Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (112)

Search Parameters:
Keywords = intestinal metaplasia

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
2 pages, 418 KB  
Correction
Correction: Qiu et al. P53 Deficiency Accelerates Esophageal Epithelium Intestinal Metaplasia Malignancy. Biomedicines 2023, 11, 882
by Quanpeng Qiu, Gang Guo, Xiaolong Guo, Xiake Hu, Tianyu Yu, Gaixia Liu, Haowei Zhang, Yinnan Chen and Junjun She
Biomedicines 2025, 13(8), 1978; https://doi.org/10.3390/biomedicines13081978 - 14 Aug 2025
Viewed by 166
Abstract
In the original publication [...] Full article
(This article belongs to the Section Cancer Biology and Oncology)
Show Figures

Figure 6

4 pages, 454 KB  
Interesting Images
Texture and Color Enhancement Imaging-Assisted Endocytoscopy Improves Characterization of Gastric Precancerous Conditions: A Set of Interesting Comparative Images
by Riccardo Vasapolli, Johannes Raphael Westphal and Christian Schulz
Diagnostics 2025, 15(15), 1925; https://doi.org/10.3390/diagnostics15151925 - 31 Jul 2025
Viewed by 307
Abstract
Chronic atrophic gastritis and intestinal metaplasia (IM) are gastric precancerous conditions (GPCs) associated with an increased risk of gastric cancer. Early detection and accurate characterization of GPC are therefore crucial for risk stratification and the implementation of preventive strategies. In the absence of [...] Read more.
Chronic atrophic gastritis and intestinal metaplasia (IM) are gastric precancerous conditions (GPCs) associated with an increased risk of gastric cancer. Early detection and accurate characterization of GPC are therefore crucial for risk stratification and the implementation of preventive strategies. In the absence of clear mucosal changes observed through white-light imaging (WLI) or virtual chromoendoscopy, endocytoscopy can help unveil the presence of GPC by enabling in vivo assessment of nuclear and cellular structures at ultra-high magnification. Endocytoscopy is typically performed using WLI following dye-based staining of the mucosa. In this case, we demonstrate that combining endocytoscopy with the texture and color enhancement imaging (TXI) mode substantially improves the assessment of the gastric mucosa. In a 61-year-old man undergoing esophagogastroduodenoscopy, WLI showed multifocal erythema in the stomach, without clearly visible lesions on either WLI or narrow-band imaging. Conventional endocytoscopy revealed multiple small spots of IM with characteristic changes in glandular structures, which were even more evident when using the TXI mode. Histological analysis of targeted biopsies confirmed small foci of IM in both the antrum and corpus. The patient was enrolled in a surveillance program because of his clinical background. The combination of endocytoscopy with the TXI mode significantly enhances the delineation of mucosal and cellular architecture, supporting a more accurate optical diagnosis. Full article
(This article belongs to the Section Medical Imaging and Theranostics)
Show Figures

Figure 1

13 pages, 1157 KB  
Review
Precision Care in Screening, Surveillance, and Overall Management of Barrett’s Esophagus
by Yeshaswini Reddy, Madhav Desai, Bernadette Tumaliuan and Nirav Thosani
J. Pers. Med. 2025, 15(8), 327; https://doi.org/10.3390/jpm15080327 - 22 Jul 2025
Viewed by 707
Abstract
Barrett’s esophagus (BE), a metaplastic transformation of an esophageal squamous epithelium into an intestinal-type columnar epithelium, is the primary precursor to esophageal adenocarcinoma (EAC). Traditional management strategies have relied heavily on selective screening, tailored surveillance intervals, and early dysplasia detection and treatment algorithms. [...] Read more.
Barrett’s esophagus (BE), a metaplastic transformation of an esophageal squamous epithelium into an intestinal-type columnar epithelium, is the primary precursor to esophageal adenocarcinoma (EAC). Traditional management strategies have relied heavily on selective screening, tailored surveillance intervals, and early dysplasia detection and treatment algorithms. However, the heterogeneity in progression risk among BE patients necessitates a more nuanced, personalized approach involving precision care, tailoring decisions to individual patient characteristics, promises to enhance outcomes in BE through more targeted screening, personalized surveillance intervals, and risk-based therapeutic strategies. This review explores the current landscape and emerging trends in precision medicine for Barrett’s esophagus, highlighting genomic markers, digital pathology, and AI-driven models as tools to transform how we approach this complex disease and prevent progression to EAC. Full article
(This article belongs to the Special Issue Clinical Updates on Personalized Upper Gastrointestinal Endoscopy)
Show Figures

