Abstract
Disulfidptosis, a recently identified cell death mechanism, plays a pivotal role in the development, progression, and treatment of digestive tract tumors, including gastric cancer, hepatocellular cancer, esophageal cancer, colorectal cancer, pancreatic cancer, cholangiocarcinoma, and neuroendocrine tumors, which have high global incidence and mortality rates. Analyzing the expression of disulfidptosis-related gene expression within the tumor microenvironment enhances our understanding of tumor biology and facilitates novel diagnostic and therapeutic strategies. Research on immune infiltration and checkpoints can identify therapeutic targets linked to disulfidptosis, thereby improving immunotherapy efficacy. Targeting genes such as SLC7A11, which are essential for maintaining glutathione levels and regulating oxidative stress, may overcome chemoresistance and enhance existing treatments. Disulfidptosis could complement current therapies as it induces cytoskeletal collapse and selective tumor cell death, especially in chemoresistant cancers. Additionally, genes like SLC7A11, RPN1, and NCKAP1 in gastric cancer correlate with poor prognosis, highlighting their potential as prognostic biomarkers. Personalized medicine approaches utilizing disulfidptosis-related biomarkers could identify patients who would benefit from therapies targeting oxidative stress regulation, leading to more precise treatments and improved outcomes. This review summarizes disulfidptosis mechanisms, advancements in digestive cancers, and the potential of related genes for prognosis, immune response evaluation, and targeted therapies, providing novel perspectives for diagnosis and personalized treatment.
Similar content being viewed by others
Avoid common mistakes on your manuscript.
1 Introduction
Digestive tract tumors, including gastric cancer, esophageal cancer, hepatocellular carcinoma (HCC), colorectal cancer (CRC), pancreatic cancer, cholangiocarcinoma, and neuroendocrine tumors, represent a significant global health burden due to their high incidence and mortality rates. In China, the incidence and mortality of these cancers are notably higher than global averages [1, 2]. Despite advances in conventional therapies like surgery, radiotherapy, and chemotherapy, along with the emergence of immunotherapy and targeted therapies, the prognosis for many patients remains poor, particularly because of challenges in early diagnosis, treatment resistance, and tumor recurrence. Consequently, there is an urgent need for novel therapeutic strategies and a deeper understanding of the underlying molecular mechanisms driving cancer progression and treatment failure [3,4,5,6,7].
Disulfidptosis, a recently discovered regulated form of cell death, has emerged as an important player in oncology. First described by Liu et al., disulfidptosis is characterized by the accumulation of disulfide bonds under glucose-deprivation stress, particularly in cancer cells with high SLC7A11 expression. This form of cell death is driven by the depletion of NADPH, disrupting the cystine-to-cysteine reduction process, leading to disulfide stress, cytoskeletal collapse, and ultimately, cell death [8,9,10]. Tumor cells often overexpress SLC7A11 to maintain glutathione (GSH) levels and counteract oxidative stress, making this pathway a critical vulnerability in cancer cells [11,12,13]. Recent studies have demonstrated that disulfidptosis-related genes (DRGs) play key roles in metabolic reprogramming, immune modulation, and therapeutic resistance, influencing tumor progression and treatment responses in digestive tract cancers. For example, genes such as SLC7A11, RPN1, and NCKAP1 have been linked to immune infiltration, modulation of the tumor microenvironment (TME), and the regulation of chemoresistance [14,15,16,17]. Emerging research suggests that targeting disulfidptosis pathways could offer promising therapeutic interventions. Bioinformatics-driven studies highlight agents like Bosutinib and Sorafenib as potential candidates for clinical application [15]. This review aims to provide an updated synthesis of the latest advances in disulfidptosis research, focusing on its molecular mechanisms, clinical significance, and therapeutic potential, as well as its prognostic predictive effects in digestive tract tumors. By exploring the integration of DRGs into metabolic regulation and immune response modulation, we offer new perspectives on developing innovative therapies and personalized treatment strategies for these malignancies (Fig. 1).
2 The mechanisms of disulfidptosis
2.1 The mechanisms and key pathways of disulfidptosis
In the 1980s, researchers discovered that certain drugs and chemicals could cause the accumulation of disulfide bonds, leading to cell death. Recently, the Gan Boyi team characterized this novel form of cell death, naming it “disulfidptosis” [8]. Their research showed that cells with high SLC7A11 expression are more susceptible to death under glucose deprivation. This susceptibility is primarily due to reduced glycolysis under glucose-starved conditions, resulting in insufficient NADPH. Without adequate NADPH, cystine cannot be reduced to cysteine, causing an accumulation of disulfide bonds and inducing disulfide stress. Disulfide bonds interact with the actin cytoskeleton, and the cross-linking of actin with cytoskeletal proteins can cause the breakdown of the actin structure, ultimately resulting in cell death (Fig. 2). Genes promoting disulfidptosis include OXSM, GYS1, NUBPL, NDUFA11, LRPPRC, and NDUFS1, while inhibitory genes include SLC7A11, SLC3A2, RPN1, and NCKAP1.
The core pathway of disulfidptosis involves multiple key regulatory factors and complex biological processes. Current research indicates that it primarily affects the actin cytoskeleton via the Rac-WAVE regulatory complex (WRC), resulting in abnormal disulfide bond formation, which disrupts the actin network structure and induces cell death. Gene Ontology (GO) enrichment analysis has shown that, under glucose starvation conditions, disulfide bond processes are mainly enriched in biological processes related to the actin cytoskeleton and cell adhesion [8]. NCK Associated Protein 1 (NCKAP1), a critical component of the WRC, is essential for regulating its function, as inhibiting NCKAP1 leads to a marked reduction in disulfidptosis occurrence [18, 19]. Additionally, redox equilibrium is another important factor in disulfidptosis. Cells with elevated SLC7A11 expression are more vulnerable to disulfidptosis when exposed to hydrogen peroxide (H2O2) or upon inhibition of thioredoxin reductase 1(TXNRD1) [20,21,22]. Additionally, the NF-κB and JNK signaling pathways play a role in both the occurrence and progression of disulfidptosis [23, 24]. These discoveries reveal the intricate mechanisms of disulfidptosis, providing valuable insights into cell death and emphasizing the necessity for further exploration of the various biological processes involved.
2.2 The relationship between disulfidptosis and other forms of cell death
Cell death is a fundamental physiological process essential for the survival and development of all organisms. It is generally categorized into two types: programmed cell death (PCD) and accidental cell death (ACD). PCD is a tightly regulated process controlled by intracellular signaling pathways that determine cell fate, whereas ACD results from external physical or chemical damage without involving intrinsic signaling mechanisms. Recent research has identified several forms of cell death associated with metabolic imbalances, including nutrient deficiencies and the excessive accumulation of metals such as iron and copper. These forms, collectively referred to as metabolic cell death, include ferroptosis, cuproptosis, and disulfidptosis [8,9,10].
