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.
Acute myeloid leukemia (AML) is a highly aggressive hematologic malignancy in adults, characterized by a high relapse rate and persistent treatment resistance, which translates to a 5-year overall survival of <10% (1,2). Although advances in molecular typing and targeted therapies have improved the prognosis of patients with specific subtypes, the heterogeneity of AML, its driving mechanisms, and complex interactions between leukemia stem cells (LSCs) and the bone marrow microenvironment remain unclear (3). High-intensity chemotherapy and allogeneic hematopoietic stem cell transplantation are the current standard of care; however, due to their severe toxic side effects, harm to normal hematopoiesis, and limitations in eliminating LSCs and overcoming drug resistance, novel therapeutic approaches that are more effective and selective are urgently required (4,5).
Targeting of regulated cell death (RCD) has attracted increasing attention from researchers in recent years (6–9). The primary benefit of this method is its ability to specifically target LSCs while reshaping the immunosuppressive bone marrow microenvironment to enhance anti-leukemic efficacy and minimize damage to normal tissues (10). Studies have shown that selective modulation of RCD pathways, such as autophagy, apoptosis, pyroptosis, necroptosis, ferroptosis and cuproptosis, can effectively target and eliminate leukemic clones resistant to traditional therapies (11–16). Furthermore, these RCD pathways form a highly interconnected regulatory network that operates through dynamic interactions rather than functioning in isolation (17). This complexity suggests that combinatorial interventions targeting multiple death pathway nodes synergistically may more effectively disrupt the death-resistant barriers of leukemia cells, thereby opening multidimensional attack pathways to overcome therapeutic bottlenecks caused by AML clonal evolution and microenvironmental sheltering (18).
The present review aims to achieve three objectives: i) To assess the latest developments and challenges in targeting RCD in AML treatment by systematically integrating the understanding of six major RCD pathways; ii) to explore the crosstalk and synergistic potential between these pathways, a critical yet underexplored area; and iii) to apply these insights in developing a framework for future combinatorial therapies. To provide novel concepts and strategies for overcoming AML treatment resistance and improving patient outcomes, the present review examines the RCD modes of action, their key functions, and potential synergistic effects in maintaining LSCs, chemoresistance and bone marrow microenvironment remodeling. The present review also explores the potential for developing a customized therapeutic system through precise targeting of the death pathway network. The profiles of each type of cell death are illustrated in Fig. 1 (8,19).
Overview of autophagy. Autophagy is a highly conserved cellular process that involves the formation of autophagosomal vesicles. These vesicles encapsulate damaged organelles, misfolded proteins and other cytoplasmic components within double-membrane structures. Cellular component recycling occurs when autophagosomes fuse with lysosomes, where the enclosed contents are enzymatically degraded into smaller molecules (20).
Autophagy can be classified into three types based on its mechanism: Microautophagy, chaperone-mediated autophagy and macroautophagy. Macroautophagy is the most widely studied type of autophagy and involves the encapsulation of damaged organelles or denatured proteins by double-membrane autophagic vesicles. These vesicles then transport the contents to fuse with lysosomes, where enzymatic degradation occurs (21). Lysosomal-associated membrane protein 2 mediates the highly specific pathway of chaperone-mediated autophagy, in which heat shock protein 70 recognizes KFERQ motifs in target proteins and unidirectionally translocates them into the lysosomal lumen for selective degradation (21). Unlike the first two autophagic pathways, microautophagy does not involve the formation of distinct autophagosomal structures. Instead, it involves creation of phagocytic vesicles through lysosomal membrane invagination, directly engulfing cytoplasmic components or organelles and delivering them to the lysosomal lumen for enzymatic degradation via membrane fusion (22).
Autophagy is a five-stage molecular process involving initiation, nucleation, membrane extension, fusion and degradation (23). When cells experience nutrient deprivation, the activity of mTOR, a key energy-sensing kinase, is suppressed, triggering the autophagy signaling cascade (24). The UNC-51 like kinase 1 (ULK1) complex, which consists of essential components including ULK1, ATG13 and FIP200, is phosphorylated by AMP-activated protein kinase. This phosphorylation reverses the inhibitory effect of mTOR on the complex (25). Upon activation, the complex migrates towards the autophagy initiation site, where ULK1 catalyzes the phosphorylation of FIP200 and ATG13 (25). This phosphorylation event recruits downstream ATG proteins to initiate the formation of the pre-autophagosome complex (26). The nucleation phase of autophagy begins with the activation of the phosphatidylinositol-3-kinase class III complex, which catalyzes the formation of a lipid-signaling platform at the membrane surface through phosphatidylinositol 3-phosphate (27). This platform recruits effector proteins containing the FYVE/PX domain, which promote the assembly of pre-autophagosomal structures (28). Membrane expansion during autophagy is driven by two ubiquitin-like modification processes: Atg7 functions as an E1-like enzyme in the Atg5-Atg12 coupling system, facilitating the covalent binding of Atg12 to Atg5 (29). This complex then associates with autophagy related 16-like 1 to regulate membrane curvature and promote the growth of autophagic vesicles (29). In the LC3 lipidation pathway, Atg4 cleaves the LC3 precursor to generate LC3-I, which is then covalently linked by Atg7 to Atg3, forming membrane-anchored LC3-II that integrates into the autophagosome membrane, promoting membrane extension and serving as a marker for substrate recognition, thereby facilitating targeted transport (30). The two pathways collaborate to complete autophagosome maturation (31). Ultimately, the autophagic vesicle membrane closes, enabling fusion with the lysosome to form an autophagic lysosome. During this process, the autophagosomes are degraded, and the resulting molecules are recycled for use by the cell (32,33). Fig. 2 illustrates the three types of autophagy and their respective processes (24).
Autophagy in AML. AML development is marked by the dynamic bidirectional regulation of autophagy (34). By eliminating oxidatively damaged DNA, autophagy maintains the metabolic balance and protects the genomic integrity of hematopoietic stem cells, preventing their malignant transformation under normal conditions (35,36). Autophagy undergoes a dynamic shift during leukemia progression: In the early stages, it promotes oncogenesis by limiting genomic instability, while in the later stages, it becomes an adaptive mechanism that supports tumor cell survival and helps leukemia cells endure stressful environments through metabolic remodeling (37,38). This dichotomy suggests that autophagy dysfunction may contribute to leukemia development, with an imbalance in its regulatory network accelerating the emergence of malignant phenotypes (39).
Research has elucidated the mechanisms by which autophagy contributes to AML development. Targeting the ATF4-dependent autophagy pathway could offer a therapeutic approach for patients with Fms-like tyrosine kinase 3 (FLT3) mutations, as Fms-like tyrosine kinase 3 internal tandem duplication (FLT3-ITD) expression increases basal autophagy, which is essential for AML cell survival (40). KIT mutations increase the likelihood of AML relapse, which is directly linked to STAT3-mediated autophagy (41,42). Nucleophosmin 1 (NPM1) mutations are common in AML, where NPM1 induces autophagy through the oncogene PML, thereby promoting AML cell proliferation (43). By inhibiting autophagy, the TP53 mutation, which is less common in hematological cancers than in solid tumors, increases p53 expression and triggers apoptotic responses dependent on p53-upregulated modulator of apoptosis and BAX, leading to AML cell destruction (44). Glycolysis, a key metabolic pathway in AML, influences the disease process by promoting abnormal invasion, proliferation and drug resistance (45). Research has demonstrated that combination of autophagy inducers with glycolysis inhibitors enhanced leukemia cell sensitivity to chemotherapy (46,47). In relapsed cases, LSCs promote mitochondrial oxidative phosphorylation by stimulating fatty acid β-oxidation, creating a distinct metabolism-dependent drug resistance mechanism (48). Fatty acid metabolism is also directly linked to AML progression (49–51). The interconnections among these metabolic pathways create a complex regulatory network that supports AML cell survival and treatment resistance. Some of the aforementioned research has translated into clinical practice, with studies showing that combination of autophagy inducers with chemotherapeutic drugs enhanced their effectiveness (52–54). For instance, the mTOR inhibitor rapamycin, when combined with chemotherapy drugs, can increase AML cell sensitivity to treatment and promote apoptosis, providing a novel approach to enhance chemotherapy efficacy (55). Combining autophagy inhibitors with targeted therapies has also improved efficacy. Inhibition of autophagy enhances the anti-leukemic effect of FLT3 inhibitors in FLT3-ITD-positive AML, offering a novel strategy to overcome resistance to targeted therapies (56). Furthermore, combination of multiple autophagy regulators can more effectively control the autophagy process in AML cells. For example, co-treatment with an mTOR inhibitor and a Beclin-1 activator increases autophagy levels, promoting cell differentiation and apoptosis, suggesting the potential therapeutic benefits of using multiple regulators simultaneously (57). Further research on the mechanisms of AML autophagy, as well as on the clinical efficacy and specific applicability of related therapies, is required.
Overview of apoptosis. Apoptosis is a genetically regulated process of programmed cell death that requires ATP to execute a series of typical events (58). Endogenous and exogenous apoptosis are two distinct pathways, and both are regulated by caspases (59).
Proteins from the Bcl-2 and caspase-9 families are crucial to the endogenous, or mitochondrial, apoptotic pathway. The members are classified into pro-apoptotic proteins, which contain only the BH3 domain, and anti-apoptotic proteins, based on their function (60). Anti-apoptotic proteins form heterodimers with pro-apoptotic proteins through the BH1-3 domains, thereby inhibiting their pro-apoptotic activity (61). Bax, a pro-apoptotic protein, is upregulated and undergoes a conformational change upon DNA damage or oxidative stress, triggering apoptotic signaling (62). Bax then translocates from the cytoplasm to the outer mitochondrial membrane, where it oligomerizes to form channels (63). Meanwhile, BCL2 antagonist/killer, originally located in the outer mitochondrial membrane, is activated by BH3-only proteins, triggering a cascade that leads to oligomerization and the formation of a transmembrane pore (58,64). The two interact to increase mitochondrial outer membrane permeability, resulting in the release of cytochrome c (CytC) from the intermembrane space into the cytoplasm (65). Upon binding to apoptotic protease activating factor-1, free CytC oligomerizes into heptameric apoptotic vesicles, driven by deoxyadenosine triphosphate. The exposed caspase recruitment domain (CARD) then recruits and activates the initiator caspase-9, which subsequently cleaves the effector caspase-3, triggering apoptosis (66).
Exogenous apoptotic pathways are mediated by transmembrane death receptors (67). Upon binding of the death ligand, the death domain of the intracellular region of the receptor recruits Fas-associated death domain protein (FADD) via the adaptor protein TNFRSF1A-associated death domain, forming the death-inducing signaling complex (68). The death effector domain of FADD then recruits and activates initiator caspase-8/10, which undergoes autocleavage to form an active dimer (69). Activated caspase-8 subsequently triggers a cascade of effector protein cleavages, driving apoptosis. Caspase-3/6/7-mediated breakdown of nuclear fibrillar proteins leads to nuclear membrane disintegration and inactivation of DNA repair enzymes (70). Genome breaks are exacerbated by poly (ADP-ribose) polymerase, leading to classic apoptotic phenotypes, including cell membrane vesiculation and apoptotic vesicle formation (71,72). This method also activates the mitochondrial system, initiating an apoptosis signaling amplification loop through cleavage of the BH3-interacting-domain death agonist (BID) protein, producing the active truncated BID fragment and ensuring irreversible cell death (73). Fig. 3 illustrates the complete process of both endogenous and exogenous apoptosis mechanisms (74).
Apoptosis in AML. In AML, the anti-apoptotic protein BCL-2 is upregulated, enabling leukemia cells to evade apoptosis and continue their growth (75,76). Based on the understanding that BCL-2 interacts with pro-apoptotic proteins, scientists have developed several BCL-2 inhibitors, including oblimersen, obatoclax mesylate (GX15-070), ABT-737, ABT-263 (navitoclax), ABT-199 (venetoclax) and S6384559 (77). Venetoclax, one of the most extensively studied medications, has demonstrated strong clinical efficacy both as a monotherapy and in combination with other drugs, particularly low-dose cytarabine or demethylating agents, improving patient survival and remission rates (78–80). Venetoclax resistance has become increasingly evident as research progresses, primarily due to dysfunction in pro-apoptotic mechanisms and the upregulation of compensatory anti-apoptotic proteins, such as myeloid cell leukemia-1 (MCL-1). To overcome venetoclax resistance, appropriate inhibitors are being developed in parallel (10,81). Despite the use of venetoclax, additional apoptosis inducers are emerging. For instance, second mitochondrial-derived activator of caspases (SMAC) analogs targeting inhibitor of apoptosis protein (IAP) can antagonize IAPs and promote apoptosis by mimicking endogenous SMAC proteins (82). A pivotal clinical study has demonstrated that SMAC analogs, when combined with conventional chemotherapies or targeted therapies, can enhance the cytotoxic effects on AML cells (83).
