Scholar : PubMed/NCBI | |
|
Doll S, Proneth B, Tyurina YY, Panzilius E, Kobayashi S, Ingold I, Irmler M, Beckers J, Aichler M, Walch A, et al: ACSL4 dictates ferroptosis sensitivity by shaping cellular lipid composition. Nat Chem Biol. 13:91–98. 2017. View Article : Google Scholar : | |
|
Gan B: ACSL4, PUFA, and ferroptosis: New arsenal in anti-tumor immunity. Signal Transduct Target Ther. 7:1282022. View Article : Google Scholar : PubMed/NCBI | |
|
Liu S, Fan S, Wang Y, Chen R, Wang Z, Zhang Y, Jiang W, Chen Y, Xu X, Yu Y, et al: ACSL4 serves as a novel prognostic biomarker correlated with immune infiltration in Cholangiocarcinoma. BMC Cancer. 23:4442023. 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 : | |
|
Wang Y, Hu M, Cao J, Wang F, Han JR, Wu TW, Li L, Yu J, Fan Y, Xie G, et al: ACSL4 and polyunsaturated lipids support metastatic extravasation and colonization. Cell. 188:412–429.e27. 2025. View Article : Google Scholar | |
|
Huang Q, Ru Y, Luo Y, Luo X, Liu D, Ma Y, Zhou X, Linghu M, Xu W, Gao F and Huang Y: Identification of a targeted ACSL4 inhibitor to treat ferroptosis-related diseases. Sci Adv. 10:eadk12002024. View Article : Google Scholar : PubMed/NCBI | |
|
Liu L, Li Y, Cao D, Qiu S, Li Y, Jiang C, Bian R, Yang Y, Li L, Li X, et al: SIRT3 inhibits gallbladder cancer by induction of AKT-dependent ferroptosis and blockade of epithelial-mesenchymal transition. Cancer Lett. 510:93–104. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Sae-Fung A, Vinayavekhin N, Fadeel B and Jitkaew S: ACSL3 is an unfavorable prognostic marker in cholangiocarcinoma patients and confers ferroptosis resistance in cholangiocarcinoma cells. NPJ Precis Oncol. 8:2842024. View Article : Google Scholar : PubMed/NCBI | |
|
Wang P, Hu Z, Yu S, Su S, Wu R, Chen C, Ye Y, Wang H, Ye X, Zhou Z, et al: A novel protein encoded by circFOXP1 enhances ferroptosis and inhibits tumor recurrence in intrahepatic cholangiocarcinoma. Cancer Lett. 598:2170922024. View Article : Google Scholar : PubMed/NCBI | |
|
Shi X, Yang J, Wang M, Xia L, Zhang L and Qiao S: Hsa_circ_0050900 affects ferroptosis in intrahepatic cholangiocarcinoma cells by targeting hsa-miR-605-3p to regulate SLC3A2. Oncol Lett. 27:22024. View Article : Google Scholar | |
|
Toshida K, Itoh S, Iseda N, Izumi T, Bekki Y, Yoshiya S, Toshima T, Iwasaki T, Oda Y and Yoshizumi T: The association of transferrin receptor with prognosis and biologic role in intrahepatic cholangiocarcinoma. Ann Surg Oncol. 31:8627–8637. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Wang X, Duan W, Ma Z, Wen H, Mao X and Liu C: ETV4/ALYREF-mediated glycolytic metabolism through PKM2 enhances resistance to ferroptosis and promotes the development of intrahepatic cholangiocarcinoma. Cancer Metab. 13:192025. View Article : Google Scholar : PubMed/NCBI | |
|
Liu C, Zhang C, Wu H, Zhao Z, Wang Z, Zhang X, Yang J, Yu W, Lian Z, Gao M and Zhou L: The AKR1C1-CYP1B1-cAMP signaling axis controls tumorigenicity and ferroptosis susceptibility of extrahepatic cholangiocarcinoma. Cell Death Differ. 32:506–520. 2025. View Article : Google Scholar | |
|
Zhang Q, Zhou J, Zhai D, Jiang Q, Yang M and Zhou M: Gut microbiota regulates the ALK5/NOX1 axis by altering glutamine metabolism to inhibit ferroptosis of intrahepatic cholangiocarcinoma cells. Biochim Biophys Acta Mol Basis Dis. 1870:1671522024. View Article : Google Scholar : PubMed/NCBI | |