Ferroptosis, an iron-dependent form of cell death, is characterized by lipid peroxidation driven by the accumulation of reactive oxygen species (ROS). Key regulators of ferroptosis include glutathione (GSH) and glutathione peroxidase 4 (GPX4), both of which play central roles in mitigating oxidative damage (Fig. 3) [9]. SLC7A11, which is a critical component of the cystine/glutamate antiporter system (System xc⁻), mediates cystine uptake and its subsequent conversion to cysteine for GSH synthesis. This process serves not only as a key regulatory pathway for ferroptosis but also links ferroptosis to disulfidptosis. Under conditions of NADPH depletion, excessive cysteine accumulation promotes disulfide bond formation, thereby triggering disulfidptosis. Conversely, GSH depletion weakens antioxidant defenses, exacerbating ferroptosis. The dual role of SLC7A11 in both ferroptosis and disulfidptosis underscores its pivotal function in regulating cellular responses to metabolic stress. These findings highlight SLC7A11 as a central node connecting ferroptosis, disulfidptosis, and the cellular antioxidant system, providing valuable insights into metabolic cell death and its therapeutic potential in disease treatment (Fig. 4) [8, 9].
The diagram of the ferroptosis mechanism. The primary mechanism of ferroptosis involves the catalytic peroxidation of highly expressed unsaturated fatty acids on the cell membrane, driven by divalent iron or lipoxygenases, ultimately leading to cell death. Furthermore, this process is marked by a reduction in GPX4, a key regulatory enzyme of the antioxidant system, specifically the glutathione system. (1) GPX4 inactivation due to GSH depletion; (2) Direct inactivation of GPX4; (3) Iron ion uptake and reduction: the introduction of iron ions into the cell, ensuring the abundant presence of ferrous iron (Fe2⁺), which initiates lipid peroxidation through the Fenton reaction. (Created in BioRender.com)
The Relationship Between Disulfidptosis and Other Forms of Cell Death. (1) Under glucose deprivation, insufficient NADPH production leads to excessive accumulation of disulfide bonds within cells, thereby triggering disulfidptosis. (2) Excessive accumulation of ROS induces lipid peroxidation, which subsequently triggers ferroptosis. (3) GSH promotes the production of GPX4, maintaining intracellular redox balance, suppressing ROS production, and consequently preventing ferroptosis. (4) Excessive accumulation of copper ions (Cu⁺) generates large amounts of ROS via the Fenton reaction, inducing cuproptosis. (5) GSH chelates copper ions (Cu⁺), thereby inhibiting the occurrence of cuproptosis. (6) Copper ions (Cu2⁺) induce autophagic degradation of GPX4, further exacerbating ferroptosis. (7) ROS depletes antioxidant defense systems (including GSH and NADPH), further aggravating the occurrence of disulfidptosis. ROS: Reactive oxygen species; NADPH: Nicotinamide Adenine Dinucleotide Phosphate; GPX4: Glutathione peroxidase 4; GSH: Glutathione; System xc⁻: The cystine/glutamate antiporter system (By Figdraw)
Cuproptosis is a form of cell death triggered by intracellular copper ion overload, primarily mediated through oxidative stress and subsequent cellular damage. Excess copper ions participate in the Fenton reaction, generating large amounts of ROS. These ROS induce lipid peroxidation, protein oxidation, and DNA damage, and ultimately disrupting normal cellular functions [10]. Research on cuproptosis has revealed that copper ions target the acylation of proteins involved in the tricarboxylic acid (TCA) cycle, thus inducing cell death and impairing the function of intracellular enzymes and other proteins. Unlike ferroptosis and disulfidptosis, SLC7A11 does not directly contribute to copper ion accumulation or mitochondrial dysfunction. However, it indirectly influences cuproptosis by maintaining GSH-mediated antioxidant defenses. When the GSH defense system is compromised, ROS clearance becomes inefficient, making cells more vulnerable to copper-induced oxidative damage and accelerating the progression of cuproptosis (Fig. 4).
Both ferroptosis and cuproptosis depend on GSH to maintain redox balance. Additionally, copper can exacerbate ferroptosis by inducing the autophagic degradation of GPX4, which creates a feedback loop between these two pathways. Furthermore, copper-induced ROS also impact both ferroptosis and disulfidptosis, positioning ROS as a central mediator across these cell death pathways. The interplay among these three forms of cell death suggests that they share common regulatory factors and interact within complex cellular networks, particularly under conditions of oxidative stress and metal ion imbalance. This interdependence highlights the therapeutic potential of targeting these pathways. For example, therapies aimed at depleting GSH—commonly employed to target oxidative stress in cancer—can sensitize cells to both ferroptosis and cuproptosis, offering novel treatment strategies. Understanding how these pathways converge through key regulators such as SLC7A11 and GPX4 can aid in the development of more precise and effective therapies, reducing off-target effects and improving efficacy (Fig. 4) [8,9,10, 25]. The relevance of these interactions in cancer treatment is increasingly recognized. For instance, a study demonstrated that the Disulfidptosis-related Ferroptosis (DRF) score, which incorporates genes such as SLC7A11, can predict prognosis and treatment response in hepatocellular carcinoma (HCC). Patients with low DRF scores showed significantly better outcomes, emphasizing the relationship between disulfidptosis and ferroptosis in cancer therapy. These insights provide new strategies for targeting metabolic pathways to address ROS and metal ion imbalances in cancer and other oxidative stress-related diseases [26].
3 Applications of disulfidptosis in digestive tract tumors
3.1 Expression of disulfidptosis-related genes in digestive tract tumors
Recent studies have identified a significant correlation between disulfidptosis-related genes (DRGs) and both the occurrence and prognosis of various digestive tract cancers. The distribution of DRGs with high aberrant expression in digestive tract tumors is detailed in (Table 1). In gastric cancer, genes such as SLC3A2, RPN1, NCKAP1, SLC7A11, PDLIM1, GLA, HIF-1α, VPS35, and CDC37 are overexpressed compared to normal tissues. Conversely, genes like DSTN, FLNA, MYH10, and MYL6 are underexpressed. These genes contribute to tumor proliferation, invasion, and migration [14,15,16,17]. In hepatocellular carcinoma (HCC), DRGs including SLC7A11, INF2, CD2AP, FLNB, ACTN4, CAPZB, ACTB, PDLIM1, FLNA, MYL6, TLN1, and DSTN show high expression levels, whereas ENO1, AGRN, and ZBTB7A exhibit low mutation frequencies. Research by Zhijian Wang’s team has demonstrated that both disulfidptosis-related and glycolysis-related genes are abnormally expressed in HCC, underscoring the critical role of disulfidptosis genes in HCC development [27, 28]. In esophageal cancer, Liu F et al. identified 443 DRGs with abnormal expression. Notably, CD96, CXCL13, IL2RG, LY96, TPK1, and ACAP1 levels are elevated, while SOX17 levels are decreased. These genes are enriched in pathways such as leukocyte adhesion, positive regulation of T cell activation, peptidase activity, and suppression of hydrolase activity [29]. Pancreatic cancer exhibits elevated expression of DRGs including SLC7A11, G6PD, PGD, PRDX1, FLNA, MYH9, TLN1, ACTB, MYH10, SLC3A2, RPN1, NCKAP1, NCKAP1L, WASF2, CYFIP1, ABI2, BAK1, and RAC1 compared to normal tissues [30,31,32]. In colon cancer, Hu G’s team identified 105 genes closely associated with disulfidptosis. Specifically, OXSM is significantly downregulated, while TRIP6, MYH3, and MYH4 are upregulated [33].