P53, a key pro-apoptotic transcription factor, is closely associated with complex karyotypes, treatment resistance and poor survival outcomes in patients with AML. This unique clinical-molecular feature suggests that dysregulation of the TP53 signaling pathway is not only a key pathogenic mechanism of AML, but also a potential marker for molecular stratification and a target for targeted therapies, supporting the optimization of precision treatment strategies (12). Thus, drugs such as MDM2 inhibitors and P53 reactivators are being investigated for patients with TP53 mutations who have poor prognoses (84–86).
Numerous studies have demonstrated that leukemia cells can also undergo apoptosis via activation of the death receptor pathway, offering a potential basis for immunotherapy in AML (87,88). AML cell apoptosis is strongly influenced by the bone marrow microenvironment (89). Components of this microenvironment, such as the extracellular matrix, regulate the expression of apoptosis-related proteins in AML cells, affecting their sensitivity to chemotherapy (90).
Future studies on apoptosis and AML should focus on enhancing the efficacy and availability of current treatments while minimizing their side effects to provide more effective AML therapies.
Overview of pyroptosis. The gasdermin (GSDM) family includes six members (GSDMA, GSDMB, GSDMC, GSDMD, GSDME and pejvakin), which are primarily expressed in tissues such as the gastrointestinal tract and skin, and serve a key role in regulating pyroptosis (91–93). GSDMD-induced pyroptosis can be categorized into classical and non-classical pathways.
The classical pyroptosis pathway begins with pattern recognition triggered by pathogen infection and proceeds through the inflammatory vesicle-GSDMD axis. When intracellular NOD-like receptors detect pathogen-associated molecular patterns, the pyrin domain (PYD) recruits the adaptor protein apoptosis-associated speck-like protein containing a CARD (ASC). This forms a platform for multimerization and recruits pro-caspase-1, which assembles into a functional inflammatory vesicle complex (94,95). When the complex activates caspase-1, it cleaves GSDMD. This cleavage releases the C-terminal domain of GSDMD, thereby relieving its inhibition of the N-terminal pore-forming domain. The released GSDMD N-terminal oligomerizes and embeds in the cell membrane, forming a nanoscale pore that disrupts cellular osmolality, causes content leakage and triggers pyroptosis, characterized by cell swelling (96–98). The primary mechanism of the anti-infective immunity of the host involves processing the precursors of IL-1β and IL-18, which are released extracellularly through the GSDMD pore in their mature forms. This recruits neutrophils and other inflammatory cells, triggering a cascade amplification effect (94,99). This is achieved through membrane pore-mediated ‘inflammatory death’, which eliminates infected cells while tightly regulating the local inflammatory response (100).
The nonclassical pyroptosis pathway is triggered by the direct sensing of lipid components, with caspase-4/5/11 detecting intracellular lipopolysaccharide (LPS) (101–103). Unlike the traditional system, this mechanism does not involve ASC bridging proteins. Caspase-4/5/11 directly binds LPS through its CARD and undergoes oligomerization for self-activation (102–104). Activated caspase-4/5/11 specifically cleaves GSDMD to release the N-terminal pore domain (NT), leading to cell membrane perforation and pyroptosis (105). However, since NT does not directly process IL-1β/IL-18 precursors, the inflammatory response is only mildly activated (102,106). In non-immune cells, caspase-4 indirectly cleaves pro-IL-18 through conformational changes, amplifying local inflammatory signals (107) Similarly, by promoting the activation of inflammatory vesicles containing NACHT, LRR and PYD domains-containing protein 3 (NLRP3), caspase-11 enhances caspase-1-mediated IL-1β maturation and secretion, forming a synergistic network of classical and non-classical regulatory mechanisms (104).
The pathophysiological relevance of pyroptosis in infections, tumors and inflammatory diseases is underscored by the ability of certain GSDM family members (specifically GSDMD, GSDMB and GSDMC) to induce focal cell death via specific protease cleavage and link programmed death with the immune response through a multidimensional regulatory network (108–110). Both the classical and non-classical pathways of pyroptosis are illustrated in Fig. 4 (100).
Pyroptosis in AML. Research has shown that inflammasomes, key molecular complexes involved in pyroptosis, serve a dual role in the occurrence, progression and treatment of leukemia, with potential clinical applications (111). Evidence suggests that pyroptosis serves a critical, bidirectional role in both the progression of AML and its treatment response (111). Mechanistic research has demonstrated that chemotherapy or targeted therapies enhance anti-AML efficacy by activating the focal cell death pathway (18). A study by Ren et al (12) showed that estrogen receptor activation enhanced the anti-AML effect of the BCL-2 inhibitor vincristine, via the pyroptosis pathway. Demethylating agents can promote venetoclax-induced AML pyroptosis by restoring GSDME expression, offering a potential strategy to overcome resistance to BCL-2 inhibitors (14). In addition to enhancing existing targeted therapies and chemotherapy, pyroptosis could provide a novel target for AML treatment. Small molecule inhibitors targeting serine dipeptidyl peptidase (DPP)8/DPP9 have been shown to induce cellular pyroptosis in a majority of a panel of human AML cell lines and primary samples, suggesting that this target may offer a novel approach for AML treatment (112). Suppression of reticulocalbin 1 gene expression reduces bone marrow mononuclear cell activity in patients with AML, further confirming the crucial role of cellular pyroptosis in AML pathogenesis (113).
The ‘immunogenic death’ feature of pyroptosis, which releases cytokines that reshape the tumor immune environment and reduce immunosuppressive cells, underlies its significance in AML therapy by helping overcome immune escape (12). For example, receptor-interacting serine/threonine-protein kinase 3 effectively inhibits aberrant bone marrow proliferation in the FLT3-ITD mutant AML model by regulating IL-1β production via inflammatory vesicles. However, it can also modify the bone marrow microenvironment, impairing normal hematopoietic stem progenitor cell function while promoting LSC development (114–116). Cytarabine promotes IL-1β secretion through NLRP3 inflammasomes, offering a novel approach to improve treatment strategies (117). Ren et al (12) also demonstrated that targeted activation of G protein-coupled estrogen receptor improved vincristine efficacy by enhancing leukemic cell pyroptosis and CD8+ T-cell immunity in patients with AML, a process mediated by IL-1β/18.
The challenge lies in the bidirectional regulation of pyroptosis, where its intensity must be carefully controlled to prevent inflammatory storms. Despite clinical studies demonstrating the therapeutic benefit and safety of pyroptosis-inducing therapies in AML, further research is needed to optimize these treatments and understand their long-term effects on patients (12,117).
Overview of necroptosis. Necroptosis is a programmed cell death mechanism independent of cysteine proteases, typically induced when the apoptotic pathway is blocked by viruses or medications (118). Its molecular core involves the receptor interacting serine/threonine kinase 1 (RIPK1)-receptor interacting serine/threonine kinase 3 (RIPK3)-mixed lineage kinase domain like pseudokinase (MLKL) signaling axis (119). The activation pathway is complex and diverse. In the classical pathway, the TNF receptor superfamily recruits RIPK1 through its death domain when caspase-8, the main executor of apoptosis, is inhibited genetically or pharmacologically. This results in the formation of an amyloid-fibril-like signaling complex (necrosome) with RIPK3, triggering an autophosphorylation cascade of RIPK3 (118,120–122). By contrast, in pattern recognition receptor-mediated bypass, toll-like receptor 3 directly recruits RIPK3 via the TIR domain-containing adapter molecule 1 (TRIF), forming a nonclassical necrosome complex that bypasses RIPK1. This facilitates signal transduction upon recognition of viral double-stranded RNA or toll-like receptor 4 in response to endotoxin LPS (118,123). Additionally, Z-DNA binding protein 1 (ZBP1) acts as a nucleic acid sensor, detecting endogenous DNA/RNA released from viral replication intermediates or mitochondrial stress. ZBP1 specifically binds to RIPK3 through its receptor-interacting protein kinase homotypic interaction motif, initiating a necroptosis program independent of RIPK1 and TRIF (118,124). The multi-pathway signals converge, indicating that RIPK3 phosphorylates the MLKL pseudokinase domain, triggering a conformational change in MLKL and the formation of transmembrane pores. This leads to intracellular calcium overload, osmotic imbalance and loss of membrane integrity, resulting in the release of cellular contents and a potent pro-inflammatory response (118,125–129). Notably, the small molecule necrostatin-1 serves as a molecular tool for precisely modulating necrotic apoptosis subtypes by blocking RIPK1-dependent necroptosis through the ATP-binding pocket of the RIPK1 kinase domain. However, it is ineffective against ZBP1- or TRIF-mediated nonclassical pathways (130). The flexibility of this signaling pathway and multiple regulatory mechanisms make necroptosis potentially bidirectional in tumor microenvironment remodeling, chemotherapeutic resistance and immune escape, namely, it can exert both antitumor and pro-tumor effects depending on the context (131). The process of necroptosis is shown in Fig. 5 (132).
Necroptosis in AML. The RIPK1/RIPK3/MLKL axis serves a crucial role in necroptosis, with its dysregulation in AML cells closely linked to disease progression (13). The role of RIPK3 in AML is complex and context-dependent. In early leukemogenesis, RIPK3 may act as a tumor suppressor by inducing necroptosis and promoting the differentiation of leukemia-initiating cells, thereby preventing myeloid leukemia progression (13,133). However, in established AML, the role of RIPK3 is often reversed. Clinical evidence shows that high RIPK3 expression is associated with poor prognosis in specific AML subtypes, such as NPM1-mutant AML (134). Wang et al (134) specifically reported that elevated RIPK3 expression was an independent predictor of poor overall survival in specific AML subtypes, such as FAB-M4/M5, normal karyotype and NPM1 mutation subtypes, a finding strongly supported by Kaplan-Meier survival analysis (log-rank P=0.021; hazard ratio, 1.8; 95% CI, 1.2–2.5). This paradox (tumor-suppressive role in initiation but oncogenic role in progression) highlights the dual role of RIPK3 and emphasizes the need for expression-based patient stratification in future therapeutic strategies (135). A study by Zhu et al (136) identified restoring RIPK3 expression to reverse R-2-hydroxyglutarate-induced necroptosis in isocitrate dehydrogenase-mutant AML cells as a potential therapeutic strategy for patients with AML. However, Hillert et al (137) suggested that targeting the RIPK1 pathway offered a more promising therapeutic approach for patients with FLT3-ITD-mutant AML. Targeting necroptosis pathways in combination with traditional chemotherapy or targeted therapies holds potential for effective treatment (138,139). Li et al (138) found that combination of the RIPK1 inhibitor 22b with cidabenamide enhanced its antileukemic effect in FLT3-ITD-positive AML cell lines and primary samples. Combining the RIPK1 inhibitor with the BCL-2 inhibitor venetoclax promotes apoptosis in AML cells and overcomes resistance to single-drug treatment (139). A study has shown that A20 deletion in AML restored sensitivity to anthracycline by inducing necroptosis (140). Although the aforementioned studies have demonstrated that targeting the necroptosis pathway is a viable strategy in AML, the clinical relevance of the RIPK1/RIPK3/MLKL axis remains unclear (135–137). Future research should focus on developing biomarkers to predict patient response to therapies targeting the necroptosis pathway, aiding in precision medicine.
Ferroptosis
Overview of ferroptosis. Ferroptosis is a form of programmed cell death driven by iron-dependent lipid peroxidation (141). Ferroptosis results from metabolic disruption caused by the imbalance of intracellular redox homeostasis (142). At physiological levels, iron serves as a key cofactor for redox enzymes, contributing to electron transport and energy metabolism (143). Excess free iron (Fe2+) generates reactive oxygen species (ROS) through the Fenton reaction, causing lipid peroxidation of the membrane (144). This disturbs cellular membrane integrity and organelle function (145).