Figure 1

15 pages, 2646 KB  
Article
Radiation Quality-Dependent Progressive Increase in Oxidative DNA Damage and Intestinal Tumorigenesis in Apc1638N/+ Mice
by Kamendra Kumar, Santosh Kumar, Jerry Angdisen, Kamal Datta, Albert J. Fornace and Shubhankar Suman
Curr. Oncol. 2025, 32(7), 382; https://doi.org/10.3390/curroncol32070382 - 1 Jul 2025
Viewed by 491
Abstract
Exposure to high-linear energy transfer (LET) heavy ions, such as 28Si, poses a significant cancer risk for astronauts. While previous studies have linked high-LET radiation exposure to persistent oxidative stress and dysregulated stress responses in intestinal crypt cells with an increased risk [...] Read more.
Exposure to high-linear energy transfer (LET) heavy ions, such as 28Si, poses a significant cancer risk for astronauts. While previous studies have linked high-LET radiation exposure to persistent oxidative stress and dysregulated stress responses in intestinal crypt cells with an increased risk of tumorigenesis, the relationship between IR-induced oxidative DNA damage and intestinal cancer risk remains incompletely understood. Here, we investigated the time-dependent effects of 28Si-ion radiation on intestinal tumorigenesis and oxidative DNA damage in Apc1638N/+ mice, a model for human intestinal cancer predisposition. Male Apc1638N/+ mice were exposed to 10 cGy of either γ-rays (low-LET) or 28Si-ions (high-LET), and intestinal tumor burden was assessed at 60 and 150 days post-irradiation. While both radiation groups showed modest, non-significant tumor increases at 60 days, 28Si-irradiated mice exhibited an approximately 2.5-fold increase in tumor incidence by 150 days, with a higher incidence of invasive carcinomas compared to γ and sham groups. Serum 8-OxodG levels, a marker of systemic oxidative stress, were significantly elevated in the 28Si-ion group, correlating with increased intestinal 8-OxodG staining. Additionally, assessment of the proliferation marker Cyclin D1 and metaplasia marker Guanylyl Cyclase C (GUCY2C) also revealed significant crypt cell hyperproliferation accompanied by increased metaplasia in 28Si-exposed mouse intestines. Positive correlations between serum 8-OxodG and tumor-associated endpoints provide compelling evidence that exposure to 28Si-ions induces progressive intestinal tumorigenesis through sustained oxidative DNA damage, crypt cell hyperproliferation, and metaplastic transformation. This study provides evidence in support of the radiation quality-dependent progressive increase in systemic and intestinal levels of 8-OxodG during intestinal carcinogenesis. Moreover, the progressive increase in oxidative DNA damage and simultaneous increase in oncogenic events after 28Si exposure also suggest that non-targeted effects might be a significant player in space radiation-induced intestinal cancer development. The correlation between serum 8-OxodG and oncogenic endpoints supports its potential utility as a predictive biomarker of high-LET IR-induced intestinal carcinogenesis, with implications for astronaut health risk monitoring during long-duration space missions. Full article
(This article belongs to the Section Gastrointestinal Oncology)
Show Figures