The abnormal expression of DRGs exhibits significant commonalities across digestive tract tumors. Highly expressed genes such as SLC3A2, RPN1, and SLC7A11 are strongly associated with increased tumor cell proliferation, invasion, and motility. In contrast, genes with low expression levels are frequently linked to reduced cellular structural integrity and weakened adhesion capabilities [14,15,16,17, 27, 28]. These DRGs play critical roles in regulating tumor development, progression, and treatment response by modulating key biological processes, including antioxidant systems, metabolic pathways, cytoskeletal dynamics, and signal transduction.
3.2 Applications of disulfidptosis in the treatment of digestive tract tumors
Disulfidptosis, a novel form of regulated cell death, has recently garnered attention for its potential in treating digestive tract tumors. However, conventional cancer therapies, such as chemotherapy and radiotherapy, are hindered by significant limitations. These treatments lack specificity, often damaging both tumor and healthy cells, leading to severe side effects, including gastrointestinal toxicity, neutropenia, and fatigue. As a result, the doses of these therapies that can be safely administered are restricted, particularly in cancers of the gastrointestinal tract, such as gastric, colorectal, and pancreatic cancers. Additionally, many tumors, including those in the digestive system, develop resistance to chemotherapy, complicating long-term remission and effective treatment. While immunotherapy holds promise, it also faces challenges, with certain cancers—such as hepatocellular carcinoma and colorectal cancer—evading immune surveillance through the expression of checkpoint molecules like PD-L1 and CTLA-4, leading to resistance. Tumor heterogeneity further complicates personalized treatment approaches. In light of these obstacles, disulfidptosis, induced by disulfide bond accumulation and SLC7A11 overexpression, emerges as a promising therapeutic target. This novel cell death mechanism offers the potential to complement existing therapies, reducing the toxicities typically associated with conventional treatments in digestive tract tumors.
3.2.1 Targeting disulfidptosis-related genes: a pathway to precision therapy in digestive tract tumors
The dysregulation of disulfidptosis-related genes (DRGs) and their associated pathways in digestive tract tumors presents promising opportunities for targeted therapeutic strategies. In gastric cancer (GC), Filamin A (FLNA) promotes tumor invasion and migration by degrading matrix metalloproteinase 9 (MMP-9). Additionally, solute carrier family 3 member 2 (SLC3A2) enhances tumor cell proliferation and survival. Ribophorin I (RPN1) and Nck-associated protein 1 (NCKAP1) similarly facilitate tumor invasion and migration through cytoskeletal regulation. In cancers with high SLC7A11 expression, glucose transporter inhibitors such as BAY-876 and KL-11743 effectively block glucose uptake, deplete intracellular NADPH levels, and induce disulfidptosis [16, 34]. Furthermore, Collagen type X alpha 1 chain (Col10A1) drives GC progression by remodeling the extracellular matrix (ECM) and activating the MEK/ERK signaling pathways [35,36,37,38,39]. CD24, a mucin-like membrane glycoprotein, regulates the epidermal growth factor receptor (EGFR) signaling pathway by inhibiting EGFR endocytosis and degradation through a RhoA-dependent mechanism [40, 41]. Neuropilin-1 (NRP1), a transmembrane glycoprotein receptor, plays a pivotal role in tumor angiogenesis and metastasis by interacting with vascular endothelial growth factor (VEGF) and semaphorins. Experimental studies demonstrate that NRP1 knockdown significantly suppresses GC cell proliferation and autophagy, potentially through the modulation of the Wnt/β-catenin signaling pathway and induction of disulfidptosis. Additionally, silencing NRP1 reduces glutamine uptake in GC cells, impairing energy production and biosynthesis. These findings suggest that NRP1 regulates glutamine transporters or metabolic enzymes, highlighting its dual role in angiogenesis and glutamine metabolism. They also underscore its therapeutic potential in GC [42,43,44,45,46]. In colorectal adenocarcinoma (COAD), POU4F1 has been identified as a key oncogenic driver that promotes tumor proliferation and migration through the ERK1/2 and MAPK pathways while enhancing sensitivity to disulfidptosis [47]. Perilipin 4 (PLIN4) regulates mitochondrial β-oxidation, generating ATP and reactive oxygen species (ROS), which may contribute to tissue damage [48,49,50]. In hepatocellular carcinoma (HCC), MYH9 (myosin heavy chain 9) has been shown to overcome sorafenib resistance by promoting disulfidptosis. Inhibition of MYH9 disrupts cytoskeletal integrity, enhances sorafenib uptake, and improves antitumor efficacy, establishing MYH9 as a critical regulator of tumor drug sensitivity [51,52,53,54].
Collectively, these findings underscore the therapeutic potential of DRGs in digestive tract tumors [8, 55]. With further validation through in vitro, in vivo, and clinical trials, key disulfidptosis-related genes could become essential targets for developing targeted therapies for digestive tract tumors.
3.2.2 Disulfidptosis-driven drug sensitivity: advancing therapies for digestive tract cancers
Integrating disulfidptosis-related gene expression patterns with bioinformatics-based drug sensitivity analyses, utilizing IC50 values, has enabled researchers to identify optimal chemotherapeutic agents for specific digestive tract tumors. These findings offer valuable insights for tailoring individualized treatment strategies.
For instance, in gastric cancer (GC), Qian Li et al. identified 89 drugs associated with treatment sensitivity, including Bosutinib and Blebbistatin. Notably, Blebbistatin enhances the efficacy of paclitaxel, while Bosutinib inhibits GC cell migration, highlighting their therapeutic potential [15]. Furthermore, Jie Li et al. demonstrated that high-risk GC patients exhibit resistance to conventional agents such as 5-Fluorouracil, Docetaxel, Erlotinib, Methotrexate, and Paclitaxel [16]. Conversely, Xing Liu et al. reported that low-risk patients respond favorably to agents like Bexarotene, Bicalutamide, Bortezomib, Dasatinib, and Imatinib. Interestingly, high-risk patients show increased sensitivity to Gemcitabine, Gefitinib, Bosutinib, Sorafenib, and Vorinostat [17]. Despite variations in study findings, Bosutinib consistently emerges as a promising therapeutic candidate, warranting further preclinical and clinical validation. In hepatocellular carcinoma (HCC), disulfidptosis-related drug sensitivity analysis revealed that patients with poor prognoses are more sensitive to chemotherapeutic agents such as Gemcitabine, Paclitaxel, and Vinorelbine [28, 56,57,58]. These findings underscore the potential of stratifying patients based on disulfidptosis-related pathways to enhance treatment outcomes. Further exploration of the relationship between disulfidptosis-related gene expression and therapeutic efficacy could optimize HCC management. Additionally, a clinical model of DRG/DRL in HCC identified AC026412.3 as a critical adverse prognostic factor linked to drug resistance and tumor proliferation. Silencing AC026412.3 enhanced the efficacy of gefitinib, highlighting its potential as a promising therapeutic target [59]. In pancreatic adenocarcinoma (PAAD), studies have identified sensitivity to agents such as Bortezomib, Dasatinib, and Cisplatin [30,31,32]. However, these findings are currently limited to preclinical analyses, emphasizing the need for further experimental validation. The potential of disulfidptosis-related pathways to improve therapeutic efficacy in PAAD highlights their importance as a focus for future research. In colorectal cancer (CRC), analyses of disulfidptosis-related ferroptosis genes indicate that high-risk patients are more sensitive to Sorafenib, Pazopanib, and Lapatinib. These findings support the development of personalized chemotherapy regimens for CRC [48]. Additionally, natural compounds such as soy isoflavones and epigallocatechin gallate (EGCG) have demonstrated potential to promote disulfidptosis by targeting glucose transporters (GLUT), reducing glucose uptake, and inhibiting tumor growth [60,61,62,63,64]. These compounds hold promise as adjuvants to existing chemotherapeutic agents.