|
Amontailak S, Titapun A, Jusakul A, Thanan R, Kimawaha P, Jamnongkan W, Thanee M, Sirithawat P and Techasen A: Prognostic values of Ferroptosis-related proteins ACSL4, SLC7A11, and CHAC1 in cholangiocarcinoma. Biomedicines. 12:20912024. View Article : Google Scholar : PubMed/NCBI | |
|
Saisomboon S, Kariya R, Boonnate P, Sawanyawisuth K, Cha'on U, Luvira V, Chamgramol Y, Pairojkul C, Seubwai W, Silsirivanit A, et al: Diminishing acetyl-CoA carboxylase 1 attenuates CCA migration via AMPK-NF-κB-snail axis. Biochim Biophys Acta Mol Basis Dis. 1869:1666942023. View Article : Google Scholar | |
|
Wang J, Zhang M, Zhang L, Cai H, Zhou S, Zhang J and Wang Y: Correlation of Nrf2, HO-1, and MRP3 in gallbladder cancer and their relationships to clinicopathologic features and survival. J Surg Res. 164:e99–e105. 2010. 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: IDH1(105GGT) single nucleotide polymorphism is associated with its activation and better prognosis. Front Med (Lausanne). 9:8862292022. View Article : Google Scholar : PubMed/NCBI | |
|
Rizzato M, Brignola S, Munari G, Gatti M, Dadduzio V, Borga C, Bergamo F, Pellino A, Angerilli V, Mescoli C, et al: Prognostic impact of FGFR2/3 alterations in patients with biliary tract cancers receiving systemic chemotherapy: The BITCOIN study. Eur J Cancer. 166:165–175. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Qian Y, Liang X, Kong P, Cheng Y, Cui H, Yan T, Wang J, Zhang L, Liu Y, Guo S, et al: Elevated DHODH expression promotes cell proliferation via stabilizing β-catenin in esophageal squamous cell carcinoma. Cell Death Dise. 11:8622020. View Article : Google Scholar | |
|
Hu J, Jiang Q, Mao W, Zhong S, Sun H and Mao K: STARD7 could be an immunological and prognostic biomarker: From pan-cancer analysis to hepatocellular carcinoma validation. Discov Oncol. 15:5432024. View Article : Google Scholar | |
|
Sun J, Zhou C, Ma Q, Chen W, Atyah M, Yin Y, Fu P, Liu S, Hu B, Ren N and Zhou H: High GCLC level in tumor tissues is associated with poor prognosis of hepatocellular carcinoma after curative resection. J Cancer. 10:3333–3343. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Wang M, Lu Y, Wang H, Wu Y, Xu X and Li Y: High ATF4 expression is associated with poor prognosis, amino acid metabolism, and autophagy in gastric cancer. Front Oncol. 11:7401202021. View Article : Google Scholar | |
|
Luis G, Godfroid A, Nishiumi S, Cimino J, Blacher S, Maquoi E, Wery C, Collignon A, Longuespée R, Montero-Ruiz L, et al: Tumor resistance to ferroptosis driven by Stearoyl-CoA Desaturase-1 (SCD1) in cancer cells and Fatty Acid Biding Protein-4 (FABP4) in tumor microenvironment promote tumor recurrence. Redox Biol. 43:1020062021. View Article : Google Scholar : PubMed/NCBI | |
|
Wei JL, Wu SY, Yang YS, Xiao Y, Jin X, Xu XE, Hu X, Li DQ, Jiang YZ and Shao ZM: GCH1 induces immunosuppression through metabolic reprogramming and IDO1 upregulation in triple-negative breast cancer. J Immunother Cancer. 9:e0023832021. View Article : Google Scholar : PubMed/NCBI | |
|
Wen F, Ling H, Ran R, Li X, Wang H, Liu Q, Li M and Yu T: LPCAT3 regulates the proliferation and metastasis of serous ovarian cancer by modulating arachidonic acid. Transl Oncol. 52:1022562025. View Article : Google Scholar : | |