Graphical abstract

23 pages, 7504 KB  
Article
Development and Validation of the Early Gastric Carcinoma Prediction Model in Post-Eradication Patients with Intestinal Metaplasia
by Wulian Lin, Guanpo Zhang, Hong Chen, Weidong Huang, Guilin Xu, Yunmeng Zheng, Chao Gao, Jin Zheng, Dazhou Li and Wen Wang
Cancers 2025, 17(13), 2158; https://doi.org/10.3390/cancers17132158 - 26 Jun 2025
Viewed by 469
Abstract
Background: Gastric cancer (GC) remains a major global health challenge, with rising incidence among patients post-Helicobacter pylori (H. pylori) eradication, particularly those with persistent intestinal metaplasia (IM). Current risk stratification tools are limited in this high-risk population. Aim: [...] Read more.
Background: Gastric cancer (GC) remains a major global health challenge, with rising incidence among patients post-Helicobacter pylori (H. pylori) eradication, particularly those with persistent intestinal metaplasia (IM). Current risk stratification tools are limited in this high-risk population. Aim: To develop, validate, and externally test a machine learning-based prediction model—termed the Early Gastric Cancer Model (EGCM)—for identifying early gastric cancer (EGC) risk in H. pylori-eradicated patients with IM, and to implement it as a web-based clinical tool. Methods: This retrospective, dual-center study enrolled 214 H. pylori-eradicated patients with histologically confirmed IM from 900 Hospital and Fujian Provincial People’s Hospital. The dataset was split into a training cohort (70%) and an internal validation cohort (30%), with an external test cohort from the second center. A total of 21 machine learning algorithms were screened using cross-validation and hyperparameter optimization. Boruta and SHAP analyses were employed for feature selection, and the final EGCM was constructed using the top five predictors: atrophy range, xanthoma, map-like redness (MLR), MLR range, and age. Model performance was evaluated via ROC curves, precision–recall curves, calibration plots, and decision curve analysis (DCA), and compared against conventional inflammatory biomarkers such as NLR and PLR. Results: The CatBoost algorithm demonstrated the best overall performance, achieving an AUC of 0.743 (95% CI: 0.70–0.80) in internal validation and 0.905 in the external test set. The EGCM exhibited superior discrimination compared to individual inflammatory markers (p < 0.01). Calibration analysis confirmed strong agreement between predicted and observed outcomes. DCA showed the EGCM yielded greater net clinical benefit. A web calculator was developed to facilitate clinical application. Conclusions: The EGCM is a validated, interpretable, and practical tool for stratifying EGC risk in H. pylori-eradicated IM patients across multiple centers. Its integration into clinical practice could improve surveillance precision and early cancer detection. Full article
(This article belongs to the Section Cancer Causes, Screening and Diagnosis)
Show Figures

Figure 1

18 pages, 2773 KB  
Article
ViSwNeXtNet Deep Patch-Wise Ensemble of Vision Transformers and ConvNeXt for Robust Binary Histopathology Classification
by Özgen Arslan Solmaz and Burak Tasci
Diagnostics 2025, 15(12), 1507; https://doi.org/10.3390/diagnostics15121507 - 13 Jun 2025
Viewed by 786
Abstract
Background: Intestinal metaplasia (IM) is a precancerous gastric condition that requires accurate histopathological diagnosis to enable early intervention and cancer prevention. Traditional evaluation of H&E-stained tissue slides can be labor-intensive and prone to interobserver variability. Recent advances in deep learning, particularly transformer-based models, [...] Read more.
Background: Intestinal metaplasia (IM) is a precancerous gastric condition that requires accurate histopathological diagnosis to enable early intervention and cancer prevention. Traditional evaluation of H&E-stained tissue slides can be labor-intensive and prone to interobserver variability. Recent advances in deep learning, particularly transformer-based models, offer promising tools for improving diagnostic accuracy. Methods: We propose ViSwNeXtNet, a novel patch-wise ensemble framework that integrates three transformer-based architectures—ConvNeXt-Tiny, Swin-Tiny, and ViT-Base—for deep feature extraction. Features from each model (12,288 per model) were concatenated into a 36,864-dimensional vector and refined using iterative neighborhood component analysis (INCA) to select the most discriminative 565 features. A quadratic SVM classifier was trained using these selected features. The model was evaluated on two datasets: (1) a custom-collected dataset consisting of 516 intestinal metaplasia cases and 521 control cases, and (2) the public GasHisSDB dataset, which includes 20,160 normal and 13,124 abnormal H&E-stained image patches of size 160 × 160 pixels. Results: On the collected dataset, the proposed method achieved 94.41% accuracy, 94.63% sensitivity, and 94.40% F1 score. On the GasHisSDB dataset, it reached 99.20% accuracy, 99.39% sensitivity, and 99.16% F1 score, outperforming individual backbone models and demonstrating strong generalizability across datasets. Conclusions: ViSwNeXtNet successfully combines local, regional, and global representations of tissue structure through an ensemble of transformer-based models. The addition of INCA-based feature selection significantly enhances classification performance while reducing dimensionality. These findings suggest the method’s potential for integration into clinical pathology workflows. Future work will focus on multiclass classification, multicenter validation, and integration of explainable AI techniques. Full article
Show Figures