Disulfidptosis-related pathways provide a robust foundation for advancing personalized therapies across various malignancies. By integrating gene expression profiles with drug sensitivity data, researchers have identified both conventional chemotherapeutics and emerging compounds as potential treatments for digestive tract tumors. Promising candidates such as Bosutinib, Sorafenib, and natural compounds like EGCG exhibit significant therapeutic potential; however, further clinical validation is essential. Future research should focus on refining patient stratification and leveraging disulfidptosis-related pathways to develop innovative and effective cancer therapies tailored to individual patient needs.
3.2.3 Disulfidptosis and immune infiltration: implications for immunotherapy in digestive tract tumors
Immune infiltration plays a critical role in shaping the tumor microenvironment (TME), profoundly influencing tumor progression, therapeutic resistance, and clinical outcomes. Exploring the relationship between immune infiltration and disulfidptosis provides valuable insights for identifying therapeutic targets, predicting patient outcomes, and designing personalized treatment strategies.
Recent studies have highlighted the impact of disulfidptosis on immune cell infiltration and function within the TME. For example, Liao Z et al. demonstrated that high-risk disulfidptosis-related gene (DRG) expression profiles in gastric cancer (GC) are associated with increased infiltration of memory B cells, plasma cells, and activated mast cells. In contrast, low-risk patients exhibited higher levels of γδ T cells, M2 macrophages, and resting mast cells [65]. Additionally, GC patients with high ACTB expression showed greater immune cell infiltration compared to those with low ACTB expression [34]. The expression of NCKAP1 was linked to the presence of M2 macrophages, T helper (Th) cells, central memory T cells (TCM), and dendritic cells (DCs). Moreover, SLC7A11 expression correlated with regulatory T cells (Treg), Th2 cells, and neutrophils. RPN1, SLC3A2, and SLC7A11 were found to positively activate mast cells, while RPN1 and SLC3A2 negatively correlated with memory B cells [14, 16]. In hepatocellular carcinoma (HCC), elevated DRG expression is associated with increased levels of induced regulatory T cells (iTregs), macrophages, natural killer (NK) cells, and T cells, along with a reduction in neutrophil levels. Patients with poor prognosis exhibited heightened infiltration of immune cells such as Tregs, Th1 and Th2 cells, immature dendritic cells (iDCs), neutrophils, M1 macrophages, CD4 + T cells, and plasma cells. Conversely, these patients showed lower levels of CD8 + T cells, naive CD4 + T cells, CD4 + central memory T cells (Tcm), M2 macrophages, and CD8 + effector memory T cells (Tem), along with a decreased CD8 + /Treg ratio. These immune profiles contribute to immunotherapy resistance and unfavorable outcomes. Interestingly, high-risk HCC patients may demonstrate improved responses to immune checkpoint inhibitors (ICIs) because of heightened immune activation within the TME, despite the presence of immunosuppressive mechanisms [58]. Notably, SLC7A11 regulates both glucose and cysteine uptake while modulating immune responses, highlighting its critical role in metabolic adaptation and immune regulation [66]. SLC7A11 shows positive associations with various immune cell types, including Th cells, macrophages, and NK cells, particularly correlating strongly with T helper cells and NK CD56 + cells. The expression of immune checkpoint molecules further illustrates the complexity of immune regulation in HCC. Molecules such as CD40 ligand (CD40LG), CD48, IDO1, CD27, and PDCD1 are highly expressed in patients with better prognoses, whereas CD276 expression is elevated in those with poor outcomes. These findings suggest that immune checkpoints play a critical role in tumor progression and may serve as promising therapeutic targets, particularly in low-risk populations [27, 28, 56,57,58, 67, 68]. In pancreatic adenocarcinoma (PAAD), Li et al. found that disulfidptosis influences immune infiltration, as Tr1 cells positively correlating with DRG expression and neutrophils showing an inverse correlation. Five key DRGs—S100A4, SLC7A11, PRDX1, SLC7A7, and DIAPH3—have been identified as potential regulators of the TME, impacting Th2 cells, macrophages, and plasmacytoid dendritic cells (pDCs). These findings underscore the potential of targeting DRGs to reprogram the TME [31]. Similarly, Yang et al. demonstrated in colorectal cancer (CRC) that tumor-associated macrophages (TAMs) and myeloid-derived suppressor cells (MDSCs) are enriched in high-risk disulfidptosis subtypes. These cells foster an immunosuppressive TME by secreting cytokines and activating immune checkpoints, thus suppressing T cell responses and facilitating tumor progression [69].
Immune checkpoint inhibitors (ICIs) targeting PD-1, PD-L1, and CTLA-4 have revolutionized cancer treatment. In disulfidptosis-associated subtypes, high expression of PD-L1 and CTLA-4 correlates with poor prognosis but also indicates that disulfidptosis may induce immunogenic cell death (ICD), converting “cold” tumors into “hot” tumors that are more responsive to ICIs. Furthermore, the elevated expression of HLA family genes in high-risk subtypes reinforces their potential as biomarkers for predicting immunotherapy responses. The dual role of disulfidptosis in regulating tumor metabolism and immune responses establishes DRGs as promising targets for precision immunotherapy. Combining ICIs targeting PD-L1, PD-1, or CTLA-4 with therapies aimed at DRG-mediated pathways, such as SLC7A11 inhibition, may enhance immune activation and overcome resistance [66]. By systematically analyzing the relationship between immune infiltration and disulfidptosis in digestive tract tumors, researchers can gain a deeper understanding of the dynamic changes within the TME. This knowledge can guide the development of more precise and effective immunotherapies, ultimately improving patient outcomes and extending survival rates.
