Open Access

New advances of nanozymes for the diagnosis and treatment of digestive system diseases (Review)

  • Authors:
    • Daihan Xie
    • Lixin Xie
    • Chao Fang
    • Zhefei Du
    • Zhenyu Cao
    • Chunxia Su
    • Yu Huo
  • View Affiliations

  • Published online on: August 28, 2025     https://doi.org/10.3892/ijmm.2025.5617
  • Article Number: 176
  • Copyright: © Xie et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Despite the significant progress that has been made in the diagnosis and treatment of digestive system diseases, these conditions continue to pose a serious public health concern worldwide. There is an ongoing need for strategies that are precise, efficient and minimally invasive. Nanozymes, engineered nanomaterials that exhibit catalytic functions, have attracted considerable interest in this context. However, clinical application of nanozymes remains limited primarily due to their diversity, targetability, biocompatibility and early‑stage clinical translation. The present review presented a comprehensive analysis of nanozymes in digestive system diseases. The main enzyme‑like activities of nanozymes are summarized to guide further material selection and characteristic exploration. Preclinical applications are highlighted with mechanisms and theranostic effects discussed alongside their potential limitations. Emerging combination therapies, including photodynamic therapy, sonodynamic therapy and biotherapy, are reviewed. Finally, the current challenges of nanozymes and possible future developments are discussed.

Introduction

Digestive system diseases are a substantial burden on contemporary society, contributing to over one-third of all reported cases (1). These conditions arise from complex mechanisms, including mucosal damage, genetic susceptibility, inflammatory response, immune dysregulation, gut microbiota dysbiosis and motility or secretory dysfunction in organs (Fig. 1) (2-5). Notable examples include gastric ulcers, liver damage, pancreatitis, inflammatory bowel disease (IBD) and tumors (1).

Diagnosing and treating digestive system diseases remain challenging. Some diagnostic methods lack the sensitivity needed for early detection (6). Although invasive procedures such as endoscopy and biopsy are valuable, they can be distressing for patients (7,8). Current treatments may suffer from poor efficacy and notable side effects (9-11). There is a pressing demand for more precise, efficient and less invasive diagnostic techniques, as well as novel therapeutic strategies tailored to the complexity of digestive system diseases.

In recent years, the biomedical community has increasingly turned its attention to nanomaterials, owing to their unique properties. Nanomaterial-based imaging and biosensing have enabled precise diagnosis (12-16). Furthermore, nanodrug delivery systems allow targeted delivery to specific tissues, effectively overcoming the limitations of conventional therapies (17,18). The concept of 'nanozyme' was originally introduced by Pasquato and Scrimin in 2004 (19). It now specifically refers to nanomaterials that emulate the catalytic behavior of natural enzymes (20). These synthetic enzyme mimics have been employed across a wide range of disciplines (21-23). Compared with natural enzymes, nanozymes have several advantages, including lower production costs, adjustable catalytic activity and greater structural stability (24-26). Although research on nanozymes for digestive system diseases is still at a relatively early stage, the growing number of studies highlights the emerging promise in this field.

Given the limited reviews on nanozymes in digestive system diseases, the aim of the present review was to provide a comprehensive analysis of their applications in diagnosis and treatment (Fig. 2). It also explores the unique advantages of nanozymes. Additionally, the current review explores the prospects and challenges of nanozymes in this field and offers insights into their potential value.

Enzyme-like activities of nanozymes in digestive system diseases

In 2014, Lin et al (27) proposed a structural-based classification system for nanozymes: Metal-based, metal oxide-based, carbon-based nanomaterials and other nanomaterials. As more nanozymes have emerged, researchers have shifted to classifying nanozymes by the types of enzymatic reactions they catalyze. The new classifications (24,26) are: i) Oxidoreductase-like family which includes oxidases, peroxidases (PODs), superoxide dismutases (SODs) and catalases (CATs) among others (Fig. 3); ii) hydrolase-like family which includes nucleases, proteases, phosphatases and esterases. This revised classification enhances our understanding of the potential applications of nanozymes in digestive system diseases. In the present review, an overview of the catalytic activities of nanozymes is provided.

Oxidase-like activity

In 2004, Comotti et al (28) reported that gold nanoparticles without a supporting matrix catalyze the oxidation of glucose to gluconate under mild conditions. However, their practical applicability was limited by their short lifespan. The catalytic mechanism involved the formation of electron-rich gold species through interactions between hydrated glucose anions and gold surface atoms, which subsequently activated molecular oxygen via nucleophilic attack. It has been proposed that intermediates such as Au+-O2 or Au2+-O22− function as electron transfer mediators (29). Clinically significant substrates include glucose, uric acid and ascorbic acid, which are used in the biosensing applications of nanozymes (30-32). A notable example involves the functionalization of MoO3 nanoparticles with dopamine as a surface ligand and triphenylphosphonium as a mitochondrial localization agent. This design enabled the selective catalytic oxidation of sulfite to sulfate within the mitochondrial environment. The positive cooperative behavior of modified MoO3 nanoparticles was confirmed by the Km and Hill coefficient, demonstrating activity comparable to goat and human sulfite oxidases (33). Some oxidase-like nanozymes generate H2O2 during reactions, which can enable H2O2-mediated cascade reactions (34-36). These nanozymes hold promise for diagnosing and treating digestive system diseases.

POD-like activity

Natural PODs use heme as a cofactor to catalyze substrate oxidation using H2O2 as an electron acceptor. Gao et al (37) first reported that Fe3O4 nanoparticles exhibited POD-like activity, following a ping-pong mechanism. Since then, a wide range of nanomaterials including metals, metal oxides, metal-organic frameworks and carbon-based systems have been investigated as POD mimetics (38-41). Most POD-like nanozymes decompose H2O2 into hydroxyl radicals (•OH), which subsequently oxidize substrates such as 3,3′,5,5′-tetramethylbenzidine, terephthalic acid and Trinder's reagents (42-44). Certain nanozymes exploit elevated H2O2 concentrations in tumor microenvironments to generate •OH species, initiating oxidative damage selectively in malignant tissues (45,46). V2O5 nanowires, studied by Vernekar et al (47), have been shown to exhibit GPx-like behavior, effectively neutralizing reactive oxygen species (ROS) while maintaining cellular redox homeostasis. Understanding these catalytic mechanisms will help identify suitable applications for POD-like nanozymes.

SOD-like activity

Superoxide can cause harmful oxidative stress in excess (48,49). SOD plays a key physiological role by converting superoxide radicals into oxygen and hydrogen H2O2, a process further detoxified by CAT (50,51). Multivalent metal ions such as Fe2+/Fe3+, Mn2+/Mn3+ and Ce3+/Ce4+ are essential for this reaction (52-55). Nanoscale CeO2, with increased surface area and oxygen vacancies, exhibits SOD-like activity. These structural features, along with lattice defects, enable CeO2 nanoparticles to regenerate, reducing the need for repeated administration compared to antioxidants such as vitamin C (56). The equations below describe this free radical scavenging mechanism, while a more comprehensive one was proposed by Celardo et al (57). Notably, the surface Ce3+/Ce4+ ratio is a determinant of catalytic performance, with higher Ce3+ content being associated with enhanced SOD-like function (58).

Fullerenes are recognized for their antioxidant and neuroprotective capabilities, but their water insolubility poses a challenge (59). Ali et al (60) addressed this with a tris-malonic acid derivative of fullerene C60 (C3), which scavenges superoxide radicals at a rate of 2×106 mol−1 s−1. Semiempirical quantum-mechanical calculations indicated that the region around malonic acid groups on C3 had the least electron distribution, attracting superoxide anions. Two O2 radicals were successively combined and dismutated in the presence of protons. The SOD-like activity of C3 is associated with the number and spatial arrangement of surface-exposed carboxyl groups on their surface (61). In addition to fullerenes, metals and their compounds such as Pt, Mn and Cu have also been found to exhibit SOD-like activity (62-64).

CAT-like activity

CAT, a natural enzyme localized primarily within cellular peroxisomes, facilitates the rapid breakdown of H2O2 into water and molecular oxygen. Kajita et al (65) synthesized gold and platinum bimetallic nanoparticles as potential CAT mimics. In recent years, numerous nanozymes that emulate CAT activity have been developed and reported (66-68). These nanozymes can mitigate oxidative stress-induced inflammation by decomposing H2O2 and, at the same time, help alleviate hypoxia within tumor tissues, thereby supporting cancer therapy (69).

Despite extensive investigation, the mechanistic underpinnings of CAT-like nanozyme catalysis remain incompletely understood. The decomposition of H2O2 by these nanozymes can occur via homolytic or heterolytic catalysis. In heterolytic catalysis, the nanozymes preferentially cleave the H-O bond in H2O2, while in homolytic catalysis, the O-O bond is more readily broken (70).

Other enzyme-like activities

Redox reactions are only part of the enzymatic spectrum in biology. Exploring enzyme-like activities beyond redox enzymes expands the range of reactions nanozymes can regulate. Nanoparticles with hydrolytic, esterase and photolyase activities have been reported, suggesting the potential for nanozymes to catalyze a broader range of reactions in the future (71-73).

Application of nanozymes in digestive system diseases

The digestive system facilitates ingestion, breakdown, absorption, transportation and production of digestive enzymes and secretions, while interacting with intestinal microbiota (74-76). Its unique pH variations support efficient catalytic activity of nanozymes. The significant surface area and microstructures of the stomach and intestines enhance contact with nanozymes (77). Due to prolonged exposure, the digestive system is biocompatible, reducing immune reactions to nanozymes, making it ideal for their application (78).

Esophageal cancer

The prognosis for esophageal cancer is generally unfavorable. In cases without metastasis, the 5-year survival rate range is 20-35% (79). Radiation therapy, which damages DNA in tumor cells to control tumor growth and metastasis, faces challenges such as inadequate X-ray energy deposition, tumor microenvironment hypoxia, upregulated antioxidant systems and DNA repair proteins in tumor cells (80-83). Developing radiosensitizers and optimizing treatment strategies are essential to improve radiation therapy efficacy. Combinations of therapies are currently used for resistant tumors but often fall short at a fundamental level (84).

Zhou et al (85) developed a nanoparticle of multifunctional covalent organic framework named triiodide-modified covalent organic framework with ferrocene (Fc), which contains iodine atoms and Fc functional groups. Iodine facilitated localized radiation dose deposition and promoted the generation of ROS, while Fc exhibited enzyme-like activities, leading to lipid peroxidation and iron deposition, reversing radiation resistance and disrupting redox homeostasis. Fc was identified as a potential radiosensitizer for resistant tumors (Fig. 4). To tackle radiation resistance stemming from the enhanced expression of antioxidant systems and DNA repair proteins within cancer cells, Zhou et al (86) designed a radio-sensitization strategy using high-Z elements, incorporating a GDY-CeO2 nanoparticle enzyme and microRNA-181a. This combination exhibited CAT-mimicking capabilities. It relieved tumor hypoxia, augmented radiation-induced DNA damage and functioned as a radiosensitizer. This was achieved through targeting RAD17 and modulating the Chk2 pathway. Additionally, there has been significant interest in intelligent phototherapy nanotherapeutic systems. Liu et al (87) introduced a BSA-MnO2/IR820@carboxylated carbon nanocages nanoparticle loaded with efficient photosensitizers, demonstrating CAT-like activity and enabling near-infrared (NIR) imaging for photothermal therapy (PTT) and photodynamic therapy (PDT) implementation.

In recent years, nanozymes have shown great promise in the diagnosis of esophageal cancer. For instance, POD-mimicking nanozymes have been successfully employed in immunohistochemical staining to enhance the visualization of esophageal tumors, offering faster and more robust color development than natural enzymes (88). Moreover, the integration of targeting ligands has allowed for enhanced recognition of early esophageal lesions (89). In addition, a carbon nanocage-based nanozyme system activated by endogenous H2O2 has been developed to generate oxygen within the tumor microenvironment, enabling magnetic resonance imaging (MRI) and fluorescence bimodal imaging (87).

However, there are potential drawbacks in treating esophageal cancer with nanozymes. Current development of nanozymes involves complex synthesis and functionalization, incorporating multiple substances, which increases costs and limits scalability. The distinct mucosal structure and function in esophageal tissue are able to influence the adsorption, dispersion and bioactivity of nanozymes. Additionally, the rapid transport of food through esophageal peristalsis reduces the time that orally administered drugs remain in the esophagus, influencing drug release and absorption (90). Further investigation is required to ascertain the effectiveness of nanozymes in specifically targeting the deeply located lesions within esophageal cancer. It is also necessary to define their role in preventing both local recurrence and distant metastasis.

Gastric diseases

Gastric diseases, including gastritis, ulcers and cancer, have a high global incidence (91). A major pathological factor is Helicobacter pylori, a microaerophilic, spiral-shaped bacterium (92,93). Standard quadruple therapy faces limitations due to the acidic environment of the stomach and effect on symbiotic bacteria (94). Nanozyme offers a novel approach to address these challenges.