The primary mechanism of ferroptosis is a dynamic imbalance of lipid peroxides, regulated by the glutathione (GSH) metabolic pathway and antioxidant defenses (146–149). Polyunsaturated fatty acid phospholipid hydroperoxide (PUFA-PL-OOH) is the key effector in this process, produced through the combined action of Fe2+, lipoxygenases (arachidonate lipoxygenases), phosphatidylethanolamine-binding protein 1 complexes, cytochrome P450 reductase and ROS. These molecules collaborate to convert PUFA-PL into the harmful PUFA-PL-OOH (147). Ferroptosis suppressor protein 1 (FSP1) and GSH peroxidase 4 (GPX4) are the main mechanisms maintaining PUFA-PL-OOH at steady state. GPX4 reduces PUFA-PL-OOH via a GSH-dependent process, converting it to its non-toxic form (147). By contrast, FSP1 scavenges lipophilic free radicals and inhibits the lipid peroxidation chain reaction by producing the reduced form of coenzyme Q10 (CoQH2) (148,149). In the absence of GPX4 inhibition or FSP1-CoQH2 system dysfunction, lipid peroxides accumulate due to impaired scavenging. Furthermore, Fe2+ catalyzes the conversion of hydrogen peroxide into hydroxyl radicals via the Fenton reaction, intensifying lipid peroxidation and ROS bursts, and ultimately causing enzyme system collapse and cellular structural damage (146,147–152).
The ferroptosis regulatory network is multilayered. At the metabolic input level, the System Xc− transporter, composed of solute carrier family 7 member 11 (SLC7A11) and solute carrier family 3 member 2, mediates cystine uptake, providing essential precursors for GSH synthesis (147,153–155). Inhibitors of this system reduce intracellular GSH levels by blocking cystine uptake, impairing GPX4 function and triggering ferroptosis (156). At the enzymatic regulatory level, GPX4 activity can be directly inhibited by small molecules such as RAS-selective lethal 3 and ML162, while GSH production is blocked by buthionine sulfoximine (157). Iron homeostasis is crucial, with divalent iron either removed by chelating agents such as desferrioxamine and bipyridine or expelled via the exosomal pathway after being stored as Fe3+ in ferritin (158,159). In addition, external ROS stimulation or disruption of lipid peroxidation detoxification can disrupt the redox balance, leading to the accumulation of toxic lipid products and triggering ferroptosis (160). These multidimensional regulatory mechanisms collectively shape the complex biological effects of iron death in cell fate decisions (145). The process of ferroptosis, with its inhibitory factors and promoting factors, is illustrated in Fig. 6 (145).
Ferroptosis, an iron-dependent form of non-apoptotic cell death, offers unique potential for pathological modulation and therapeutic intervention in AML (15). AML cells are more susceptible to ferroptosis due to metabolic reprogramming and disrupted redox homeostasis (161). This susceptibility is further supported by significantly lower GPX4 expression in primary AML samples compared with normal bone marrow (P=1.3×10−6), along with reduced GSH levels and increased mitochondrial lipid peroxidation (162).
Ferroptosis holds significant potential in targeted therapy for AML. Research suggests that several natural products possess the potential to induce ferroptosis in cancer treatment (163). Crotonoside, a key constituent of Crotonus sativus listed in the Chinese Pharmacopoeia, induces ferroptosis in AML cells by stimulating lipid peroxidation and increasing autophagy, offering a novel approach for AML treatment (164). Birsen et al (165) demonstrated that APR-246-induced AML cell death was inhibited by iron chelators, lipophilic antioxidants and lipid peroxidation inhibitors, suggesting that APR-246 induced ferroptosis in AML cells. Lin et al (166) demonstrated that biomimetic high-density lipoprotein nanoparticles could disrupt intracellular cholesterol homeostasis by targeting the scavenger receptor class B type 1 receptor, which was upregulated in AML cells. This inhibited the GPX4-mediated antioxidant pathway and induced ferroptosis in AML cells at nanomolar concentrations, representing a major innovation in AML treatment.
Research indicates that M2 macrophage-derived growth differentiation factor 15 confers ferroptosis resistance in the tumor microenvironment, identifying a key drug resistance mechanism (167). AML cells are more resistant to mitoxantrone when co-cultured with M2 macrophages, providing a novel approach for overcoming AML microenvironment-mediated drug resistance (168). As a potent ferroptosis inducer and chemotherapeutic drug carrier for AML treatment, Yu et al (169) showed that the ferroptosis-inducing nanomedicine glutathione-capped ferrite nanoparticles successfully overcame AML resistance. Inhibition of nuclear factor erythroid 2-related factor 2 (NRF2) expression enhanced the anti-AML effect of vinpocetine by inducing AML cell death via the ferroptosis pathway (170). Pardieu et al (171) determined that SLC7A11 promoted AML cell survival by encoding the xCT cystine importer. The authors also demonstrated that combining daunorubicin with xCT inhibition achieved optimal anti-AML efficacy, suggesting that xCT inhibition combined with chemotherapy could be a promising strategy for AML treatment.
While ferroptosis has shown considerable promise in AML treatment, several challenges remain (172). These include the lack of techniques for monitoring ferroptosis dynamics, tumor heterogeneity-related differences in response and unclear mechanisms of microenvironment regulation. Addressing these issues is crucial for improving the long-term survival of patients with AML.
Overview of cuproptosis. Cuproptosis, a copper-dependent form of RCD, was first proposed by Tsvetkov et al (173) in 2022. The unique mechanism distinguishes cuproptosis from other death pathways, such as apoptosis, pyroptosis, necroptosis and ferroptosis. Cuproptosis is based on the direct binding of copper ions to lipoylase proteins in the tricarboxylic acid cycle, leading to abnormal aggregation (174). This copper-mediated interaction destabilizes iron-sulfur cluster proteins, disrupting key components of the mitochondrial respiratory chain and inducing protein homeostatic imbalance and oxidative stress, ultimately driving cell death via the proteotoxic stress pathway (175).
A key breakthrough in cuproptosis research was the 2019 discovery by Massachusetts Institute of Technology and Harvard teams of the copper ion carriers disulfiram and ilithromycin (ES), two small molecules that can specifically mediate copper ion transport across membranes, providing a central tool for studying copper-dependent cell death (176). As an anticancer treatment targeting mitochondrial metabolism, ES not only increases oxidative stress and ROS formation, but also triggers a novel cell death pathway through the accumulation of copper ions, according to recent findings (173). Tsvetkov et al (173) demonstrated that ES-induced cell death occurred without caspase-3 activation and was resistant to both classical apoptosis inhibitors and other death pathway blockers, confirming its independence from known death mechanisms. In-depth mechanistic studies revealed that ES penetrates the cell membrane by forming a complex with extracellular Cu2+, reducing Cu2+ to Cu+, and releasing highly reactive ROS in the mitochondria. Free ES then re-enters the circulatory system to transport Cu2+, ultimately leading to copper overloading in mitochondria through the ‘ion shuttle’ mechanism (177–179). The lethal effects of ES were reduced by electron transport chain complexes and the mitochondrial pyruvate carrier, but not by mitochondrial uncouplers. Metabolomics analyses revealed a time-dependent increase in tricarboxylic acid cycle intermediates during ES treatment, suggesting that copper toxicity primarily disrupts the tricarboxylic acid cycle rather than oxidative phosphorylation (180,181).
Genetic evidence has indicated that ferredoxin 1 (FDX1)-knockout cells were resistant to cuproptosis, confirming that loss of FDX1, a key regulator of copper toxicity signaling, prevents the lipoylated protein aggregation cascade triggered by copper accumulation. This established that FDX1-mediated metabolic disruption is a core molecular pathway for cuproptosis execution (182). FDX1-mediated toxicity in cuproptosis occurs through the specific interaction of Cu+ with the mitochondrial lipoylase system, which triggers deleterious protein aggregation (173). Cu+ binds to the fatty acylated pyruvate dehydrogenase complex, inducing abnormal oligomerization and inactivation of the enzyme, which prevents pyruvate conversion to acetyl-CoA and disrupts metabolic flow via the tricarboxylic acid cycle (173,183). FDX1 serves multiple roles in this process: It catalyzes the lipid acylation of pyruvate dehydrogenase and α-ketoglutarate dehydrogenase, and enhances copper ion toxicity by converting Cu2+ to the more toxic Cu+ (184). Lipoic acid synthase, meanwhile, facilitates lipid acylation by synthesizing lipoic acid and aids the assembly of Fe-S cluster proteins in collaboration with FDX1, together forming the molecular network underlying copper ion toxicity (184). In this process, depletion of large Fe-S clusters disrupts the electron transport chain, while the blockage of the tricarboxylic acid cycle further impairs mitochondrial energy metabolism, ultimately leading to cell death through proteotoxic stress and metabolic disruption (173). This pathway aligns closely with the previously identified ES-induced cuproptosis pathway (173). Loss of FDX1 function inhibits both copper ion-mediated lipoylated protein aggregation and metabolic reprogramming, highlighting its critical role in cuproptosis signaling (185). The entire process of the cuproptosis pathway is illustrated in Fig. 7 (174).
The copper-disulfide complex (Cu-DSF) binding system selectively targets cancer cells and cancer stem cells (186). The Cu-DSF exhibits dose-dependent cytotoxicity in LSCs, with minimal effects on normal hematopoietic progenitor cells. This selective toxicity is likely due to the enhanced copper ion enrichment in cancer cells (16). A study has demonstrated that patients with AML had reduced serum levels of zinc and selenium, accompanied by elevated copper levels, suggesting that disruptions in copper metabolism contribute to disease progression (187). Since LSCs contain higher copper levels than normal tissues, researchers have suggested using copper ion carriers to selectively transport copper ions into tumor cell mitochondria. This induces intracellular copper overload, triggering lethal effects such as oxidative stress, proteotoxicity and mitochondrial dysfunction, ultimately achieving the specific killing of tumor cells (188).
Current copper-targeted therapies for AML focus on the antitumor effects of copper chelators in hematologic malignancies (189–191). Disulfiram-copper complexes demonstrate anti-AML activity in both in vitro and in vivo assays, and partially overcome drug resistance in AML cells (192,193). Xu et al (16) demonstrated that disulfiram, either alone or in combination with copper, inhibited AML cell proliferation and induced apoptosis through mechanisms involving inactivation of the NRF2-NF-κB signaling pathway and activation of the ROS-JNK stress pathway due to copper accumulation. Furthermore, in the non-obese diabetic/severe combined immune deficiency mouse model, disulfiram also suppressed the growth of human CD34+/CD38+ AML cell xenograft tumors, further confirming its therapeutic potential in vivo. The clinical translation of copper-targeted therapies faces challenges, including off-target effects, systemic toxicity and drug resistance (194). For instance, copper chelators damage normal cells due to nonspecific binding, while long-term use of copper transporter inhibitors may lead to compensatory metabolic adaptation (195). A growing body of evidence underscores a strong association between cuproptosis and the pathogenesis of AML (196–198). This includes the dynamic expression of copper transporter proteins, the regulation of copper storage proteins, and the interplay between copper metabolism and the tumor microenvironment (188,194). Further research is needed to optimize copper carrier targeting with nanodelivery technologies and explore the synergistic effects of copper overload on cell death pathways, aiming to enhance therapeutic specificity and minimize the toxicity risk (194).
Cell death pathways are intricately interlinked, forming a complex signaling network that collectively governs cell fate (199). Autophagy also mitigates pyroptosis and its associated inflammation by removing inflammatory vesicle components, inflammatory factor precursors and damaged mitochondria (200). Autophagy serves a critical and bidirectional role in ferroptosis: Lipophagy scavenges lipid peroxides, while ferritin phagocytosis, a selective form of autophagy, releases ferric ions and drives ferroptosis (201). In the context of cuproptosis, autophagy may regulate the process by removing copper-induced aggregation of proteins or damaged mitochondria (173). Apoptosis, a conventional form of programmed cell death, is reciprocally suppressed by necroptosis: The apoptotic executor caspase-8 cleaves and inactivates RIPK1/RIPK3, key kinases involved in necroptosis (202). Necroptosis often serves as an alternative pathway when apoptosis is inhibited (203). By contrast, apoptosis and pyroptosis are linked by GSDM proteins; caspase-8 cleavage of GSDME converts apoptosis into an inflammatory form of cell death characterized by pyroptosis (203). Apoptosis and ferroptosis share common regulators, such as p53, which initiate both processes. Additionally, anti-apoptotic proteins can suppress ferroptosis, while mitochondrial damage induced by ferroptosis may also trigger apoptosis (204,205). Oxidative or endoplasmic reticulum stress caused by copper ion accumulation may indirectly activate apoptotic pathways (178). Necroptosis and pyroptosis are forms of inflammatory programmed necrosis that share the upstream signal RIPK1 (206). At the execution level, their terminal effectors, MLKL and GSDMD, can further interact through synergistic or competitive mechanisms during pore formation (207). Both necroptosis and pyroptosis share ROS triggers with ferroptosis. TNF produced by necroptosis or inflammatory factors released during pyroptosis can create a pro-oxidative environment that indirectly promotes ferroptosis (208). Additionally, damage-associated molecular patterns released by ferroptosis can activate inflammatory vesicles, triggering pyroptosis through a feedback mechanism (209). Although ferroptosis and cuproptosis have distinct core mechanisms, both involve metal ions and oxidative stress. Iron and copper metabolism are interconnected, with key regulatory nodes, such as mitochondrial function, ROS levels, inflammatory signaling and the balance of iron/copper/cystine, serving as pivotal points in the crosstalk between these death pathways (210). Understanding this complex network is crucial for explaining cell fate regulation during cancer pathogenesis and for developing precision treatments that target specific death pathways, as inhibition of one may trigger compensatory activation of another.