3.3 Applications of disulfidptosis in prognosis of digestive tract tumors
Disulfidptosis, a recently identified form of regulated cell death driven by the accumulation of disulfide bonds under stress conditions, has emerged as a promising prognostic factor in digestive tract cancers. Studies have demonstrated that the expression profiles of disulfidptosis-related genes (DRGs) are closely associated with clinical outcomes. For instance, in gastric cancer (GC), high expression levels of GLA, CDC37, VAMP7, ALG1, and ANKZF1 are linked to favorable prognosis. In contrast, elevated levels of CD24, MAGE-A3, SERPINE1, IRGM, NRP1, HIF-1α, VPS35, PLS3, GRP, APOD, SGCE, COL8A1, NCKAP1, VCAN, NT5E, and SLC7A11 are associated with poor prognosis and advanced clinical stages [14,15,16, 40, 65]. In hepatocellular carcinoma (HCC), DRG expression exhibits significant prognostic variability. High expression levels of CBR4, SEC31B, SPP2, RDH16, LCAT, TRIM55, GHR, OGN, TCP10L, and DNASE1L3 are associated with improved prognosis. Conversely, elevated levels of GNL2, NDRG1, TMCO3, TRIB3, SLC7A11, LRPPRC, CDCA8, GAGE1, PPP2R2C, TNFRSF11B, IL8, TREM1, SLC2A1, SCIN, AKR1B15, MMP1, CORIN, SLC1A5, GAGE4, and NEIL3 correlate with poor outcomes (Table 2) [27, 56, 57, 67, 68]. Similarly, in colorectal adenocarcinoma (COAD), POU4F1 has been identified as a critical prognostic marker, with its overexpression significantly correlating with reduced survival rates [47].
These findings highlight the potential of integrating DRG expression profiles with clinical tools to establish a robust framework for individualized cancer prognosis and treatment strategies. Key biomarkers such as SLC7A11, POU4F1, CD24, and NRP1 show considerable promise in enhancing prognostic accuracy and guiding targeted therapies. Compared to single-gene analyses, risk scores that incorporate multiple DRGs demonstrate superior predictive capability, providing more comprehensive insights for clinical evaluation. Furthermore, combining these risk scores with clinical features, laboratory findings, and imaging data could facilitate the development of multidimensional prognostic models. Such approaches have the potential to enable precise and personalized cancer therapies, thereby advancing the broader field of oncology.
4 Conclusions and future perspectives
In conclusion, disulfidptosis, a recently identified form of regulated cell death, plays a crucial role in the development and treatment of digestive tract tumors. This review underscores the therapeutic and prognostic potential of disulfidptosis, particularly through key genes such as SLC7A11, SLC3A2, NCKAP1, and NRP1, which serve as significant biomarkers and therapeutic targets. These genes influence tumor progression, immune modulation, and therapeutic resistance, providing novel insights into treatment strategies. The unique focus of this review is on emphasizing disulfidptosis’ role in overcoming chemoresistance and enhancing immunotherapy efficacy, offering new therapeutic avenues for treating difficult-to-target cancers.
Future research should give priority to validating the functional roles of these genes in various models and conducting large-scale clinical trials to confirm their predictive value as biomarkers for patient stratification and treatment response. Further studies into the interactions between disulfidptosis and other cell death pathways, such as ferroptosis and cuproptosis, will deepen our understanding of their collective impact on tumor progression and therapy. This exploration is crucial for developing combination therapies that optimize the benefits of each mechanism.
Additionally, identifying additional regulators of disulfidptosis and exploring its molecular pathways may lead to new therapeutic strategies. For instance, small molecule inhibitors targeting oxidative stress pathways or disulfide bond formation could not only induce disulfidptosis but also sensitize cancer cells to conventional therapies. Combining disulfidptosis induction with chemotherapy or immunotherapy has the potential to improve treatment outcomes, particularly in drug-resistant tumors. Investigating the role of immune checkpoint inhibitors in modulating disulfidptosis may provide new opportunities for combination immunotherapies.
In summary, advancing disulfidptosis research through interdisciplinary collaboration and technological innovation is essential for developing more precise diagnostic tools and therapeutic approaches. By exploring the complex relationships between disulfidptosis, immune infiltration, and tumor progression, we can enable the development of personalized treatment strategies, improving patient outcomes and the quality of life for individuals with digestive tract cancers.
Data availability
No datasets were generated or analysed during the current study.
References
He J, Zhu YJ, Fan XK, et al. Progress in systematic epidemiological studies on the risk of digestive tract tumor incidence. Chin J Oncol. 1–8.
Huang J, Lucero-Prisno DE 3rd, Zhang L, Xu W, Wong SH, Ng SC, Wong MCS. Updated epidemiology of gastrointestinal cancers in East Asia. Nat Rev Gastroenterol Hepatol. 2023;20(5):271–87. https://doi.org/10.1038/s41575-022-00726-3.
Yang YM, Hong P, Xu WW, He QY, Li B. Advances in targeted therapy for esophageal cancer. Signal Transduct Target Ther. 2020;5(1):229. https://doi.org/10.1038/s41392-020-00323-3.
Joshi SS, Badgwell BD. Current treatment and recent progress in gastric cancer. CA Cancer J Clin. 2021;71(3):264–79. https://doi.org/10.3322/caac.21657.
Ganesan P, Kulik LM. Hepatocellular carcinoma: new developments. Clin Liver Dis. 2023;27(1):85–102. https://doi.org/10.1016/j.cld.2022.08.004.
Biller LH, Schrag D. Diagnosis and treatment of metastatic colorectal cancer: a review. JAMA. 2021;325(7):669–85. https://doi.org/10.1001/jama.2021.0106.
Wood LD, Canto MI, Jaffee EM, Simeone DM. Pancreatic cancer: pathogenesis, screening, diagnosis, and treatment. Gastroenterology. 2022;163(2):386-402.e1. https://doi.org/10.1053/j.gastro.2022.03.056.
Liu X, Nie L, Zhang Y, Yan Y, Wang C, Colic M, Olszewski K, Horbath A, Chen X, Lei G, Mao C, Wu S, Zhuang L, Poyurovsky MV, James You M, Hart T, Billadeau DD, Chen J, Gan B. Actin cytoskeleton vulnerability to disulfide stress mediates disulfidptosis. Nat Cell Biol. 2023;25(3):404–14. https://doi.org/10.1038/s41556-023-01091-2.
Mou Y, Wang J, Wu J, He D, Zhang C, Duan C, Li B. Ferroptosis, a new form of cell death: opportunities and challenges in cancer. J Hematol Oncol. 2019;12(1):34. https://doi.org/10.1186/s13045-019-0720-y.
Tsvetkov P, Coy S, Petrova B, Dreishpoon M, Verma A, Abdusamad M, Rossen J, Joesch-Cohen L, Humeidi R, Spangler RD, Eaton JK, Frenkel E, Kocak M, Corsello SM, Lutsenko S, Kanarek N, Santagata S, Golub TR. Copper induces cell death by targeting lipoylated TCA cycle proteins. Science. 2022;375(6586):1254–61. https://doi.org/10.1126/science.abf0529.