Zhang et al (95) synthesized a pH-responsive graphitic oxidase-like nanozyme (PtCo@G@CPB), which is made to selectively target and combat H. pylori without disrupting intestinal microbiota. The oxidase-like activity of PtCo@G was stably activated under gastric acid conditions (pH 0.9-1.5), producing superoxide radicals from H2O2. These radicals attacked the bacterial membrane, causing cell death. The enzyme activity was inhibited under the neutral conditions of the intestine, minimizing toxicity to normal tissues and commensal bacteria. Although in vivo short-term toxicity studies (lasting 6 days) indicated low toxicity levels, there is a dearth of comprehensive investigations regarding the long-term retention toxicity associated with heavy metals (96). Yan et al (97) developed a stable, pH-sensitive, persistent luminescence (PL) nanozyme (MSPLNP-Au-CB). It allows for autofluorescence-free long-term and real-time imaging in vivo and is aimed at targeted eradication of H. pylori. The PL nanozyme platform facilitated bioimaging as well as the treatment of bacterial infections under challenging circumstances.

Nanozymes also have extensive applications in the diagnosis and treatment for gastric cancer, achieving highly efficient tumor treatment by integrating PTT, PDT and nanozyme oxidation (98). Zhang et al (99) introduced a nanocomposite material, gold-doped mesoporous carbon spheres (OMCAPs@rBSA-FA@IR780), which exhibited POD-like activity from internally-doped gold nanoparticles that generated ROS, leading to oxidative damage and apoptosis in gastric tumor cells. Additionally, internal incorporation of NIR dye IR780 enabled PTT under 808 nm laser irradiation. A novel DNA-templated Ag@Pd alloy nanocluster (DNA-Ag@Pd NC) was developed with excellent stability in vitro and in vivo. It exhibited an impressively photothermal conversion efficiency reaching 59.32% and enhanced POD-like activity, enabling highly efficient photothermal-enhanced nanocatalytic therapy for gastric cancer, guided by high-contrast NIR-II photoacoustic (PA) imaging (100).

Recent advances have demonstrated the potential of nanozyme-based platforms in the early diagnosis of gastric diseases. Various strategies have been employed to enhance sensitivity and specificity, including MoS2-based nanozymes combined with hybridization chain reactions for microRNA detection and chiral POD-like nanozymes for enantiomer identification in gastric cancer (101,102). Additionally, dual-functional systems integrating nanozymes with catalytic hairpin assembly or plasmonic surface-enhanced Raman scattering have been applied for the ultrasensitive detection of gastric precancerous lesions and D-amino acid biomarkers in saliva (103,104). These innovations highlight the versatility of nanozymes in molecular sensing and their emerging role in non-invasive and precise gastric cancer diagnostics.

As previously mentioned, the highly acidic environment and unique microbial community in stomach present challenges for nanozyme treatments. Current research focuses on short-term toxicity, lacking comprehensive long-term assessments. It is unclear whether exposure to normal tissues outside the affected area causes irritation, disrupts digestive function, or affects nutrient absorption. Some nanozymes are activated under acidic conditions, but prolonged exposure to low pH levels may cause structural denaturation or deactivation, affecting treatment efficacy. While certain nanozymes selectively eradicate H. pylori, more in-depth research is required to comprehend their long-term impact on the microbial community.

Liver diseases

Acute liver injury (ALI) represents a serious condition marked by necrosis of liver cells and impairment of liver function, caused by drug exposure, viral infection and alcohol abuse (105-107). N-acetylcysteine is a common therapeutic drug but has a narrow therapeutic window (108). Nanozymes show potential in ALI diagnosis and treatment. Zhou et al (109) designed the ultrasmall gold nanoparticles-tannic acid hybrid nanozyme, SAuPTB, that activated the Nrf2 pathway, offering anti-inflammatory and antioxidant effects. Similarly, Prussian blue nanozymes have shown protective effects on anthracene-induced ALI (110). Xia et al (111) demonstrated that reducing ruthenium nanoparticles to 2.0 nm markedly enhanced antioxidant activity and upregulated regulatory T cells. PA imaging is coming as a potent means for imaging intracellular ROS. A ROS-sensitive nanozyme-enhanced PA nanoprobe catalyzes oxygen microbubble production in the presence of ROS, amplifying the PA signal and reversing the hypoxic microenvironment for simultaneous treatment (112). In alcoholic liver injury, current approaches focus on supportive care rather than directly metabolizing excess alcohol (113). Geng et al (114) proposed an alcohol detoxification strategy using an alcohol dehydrogenase/CAT/aldehyde dehydrogenase-cascaded nanoreactor (AA@mMOF) for targeted detoxification and ALI management.

Liver fibrosis, which results from chronic liver injury, has the potential to progress to cirrhosis and hepatocellular carcinoma. This process entails the generation of ROS, the activation of hepatic stellate cells (HSCs) and the overproduction of extracellular matrix (115-118). Effective anti-fibrotic methods are limited, with transplantation as the last resort. Addressing excessive ROS generation, Zhang et al (119) showed that MoS2 nanozyme mimicked cellular enzymes and cleared ROS for liver fibrosis treatment. Lu et al (120) developed a sequential delivery strategy using a star-shaped nanozyme (Au NS@CAR-HA) loaded with carvedilol to clear ROS, inhibit autophagy and target HSCs for fibrosis treatment.

Nanozymes have garnered attention in liver cancer. While ferrotherapy shows promise, it faces limitations in catalytic efficiency (121,122). Jiang et al (123) reported the first hybrid semiconducting polymer nanozyme (HSN). It has augmented catalytic activity when under light irradiation, which combined NIR-II PA imaging with photothermal ferrotherapy to enhance the Fenton reaction, promoting cell apoptosis and ferroptosis. Anticancer mechanism of nanozymes lies mainly in the ROS generation triggered by changes in the metal element oxidation state. Meanwhile, some studies have found that metals with unchanged oxidation states can be explored and developed for catalytic activity. A CaF2 nanozyme was designed with ultrasound enhanced POD-mimicking activity, inducing mitochondrial dysfunction through calcium-pumping channel regulation, achieving effective antitumor effects in an H22 liver cancer model (124). Metals such as Os, commonly used in battery technologies, have shown potential in cancer treatment. Os-Te nanorods (OsTeNRs) exhibited excellent photothermal conversion and photocatalytic activities (125).

Apart from its therapeutic applications, nanozymes can also be used for the diagnosis of liver diseases. Various nanozyme platforms have been developed to detect key pathological features associated with hepatic disorders, such as elevated ROS, abnormal enzyme activity and specific biomarkers (112,126). For example, POD- and oxidase-mimicking nanozymes have been used for colorimetric and fluorescent detection of alanine aminotransferase and aspartate aminotransferase, which are essential indicators of liver injury (127). Furthermore, nanozymes integrated with imaging modalities such as MRI, PA imaging and chemiluminescence imaging have enabled non-invasive and real-time monitoring of liver fibrosis, hepatitis and hepatocellular carcinoma (128,129). These advances highlight the potential of nanozyme-based diagnostic platforms to improve the accuracy, efficiency and accessibility of liver disease diagnostics.

The liver, essential for metabolism, detoxification and immunity, lacks comprehensive research on nanozyme degradation, clearance and potential functional effects. The immunogenicity of nanozymes and their compatibility with the immune system in liver are also unclear (130). The complex microenvironment in liver, with its pH variations, redox potential, nutrient concentrations and metabolic byproducts, poses challenges for nanozymes due to inadequate organ-specific targeting, leading to uneven distribution and potential toxicity. Liver diseases involve diverse processes like inflammation and fibrosis. Current research often focuses on specific treatment aspects, highlighting the need for multifunctional nanozymes capable of addressing multiple pathological pathways simultaneously.

Pancreatic diseases

The pancreas is essential for digestion and hormone regulation. Acute pancreatitis (AP) causes severe abdominal pain due to the activation of pancreatic enzymes, leading to tissue damage and systemic inflammation (131-136). Oxidative stress plays a crucial role in AP progression (137,138). Nanozymes, which mimic antioxidant enzymes, present a promising approach for AP treatment (26).

In AP, the triggering of the NF-κB signaling pathway results in the upregulation of inflammatory cytokines such as IL-1, IL-6 and TNF-α (133,139,140). Xie et al (141) reported that Prussian blue nanoclearance agents (PBzymes) effectively neutralize ROS, converting them into harmless H2O and O2 (Fig. 5). PBzymes demonstrate significant removal efficiency of •OH and •OOH (59.1 and 41.9% at 30 μg/ml, respectively), exhibiting intrinsic antioxidant and anti-inflammatory properties through the inhibition of the TLR/NF-κB signaling pathway. Severe AP (SAP) affects 20-30% of patients, leading to multi-organ damage and high mortality (131). Ferroptosis and oxidative stress are key factors in SAP pathogenesis (142-144). Ca/Fe-based nanozymes have been shown to reduce SAP-related biomarkers and inflammation, increasing GPx4 and FTH1, key factors in ferroptosis and intracellular iron regulation (145). To address potential biocompatibility concerns, Xie et al (146) introduced MoSe2-PVP nanoparticles with high biocompatibility, ROS/RNS clearance, biodegradability and pancreatic accumulation properties.

Pancreatic cancer (PC) poses a severe threat, with a low 5-year survival rate of 8% (147). The low-oxygen tumor microenvironment promotes treatment resistance and malignant progression (148). A wide array of therapeutic strategies, like PTT, PDT, chemo-dynamic therapy and sonodynamic therapy (SDT), have been explored to combat PC. NIR lasers in phototherapy boost the catalytic activity of nanozymes and augment therapeutic effectiveness (113,149). Li et al (150) developed a dual-enzyme-mimicking nanozyme (PtFe@Fe3O4) with NIR enhancement, showing electron transfer between PtFe nanorods and Fe3O4 nanoparticles to boost catalytic activity. NIR laser-triggered POD and CAT-like activity achieved notable 99.8% inhibition of deeply located PC. Kang et al (148) presented a ruthenium-tellurium hollow nanorod for combined PDT and PTT in PC treatment, demonstrating excellent performance under UV-vis-NIR laser irradiation. Chen et al (151) developed a synergistic treatment platform utilizing ZIF-90 nanoparticles nucleating on platinum nanoparticles. This platform enhanced chemotherapy and SDT by relieving hypoxia, increasing chemotherapy sensitivity boosting ROS and singlet oxygen production under US irradiation, achieving a synergistic strategy between chemotherapy and SDT.

At present, the application of nanozymes in the diagnosis of pancreatic diseases is still limited. Zhang et al (152) developed gemcitabine-loaded carbonaceous nanozymes with POD-like and glutathione POD-like activities, thereby enabling MRI-guided imaging and enhancing diagnostic precision by responding to the tumor microenvironment.

The pancreas, essential for secreting digestive enzymes and hormones like insulin, presents unique challenges for nanozyme therapies. Current treatments may interfere with normal digestive and hormonal functions. PC often involves a hypoxic tumor microenvironment, where traditional nanozymes may be less effective, highlighting the need for design improvements to function in low oxygen conditions.

IBD

IBD, characterized by inflammation, oxidative stress, immune dysregulation and intestinal microbial imbalance, poses complex therapeutic challenges due to the intricate interplay of these factors (153,154). Excessive ROS accumulation in IBD lesions exacerbates the condition by directly damaging intestinal epithelial cells and altering intestinal microbiota composition (155,156). Nanozyme-based strategies targeting ROS have garnered attention as potential treatments for IBD.

Monitoring IBD activity is crucial for treatment evaluation and surgical timing (157). Cerium oxide (CeO2) nanoparticles have emerged as promising CT contrast agents for gastrointestinal tract imaging. Cao et al (158) developed dextran-coated CeO2 nanoparticles (D-CeO2) for IBD surveillance. D-CeO2 exhibited slightly higher CT values compared with iohexol at equivalent concentrations in vitro. In murine colitis models, D-CeO2 enabled extended imaging windows and preferentially accumulation at inflamed sites, outperforming iohexol in lesion targeting. Similarly, inulin-coated CeO2 nanoparticles (CeO2@IN nanoparticles) demonstrated excellent in vivo imaging performance, markedly extending the imaging duration by up to 12 h (159). Notably, due to their intrinsic SOD- and CAT-like activities, these nanozymes are capable of scavenging ROS, thereby also contributing to the mitigation of colitis-related tissue damage. In another approach, a BaSO4-based imaging platform, BaSO4@PDA@CeO2/DSP, was developed to enhance adhesion to inflamed regions. The inherent gastrointestinal stability of BaSO4, combined with its functional modifications, offers a promising strategy for IBD diagnosis (160). Deng et al (161) introduced a multimodal imaging probe, metal-polyphenol network (MPN@CeOx), in which the exposed MPN serves as an MRI contrast agent, enabling MRI-guided diagnosis and potentially facilitating image-guided therapy for colitis. Another innovative approach involves a ROS-targeting nano-sensor, PPNCP, designed to release platinum nanoclusters (PtNCs) at inflammation sites. After penetrating the intestinal barrier, PtNCs could be traced in urine through their enzyme-mimicking activity, facilitating non-invasive monitoring (Fig. 6) (162). However, concerns remain regarding nephrotoxicity of platinum derivatives, especially in geriatric populations, highlighting the need for patient-specific approaches despite the non-invasive advantage (163). Additionally, carbon dots (C-dots), owing to their favorable fluorescence properties and stability under inflammation-mimicking conditions have shown promise as imaging agents (164). However, limited differences in biodistribution between healthy and inflamed mice in major organs and the colon suggest that further optimization is required to harness their potential for inflammation-specific imaging.

