Spandidos Publications Logo
  • About
    • About Spandidos
    • Aims and Scopes
    • Abstracting and Indexing
    • Editorial Policies
    • Reprints and Permissions
    • Job Opportunities
    • Terms and Conditions
    • Contact
  • Journals
    • All Journals
    • Oncology Letters
      • Oncology Letters
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Oncology
      • International Journal of Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular and Clinical Oncology
      • Molecular and Clinical Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Experimental and Therapeutic Medicine
      • Experimental and Therapeutic Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Molecular Medicine
      • International Journal of Molecular Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Biomedical Reports
      • Biomedical Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Reports
      • Oncology Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular Medicine Reports
      • Molecular Medicine Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • World Academy of Sciences Journal
      • World Academy of Sciences Journal
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Functional Nutrition
      • International Journal of Functional Nutrition
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Epigenetics
      • International Journal of Epigenetics
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Medicine International
      • Medicine International
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
  • Articles
  • Information
    • Information for Authors
    • Information for Reviewers
    • Information for Librarians
    • Information for Advertisers
    • Conferences
  • Language Editing
Spandidos Publications Logo
  • About
    • About Spandidos
    • Aims and Scopes
    • Abstracting and Indexing
    • Editorial Policies
    • Reprints and Permissions
    • Job Opportunities
    • Terms and Conditions
    • Contact
  • Journals
    • All Journals
    • Biomedical Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Experimental and Therapeutic Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Epigenetics
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Functional Nutrition
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Molecular Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Medicine International
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular and Clinical Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular Medicine Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Letters
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • World Academy of Sciences Journal
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
  • Articles
  • Information
    • For Authors
    • For Reviewers
    • For Librarians
    • For Advertisers
    • Conferences
  • Language Editing
Login Register Submit
  • This site uses cookies
  • You can change your cookie settings at any time by following the instructions in our Cookie Policy. To find out more, you may read our Privacy Policy.

    I agree
Search articles by DOI, keyword, author or affiliation
Search
Advanced Search
presentation
International Journal of Molecular Medicine
Join Editorial Board Propose a Special Issue
Print ISSN: 1107-3756 Online ISSN: 1791-244X
Journal Cover
March-2026 Volume 57 Issue 3

Full Size Image

Sign up for eToc alerts
Recommend to Library

Journals

International Journal of Molecular Medicine

International Journal of Molecular Medicine

International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.

International Journal of Oncology

International Journal of Oncology

International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.

Molecular Medicine Reports

Molecular Medicine Reports

Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.

Oncology Reports

Oncology Reports

Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.

Oncology Letters

Oncology Letters

Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.

Biomedical Reports

Biomedical Reports

Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.

Molecular and Clinical Oncology

Molecular and Clinical Oncology

International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.

World Academy of Sciences Journal

World Academy of Sciences Journal

Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.

International Journal of Functional Nutrition

International Journal of Functional Nutrition

Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.

International Journal of Epigenetics

International Journal of Epigenetics

Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.

Medicine International

Medicine International

An International Open Access Journal Devoted to General Medicine.

Journal Cover
March-2026 Volume 57 Issue 3

Full Size Image

Sign up for eToc alerts
Recommend to Library

  • Article
  • Citations
    • Cite This Article
    • Download Citation
    • Create Citation Alert
    • Remove Citation Alert
    • Cited By
  • Similar Articles
    • Related Articles (in Spandidos Publications)
    • Similar Articles (Google Scholar)
    • Similar Articles (PubMed)
  • Download PDF
  • Download XML
  • View XML
Review Open Access

Role of cuproptosis in digestive system tumors (Review)

  • Authors:
    • Li Zhang
    • Yongpeng Cheng
    • Lulu Tang
    • Jiaxing Zhu
    • Biguang Tuo
  • View Affiliations / Copyright

    Affiliations: Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China, Department of Hepatobiliary Surgery, Liupanshui Municipal People's Hospital, Liupanshui, Guizhou 553000, P.R. China, Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
    Copyright: © Zhang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
  • Article Number: 56
    |
    Published online on: January 2, 2026
       https://doi.org/10.3892/ijmm.2026.5727
  • Expand metrics +
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Metrics: Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )
Cited By (CrossRef): 0 citations Loading Articles...

This article is mentioned in:


Abstract

In cells, copper levels are tightly regulated because copper deficiency leads to Menkes disease, anemia and neurodegeneration, whereas copper overload is associated with Wilson disease, liver injury, neurodegeneration and several cancers. Cuproptosis, a form of regulated cell death, depends on the intracellular accumulation of excessive copper. This process induces mitochondrial dysfunction and cell death by disrupting the stability of mitochondrial lipoylated proteins and iron‑sulfur cluster proteins. The present review aimed to summarize the mechanisms underlying cuproptosis in gastrointestinal cancer, with a focus on the relationship between copper metabolism imbalance and tumor initiation and progression, as well as the potential therapeutic applications of cuproptosis‑associated agents in oncology. The application prospects of cuproptosis in gastrointestinal tumor therapy are broad, offering novel therapeutic options that may improve prognosis in patients and survival outcomes.

Introduction

Copper is an essential trace mineral required for numerous physiological processes, including aerobic respiration, oxidative stress regulation and biosynthesis (1-4). Despite its key role in biological activities, intracellular copper levels are tightly regulated because both copper deficiency, such as in Menkes disease, anemia, and neurodegeneration, and copper excess, as seen in Wilson disease, liver injury, neurodegenerative disorder and several types of cancer, can lead to severe pathological conditions (5-9). When intracellular copper accumulates excessively, the organism initiates specific regulatory programs to decrease the copper content (10,11). Programmed cell death (PCD) refers to an orderly, gene-regulated process that maintains cell and systemic homeostasis and is key for both physiological and pathological cell turnover (12). The classical forms of PCD include apoptosis, necroptosis, pyroptosis, ferroptosis and autophagy (13). Apoptosis is characterized primarily by caspase activation and the release of cytochrome C from mitochondria under genetic control (14). Necroptosis involves both necrosis and apoptosis and is typically triggered by the binding of death receptors (such as tumor necrosis factor receptor 1 and the Fas receptor) to their respective ligands (15). Pyroptosis is mediated by pore-forming gasdermin proteins, which are activated by caspase-1 and the inflammasome complex, leading to cell membrane rupture (16). Ferroptosis is an iron-dependent process driven by lipid peroxidation of unsaturated fatty acids under the action of ferrous ions or lipoxygenase (17). Autophagy promotes the degradation and recycling of intracellular components under stress conditions to maintain cell homeostasis (18). In recent years, as the mechanisms of cell death have been elucidated, cuproptosis has emerged as a newly discovered form of PCD (19). Unlike traditional pathways such as apoptosis, necroptosis, autophagy or ferroptosis, cuproptosis depends on the intracellular accumulation of copper, which directly binds lipoylated proteins in the tricarboxylic acid (TCA) cycle, causing protein aggregation and iron-sulfur cluster degradation. This results in proteotoxic stress, mitochondrial dysfunction and cell death (19). In addition to direct copper accumulation, copper also induces other forms of cell death. For example, copper-induced apoptosis occurs via the catalytic generation of reactive oxygen species (ROS), leading to oxidative stress, DNA damage and the activation of apoptotic pathways (20). Copper can also directly or indirectly modulate apoptosis-associated proteins, such as by activating p53 and enhancing its pro-death function (21). Copper-induced ferroptosis may occur via promotion of iron absorption and utilization, increasing intracellular iron levels and exacerbating lipid peroxidation, thereby increasing ferroptotic death (22). Copper also produces hydroxyl radicals through Fenton-like reactions, triggering lipid peroxidation and ferroptosis (23). In addition, copper may induce the autophagic degradation of GPX4, facilitating ferroptosis (24). Copper-induced autophagy is triggered by the activation of AMPK, inhibition of mTOR or direct interaction with UNC-51-like kinase ½ (25). This process also occurs via upregulation of autophagy-related genes and activation of the transcription factor (TF)EB, promoting autophagosome and autolysosome formation, which may lead to autophagy-dependent cell death (26). Moreover, copper-induced ROS generation and endoplasmic reticulum stress promote NLRP3 inflammasome assembly and gasdermin D activation, thus triggering pyroptosis (6). Recent research has revealed an association between copper metabolism dysregulation and tumor initiation and progression (6). Copper serves a dual role in cancer: Imbalanced copper metabolism promotes tumor cell proliferation and survival by activating the receptor tyrosine kinase, PI3K/Akt/mTOR, and MAPK/ERK signaling pathways (6,27) and modulating the tumor microenvironment (TME) through angiogenesis and immune evasion (28), while cuproptosis suppresses tumor growth by inducing cell death and activating immune responses (6). Copper stimulates apoptosis, necrosis, autophagy, ferroptosis and cuproptosis and enhances antitumor immunity by activating immune cells (11). As copper dysregulation is common in cancer cells, targeting copper levels or metabolic pathways can trigger cuproptosis, thereby inhibiting tumor growth and progression. Cuproptosis may thus represent a promising anticancer strategy. Furthermore, the potential effects of cuproptosis and other forms of cell death, such as ferroptosis and apoptosis, provide a theoretical basis for combination therapy (22). In recent years, therapeutic strategies for gastrointestinal cancer have shifted from local surgical resection to systemic, multimodal therapy (29). Although progress has been made in terms of conventional chemotherapy and targeted therapies in certain patients, drug resistance and relapse remain challenges. With the advent of precision medicine and immunotherapy, gastrointestinal cancer management has evolved from single-modality chemotherapy to integrated metabolic regulation, immune remodeling, and microenvironmental intervention. Immune checkpoint inhibitors (PD-1/PD-L1 and CTLA-4 antibodies) and antiangiogenic agents have demonstrated promising efficacy and potential for application in patients with gastrointestinal and hepatocellular tumors (30,31). Concurrently, metabolism-immune integrated therapy has been proposed to enhance immune responses by modulating tumor energy metabolism and redox balance (32). The regulation of metal ion homeostasis has been recognized as a key component of systemic therapy, with copper serving a pivotal role in the regulation of mitochondrial respiration and oxidative stress (19). As a copper-dependent PCD pathway, cuproptosis may serve as a link between metabolism and immunity. Studies (30,31,33) have highlighted the interplay between ion metabolism, oxidative stress and immune signaling as a central axis in systemic therapy for gastrointestinal tumors, thereby providing a theoretical foundation for incorporating cuproptosis into therapeutic frameworks. Accordingly, the present review systematically summarizes the mechanisms of copper metabolism and cuproptosis to understand metabolic and immune microenvironmental remodeling in gastrointestinal cancer, offering conceptual and theoretical support for metabolic targeting and systemic treatment strategies.

Copper metabolism, transport, regulation and function

Copper metabolism

Copper, a widely distributed metallic element in nature, is an essential trace element for the human body. Most dietary copper exists in the form of Cu2+ and is absorbed primarily in the duodenum and small intestine (34). After absorption through the gastrointestinal tract into the bloodstream, ~90% of copper binds to ceruloplasmin in the plasma, while the remaining portion is associated with albumin, transcuprein and histidine. These copper complexes are transported via the portal vein to the liver and other organs where copper exerts physiological effects (35). The liver serves as the primary storage organ for copper (5). Mitochondria are the notable sites of copper utilization due to the presence of copper-dependent enzymes such as cytochrome c oxidase, which is involved in oxidative phosphorylation, and 1-5% of total cellular superoxide dismutase 1 (SOD1), which serves a critical role in mitigating oxidative stress within the mitochondrial matrix. These enzymes highlight the essential role of copper in maintaining mitochondrial function and cellular health (36). Excess copper is secreted into the bile and blood, excreted via the intestine or delivered to peripheral tissues to catalyze physiological reactions (37). A small portion is excreted in urine, sweat and menstrual fluid (37). Extracellular copper exists predominantly in the divalent Cu2+ state and cannot be directly used by cells (38,39). At the cellular level, Cu2+ enters through divalent metal transporter 1 or is reduced to Cu+ by six-transmembrane epithelial antigens such as Six-Transmembrane Epithelial Antigen of the Prostate 1 (STEAP1) or duodenal cytochrome b, after which Cu+ is imported into the cytoplasm via the high-affinity copper transporter 1 (CTR1) (40,41). Once inside the cytosol, excess copper is sequestered by metallothioneins (34) or delivered to specific organelles by copper chaperone proteins. For example, the copper chaperone for SOD (CCS) transfers copper to SOD1, enabling copper insertion, disulfide bond formation and the localization of SOD1 to the cytosol or mitochondria (42). Cytochrome c oxidase 17 (COX17) transfers copper to the mitochondrial inner membrane subunits COX1 and COX2 (38), which participate in the assembly of the respiratory chain. In addition, copper is transported through the secretory pathway via antioxidant-1 copper chaperone (ATOX1), which delivers copper to the trans-Golgi network (TGN) and copper-transporting ATPases (ATP7A and ATP7B) located in the Golgi or plasma membrane, maintaining cell copper homeostasis (26). The overall process of copper metabolism is illustrated in Fig. 1.