Chen Q, Zheng W, Guan J, Liu H, Dan Y, Zhu L, Song Y, Zhou Y, Zhao X, Zhang Y, Bai Y, Pan Y, Zhang J, Shao C. SOCS2-enhanced ubiquitination of SLC7A11 promotes ferroptosis and radiosensitization in hepatocellular carcinoma. Cell Death Differ. 2023;30(1):137–51. https://doi.org/10.1038/s41418-022-01051-7.
Zhang W, Sun Y, Bai L, Zhi L, Yang Y, Zhao Q, Chen C, Qi Y, Gao W, He W, Wang L, Chen D, Fan S, Chen H, Piao HL, Qiao Q, Xu Z, Zhang J, Zhao J, Zhang S, et al. RBMS1 regulates lung cancer ferroptosis through translational control of SLC7A11. J Clin Investig. 2021;131(22):e152067. https://doi.org/10.1172/JCI152067.
Yang J, Zhou Y, Xie S, Wang J, Li Z, Chen L, Mao M, Chen C, Huang A, Chen Y, Zhang X, Khan NUH, Wang L, Zhou J. Metformin induces ferroptosis by inhibiting UFMylation of SLC7A11 in breast cancer. J Exp Clin Cancer Res CR. 2021;40(1):206. https://doi.org/10.1186/s13046-021-02012-7.
Yan J, Fang Z, Shi M, Tu C, Zhang S, Jiang C, Li Q, Shao Y. Clinical significance of disulfidptosis-related genes and functional analysis in gastric cancer. J Cancer. 2024;15(4):1053–66. https://doi.org/10.7150/jca.91796.
Li Q, Yin LK. Comprehensive analysis of disulfidptosis related genes and prognosis of gastric cancer. World J Clin Oncol. 2023;14(10):373–99. https://doi.org/10.5306/wjco.v14.i10.373.
Li J, Yu T, Sun J, Ma M, Zheng Z, He Y, Kang W, Ye X. Integrated analysis of disulfidptosis-related immune genes signature to boost the efficacy of prognostic prediction in gastric cancer. Cancer Cell Int. 2024;24(1):112. https://doi.org/10.1186/s12935-024-03294-5.
Liu X, Ou J. The development of prognostic gene markers associated with disulfidptosis in gastric cancer and their application in predicting drug response. Heliyon. 2024;10(4): e26013. https://doi.org/10.1016/j.heliyon.2024.e26013.
Kunda P, Craig G, Dominguez V, Baum B. Abi, Sra1, and Kette control the stability and localization of SCAR/WAVE to regulate the formation of actin-based protrusions. Curr Biol CB. 2003;13(21):1867–75. https://doi.org/10.1016/j.cub.2003.10.005.
Miki H, Suetsugu S, Takenawa T. WAVE, a novel WASP-family protein involved in actin reorganization induced by Rac. EMBO J. 1998;17(23):6932–41. https://doi.org/10.1093/emboj/17.23.6932.
Zhong Z, Zhang C, Ni S, Ma M, Zhang X, Sang W, Lv T, Qian Z, Yi C, Yu B. NFATc1-mediated expression of SLC7A11 drives sensitivity to TXNRD1 inhibitors in osteoclast precursors. Redox Biol. 2023;63: 102711. https://doi.org/10.1016/j.redox.2023.102711.
Meyer Y, Belin C, Delorme-Hinoux V, Reichheld JP, Riondet C. Thioredoxin and glutaredoxin systems in plants: molecular mechanisms, crosstalks, and functional significance. Antioxid Redox Signal. 2012;17(8):1124–60. https://doi.org/10.1089/ars.2011.4327.
Al-Yafee YA, Al-Ayadhi LY, Haq SH, El-Ansary AK. Novel metabolic biomarkers related to sulfur-dependent detoxification pathways in autistic patients of Saudi Arabia. BMC Neurol. 2011;11:139. https://doi.org/10.1186/1471-2377-11-139.
Ji PY, Li ZY, Wang H, Dong JT, Li XJ, Yi HL. Arsenic and sulfur dioxide co-exposure induce renal injury via activation of the NF-κB and caspase signaling pathway. Chemosphere. 2019;224:280–8. https://doi.org/10.1016/j.chemosphere.2019.02.111.
Brancaccio M, Milito A, Viegas CA, Palumbo A, Simes DC, Castellano I. First evidence of dermo-protective activity of marine sulfur-containing histidine compounds. Free Radical Biol Med. 2022;192:224–34. https://doi.org/10.1016/j.freeradbiomed.2022.09.017.
Mao C, Wang M, Zhuang L, Gan B. Metabolic cell death in cancer: ferroptosis, cuproptosis, disulfidptosis, and beyond. Protein Cell. 2024;15(9):642–60. https://doi.org/10.1093/procel/pwae003.
Zhang C, Xu T, Ji K, Cao S, Ai J, Pan J, Cao Y, Yang Y, Jing L, Sun JH. Development and experimental validation of a machine learning-based disulfidptosis-related ferroptosis score for hepatocellular carcinoma. Apoptosis. 2024;29(1–2):103–20. https://doi.org/10.1007/s10495-023-01900-x.
Yang T, Liu J, Liu F, Lei J, Chen S, Ma Z, Ke P, Yang Q, Wen J, He Y, Duan J, Zeng X. Integrative analysis of disulfidptosis and immune microenvironment in hepatocellular carcinoma: a putative model and immunotherapeutic strategies. Front Immunol. 2024;14:1294677. https://doi.org/10.3389/fimmu.2023.1294677.
Wang Z, Chen X, Zhang J, Chen X, Peng J, Huang W. Based on disulfidptosis-related glycolytic genes to construct a signature for predicting prognosis and immune infiltration analysis of hepatocellular carcinoma. Front Immunol. 2023;14:1204338. https://doi.org/10.3389/fimmu.2023.1204338.
Liu F, Yuan D, Liu X, Zhuo S, Liu X, Sheng H, Sha M, Ye J, Yu H. A demonstration based on multi-omics transcriptome sequencing data revealed disulfidptosis heterogeneity within the tumor microenvironment of esophageal squamous cell carcinoma. Discover Oncol. 2023;14(1):96. https://doi.org/10.1007/s12672-023-00711-5.
Xiong Y, Kong X, Mei H, Wang J, Zhou S. Bioinformatics-based analysis of the relationship between disulfidptosis and prognosis and treatment response in pancreatic cancer. Sci Rep. 2023;13(1):22218. https://doi.org/10.1038/s41598-023-49752-4.
Li Y, Chen MX, Li HT, Cai XM, Chen B, Xie ZF. Comprehensive analysis based on the disulfidptosis-related genes identifies hub genes and immune infiltration for pancreatic adenocarcinoma. Open Med (Warsaw, Poland). 2024;19(1):20240906. https://doi.org/10.1515/med-2024-0906.