Nanozymes have been strategically designed for direct therapy in IBD, functioning to neutralize ROS and mitigate inflammation. Zhao et al (165) introduced an innovative approach with CeO2@montmorillonite (MMT), which combines ROS-scavenging CeO2 nanoparticles with MMT. The system not only maintains catalytic efficiency, but also minimizes potential intestinal toxicity, exhibiting superior therapeutic outcomes in a mouse model by alleviating symptoms and suppressing inflammatory markers. To tackle the issue of optimizing multiple enzymatic functions within a nanozyme, researchers employed a valence-engineering strategy, using spinel oxide ZnMn2O4 as a model. This led to the construction of LiMn2O4, which excelled in various activity assays, including SOD, CAT and GPx further validating its performance in preclinical settings (166). Other nanozyme formulations, such as Pt@PCN222-Mn, Ni3S4 and MeNPs, have also been successfully tested in preclinical models for IBD treatment (167-169).

Nanozymes demonstrate synergistic effects in combination therapies, emphasizing the need for well-designed drug delivery systems. One approach involves decorating the anti-inflammatory agent curcumin (CCM) with a Prussian blue analog (CoFe PBA), resulting in a core-shell structure (CCM-CoFe PBA). Under acidic conditions, the interaction between CCM and CoFe PBA weakens due to the disruption of the coordination bond between Fe3+ in CoFe PBA and the keto-type structure. This feature facilitates targeted drug delivery in mildly acidic inflammatory environments, although it may reduce efficacy in the acidic gastric environment. In a DSS-induced colitis mouse model, CCM-CoFe PBA treatment alleviated symptoms, polarized macrophages toward the M2 phenotype and improved the histological structure of the colonic mucosa (170). Cao et al (171) developed a single-atom nanozyme modified with Bifidobacterium longum (BL) to create a therapeutic system (BL@B-SA50). This hybrid system exhibits both SOD- and CAT-like activities, effectively neutralizing ROS and shielding anaerobic probiotic bacteria from oxidative stress in inflamed intestinal environments. In murine colitis models, treatment with BL@B-SA50 resulted in decreased levels of ROS and pro-inflammatory mediators (IL-6, IFN-γ, MPO), along with a notable elevation of the anti-inflammatory cytokine IL-10. Oral administration of this formulation in a canine model of ulcerative colitis facilitated mucosal repair in the colon, suggesting its translational therapeutic promise. Furthermore, C-dots synthesized from glutathione and biotin through a solvothermal reaction have demonstrated dual functionalities, mitigating colonic inflammation and enabling non-invasive imaging within the gastrointestinal tract (164).

Beyond ROS, RNS also play a role in mediating biomolecular damage in inflamed tissues (172). PEG-coated rhodium nanodots (Rh-PEG NDs) have shown effective scavenging capabilities against both ROS and RNS. In a murine colitis model, Rh-PEG NDs reduced tissue damage, inflammatory cytokine levels and CD45+ cell counts and demonstrated excellent blood compatibility with a hemolysis rate <0.5% (173). Ultrathin trimetallic nanosheets (TMNSs; Ru38Pd34Ni28) feature Ru-O and Ni-O bonds that efficiently clear ROS and RNS, with enhanced activity due to atomic vacancies on the surface. In a DSS-induced colitis model, mice treated with TMNSs showed weight recovery, improved colon length and reduced ROS and inflammatory factors (174).

Nanozymes have shown extensive application in preclinical models for IBD treatment. Enhancing nanozyme efficacy within the intestinal microenvironment remains crucial, necessitating the development of nanozyme therapeutics tailored to different IBD biological subtypes (175,176). Evidence suggests that Fe may worsen ulcerative colitis, while Se and Zn might help prevent its development and the roles of Cu and Mn in ulcerative colitis remain unclear (177,178). Additionally, microplastics and endocrine disruptors from nanozyme synthesis could negatively impact IBD patients (178). A metal-free melanin nanozyme has been proposed to avoid the risks associated with heavy metals (169). Improving synthesis processes and reducing impurities through separation steps are needed to address reagent residue issues.

The high manufacturing and storage costs of precious metal-containing nanozymes limit their clinical use (179,180). Probiotics and algae have ideal living conditions (181,182). While they can treat IBD in vivo, preserving probiotic- and algae-based nanozyme systems for large-scale production remains challenging. Gene editing technology might enhance their survival under adverse conditions.

The NF-κB and JAK-STAT pathways are key targets in IBD treatment with nanozymes (183,184). Novel mechanisms, such as inhibiting T-cell differentiation and disrupting neutrophil extracellular traps, have been proposed to alleviate IBD symptoms (185,186). Enhancing nanozyme selectivity for inflammatory loci is crucial. Current strategies, such as absorption by charge or pH in the lesion sites, do not fully align with precision medicine. Ma et al (187) developed a macrophage membrane-coated nanozyme system with CRISPR/Cas9 to reduce CD98 expression. Future research should focus on precisely targeting disease sites with nanozyme systems.

Colon cancer

Colon cancer, a prevalent malignancy in the digestive system, involves a multistep and multigenic disorder (188). There is a strong link between IBD and the onset of colon cancer (189). Nanozymes target colon cancer by mitigating the hypoxic microenvironment, inducing ROS overload and initiating immunotherapy.

PTT employs NIR light in junction with photothermal agents to induce localized hyperthermia, effectively eliminating tumor cells (190). Zhu et al (191) introduced a multifunctional system [Ru@CeO2-ruthenium complex (RBT)/resveratrol (Res)-DPEG) that integrates PTT with chemotherapy. The yolk-shell structured nanozyme demonstrated excellent catalytic efficiency and photothermal conversion capability. Additionally, it was co-loaded with RBT and Res. The dual-layer PEG-coating extended circulation time by reducing plasma protein adsorption. In both subcutaneous and orthotopic colon cancer models, Ru@CeO2-RBT/Res-DPEG + NIR exhibited strong antitumor effects and reduced HIF-1α levels in tumor tissues. Another tumor microenvironment-responsive platform, the POD nanocrystals (CatCry-AgNP-DOX), which does not fall within the category of nanozymes, may present a novel approach for future drug delivery (192).

PDT selectively targets tumors with minimal invasiveness (193). Hao et al (194) developed a ROS-sensitive nanoplatform [camptothecin (CPT)-TK-HPPH/Pt nanoparticle], which integrates CPT with the photosensitizer HPPH. Pt nanoparticle-generated oxygen-facilitated HPPH consumption <660 nm laser irradiation, cleaving the thioketal bond to release CPT simultaneously (Fig. 7). In vivo experiments showed a marked tumor volume reduction and pronounced tumor necrosis in the CPT-TK-HPPH/Pt nanoparticle + laser group. However, the effectiveness of PDT is limited by the penetration depth of laser light. Duo et al (195) addressed this by inserting an optical fiber into the peritoneal cavity of mice for PDT treatment with orthotopic tumors, showing promising results. SDT offers high tissue penetration and non-invasiveness (196). A cascading nano-reactor [CCP@ hollow polydopamine (HP)@M] was designed, incorporating the sonosensitizer Ce6 and autophagy inhibitor chloroquine. The HP with metal-organic framework core demonstrated drug-loading capability and SOD-like activity. Additionally, platinum nanoparticles were doped onto the carrier HP, endowing the system with CAT- and POD-like activities to alleviate tumor hypoxia while amplifying ROS generation. In HCT-116 xenograft models, the combined approach of ROS elevation and autophagy inhibition, together with SDT, resulted in suppression of tumor. RNA-sequencing analysis showed downregulation of cancer, autophagy, apoptosis, ferroptosis pathways and HIF-1α, aligning with the expected nano-reactor mechanism (197).

In recent years, growing evidence has highlighted the role of tumor-resident microbiota in tumor development and therapeutic resistance, opening new avenues for microbial-targeted strategies (198). Researchers developed a protein-supported copper single-atom nanozyme (BSA-Cu SAN) facilitating the conversion of H2O2 into highly reactive hydroxyl radicals (•OH). This oxidative stress was directed at Fusobacterium nucleatum within tumors, impairing its viability. In HCT-116 xenograft mouse models, BSA-Cu SAN administered intravenously or intratumorally effectively inhibited tumor growth. Of note, 48 h after intravenous injection, no notable drug accumulation was observed, reducing tissue damage from prolonged heavy metal retention (199). Another noteworthy therapeutic approach involves utilizing pH-dependent POD-like nanozymes that, upon sensing the tumor microenvironment, generate a small amount of ROS around the engineered probiotic BL999. This prompted the probiotic to produce acidic metabolites, feedback enhancing the POD-like nanozyme activity, ultimately generating a substantial amount of ROS to counteract tumor tissues (200).

Nanozyme-based diagnostic platforms facilitate the precision of cancer diagnosis. One feasible approach involves the use of multifunctional CeO2 nanoparticles for the detection of T-cell immunoglobulin and mucin domain 1, a membrane glycoprotein associated with tumor angiogenesis. Compared with conventional diagnostic platforms, this method provides a moderate enhancement in sensitivity, a significant expansion of the linear detection range and a reduction in background current, thereby contributing to more accurate diagnosis and prognostic stratification in patients (201). Another diagnostic platform enables direct detection of circulating cancer stem cells from blood and fecal samples (202). Additionally, nanozymes have been employed in the detection of global DNA methylation in colorectal cancer cells using mesoporous iron oxide. Compared with traditional procedures, this method offers lower cost and higher detection sensitivity, making it a promising platform for rapid clinical screening (203).

Nanozymes, while promising, have shown relatively low efficiency compared with surgical treatments in eliminating tumors. As aforementioned, current strategies combine nanozyme with traditional treatments to enhance efficacy and minimize damage. Developing combinatory protocols that exploit complementary mechanisms is crucial for advancing cancer treatment.

The biological behavior of colon cancer varies, necessitating more specific approaches (204,205). Nanozyme systems need to target specific biomarkers and pathways unique to colon cancer, informed by molecular and genetic profiles, to improve effectiveness and reduce off-target effects.

Current research focuses on alleviating tumor hypoxia by enhancing oxygen supply (206-208). Exploring unconventional mechanisms, such as ferroptosis and amino acid depletion, has opened up promising strategies, particularly by disrupting metabolic processes and survival signaling in tumors (209,210).

Other digestive system diseases

Nanozymes have been used in the functionalization of stem cells for treating digestive diseases (211,212). Additionally, they have been applied in RNA interference technology for antiviral therapy (213). However, additional research is essential to determine the full extent of their applicability across various digestive system diseases.

Conclusions, challenges and perspectives

The present review highlighted the latest advances of nanozymes in preclinical models of major digestive system diseases, encompassing diagnostics, monitoring and, most importantly, therapeutic applications. A detailed overview of nanozyme applications in diseases affecting the esophagus, stomach, liver, pancreas and colon was provided. Primary enzyme-like activities were also discussed. Despite significant progress, the application in digestive system diseases is still in its early stages. In the current review, the challenges and potential prospects were outlined.

At present, nanozymes continue to encounter several critical shortcomings and unresolved challenges in both research and clinical applications. One of the primary issues is the lack of sufficient targeting specificity. A number of nanozymes exhibit poor tissue or cellular selectivity and are readily sequestered by non-target organs, such as the liver and spleen. In the gastrointestinal tract, their catalytic activity can be compromised by physiological barriers including mucus layers, gastric acid and digestive enzymes. Furthermore, concerns regarding the biocompatibility and potential toxicity of nanozymes, particularly those based on metal oxides, persist. Long-term accumulation or high-dose administration of such materials may induce cytotoxic effects or elicit inflammatory responses. Compared with natural enzymes, nanozymes often display suboptimal catalytic efficiency under physiologically relevant conditions, especially in environments with fluctuating pH or redox states. In addition, the in vivo biodistribution, metabolism and excretion of nanozymes are difficult to monitor, complicating dose optimization and therapeutic outcome evaluation. Immunological responses and clearance mechanisms triggered by nanozymes have not yet been fully elucidated, raising concerns about the potential for immune tolerance or immune-related toxicity with prolonged administration.

To address these limitations, future research should focus on several key directions. First, surface modification strategies such as conjugation with antibodies, peptides, or oligonucleotides can enhance the targeting specificity of nanozymes. The development of stimuli-responsive nanozymes that are activated by pathological microenvironments such as acidic pH and high ROS levels may further improve site-specific efficacy. Second, the use of biocompatible and biodegradable materials to encapsulate or functionalize nanozymes can reduce exposure of the metal core and mitigate toxicity. Third, optimization of nanozyme morphology, particle size and surface catalytic sites, or the construction of synergistic systems with natural enzymes, may help improve catalytic performance. Last, designing nanozymes with strong mucosal adhesion and protective, pH-responsive coatings or integrating them with probiotics and microbiota-targeted therapies may enhance their stability and therapeutic effect within the gastrointestinal tract.

In conclusion, nanozymes hold tremendous potential as a next-generation tool for the diagnosis and treatment of digestive system diseases. However, their successful clinical application will rely on interdisciplinary efforts to overcome current limitations in biology, chemistry and related fields. Continued innovation in material design, targeting strategies and in vivo evaluation methodologies will be key to unlocking the full potential of nanozymes. In the future, nanozymes are expected to transition from promising preclinical agents to clinically viable therapeutic modalities.