Copper metabolism. Extracellular
Cu2+ is reduced to Cu+ by the reductase
STEAP. Some Cu+ is transported to the cell through the
transporter CTR1 to bind to CCS and SOD1, and the remaining portion
is transported to the mitochondria, TGN and nucleus. In
mitochondria, Cu+ participates in the respiratory chain
and redox pathway by binding CCO. In the mitochondrial
intermembrane space, COX17 binds and transfers Cu+ to
SCO1 or COX11, which transfers Cu+ to the cytochrome
oxidase subunit. In the nucleus, Cu+ binds to
transcription factors and drives gene expression. In the TGN, the
Cu+-ATPase transporters ATP7A and ATP7B transfer
Cu+ from the cytoplasm to the lumen of the TGN, where
Cu+ activates Cu-dependent enzymes in the secretory
pathway. When the intracellular Cu+ concentration is
high, ATP7A and ATP7B withdraw from the TGN and promote
Cu+ output. ATOX1, antioxidant 1 copper chaperone;
ATP7A, ATPase copper-transporting α; CCO, cytochrome c oxidase;
CCS, copper chaperone superoxide dismutase; COX17, cytochrome c
oxidase copper chaperone protein 17; SCO1, synthetic cytochrome c
oxidase 1; SOD1, superoxide dismutase 1; STEAP, prostate
six-transmembrane epithelial antigen; SLC31A1, solute carrier
family 31 member 1; TGN, trans-Golgi network.

Figure 1

Copper metabolism. Extracellular Cu2+ is reduced to Cu+ by the reductase STEAP. Some Cu+ is transported to the cell through the transporter CTR1 to bind to CCS and SOD1, and the remaining portion is transported to the mitochondria, TGN and nucleus. In mitochondria, Cu+ participates in the respiratory chain and redox pathway by binding CCO. In the mitochondrial intermembrane space, COX17 binds and transfers Cu+ to SCO1 or COX11, which transfers Cu+ to the cytochrome oxidase subunit. In the nucleus, Cu+ binds to transcription factors and drives gene expression. In the TGN, the Cu+-ATPase transporters ATP7A and ATP7B transfer Cu+ from the cytoplasm to the lumen of the TGN, where Cu+ activates Cu-dependent enzymes in the secretory pathway. When the intracellular Cu+ concentration is high, ATP7A and ATP7B withdraw from the TGN and promote Cu+ output. ATOX1, antioxidant 1 copper chaperone; ATP7A, ATPase copper-transporting α; CCO, cytochrome c oxidase; CCS, copper chaperone superoxide dismutase; COX17, cytochrome c oxidase copper chaperone protein 17; SCO1, synthetic cytochrome c oxidase 1; SOD1, superoxide dismutase 1; STEAP, prostate six-transmembrane epithelial antigen; SLC31A1, solute carrier family 31 member 1; TGN, trans-Golgi network.

Copper import proteins

In addition to the aforementioned transporters, CTR1 is the primary plasma membrane protein responsible for cellular copper uptake. CTR1, which has the highest copper-binding affinity among known transporters, specifically binds extracellular Cu2+ and transports it into the cytoplasm, where it serves as a major copper chaperone for enzymes such as SOD (5,43) (Table I). Following exposure to extracellular copper, CTR1 recognizes and binds copper, undergoes conformational changes and transports copper into cells to sustain normal physiological function (44). Once internalized, copper binds to chaperones such as ATOX1 and COX17, which deliver copper to target enzymes, mitochondria or metalloproteins in the endoplasmic reticulum, ensuring proper copper distribution (45). The upregulation of CTR1 leads to cellular copper overload (46). Additionally, solute carrier family 25 member 3 (SLC25A3), a mitochondrial phosphate carrier, is capable of copper transport, and its upregulation results in mitochondrial matrix copper overload (47).

Table I

Functions of copper death-associated genes and their roles in cuproptosis.

Table I

Functions of copper death-associated genes and their roles in cuproptosis.

GeneSubcellular locationFunctionRole in cuproptosis(Refs.)
CTR1Cell membraneTransports extracellular copper intracellularly; copper chaperones of enzymes such as superoxide dismutaseIts increase leads to intracellular copper accumulation(5,43,46)
FDX1Mitochondrial matrixUpstream regulator of lipid acylation of mitochondrial proteins; Fe-S cluster biosynthesis; reduces Cu2+ to Cu+; synthesizes steroid hormones and electron transport intermediates for mitochondrial cytochrome P450Cu2+ to Cu+(78-83)
LIASMitochondriaConverts octanoylated domains into lipoylated derivativesInvolved in the lipoylation pathway(84,85)
DLATMitochondrial matrixMediates the conversion of pyruvate to acetyl-CoALipoylated DLAT oligomerization leads to cell death(86,87)
GLSMitochondria, cytoplasm and cytosolCatalyzes the catabolism of glutamineIts decrease leads to sensitivity to cuproptosis(88)
CDKN2ANucleus, cytosolInduces cell cycle arrest in G1 and G2 phaseIts decrease leads to sensitivity to cuproptosis(89-91)
ATP7ACell membrane, trans-Golgi network membrane, plasma membraneRegulates the excretion or distribution of copper in the intracellular spaceIts decrease leads to intracellular copper accumulation(92,93)
ATP7BCell membrane, trans-Golgi network and membraneRegulates the excretion or distribution of copper in the intracellular spaceIts decrease leads to intracellular copper accumulation(94,95)

[i] FDX1, ferredoxin 1; LIAS, Lipoic Acid Synthetase; DLAT, Dihydrolipoamide S-Acyltransferase; GLS, Glutaminase; CDKN2A, Cyclin-Dependent Kinase Inhibitor 2A; ATP7A, ATPase, Cu2+ Transporting, Alpha Polypeptide; CTR, Copper Transporter.

Copper export proteins

ATPase copper-transporting α (ATP7A) and β (ATP7B) are P-type copper-transporting ATPases that regulate copper efflux and intracellular distribution (48). They use the energy derived from ATP hydrolysis to actively transport copper across membranes against their concentration gradients, thereby maintaining copper homeostasis (48,49). ATP7A is expressed ubiquitously, whereas ATP7B is expressed primarily in the liver (50). In hepatocytes, ATP7B mediates the excretion of excess copper into bile, while unabsorbed copper is eliminated through feces (51). Copper efflux is essential for preventing copper-induced cytotoxicity. When the intracellular Cu+ concentration increases above a threshold, ATOX1 mediates Cu+ transfer to ATP7A/B in the TGN, after which these proteins relocate to the plasma membrane to export Cu+ (26). ATP7A and ATP7B thus serve central roles in maintaining systemic copper balance. Dysfunction of these ATPases leads to severe multisystem disorders such as Menkes (52) and Wilson's disease (53). Copper imbalance contributes to cardiovascular disease (54), retinal disorders (55) and tumorigenesis (43) and also perturbs mitochondrial respiration, glycolysis, insulin resistance and lipid metabolism (56-58).

Copper function

As a heavy metal element, copper is typically associated with toxicity, however, copper also has essential physiological functions as a cofactor for numerous proteins and enzymes (5). For example, cytochrome c oxidase is a copper-dependent enzyme that is key for the final step in the electron transport chain, where it facilitates the transfer of electrons to oxygen during oxidative phosphorylation (59). SOD1), another copper-containing enzyme, protects cells from oxidative damage by converting superoxide radicals to hydrogen peroxide and oxygen (60). Its versatile redox activity, which involves cycling between Cu+ and Cu2+, enables copper to serve as a crucial catalytic cofactor in biochemical reactions (61), including oxidative stress regulation (1,2), cellular respiration (56), neurotransmitter synthesis and metabolism (62) and epigenetic modification (63). In addition to redox functions, copper contributes to hematopoiesis, immune regulation, melanin and connective tissue formation and central nervous system protection (8,64,65). Copper has also been implicated in cancer diagnosis and therapy. Advances in metal-based medicine have focused on achieving targeted toxicity through chemical coordination and controlled drug delivery (19,66). Conversely, copper-depleting therapy [tetrathiomolybdate (TTM)] exerts antitumor effects by chelating copper in the serum, decreasing vascular endothelial growth factor expression and modulating the immunosuppressive TME. These mechanisms demonstrate antimetastatic and antiangiogenic effects in models of breast, colorectal and hepatocellular carcinoma (HCC) (67-69). Although other metals, such as platinum and technetium, have been applied in chemotherapy, imaging and radiotherapy (70,71), the dual nature of copper, in which it is both physiologically essential and potentially toxic, links it with energy metabolism and immune regulation. Consequently, copper homeostasis has emerged as a frontier topic in the study of metal-based anticancer therapeutics (72,73).

Mechanisms of cuproptosis

Mitochondrial damage and lipoylated protein aggregation

Copper can trigger multiple forms of cell death, including apoptosis, oxidative stress-induced necrosis, autophagy and ferroptosis (74). Recent study have revealed that under the action of copper ionophores such as elesclomol (ES), copper induces cuproptosis, a distinct form of PCD, through unique mechanisms involving the disruption of iron-sulfur (Fe-S) cluster proteins and the induction of lipid peroxidation (19). The key hallmark of cuproptosis is the abnormal aggregation of lipoylated proteins and depletion of Fe-S cluster proteins, leading to mitochondrial contraction, chromatin fragmentation and cell membrane rupture (75). Mechanistically, ES transports Cu2+ into the cytoplasm, where it is reduced to Cu+ by ferredoxin 1 (FDX1). The reduced Cu+ enters mitochondria and binds directly to key lipoylated enzymes in the TCA cycle, such as dihydrolipoamide S-acetyltransferase (DLAT) and dihydrolipoamide S-succinyltransferase (DLST). This interaction induces protein aggregation and the loss of Fe-S clusters (19). Fe-S clusters are key cofactors for numerous mitochondrial enzymes and respiratory chain complexes, such as succinate dehydrogenase subunit A and NADH:ubiquinone oxidoreductase subunit S1. Their disruption impairs the electron transport chain and decreases ATP synthesis and mitochondrial membrane potential (76). Consequently, mitochondrial energy production decreases, accompanied by increased inner membrane permeability, elevated Ca2+ concentration and the accumulation of ROS (77). The aggregation of lipoylated proteins further induces proteotoxic stress, disrupting proteostasis and exacerbating mitochondrial injury. Together, these molecular events define the mitochondrial mechanism of cuproptosis (Fig. 2).

Molecular mechanism of cuproptosis.
Excess Cu2+ is imported into cells via CTR/SLC31A1 and
as well as copper ionophores elesclomol and DSF. Cu2+ is
reduced to Cu+ mainly through FDX1 and STEAP, increasing
the bioactive Cu+ pool. Mitochondrial Cu+
directly binds lipoylated TCA-cycle enzymes, particularly the
pyruvate dehydrogenase complex, inducing aberrant aggregation of
DLAT and other lipoylated proteins. This aggregation disrupts
lipoylation-dependent TCA activity and Ac-CoA production, and is
accompanied by loss of Fe-S clusters, thereby impairing
Fe-S-dependent enzymes and the ETC. These events drive excessive
ROS generation, oxidative stress, mitochondrial dysfunction and
ultimately cuproptosis-related cell death. Copper homeostasis is
counterbalanced by ATP7A/B-mediated copper efflux/redistribution,
while GSH buffering mitigates copper-associated oxidative stress.
FDX1, ferredoxin 1; TCA, tricarboxylic acid cycle; DLAT,
Dihydrolipoamide S-Acyltransferase; ROS, Reactive Oxygen Species;
DSF, Disulfiram; STEAP, Six-Transmembrane epithelial Antigen of the
Prostate; CTR, Copper Transporter; SLC31A1, Solute Carrier Family
31 Member 1; ATP7A/B, ATPase, Cu2+ Transporting, α/β
polypeptide; Ac-CoA, Acetyl-Coenzyme A; ETC, Electron Transport
Chain; GSH, Glutathione; LIAS, Lipoic Acid Synthase.