Wu Y, Shang J, Ruan Q, Tan X. Integrated single-cell and bulk RNA sequencing in pancreatic cancer identifies disulfidptosis-associated molecular subtypes and prognostic signature. Sci Rep. 2023;13(1):17577. https://doi.org/10.1038/s41598-023-43036-7.
Hu G, Yao H, Wei Z, Li L, Yu Z, Li J, Luo X, Guo Z. A bioinformatics approach to identify a disulfidptosis-related gene signature for prognostic implication in colon adenocarcinoma. Sci Rep. 2023;13(1):12403. https://doi.org/10.1038/s41598-023-39563-y.
Zhang P, Chen Z, Lin X, Yu S, Yu X, Chen Z. Unravelling diagnostic clusters and immune landscapes of disulfidptosis patterns in gastric cancer through bioinformatic assay. Aging. 2023;15(24):15434–50.
Liu Z, Sun L, Zhu W, Zhu J, Wu C, Peng X, Tian H, Huang C, Zhu Z. Disulfidptosis signature predicts immune microenvironment and prognosis of gastric cancer. Biol Direct. 2024;19(1):65. https://doi.org/10.1186/s13062-024-00518-6.
Wang X, Bai Y, Zhang F, Li D, Chen K, Wu R, Tang Y, Wei X, Han P. Prognostic value of COL10A1 and its correlation with tumor-infiltrating immune cells in urothelial bladder cancer: a comprehensive study based on bioinformatics and clinical analysis validation. Front Immunol. 2023;14: 955949. https://doi.org/10.3389/fimmu.2023.955949.
Zhou W, Li Y, Gu D, Xu J, Wang R, Wang H, Liu C. High expression COL10A1 promotes breast cancer progression and predicts poor prognosis. Heliyon. 2022;8(10): e11083. https://doi.org/10.1016/j.heliyon.2022.e11083.
Wen Z, Sun J, Luo J, Fu Y, Qiu Y, Li Y, Xu Y, Wu H, Zhang Q. COL10A1-DDR2 axis promotes the progression of pancreatic cancer by regulating MEK/ERK signal transduction. Front Oncol. 2022;12:1049345. https://doi.org/10.3389/fonc.2022.1049345.
Liang Y, Xia W, Zhang T, Chen B, Wang H, Song X, Zhang Z, Xu L, Dong G, Jiang F. Upregulated collagen COL10A1 remodels the extracellular matrix and promotes malignant progression in lung adenocarcinoma. Front Oncol. 2020;10: 573534. https://doi.org/10.3389/fonc.2020.573534.
Su W, Shi X, Wen X, Li X, Zhou J, Zhou Y, Ren F, Kang K. Integrative analysis of multiple cell death model for precise prognosis and drug response prediction in gastric cancer. Discover Oncol. 2024;15(1):532. https://doi.org/10.1007/s12672-024-01411-4.
Deng W, Gu L, Li X, Zheng J, Zhang Y, Duan B, Cui J, Dong J, Du J. CD24 associates with EGFR and supports EGF/EGFR signaling via RhoA in gastric cancer cells. J Transl Med. 2016;14:32. https://doi.org/10.1186/s12967-016-0787-y.
Li Q, Shi G, Li Y, Lu R, Liu Z. Integrated analysis of disulfidoptosis-related genes identifies NRP1 as a novel biomarker promoting proliferation of gastric cancer via glutamine mediated energy metabolism. Discover Oncol. 2024;15(1):337. https://doi.org/10.1007/s12672-024-01217-4.
Fernández-Palanca P, Payo-Serafín T, Méndez-Blanco C, San-Miguel B, Tuñón MJ, González-Gallego J, Mauriz JL. Neuropilins as potential biomarkers in hepatocellular carcinoma: a systematic review of basic and clinical implications. Clin Mol Hepatol. 2023;29(2):293–319. https://doi.org/10.3350/cmh.2022.0425.
Zhang P, Chen L, Zhou F, He Z, Wang G, Luo Y. NRP1 promotes prostate cancer progression via modulating EGFR-dependent AKT pathway activation. Cell Death Dis. 2023;14(2):159. https://doi.org/10.1038/s41419-023-05696-1.
Yu QY, Han Y, Lu JH, Sun YJ, Liao XH. NRP1 regulates autophagy and proliferation of gastric cancer through Wnt/β-catenin signaling pathway. Aging. 2023;15(17):8613–29. https://doi.org/10.18632/aging.204560.
Cruzat V, Macedo Rogero M, Noel Keane K, Curi R, Newsholme P. Glutamine: metabolism and immune function, supplementation and clinical translation. Nutrients. 2018;10(11):1564. https://doi.org/10.3390/nu10111564.
Li M, Wang J, Zhao Y, Lin C, Miao J, Ma X, Ye Z, Chen C, Tao K, Zhu P, Hu Q, Sun J, Gu J, Wei S. Identifying and evaluating a disulfidptosis-related gene signature to predict prognosis in colorectal adenocarcinoma patients. Front Immunol. 2024;15:1344637. https://doi.org/10.3389/fimmu.2024.1344637.
Liu X, Li D, Gao W, Chen P, Liu H, Zhao Y, Zhao W, Dong G. Molecular characterization, clinical value, and cancer-immune interactions of genes related to disulfidptosis and ferroptosis in colorectal cancer. Discover Oncol. 2024;15(1):183. https://doi.org/10.1007/s12672-024-01031-y.
Han X, Zhu J, Zhang X, Song Q, Ding J, Lu M, Sun S, Hu G. Plin4-dependent lipid droplets hamper neuronal mitophagy in the MPTP/p-induced mouse model of Parkinson’s disease. Front Neurosci. 2018;12:397. https://doi.org/10.3389/fnins.2018.00397.
Qu LW, Zhou B, Wang GZ, Chen Y, Zhou GB. Genomic variations in paired normal controls for lung adenocarcinomas. Oncotarget. 2017;8(61):104113–22. https://doi.org/10.18632/oncotarget.22020.
Zhang K, Zhu Z, Zhou J, Shi M, Wang N, Yu F, Xu L. Disulfidptosis-related gene expression reflects the prognosis of drug-resistant cancer patients and inhibition of MYH9 reverses sorafenib resistance. Transl Oncol. 2024;49: 102091. https://doi.org/10.1016/j.tranon.2024.102091.
Lin X, Li AM, Li YH, Luo RC, Zou YJ, Liu YY, Liu C, Xie YY, Zuo S, Liu Z, Liu Z, Fang WY. Silencing MYH9 blocks HBx-induced GSK3β ubiquitination and degradation to inhibit tumor stemness in hepatocellular carcinoma. Signal Transduct Target Ther. 2020;5(1):13. https://doi.org/10.1038/s41392-020-0111-4.
Hou R, Li Y, Luo X, Zhang W, Yang H, Zhang Y, Liu J, Liu S, Han S, Liu C, Huang Y, Liu Z, Li A, Fang W. ENKUR expression induced by chemically synthesized cinobufotalin suppresses malignant activities of hepatocellular carcinoma by modulating β-catenin/c-Jun/MYH9/USP7/c-Myc axis. Int J Biol Sci. 2022;18(6):2553–67. https://doi.org/10.7150/ijbs.67476.