Figure 1

22 pages, 16693 KB  
Article
The Therapeutic Potential of Bombyx Batryticatus for Chronic Atrophic Gastritis Precancerous Lesions via the PI3K/AKT/mTOR Pathway Based on Network Pharmacology of Blood-Entering Components
by Xiaojie Wang, Miaomiao Chang, Kun Feng, Qingyue Wang, Bowen Li and Weijuan Gao
Pharmaceuticals 2025, 18(6), 791; https://doi.org/10.3390/ph18060791 - 25 May 2025
Viewed by 869
Abstract
Background: Chronic atrophic gastritis precancerous lesions (PL-CAG) are characterized by the atrophy of gastric mucosal glands, often accompanied by intestinal metaplasia or dysplasia. Timely intervention and treatment can effectively reverse its malignant progression and prevent the onset of gastric cancer. Bombyx Batryticatus (BB) [...] Read more.
Background: Chronic atrophic gastritis precancerous lesions (PL-CAG) are characterized by the atrophy of gastric mucosal glands, often accompanied by intestinal metaplasia or dysplasia. Timely intervention and treatment can effectively reverse its malignant progression and prevent the onset of gastric cancer. Bombyx Batryticatus (BB) exhibits a range of pharmacological effects, including anticoagulation, antiepileptic properties, anticancer activity, and antibacterial effects. However, the pharmacological basis and mechanisms underlying BB’s efficacy in treating PL-CAG remain unclear. Methods: A three-factor modeling approach was implemented to develop a rat PL-CAG model, while the MNNG-induced PLGC (precancerous lesions of gastric cancer) cell model was served as a cell PL-CAG model. UPLC-QE-Orbitrap-MS/MS (Ultra performance liquid chromatography-quadrupole-electrostatic field orbital trap high-resolution mass spectrometry) was utilized to perform an in-depth analysis of the components in the plasma extract of BB. Leveraging network pharmacology, molecular docking analyses, and experimental validation, we initially elucidated the potential mechanisms through which BB mediates its therapeutic effects on PL-CAG at both in vivo and in vitro levels. Results: Prototype compounds of 42 blood-entering components were identified by UPLC-QE-Orbitrap-MS/MS analysis. Network pharmacology analysis and molecular docking studies indicate that the core targets are primarily enriched in the PI3K-Akt signaling pathway, and the key components, including Nepitrin, Quercetin 3-O-neohesperidoside, Rutin, and others, exhibited stable docking conformations with the first eleven pivotal targets. Both in vivo and in vitro experiments validated that BB may effectively treat PL-CAG via modulation of the PI3K-Akt signaling pathway. Conclusions: The therapeutic efficacy of BB in the management of PL-CAG may be achieved through the synergistic interaction of multiple components and targets, which may be more closely related to the inhibition of the PI3K/AKT signaling pathway. This approach will establish a solid experimental foundation and provide essential data for the clinical application of BB in treating PL-CAG, while also facilitating further research initiatives. Full article
(This article belongs to the Section Natural Products)
Show Figures

Figure 1

15 pages, 6140 KB  
Article
Spatial Study of TLR4, TLR5 and TLR9 in Gastric Premalignant Lesions Before and After Helicobacter pylori Eradication
by Franz Villarroel-Espíndola, Leyla Jaupi, Joaquín Reyes, Carlos Barrientos, Celia Podestá, Carolina Selman, Carolina Bizama, Alejandro Corvalan, Roxana Gonzalez-Stegmaier, Sergio Jara-Rosales and Pia Bascur
Int. J. Mol. Sci. 2025, 26(9), 4059; https://doi.org/10.3390/ijms26094059 - 25 Apr 2025
Viewed by 804
Abstract
The histological changes in the gastric epithelium are crucial in the progression from premalignant to neoplastic lesions. TLR4, TLR5 and TLR9 have been localized in the gastric epithelium and studied separately using conventional histological techniques without a focus on the protein or cell [...] Read more.
The histological changes in the gastric epithelium are crucial in the progression from premalignant to neoplastic lesions. TLR4, TLR5 and TLR9 have been localized in the gastric epithelium and studied separately using conventional histological techniques without a focus on the protein or cell interactions within the microenvironment. Therefore, we developed a multiplex immunohistochemistry/immunofluorescence (mIHC/IF) technology for the simultaneous detection of TLR4, TLR5 and TLR9 on a single tissue section of human gastric biopsies from 10 paired cases collected in two independent visits, and its correlation with the OLGA/OLGIM scoring and H. pylori status after eradication. The results confirmed that mIHC/IF is useful for simultaneously interrogating six biomarkers and demonstrated that TLR4 and TLR9 are significantly associated with H. pylori infection. However, only TLR9 is positively related to the presence of intestinal metaplasia. TLR5 was mainly present in goblet cells (TFF3+) but did not show any significant association with H. pylori or the presence of intestinal metaplasia. Our results suggest that a more comprehensive strategy to interrogate the tissue microenvironment in premalignant lesions may improve the interpretation of the earned risk of gastric cancer in patients with chronic gastritis and evidence of failure in H. pylori eradication. Full article
Show Figures