Figure 2

Molecular mechanism of cuproptosis. Excess Cu2+ is imported into cells via CTR/SLC31A1 and as well as copper ionophores elesclomol and DSF. Cu2+ is reduced to Cu+ mainly through FDX1 and STEAP, increasing the bioactive Cu+ pool. Mitochondrial Cu+ directly binds lipoylated TCA-cycle enzymes, particularly the pyruvate dehydrogenase complex, inducing aberrant aggregation of DLAT and other lipoylated proteins. This aggregation disrupts lipoylation-dependent TCA activity and Ac-CoA production, and is accompanied by loss of Fe-S clusters, thereby impairing Fe-S-dependent enzymes and the ETC. These events drive excessive ROS generation, oxidative stress, mitochondrial dysfunction and ultimately cuproptosis-related cell death. Copper homeostasis is counterbalanced by ATP7A/B-mediated copper efflux/redistribution, while GSH buffering mitigates copper-associated oxidative stress. FDX1, ferredoxin 1; TCA, tricarboxylic acid cycle; DLAT, Dihydrolipoamide S-Acyltransferase; ROS, Reactive Oxygen Species; DSF, Disulfiram; STEAP, Six-Transmembrane epithelial Antigen of the Prostate; CTR, Copper Transporter; SLC31A1, Solute Carrier Family 31 Member 1; ATP7A/B, ATPase, Cu2+ Transporting, α/β polypeptide; Ac-CoA, Acetyl-Coenzyme A; ETC, Electron Transport Chain; GSH, Glutathione; LIAS, Lipoic Acid Synthase.

Oxidative stress

Cu2+ catalyzes the Fenton-like reaction, generating hydroxyl radicals that initiate oxidative stress, leading to extensive cell damage and death (96). Hydroxyl radicals are highly reactive and attack DNA molecules to cause strand breaks, base modification and cross-linking, thereby interfering with DNA replication and transcription (97,98). These radicals also induce lipid peroxidation, compromising the integrity and fluidity of cell membranes and increasing membrane permeability (99). Additionally, oxidative modification alters protein structure and function, thereby disrupting intracellular protein homeostasis (100). Mitochondria are notable targets of copper-induced oxidative stress. Excess ROS decrease the mitochondrial membrane potential and ATP synthesis and promote cytochrome c release, ultimately activating caspase cascades and triggering apoptosis (101). Oxidative stress also activates TFs such asp53 and Nrf2, which regulate antioxidant responses and cell death pathways (102). Although Nrf2 activation enhances the antioxidant capacity by promoting the expression of antioxidant genes, when oxidative stress becomes excessive, these defense mechanisms are overwhelmed, leading to the accumulation of reactive oxygen species (ROS) and disruption of redox balance, ultimately resulting in cell death. Collectively, these molecular events constitute the oxidative stress-mediated mechanism of cuproptosis.

Cuproptosis in digestive system tumors

Increasing evidence has demonstrated that the dysregulation of copper metabolism is associated with the onset and progression of various diseases, particularly malignancy (6,7,103). As a key signaling metal, copper participates in cancer development by promoting cell proliferation, angiogenesis and metastasis (104). Copper serves dual roles in cancer biology; it is indispensable for cellular metabolism, but its dysregulation is associated with oncogenesis. Elevated copper levels have been detected in tumor tissue or serum in patients with multiple types of cancer, including breast (105-109), lung (110-112) and gastrointestinal cancer (113-116), oral (117), thyroid (118) and gallbladder carcinoma (119) and gynecological (115,116) and prostate cancer (120). These findings indicate that copper is not only a key factor in tumor growth and metastasis but also a necessary micronutrient for tumor cells (73,121). Mechanistically, copper promotes tumor progression through multiple pathways. First, copper stimulates angiogenesis by activating angiogenic factors and enhancing the proliferation and migration of vascular endothelial cells (122), thereby supporting tumor initiation, growth and metastasis (73,74,123,124). Newly formed vasculature provides key nutrients and serves as a conduit for tumor cell dissemination. Second, copper serves as a cofactor for several metalloenzymes, including MMP-9, SOD1, vascular adhesion protein-1 and lysyl oxidase (LOX), all of which are key for cancer invasion and metastasis (125-128). The ATOX1-ATP7A-LOX axis promotes metastatic dissemination by facilitating the copper-dependent activation of LOX and LOX-like enzymes, which remodel the extracellular matrix and increase tumor invasiveness (73,129). Third, copper activates the MAPK/ERK signaling pathway, thereby promoting tumor cell proliferation (130). In addition to these direct mechanisms, copper also modulates the TME to promote cancer progression. Copper influences tumor metabolic reprogramming, enhancing cell survival under hypoxic conditions (6). Moreover, copper contributes to immune evasion by regulating immune cell activity or promoting the expansion of immunosuppressive cell populations, allowing tumor cells to escape host immune surveillance (131). For example, copper alters macrophage polarization, shifting tumor-associated macrophages toward the M2 phenotype, which enhances immune suppression and facilitates tumor invasion and metastasis (11,132). Collectively, these findings suggest that copper serves multiple roles in tumorigenesis and disease progression, including roles in cell proliferation, angiogenesis, metastasis, metabolic reprogramming and immune escape, positioning copper as both a potential biomarker and therapeutic target in cancer biology.

Esophageal cancer (ESCA)

The global incidence of ESCA in 2020 was ~604,000 new cases, and about 544,000 people died from ESCA; ESCA ranks 11th in terms of global cancer incidence and 7th in terms of cancer-associated mortality (133). Despite notable advances in diagnosis and therapy, the 5-year survival rate of patients with esophageal squamous cell carcinoma remains 20% owing to its late detection and rapid progression (134). Therefore, identifying novel diagnostic biomarkers and therapeutic strategies is key for improving prognosis and survival outcomes. Methyltransferase-like 3 (METTL3) expression is markedly elevated in ESCA tissue compared with that in normal esophageal epithelium, particularly in highly malignant tumors, and is associated with disease progression (135). Liu et al (136) investigated the role of METTL3 in ESCA and revealed its association with glycolysis, cuproptosis and the competing endogenous (ce) RNA regulatory network. These findings demonstrated that METTL3 serves as a critical mediator of ESCA progression by modulating glycolysis-associated gene expression. Upregulation of METTL3 in esophageal carcinoma enhances glycolytic flux, increases glucose uptake and lactate production and promotes tumor growth (128). Moreover, METTL3 expression is associated with the expression of genes associated with cuproptosis. Mechanistically, METTL3 serves dual regulatory roles in glycolysis and cuproptosis via N6-methyladenosine (m6A) RNA modification. Specifically, METTL3 alters and stabilizes glycolysis-associated mRNAs, thereby altering tumor cell energy metabolism and proliferation. Concurrently, METTL3 may regulate genes associated with copper homeostasis, affecting intracellular copper levels and inducing cell death. Additionally, METTL3 is involved in the ceRNA network, in which long non-coding (lnc) and circular RNAs compete for shared microRNAs (miRNAs or miRs), influencing posttranscriptional regulation. By modulating the stability or function of ceRNA components, METTL3 indirectly affects gene expression and tumor progression. These findings indicate that targeting METTL3 and its associated pathways may represent a promising therapeutic strategy for ESCA, especially in highly glycolytic and copper-enriched subtypes. The aforementioned study provides valuable insights into the epigenetic and metabolic regulation of ESCA, underscoring the potential of METTL3 as a therapeutic target that links epigenetic modification, metabolic reprogramming and copper-induced cell death. Further study have identified the cuproptosis-associated genes Centromere Protein E and SHC SH2 Domain-Binding Protein 1 as key biomarkers of Barrett's esophagus (BE) progression to esophageal adenocarcinoma (EAC) (137). These genes may influence the immune microenvironment and promote the transformation from BE to EAC, offering molecular targets for early diagnosis and treatment. Metabolic abnormalities (lactate accumulation and mitochondrial dysfunction) within tumor cells and the surrounding microenvironment impair immune responses and promote immune evasion (33). Moreover, disruption of metal ion homeostasis, particularly copper imbalance, alters oxidative stress and mitochondrial metabolism, further influencing tumor survival and therapeutic resistance (32,138). Therefore, integrating cuproptosis pathways into chemoradiotherapy or immunotherapy regimens may enhance radiosensitivity and remodel the tumor immune microenvironment, offering a rational framework for combination treatment strategies in ESCA.

Gastric cancer (GC)

GC is an epithelial malignancy originating in the stomach. In 2022, there were ~968,000 new cases of GC worldwide, accounting for 4.9% of all new cancer cases, making it the fifth most common cancer by incidence globally (139). At the same time, gastric cancer accounted for approximately 6.8% of cancer-related deaths globally, with around 660,000 deaths, ranking fifth among the leading causes of cancer death worldwide (139). Disruptions in PCD serve a crucial role in GC pathogenesis (140,141). Copper levels are notably elevated in gastric tumor tissue compared with normal gastric mucosa, particularly in high-grade malignancy. Moreover, the copper content is positively associated with the TNM stage of GC (142). In a recent study, Sun et al (79) investigated the mechanism of cuproptosis in GC, identifying its association with METTL16, an atypical m6A RNA METTL involved in m6A modification. METTL16 serves as a key mediator of cuproptosis by regulating FDX1 mRNA through m6A modification (79). Elevated copper levels in GC tissue promote the lactylation of METTL16 at lysine residue K229, which enhances its enzymatic activity and upregulates FDX1 expression, inducing cell death. Mechanistically, SIRT2 serves as a critical delactylase, removing lactyl groups from METTL16-K229 and inhibiting METTL16 activity. Treatment with (Z)-2-cyano-3-[5-(2,5-dichlorophenyl) furan-2-yl]-N-quinolin-5-ylprop-2-enamide, a selective SIRT2 inhibitor, increases METTL16 lactylation and FDX1 expression, thereby promoting cuproptosis. These findings suggest that the combined use of copper ionophores (such as ES) and SIRT2 inhibitors may represent a promising therapeutic strategy for GC, particularly in aggressive, high-copper and high-lactate subtypes such as mucinous adenocarcinoma. Sun et al provided valuable mechanistic insight into the role of METTL16-mediated RNA modification and lactylation in copper-induced cell death, highlighting the therapeutic potential of targeting the METTL16-SIRT2-FDX1 axis in GC treatment. Polypyrimidine tract-binding protein 3 (PTBP3) is markedly upregulated in peritoneal metastases of GC and is associated with poor prognosis (143). Single-cell RNA sequencing and transcriptome analysis reveal that PTBP3 regulates its downstream target COX11, impairing its function and decreasing the mitochondrial copper content, which enables tumor cells to evade cuproptosis. Researchers have developed an antisense oligonucleotide (ASO) targeting the short isoform of COX11 pre-mRNA exon 4 (136), effectively degrading COX11 mRNA and disrupting copper homeostasis. In a patient-derived organoid xenograft model combination therapy using exogenous copper ionophores and ASO drugs leads to excessive mitochondrial copper accumulation, proteotoxic stress and the induction of cuproptosis, thereby suppressing peritoneal metastasis (143). This provides a new therapeutic strategy targeting PTBP3-mediated COX11 splicing to restore copper-dependent cell death in metastatic GC. In stomach adenocarcinoma (STAD), FDX1, lipoic Acid Synthase (LIAS), Metal Regulatory Transcription Factor 1 (MTF1) and Pyruvate Dehydrogenase E1 Subunit Alpha 1 have been identified as key genes associated with cuproptosis (144). FDX1 is highly expressed in STAD tumor tissues, associated with poor prognosis and increased chemosensitivity to cisplatin and 5-fluorouracil, making FDX1 a potential predictive biomarker for chemotherapy response (145). LIAS and MTF1 exhibit notable prognostic value, where higher expression levels are associated with improved survival. Collectively, these molecular markers not only contribute to prognostic evaluation but also provide a foundation for the development of copper-targeted anticancer drugs. Aberrant copper homeostasis in GC is associated with mitochondrial dysfunction and ROS accumulation, whereas cuproptosis promotes energy metabolism collapse and cell death (146,147). Future studies should explore the potential synergy between copper-modulating drugs (such as ES) and immunotherapy or antiangiogenic therapy with the aim of achieving a coordinated antitumor effect through the simultaneous regulation of metabolic, immune and redox networks.