Availability of data and materials

Not applicable.

Authors' contributions

Conceptualization was by YH and CS. Investigation was by DX, LX and CF. DX and LX were responsible for writing and original draft preparation. DX, LX, CF, ZD, ZC and YH were responsible for writing, reviewing and editing. Visualization was by DX and LX. Supervision was by YH and CS. Project administration was by YH and CS. CS made a significant contribution to the manuscript revision. Data authentication is not applicable. All authors 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.

Abbreviations:

CAT

catalase

GPx

glutathione peroxidase

NIR

near-infrared

ROS

reactive oxygen species

SOD

superoxide dismutase

PDT

photodynamic therapy

POD

peroxidase

PTT

photothermal therapy

RNS

reactive nitrogen species

SDT

sonodynamic therapy

Acknowledgements

Not applicable.

Funding

The present study was funded by National Key R&D Program of China (grant nos. 2023YFC2508601, 2023Y FC2508604 and 2023YFC2508605), Tongji University Medicine-X Interdisciplinary Research Initiative (grant no. 2025-0554-ZD-08), Shanghai Hospital Development Center Foundation (grant no. SHDC22025208), Shanghai Hospital Development Center Foundation (grant no. SHDC12024125), Clinical Research Foundation of Shanghai Pulmonary Hospital (grant no. LYRC202401), Innovation Team Project of the Faculty of Chinese Medicine Science, Guangxi University of Chinese Medicine (grant nos. 2023CX001 and 2024ZZA004).

References

1 

Wang Y, Huang Y, Chase RC, Li T, Ramai D, Li S, Huang X, Antwi SO, Keaveny AP and Pang M: Global burden of digestive diseases: A systematic analysis of the global burden of diseases study, 1990 to 2019. Gastroenterology. 165:773–783.e15. 2023. View Article : Google Scholar : PubMed/NCBI

2 

Lee M and Chang EB: Inflammatory bowel diseases (IBD) and the microbiome-searching the crime scene for clues. Gastroenterology. 160:524–537. 2021. View Article : Google Scholar

3 

Mayerle J, Sendler M, Hegyi E, Beyer G, Lerch MM and Sahin-Tóth M: Genetics, cell biology, and pathophysiology of pancreatitis. Gastroenterology. 156:1951–1968.e1. 2019. View Article : Google Scholar : PubMed/NCBI

4 

Zhou WC, Zhang QB and Qiao L: Pathogenesis of liver cirrhosis. World J Gastroenterol. 20:7312–7324. 2014. View Article : Google Scholar : PubMed/NCBI

5 

Gordon-Weeks AN, Lim SY, Yuzhalin AE, Jones K and Muschel R: Macrophage migration inhibitory factor: A key cytokine and therapeutic target in colon cancer. Cytokine Growth Factor Rev. 26:451–461. 2015. View Article : Google Scholar : PubMed/NCBI

6 

Ku G, Tan IB, Yau T, Boku N, Laohavinij S, Cheng AL, Kang YK and de Lima Lopes G Jr: Management of colon cancer: Resource-stratified guidelines from the Asian Oncology Summit 2012. Lancet Oncol. 13:e470–e481. 2012. View Article : Google Scholar : PubMed/NCBI

7 

Lauriola M, Tomai M, Palma R, La Spina G, Foglia A, Panetta C, Raniolo M and Pontone S: Intolerance of uncertainty and anxiety-related dispositions predict pain during upper endoscopy. Front Psychol. 10:11122019. View Article : Google Scholar : PubMed/NCBI

8 

Eisenberg E, Konopniki M, Veitsman E, Kramskay R, Gaitini D and Baruch Y: Prevalence and characteristics of pain induced by percutaneous liver biopsy. Anesth Analg. 96:1392–1396. 2003. View Article : Google Scholar : PubMed/NCBI

9 

Zedan AH, Hansen TF, Fex Svenningsen A and Vilholm OJ: Oxaliplatin-induced neuropathy in colorectal cancer: Many questions with few answers. Clin Colorectal Cancer. 13:73–80. 2014.

10 

Click B and Regueiro M: A practical guide to the safety and monitoring of new IBD therapies. Inflamm Bowel Dis. 25:831–842. 2019. View Article : Google Scholar :

11 

Cosnes J, Bourrier A, Laharie D, Nahon S, Bouhnik Y, Carbonnel F, Allez M, Dupas JL, Reimund JM, Savoye G, et al: Early administration of azathioprine vs conventional management of Crohn's Disease: A randomized controlled trial. Gastroenterology. 145:758–765.e52; quiz e14-e55. 2013. View Article : Google Scholar : PubMed/NCBI

12 

Fan K, Cao C, Pan Y, Lu D, Yang D, Feng J, Song L, Liang M and Yan X: Magnetoferritin nanoparticles for targeting and visualizing tumour tissues. Nat Nanotechnol. 7:459–464. 2012. View Article : Google Scholar : PubMed/NCBI

13 

Gazouli M, Bouziotis P, Lyberopoulou A, Ikonomopoulos J, Papalois A, Anagnou NP and Efstathopoulos EP: Quantum dots-bevacizumab complexes for in vivo imaging of tumors. In Vivo. 28:1091–1095. 2014.PubMed/NCBI

14 

Zhong Y, Ma Z, Wang F, Wang X, Yang Y, Liu Y, Zhao X, Li J, Du H, Zhang M, et al: In vivo molecular imaging for immunotherapy using ultra-bright near-infrared-IIb rare-earth nanoparticles. Nat Biotechnol. 37:1322–1331. 2019. View Article : Google Scholar : PubMed/NCBI

15 

Idris AO, Mabuba N and Arotiba OA: An exfoliated graphite-based electrochemical immunosensor on a dendrimer/carbon nanodot platform for the detection of carcinoembryonic antigen cancer biomarker. Biosensors (Basel). 9:392019. View Article : Google Scholar : PubMed/NCBI

16 

Li Y, Huang Z, Li Z, Li C, Liu R and Lv Y: Mass spectrometric multiplex detection of MicroRNA and protein biomarkers for liver cancer. Anal Chem. 94:17248–17254. 2022. View Article : Google Scholar : PubMed/NCBI

17 

Guo J, Yu Z, Sun D, Zou Y, Liu Y and Huang L: Two nanoformulations induce reactive oxygen species and immunogenetic cell death for synergistic chemo-immunotherapy eradicating colorectal cancer and hepatocellular carcinoma. Mol Cancer. 20:102021. View Article : Google Scholar : PubMed/NCBI

18 

Song W, Tiruthani K, Wang Y, Shen L, Hu M, Dorosheva O, Qiu K, Kinghorn KA, Liu R and Huang L: Trapping of lipopolysaccharide to promote immunotherapy against colorectal cancer and attenuate liver metastasis. Adv Mater. 30:e18050072018. View Article : Google Scholar : PubMed/NCBI

19 

Manea F, Houillon FB, Pasquato L and Scrimin P: Nanozymes: Gold-nanoparticle-based transphosphorylation catalysts. Angew Chem Int Ed Engl. 43:6165–6169. 2004. View Article : Google Scholar : PubMed/NCBI

20 

Wei H and Wang E: Nanomaterials with enzyme-like characteristics (nanozymes): Next-generation artificial enzymes. Chem Soc Rev. 42:6060–6093. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Mahmudunnabi RG, Farhana FZ, Kashaninejad N, Firoz SH, Shim YB and Shiddiky MJA: Nanozyme-based electrochemical biosensors for disease biomarker detection. Analyst. 145:4398–4420. 2020. View Article : Google Scholar : PubMed/NCBI

22 

Luo Q, Shao N, Zhang AC, Chen CF, Wang D, Luo LP and Xiao ZY: Smart biomimetic nanozymes for precise molecular imaging: Application and challenges. Pharmaceuticals (Basel). 16:2492023. View Article : Google Scholar : PubMed/NCBI

23 

Alvarado-Ramírez L, Rostro-Alanis M, Rodríguez-Rodríguez J, Sosa-Hernández JE, Melchor-Martínez EM, Iqbal HMN and Parra-Saldívar R: Enzyme (Single and Multiple) and nanozyme biosensors: Recent developments and their novel applications in the water-food-health nexus. Biosensors (Basel). 11:4102021. View Article : Google Scholar : PubMed/NCBI

24 

Wu J, Wang X, Wang Q, Lou Z, Li S, Zhu Y, Qin L and Wei H: Nanomaterials with enzyme-like characteristics (nanozymes): next-generation artificial enzymes (II). Chem Soc Rev. 48:1004–1076. 2019. View Article : Google Scholar

25 

Jiang D, Ni D, Rosenkrans ZT, Huang P, Yan X and Cai W: Nanozyme: new horizons for responsive biomedical applications. Chem Soc Rev. 48:3683–3704. 2019. View Article : Google Scholar : PubMed/NCBI

26 

Huang Y, Ren J and Qu X: Nanozymes: Classification, catalytic mechanisms, activity regulation, and applications. Chem Rev. 119:4357–4412. 2019. View Article : Google Scholar : PubMed/NCBI

27 

Lin Y, Ren J and Qu X: Nano-gold as artificial enzymes: Hidden talents. Adv Mater. 26:4200–4217. 2014. View Article : Google Scholar : PubMed/NCBI

28 

Comotti M, Della Pina C, Matarrese R and Rossi M: The catalytic activity of 'naked' gold particles. Angew Chem Int Ed Engl. 43:5812–5815. 2004. View Article : Google Scholar : PubMed/NCBI

29 

Comotti M, Della Pina C, Falletta E and Rossi M: Aerobic oxidation of glucose with gold catalyst: Hydrogen peroxide as intermediate and reagent. Adv Synth Catal. 348:313–316. 2006. View Article : Google Scholar

30 

Sengupta P, Pramanik K, Datta P and Sarkar P: Chemically modified carbon nitride-chitin-acetic acid hybrid as a metal-free bifunctional nanozyme cascade of glucose oxidase-peroxidase for 'click off' colorimetric detection of peroxide and glucose. Biosens Bioelectron. 154:1120722020. View Article : Google Scholar

31 

Zhang K, Zhuo Z, Fan G, Wang Z, Chen S, Xu L, Wen Y and Wang P: Nano-ZnS decorated hierarchically porous carbon electrocatalyst with multiple enzyme-like activities as a nanozyme sensing platform for simultaneous detection of dopamine, uric acid, guanine, and adenine. Nanoscale. 13:20078–20090. 2021. View Article : Google Scholar : PubMed/NCBI

32 

Wu R, Sun M, Liu X, Qin F, Zhang X, Qian Z, Huang J, Li Y, Tan T, Chen W and Chen Z: Oxidase-like ZnCoFe three-atom nanozyme as a colorimetric platform for ascorbic acid sensing. Anal Chem. 94:14308–14316. 2022. View Article : Google Scholar : PubMed/NCBI

33 

Ragg R, Natalio F, Tahir MN, Janssen H, Kashyap A, Strand D, Strand S and Tremel W: Molybdenum trioxide nanoparticles with intrinsic sulfite oxidase activity. ACS Nano. 8:5182–5189. 2014. View Article : Google Scholar : PubMed/NCBI

34 

Mu Q, Sun Y, Guo A, Xu X, Qin B and Cai A: A bifunctionalized NiCo2O4-Au composite: Intrinsic peroxidase and oxidase catalytic activities for killing bacteria and disinfecting wound. J Hazard Mater. 402:1239392021. View Article : Google Scholar

35 

Lei L and Wang K: Synergistic combination of an intelligent nanozyme and radiotherapy for treating renal cancer. Int J Nanomedicine. 19:699–707. 2024. View Article : Google Scholar : PubMed/NCBI

36 

Chen L, Xing S, Lei Y, Chen Q, Zou Z, Quan K, Qing Z, Liu J and Yang R: A glucose-powered activatable nanozyme breaking pH and H2O2 limitations for treating diabetic infections. Angew Chem Int Ed Engl. 60:23534–23539. 2021. View Article : Google Scholar : PubMed/NCBI

37 

Gao L, Zhuang J, Nie L, Zhang J, Zhang Y, Gu N, Wang T, Feng J, Yang D, Perrett S and Yan X: Intrinsic peroxidase-like activity of ferromagnetic nanoparticles. Nat Nanotechnol. 2:577–583. 2007. View Article : Google Scholar

38 

Chen J, Liu X, Zheng G, Feng W, Wang P, Gao J, Liu J, Wang M and Wang Q: Detection of glucose based on noble metal nanozymes: Mechanism, activity regulation, and enantioselective recognition. Small. 19:e22059242023. View Article : Google Scholar

39 

Li P, Feng Y, Cheng D and Wei J: Self-template synthesis of mesoporous vanadium oxide nanospheres with intrinsic peroxidase-like activity and high antibacterial performance. J Colloid Interface Sci. 625:435–445. 2022. View Article : Google Scholar : PubMed/NCBI

40 

Yang QY, Wan CQ, Wang YX, Shen XF and Pang YH: Bismuth-based metal-organic framework peroxidase-mimic nanozyme: Preparation and mechanism for colorimetric-converted ultra-trace electrochemical sensing of chromium ion. J Hazard Mater. 451:1311482023. View Article : Google Scholar : PubMed/NCBI