Graphical abstract

19 pages, 1289 KB  
Review
Molecular Alterations in Gastric Intestinal Metaplasia Shed Light on Alteration of Methionine Metabolism: Insight into New Diagnostic and Treatment Approaches
by Nigatu Tadesse Gebrehiwot, Ying Liu, Juan Li and Hong-Min Liu
Biomedicines 2025, 13(4), 964; https://doi.org/10.3390/biomedicines13040964 - 15 Apr 2025
Viewed by 1270
Abstract
Gastric intestinal metaplasia (GIM) is a precancerous lesion and the key risk factor in the development of gastric cancer (GC), but early detection and treatment remain challenging. The traditional endoscopic diagnosis of metaplastic lesions is complicated by an increased rate of inappropriateness and [...] Read more.
Gastric intestinal metaplasia (GIM) is a precancerous lesion and the key risk factor in the development of gastric cancer (GC), but early detection and treatment remain challenging. The traditional endoscopic diagnosis of metaplastic lesions is complicated by an increased rate of inappropriateness and false negativity. Although early interventions with H. pylori eradication, as well as endoscopic therapy results, were promising, there is still a significant unmet need to control GIM progression and recurrences. Molecular alterations, such as an increased DNA methylation index, have been identified as a crucial factor in the downregulation of tumor suppressor genes, such as the caudal-type homeobox (CDX2) gene, which regulates epithelial cell proliferation and GIM progression and is associated with treatment failure. CDX2 is downregulated by promoter hypermethylation in the colonic-type epithelium, in which the methylation was correlated with reduced intake of dietary folate sources. Tumor cells alter to dietary methionine sources in the biosynthesis of S-Adenosylmethionine, a universal methyl donor for transmethylation, under the conditions of limited folate and B12 availability. The gut microbiota also exhibited a shift in microbial composition, which could influence the host’s dietary methionine metabolism. Meanwhile, activated oncogenic signaling via the PI3K/Akt/mTORC1/c-MYC pathway could promotes rewiring dietary methionine and cellular proliferation. Tumor methionine dependence is a metabolic phenotype that could be helpful in predictive screening of tumorigenesis and as a target for preventive therapy to enhance precision oncology. This review aimed to discuss the molecular alterations in GIM to shed light on the alteration of methionine metabolism, with insight into new diagnostic and treatment approaches and future research directions. Full article
(This article belongs to the Special Issue Feature Reviews in Gastrointestinal Diseases)
Show Figures