|
Takahara H, Kanazawa T, Oshita H, Tomita Y, Hananoi Y, Ishibashi S, Ikeda M, Furukawa A, Kinoshita M, Yamamoto K, et al: GPX4 and FSP1 expression in lung adenocarcinoma: Prognostic implications and Ferroptosis-based therapeutic strategies. Cancers (Basel). 16:38882024. View Article : Google Scholar : PubMed/NCBI | |
|
Su L, Huang Y, Zheng L, Zhu Z, Wu Y and Li P: Isocitrate dehydrogenase 1 mutation in cholangiocarcinoma impairs tumor progression by sensitizing cells to ferroptosis. Open Med (Wars). 17:863–870. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Abou-Alfa GK, Macarulla T, Javle MM, Kelley RK, Lubner SJ, Adeva J, Cleary JM, Catenacci DV, Borad MJ, Bridgewater J, et al: Ivosidenib in IDH1-mutant, chemotherapy-refractory cholangiocarcinoma (ClarIDHy): A multicentre, randomised, double-blind, placebo-controlled, phase 3 study. Lancet Oncol. 21:796–807. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Dong S, An S, Liu Q, Wang X, Hu Y and Jiang A: Study on the synergistic mechanism of photodynamic therapy combined with ferroptosis inducer to induce ferroptosis in cholangiocarcinoma. Lancet Oncol. 56:845–853. 2024. | |
|
Li S, Chen X, Shi H, Yi M, Xiong B and Li T: Tailoring traditional Chinese medicine in cancer therapy. Mol Cancer. 24:272025. View Article : Google Scholar : PubMed/NCBI | |
|
Xi Z, Dai R, Ze Y, Jiang X, Liu M and Xu H: Traditional Chinese medicine in lung cancer treatment. Mol Cancer. 24:572025. View Article : Google Scholar : PubMed/NCBI | |
|
Wu J, Tang G, Cheng CS, Yeerken R, Chan YT, Fu Z, Zheng YC, Feng Y and Wang N: Traditional Chinese medicine for the treatment of cancers of hepatobiliary system: From clinical evidence to drug discovery. Mol Cancer. 23:2182024. View Article : Google Scholar : PubMed/NCBI | |
|
Kim HI, Lee SJ, Choi YJ, Kim MJ, Kim TY and Ko SG: Quercetin induces apoptosis in glioblastoma cells by suppressing Axl/IL-6/STAT3 signaling pathway. Am J Chin Med. 49:767–784. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Mohammed HA, Sulaiman GM, Anwar SS, Tawfeeq AT, Khan RA, Mohammed SAA, Al-Omar MS, Alsharidah M, Rugaie OA and Al-Amiery AA: Quercetin against MCF7 and CAL51 breast cancer cell lines: Apoptosis, gene expression and cytotoxicity of nano-quercetin. Nanomedicine (Lond). 16:1937–1961. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Song Y, Zhang Z, Chai Q, Zheng H, Qi Y, Xia G, Yu Z, Yang R, Huang J, Li Y, et al: Quercetin inhibits intrahepatic cholangiocarcinoma by inducing ferroptosis and inhibiting invasion via the NF-[Formula: See text]B pathway. Am J Chin Med. 51:701–721. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Song Y, Zhang Z, Chai Q, Zheng H, Qi Y, Xia G, Yu Z, Yang R, Huang J, Li Y, et al: ERRATUM: Quercetin inhibits intrahepatic cholangiocarcinoma by inducing ferroptosis and inhibiting invasion via the NF-[Formula: See text]B pathway. Am J Chin Med. 51:1613–1614. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Chen C, Shenoy AK, Padia R, Fang D, Jing Q, Yang P, Su SB and Huang S: Suppression of lung cancer progression by isoliquiritigenin through its metabolite 2, 4, 2', 4'-Tetrahydroxychalcone. J Exp Clin Cancer Res. 37:2432018. View Article : Google Scholar : PubMed/NCBI | |
|