41 

Liu X, Huang D, Lai C, Qin L, Zeng G, Xu P, Li B, Yi H and Zhang M: Peroxidase-like activity of smart nanomaterials and their advanced application in colorimetric glucose biosensors. Small. 15:e19001332019. View Article : Google Scholar : PubMed/NCBI

42 

Wan K, Jiang B, Tan T, Wang H and Liang M: Surface-mediated production of complexed •oh radicals and Fe•O species as a mechanism for iron oxide peroxidase-like nanozymes. Small. 18:e22043722022. View Article : Google Scholar

43 

Yao J, Cheng Y, Zhou M, Zhao S, Lin S, Wang X, Wu J, Li S and Wei H: ROS scavenging Mn3O4 nanozymes for in vivo anti-inflammation. Chem Sci. 9:2927–2933. 2018. View Article : Google Scholar : PubMed/NCBI

44 

Tong L, Wu L, Zai Y, Zhang Y, Su E and Gu N: Paper-based colorimetric glucose sensor using Prussian blue nanoparticles as mimic peroxidase. Biosens Bioelectron. 219:1147872023. View Article : Google Scholar

45 

Wu F, Du Y, Yang J, Shao B, Mi Z, Yao Y, Cui Y, He F, Zhang Y and Yang P: Peroxidase-like active nanomedicine with dual glutathione depletion property to restore oxaliplatin chemosensitivity and promote programmed cell death. ACS Nano. 16:3647–3663. 2022. View Article : Google Scholar : PubMed/NCBI

46 

Guo JJ, Wang Y and Zhao M: Target-directed functionalized ferrous phosphate-carbon dots fluorescent nanostructures as peroxidase mimetics for cancer cell detection and ROS-mediated therapy. Sens Actuat B-Chem. 297:1267392019. View Article : Google Scholar

47 

Vernekar AA, Sinha D, Srivastava S, Paramasivam PU, D'Silva P and Mugesh G: An antioxidant nanozyme that uncovers the cytoprotective potential of vanadia nanowires. Nat Commun. 5:53012014. View Article : Google Scholar : PubMed/NCBI

48 

Griendling KK, Camargo LL, Rios FJ, Alves-Lopes R, Montezano AC and Touyz RM: Oxidative stress and hypertension. Circ Res. 128:993–1020. 2021. View Article : Google Scholar : PubMed/NCBI

49 

Eftekharpour E and Fernyhough P: Oxidative stress and mitochondrial dysfunction associated with peripheral neuropathy in type 1 diabetes. Antioxid Redox Signal. 37:578–596. 2022. View Article : Google Scholar

50 

Zhao H, Zhang R, Yan X and Fan K: Superoxide dismutase nanozymes: An emerging star for anti-oxidation. J Mater Chem B. 9:6939–6957. 2021. View Article : Google Scholar : PubMed/NCBI

51 

Muzykantov VR: Targeting of superoxide dismutase and catalase to vascular endothelium. J Control Release. 71:1–21. 2001. View Article : Google Scholar : PubMed/NCBI

52 

Zelko IN, Mariani TJ and Folz RJ: Superoxide dismutase multigene family: A comparison of the CuZn-SOD (SOD1), Mn-SOD (SOD2), and EC-SOD (SOD3) gene structures, evolution, and expression. Free Radic Biol Med. 33:337–349. 2002. View Article : Google Scholar : PubMed/NCBI

53 

Miller AF: Superoxide dismutases: Ancient enzymes and new insights. FEBS Lett. 586:585–595. 2012. View Article : Google Scholar

54 

Jomova K, Alomar SY, Alwasel SH, Nepovimova E, Kuca K and Valko M: Several lines of antioxidant defense against oxidative stress: Antioxidant enzymes, nanomaterials with multiple enzyme-mimicking activities, and low-molecular-weight antioxidants. Arch Toxicol. 98:1323–1367. 2024. View Article : Google Scholar : PubMed/NCBI

55 

Chen BH and Stephen Inbaraj B: Various physicochemical and surface properties controlling the bioactivity of cerium oxide nanoparticles. Crit Rev Biotechnol. 38:1003–1024. 2018. View Article : Google Scholar : PubMed/NCBI

56 

Chen J, Patil S, Seal S and McGinnis JF: Rare earth nanoparticles prevent retinal degeneration induced by intracellular peroxides. Nat Nanotechnol. 1:142–150. 2006. View Article : Google Scholar

57 

Celardo I, Pedersen JZ, Traversa E and Ghibelli L: Pharmacological potential of cerium oxide nanoparticles. Nanoscale. 3:1411–1420. 2011. View Article : Google Scholar : PubMed/NCBI

58 

Xu C and Qu X: Cerium oxide nanoparticle: A remarkably versatile rare earth nanomaterial for biological applications. NPG Asia Mater. 6:e902014. View Article : Google Scholar

59 

Dugan LL, Turetsky DM, Du C, Lobner D, Wheeler M, Almli CR, Shen CK, Luh TY, Choi DW and Lin TS: Carboxyfullerenes as neuroprotective agents. Proc Natl Acad Sci USA. 94:9434–9439. 1997. View Article : Google Scholar : PubMed/NCBI

60 

Ali SS, Hardt JI, Quick KL, Kim-Han JS, Erlanger BF, Huang TT, Epstein CJ and Dugan LL: A biologically effective fullerene (C60) derivative with superoxide dismutase mimetic properties. Free Radic Biol Med. 37:1191–1202. 2004. View Article : Google Scholar : PubMed/NCBI

61 

Ali SS, Hardt JI and Dugan LL: SOD activity of carboxyfullerenes predicts their neuroprotective efficacy: A structure-activity study. Nanomedicine. 4:283–294. 2008. View Article : Google Scholar : PubMed/NCBI

62 

Xue B, Ge M, Fan K, Huang X, Yan X, Jiang W, Jiang B and Yang Z: Mitochondria-targeted nanozymes eliminate oxidative damage in retinal neovascularization disease. J Control Release. 350:271–283. 2022. View Article : Google Scholar : PubMed/NCBI

63 

Song C, Sheng L and Zhang X: Preparation and characterization of a thermostable enzyme (Mn-SOD) immobilized on supermagnetic nanoparticles. Appl Microbiol Biotechnol. 96:123–132. 2012. View Article : Google Scholar : PubMed/NCBI

64 

Korschelt K, Ragg R, Metzger CS, Kluenker M, Oster M, Barton B, Panthöfer M, Strand D, Kolb U, Mondeshki M, et al: Glycine-functionalized copper(ii) hydroxide nanoparticles with high intrinsic superoxide dismutase activity. Nanoscale. 9:3952–3960. 2017. View Article : Google Scholar : PubMed/NCBI

65 

Kajita M, Hikosaka K, Iitsuka M, Kanayama A, Toshima N and Miyamoto Y: Platinum nanoparticle is a useful scavenger of superoxide anion and hydrogen peroxide. Free Radic Res. 41:615–626. 2007. View Article : Google Scholar : PubMed/NCBI

66 

Li Z, Zhao Y, Huang H, Zhang C, Liu H, Wang Z, Yi M, Xie N, Shen Y, Ren X, et al: A Nanozyme-immobilized hydrogel with endogenous ROS-scavenging and oxygen generation abilities for significantly promoting oxidative diabetic wound healing. Adv Healthc Mater. 11:e22015242022. View Article : Google Scholar : PubMed/NCBI

67 

Lin A, Sun Z, Xu X, Zhao S, Li J, Sun H, Wang Q, Jiang Q, Wei H and Shi D: Self-cascade uricase/catalase mimics alleviate acute gout. Nano Lett. 22:508–516. 2022. View Article : Google Scholar

68 

Cai G, Li R, Chai X, Cai X, Zheng K, Wang Y, Fan K, Guo Z, Guo J and Jiang W: Catalase-templated nanozyme-loaded microneedles integrated with polymyxin B for immunoregulation and antibacterial activity in diabetic wounds. J Colloid Interface Sci. 667:529–542. 2024. View Article : Google Scholar : PubMed/NCBI

69 

Wang C, Li Y, Yang W, Zhou L and Wei S: Nanozyme with robust catalase activity by multiple mechanisms and its application for hypoxic tumor treatment. Adv Healthc Mater. 10:e21006012021. View Article : Google Scholar : PubMed/NCBI

70 

Xu D, Wu L, Yao H and Zhao L: Catalase-like nanozymes: classification, catalytic mechanisms, and their applications. Small. 18:e22034002022. View Article : Google Scholar : PubMed/NCBI

71 

Fan C, Tang Y, Wang H, Huang Y, Xu F, Yang Y, Huang Y, Rong W and Lin Y: ZIF-90 with biomimetic Zn-N coordination structures as an effective nanozyme to mimic natural hydrolase. Nanoscale. 14:7985–7990. 2022. View Article : Google Scholar : PubMed/NCBI

72 

Sun S, Zhang Z, Xiang Y, Cao M and Yu D: Amino acid-mediated synthesis of the ZIF-8 nanozyme that reproduces both the zinc-coordinated active center and hydrophobic pocket of natural carbonic anhydrase. Langmuir. 38:1621–1630. 2022. View Article : Google Scholar : PubMed/NCBI

73 

Zhou Q, Zhang T, Jie J, Hou Y, Hu Z, Jiao Z and Su H: TiO2 as a nanozyme mimicking photolyase to repair DNA damage. J Phys Chem Lett. 13:10929–10935. 2022. View Article : Google Scholar : PubMed/NCBI

74 

O'Hara AM and Shanahan F: The gut flora as a forgotten organ. EMBO Rep. 7:688–693. 2006. View Article : Google Scholar : PubMed/NCBI

75 

Wang Y, Shen J, Handschuh-Wang S, Qiu M, Du S and Wang B: Microrobots for targeted delivery and therapy in digestive system. ACS Nano. 17:27–50. 2023. View Article : Google Scholar

76 

Janowitz HD: Digestive system. Annu Rev Physiol. 23:153–182. 1961. View Article : Google Scholar : PubMed/NCBI

77 

Feng K, Wang G, Wang S, Ma J, Wu H, Ma M and Zhang Y: Breaking the pH limitation of nanozymes: Mechanisms, methods, and applications. Adv Mater. 36:e24016192024. View Article : Google Scholar : PubMed/NCBI

78 

Hua S, Dong X, Peng Q, Zhang K, Zhang X and Yang J: Single-atom nanozymes shines diagnostics of gastrointestinal diseases. J Nanobiotechnology. 22:2862024. View Article : Google Scholar : PubMed/NCBI

79 

Kelly RJ: Emerging multimodality approaches to treat localized esophageal cancer. J Natl Compr Canc Netw. 17:1009–1014. 2019. View Article : Google Scholar : PubMed/NCBI

80 

Barker HE, Paget JT, Khan AA and Harrington KJ: The tumour microenvironment after radiotherapy: Mechanisms of resistance and recurrence. Nat Rev Cancer. 15:409–425. 2015. View Article : Google Scholar : PubMed/NCBI

81 

Diehn M, Cho RW, Lobo NA, Kalisky T, Dorie MJ, Kulp AN, Qian D, Lam JS, Ailles LE, Wong M, et al: Association of reactive oxygen species levels and radioresistance in cancer stem cells. Nature. 458:780–783. 2009. View Article : Google Scholar : PubMed/NCBI

82 

Xie J, Gong L, Zhu S, Yong Y, Gu Z and Zhao Y: Emerging strategies of nanomaterial-mediated tumor radiosensitization. Adv Mater. 31:e18022442019. View Article : Google Scholar

83 

Petroni G, Cantley LC, Santambrogio L, Formenti SC and Galluzzi L: Radiotherapy as a tool to elicit clinically actionable signalling pathways in cancer. Nat Rev Clin Oncol. 19:114–131. 2022. View Article : Google Scholar :

84 

Worrell SG, Goodman KA, Altorki NK, Ashman JB, Crabtree TD, Dorth J, Firestone S, Harpole DH, Hofstetter WL, Hong TS, et al: The society of thoracic surgeons/american society for radiation oncology updated clinical practice guidelines on multimodality therapy for locally advanced cancer of the esophagus or gastroesophageal junction. Ann Thorac Surg. 117:15–32. 2024. View Article : Google Scholar

85 

Zhou LL, Guan Q, Zhou W, Kan JL, Teng K, Hu M and Dong YB: A multifunctional covalent organic framework nanozyme for promoting ferroptotic radiotherapy against esophageal cancer. ACS Nano. 17:20445–20461. 2023. View Article : Google Scholar : PubMed/NCBI

86 

Zhou X, You M, Wang F, Wang Z, Gao X, Jing C, Liu J, Guo M, Li J, Luo A, et al: Multifunctional graphdiyne-cerium oxide nanozymes facilitate MicroRNA delivery and attenuate tumor hypoxia for highly efficient radiotherapy of esophageal cancer. Adv Mater. 33:e21005562021. View Article : Google Scholar : PubMed/NCBI

87 

Liu J, Gao J, Zhang A, Guo Y, Fan S, He Y, Yang K, Wang J, Cui D and Cheng Y: Carbon nanocage-based nanozyme as an endogenous H2O2-activated oxygenerator for real-time bimodal imaging and enhanced phototherapy of esophageal cancer. Nanoscale. 12:21674–21686. 2020. View Article : Google Scholar : PubMed/NCBI