HCC

Based on GLOBOCAN 2020, HCC ranks sixth in incidence and third in cancer-associated mortality worldwide (148). Although notable therapeutic progress has been made over the past decade, the prognosis of HCC remains poor, largely because most patients are diagnosed at advanced stages, precluding surgical or localized treatment (149,150). Hence, identifying effective molecular targets and therapeutic strategies for HCC is key. Clinical studies have reported significantly elevated serum copper levels (151,152), increased copper-protein complexes and enhanced expression of copper-binding proteins in patients with HCC (153,154), as well as the downregulation of copper transporters such as ATP7A/B and SLC31A1/2 (155). Copper concentrations in HCC tissue are markedly higher than those in normal liver tissue and elevated serum copper levels are associated with tumor progression (156). In a recent study, Li et al (157) investigated the mechanism of cuproptosis in HCC, focusing on DLAT, a key gene associated with copper-induced cell death. The aforementioned study reported that maternal embryonic leucine zipper kinase (MELK) serves as a key mediator of cuproptosis by activating the PI3K/mTOR signaling pathway. Elevated copper levels in HCC promote MELK expression and activity, which upregulate DLAT expression and support mitochondrial function, thus facilitating HCC progression. Mechanistically, treatment with the copper ionophore ES decreases the expression of translocase of outer mitochondrial membrane 20 and enhances DLAT oligomerization, thereby suppressing MELK activity and triggering cuproptosis. These findings indicate that the combination of copper ionophores and PI3K/mTOR pathway inhibitors may represent a promising therapeutic approach for treating HCC. Li et al (157) provided key mechanistic insight into the MELK-DLAT regulatory axis, highlighting its potential as a therapeutic target in liver cancer. Exposure of Hep3B hepatoma cells to Cu2+ suppresses histone acetyltransferase activity, leading to hypoacetylation of histones H3 and H4, which promotes cell proliferation (19). Copper also binds pyruvate dehydrogenase kinase 1 (PDK1), enhancing its interaction with AKT, thereby activating the PDK1-AKT oncogenic signaling cascade and promoting HCC progression (158). Conversely, copper chelators or CTR1 inhibitors downregulate the CTR1-AKT axis, thus inhibiting tumor growth (158). CTR1 is aberrantly upregulated in breast cancer and is negatively regulated by NEDD4-like E3 ubiquitin ligase (NEDD4l), which promotes CTR1 ubiquitination and degradation. NEDD4l exerts tumor-suppressive effects by inhibiting CTR1-mediated AKT signaling. These findings reveal an association between the CTR1/Cu pathway and PDK1-AKT oncogenic signaling, underscoring the therapeutic potential of targeting the CTR1-Cu axis in cancers driven by AKT hyperactivation. By contrast with copper accumulation, FDX1 expression is notably lower in HCC tissues than in normal liver tissue, and its expression is positively associated with patient survival, with higher FDX1 expression predicting longer overall survival. Moreover, FDX1 levels are positively associated with oxaliplatin sensitivity. In a recent study, Quan et al (159) elucidated the molecular mechanism of cuproptosis in HCC, focusing on the FDX1-associated lncRNA/miRNA regulatory axis. Through bioinformatic analyses, the aforementioned study identified LINC02362 as a ceRNA that modulates miR-18a-5p, which directly targets FDX1. Upregulation of the LINC02362/miR-18a-5p/FDX1 pathway signaling suppresses HCC cell proliferation. Conversely, LINC02362 knockdown decreases intracellular copper concentrations and induces resistance to ES-Cu-mediated cell death. Additionally, upregulation of this axis enhances the sensitivity of HCC to oxaliplatin by promoting cuproptosis. These findings suggest that LINC02362/miR-18a-5p/FDX1 is a novel regulatory pathway capable of overcoming oxaliplatin resistance in HCC through cuproptotic mechanisms. Recent preclinical studies have shown that copper chelators, including triethylenetetramine and D-penicillamine, significantly decrease copper levels and inhibit tumor growth in HCC (160). By restoring copper homeostasis, these agents may also overcome resistance to conventional therapy, supporting their potential clinical use in HCC treatment. Emerging research indicates that metabolic reprogramming and copper imbalance are common features of HCC (161). Excess copper disrupts mitochondrial oxidative phosphorylation and lipoyl enzyme complex activity, leading to lipid metabolic disorder and energy collapse (19). Moreover, the expression of genes associated with cuproptosis, such as FDX1, LIAS and DLAT, is downregulated in HCC tissue and the expression of these genes is significantly associated with immune phenotypes and overall survival (162). In conclusion, copper serves as both a metabolic cofactor and a potential therapeutic target in HCC. Regulation of copper levels or exploitation of its bioactive properties offers a promising avenue for novel anticancer strategies. Future studies should elucidate the mechanisms of copper-targeted therapy, optimize therapeutic regimens and conduct rigorous clinical trials to validate their safety and efficacy.

Pancreatic cancer

According to GLOBOCAN 2020, pancreatic cancer is a highly aggressive malignancy, ranking twelfth in incidence but sixth in overall cancer-associated mortality worldwide (163). Despite advances in treatment, the 5-year overall survival rate remains ~ 10% (164). Pancreatic cancer remains among the most common types of treatment-refractory malignancy. Elevated serum copper levels may contribute to pancreatic cancer development (165). Novel nanomaterial-based strategies to exploit cuproptosis for therapeutic benefit (166). For example, tussah silk fibroin (TSF)-based nanoparticles (NPs) use TME-responsive release mechanisms to deliver copper and the cuproptosis-inducing drug ES directly to pancreatic cancer cells. Upon targeted delivery, TSF@ ES-Cu NPs induce cuproptosis, releasing damage-associated molecular patterns that activate antitumor immunity (166). This promotes dendritic cell maturation and macrophage M1 polarization, thereby reshaping the TME and enhancing immune responses. Collectively, these findings demonstrate that TSF@ ES-Cu NPs suppress pancreatic cancer growth through a dual mechanism of cuproptosis induction and immune microenvironment remodeling, offering a promising avenue for clinical translation. In addition, other research have explored the prognostic and therapeutic roles of cuproptosis-associated lncRNAs in pancreatic ductal adenocarcinoma (PAAD) (167). Researchers have developed a cuproptosis-immune-related (CIR) score to characterize the interaction between cuproptosis and the tumor immune microenvironment (168). By integrating single-cell sequencing and transcriptomic data, researchers have identified immune- and cuproptosis-related genes associated with PAAD and constructed a CIR score model (168). This score not only predicts prognosis in patients and the immune landscape but also reflects the tumor mutational burden (TMB), immune checkpoint sensitivity, and drug responsiveness. Patients in the high CIR score group exhibit higher TMB and poorer survival, whereas those in the low CIR score group exhibit stronger immune activation and greater potential responsiveness to immunotherapy. Mouse model experiments have validated the predictive power of the CIR score in guiding combination therapy involving immunotherapy, targeted therapy and chemotherapy (168). Such combined regimens significantly inhibit PAAD progression, highlighting the translational potential of cuproptosis-based prognostic markers in personalized immunotherapy. Mechanistically, these findings indicate that the cuproptosis-metabolic reprogramming-immune activation axis represents a novel paradigm for the systemic treatment of pancreatic cancer. The integration of copper homeostasis regulation into individualized therapeutic frameworks provides both theoretical and practical foundations for precision medicine in PAAD.

Colorectal cancer (CRC)

CRC, one of the most common malignant tumors of the digestive system, is the third most commonly diagnosed cancer worldwide, with approximately 1.9 million new cases annually (169). Cuproptosis-related genes, such as FDX1, SDHB, DLAT and DLST, are expressed at higher levels in normal compared with tumor tissues (170). Moreover, higher expression of these genes in tumor tissue is associated with better prognosis (170). When ES, a copper carrier, combines with copper, the proliferation of CRC cells is significantly inhibited and apoptosis is promoted. This effect is markedly suppressed by the copper chelator TTM, further confirming the mechanism of copper-induced cell death. Furthermore, 2-deoxy-D-glucose, a glycolysis inhibitor, can significantly enhance cuproptosis (170). In addition, galactose promotes oxidative phosphorylation by inhibiting the glycolytic pathway in tumor cells, thereby enhancing copper-induced cell death (170). These results indicate that the inhibition of glycolysis can increase the sensitivity of tumor cells to cuproptosis. Studies have also shown that 4-octyl itaconate (4-OI), a cell-permeable derivative of itaconic acid, inhibits glycolysis by targeting the key glycolytic enzyme GAPDH, thereby enhancing cuproptosis. 4-OI suppresses GAPDH activity, decreases lactate production and subsequently promotes copper-induced cell death. Furthermore, in vivo experiments demonstrated that 4-OI has significant antitumor effects and that its combination with ES markedly decreases tumor volume. Yang et al (170) provided new insights into the role of cuproptosis in CRC and revealed that 4-OI enhances copper-induced cell death by inhibiting glycolysis. These findings not only provide a novel therapeutic strategy for CRC but also establish theoretical support for cuproptosis as a potential anticancer approach. Cuproptosis-associated genes demonstrate important prognostic value in colon adenocarcinoma because their expression levels are associated with patient survival, TME characteristics and drug sensitivity (171). To the best of our knowledge, the aforementioned study was the first to systematically analyze the roles of genes associated with cuproptosis in colon adenocarcinoma and to elucidate their potential mechanisms in tumor progression and immune microenvironment regulation. Similarly, genes associated with cuproptosis serve a significant role in the prognosis and treatment of rectal adenocarcinoma (172). Copper chelators or carriers may synergize with chemotherapy or immunotherapy, thereby improving the prognosis of liver-metastatic CRC (173).

Status and prospects for the clinical application of cuproptosis-associated drugs

Cuproptosis-associated drugs have demonstrated antitumor potential in clinical trials for gastrointestinal cancer (174,175). For example, the disulfiram/copper complex inhibits the proliferation of GC cells by inducing oxidative stress and DNA damage while increasing the sensitivity of tumor cells to chemotherapeutic agents (174). In addition, TTM, a copper chelator, exerts antiangiogenic and chemosensitizing effects in metastatic CRC (175). The potential clinical applications of these drugs include combining them with chemotherapy to enhance therapeutic efficacy, targeting copper metabolism-associated proteins to improve treatment precision and using biomarkers such as serum copper levels to guide medication strategies. Nevertheless, further research is needed to optimize the design of cuproptosis-associated drugs to improve their efficacy and decrease side effects. However, future in-depth studies on the molecular mechanisms of these drugs and additional clinical trials to verify their safety and effectiveness are needed.

Conclusion

As a newly identified form of regulated cell death, cuproptosis has become a prominent focus in tumor biology, particularly in gastrointestinal cancer. The present review summarizes the mechanistic pathways and therapeutic potential of cuproptosis, emphasizing key molecular events such as mitochondrial damage, oxidative stress and protein lipoylation, as well as copper imbalance and its association with tumor initiation and development. Current evidence indicates that disruption of copper metabolism is associated with tumor growth and progression. By modulating intracellular copper levels, cuproptosis inhibits tumor cell proliferation through various mechanisms, including the induction of PCD and the enhancement of antitumor immune responses. Moreover, cuproptosis-associated drugs, such as disulfiram/copper complexes and TTM, have demonstrated promising anticancer potential in preclinical and clinical studies because they induce oxidative stress, increase chemosensitivity and inhibit angiogenesis. However, the clinical application of these drugs faces several challenges, including limited efficacy, toxicity management and a lack of large-scale validation. Future studies should further clarify the molecular mechanisms of cuproptosis, develop novel copper-dependent therapeutic agents with improved selectivity and safety and conduct clinical trials to assess their translational value. In conclusion, cuproptosis offers a novel conceptual and therapeutic direction for gastrointestinal oncology, holding promise for treatment options that could improve prognosis and survival outcomes.