Chen C, Huang S, Chen CL, Su SB and Fang DD: Isoliquiritigenin inhibits ovarian cancer metastasis by reversing Epithelial-to-mesenchymal transition. Molecules. 24:37252019. View Article : Google Scholar : PubMed/NCBI | |
|
Wang Z, Li W, Wang X, Zhu Q, Liu L, Qiu S, Zou L, Liu K, Li G, Miao H, et al: Isoliquiritigenin induces HMOX1 and GPX4-mediated ferroptosis in gallbladder cancer cells. Chin Med J (Engl). 136:2210–2220. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Wang Z, Zhang Y, Shen Y, Zhu C, Qin X and Gao Y: Liquidambaric acid inhibits cholangiocarcinoma progression by disrupting the STAMBPL1/NRF2 positive feedback loop. Phytomedicine. 136:1563032025. View Article : Google Scholar | |
|
Zhao J, Shi L, Yang Y, Zhu J, Zhou Z, Dong P, Liu S, Yang Z and Gong W: Wu-Mei-Wan promotes ferroptosis in gallbladder cancer through STAT3 negative regulation: An integrated HPLC, proteomics, network pharmacology, and experimental validation study. J Ethnopharmacol. 347:1196712025. View Article : Google Scholar : PubMed/NCBI | |
|
Zhu J, Xiong Y, Zhang Y, Liang H, Cheng K, Lu Y, Cai G, Wu Y, Fan Y, Chen X, et al: Simvastatin overcomes the pPCK1-pLDHA-SPRINGlac axis-mediated ferroptosis and chemo-immunotherapy resistance in AKT-hyperactivated intrahepatic cholangiocarcinoma. Cancer Commun (Lond). 45:1038–1071. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Li W, Wang Z, Lin R, Huang S, Miao H, Zou L, Liu K, Cui X, Wang Z, Zhang Y, et al: Lithocholic acid inhibits gallbladder cancer proliferation through interfering glutaminase-mediated glutamine metabolism. Biochem Pharmacol. 205:1152532022. View Article : Google Scholar : PubMed/NCBI | |
|
Pham TC, Nguyen VN, Choi Y, Lee S and Yoon J: Recent strategies to develop innovative photosensitizers for enhanced photodynamic therapy. Chem Rev. 121:13454–13619. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Brown SB, Brown EA and Walker I: The present and future role of photodynamic therapy in cancer treatment. Lancet Oncol. 5:497–508. 2004. View Article : Google Scholar : PubMed/NCBI | |
|
Alzeibak R, Mishchenko TA, Shilyagina NY, Balalaeva IV, Vedunova MV and Krysko DV: Targeting immunogenic cancer cell death by photodynamic therapy: Past, present and future. J Immunother Cancer. 9:e0019262021. View Article : Google Scholar : PubMed/NCBI | |
|
Yan X, Li Z, Chen H, Yang F, Tian Q and Zhang Y: Photodynamic therapy inhibits cancer progression and induces ferroptosis and apoptosis by targeting P53/GPX4/SLC7A11 signaling pathways in cholangiocarcinoma. Photodiagnosis Photodyn Ther. 47:1041042024. View Article : Google Scholar : PubMed/NCBI | |
|
An W, Zhang K, Li G, Zheng S, Cao Y and Liu J: Hypericin mediated photodynamic therapy induces ferroptosis via inhibiting the AKT/mTORC1/GPX4 axis in cholangiocarcinoma. Transl Oncol. 52:1022342025. View Article : Google Scholar : | |
|
Huang YP, Wang YX, Zhou H, Liu ZT, Zhang ZJ, Xiong L, Zou H and Wen Y: Surufatinib combined with photodynamic therapy induces ferroptosis to inhibit cholangiocarcinoma in vitro and in tumor models. Front Pharmacol. 15:12882552024. View Article : Google Scholar : PubMed/NCBI | |
|
Wang W, Gao Y, Xu J, Zou T, Yang B, Hu S, Cheng X, Xia Y and Zheng Q: A NRF2 regulated and the immunosuppressive microenvironment reversed nanoplatform for cholangiocarcinoma Photodynamic-Gas therapy. Adv Sci (Weinh). 11:e23071432024. View Article : Google Scholar : PubMed/NCBI |