88 

Wu Y, Song M, Xin Z, Zhang X, Zhang Y, Wang C, Li S and Gu N: Ultra-small particles of iron oxide as peroxidase for immunohistochemical detection. Nanotechnology. 22:2257032011. View Article : Google Scholar : PubMed/NCBI

89 

Wang P, Wang T, Hong J, Yan X and Liang M: Nanozymes: A new disease imaging strategy. Front Bioeng Biotechnol. 8:152020. View Article : Google Scholar : PubMed/NCBI

90 

Krause J, Brokmann F, Rosenbaum C and Weitschies W: The challenges of drug delivery to the esophagus and how to overcome them. Expert Opin Drug Deliv. 19:119–131. 2022. View Article : Google Scholar : PubMed/NCBI

91 

Senchukova MA: Helicobacter pylori and gastric cancer progression. Curr Microbiol. 79:3832022. View Article : Google Scholar : PubMed/NCBI

92 

Marshall BJ and Warren JR: Unidentified curved bacilli in the stomach of patients with gastritis and peptic ulceration. Lancet. 1:1311–1315. 1984. View Article : Google Scholar : PubMed/NCBI

93 

Ansari S and Yamaoka Y: Helicobacter pylori infection, its laboratory diagnosis, and antimicrobial resistance: A perspective of clinical relevance. Clin Microbiol Rev. 35:e00258212022. View Article : Google Scholar : PubMed/NCBI

94 

Hu Y, Zhu Y and Lu NH: The management of Helicobacter pylori infection and prevention and control of gastric cancer in China. Front Cell Infect Microbiol. 12:10492792022. View Article : Google Scholar : PubMed/NCBI

95 

Zhang L, Zhang L, Deng H, Li H, Tang W, Guan L, Qiu Y, Donovan MJ, Chen Z and Tan W: In vivo activation of pH-responsive oxidase-like graphitic nanozymes for selective killing of Helicobacter pylori. Nat Commun. 12:20022021. View Article : Google Scholar : PubMed/NCBI

96 

Efentakis M and Dressman JB: Gastric juice as a dissolution medium: Surface tension and pH. Eur J Drug Metab Pharmacokinet. 23:97–102. 1998. View Article : Google Scholar : PubMed/NCBI

97 

Yan LX, Wang BB, Zhao X, Chen LJ and Yan XP: A pH-Responsive persistent luminescence nanozyme for selective imaging and killing of helicobacter pylori and common resistant bacteria. ACS Appl Mater Interfaces. 13:60955–60965. 2021. View Article : Google Scholar : PubMed/NCBI

98 

Guo Z, Zhu AT, Fang RH and Zhang L: Recent developments in nanoparticle-based photo-immunotherapy for cancer treatment. Small Methods. 7:e23002522023. View Article : Google Scholar : PubMed/NCBI

99 

Zhang A, Pan S, Zhang Y, Chang J, Cheng J, Huang Z, Li T, Zhang C, de la Fuentea JM, Zhang Q and Cui D: Carbon-gold hybrid nanoprobes for real-time imaging, photothermal/photodynamic and nanozyme oxidative therapy. Theranostics. 9:3443–3458. 2019. View Article : Google Scholar : PubMed/NCBI

100 

Zhang Y, Li Y, Li J, Mu F, Wang J, Shen C, Wang H, Huang F, Chen B, Luo Z and Wang L: DNA-Templated Ag@ Pd nanoclusters for NIR-II photoacoustic imaging-guided photothermal-augmented nanocatalytic therapy. Adv Healthc Mater. 12:e23002672023. View Article : Google Scholar

101 

Ma J, Yao Q, Lv S, Yi J, Zhu D, Zhu C, Wang L and Su S: Integrated triple signal amplification strategy for ultrasensitive electrochemical detection of gastric cancer-related microRNA utilizing MoS2-based nanozyme, hybridization chain reaction, and horseradish peroxidase. J Nanobiotechnology. 22:5962024. View Article : Google Scholar :

102 

Yao QH, Zeng MH, Zhang C, Zheng F, Jin JW, Ye TX, Wang YT, Chen XM, Guo ZY and Chen X: Polyphenol briged bimetallic-heterostructure inducing chiral peroxidase nanozyme activity for enantiomers identification in gastric cancer. Biosens Bioelectron. 282:1174492025. View Article : Google Scholar : PubMed/NCBI

103 

Zhuang Y, Yin H, Huang Y, Jiang F, Li L, Wu Z, Yang Y, Cao X and Wei W: Catalytic hairpin assembly-powered nanozyme-SERS dual-function sensing system for ultrasensitive detection of gastric precancerous lesions. Biosens Bioelectron. 283:1175362025. View Article : Google Scholar : PubMed/NCBI

104 

Deng X, Wu J, Jiang ZW, Liu W, Gong X, Cao Y, Zhang P and Wang Y: Detection of d-Amino acids in saliva for gastric cancer diagnosis using Pt/MXene plasmonic nanozymes. Anal Chem. 97:10289–10298. 2025. View Article : Google Scholar : PubMed/NCBI

105 

Asrani SK, Devarbhavi H, Eaton J and Kamath PS: Burden of liver diseases in the world. J Hepatol. 70:151–171. 2019. View Article : Google Scholar

106 

Hassan A and Fontana RJ: The diagnosis and management of idiosyncratic drug-induced liver injury. Liver Int. 39:31–41. 2019. View Article : Google Scholar

107 

Coccolini F, Coimbra R, Ordonez C, Kluger Y, Vega F, Moore EE, Biffl W, Peitzman A, Horer T, Abu-Zidan FM, et al: Liver trauma: WSES 2020 guidelines. World J Emerg Surg. 15:242020. View Article : Google Scholar : PubMed/NCBI

108 

Yan M, Huo Y, Yin S and Hu H: Mechanisms of acetaminophen-induced liver injury and its implications for therapeutic interventions. Redox Biol. 17:274–283. 2018. View Article : Google Scholar : PubMed/NCBI

109 

Zhou C, Zhang L, Xu Z, Sun T, Gong M, Liu Y and Zhang D: Self-propelled ultrasmall AuNPs-tannic acid hybrid nanozyme with ROS-scavenging and anti-inflammatory activity for drug-induced liver injury alleviation. Small. 19:e22064082023. View Article : Google Scholar : PubMed/NCBI

110 

Bai H, Kong F, Feng K, Zhang X, Dong H, Liu D, Ma M, Liu F, Gu N and Zhang Y: Prussian blue nanozymes prevent anthracycline-induced liver injury by attenuating oxidative stress and regulating inflammation. ACS Appl Mater Interfaces. 13:42382–42395. 2021. View Article : Google Scholar : PubMed/NCBI

111 

Xia F, Hu X, Zhang B, Wang X, Guan Y, Lin P, Ma Z, Sheng J, Ling D and Li F: Ultrasmall ruthenium nanoparticles with boosted antioxidant activity upregulate regulatory T cells for highly efficient liver injury therapy. Small. 18:e22015582022. View Article : Google Scholar : PubMed/NCBI

112 

Wu H, Xia F, Zhang L, Fang C, Lee J, Gong L, Gao J, Ling D and Li F: A ROS-Sensitive nanozyme-augmented photoacoustic nanoprobe for early diagnosis and therapy of acute liver failure. Adv Mater. 34:e21083482022. View Article : Google Scholar

113 

Marroni CA, Fleck AM Jr, Fernandes SA, Galant LH, Mucenic M, de Mattos Meine MH, Mariante-Neto G and Brandão ABM: Liver transplantation and alcoholic liver disease: History, controversies, and considerations. World J Gastroenterol. 24:2785–2805. 2018. View Article : Google Scholar : PubMed/NCBI

114 

Geng X, Du X, Wang W, Zhang C, Liu X, Qu Y, Zhao M, Li W, Zhang M, Tu K and Li YQ: Confined cascade metabolic reprogramming nanoreactor for targeted alcohol detoxification and alcoholic liver injury management. ACS Nano. 17:7443–7455. 2023. View Article : Google Scholar : PubMed/NCBI

115 

Friedman SL and Pinzani M: Hepatic fibrosis 2022: Unmet needs and a blueprint for the future. Hepatology. 75:473–488. 2022. View Article : Google Scholar

116 

Zhang M, Serna-Salas S, Damba T, Borghesan M, Demaria M and Moshage H: Hepatic stellate cell senescence in liver fibrosis: Characteristics, mechanisms and perspectives. Mech Ageing Dev. 199:1115722021. View Article : Google Scholar : PubMed/NCBI

117 

Carloni V, Luong TV and Rombouts K: Hepatic stellate cells and extracellular matrix in hepatocellular carcinoma: More complicated than ever. Liver Int. 34:834–843. 2014. View Article : Google Scholar : PubMed/NCBI

118 

Ezhilarasan D, Sokal E and Najimi M: Hepatic fibrosis: It is time to go with hepatic stellate cell-specific therapeutic targets. Hepatobiliary Pancreat Dis Int. 17:192–197. 2018. View Article : Google Scholar : PubMed/NCBI

119 

Zhang X, Zhang S, Yang Z, Wang Z, Tian X and Zhou R: Self-cascade MoS(2) nanozymes for efficient intracellular antioxidation and hepatic fibrosis therapy. Nanoscale. 13:12613–12622. 2021. View Article : Google Scholar : PubMed/NCBI

120 

Lu Q, Zhou Y, Xu M, Liang X, Jing H, Wang X and Li N: Sequential delivery for hepatic fibrosis treatment based on carvedilol loaded star-like nanozyme. J Control Release. 341:247–260. 2022. View Article : Google Scholar

121 

Tang Z, Liu Y, He M and Bu W: Chemodynamic therapy: Tumour microenvironment-mediated fenton and fenton-like reactions. Angew Chem Int Ed Engl. 58:946–956. 2019. View Article : Google Scholar

122 

Tarangelo A and Dixon SJ: Nanomedicine: An iron age for cancer therapy. Nat Nanotechnol. 11:921–922. 2016. View Article : Google Scholar : PubMed/NCBI

123 

Jiang Y, Zhao X, Huang J, Li J, Upputuri PK, Sun H, Han X, Pramanik M, Miao Y, Duan H, et al: Transformable hybrid semiconducting polymer nanozyme for second near-infrared photothermal ferrotherapy. Nat Commun. 11:18572020. View Article : Google Scholar : PubMed/NCBI

124 

Dong C, Dai X, Wang X, Lu Q, Chen L, Song X, Ding L, Huang H, Feng W, Chen Y and Chang M: A Calcium fluoride nanozyme for ultrasound-amplified and Ca2+ -overload-enhanced catalytic tumor nanotherapy. Adv Mater. 34:e22056802022. View Article : Google Scholar

125 

Kang S, Gil Y, Yim G, Min D and Jang H: Osmium-tellurium nanozymes for pentamodal combinatorial cancer therapy. ACS Appl Mater Interfaces. 13:44124–44135. 2021. View Article : Google Scholar : PubMed/NCBI

126 

Jiang B, Yan L, Zhang J, Zhou M, Shi G, Tian X, Fan K, Hao C and Yan X: Biomineralization synthesis of the cobalt nanozyme in SP94-ferritin nanocages for prognostic diagnosis of hepatocellular carcinoma. ACS Appl Mater Interfaces. 11:9747–9755. 2019. View Article : Google Scholar : PubMed/NCBI

127 

Liu X, Mei X, Yang J and Li Y: Hydrogel-involved colorimetric platforms based on layered double oxide nanozymes for point-of-care detection of liver-related biomarkers. ACS Appl Mater Interfaces. 14:6985–6993. 2022. View Article : Google Scholar : PubMed/NCBI

128 

He C, Ke Z, Liu K, Peng J, Yang Q, Wang L, Feng G and Fang J: Nanozyme-based dual-signal sensing system for colorimetric and photothermal detection of AChE activity in the blood of liver-injured mice. Anal Bioanal Chem. 415:2655–2664. 2023. View Article : Google Scholar : PubMed/NCBI

129 

Liu J, Shi X, Qu Y and Wang G: Functionalized ZnMnFe2O4-PEG-FA nanoenzymes integrating diagnosis and therapy for targeting hepatic carcinoma guided by multi-modality imaging. Nanoscale. 15:11013–11025. 2023. View Article : Google Scholar : PubMed/NCBI

130 

Alimardani V, Rahiminezhad Z, DehghanKhold M, Farahavar G, Jafari M, Abedi M, Moradi L, Niroumand U, Ashfaq M, Abolmaali SS and Yousefi G: Nanotechnology-based cell-mediated delivery systems for cancer therapy and diagnosis. Drug Deliv Transl Res. 13:189–221. 2023. View Article : Google Scholar

131 

Mederos MA, Reber HA and Girgis MD: Acute pancreatitis: A review. JAMA. 325:382–390. 2021. View Article : Google Scholar : PubMed/NCBI

132 

Tan JH, Cao RC, Zhou L, Zhou ZT, Chen HJ, Xu J, Chen XM, Jin YC, Lin JY, Zeng JL, et al: ATF6 aggravates acinar cell apoptosis and injury by regulating p53/AIFM2 transcription in severe acute pancreatitis. Theranostics. 10:8298–8314. 2020. View Article : Google Scholar : PubMed/NCBI