Availability of data and materials

Not applicable.

Authors' contributions

LZ and YC conceived the study. LT, JZ and BT edited the manuscript. Data authentication is not applicable. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgements

Not applicable.

Funding

The present study was supported by grants from the National Natural Science Foundation of China (grant no. 82073087) and Medical Research Union Fund for High-quality Health Development of Guizhou Province (grant no. 2024GZYXKYJJXM0019).

References

1 

Lutsenko S, Roy S and Tsvetkov P: Mammalian copper homeostasis: Physiological roles and molecular mechanisms. Physiol Rev. 105:441–491. 2025. View Article : Google Scholar :

2 

Locatelli M and Farina C: Role of copper in central nervous system physiology and pathology. Neural Regen Res. 20:1058–1068. 2025. View Article : Google Scholar

3 

Chen G, Li J, Han H, Du R and Wang X: Physiological and molecular mechanisms of plant responses to copper stress. Int J Mol Sci. 23:129502022. View Article : Google Scholar : PubMed/NCBI

4 

Verdejo-Torres O, Klein DC, Novoa-Aponte L, Carrazco-Carrillo J, Bonilla-Pinto D, Rivera A, Bakhshian A, Fitisemanu FM, Jiménez-González ML, Flinn L, et al: Cysteine rich intestinal protein 2 is a copper-responsive regulator of skeletal muscle differentiation and metal homeostasis. PLoS Genet. 20:e10114952024. View Article : Google Scholar : PubMed/NCBI

5 

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

6 

Guo Z, Chen D, Yao L, Sun Y, Li D, Le J, Dian Y, Zeng F, Chen X and Deng G: The molecular mechanism and therapeutic landscape of copper and cuproptosis in cancer. Signal Transduct Target Ther. 10:1492025. View Article : Google Scholar : PubMed/NCBI

7 

Yang Y, Wu J, Wang L, Ji G and Dang Y: Copper homeostasis and cuproptosis in health and disease. MedComm (2020). 5:e7242024. View Article : Google Scholar : PubMed/NCBI

8 

Myint ZW, Oo TH, Thein KZ, Tun AM and Saeed H: Copper deficiency anemia: Review article. Ann Hematol. 97:1527–1534. 2018. View Article : Google Scholar : PubMed/NCBI

9 

Gromadzka G, Tarnacka B, Flaga A and Adamczyk A: Copper dyshomeostasis in neurodegenerative diseases-therapeutic implications. Int J Mol Sci. 21:92592020. View Article : Google Scholar : PubMed/NCBI

10 

Sailer J, Nagel J, Akdogan B, Jauch AT, Engler J, Knolle PA and Zischka H: Deadly excess copper. Redox Biol. 75:1032562024. View Article : Google Scholar : PubMed/NCBI

11 

Zhang S, Huang Q, Ji T, Li Q and Hu C: Copper homeostasis and copper-induced cell death in tumor immunity: Implications for therapeutic strategies in cancer immunotherapy. Biomark Res. 12:1302024. View Article : Google Scholar : PubMed/NCBI

12 

Wang XR and Cull B: Apoptosis and autophagy: Current understanding in tick-pathogen interactions. Front Cell Infect Microbiol. 12:7844302022. View Article : Google Scholar : PubMed/NCBI

13 

Hong Y, He J, Deng D, Liu Q, Zu X and Shen Y: Targeting kinases that regulate programmed cell death: A new therapeutic strategy for breast cancer. J Transl Med. 23:4392025. View Article : Google Scholar : PubMed/NCBI

14 

Bertheloot D, Latz E and Franklin BS: Necroptosis, pyroptosis and apoptosis: An intricate game of cell death. Cell Mol Immunol. 18:1106–1121. 2021. View Article : Google Scholar : PubMed/NCBI

15 

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

16 

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

17 

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

18 

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

19 

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

20 

Zhao G, Sun H, Zhang T and Liu JX: Copper induce zebrafish retinal developmental defects via triggering stresses and apoptosis. Cell Commun Signal. 18:452020. View Article : Google Scholar : PubMed/NCBI

21 

Ostrakhovitch EA and Cherian MG: Role of p53 and reactive oxygen species in apoptotic response to copper and zinc in epithelial breast cancer cells. Apoptosis. 10:111–121. 2005. View Article : Google Scholar : PubMed/NCBI

22 

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

23 

Vana F, Szabo Z, Masarik M and Kratochvilova M: The interplay of transition metals in ferroptosis and pyroptosis. Cell Div. 19:242024. View Article : Google Scholar : PubMed/NCBI

24 

Xue Q, Yan D, Chen X, Li X, Kang R, Klionsky DJ, Kroemer G, Chen X, Tang D and Liu J: Copper-dependent autophagic degradation of GPX4 drives ferroptosis. Autophagy. 19:1982–1996. 2023. View Article : Google Scholar : PubMed/NCBI

25 

Fu Y, Zeng S, Wang Z, Huang H, Zhao X and Li M: Mechanisms of copper-induced autophagy and links with human diseases. Pharmaceuticals (Basel). 18:992025. View Article : Google Scholar : PubMed/NCBI

26 

Xue Q, Kang R, Klionsky DJ, Tang D, Liu J and Chen X: Copper metabolism in cell death and autophagy. Autophagy. 19:2175–2195. 2023. View Article : Google Scholar : PubMed/NCBI

27 

Wang Y, Qiao S, Wang P, Li M, Ma X, Wang H and Dong J: Copper's new role in cancer: How cuproptosis-related genes could revolutionize glioma treatment. BMC Cancer. 25:8592025. View Article : Google Scholar : PubMed/NCBI

28 

Tang X, Yan Z, Miao Y, Ha W, Li Z, Yang L and Mi D: Copper in cancer: From limiting nutrient to therapeutic target. Front Oncol. 13:12091562023. View Article : Google Scholar : PubMed/NCBI

29 

Shoda K, Kawaguchi Y, Maruyama S and Ichikawa D: Essential updates 2023/2024: Recent advances of multimodal approach in patients for gastric cancer. Ann Gastroenterol Surg. 9:1119–1127. 2025. View Article : Google Scholar : PubMed/NCBI

30 

Ricci AD, Rizzo A and Brandi G: DNA damage response alterations in gastric cancer: knocking down a new wall. Future Oncol. 17:865–868. 2021. View Article : Google Scholar : PubMed/NCBI

31 

Rizzo A and Ricci AD: Challenges and future trends of hepatocellular carcinoma immunotherapy. Int J Mol Sci. 23:113632022. View Article : Google Scholar : PubMed/NCBI

32 

Vitale E, Rizzo A, Santa K and Jirillo E: Associations between 'Cancer Risk', 'Inflammation' and 'Metabolic Syndrome': A scoping review. Biology (Basel). 13:3522024.

33 

Brandi G, Ricci AD, Rizzo A, Zanfi C, Tavolari S, Palloni A, De Lorenzo S, Ravaioli M and Cescon M: Is post-transplant chemotherapy feasible in liver transplantation for colorectal cancer liver metastases? Cancer Commun (Lond). 40:461–464. 2020. View Article : Google Scholar : PubMed/NCBI

34 

Mason KE: A conspectus of research on copper metabolism and requirements of man. J Nutr. 109:1979–2066. 1979. View Article : Google Scholar : PubMed/NCBI

35 

Eisses JF and Kaplan JH: The mechanism of copper uptake mediated by human CTR1: A mutational analysis. J Biol Chem. 280:37159–37168. 2005. View Article : Google Scholar : PubMed/NCBI

36 

Zischka H and Einer C: Mitochondrial copper homeostasis and its derailment in Wilson disease. Int J Biochem Cell Biol. 102:71–75. 2018. View Article : Google Scholar : PubMed/NCBI

37 

Jiayi H, Ziyuan T, Tianhua X, Mingyu Z, Yutong M, Jingyu W, Hongli Z and Li S: Copper homeostasis in chronic kidney disease and its crosstalk with ferroptosis. Pharmacol Res. 202:1071392024. View Article : Google Scholar : PubMed/NCBI

38 

Chen J, Jiang Y, Shi H, Peng Y, Fan X and Li C: The molecular mechanisms of copper metabolism and its roles in human diseases. Pflugers Arch. 472:1415–1429. 2020. View Article : Google Scholar : PubMed/NCBI

39 

Lutsenko S: Dynamic and cell-specific transport networks for intracellular copper ions. J Cell Sci. 134:jcs2405232021. View Article : Google Scholar : PubMed/NCBI

40 

McKie AT, Barrow D, Latunde-Dada GO, Rolfs A, Sager G, Mudaly E, Mudaly M, Richardson C, Barlow D, Bomford A, et al: An iron-regulated ferric reductase associated with the absorption of dietary iron. Science. 291:1755–1759. 2001. View Article : Google Scholar : PubMed/NCBI

41 

Mastrogiannaki M, Matak P, Keith B, Simon MC, Vaulont S and Peyssonnaux C: HIF-2alpha, but not HIF-1alpha, promotes iron absorption in mice. J Clin Invest. 119:1159–1166. 2009. View Article : Google Scholar : PubMed/NCBI

42 

Kawamata H and Manfredi G: Import, maturation, and function of SOD1 and its copper chaperone CCS in the mitochondrial intermembrane space. Antioxid Redox Signal. 13:1375–1384. 2010. View Article : Google Scholar : PubMed/NCBI

43 

Wang Z, Jin D, Zhou S, Dong N, Ji Y, An P, Wang J, Luo Y and Luo J: Regulatory roles of copper metabolism and cuproptosis in human cancers. Front Oncol. 13:11234202023. View Article : Google Scholar : PubMed/NCBI

44 

Ohrvik H and Thiele DJ: How copper traverses cellular membranes through the mammalian copper transporter 1, Ctr1. Ann N Y Acad Sci. 1314:32–41. 2014. View Article : Google Scholar : PubMed/NCBI

45 

Yu Q, Xiao Y, Guan M, Zhang X, Yu J, Han M and Li Z: Copper metabolism in osteoarthritis and its relation to oxidative stress and ferroptosis in chondrocytes. Front Mol Biosci. 11:14724922024. View Article : Google Scholar : PubMed/NCBI

46 

Kim H, Wu X and Lee J: SLC31 (CTR) family of copper transporters in health and disease. Mol Aspects Med. 34:561–570. 2013. View Article : Google Scholar : PubMed/NCBI

47 

Cobine PA, Moore SA and Leary SC: Getting out what you put in: Copper in mitochondria and its impacts on human disease. Biochim Biophys Acta Mol Cell Res. 1868:1188672021. View Article : Google Scholar

48 

Chen Z, Li YY and Liu X: Copper homeostasis and copper-induced cell death: Novel targeting for intervention in the pathogenesis of vascular aging. Biomed Pharmacother. 169:1158392023. View Article : Google Scholar

49 

Andersson M, Mattle D, Sitsel O, Klymchuk T, Nielsen AM, Moller LB, White SH, Nissen P and Gourdon P: Copper-transporting P-type ATPases use a unique ion-release pathway. Nat Struct Mol Biol. 21:43–48. 2014. View Article : Google Scholar :

50 

Telianidis J, Hung YH, Materia S and Fontaine SL: Role of the P-Type ATPases, ATP7A and ATP7B in brain copper homeostasis. Front Aging Neurosci. 5:442013. View Article : Google Scholar : PubMed/NCBI

51 

Fanni D, Pilloni L, Orru S, Coni P, Liguori C, Serra S, Lai ML, Uccheddu A, Contu L, Van Eyken P and Faa G: Expression of ATP7B in normal human liver. Eur J Histochem. 49:371–378. 2005. View Article : Google Scholar : PubMed/NCBI

52 

Kaler SG: ATP7A-related copper transport diseases-emerging concepts and future trends. Nat Rev Neurol. 7:15–29. 2011. View Article : Google Scholar : PubMed/NCBI

53 

Yang GM, Xu L, Wang RM, Tao X, Zheng ZW, Chang S, Ma D, Zhao C, Dong Y, Wu S, et al: Structures of the human Wilson disease copper transporter ATP7B. Cell Rep. 42:1124172023. View Article : Google Scholar : PubMed/NCBI