Ren X, Zhu H, Deng K, Ning X, Li L, Liu D, Yang B, Shen C, Wang X, Wu N, Chen S, Gu D, Wang L. Long noncoding RNA TPRG1-AS1 suppresses migration of vascular smooth muscle cells and attenuates atherogenesis via interacting with MYH9 protein. Arterioscler Thromb Vasc Biol. 2022;42(11):1378–97. https://doi.org/10.1161/ATVBAHA.122.318158.
Liu X, Olszewski K, Zhang Y, Lim EW, Shi J, Zhang X, Zhang J, Lee H, Koppula P, Lei G, Zhuang L, You MJ, Fang B, Li W, Metallo CM, Poyurovsky MV, Gan B. Cystine transporter regulation of pentose phosphate pathway dependency and disulfide stress exposes a targetable metabolic vulnerability in cancer. Nat Cell Biol. 2020;22(4):476–86. https://doi.org/10.1038/s41556-020-0496-x.
Tang J, Peng X, Xiao D, Liu S, Tao Y, Shu L. Disulfidptosis-related signature predicts prognosis and characterizes the immune microenvironment in hepatocellular carcinoma. Cancer Cell Int. 2024;24(1):19. https://doi.org/10.1186/s12935-023-03188-y.
Zhao J, Luo Z, Fu R, Zhou J, Chen S, Wang J, Chen D, Xie X. Disulfidptosis-related signatures for prognostic and immunotherapy reactivity evaluation in hepatocellular carcinoma. Eur J Med Res. 2023;28(1):571. https://doi.org/10.1186/s40001-023-01535-3.
Wang T, Guo K, Zhang D, Wang H, Yin J, Cui H, Wu W. Disulfidptosis classification of hepatocellular carcinoma reveals correlation with clinical prognosis and immune profile. Int Immunopharmacol. 2023;120: 110368. https://doi.org/10.1016/j.intimp.2023.110368.
Xu K, Dai C, Yang J, Xu J, Xia C, Li J, Zhang C, Xu N, Wu T. Disulfidptosis-related lncRNA signatures assess immune microenvironment and drug sensitivity in hepatocellular carcinoma. Comput Biol Med. 2024;169: 107930. https://doi.org/10.1016/j.compbiomed.2024.107930.
Li X, Xu J, Yan L, Tang S, Zhang Y, Shi M, Liu P. Targeting disulfidptosis with potentially bioactive natural products in metabolic cancer therapy. Metabolites. 2024;14(11):604. https://doi.org/10.3390/metabo14110604.
Yang C, Wu A, Tan L, Tang D, Chen W, Lai X, Gu K, Chen J, Chen D, Tang Q. Epigallocatechin-3-gallate alleviates liver oxidative damage caused by iron overload in mice through inhibiting ferroptosis. Nutrients. 2023;15(8):1993. https://doi.org/10.3390/nu15081993.
Farhan M, Rizvi A. Understanding the prooxidant action of plant polyphenols in the cellular microenvironment of malignant cells: role of copper and therapeutic implications. Front Pharmacol. 2022;13: 929853. https://doi.org/10.3389/fphar.2022.929853.
Ge EJ, Bush AI, Casini A, Cobine PA, Cross JR, DeNicola GM, Dou QP, Franz KJ, Gohil VM, Gupta S, Kaler SG, Lutsenko S, Mittal V, Petris MJ, Polishchuk R, Ralle M, Schilsky ML, Tonks NK, Vahdat LT, Van Aelst L, et al. Connecting copper and cancer: from transition metal signalling to metalloplasia. Nat Rev Cancer. 2022;22(2):102–13. https://doi.org/10.1038/s41568-021-00417-2.
Vera JC, Reyes AM, Cárcamo JG, Velásquez FV, Rivas CI, Zhang RH, Strobel P, Iribarren R, Scher HI, Slebe JC. Genistein is a natural inhibitor of hexose and dehydroascorbic acid transport through the glucose transporter, GLUT1. J Biol Chem. 1996;271(15):8719–24. https://doi.org/10.1074/jbc.271.15.8719.
Liao Z, Xie Z. Construction of a disulfidptosis-related glycolysis gene risk model to predict the prognosis and immune infiltration analysis of gastric adenocarcinoma. Clin Transl Oncol. 2024;26(9):2309–22. https://doi.org/10.1007/s12094-024-03457-w.
Qu J, Guan H, Zheng Q, Sun F. Molecular subtypes of disulfidptosis regulated genes and prognosis models for predicting prognosis, tumor microenvironment infiltration, and therapeutic response in hepatocellular carcinoma. Int J Biol Macromol. 2024;261(Pt 1): 129584. https://doi.org/10.1016/j.ijbiomac.2024.129584.
Li XM, Liu SP, Li Y, Cai XM, Zhang SB, Xie ZF. Identification of disulfidptosis-related genes with immune infiltration in hepatocellular carcinoma. Heliyon. 2023;9(8): e18436. https://doi.org/10.1016/j.heliyon.2023.e18436.
Yang L, Zhang W, Yan Y. Identification and characterization of a novel molecular classification based on disulfidptosis-related genes to predict prognosis and immunotherapy efficacy in hepatocellular carcinoma. Aging. 2023;15(13):6135–51. https://doi.org/10.18632/aging.204809.
Yang R, Lai C, Huang L, Li F, Peng W, Wu M, Xin J, Lu Y, Ouyang M, Bai Y, Lei H, He S, Lin Y. Role of disulfidptosis in colorectal adenocarcinoma: implications for prognosis and immunity. Front Immunol. 2024;15:1409149. https://doi.org/10.3389/fimmu.2024.1409149.
Acknowledgements
We thank Figdraw (http://www.figdraw.com) and BioRender (Scientific Image and Illustration Software | BioRender ) for helping us with our drawing.
Funding
This study was funded by Changzhou Sci & Tech Program (Grant No. CJ20220064), the 2023 Changzhou Health Commission Science and Technology Project (Grant No. QY202301) and Top Talent of Changzhou “The 14th Five-Year Plan” High-level Health Personnel Training Project (Grant No.2022260).
Author information
Authors and Affiliations
Contributions
Y.C: Writing—original draft & editing, Visualization. D.C.Z, H.N.Y and J.W: Provided valuable suggestions, software, Validation and editing. W.T.H: Review & editing. Each author agrees to take responsibility for their respective contributions and to ensure the accuracy and integrity of any part of the review.
Corresponding author
Ethics declarations
Ethics approval and consent to participate
Not applicable.
Consent for publication
Not applicable.
Competing interests
The authors declare no competing interests.
Additional information
Publisher's Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.
About this article
Cite this article
Chen, Y., Zhang, D., Yang, H. et al. Advances in the study of disulfidptosis in digestive tract tumors. Discov Onc 16, 186 (2025). https://doi.org/10.1007/s12672-025-01875-y
Received:
Accepted:
Published:
DOI: https://doi.org/10.1007/s12672-025-01875-y