133 

Sah RP, Garg P and Saluja AK: Pathogenic mechanisms of acute pancreatitis. Curr Opin Gastroenterol. 28:507–515. 2012. View Article : Google Scholar : PubMed/NCBI

134 

Bhatia M, Wong FL, Cao Y, Lau HY, Huang J, Puneet P and Chevali L: Pathophysiology of acute pancreatitis. Pancreatology. 5:132–144. 2005. View Article : Google Scholar : PubMed/NCBI

135 

Escobar J, Pereda J, López-Rodas G and Sastre J: Redox signaling and histone acetylation in acute pancreatitis. Free Radic Biol Med. 52:819–837. 2012. View Article : Google Scholar

136 

Pasari LP, Khurana A, Anchi P, Aslam Saifi M, Annaldas S and Godugu C: Visnagin attenuates acute pancreatitis via Nrf2/NFκB pathway and abrogates associated multiple organ dysfunction. Biomed Pharmacother. 112:1086292019. View Article : Google Scholar

137 

Pérez S, Pereda J, Sabater L and Sastre J: Redox signaling in acute pancreatitis. Redox Biol. 5:1–14. 2015. View Article : Google Scholar : PubMed/NCBI

138 

Yao Q, Jiang X, Zhai YY, Luo LZ, Xu HL, Xiao J, Kou L and Zhao YZ: Protective effects and mechanisms of bilirubin nanomedicine against acute pancreatitis. J Control Release. 322:312–325. 2020. View Article : Google Scholar : PubMed/NCBI

139 

Jakkampudi A, Jangala R, Reddy BR, Mitnala S, Nageshwar Reddy D and Talukdar R: NF-κB in acute pancreatitis: Mechanisms and therapeutic potential. Pancreatology. 16:477–488. 2016. View Article : Google Scholar : PubMed/NCBI

140 

Rakonczay Z Jr, Hegyi P, Takács T, McCarroll J and Saluja AK: The role of NF-kappaB activation in the pathogenesis of acute pancreatitis. Gut. 57:259–267. 2008. View Article : Google Scholar

141 

Xie X, Zhao J, Gao W, Chen J, Hu B, Cai X and Zheng Y: Prussian blue nanozyme-mediated nanoscavenger ameliorates acute pancreatitis via inhibiting TLRs/NF-κB signaling pathway. Theranostics. 11:3213–3228. 2021. View Article : Google Scholar :

142 

Yang L, Ye F, Liu J, Klionsky DJ, Tang D and Kang R: Extracellular SQSTM1 exacerbates acute pancreatitis by activating autophagy-dependent ferroptosis. Autophagy. 19:1733–1744. 2023. View Article : Google Scholar :

143 

Li H, Lin Y, Zhang L, Zhao J and Li P: Ferroptosis and its emerging roles in acute pancreatitis. Chin Med J (Engl). 135:2026–2034. 2022. View Article : Google Scholar : PubMed/NCBI

144 

Fan R, Sui J, Dong X, Jing B and Gao Z: Wedelolactone alleviates acute pancreatitis and associated lung injury via GPX4 mediated suppression of pyroptosis and ferroptosis. Free Radic Biol Med. 173:29–40. 2021. View Article : Google Scholar : PubMed/NCBI

145 

Li Y, Cai R, Chen K, Zhang Y, Chen X, Sun B, Jiang Y, Tan C, Peng C, Song Y, et al: Ca/Fe-Based nanozymes relieve severe acute pancreatitis by ferroptosis regulation and reactive oxygen species scavenging. ACS Appl. Nano Mater. 14:12968–12979. 2023. View Article : Google Scholar

146 

Xie P, Zhang L, Shen H, Wu H, Zhao J, Wang S and Hu L: Biodegradable MoSe2-polyvinylpyrrolidone nanoparticles with multi-enzyme activity for ameliorating acute pancreatitis. J Nanobiotechnology. 20:1132022. View Article : Google Scholar :

147 

Sarvepalli D, Rashid MU, Rahman AU, Ullah W, Hussain I, Hasan B, Jehanzeb S, Khan AK, Jain AG, Khetpal N and Ahmad S: Gemcitabine: A review of chemoresistance in pancreatic cancer. Crit Rev Oncog. 24:199–212. 2019. View Article : Google Scholar : PubMed/NCBI

148 

Kang S, Gil YG, Min DH and Jang H: Nonrecurring circuit nanozymatic enhancement of hypoxic pancreatic cancer phototherapy using speckled Ru-Te hollow nanorods. ACS Nano. 14:4383–4394. 2020. View Article : Google Scholar : PubMed/NCBI

149 

Chen H, Qiu Y, Ding D, Lin H, Sun W, Wang GD, Huang W, Zhang W, Lee D, Liu G, et al: Gadolinium-encapsulated graphene carbon nanotheranostics for imaging-guided photodynamic therapy. Adv Mater. Jul 23–2018.Epub ahead of print. View Article : Google Scholar

150 

Li S, Shang L, Xu B, Wang S, Gu K, Wu Q, Sun Y, Zhang Q, Yang H, Zhang F, et al: A nanozyme with photo-enhanced dual enzyme-like activities for deep pancreatic cancer therapy. Angew Chem Int Ed Engl. 58:12624–12631. 2019. View Article : Google Scholar : PubMed/NCBI

151 

Chen J, Bao Y, Song Y, Zhang C, Qiu F, Sun Y, Xin L, Cao J, Jiang Y, Luo J, et al: Hypoxia-alleviated nanoplatform to enhance chemosensitivity and sonodynamic effect in pancreatic cancer. Cancer Lett. 520:100–108. 2021. View Article : Google Scholar : PubMed/NCBI

152 

Zhang G, Li N, Qi Y, Zhao Q, Zhan J and Yu D: Synergistic ferroptosis-gemcitabine chemotherapy of the gemcitabine loaded carbonaceous nanozymes to enhance the treatment and magnetic resonance imaging monitoring of pancreatic cancer. Acta Biomater. 142:284–297. 2022. View Article : Google Scholar : PubMed/NCBI

153 

Ramos GP and Papadakis KA: Mechanisms of disease: Inflammatory bowel diseases. Mayo Clin Proc. 94:155–165. 2019. View Article : Google Scholar : PubMed/NCBI

154 

Xavier RJ and Podolsky DK: Unravelling the pathogenesis of inflammatory bowel disease. Nature. 448:427–434. 2007. View Article : Google Scholar : PubMed/NCBI

155 

Liu P, Li Y, Wang R, Ren F and Wang X: Oxidative stress and antioxidant nanotherapeutic approaches for inflammatory bowel disease. Biomedicines. 10:852021. View Article : Google Scholar

156 

Weiss GA and Hennet T: Mechanisms and consequences of intestinal dysbiosis. Cell Mol Life Sci. 74:2959–2977. 2017. View Article : Google Scholar : PubMed/NCBI

157 

Eliakim R and Magro F: Imaging techniques in IBD and their role in follow-up and surveillance. Nat Rev Gastroenterol Hepatol. 11:722–736. 2014. View Article : Google Scholar : PubMed/NCBI

158 

Cao Y, Cheng K, Yang M, Deng Z, Ma Y, Yan X, Zhang Y, Jia Z, Wang J, Tu K, et al: Orally administration of cerium oxide nanozyme for computed tomography imaging and anti-inflammatory/anti-fibrotic therapy of inflammatory bowel disease. J Nanobiotechnology. 21:212023. View Article : Google Scholar : PubMed/NCBI

159 

Li X, Cao L, Li J, Li Z, Ma H, Cheng S, Xu H and Zhao Y: Orally Administrated Inulin-Modified Nanozymes for CT-Guided IBD Theranostics. Int J Nanomedicine. 20:2119–2131. 2025. View Article : Google Scholar : PubMed/NCBI

160 

He Y, Jin Z, Wang Y, Wu C, He X, Weng W, Cai X and Cheng K: Multifunctional double-loaded oral nanoplatform for computed tomography imaging-guided and integrated treatment of inflammatory bowel disease. ACS Nano. 19:14893–14913. 2025. View Article : Google Scholar : PubMed/NCBI

161 

Deng Z, Ma W, Ding C, Wei C, Gao B, Zhu M, Zhang Y, Wu F and Zhang M, Li R and Zhang M: Metal polyphenol network/cerium oxide artificial enzymes therapeutic nanoplatform for MRI/CT-aided intestinal inflammation management. Nano Today. 53:1020442023. View Article : Google Scholar

162 

Zhou D, Yin Y, Zhu Z, Gao Y, Yang J, Pan Y and Song Y: Orally administered platinum nanomarkers for urinary monitoring of inflammatory bowel disease. ACS Nano. 16:18503–18514. 2022. View Article : Google Scholar : PubMed/NCBI

163 

Duan ZY, Liu JQ, Yin P, Li JJ, Cai GY and Chen XM: Impact of aging on the risk of platinum-related renal toxicity: A systematic review and meta-analysis. Cancer Treat Rev. 69:243–253. 2018. View Article : Google Scholar : PubMed/NCBI

164 

Ma Y, Zhao J, Cheng L, Li C, Yan X, Deng Z, Zhang Y, Liang J, Liu C and Zhang M: Versatile carbon dots with superoxide dismutase-like nanozyme activity and red fluorescence for inflammatory bowel disease therapeutics. Carbon. 204:526–537. 2023. View Article : Google Scholar

165 

Zhao S, Li Y, Liu Q, Li S, Cheng Y, Cheng C, Sun Z, Du Y, Butch CJ and Wei H: An Orally Administered CeO2@montmorillonite nanozyme targets inflammation for inflammatory bowel disease therapy. Adv Funct Mater. 30:20046922020. View Article : Google Scholar

166 

Wang Q, Cheng C, Zhao S, Liu Q, Zhang Y, Liu W, Zhao X, Zhang H, Pu J, Zhang S, et al: A valence-engineered self-cascading antioxidant nanozyme for the therapy of inflammatory bowel disease. Angew Chem Int Ed Engl. 61:e2022011012022. View Article : Google Scholar : PubMed/NCBI

167 

Liu Y, Cheng Y, Zhang H, Zhou M, Yu Y, Lin S, Jiang B, Zhao X, Miao L, Wei CW, et al: Integrated cascade nanozyme catalyzes in vivo ROS scavenging for anti-inflammatory therapy. Sci Adv. 6:eabb26952020. View Article : Google Scholar : PubMed/NCBI

168 

Yu Y, Zhao X, Xu X, Cai C, Tang X, Zhang Q, Zhong L, Zhou F, Yang D and Zhu Z: Rational design of orally administered cascade nanozyme for inflammatory bowel disease therapy. Adv Mater. 35:e23049672023. View Article : Google Scholar : PubMed/NCBI

169 

Huang Q, Yang Y, Zhu Y, Chen Q, Zhao T, Xiao Z, Wang M, Song X, Jiang Y, Yang Y, et al: Oral metal-free melanin nanozymes for natural and durable targeted treatment of inflammatory bowel disease (IBD). Small. 19:e22073502023. View Article : Google Scholar : PubMed/NCBI

170 

Yao H, Wang F, Chong H, Wang J, Bai Y, Du M, Yuan X, Yang X, Wu M, Li Y and Pang H: A Curcumin-modified coordination polymers with ros scavenging and macrophage phenotype regulating properties for efficient ulcerative colitis treatment. Adv Sci (Weinh). 10:e23006012023. View Article : Google Scholar : PubMed/NCBI

171 

Cao F, Jin L, Gao Y, Ding Y, Wen H, Qian Z, Zhang C, Hong L, Yang H, Zhang J, et al: Artificial-enzymes-armed Bifidobacterium longum probiotics for alleviating intestinal inflammation and microbiota dysbiosis. Nat Nanotechnol. 18:617–627. 2023. View Article : Google Scholar : PubMed/NCBI

172 

Zhu H and Li YR: Oxidative stress and redox signaling mechanisms of inflammatory bowel disease: Updated experimental and clinical evidence. Exp Biol Med (Maywood). 237:474–480. 2012. View Article : Google Scholar : PubMed/NCBI

173 

Miao Z, Jiang S, Ding M, Sun S, Ma Y, Younis MR, He G, Wang J, Lin J, Cao Z, et al: Ultrasmall rhodium nanozyme with RONS scavenging and photothermal activities for anti-inflammation and antitumor theranostics of colon diseases. Nano Lett. 20:3079–3089. 2020. View Article : Google Scholar : PubMed/NCBI

174 

Wang Y, Dai X, Wu L, Xiang H, Chen Y and Zhang R: Atomic vacancies-engineered ultrathin trimetallic nanozyme with anti-inflammation and antitumor performances for intestinal disease treatment. Biomaterials. 299:1221782023. View Article : Google Scholar : PubMed/NCBI

175 

Chen J, Xu F, Ruan X, Sun J, Zhang Y, Zhang H, Zhao J, Zheng J, Larsson SC, Wang X, et al: Therapeutic targets for inflammatory bowel disease: Proteome-wide Mendelian randomization and colocalization analyses. EBioMedicine. 89:1044942023. View Article : Google Scholar : PubMed/NCBI