54 

Fukai T, Ushio-Fukai M and Kaplan JH: Copper transporters and copper chaperones: Roles in cardiovascular physiology and disease. Am J Physiol Cell Physiol. 315:C186–C201. 2018. View Article : Google Scholar : PubMed/NCBI

55 

Ugarte M, Osborne NN, Brown LA and Bishop PN: Iron, zinc, and copper in retinal physiology and disease. Surv Ophthalmol. 58:585–609. 2013. View Article : Google Scholar : PubMed/NCBI

56 

Ishida S, Andreux P, Poitry-Yamate C, Auwerx J and Hanahan D: Bioavailable copper modulates oxidative phosphorylation and growth of tumors. Proc Natl Acad Sci USA. 110:19507–19512. 2013. View Article : Google Scholar : PubMed/NCBI

57 

Wooton-Kee CR, Robertson M, Zhou Y, Dong B, Sun Z, Kim KH, Liu H, Xu Y, Putluri N, Saha P, et al: Metabolic dysregulation in the Atp7b(-/-) Wilson's disease mouse model. Proc Natl Acad Sci USA. 117:2076–2083. 2020. View Article : Google Scholar : PubMed/NCBI

58 

Yang H, Ralle M, Wolfgang MJ, Dhawan N, Burkhead JL, Rodriguez S, Kaplan JH, Wong GW, Haughey N and Lutsenko S: Copper-dependent amino oxidase 3 governs selection of metabolic fuels in adipocytes. PLoS Biol. 16:e20065192018. View Article : Google Scholar : PubMed/NCBI

59 

Kim E, Chufan EE, Kamaraj K and Karlin KD: Synthetic models for heme-copper oxidases. Chem Rev. 104:1077–1133. 2004. View Article : Google Scholar : PubMed/NCBI

60 

Anwar S, Sarwar T, Khan AA and Rahmani AH: Therapeutic applications and mechanisms of superoxide dismutase (SOD) in different pathogenesis. Biomolecules. 15:11302025. View Article : Google Scholar : PubMed/NCBI

61 

Grubman A and White AR: Copper as a key regulator of cell signalling pathways. Expert Rev Mol Med. 16:e112014. View Article : Google Scholar : PubMed/NCBI

62 

Heffern MC, Park HM, Au-Yeung HY, Van de Bittner GC, Ackerman CM, Stahl A and Chang CJ: In vivo bioluminescence imaging reveals copper deficiency in a murine model of nonalcoholic fatty liver disease. Proc Natl Acad Sci USA. 113:14219–14224. 2016. View Article : Google Scholar : PubMed/NCBI

63 

Herranz N, Dave N, Millanes-Romero A, Morey L, Diaz VM, Lorenz-Fonfria V, Gutierrez-Gallego R, Jerónimo C, Di Croce L, García de Herreros A and Peiró S: Lysyl oxidase-like 2 deaminates lysine 4 in histone H3. Mol Cell. 46:369–376. 2012. View Article : Google Scholar : PubMed/NCBI

64 

Percival SS: Copper and immunity. Am J Clin Nutr. 67(5 Suppl): 1064S–1068S. 1998. View Article : Google Scholar : PubMed/NCBI

65 

An Y, Li S, Huang X, Chen X, Shan H and Zhang M: The role of copper homeostasis in brain disease. Int J Mol Sci. 23:138502022. View Article : Google Scholar : PubMed/NCBI

66 

Cheng YF, Zhao YJ, Chen C and Zhang F: Heavy metals toxicity: Mechanism, health effects, and therapeutic interventions. MedComm (2020). 6:e702412025. View Article : Google Scholar : PubMed/NCBI

67 

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

68 

Kong R and Sun G: Targeting copper metabolism: A promising strategy for cancer treatment. Front Pharmacol. 14:12034472023. View Article : Google Scholar : PubMed/NCBI

69 

Nan L, Yuan W, Guodong C and Yonghui H: Multitargeting strategy using tetrathiomolybdate and lenvatinib: Maximizing antiangiogenesis activity in a preclinical liver cancer model. Anticancer Agents Med Chem. 23:786–793. 2023. View Article : Google Scholar

70 

Cohen R, Raeisi M, Chibaudel B, Yothers G, Goldberg RM, Bachet JB, Wolmark N, Yoshino T, Schmoll HJ, Haller DG, et al: Impact of tumor and node stages on the efficacy of adjuvant oxaliplatin-based chemotherapy in stage III colon cancer patients: An ACCENT pooled analysis. ESMO Open. 10:1044812025. View Article : Google Scholar

71 

Nawar MF and Turler A: New strategies for a sustainable (99m) Tc supply to meet increasing medical demands: Promising solutions for current problems. Front Chem. 10:9262582022. View Article : Google Scholar

72 

Wang W, Mo W, Hang Z, Huang Y, Yi H, Sun Z and Lei A: Cuproptosis: Harnessing transition metal for cancer therapy. ACS Nano. 17:19581–19599. 2023. View Article : Google Scholar : PubMed/NCBI

73 

Ge EJ, Bush AI, Casini A, Cobine PA, Cross JR, DeNicola GM, Dou QP, Franz KJ, Gohil VM, Gupta S, et al: Connecting copper and cancer: From transition metal signalling to metalloplasia. Nat Rev Cancer. 22:102–113. 2022. View Article : Google Scholar :

74 

Li Y: Copper homeostasis: Emerging target for cancer treatment. IUBMB Life. 72:1900–1908. 2020. View Article : Google Scholar : PubMed/NCBI

75 

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

76 

Shim D and Han J: Coordination chemistry of mitochondrial copper metalloenzymes: Exploring implications for copper dyshomeostasis in cell death. BMB Rep. 56:575–583. 2023. View Article : Google Scholar : PubMed/NCBI

77 

Cheng H, Yang B, Ke T, Li S, Yang X, Aschner M and Chen P: Mechanisms of metal-induced mitochondrial dysfunction in neurological disorders. Toxics. 9:1422021. View Article : Google Scholar : PubMed/NCBI

78 

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

79 

Sun L, Zhang Y, Yang B, Sun S, Zhang P, Luo Z, Feng T, Cui Z, Zhu T, Li Y, et al: Lactylation of METTL16 promotes cuproptosis via m(6)A-modification on FDX1 mRNA in gastric cancer. Nat Commun. 14:65232023. View Article : Google Scholar : PubMed/NCBI

80 

Polishchuk EV, Concilli M, Iacobacci S, Chesi G, Pastore N, Piccolo P, Paladino S, Baldantoni D, van IJzendoorn SC, Chan J, et al: Wilson disease protein ATP7B utilizes lysosomal exocytosis to maintain copper homeostasis. Dev Cell. 29:686–700. 2014. View Article : Google Scholar : PubMed/NCBI

81 

Yang L, Zhang Y, Wang Y, Jiang P, Liu F and Feng N: Ferredoxin 1 is a cuproptosis-key gene responsible for tumor immunity and drug sensitivity: A pan-cancer analysis. Front Pharmacol. 13:9381342022. View Article : Google Scholar : PubMed/NCBI

82 

Sheftel AD, Stehling O, Pierik AJ, Elsasser HP, Muhlenhoff U, Webert H, Hobler A, Hannemann F, Bernhardt R and Lill R: Humans possess two mitochondrial ferredoxins, Fdx1 and Fdx2, with distinct roles in steroidogenesis, heme, and Fe/S cluster biosynthesis. Proc Natl Acad Sci USA. 107:11775–11780. 2010. View Article : Google Scholar : PubMed/NCBI

83 

Huang X, Wang T, Ye J, Feng H and Zhang X, Ma X, Wang B, Huang Y and Zhang X: FDX1 expression predicts favourable prognosis in clear cell renal cell carcinoma identified by bioinformatics and tissue microarray analysis. Front Genet. 13:9947412022. View Article : Google Scholar : PubMed/NCBI

84 

Mayr JA, Zimmermann FA, Fauth C, Bergheim C, Meierhofer D, Radmayr D, Zschocke J, Koch J and Sperl W: Lipoic acid synthetase deficiency causes neonatal-onset epilepsy, defective mitochondrial energy metabolism, and glycine elevation. Am J Hum Genet. 89:792–797. 2011. View Article : Google Scholar : PubMed/NCBI

85 

Cai Y, He Q, Liu W, Liang Q, Peng B, Li J, Zhang W, Kang F, Hong Q, Yan Y, et al: Comprehensive analysis of the potential cuproptosis-related biomarker LIAS that regulates prognosis and immunotherapy of pan-cancers. Front Oncol. 12:9521292022. View Article : Google Scholar : PubMed/NCBI

86 

Casteel J, Miernyk JA and Thelen JJ: Mapping the lipoylation site of Arabidopsis thaliana plastidial dihydrolipoamide S-acetyltransferase using mass spectrometry and site-directed mutagenesis. Plant Physiol Biochem. 49:1355–1361. 2011. View Article : Google Scholar : PubMed/NCBI

87 

Rumping L, Tessadori F, Pouwels PJW, Vringer E, Wijnen JP, Bhogal AA, Savelberg SMC, Duran KJ, Bakkers MJG, Ramos RJJ, et al: GLS hyperactivity causes glutamate excess, infantile cataract and profound developmental delay. Hum Mol Genet. 28:96–104. 2019. View Article : Google Scholar

88 

Rumping L, Buttner B, Maier O, Rehmann H, Lequin M, Schlump JU, Schmitt B, Schiebergen-Bronkhorst B, Prinsen HCMT, Losa M, et al: Identification of a loss-of-function mutation in the context of glutaminase deficiency and neonatal epileptic encephalopathy. JAMA Neurol. 76:342–350. 2019. View Article : Google Scholar :

89 

Agarwal P, Sandey M, DeInnocentes P and Bird RC: Tumor suppressor gene p16/INK4A/CDKN2A-dependent regulation into and out of the cell cycle in a spontaneous canine model of breast cancer. J Cell Biochem. 114:1355–1363. 2013. View Article : Google Scholar

90 

Serrano M, Hannon GJ and Beach D: A new regulatory motif in cell-cycle control causing specific inhibition of cyclin D/CDK4. Nature. 366:704–407. 1993. View Article : Google Scholar : PubMed/NCBI

91 

Bian Z, Yu Y, Yang T, Quan C, Sun W and Fu S: Effect of tumor suppressor gene cyclin-dependent kinase inhibitor 2A wild-type and A148T mutant on the cell cycle of human ovarian cancer cells. Oncol Lett. 7:1229–1232. 2014. View Article : Google Scholar : PubMed/NCBI

92 

Samimi G, Safaei R, Katano K, Holzer AK, Rochdi M, Tomioka M, Goodman M and Howell SB: Increased expression of the copper efflux transporter ATP7A mediates resistance to cisplatin, carboplatin, and oxaliplatin in ovarian cancer cells. Clin Cancer Res. 10:4661–4669. 2004. View Article : Google Scholar : PubMed/NCBI

93 

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

94 

Mangala LS, Zuzel V, Schmandt R, Leshane ES, Halder JB, Armaiz-Pena GN, Spannuth WA, Tanaka T, Shahzad MM, Lin YG, et al: Therapeutic targeting of ATP7B in ovarian carcinoma. Clin Cancer Res. 15:3770–3780. 2009. View Article : Google Scholar : PubMed/NCBI

95 

Yun Y, Wang Y, Yang E and Jing X: Cuproptosis-related gene - SLC31A1, FDX1 and ATP7B-polymorphisms are associated with risk of lung cancer. Pharmgenomics Pers Med. 15:733–742. 2022.