Although preclinical evidence strongly supports targeting multiple RCD pathways in AML, clinical translation faces challenges due to variable patient responses (18). The most successful approach to date focuses on apoptosis, where the combination of the BCL-2 inhibitor venetoclax and hypomethylating agents has improved outcomes for elderly patients with AML or those ineligible for intensive chemotherapy. This strategy has been validated in phase III trials and has received regulatory approval, providing an effective means to overcome apoptosis escape (204). Current research is focused on overcoming venetoclax resistance, particularly by targeting complementary anti-apoptotic proteins such as MCL-1 or by combining it with other targeted therapies, such as FLT3 inhibitors. Several clinical trials are actively exploring these approaches (204,211,212).
By contrast, direct modulation of other RCD pathways, such as autophagy, necroptosis, pyroptosis and ferroptosis, is still in its early stages. Current clinical evidence primarily comes from observations where these death pathways are incidentally induced by conventional chemotherapies or targeted therapies. Targeting autophagy has been particularly challenging due to its context-dependent dual role, with non-specific inhibitors such as chloroquine showing limited efficacy, highlighting the need for more precise agents (213). For example, hypomethylating agents promote GSDME-mediated pyroptosis, while the p53 reactivator APR-246 induces ferroptosis, suggesting that the partial therapeutic efficacy of these agents may stem from non-apoptotic death mechanisms (14,158). However, first-in-class drugs designed to activate these pathways, such as DPP8/9 inhibitors for pyroptosis, RIPK1 activators for necroptosis and GPX4 inhibitors for ferroptosis, have yet to enter clinical trials for AML, highlighting a translational gap and the need for further research (13,103,159).
The copper ionophore disulfiram has been used for decades with sporadic anticancer activity, while its systematic evaluation in AML is still confined to an early-stage trial, highlighting a significant translational challenge for the cuproptosis pathway (214). The novel copper ionophore electrochlorol is currently under early-stage investigation for solid tumors, and its potential to target mitochondria-rich LSCs in AML remains a hypothesis awaiting clinical validation (188).
Advancing this field requires overcoming several key challenges: The lack of reliable biomarkers for predicting treatment response and enabling precise patient stratification; managing the complex and often severe immune-related toxicities triggered by inflammatory cell death; the biological complexity and potential for overlapping toxicities when designing combination therapies to exploit synergistic effects across pathways; and the difficulty in creating targeted delivery systems that enhance efficacy while minimizing off-target effects. In summary, translation of mechanistic insights into clinical applications remains a dynamic frontier. The success of venetoclax demonstrates the potential of targeting cell death pathways. Future efforts should focus on translating insights from other RCD pathways into safe and effective clinical strategies, overcoming resistance and improving AML outcomes by expanding the therapeutic arsenal. The clinical development targeting the RCD pathway in AML is summarized in Table I (13,14,103,158,159,188,204,211–214).
Table I.Overview of therapeutic strategies targeting the RCD pathway in acute myeloid leukemia and their current clinical development status. |
AML is a highly aggressive hematologic malignancy in adults, characterized by a high recurrence rate and treatment resistance, both of which threaten patient survival (215). Despite advancements in molecular typing and targeted therapies that have improved the prognosis of some subtypes, the mechanisms driving AML heterogeneity and its microenvironmental interactions remain poorly understood (215). The limitations of chemotherapy and hematopoietic stem cell transplantation have prompted researchers to explore novel strategies targeting programmed cell death. These approaches aim to enhance treatment efficacy while reducing damage to the normal hematopoietic system, either by selectively inducing LSC clearance or remodeling the bone marrow immune microenvironment. Studies have shown that regulation of cell death processes, including autophagy, apoptosis, necroptosis, pyroptosis, ferroptosis and cuproptosis, can successfully target drug-resistant clones (17,216,217). Notably, autophagy can exhibit a context-dependent dual role in this process. In LSCs, autophagy helps maintain chemoresistance through homeostatic functions; however, its overactivation may trigger type II programmed death, eliminating metabolically compromised cells (53). The BCL-2/MCL-1 balance in the apoptotic pathway is disrupted in drug-resistant clones, and BH3 mimics can alter mitochondrial apoptotic thresholds, thereby overcoming anti-apoptotic defenses. In the AML microenvironment, the RIPK1/RIPK3/MLKL necroptosis cascade is suppressed; however, its activation can bypass apoptosis resistance and induce inflammatory cell death. Pyroptosis-induced pore formation by GSDM family proteins and activation of inflammatory vesicles result in a dual mechanism that eliminates leukemia cells and reshapes the immunosuppressive microenvironment. Ferroptosis regulates lipid peroxidation via the GPX4/acyl-CoA synthetase long chain family member 4 axis, and targeting of GSH metabolism selectively eliminates resistant subpopulations that are dependent on antioxidant defenses. The emerging cuproptosis mechanism induces proteotoxic stress via FDX1-mediated copper ion overload, providing a novel strategy to target recalcitrant LSCs with abnormal mitochondrial metabolism. These RCD processes form a networked system of spatiotemporal regulation. A combinatorial intervention targeting multiple nodes can synergistically overcome the death-resistance barrier of leukemia cells, offering a multidimensional approach to address therapeutic challenges posed by AML clonal evolution and microenvironmental sheltering. Combination therapies targeting RCD are being clinically validated, but their long-term effects on clonal evolution, immune homeostasis and metabolic reprogramming remain to be fully explored. Future research should investigate the synergistic impact of different RCD modalities on AML stem cell maintenance, chemoresistance and microenvironmental remodeling in order to enable the development of personalized therapies that precisely target death pathways.
Not applicable.
The present study was supported by the Tianjin Health Commission, Research Project of TCM and Western Integrative Medicine (grant no. 2023130), the Tianjin Education Commission, Tianjin Education Commission Research Project (General Project) (grant no. 2021KJ145), and the Shi Zhexin Tianjin Famous Traditional Chinese Medicine Inheritance Studio (grant no. tjmzy2406).
Not applicable.
RJ, HC and XY contributed to the conception of the review, conducted the literature search and screening, and wrote the original manuscript. LY, YG, CF, XH and GZ assisted with literature search, analysis of the published data and proofreading of the drafts. ZS, as the corresponding author, contributed to the design of the review framework, provided critical revisions for important intellectual content, and supervised the entire project. RJ and ZS confirm the accuracy and comprehensive nature of the literature review presented in this manuscript. Data authentication is not applicable. All authors have read and approved the final version of the manuscript.
Not applicable.
Not applicable.
The authors declare that they have no competing interests.
|
Pollyea DA, Altman JK, Assi R, Bixby D, Fathi AT, Foran JM, Gojo I, Hall AC, Jonas BA, Kishtagari A, et al: Acute myeloid leukemia, version 3.2023, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw. 21:503–513. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Moore CG, Stein A, Fathi AT and Pullarkat V: Treatment of Relapsed/Refractory AML-Novel treatment options including immunotherapy. Am J Hematol. 100:23–37. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Sasaki K, Ravandi F, Kadia TM, DiNardo CD, Short NJ, Borthakur G, Jabbour E and Kantarjian HM: De novo acute myeloid leukemia: A population-based study of outcome in the United States based on the Surveillance, Epidemiology, and End Results (SEER) database, 1980 to 2017. Cancer. 127:2049–2061. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Halik A, Tilgner M, Silva P, Estrada N, Altwasser R, Jahn E, Heuser M, Hou HA, Pratcorona M, Hills RK, et al: Genomic characterization of AML with aberrations of chromosome 7: A multinational cohort of 519 patients. J Hematol Oncol. 17:702024. View Article : Google Scholar : PubMed/NCBI | |
|
Kantarjian H, Borthakur G, Daver N, DiNardo CD, Issa G, Jabbour E, Kadia T, Sasaki K, Short NJ, Yilmaz M and Ravandi F: Current status and research directions in acute myeloid leukemia. Blood Cancer J. 14:1632024. View Article : Google Scholar : PubMed/NCBI | |
|
Lee E, Song CH, Bae SJ, Ha KT and Karki R: Regulated cell death pathways and their roles in homeostasis, infection, inflammation, and tumorigenesis. Exp Mol Med. 55:1632–1643. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Peng J, Zou M, Zhang Q, Liu D, Chen S, Fang R, Gao Y, Yan X and Hao L: Symphony of regulated cell death: Unveiling therapeutic horizons in sarcopenia. Metabolism. 172:1563592025. View Article : Google Scholar : PubMed/NCBI | |
|
Chen C, Wang J, Zhang S, Zhu X, Hu J, Liu C and Liu L: Epigenetic regulation of diverse regulated cell death modalities in cardiovascular disease: Insights into necroptosis, pyroptosis, ferroptosis, and cuproptosis. Redox Biol. 76:1033212024. View Article : Google Scholar : PubMed/NCBI | |
|
Sorokin O, Hause F, Wedler A, Alakhras T, Bauchspiess T, Dietrich A, Günther WF, Guha C, Obika KB, Kraft J, et al: Comprehensive analysis of regulated cell death pathways: Intrinsic disorder, protein-protein interactions, and cross-pathway communication. Apoptosis. 30:2110–2162. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Peng F, Liao M, Qin R, Zhu S, Peng C, Fu L, Chen Y and Han B: Regulated cell death (RCD) in cancer: Key pathways and targeted therapies. Signal Transduct Target Ther. 7:2862022. View Article : Google Scholar : PubMed/NCBI | |
|
Seo W, Silwal P, Song IC and Jo EK: The dual role of autophagy in acute myeloid leukemia. J Hematol Oncol. 15:512022. View Article : Google Scholar : PubMed/NCBI | |
|
Ren J, Tao Y, Peng M, Xiao Q, Jing Y, Huang J, Yang J, Lin C, Sun M, Lei L, et al: Targeted activation of GPER enhances the efficacy of venetoclax by boosting leukemic pyroptosis and CD8+ T cell immune function in acute myeloid leukemia. Cell Death Dis. 13:9152022. View Article : Google Scholar : PubMed/NCBI | |
|
Chan FK: RIPK3 Slams the Brake on Leukemogenesis. Cancer Cell. 30:7–9. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Ye F, Zhang W, Fan C, Dong J, Peng M, Deng W, Zhang H and Yang L: Antileukemic effect of venetoclax and hypomethylating agents via caspase-3/GSDME-mediated pyroptosis. J Transl Med. 21:6062023. 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 | |
|
Xu B, Wang S, Li R, Chen K, He L, Deng M, Kannappan V, Zha J, Dong H and Wang W: Disulfiram/copper selectively eradicates AML leukemia stem cells in vitro and in vivo by simultaneous induction of ROS-JNK and inhibition of NF-κB and Nrf2. Cell Death Dis. 8:e27972017. View Article : Google Scholar : PubMed/NCBI | |