176 

Gonzalez CG, Mills RH, Zhu Q, Sauceda C, Knight R, Dulai PS and Gonzalez DJ: Location-specific signatures of Crohn's disease at a multi-omics scale. Microbiome. 10:1332022. View Article : Google Scholar : PubMed/NCBI

177 

Bagherzadeh F and Mohammadi-Moghadam F: New insights into the role of metal(loid)s in the development of ulcerative colitis: A systematic review. Environ Sci Pollut Res Int. 30:66486–66493. 2023. View Article : Google Scholar : PubMed/NCBI

178 

Chen X, Wang S, Mao X, Xiang X, Ye S, Chen J, Zhu A, Meng Y, Yang X, Peng S, et al: Adverse health effects of emerging contaminants on inflammatory bowel disease. Front Public Health. 11:11407862023. View Article : Google Scholar : PubMed/NCBI

179 

Li M, Liu J, Shi L, Zhou C, Zou M, Fu D, Yuan Y, Yao C, Zhang L, Qin S, et al: Gold nanoparticles-embedded ceria with enhanced antioxidant activities for treating inflammatory bowel disease. Bioact Mater. 25:95–106. 2023.PubMed/NCBI

180 

Kim HS, Lee S and Lee DY: Aurozyme: A revolutionary nanozyme in colitis, switching peroxidase-like to catalase-like activity. Small. 19:e23023312023. View Article : Google Scholar : PubMed/NCBI

181 

Zhao H, Du Y, Liu L, Du Y, Cui K, Yu P, Li L, Zhu Y, Jiang W, Li Z, et al: Oral nanozyme-engineered probiotics for the treatment of ulcerative colitis. J Mater Chem B. 10:4002–4011. 2022. View Article : Google Scholar : PubMed/NCBI

182 

Chen QW, Cao MW, Qiao JY, Li QR and Zhang XZ: Integrated cascade catalysis of microalgal bioenzyme and inorganic nanozyme for anti-inflammation therapy. Nanoscale Horiz. 8:489–498. 2023. View Article : Google Scholar : PubMed/NCBI

183 

Salas A, Hernandez-Rocha C, Duijvestein M, Faubion W, McGovern D, Vermeire S, Vetrano S and Vande Casteele N: JAK-STAT pathway targeting for the treatment of inflammatory bowel disease. Nat Rev Gastroenterol Hepatol. 17:323–337. 2020. View Article : Google Scholar : PubMed/NCBI

184 

Wang B and Shen J: NF-κB inducing kinase regulates intestinal immunity and homeostasis. Front Immunol. 13:8956362022. View Article : Google Scholar

185 

Zhu D, Wu H, Jiang K, Xu Y, Miao Z, Wang H and Ma Y: Zero-valence selenium-enriched prussian blue nanozymes reconstruct intestinal barrier against inflammatory bowel disease via inhibiting ferroptosis and T cells differentiation. Adv Healthc Mater. 12:e22031602023. View Article : Google Scholar : PubMed/NCBI

186 

Wang CJ, Ko GR, Lee YY, Park J, Park W, Park TE, Jin Y, Kim SN, Lee JS and Park CG: Polymeric DNase-I nanozymes targeting neutrophil extracellular traps for the treatment of bowel inflammation. Nano Converg. 11:62024. View Article : Google Scholar : PubMed/NCBI

187 

Ma Y, Gao W, Zhang Y, Yang M, Yan X, Zhang Y, Li G, Liu C, Xu C and Zhang M: Biomimetic MOF nanoparticles delivery of C-Dot nanozyme and CRISPR/Cas9 system for site-specific treatment of ulcerative colitis. ACS Appl Mater Interfaces. 14:6358–6369. 2022. View Article : Google Scholar : PubMed/NCBI

188 

Stoffel EM and Murphy CC: Epidemiology and mechanisms of the increasing incidence of colon and rectal cancers in young adults. Gastroenterology. 158:341–353. 2020. View Article : Google Scholar

189 

Clarke WT and Feuerstein JD: Colorectal cancer surveillance in inflammatory bowel disease: Practice guidelines and recent developments. World J Gastroenterol. 25:4148–4157. 2019. View Article : Google Scholar : PubMed/NCBI

190 

Hou YJ, Yang XX, Liu RQ, Zhao D, Guo CX, Zhu AC, Wen MN, Liu Z, Qu GF and Meng HX: Pathological mechanism of photodynamic therapy and photothermal therapy based on nanoparticles. Int J Nanomedicine. 15:6827–6838. 2020. View Article : Google Scholar : PubMed/NCBI

191 

Zhu X, Gong Y, Liu Y, Yang C, Wu S, Yuan G, Guo X, Liu J and Qin X: Ru@CeO2 yolk shell nanozymes: Oxygen supply in situ enhanced dual chemotherapy combined with photothermal therapy for orthotopic/subcutaneous colorectal cancer. Biomaterials. 242:1199232020. View Article : Google Scholar

192 

Zhou R, Ohulchanskyy TY, Xu Y, Ziniuk R, Xu H, Liu L and Qu J: Tumor-microenvironment-activated NIR-II nanotheranostic platform for precise diagnosis and treatment of colon cancer. ACS Appl Mater Interfaces. 14:23206–23218. 2022. View Article : Google Scholar : PubMed/NCBI

193 

Agostinis P, Berg K, Cengel KA, Foster TH, Girotti AW, Gollnick SO, Hahn SM, Hamblin MR, Juzeniene A, Kessel D, et al: Photodynamic therapy of cancer: An update. CA Cancer J Clin. 61:250–281. 2011.PubMed/NCBI

194 

Hao Y, Chen Y, He X, Yu Y, Han R, Li Y, Yang C, Hu D and Qian Z: Polymeric nanoparticles with ROS-responsive prodrug and platinum nanozyme for enhanced chemophotodynamic therapy of colon cancer. Adv Sci (Weinh). 7:20018532020. View Article : Google Scholar : PubMed/NCBI

195 

Duo Y, Suo M, Zhu D, Li Z, Zheng Z and Tang BZ: AIEgen-based bionic nanozymes for the interventional photodynamic therapy-based treatment of orthotopic colon cancer. ACS Appl Mater Interfaces. 14:26394–26403. 2022. View Article : Google Scholar : PubMed/NCBI

196 

Son S, Kim JH, Wang X, Zhang C, Yoon SA, Shin J, Sharma A, Lee MH, Cheng L, Wu J and Kim JS: Multifunctional sonosensitizers in sonodynamic cancer therapy. Chem Soc Rev. 49:3244–3261. 2020. View Article : Google Scholar : PubMed/NCBI

197 

Zhang Y, Zhao J, Zhang L, Zhao Y, Zhang Y, Cheng L, Wang D, Liu C and Zhang M, Fan K and Zhang M: A cascade nanoreactor for enhancing sonodynamic therapy on colorectal cancer via synergistic ROS augment and autophagy blockage. Nano Today. 49:1017982023. View Article : Google Scholar

198 

Wang G, He X and Wang Q: Intratumoral bacteria are an important 'accomplice' in tumor development and metastasis. Biochim Biophys Acta Rev Cancer. 1878:1888462023. View Article : Google Scholar

199 

Wang X, Chen Q, Zhu Y, Wang K, Chang Y, Wu X, Bao W, Cao T, Chen H, Zhang Y and Qin H: Destroying pathogen-tumor symbionts synergizing with catalytic therapy of colorectal cancer by biomimetic protein-supported single-atom nanozyme. Signal Transduct Target Ther. 8:2772023. View Article : Google Scholar : PubMed/NCBI

200 

Cao F, Jin L, Zhang C, Gao Y, Qian Z, Wen H, Yang S, Ye Z, Hong L, Yang H, et al: engineering clinically relevant probiotics with switchable 'nano-promoter' and 'nano-effector' for precision tumor therapy. Adv Mater. 36:e23042572023. View Article : Google Scholar

201 

Cabrero-Martín A, Santiago S, Serafín V, Pedrero M, Montero-Calle A, Pingarrón JM, Barderas R and Campuzano S: Multifunctional cerium nanolabels in electrochemical immunosensing with improved robustness and performance: Determination of TIM-1 in colorectal cancer scenarios as a case study. Mikrochim Acta. 192:2432025. View Article : Google Scholar : PubMed/NCBI

202 

Liu X, Fang Y, Liu J, Chen X, Teng F and Li C: Nanozyme-based pump-free microfluidic chip for colorectal cancer diagnosis via circulating cancer stem cell detection. ACS Sens. 9:5090–5098. 2024. View Article : Google Scholar : PubMed/NCBI

203 

Bhattacharjee R, Tanaka S, Moriam S, Masud MK, Lin J, Alshehri SM, Ahamad T, Salunkhe RR, Nguyen NT, Yamauchi Y, et al: Porous nanozymes: The peroxidase-mimetic activity of mesoporous iron oxide for the colorimetric and electrochemical detection of global DNA methylation. J Mater Chem B. 6:4783–4791. 2018. View Article : Google Scholar : PubMed/NCBI

204 

Lee MS, Menter DG and Kopetz S: Right versus left colon cancer biology: Integrating the consensus molecular subtypes. J Natl Compr Canc Netw. 15:411–419. 2017. View Article : Google Scholar : PubMed/NCBI

205 

Marisa L, de Reyniès A, Duval A, Selves J, Gaub MP, Vescovo L, Etienne-Grimaldi MC, Schiappa R, Guenot D, Ayadi M, et al: Gene expression classification of colon cancer into molecular subtypes: Characterization, validation, and prognostic value. PLoS Med. 10:e10014532013. View Article : Google Scholar : PubMed/NCBI

206 

Hao Y, Liu T, Zhou H, Xu R, Li K, Chen M and Chen Y: Oxygen-supplying ROS-responsive prodrug for synergistic chemotherapy and photodynamic therapy of colon cancer. Front Pharmacol. 15:13255442024. View Article : Google Scholar : PubMed/NCBI

207 

Kim S, Sundaram A, Mathew AP, Hareshkumar VS, Mohapatra A, Thomas RG, Bui TTM, Moon K, Kweon S, Park IK and Jeong YY: In situ hypoxia modulating nano-catalase for amplifying DNA damage in radiation resistive colon tumors. Biomater Sci. 11:6177–6192. 2023. View Article : Google Scholar : PubMed/NCBI

208 

Sun L, Gao W, Liu J, Wang J, Li L, Yu H and Xu ZP: O2-supplying nanozymes alleviate hypoxia and deplete lactate to eliminate tumors and activate antitumor immunity. ACS Appl Mater Interfaces. 14:56644–56657. 2022. View Article : Google Scholar : PubMed/NCBI

209 

Zhang C, Chen L, Bai Q, Wang L, Li S, Sui N, Yang D and Zhu Z: Nonmetal graphdiyne nanozyme-based ferroptosis-apoptosis strategy for colon cancer therapy. ACS Appl Mater Interfaces. 14:27720–27732. 2022. View Article : Google Scholar : PubMed/NCBI

210 

Du J, Liu J, Zhao Z, Dai J, Li K and Lin Y: Nonmetallic N/C nanozyme performs continuous consumption of glu for inhibition of colorectal cancer cells. ACS Appl Bio Mater. 6:267–276. 2023. View Article : Google Scholar

211 

Jin Y, Zhang J, Xu Y, Yi K, Li F, Zhou H, Wang H, Chan HF, Lao YH, Lv S, et al: Stem cell-derived hepatocyte therapy using versatile biomimetic nanozyme incorporated nanofiber-reinforced decellularized extracellular matrix hydrogels for the treatment of acute liver failure. Bioact Mater. 28:112–131. 2023.PubMed/NCBI

212 

Sahu A, Jeon J, Lee MS, Yang HS and Tae G: nanozyme impregnated mesenchymal stem cells for hepatic ischemia-reperfusion injury alleviation. ACS Appl Mater Interfaces. 13:25649–25662. 2021. View Article : Google Scholar : PubMed/NCBI

213 

Wang Z, Liu H, Yang SH, Wang T, Liu C and Cao YC: Nanoparticle-based artificial RNA silencing machinery for antiviral therapy. Proc Natl Acad Sci USA. 109:12387–12392. 2012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2025
Volume 56 Issue 5

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Xie D, Xie L, Fang C, Du Z, Cao Z, Su C and Huo Y: New advances of nanozymes for the diagnosis and treatment of digestive system diseases (Review). Int J Mol Med 56: 176, 2025.
APA
Xie, D., Xie, L., Fang, C., Du, Z., Cao, Z., Su, C., & Huo, Y. (2025). New advances of nanozymes for the diagnosis and treatment of digestive system diseases (Review). International Journal of Molecular Medicine, 56, 176. https://doi.org/10.3892/ijmm.2025.5617
MLA
Xie, D., Xie, L., Fang, C., Du, Z., Cao, Z., Su, C., Huo, Y."New advances of nanozymes for the diagnosis and treatment of digestive system diseases (Review)". International Journal of Molecular Medicine 56.5 (2025): 176.
Chicago
Xie, D., Xie, L., Fang, C., Du, Z., Cao, Z., Su, C., Huo, Y."New advances of nanozymes for the diagnosis and treatment of digestive system diseases (Review)". International Journal of Molecular Medicine 56, no. 5 (2025): 176. https://doi.org/10.3892/ijmm.2025.5617