96 

Wang D, Tian Z, Zhang P, Zhen L, Meng Q, Sun B, Xu X, Jia T and Li S: The molecular mechanisms of cuproptosis and its relevance to cardiovascular disease. Biomed Pharmacother. 163:1148302023. View Article : Google Scholar : PubMed/NCBI

97 

Halliwell B, Adhikary A, Dingfelder M and Dizdaroglu M: Hydroxyl radical is a significant player in oxidative DNA damage in vivo. Chem Soc Rev. 50:8355–8360. 2021. View Article : Google Scholar : PubMed/NCBI

98 

Chandimali N, Bak SG, Park EH, Lim HJ, Won YS, Kim EK, Park SI and Lee SJ: Free radicals and their impact on health and antioxidant defenses: A review. Cell Death Discov. 11:192025. View Article : Google Scholar : PubMed/NCBI

99 

Wong-Ekkabut J, Xu Z, Triampo W, Tang IM, Tieleman DP and Monticelli L: Effect of lipid peroxidation on the properties of lipid bilayers: A molecular dynamics study. Biophys J. 93:4225–4236. 2007. View Article : Google Scholar : PubMed/NCBI

100 

Kehm R, Baldensperger T, Raupbach J and Höhn A: Protein oxidation - Formation mechanisms, detection and relevance as biomarkers in human diseases. Redox Biol. 42:1019012021. View Article : Google Scholar : PubMed/NCBI

101 

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

102 

Chen W, Jiang T, Wang H, Tao S, Lau A, Fang D and Zhang DD: Does Nrf2 contribute to p53-mediated control of cell survival and death? Antioxid Redox Signal. 17:1670–1675. 2012. View Article : Google Scholar : PubMed/NCBI

103 

Shan D, Song J, Ren Y, Zhang Y, Ba Y, Luo P, Cheng Q, Xu H, Weng S, Zuo A, et al: Copper in cancer: Friend or foe? Metabolism, dysregulation, and therapeutic opportunities. Cancer Commun (Lond). 45:577–607. 2025. View Article : Google Scholar : PubMed/NCBI

104 

Dow JA: The essential roles of metal ions in insect homeostasis and physiology. Curr Opin Insect Sci. 23:43–50. 2017. View Article : Google Scholar : PubMed/NCBI

105 

Ding X, Jiang M, Jing H, Sheng W, Wang X, Han J and Wang L: Analysis of serum levels of 15 trace elements in breast cancer patients in Shandong, China. Environ Sci Pollut Res Int. 22:7930–7935. 2015. View Article : Google Scholar

106 

Kuo HW, Chen SF, Wu CC, Chen DR and Lee JH: Serum and tissue trace elements in patients with breast cancer in Taiwan. Biol Trace Elem Res. 89:1–11. 2002. View Article : Google Scholar : PubMed/NCBI

107 

Pavithra V, Sathisha TG, Kasturi K, Mallika DS, Amos SJ and Ragunatha S: Serum levels of metal ions in female patients with breast cancer. J Clin Diagn Res. 9:BC25–c27. 2015.PubMed/NCBI

108 

Feng JF, Lu L, Zeng P, Yang YH, Luo J, Yang YW and Wang D: Serum total oxidant/antioxidant status and trace element levels in breast cancer patients. Int J Clin Oncol. 17:575–583. 2012. View Article : Google Scholar

109 

Zowczak M, Iskra M, Torlinski L and Cofta S: Analysis of serum copper and zinc concentrations in cancer patients. Biol Trace Elem Res. 82:1–8. 2001. View Article : Google Scholar : PubMed/NCBI

110 

Diez M, Cerdan FJ, Arroyo M and Balibrea JL: Use of the copper/zinc ratio in the diagnosis of lung cancer. Cancer. 63:726–730. 1989. View Article : Google Scholar : PubMed/NCBI

111 

Jin Y, Zhang C, Xu H, Xue S, Wang Y, Hou Y, Kong Y and Xu Y: Combined effects of serum trace metals and polymorphisms of CYP1A1 or GSTM1 on non-small cell lung cancer: A hospital based case-control study in China. Cancer Epidemiol. 35:182–187. 2011. View Article : Google Scholar

112 

Oyama T, Matsuno K, Kawamoto T, Mitsudomi T, Shirakusa T and Kodama Y: Efficiency of serum copper/zinc ratio for differential diagnosis of patients with and without lung cancer. Biol Trace Elem Res. 42:115–127. 1994. View Article : Google Scholar : PubMed/NCBI

113 

Stepien M, Jenab M, Freisling H, Becker NP, Czuban M, Tjonneland A, Olsen A, Overvad K, Boutron-Ruault MC, Mancini FR, et al: Pre-diagnostic copper and zinc biomarkers and colorectal cancer risk in the European Prospective Investigation into Cancer and Nutrition cohort. Carcinogenesis. 38:699–707. 2017. View Article : Google Scholar : PubMed/NCBI

114 

Sohrabi M, Gholami A, Azar MH, Yaghoobi M, Shahi MM, Shirmardi S, Nikkhah M, Kohi Z, Salehpour D, Khoonsari MR, et al: Trace element and heavy metal levels in colorectal cancer: Comparison between cancerous and non-cancerous tissues. Biol Trace Elem Res. 183:1–8. 2018. View Article : Google Scholar

115 

Margalioth EJ, Schenker JG and Chevion M: Copper and zinc levels in normal and malignant tissues. Cancer. 52:868–872. 1983. View Article : Google Scholar : PubMed/NCBI

116 

Yaman M, Kaya G and Yekeler H: Distribution of trace metal concentrations in paired cancerous and non-cancerous human stomach tissues. World J Gastroenterol. 13:612–618. 2007. View Article : Google Scholar : PubMed/NCBI

117 

Khanna SS and Karjodkar FR: Circulating immune complexes and trace elements (Copper, Iron and Selenium) as markers in oral precancer and cancer: A randomised, controlled clinical trial. Head Face Med. 2:332006. View Article : Google Scholar

118 

Baltaci AK, Dundar TK, Aksoy F and Mogulkoc R: Changes in the serum levels of trace elements before and after the operation in thyroid cancer patients. Biol Trace Elem Res. 175:57–64. 2017. View Article : Google Scholar

119 

Basu S, Singh MK, Singh TB, Bhartiya SK, Singh SP and Shukla VK: Heavy and trace metals in carcinoma of the gallbladder. World J Surg. 37:2641–2646. 2013. View Article : Google Scholar : PubMed/NCBI

120 

Saleh SAK, Adly HM, Abdelkhaliq AA and Nassir AM: Serum levels of selenium, zinc, copper, manganese, and iron in prostate cancer patients. Curr Urol. 14:44–49. 2020. View Article : Google Scholar : PubMed/NCBI

121 

Blockhuys S, Celauro E, Hildesjo C, Feizi A, Stal O, Fierro-Gonzalez JC and Wittung-Stafshede P: Defining the human copper proteome and analysis of its expression variation in cancers. Metallomics. 9:112–123. 2017. View Article : Google Scholar

122 

Wang W, Lu K, Jiang X, Wei Q, Zhu L, Wang X, Jin H and Feng L: Ferroptosis inducers enhanced cuproptosis induced by copper ionophores in primary liver cancer. J Exp Clin Cancer Res. 42:1422023. View Article : Google Scholar : PubMed/NCBI

123 

Harris ED: A Requirement for Copper in Angiogenesis. Nutr Rev. 62:60–64. 2004. View Article : Google Scholar : PubMed/NCBI

124 

De Luca A, Barile A, Arciello M and Rossi L: Copper homeostasis as target of both consolidated and innovative strategies of anti-tumor therapy. J Trace Elem Med Biol. 55:204–213. 2019. View Article : Google Scholar : PubMed/NCBI

125 

Barker HE, Cox TR and Erler JT: The rationale for targeting the LOX family in cancer. Nat Rev Cancer. 12:540–552. 2012. View Article : Google Scholar : PubMed/NCBI

126 

Groleau J, Dussault S, Haddad P, Turgeon J, Menard C, Chan JS and Rivard A: Essential role of copper-zinc superoxide dismutase for ischemia-induced neovascularization via modulation of bone marrow-derived endothelial progenitor cells. Arterioscler Thromb Vasc Biol. 30:2173–2181. 2010. View Article : Google Scholar : PubMed/NCBI

127 

Lowndes SA and Harris AL: Copper chelation as an antiangiogenic therapy. Oncol Res. 14:529–539. 2004. View Article : Google Scholar

128 

Pannecoeck R, Serruys D, Benmeridja L, Delanghe JR, van Geel N, Speeckaert R and Speeckaert MM: Vascular adhesion protein-1: Role in human pathology and application as a biomarker. Crit Rev Clin Lab Sci. 52:284–300. 2015. View Article : Google Scholar : PubMed/NCBI

129 

Wong CC, Tse AP, Huang YP, Zhu YT, Chiu DK, Lai RK, Au SL, Kai AK, Lee JM, Wei LL, et al: Lysyl oxidase-like 2 is critical to tumor microenvironment and metastatic niche formation in hepatocellular carcinoma. Hepatology. 60:1645–1658. 2014. View Article : Google Scholar : PubMed/NCBI

130 

Salvador F, Martin A, Lopez-Menendez C, Moreno-Bueno G, Santos V, Vazquez-Naharro A, Santamaria PG, Morales S, Dubus PR, Muinelo-Romay L, et al: Lysyl oxidase-like protein LOXL2 promotes lung metastasis of breast cancer. Cancer Res. 77:5846–5859. 2017. View Article : Google Scholar : PubMed/NCBI

131 

Cheng F, Peng G, Lu Y, Wang K, Ju Q, Ju Y and Ouyang M: Relationship between copper and immunity: The potential role of copper in tumor immunity. Front Oncol. 12:10191532022. View Article : Google Scholar : PubMed/NCBI

132 

Kang J, Lin C, Chen J and Liu Q: Copper induces histone hypoacetylation through directly inhibiting histone acetyltransferase activity. Chem Biol Interact. 148:115–123. 2004. View Article : Google Scholar : PubMed/NCBI

133 

International Agency for Research on Cancer: Oesophageal cancer. GLOBOCAN. 2022, https://www.iarc.who.int/cancer-type/oesophageal-cancer/. Accessed Dec 2, 2025

134 

Zhang Y, Liu S, Zhou S, Yu D, Gu J, Qin Q, Cheng Y and Sun X: Focal adhesion kinase: Insight into its roles and therapeutic potential in oesophageal cancer. Cancer Lett. 496:93–103. 2021. View Article : Google Scholar

135 

Liu XS, Yuan LL, Gao Y, Zhou LM, Yang JW and Pei ZJ: Overexpression of METTL3 associated with the metabolic status on (18)F-FDG PET/CT in patients with esophageal carcinoma. J Cancer. 11:4851–4860. 2020. View Article : Google Scholar : PubMed/NCBI

136 

Liu XS, Zhang Y, Liu ZY, Gao Y, Yuan LL, Zeng DB, Tan F, Wan HB and Pei ZJ: METTL3 as a novel diagnosis and treatment biomarker and its association with glycolysis, cuproptosis and ceRNA in oesophageal carcinoma. J Cell Mol Med. 28:e181952024. View Article : Google Scholar : PubMed/NCBI

137 

Lin K, Hu K, Chen Q and Wu J: The function and immune role of cuproptosis associated hub gene in Barrett's esophagus and esophageal adenocarcinoma. Biosci Trends. 17:381–392. 2023. View Article : Google Scholar : PubMed/NCBI

138 

Bas O, Sahin TK, Karahan L, Rizzo A and Guven DC: Prognostic significance of the cachexia index (CXI) in patients with cancer: A systematic review and meta-analysis. Clin Nutr ESPEN. 68:240–247. 2025. View Article : Google Scholar : PubMed/NCBI

139 

International Agency for Research on Cancer: Stomach cancer. GLOBOCAN. 2022, https://www.iarc.who.int/cancer-type/stomach-cancer/. Accessed Dec 2, 2025

140 

Wang H, Liu M, Zeng X, Zheng Y, Wang Y and Zhou Y: Cell death affecting the progression of gastric cancer. Cell Death Discov. 8:3772022. View Article : Google Scholar : PubMed/NCBI

141 

Song Q, Liu S, Wu D and Cai A: Multiple programmed cell death patterns predict the prognosis and drug sensitivity in gastric cancer. Front Immunol. 16:15114532025. View Article : Google Scholar : PubMed/NCBI

142 

Li Y, Sun W, Yuan S, Liu X, Zhang Z, Gu R, Li P and Gu X: The role of cuproptosis in gastric cancer. Front Immunol. 15:14356512024. View Article : Google Scholar : PubMed/NCBI