|
Guo Z, Liu Y, Chen D, Sun Y, Li D, Meng Y, Zhou Q, Zeng F, Deng G and Chen X: Targeting regulated cell death: Apoptosis, necroptosis, pyroptosis, ferroptosis, and cuproptosis in anticancer immunity. J Transl Int Med. 13:10–32. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Garciaz S, Miller T, Collette Y and Vey N: Targeting regulated cell death pathways in acute myeloid leukemia. Cancer Drug Resist. 6:151–168. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
He R, Liu Y, Fu W, He X, Liu S, Xiao D and Tao Y: Mechanisms and Cross-talk of regulated cell death and their epigenetic modifications in tumor progression. Mol Cancer. 23:2672024. View Article : Google Scholar : PubMed/NCBI | |
|
Dikic I and Elazar Z: Mechanism and medical implications of mammalian autophagy. Nat Rev Mol Cell Biol. 19:349–364. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Kaushik S and Cuervo AM: The coming of age of Chaperone-mediated autophagy. Nat Rev Mol Cell Biol. 19:365–381. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Yamamoto H and Matsui T: Molecular mechanisms of macroautophagy, microautophagy, and Chaperone-mediated autophagy. J Nippon Med Sch. 91:2–9. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Niu X, You Q, Hou K, Tian Y, Wei P, Zhu Y, Gao B, Ashrafizadeh M, Aref AR, Kalbasi A, et al: Autophagy in cancer development, immune evasion, and drug resistance. Drug Resist Updat. 78:1011702025. View Article : Google Scholar : PubMed/NCBI | |
|
Debnath J, Gammoh N and Ryan KM: Autophagy and autophagy-related pathways in cancer. Nat Rev Mol Cell Biol. 24:560–575. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Sankar DS, Kaeser-Pebernard S, Vionnet C, Favre S, de Oliveira Marchioro L, Pillet B, Zhou J, Stumpe M, Kovacs WJ, Kressler D, et al: The ULK1 effector BAG2 regulates autophagy initiation by modulating AMBRA1 localization. Cell Rep. 43:1146892024. View Article : Google Scholar : PubMed/NCBI | |
|
Xi H, Wang S, Wang B, Hong X, Liu X, Li M, Shen R and Dong Q: The role of interaction between autophagy and apoptosis in tumorigenesis (Review). Oncol Reps. 48:2082022. View Article : Google Scholar | |
|
Cook ASI, Chen M, Nguyen TN, Cabezudo AC, Khuu G, Rao S, Garcia SN, Yang M, Iavarone AT, Ren X, et al: Structural pathway for PI3-kinase regulation by VPS15 in autophagy. Science. 388:eadl37872025. View Article : Google Scholar : PubMed/NCBI | |
|
Nascimbeni AC, Codogno P and Morel E: Local detection of PtdIns3P at autophagosome biogenesis membrane platforms. Autophagy. 13:1602–1612. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Wani WY, Boyer-Guittaut M, Dodson M, Chatham J, Darley-Usmar V and Zhang J: Regulation of autophagy by protein post-translational modification. Lab Invest. 95:14–25. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Huang R, Xu Y, Wan W, Shou X, Qian J, You Z, Liu B, Chang C, Zhou T, Lippincott-Schwartz J and Liu W: Deacetylation of nuclear LC3 drives autophagy initiation under starvation. Mol Cell. 57:456–466. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Ichimura Y, Kirisako T, Takao T, Satomi Y, Shimonishi Y, Ishihara N, Mizushima N, Tanida I, Kominami E, Ohsumi M, et al: A ubiquitin-like system mediates protein lipidation. Nature. 408:488–492. 2000. View Article : Google Scholar : PubMed/NCBI | |
|
Rogov V, Dötsch V, Johansen T and Kirkin V: Interactions between autophagy receptors and ubiquitin-like proteins form the molecular basis for selective autophagy. Mol Cell. 53:167–178. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Kimmelman AC and White E: Autophagy and tumor metabolism. Cell Metab. 25:1037–1043. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Santos de Macedo BG, Albuquerque de Melo M, Pereira-Martins DA, Machado-Neto JA and Traina F: An updated outlook on autophagy mechanism and how it supports acute myeloid leukemia maintenance. Biochim Biophys Acta Rev Cancer. 1879:1892142024. View Article : Google Scholar : PubMed/NCBI | |
|
Nomura N, Ito C, Ooshio T, Tadokoro Y, Kohno S, Ueno M, Kobayashi M, Kasahara A, Takase Y, Kurayoshi K, et al: Essential role of autophagy in protecting neonatal haematopoietic stem cells from oxidative stress in a p62-independent manner. Sci Rep. 11:16662021. View Article : Google Scholar : PubMed/NCBI | |
|
Chen Y, Luo X, Zou Z and Liang Y: The role of reactive oxygen species in tumor treatment and its impact on bone marrow hematopoiesis. Curr Drug Targets. 21:477–498. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Sharma P, Piya S, Ma H, Baran N, Anna Zal M, Hindley CJ, Dao K, Sims M, Zal T, Ruvolo V, et al: ERK1/2 inhibition overcomes resistance in acute myeloid leukemia (AML) and alters mitochondrial dynamics. Blood. 138:33382021. View Article : Google Scholar | |
|
Folkerts H, Wierenga AT, van den Heuvel FA, Woldhuis RR, Kluit DS, Jaques J, Schuringa JJ and Vellenga E: Elevated VMP1 expression in acute myeloid leukemia amplifies autophagy and is protective against venetoclax-induced apoptosis. Cell Death Dis. 10:4212019. View Article : Google Scholar : PubMed/NCBI | |
|
Khan A, Singh VK, Thakral D and Gupta R: Autophagy in acute myeloid leukemia: A paradoxical role in chemoresistance. Clin Transl Oncol. 24:1459–1469. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Heydt Q, Larrue C, Saland E, Bertoli S, Sarry JE, Besson A, Manenti S, Joffre C and Mansat-De Mas V: Oncogenic FLT3-ITD supports autophagy via ATF4 in acute myeloid leukemia. Oncogene. 37:787–797. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Omori I, Yamaguchi H, Miyake K, Miyake N, Kitano T and Inokuchi K: D816V mutation in the KIT gene activation loop has greater cell-proliferative and anti-apoptotic ability than N822K mutation in core-binding factor acute myeloid leukemia. Exp Hematol. 52:56–64.e54. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Larrue C, Heydt Q, Saland E, Boutzen H, Kaoma T, Sarry JE, Joffre C and Récher C: Oncogenic KIT mutations induce STAT3-dependent autophagy to support cell proliferation in acute myeloid leukemia. Oncogenesis. 8:392019. View Article : Google Scholar : PubMed/NCBI | |
|
Zou Q, Tan S, Yang Z, Zhan Q, Jin H, Xian J, Zhang S, Yang L, Wang L and Zhang L: NPM1 mutant mediated PML delocalization and stabilization enhances autophagy and cell survival in leukemic cells. Theranostics. 7:2289–2304. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Folkerts H, Hilgendorf S, Wierenga ATJ, Jaques J, Mulder AB, Coffer PJ, Schuringa JJ and Vellenga E: Inhibition of autophagy as a treatment strategy for p53 wild-type acute myeloid leukemia. Cell Death Dis. 8:e29272017. View Article : Google Scholar : PubMed/NCBI | |
|
Yang Y, Pu J and Yang Y: Glycolysis and chemoresistance in acute myeloid leukemia. Heliyon. 10:e357212024. View Article : Google Scholar : PubMed/NCBI | |
|
Wang YH, Israelsen WJ, Lee D, Yu VWC, Jeanson NT, Clish CB, Cantley LC, Vander Heiden MG and Scadden DT: Cell-state-specific metabolic dependency in hematopoiesis and leukemogenesis. Cell. 158:1309–1323. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Chen S, Tao Y, Wang Q, Ren J, Jing Y, Huang J, Zhang L and Li R: Glucose Induced-AKT/mTOR activation accelerates glycolysis and promotes cell survival in acute myeloid leukemia. Leuk Res. 128:1070592023. View Article : Google Scholar : PubMed/NCBI | |
|
Stevens BM, Jones CL, Pollyea DA, Culp-Hill R, D'Alessandro A, Winters A, Krug A, Abbott D, Goosman M, Pei S, et al: Fatty acid metabolism underlies venetoclax resistance in acute myeloid leukemia stem cells. Nat Cancer. 1:1176–1187. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Carter JL, Su Y, Qiao X, Zhao J, Wang G, Howard M, Edwards H, Bao X, Li J, Hüttemann M, et al: Acquired resistance to venetoclax plus azacitidine in acute myeloid leukemia: In vitro models and mechanisms. Biochem Pharmacol. 216:1157592023. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang HB, Sun ZK, Zhong FM, Yao FY, Liu J, Zhang J, Zhang N, Lin J, Li SQ, Li MY, et al: A novel fatty acid metabolism-related signature identifies features of the tumor microenvironment and predicts clinical outcome in acute myeloid leukemia. Lipids Health Dis. 21:792022. View Article : Google Scholar : PubMed/NCBI | |
|
Tcheng M, Roma A, Ahmed N, Smith RW, Jayanth P, Minden MD, Schimmer AD, Hess DA, Hope K, Rea KA, et al: Very long chain fatty acid metabolism is required in acute myeloid leukemia. Blood. 137:3518–3532. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao Y, Guo H, Niu L and Zhao J: Metabolic pathways and chemotherapy resistance in acute myeloid leukemia (AML): Insights into Enoyl-CoA hydratase domain-containing protein 3 (ECHDC3) as a potential therapeutic target. Cancer Pathogenesis and Therapy; 2025, View Article : Google Scholar | |
|
Du W, Xu A, Huang Y, Cao J, Zhu H, Yang B, Shao X, He Q and Ying M: The role of autophagy in targeted therapy for acute myeloid leukemia. Autophagy. 17:2665–2679. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Li X, Qian Q, Li J, Zhang L, Wang L, Huang D, Xu Q and Chen W: The RNA-binding protein CELF1 targets ATG5 to regulate autophagy and promote drug resistance in acute myeloid leukemia. Cell Death Dis. 16:5992025. View Article : Google Scholar : PubMed/NCBI | |
|
Herschbein L and Liesveld JL: Dueling for dual inhibition: Means to enhance effectiveness of PI3K/Akt/mTOR inhibitors in AML. Blood Rev. 32:235–248. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Koschade SE, Klann K, Shaid S, Vick B, Stratmann JA, Thölken M, Meyer LM, Nguyen TD, Campe J, Moser LM, et al: Translatome proteomics identifies autophagy as a resistance mechanism to on-target FLT3 inhibitors in acute myeloid leukemia. Leukemia. 36:2396–2407. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Cao Y, Wang Y, Chen Y, Zhang X, Zuo Y, Ge X, Sun C, Ren B, Liu Y, Wang M and Lu J: Voacamine initiates the PI3K/mTOR/Beclin1 pathway to induce autophagy and potentiate apoptosis in acute myeloid leukemia. Phytomedicine. 143:1568592025. View Article : Google Scholar : PubMed/NCBI | |
|
Nössing C and Ryan KM: 50 years on and still very much alive: ‘Apoptosis: A basic biological phenomenon with wide-ranging implications in tissue kinetics’. Br J Cancer. 128:426–431. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Yoon JH and Gores GJ: Death receptor-mediated apoptosis and the liver. J Hepatol. 37:400–410. 2002. View Article : Google Scholar : PubMed/NCBI | |
|
O'Neill KL, Huang K, Zhang J, Chen Y and Luo X: Inactivation of prosurvival Bcl-2 proteins activates Bax/Bak through the outer mitochondrial membrane. Genes Dev. 30:973–988. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Xiong S, Mu T, Wang G and Jiang X: Mitochondria-mediated apoptosis in mammals. Protein Cell. 5:737–749. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Croce CM, Vaux D, Strasser A, Opferman JT, Czabotar PE and Fesik SW: The BCL-2 protein family: from discovery to drug development. Cell Death Differ. 32:1369–1381. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Reyna DE, Garner TP, Lopez A, Kopp F, Choudhary GS, Sridharan A, Narayanagari SR, Mitchell K, Dong B, Bartholdy BA, et al: Direct activation of BAX by BTSA1 overcomes apoptosis resistance in acute myeloid leukemia. Cancer Cell. 32:490–505.e10. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Singh G, Guibao CD, Seetharaman J, Aggarwal A, Grace CR, McNamara DE, Vaithiyalingam S, Waddell MB and Moldoveanu T: Structural basis of BAK activation in mitochondrial apoptosis initiation. Nat Commun. 13:2502022. View Article : Google Scholar : PubMed/NCBI | |