143 

Zhou Y, Dong C, Shen X, Wang P, Chen T, Li W, Sun X, Li P, Xu C, Duan K, et al: Targeting PTBP3-Mediated alternative splicing of COX11 induces cuproptosis for inhibiting gastric cancer peritoneal metastasis. Adv Sci (Weinh). 12:e24159832025. View Article : Google Scholar : PubMed/NCBI

144 

Zuo X, Lei Y, Ou S, Yuan X, Shi P, Li Q and Xu Y: Integration of cuproptosis-related gene signatures in stomach adenocarcinoma: Implications for prognostic prediction and therapeutic strategies in cancer drug resistance. Discov Oncol. 16:8852025. View Article : Google Scholar : PubMed/NCBI

145 

Xie XZ, Zuo L, Huang W, Fan QM, Weng YY, Yao WD, Jiang JL and Jin JQ: FDX1 as a novel biomarker and treatment target for stomach adenocarcinoma. World J Gastrointest Surg. 16:1803–1824. 2024. View Article : Google Scholar : PubMed/NCBI

146 

Lu L, Yang W, Gu Y, Jin L and Liang Z: The role of cuproptosis in the occurrence and development of gastric cancer. Front Pharmacol. 16:16642002025. View Article : Google Scholar : PubMed/NCBI

147 

Chen Y, Liao Y, Huang L and Luo Z: Exploring copper metabolism-induced cell death in gastric cancer: A single-cell RNA sequencing study and prognostic model development. Discov Oncol. 15:4822024. View Article : Google Scholar : PubMed/NCBI

148 

International Agency for Research on Cancer: Liver cancer. GLOBOCAN. 2022, https://www.iarc.who.int/cancer-type/liver-cancer/. Accessed Dec 2, 2025

149 

Yu M, Chen Z, Zhou Q, Zhang B, Huang J, Jin L, Zhou B, Liu S, Yan J, Li X, et al: PARG inhibition limits HCC progression and potentiates the efficacy of immune checkpoint therapy. J Hepatol. 77:140–151. 2022. View Article : Google Scholar : PubMed/NCBI

150 

Huang H, Tsui YM and Ng IO: Fueling HCC dynamics: Interplay between tumor microenvironment and tumor initiating cells. Cell Mol Gastroenterol Hepatol. 15:1105–1116. 2023. View Article : Google Scholar : PubMed/NCBI

151 

Fang AP, Chen PY, Wang XY, Liu ZY, Zhang DM, Luo Y, Liao GC, Long JA, Zhong RH, Zhou ZG, et al: Serum copper and zinc levels at diagnosis and hepatocellular carcinoma survival in the Guangdong liver cancer cohort. Int J Cancer. 144:2823–2832. 2019. View Article : Google Scholar

152 

Gunjan D, Shalimar, Nadda N, Kedia S, Nayak B, Paul SB, Gamanagatti SR and Acharya SK: Hepatocellular carcinoma: An unusual complication of longstanding wilson disease. J Clin Exp Hepatol. 7:152–154. 2017. View Article : Google Scholar : PubMed/NCBI

153 

Haratake J, Horie A and Takeda S: Histochemical and ultrastructural study of copper-binding protein in hepatocellular carcinoma. Cancer. 60:1269–1274. 1987. View Article : Google Scholar : PubMed/NCBI

154 

Lefkowitch JH, Muschel R, Price JB, Marboe C and Braunhut S: Copper and copper-binding protein in fibrolamellar liver cell carcinoma. Cancer. 51:97–100. 1983. View Article : Google Scholar : PubMed/NCBI

155 

Davis CI, Gu X, Kiefer RM, Ralle M, Gade TP and Brady DC: Altered copper homeostasis underlies sensitivity of hepatocellular carcinoma to copper chelation. Metallomics. 12:1995–2008. 2020. View Article : Google Scholar : PubMed/NCBI

156 

Poznanski J, Soldacki D, Czarkowska-Paczek B, Bonna A, Kornasiewicz O, Krawczyk M, Bal W and Pączek L: Cirrhotic liver of liver transplant recipients accumulate silver and co-accumulate copper. Int J Mol Sci. 22:17822021. View Article : Google Scholar : PubMed/NCBI

157 

Li Z, Zhou H, Zhai X, Gao L, Yang M, An B, Xia T, Du G, Li X, Wang W and Jin B: MELK promotes HCC carcinogenesis through modulating cuproptosis-related gene DLAT-mediated mitochondrial function. Cell Death Dis. 14:7332023. View Article : Google Scholar : PubMed/NCBI

158 

Guo J, Cheng J, Zheng N, Zhang X, Dai X, Zhang L, Hu C, Wu X, Jiang Q, Wu D, et al: Copper promotes tumorigenesis by activating the PDK1-AKT oncogenic pathway in a copper transporter 1 dependent manner. Adv Sci (Weinh). 8:e20043032021. View Article : Google Scholar : PubMed/NCBI

159 

Quan B, Liu W, Yao F, Li M, Tang B, Li J, Ren Z and Yin X: LINC02362/hsa-miR-18a-5p/FDX1 axis suppresses proliferation and drives cuproptosis and oxaliplatin sensitivity of hepatocellular carcinoma. Am J Cancer Res. 13:5590–5609. 2023.PubMed/NCBI

160 

Oliveri V: Selective targeting of cancer cells by copper ionophores: An overview. Front Mol Biosci. 9:8418142022. View Article : Google Scholar : PubMed/NCBI

161 

Zhou C, Yang J, Liu T, Jia R, Yang L, Sun P and Zhao W: Copper metabolism and hepatocellular carcinoma: Current insights. Front Oncol. 13:11866592023. View Article : Google Scholar : PubMed/NCBI

162 

Zhao X, Chen J, Yin S, Shi J, Zheng M, He C, Meng H, Han Y, Han J, Guo J, et al: The expression of cuproptosis-related genes in hepatocellular carcinoma and their relationships with prognosis. Front Oncol. 12:9924682022. View Article : Google Scholar : PubMed/NCBI

163 

International Agency for Research on Cancer: Pancreatic cancer. GLOBOCAN. 2022, https://www.iarc.who.int/cancer-type/pancreatic-cancer/. Accessed Dec 2, 2025

164 

Leiphrakpam PD, Chowdhury S, Zhang M, Bajaj V, Dhir M and Are C: Trends in the global incidence of pancreatic cancer and a brief review of its histologic and molecular subtypes. J Gastrointest Cancer. 56:712025. View Article : Google Scholar : PubMed/NCBI

165 

Lener MR, Scott RJ, Wiechowska-Kozlowska A, Serrano-Fernandez P, Baszuk P, Jaworska-Bieniek K, Sukiennicki G, Marciniak W, Muszyńska M, Kładny J, et al: Serum concentrations of selenium and copper in patients diagnosed with pancreatic cancer. Cancer Res Treat. 48:1056–1064. 2016. View Article : Google Scholar : PubMed/NCBI

166 

Gao S, Ge H, Gao L, Gao Y, Tang S, Li Y, Yuan Z and Chen W: Silk fibroin nanoparticles for enhanced cuproptosis and immunotherapy in pancreatic cancer treatment. Adv Sci (Weinh). 12:e24176762025. View Article : Google Scholar : PubMed/NCBI

167 

Zhou C, Yi C, Yi Y, Qin W, Yan Y, Dong X, Zhang X, Huang Y, Zhang R, Wei J, et al: LncRNA PVT1 promotes gemcitabine resistance of pancreatic cancer via activating Wnt/β-catenin and autophagy pathway through modulating the miR-619-5p/Pygo2 and miR-619-5p/ATG14 axes. Mol Cancer. 19:1182020. View Article : Google Scholar

168 

Sun Y, Yao L, Man C, Gao Z, He R and Fan Y: Development and validation of cuproptosis-related lncRNAs associated with pancreatic cancer immune microenvironment based on single-cell. Front Immunol. 14:12207602023. View Article : Google Scholar : PubMed/NCBI

169 

International Agency for Research on Cancer: Colorectal cancer. GLOBOCAN. 2022, https://www.iarc.who.int/cancer-type/colorectal-cancer/. Accessed Dec 2, 2025

170 

Yang W, Wang Y, Huang Y, Yu J, Wang T, Li C, Yang L, Zhang P, Shi L, Yin Y, et al: 4-Octyl itaconate inhibits aerobic glycolysis by targeting GAPDH to promote cuproptosis in colorectal cancer. Biomed Pharmacother. 159:1143012023. View Article : Google Scholar : PubMed/NCBI

171 

Gu Y, Li C, Yan Y, Ming J, Li Y, Chao X and Wang T: Comprehensive analysis and verification of the prognostic significance of cuproptosis-related genes in colon adenocarcinoma. Int J Mol Sci. 25:118302024. View Article : Google Scholar : PubMed/NCBI

172 

Ma J, Lin H, Wang Y, Zhang Y, Zhou C, Tang D, Kagawa Y, Hou D and Jiang G: The unique role of cuproptosis in the prognosis and treatment of rectum adenocarcinoma. J Gastrointest Oncol. 16:367–385. 2025. View Article : Google Scholar : PubMed/NCBI

173 

Wang Y, Pei P, Yang K, Guo L and Li Y: Copper in colorectal cancer: From copper-related mechanisms to clinical cancer therapies. Clin Transl Med. 14:e17242024. View Article : Google Scholar : PubMed/NCBI

174 

Li Y, Chen F, Chen J, Chan S, He Y, Liu W and Zhang G: Disulfiram/Copper induces antitumor activity against both nasopharyngeal cancer cells and cancer-associated fibroblasts through ROS/MAPK and ferroptosis pathways. Cancers (Basel). 12:1382020. View Article : Google Scholar : PubMed/NCBI

175 

Chan N, Willis A, Kornhauser N, Ward MM, Lee SB, Nackos E, Seo BR, Chuang E, Cigler T, Moore A, et al: Correction: Influencing the tumor microenvironment: A phase II study of copper depletion using tetrathiomolybdate in patients with breast cancer at high risk for recurrence and in preclinical models of lung metastases. Clin Cancer Res. 26:50512020. View Article : Google Scholar : PubMed/NCBI

Related Articles

  • Abstract
  • View
  • Download
  • Twitter
Copy and paste a formatted citation
Spandidos Publications style
Zhang L, Cheng Y, Tang L, Zhu J and Tuo B: Role of cuproptosis in digestive system tumors (Review). Int J Mol Med 57: 56, 2026.
APA
Zhang, L., Cheng, Y., Tang, L., Zhu, J., & Tuo, B. (2026). Role of cuproptosis in digestive system tumors (Review). International Journal of Molecular Medicine, 57, 56. https://doi.org/10.3892/ijmm.2026.5727
MLA
Zhang, L., Cheng, Y., Tang, L., Zhu, J., Tuo, B."Role of cuproptosis in digestive system tumors (Review)". International Journal of Molecular Medicine 57.3 (2026): 56.
Chicago
Zhang, L., Cheng, Y., Tang, L., Zhu, J., Tuo, B."Role of cuproptosis in digestive system tumors (Review)". International Journal of Molecular Medicine 57, no. 3 (2026): 56. https://doi.org/10.3892/ijmm.2026.5727
Copy and paste a formatted citation
x
Spandidos Publications style
Zhang L, Cheng Y, Tang L, Zhu J and Tuo B: Role of cuproptosis in digestive system tumors (Review). Int J Mol Med 57: 56, 2026.
APA
Zhang, L., Cheng, Y., Tang, L., Zhu, J., & Tuo, B. (2026). Role of cuproptosis in digestive system tumors (Review). International Journal of Molecular Medicine, 57, 56. https://doi.org/10.3892/ijmm.2026.5727
MLA
Zhang, L., Cheng, Y., Tang, L., Zhu, J., Tuo, B."Role of cuproptosis in digestive system tumors (Review)". International Journal of Molecular Medicine 57.3 (2026): 56.
Chicago
Zhang, L., Cheng, Y., Tang, L., Zhu, J., Tuo, B."Role of cuproptosis in digestive system tumors (Review)". International Journal of Molecular Medicine 57, no. 3 (2026): 56. https://doi.org/10.3892/ijmm.2026.5727
Follow us
  • Twitter
  • LinkedIn
  • Facebook
About
  • Spandidos Publications
  • Careers
  • Cookie Policy
  • Privacy Policy
How can we help?
  • Help
  • Live Chat
  • Contact
  • Email to our Support Team