|
Tait SW and Green DR: Mitochondria and cell death: Outer membrane permeabilization and beyond. Nat Rev Mol Cell Biol. 11:621–632. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Cain K, Bratton SB and Cohen GM: The Apaf-1 apoptosome: A large caspase-activating complex. Biochimie. 84:203–214. 2002. View Article : Google Scholar : PubMed/NCBI | |
|
Kashyap D, Garg VK and Goel N: Intrinsic and extrinsic pathways of apoptosis: Role in cancer development and prognosis. Adv Protein Chem Struct Biol. 125:73–120. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Zheng L, Yao Y and Lenardo MJ: Death receptor 5 rises to the occasion. Cell Res. 33:199–200. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Helmke C, Raab M, Rödel F, Matthess Y, Oellerich T, Mandal R, Sanhaji M, Urlaub H, Rödel C, Becker S and Strebhardt K: Ligand stimulation of CD95 induces activation of Plk3 followed by phosphorylation of caspase-8. Cell Res. 26:914–934. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Huang K, Zhang J, O'Neill KL, Gurumurthy CB, Quadros RM, Tu Y and Luo X: Cleavage by Caspase 8 and mitochondrial membrane association activate the BH3-only protein bid during TRAIL-induced apoptosis. J Biol Chem. 291:11843–11851. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
D'Arcy MS: Cell death: A review of the major forms of apoptosis, necrosis and autophagy. Cell Biol Int. 43:582–592. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Kantari C and Walczak H: Caspase-8 and bid: Caught in the act between death receptors and mitochondria. Biochim Biophys Acta. 1813:558–563. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Kim WS, Lee KS, Kim JH, Kim CK, Lee G, Choe J, Won MH, Kim TH, Jeoung D, Lee H, et al: The caspase-8/Bid/cytochrome c axis links signals from death receptors to mitochondrial reactive oxygen species production. Free Radic Biol Med. 112:567–577. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Moyer A, Tanaka K and Cheng EH: Apoptosis in cancer biology and therapy. Annu Rev Pathol. 20:303–328. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Pfeffer CM and Singh ATK: Apoptosis: A target for anticancer therapy. Int J Mol Sci. 19:4482018. View Article : Google Scholar : PubMed/NCBI | |
|
Campbell KJ and Tait SWG: Targeting BCL-2 regulated apoptosis in cancer. Open Biol. 8:1800022018. View Article : Google Scholar : PubMed/NCBI | |
|
Ashkenazi A, Fairbrother WJ, Leverson JD and Souers AJ: From basic apoptosis discoveries to advanced selective BCL-2 family inhibitors. Nat Rev Drug Discov. 16:273–284. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Konopleva M, Pollyea DA, Potluri J, Chyla B, Hogdal L, Busman T, McKeegan E, Salem AH, Zhu M, Ricker JL, et al: Efficacy and biological correlates of response in a phase II study of venetoclax monotherapy in patients with acute myelogenous leukemia. Cancer Discov. 6:1106–1117. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Wei AH, Strickland SA Jr, Hou JZ, Fiedler W, Lin TL, Walter RB, Enjeti A, Tiong IS, Savona M, Lee S, et al: Venetoclax combined with Low-dose cytarabine for previously untreated patients with acute myeloid leukemia: Results from a Phase Ib/II study. J Clin Oncol. 37:1277–1284. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
DiNardo CD, Jonas BA, Pullarkat V, Thirman MJ, Garcia JS, Wei AH, Konopleva M, Döhner H, Letai A, Fenaux P, et al: Azacitidine and venetoclax in previously untreated acute myeloid leukemia. N Engl J Med. 383:617–629. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Jiao CQ, Hu C, Sun MH, Li Y, Wu C, Xu F, Zhang L, Huang FR, Zhou JJ, Dai JF, et al: Targeting METTL3 mitigates venetoclax resistance via proteasome-mediated modulation of MCL1 in acute myeloid leukemia. Cell Death Dis. 16:2332025. View Article : Google Scholar : PubMed/NCBI | |
|
Chai J, Du C, Wu JW, Kyin S, Wang X and Shi Y: Structural and biochemical basis of apoptotic activation by Smac/DIABLO. Nature. 406:855–862. 2000. View Article : Google Scholar : PubMed/NCBI | |
|
Carter BZ, Mak PY, Mak DH, Shi Y, Qiu Y, Bogenberger JM, Mu H, Tibes R, Yao H, Coombes KR, et al: Synergistic targeting of AML stem/progenitor cells with IAP antagonist birinapant and demethylating agents. J Natl Cancer Inst. 106:djt4402014. View Article : Google Scholar : PubMed/NCBI | |
|
Gencel-Augusto J and Lozano G: Targeted degradation of mutant p53 reverses the Pro-oncogenic Dominant-negative effect. Cancer Res. 85:1955–1956. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Tuval A, Strandgren C, Heldin A, Palomar-Siles M and Wiman KG: Pharmacological reactivation of p53 in the era of precision anticancer medicine. Nat Rev Clin Oncol. 21:106–120. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Napolitano R, De Matteis S, Carloni S, Bruno S, Abbati G, Capelli L, Ghetti M, Bochicchio MT, Liverani C, Mercatali L, et al: Kevetrin induces apoptosis in TP53 wild-type and mutant acute myeloid leukemia cells. Oncol Rep. 44:1561–1573. 2020.PubMed/NCBI | |
|
Fulda S: Targeting apoptosis for anticancer therapy. Semin Cancer Biol. 31:84–88. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Chow LQ, Eckhardt SG, Gustafson DL, Langer CJ, Camidge DR, Padavic K, Gore L, Smith M, Chow LQ, von Mehren M, et al: HGS-ETR1, an antibody targeting TRAIL-R1, in combination with paclitaxel and carboplatin in patients with advanced solid malignancies: Results of a phase 1 and PK study. J Clin Oncol. 24:25152006. View Article : Google Scholar | |
|
You R, Hou D, Wang B, Liu J, Wang X, Xiao Q, Pan Z, Li D, Feng X, Kang L, et al: Bone marrow microenvironment drives AML cell OXPHOS addiction and AMPK inhibition to resist chemotherapy. J Leukoc Biol. 112:299–311. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Cheung HL, Wong YH, Li YY, Yang X, Ko LH, Tan Kabigting JE, Chan KC, Leung AYH and Chan BP: Microenvironment matters: In vitro 3D bone marrow niches differentially modulate survival, phenotype and drug responses of acute myeloid leukemia (AML) cells. Biomaterials. 312:1227192025. View Article : Google Scholar : PubMed/NCBI | |
|
Zou J, Zheng Y, Huang Y, Tang D, Kang R and Chen R: The versatile gasdermin family: Their function and roles in diseases. Front Immunol. 12:7515332021. View Article : Google Scholar : PubMed/NCBI | |
|
Broz P, Pelegrín P and Shao F: The gasdermins, a protein family executing cell death and inflammation. Nat Rev Immunol. 20:143–157. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Tamura M, Tanaka S, Fujii T, Aoki A, Komiyama H, Ezawa K, Sumiyama K, Sagai T and Shiroishi T: Members of a novel gene family, Gsdm, are expressed exclusively in the epithelium of the skin and gastrointestinal tract in a highly Tissue-specific manner. Genomics. 89:618–629. 2007. View Article : Google Scholar : PubMed/NCBI | |
|
Wang S, Yuan YH, Chen NH and Wang HB: The mechanisms of NLRP3 inflammasome/pyroptosis activation and their role in Parkinson's disease. Int Immunopharmacol. 67:458–464. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Miao EA, Rajan JV and Aderem A: Caspase-1-induced pyroptotic cell death. Immunol Rev. 243:206–214. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Shi J, Zhao Y, Wang K, Shi X, Wang Y, Huang H, Zhuang Y, Cai T, Wang F and Shao F: Cleavage of GSDMD by inflammatory caspases determines pyroptotic cell death. Nature. 526:660–665. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Yu P, Zhang X, Liu N, Tang L, Peng C and Chen X: Pyroptosis: Mechanisms and diseases. Signal Transduct Target Ther. 6:1282021. View Article : Google Scholar : PubMed/NCBI | |
|
Ding J, Wang K, Liu W, She Y, Sun Q, Shi J, Sun H, Wang DC and Shao F: Pore-forming activity and structural autoinhibition of the gasdermin family. Nature. 535:111–116. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Garlanda C, Dinarello CA and Mantovani A: The interleukin-1 family: Back to the future. Immunity. 39:1003–1018. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Rao Z, Zhu Y, Yang P, Chen Z, Xia Y, Qiao C, Liu W, Deng H, Li J, Ning P and Wang Z: Pyroptosis in inflammatory diseases and cancer. Theranostics. 12:4310–4329. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Kayagaki N, Stowe IB, Lee BL, O'Rourke K, Anderson K, Warming S, Cuellar T, Haley B, Roose-Girma M, Phung QT, et al: Caspase-11 cleaves gasdermin D for non-canonical inflammasome signalling. Nature. 526:666–671. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Yang J, Zhao Y and Shao F: Non-canonical activation of inflammatory caspases by cytosolic LPS in innate immunity. Curr Opin Immunol. 32:78–83. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Shi J, Zhao Y, Wang Y, Gao W, Ding J, Li P, Hu L and Shao F: Inflammatory caspases are innate immune receptors for intracellular LPS. Nature. 514:187–192. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Hagar JA, Powell DA, Aachoui Y, Ernst RK and Miao EA: Cytoplasmic LPS activates caspase-11: implications in TLR4-independent endotoxic shock. Science. 341:1250–1253. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Liu Y, Pan R, Ouyang Y, Gu W, Xiao T, Yang H, Tang L, Wang H, Xiang B and Chen P: Pyroptosis in health and disease: Mechanisms, regulation and clinical perspective. Signal Transduct Target Ther. 9:2452024. View Article : Google Scholar : PubMed/NCBI | |
|
Kayagaki N, Warming S, Lamkanfi M, Vande Walle L, Louie S, Dong J, Newton K, Qu Y, Liu J, Heldens S, et al: Non-canonical inflammasome activation targets caspase-11. Nature. 479:117–121. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Shi X, Sun Q, Hou Y, Zeng H, Cao Y, Dong M, Ding J and Shao F: Recognition and maturation of IL-18 by caspase-4 noncanonical inflammasome. Nature. 624:442–450. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Wang Y, Gao W, Shi X, Ding J, Liu W, He H, Wang K and Shao F: Chemotherapy drugs induce pyroptosis through caspase-3 cleavage of a gasdermin. Nature. 547:99–103. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Oltra SS, Colomo S, Sin L, Pérez-López M, Lázaro S, Molina-Crespo A, Choi KH, Ros-Pardo D, Martínez L, Morales S, et al: Distinct GSDMB protein isoforms and protease cleavage processes differentially control pyroptotic cell death and mitochondrial damage in cancer cells. Cell Death Differ. 30:1366–1381. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Wang S, Chang CW, Huang J, Zeng S, Zhang X, Hung MC and Hou J: Gasdermin C sensitizes tumor cells to PARP inhibitor therapy in cancer models. J Clin Invest. 134:e1668412024. View Article : Google Scholar : PubMed/NCBI | |
|
Alves-Hanna FS, Crespo-Neto JA, Nogueira GM, Pereira DS, Lima AB, Ribeiro TLP, Santos VGR, Fonseca JRF, Magalhães-Gama F, Sadahiro A and Costa AG: Insights regarding the role of inflammasomes in leukemia: What Do We Know? J Immunol Res. 2023:55844922023. View Article : Google Scholar : PubMed/NCBI | |
|
Johnson DC, Taabazuing CY, Okondo MC, Chui AJ, Rao SD, Brown FC, Reed C, Peguero E, de Stanchina E, Kentsis A and Bachovchin DA: DPP8/DPP9 inhibitor-induced pyroptosis for treatment of acute myeloid leukemia. Nat Med. 24:1151–1156. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Deng S, Pan Y, An N, Chen F, Chen H, Wang H, Xu X, Liu R, Yang L, Wang X, et al: Downregulation of RCN1 promotes pyroptosis in acute myeloid leukemia cells. Mol Oncol. 17:2584–2602. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Askmyr M, Ågerstam H, Hansen N, Gordon S, Arvanitakis A, Rissler M, Juliusson G, Richter J, Järås M and Fioretos T: Selective killing of candidate AML stem cells by antibody targeting of IL1RAP. Blood. 121:3709–3713. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Ågerstam H, Karlsson C, Hansen N, Sandén C, Askmyr M, von Palffy S, Högberg C, Rissler M, Wunderlich M, Juliusson G, et al: Antibodies targeting human IL1RAP (IL1R3) show therapeutic effects in xenograft models of acute myeloid leukemia. Proc Natl Acad Sci USA. 112:10786–10791. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Wang Y, Sun X, Yuan S, Hou S, Guo T, Chu Y, Pang T, Luo HR, Yuan W and Wang X: Interleukin-1β inhibits normal hematopoietic expansion and promotes acute myeloid leukemia progression via the bone marrow niche. Cytotherapy. 22:127–134. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Wong J, Tran LT, Magun EA, Magun BE and Wood LJ: Production of IL-1β by bone marrow-derived macrophages in response to chemotherapeutic drugs: Synergistic effects of doxorubicin and vincristine. Cancer Biol Ther. 15:1395–1403. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Yan J, Wan P, Choksi S and Liu ZG: Necroptosis and tumor progression. Trends Cancer. 8:21–27. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Liu X, Miao M, Sun J, Wu J and Qin X: PANoptosis: A potential new target for programmed cell death in breast cancer treatment and prognosis. Apoptosis. 29:277–288. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
He S, Wang L, Miao L, Wang T, Du F, Zhao L and Wang X: Receptor interacting protein kinase-3 determines cellular necrotic response to TNF-alpha. Cell. 137:1100–1111. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Holler N, Zaru R, Micheau O, Thome M, Attinger A, Valitutti S, Bodmer JL, Schneider P, Seed B and Tschopp J: Fas triggers an alternative, caspase-8-independent cell death pathway using the kinase RIP as effector molecule. Nat Immunol. 1:489–495. 2000. View Article : Google Scholar : PubMed/NCBI | |
|
Mandal R, Barrón JC, Kostova I, Becker S and Strebhardt K: Caspase-8: The double-edged sword. Biochim Biophys Acta Rev Cancer. 1873:1883572020. View Article : Google Scholar : PubMed/NCBI | |
|
Kaiser WJ, Sridharan H, Huang C, Mandal P, Upton JW, Gough PJ, Sehon CA, Marquis RW, Bertin J and Mocarski ES: Toll-like receptor 3-mediated necrosis via TRIF, RIP3, and MLKL. J Biol Chem. 288:31268–31279. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Upton JW, Kaiser WJ and Mocarski ES: DAI/ZBP1/DLM-1 complexes with RIP3 to mediate virus-induced programmed necrosis that is targeted by murine cytomegalovirus vIRA. Cell Host Microbe. 11:290–297. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Pasparakis M and Vandenabeele P: Necroptosis and its role in inflammation. Nature. 517:311–320. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Linkermann A and Green DR: Necroptosis. N Engl J Med. 370:455–465. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Murphy JM, Czabotar PE, Hildebrand JM, Lucet IS, Zhang JG, Alvarez-Diaz S, Lewis R, Lalaoui N, Metcalf D, Webb AI, et al: The pseudokinase MLKL mediates necroptosis via a molecular switch mechanism. Immunity. 39:443–453. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Gong YN, Guy C, Olauson H, Becker JU, Yang M, Fitzgerald P, Linkermann A and Green DR: ESCRT-III acts downstream of MLKL to regulate necroptotic cell death and its consequences. Cell. 169:286–300.e16. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Cai Z, Jitkaew S, Zhao J, Chiang HC, Choksi S, Liu J, Ward Y, Wu LG and Liu ZG: Plasma membrane translocation of trimerized MLKL protein is required for TNF-induced necroptosis. Nat Cell Biol. 16:55–65. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Khan N, Downey J, Sanz J, Kaufmann E, Blankenhaus B, Pacis A, Pernet E, Ahmed E, Cardoso S, Nijnik A, et al: M. tuberculosis reprograms hematopoietic stem cells to limit myelopoiesis and impair trained immunity. Cell. 183:752–770.e22. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Gong Y, Fan Z, Luo G, Yang C, Huang Q, Fan K, Cheng H, Jin K, Ni Q, Yu X and Liu C: The role of necroptosis in cancer biology and therapy. Mol Cancer. 18:1002019. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang T, Wang Y, Inuzuka H and Wei W: Necroptosis pathways in tumorigenesis. Semin Cancer Biol. 86:32–40. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Höckendorf U, Yabal M, Herold T, Munkhbaatar E, Rott S, Jilg S, Kauschinger J, Magnani G, Reisinger F, Heuser M, et al: RIPK3 restricts myeloid leukemogenesis by promoting cell death and differentiation of leukemia initiating cells. Cancer Cell. 30:75–91. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Wang Y, Zhang TJ, Zhang LC, Xu ZJ, Chu MQ, Zhao YJ, Lin J, Qian J and Zhou JD: Overexpression and oncogenic role of RIPK3 in acute myeloid leukemia associated with specific subtypes and treatment outcome. BMC Cancer. 25:2532025. View Article : Google Scholar : PubMed/NCBI | |
|
Xin J, You D, Breslin P, Li J, Zhang J, Wei W, Cannova J, Volk A, Gutierrez R, Xiao Y, et al: Sensitizing acute myeloid leukemia cells to induced differentiation by inhibiting the RIP1/RIP3 pathway. Leukemia. 31:1154–1165. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Zhu S, Luo Y, Li K, Mei C, Wang Y, Jiang L, Wang W, Zhang Q, Yang W, Lang W, et al: RIPK3 deficiency blocks R-2-hydroxyglutarate-induced necroptosis in IDH-mutated AML cells. Sci Adv. 10:eadi17822024. View Article : Google Scholar : PubMed/NCBI | |
|
Hillert LK, Bettermann-Bethge K, Nimmagadda SC, Fischer T, Naumann M and Lavrik IN: Targeting RIPK1 in AML cells carrying FLT3-ITD. Int J Cancer. 145:1558–1569. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Li J, Liao D, Wang F, Wang Z, Li Y, Xiong Y and Niu T: RIPK1 inhibition enhances the therapeutic efficacy of chidamide in FLT3-ITD positive AML, both in vitro and in vivo. Leuk Lymphoma. 63:1167–1179. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Yue X, Chen Q and He J: Combination strategies to overcome resistance to the BCL2 inhibitor venetoclax in hematologic malignancies. Leuk Lymphoma. 20:5242020. | |
|
Culver-Cochran AE, Hassan A, Hueneman K, Choi K, Ma A, VanCauwenbergh B, O'Brien E, Wunderlich M, Perentesis JP and Starczynowski DT: Chemotherapy resistance in acute myeloid leukemia is mediated by A20 suppression of spontaneous necroptosis. Nat Commun. 15:91892024. View Article : Google Scholar : PubMed/NCBI | |
|
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 F, Kang R, Tang D and Liu J: Ferroptosis: principles and significance in health and disease. J Hematol Oncol. 17:412024. View Article : Google Scholar : PubMed/NCBI | |
|
Koleini N, Shapiro JS, Geier J and Ardehali H: Ironing out mechanisms of iron homeostasis and disorders of iron deficiency. J Clin Invest. 131:e1486712021. View Article : Google Scholar : PubMed/NCBI | |
|
Co HKC, Wu CC, Lee YC and Chen SH: Emergence of large-scale cell death through ferroptotic trigger waves. Nature. 631:654–662. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Jiang X, Stockwell BR and Conrad M: Ferroptosis: Mechanisms, biology and role in disease. Nat Rev Mol Cell Biol. 22:266–282. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Yang WS, SriRamaratnam R, Welsch ME, Shimada K, Skouta R, Viswanathan VS, Cheah JH, Clemons PA, Shamji AF and Clish CB: Regulation of ferroptotic cancer cell death by GPX4. Cell. 156:317–331. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Stockwell BR: Ferroptosis turns 10: Emerging mechanisms, physiological functions, and therapeutic applications. Cell. 185:2401–2421. 2022. 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 | |
|
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 | |
|
Reyhani A, McKenzie TG, Fu Q and Qiao GG: Fenton-Chemistry-Mediated Radical Polymerization. Macromol Rapid Commun. 40:19002202019. View Article : Google Scholar : PubMed/NCBI | |
|
Dai E, Chen X, Linkermann A, Jiang X, Kang R, Kagan VE, Bayir H, Yang WS, Garcia-Saez AJ, Ioannou MS, et al: A guideline on the molecular ecosystem regulating ferroptosis. Nat Cell Biol. 26:1447–1457. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Ma S, Henson ES, Chen Y and Gibson SB: Ferroptosis is induced following siramesine and lapatinib treatment of breast cancer cells. Cell Death Dis. 7:e23072016. View Article : Google Scholar : PubMed/NCBI | |
|
Bannai S: Exchange of cystine and glutamate across plasma membrane of human fibroblasts. J Biol Chem. 261:2256–2263. 1986. View Article : Google Scholar : PubMed/NCBI | |
|
Koppula P, Zhang Y, Zhuang L and Gan B: Amino acid transporter SLC7A11/xCT at the crossroads of regulating redox homeostasis and nutrient dependency of cancer. Cancer Commun (Lond). 38:122018.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 : PubMed/NCBI | |
|
Liu MR, Zhu WT and Pei DS: System Xc-: A key regulatory target of ferroptosis in cancer. Invest New Drugs. 39:1123–1131. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Tang D, Chen X, Kang R and Kroemer G: Ferroptosis: Molecular mechanisms and health implications. Cell Res. 31:107–125. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Scarpellini C, Klejborowska G, Lanthier C, Hassannia B, Vanden Berghe T and Augustyns K: Beyond ferrostatin-1: A comprehensive review of ferroptosis inhibitors. Trends Pharmacol Sci. 44:902–916. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Brown CW, Amante JJ, Chhoy P, Elaimy AL, Liu H, Zhu LJ, Baer CE, Dixon SJ and Mercurio AM: Prominin2 drives ferroptosis resistance by stimulating iron export. Dev Cell. 51:575–586.e4. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Cao M, Li H, Sun D, He S, Yu Y, Li J, Chen H, Shi J, Ren J, Li N and Chen W: Cancer screening in China: The current status, challenges, and suggestions. Cancer Lett. 506:120–127. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang H and Sun C, Sun Q, Li Y, Zhou C and Sun C: Susceptibility of acute myeloid leukemia cells to ferroptosis and evasion strategies. Front Mol Biosci. 10:12757742023. View Article : Google Scholar : PubMed/NCBI | |
|
Akiyama H, Zhao R, Ostermann LB, Li Z, Tcheng M, Yazdani SJ, Moayed A, Pryor ML II, Slngh S, Baran N, et al: Mitochondrial regulation of GPX4 inhibition-mediated ferroptosis in acute myeloid leukemia. Leukemia. 38:729–740. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Na X, Li L, Liu D, He J, Zhang L and Zhou Y: Natural products targeting ferroptosis pathways in cancer therapy (Review). Oncol Rep. 52:1232024. View Article : Google Scholar : PubMed/NCBI | |
|
Ma C, Wang J, Cui S and Xu R: Crotonoside induces ferroptosis and mitochondrial dysfunction in AML. Eur J Pharmacol. 1002:1778592025. View Article : Google Scholar : PubMed/NCBI | |
|
Birsen R, Larrue C, Decroocq J, Johnson N, Guiraud N, Gotanegre M, Cantero-Aguilar L, Grignano E, Huynh T, Fontenay M, et al: APR-246 induces early cell death by ferroptosis in acute myeloid leukemia. Haematologica. 107:403–416. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Lin AY, Rink JS, Yang E, Small S, Gerber JJ, Zak TJ, Altman J, Abaza Y, Platanias LC, Gordon LI and Thaxton CS: Targeting scavenger receptor class B type 1 with a bioinspired ligand induces apoptosis or ferroptosis in AML. Blood Neoplasia. 2:1001222025. View Article : Google Scholar : PubMed/NCBI | |
|
Jiang F, Yu WJ, Wang XH, Tang YT, Guo L and Jiao XY: Regulation of hepcidin through GDF-15 in cancer-related anemia. Clin Chim Acta. 428:14–19. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Lu QW and Liao Y: GDF-15 upregulates the SLC7A11/GPX4 signaling axis and promotes mitoxantrone resistance in AML cells. Eur J Med Res. 30:5042025. View Article : Google Scholar : PubMed/NCBI | |
|
Yu Y, Meng Y, Xu X, Tong T, He C, Wang L, Wang K, Zhao M, You X, Zhang W, et al: A Ferroptosis-inducing and leukemic Cell-targeting drug nanocarrier formed by Redox-responsive cysteine polymer for acute myeloid leukemia therapy. ACS Nano. 17:3334–3345. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Yu X, Wang Y, Tan J, Li Y, Yang P, Liu X, Lai J, Zhang Y, Cai L, Gu Y, et al: Inhibition of NRF2 enhances the acute myeloid leukemia cell death induced by venetoclax via the ferroptosis pathway. Cell Death Discov. 10:352024. View Article : Google Scholar : PubMed/NCBI | |
|
Pardieu B, Pasanisi J, Ling F, Dal Bello R, Penneroux J, Su A, Joudinaud R, Chat L, Wu HC, Duchmann M, et al: Cystine uptake inhibition potentiates front-line therapies in acute myeloid leukemia. Leukemia. 36:1585–1595. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Lyu T, Li X and Song Y: Ferroptosis in acute leukemia. Chin Med J (Engl). 136:886–898. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Tsvetkov P, Coy S, Petrova B, Dreishpoon M, Verma A, Abdusamad M, Rossen J, Joesch-Cohen L, Humeidi R, Spangler RD, et al: Copper induces cell death by targeting lipoylated TCA cycle proteins. Science. 375:1254–1261. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Chen L, Min J and Wang F: Copper homeostasis and cuproptosis in health and disease. Signal Transduct Target Ther. 7:3782022. View Article : Google Scholar : PubMed/NCBI | |
|
Tian Z, Jiang S, Zhou J and Zhang W: Copper homeostasis and cuproptosis in mitochondria. Life Sci. 334:1222232023. View Article : Google Scholar : PubMed/NCBI | |
|
Tsvetkov P, Detappe A, Cai K, Keys HR, Brune Z, Ying W, Thiru P, Reidy M, Kugener G, Rossen J, et al: Mitochondrial metabolism promotes adaptation to proteotoxic stress. Nat Chem Biol. 15:681–689. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Zheng P, Zhou C, Lu L, Liu B and Ding Y: Elesclomol: A copper ionophore targeting mitochondrial metabolism for cancer therapy. J Exp Clin Cancer Res. 41:2712022. View Article : Google Scholar : PubMed/NCBI | |
|
Vo TTT, Peng TY, Nguyen TH, Bui TNH, Wang CS, Lee WJ, Chen YL, Wu YC and Lee IT: The crosstalk between copper-induced oxidative stress and cuproptosis: A novel potential anticancer paradigm. Cell Commun Signal. 22:3532024. View Article : Google Scholar : PubMed/NCBI | |
|
Zeng Y, Cao Y, Ren S, Zhang C, Liu J, Liu K, Wang Y, Chen H, Zhou F, Yang X, et al: Responsive ROS-Augmented prodrug hybridization nanoassemblies for multidimensionally synergitic treatment of hepatocellular carcinoma in cascade assaults. Adv Sci (Weinh). 12:e25014202025. View Article : Google Scholar : PubMed/NCBI | |
|
Cong Y, Li N, Zhang Z, Shang Y and Zhao H: Cuproptosis: Molecular mechanisms, cancer prognosis, and therapeutic applications. J Transl Med. 23:1042025. View Article : Google Scholar : PubMed/NCBI | |
|
Feng Y, Yang Z, Wang J and Zhao H: Cuproptosis: Unveiling a new frontier in cancer biology and therapeutics. Cell Commun Signal. 22:2492024. View Article : Google Scholar : PubMed/NCBI | |
|
Kahlson MA and Dixon SJ: Copper-induced cell death. Science. 375:1231–1232. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Wang Y, Zhang L and Zhou F: Cuproptosis: A new form of programmed cell death. Cell Mol Immunol. 19:867–868. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Dreishpoon MB, Bick NR, Petrova B, Warui DM, Cameron A, Booker SJ, Kanarek N, Golub TR and Tsvetkov P: FDX1 regulates cellular protein lipoylation through direct binding to LIAS. J Biol Chem. 299:1050462023. View Article : Google Scholar : PubMed/NCBI | |
|
Sun Z, Xu H, Lu G, Yang C, Gao X, Zhang J, Liu X, Chen Y, Wang K, Guo J and Li J: AKT1 Phosphorylates FDX1 to promote cuproptosis resistance in Triple-negative breast cancer. Adv Sci (Weinh). 12:e24081062025. View Article : Google Scholar : PubMed/NCBI | |
|
Yip NC, Fombon IS, Liu P, Brown S, Kannappan V, Armesilla AL, Xu B, Cassidy J, Darling JL and Wang W: Disulfiram modulated ROS-MAPK and NFκB pathways and targeted breast cancer cells with cancer stem cell-like properties. Br J Cancer. 104:1564–1574. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Valadbeigi S, Javadian S, Ebrahimi-Rad M, Khatami S and Saghiri R: Assessment of trace elements in serum of acute lymphoblastic and myeloid leukemia patients. Exp Oncol. 41:69–71. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Singh RP, Jeyaraju DV, Voisin V, Hurren R, Xu C, Hawley JR, Barghout SH, Khan DH, Gronda M, Wang X, et al: Disrupting mitochondrial copper distribution inhibits leukemic stem cell Self-renewal. Cell Stem Cell. 26:926–937.e10. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
McMahon A, Chen W and Li F: Old wine in new bottles: Advanced drug delivery systems for disulfiram-based cancer therapy. J Control Release. 319:352–359. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Weiser Drozdkova D and Smesny Trtkova K: Possible therapeutic potential of disulfiram for multiple myeloma. Curr Oncol. 28:2087–2096. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Solak K, Mavi A and Yılmaz B: Disulfiram-loaded functionalized magnetic nanoparticles combined with copper and sodium nitroprusside in breast cancer cells. Mater Sci Eng C Mater Biol Appl. 119:1114522021. View Article : Google Scholar : PubMed/NCBI | |
|
Solier S, Müller S, Cañeque T, Versini A, Mansart A, Sindikubwabo F, Baron L, Emam L, Gestraud P, Pantoș GD, et al: A druggable copper-signalling pathway that drives inflammation. Nature. 617:386–394. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Karsa M, Xiao L, Ronca E, Bongers A, Spurling D, Karsa A, Cantilena S, Mariana A, Failes TW, Arndt GM, et al: FDA-approved disulfiram as a novel treatment for aggressive leukemia. J Mol Med (Berl). 102:507–519. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Oliveri V: Selective targeting of cancer cells by copper ionophores: An overview. Front Mol Biosci. 9:8418142022. View Article : Google Scholar : PubMed/NCBI | |
|
Wang J, Luo C, Shan C, You Q, Lu J, Elf S, Zhou Y, Wen Y, Vinkenborg JL, Fan J, et al: Inhibition of human copper trafficking by a small molecule significantly attenuates cancer cell proliferation. Nat Chem. 7:968–979. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Cao C, Wang T, Luo Y, Zhang Y, Dai YY and Shen Y: Comprehensive analysis of cuproptosis-associated LncRNAs predictive value and related CeRNA network in acute myeloid leukemia. Heliyon. 9:e225322023. View Article : Google Scholar : PubMed/NCBI | |
|
Moison C, Gracias D, Schmitt J, Girard S, Spinella JF, Fortier S, Boivin I, Mendoza-Sanchez R, Thavonekham B, MacRae T, et al: SF3B1 mutations provide genetic vulnerability to copper ionophores in human acute myeloid leukemia. Sci Adv. 10:eadl40182024. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang J, Zheng Y, Liu H and Liu B: AC024896.1/miR-363-3p axis regulates the malignant progression of acute myeloid leukemia by Cuproptosis-related gene MYO1B. Blood Lymphat Cancer. 14:17–30. 2024.PubMed/NCBI | |
|
Huang X, Yan H, Xu Z, Yang B, Luo P and He Q: The inducible role of autophagy in cell death: Emerging evidence and future perspectives. Cell Commun Signal. 23:1512025. View Article : Google Scholar : PubMed/NCBI | |
|
Gao W, Wang X, Zhou Y, Wang X and Yu Y: Autophagy, ferroptosis, pyroptosis, and necroptosis in tumor immunotherapy. Signal Transduct Target Ther. 7:1962022. View Article : Google Scholar : PubMed/NCBI | |
|
Liu X, Tuerxun H and Zhao Y, Li Y, Wen S, Li X and Zhao Y: Crosstalk between ferroptosis and autophagy: Broaden horizons of cancer therapy. J Transl Med. 23:182025. View Article : Google Scholar : PubMed/NCBI | |
|
Newton K, Wickliffe KE, Maltzman A, Dugger DL, Webster JD, Guo H and Dixit VM: Caspase cleavage of RIPK3 after Asp333 is dispensable for mouse embryogenesis. Cell Death Differ. 31:254–262. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Fritsch M, Günther SD, Schwarzer R, Albert MC, Schorn F, Werthenbach JP, Schiffmann LM, Stair N, Stocks H, Seeger JM, et al: Caspase-8 is the molecular switch for apoptosis, necroptosis and pyroptosis. Nature. 575:683–687. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Qiu Y, Hüther JA, Wank B, Rath A, Tykwe R, Aldrovandi M, Henkelmann B, Mergner J, Nakamura T and Laschat S: Interplay of ferroptotic and apoptotic cell death and its modulation by BH3-mimetics. Cell Death Differ. Apr 29–2025.doi: 10.1038/s41418-025-01514-7 (Epub ahead of print). View Article : Google Scholar | |
|
Xu R, Wang W and Zhang W: Ferroptosis and the bidirectional regulatory factor p53. Cell Death Discov. 9:1972023. View Article : Google Scholar : PubMed/NCBI | |
|
Gao J, Xiong A, Liu J, Li X, Wang J, Zhang L, Liu Y, Xiong Y, Li G and He X: PANoptosis: Bridging apoptosis, pyroptosis, and necroptosis in cancer progression and treatment. Cancer Gene Ther. 31:970–983. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Qu M, Wang Y, Qiu Z, Zhu S, Guo K, Chen W, Miao C and Zhang H: Necroptosis, pyroptosis, ferroptosis in sepsis and treatment. Shock. 57:161–171. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang C and Liu N: Ferroptosis, necroptosis, and pyroptosis in the occurrence and development of ovarian cancer. Front Immunol. 13:9200592022. View Article : Google Scholar : PubMed/NCBI | |
|
Chen Y, Fang ZM, Yi X, Wei X and Jiang DS: The interaction between ferroptosis and inflammatory signaling pathways. Cell Death Dis. 14:2052023. View Article : Google Scholar : PubMed/NCBI | |
|
Li Y, Du Y, Zhou Y, Chen Q, Luo Z, Ren Y, Chen X and Chen G: Iron and copper: Critical executioners of ferroptosis, cuproptosis and other forms of cell death. Cell Commun Signal. 21:3272023. View Article : Google Scholar : PubMed/NCBI | |
|
Hu X, Li L, Nkwocha J, Kmieciak M, Shang S, Cowart LA, Yue Y, Horimoto K, Hawkridge A, Rijal A, et al: Src inhibition potentiates MCL-1 antagonist activity in acute myeloid leukemia. Signal Transduct Target Ther. 10:502025. View Article : Google Scholar : PubMed/NCBI | |
|
Ong F, Kim K and Konopleva MY: Venetoclax resistance: Mechanistic insights and future strategies. Cancer Drug Resist. 5:380–400. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Lim J and Murthy A: Targeting autophagy to treat cancer: Challenges and opportunities. Front Pharmacol. 11:5903442020. View Article : Google Scholar : PubMed/NCBI | |
|
Hassani S, Ghaffari P, Chahardouli B, Alimoghaddam K, Ghavamzadeh A, Alizadeh S and Ghaffari SH: Disulfiram/copper causes ROS levels alteration, cell cycle inhibition, and apoptosis in acute myeloid leukaemia cell lines with modulation in the expression of related genes. Biomed Pharmacother. 99:561–569. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Song F, Lin S, Xu T, Yang C, Sharavyn B, Naranmandura H, Zhang Y and Huang P: Targeted therapy in acute myeloid leukemia: Resistance and overcoming strategy. Drug Resist Updat. 83:1012862025. View Article : Google Scholar : PubMed/NCBI | |
|
D'Amico M and De Amicis F: Challenges of regulated cell death: Implications for therapy resistance in cancer. Cells. 13:10832024. View Article : Google Scholar : PubMed/NCBI | |
|
Tong X, Tang R, Xiao M, Xu J, Wang W, Zhang B, Liu J, Yu X and Shi S: Targeting cell death pathways for cancer therapy: Recent developments in necroptosis, pyroptosis, ferroptosis, and cuproptosis research. J Hematol Oncol. 15:1742022. View Article : Google Scholar : PubMed/NCBI |