Graphical abstract

15 pages, 1819 KB  
Article
Methylated Reprimo Cell-Free DNA as a Non-Invasive Biomarker for Gastric Cancer
by María José Maturana, Oslando Padilla, Pablo M. Santoro, Maria Alejandra Alarcón, Wilda Olivares, Alejandro Blanco, Ricardo Armisen, Marcelo Garrido, Edmundo Aravena, Carlos Barrientos, Alfonso Calvo-Belmar and Alejandro H. Corvalán
Int. J. Mol. Sci. 2025, 26(7), 3333; https://doi.org/10.3390/ijms26073333 - 3 Apr 2025
Viewed by 1114
Abstract
Restrictions resulting from the COVID-19 pandemic abruptly reversed the slow decline of the diagnosis and mortality rates of gastric cancer (GC). This scenario highlights the importance of developing cost-effective methods for mass screening and evaluation of treatment response. In this study, we evaluated [...] Read more.
Restrictions resulting from the COVID-19 pandemic abruptly reversed the slow decline of the diagnosis and mortality rates of gastric cancer (GC). This scenario highlights the importance of developing cost-effective methods for mass screening and evaluation of treatment response. In this study, we evaluated a non-invasive method based on the circulating methylated cell-free DNA (cfDNA) of Reprimo (RPRM), a tumor suppressor gene associated with the development of GC. Methylated RPRM cfDNA was analyzed in three de-identified cohorts: Cohort 1 comprised 81 participants with GC and 137 healthy donors (HDs); Cohort 2 comprised 27 participants with GC undergoing gastrectomy and/or chemotherapy analyzed at the beginning and after three months of treatment; and Cohort 3 comprised 1105 population-based participants in a secondary prevention program who underwent esophagogastroduodenal (EGD) endoscopy. This cohort includes 180 normal participants, 845 participants with premalignant conditions (692 with chronic atrophic gastritis [AG] and 153 with gastric intestinal metaplasia/low-grade dysplasia [GIM/LGD]), 21 with high-grade dysplasia/early GC [HGD/eGC], and 59 with advanced GC [aGC]). A nested case-control substudy was performed using a combination of methylated RPRM cfDNA and pepsinogens (PG)-I/II ratio. The dense CpG island of the promoter region of the RPRM gene was bisulfite sequenced and analyzed to develop a fluorescence-based real-time PCR assay (MethyLight). This assay allows the determination of the absolute number of copies of methylated RPRM cfDNA. A targeted sequence of PCR amplicon products confirmed the gastric origin of the plasma-isolated samples. In Cohort 1, the mean value of GCs (32,240.00 copies/mL) was higher than that of the HD controls (139.00 copies/mL) (p < 0.0001). After dividing this cohort into training–validation subcohorts, we identified an area under the curve of 0.764 (95% confidence interval (CI) = 0.683–0.845) in the training group. This resulted in a cut-off value of 87.37 copies/mL (sensitivity 70.0% and specificity 80.2%). The validation subcohort predicted a sensitivity of 66.67% and a specificity of 83.33%. In Cohort 2 (monitoring treatment response), RPRM levels significantly decreased in responders (p = 0.0042) compared to non-responders. In Cohort 3 (population-based participants), 18.9% %, 24.1%, 30.7%, 47.0%, and 71.2% of normal, AG, GIM/LGD, HGD/eGC, and aGC participants tested positive for methylated RPRM cfDNA, respectively. Overall sensitivity and specificity in distinguishing normal/premalignant conditions vs. GC were 65.0% (95% CI 53.52% to 75.33%) and 75.9% (95% CI 73.16% to 78.49%), respectively, with an accuracy of 75.11% (95% CI 72.45% to 77.64%). Logistic regression analyses revealed an OR of 1.85 (95% CI 1.11–3.07, p = 0.02) and an odds ratio (OR) of 3.9 (95% CI 1.53–9.93, p = 0.004) for the risk of developing GIM/LGD and HGD/eGC, respectively. The combined methylated RPRM cfDNA and PG-I/II ratio reached a sensitivity of 78.9% (95% CI 54.43% to 93.95%) and specificity of 63.04% (95% CI 52.34% to 72.88%) for detecting HGD/eGC vs. three to six age- and sex-matched participants with premalignant conditions. Our results demonstrate that methylated RPRM cfDNA should be considered a direct biomarker for the non-invasive detection of GC and a predictive biomarker for treatment response. Full article
(This article belongs to the Section Molecular Oncology)
Show Figures

Figure 1

20 pages, 4911 KB  
Article
Therapeutic Potential of Qilianxiaopi Formula: Targeting ADAM17-Mediated Chronic Inflammation in Atrophic Gastritis
by Sijing Du, Tianxiang Wang, Zhiqiang Li, Ting Li, Zelong Miao, Yuling Chen, Songbiao Zhu, Wei Wei and Haiteng Deng
Pharmaceuticals 2025, 18(3), 435; https://doi.org/10.3390/ph18030435 - 19 Mar 2025
Cited by 1 | Viewed by 1140
Abstract
Background: Gastric cancer (GC) is a leading cause of mortality worldwide, particularly in China. Chronic atrophic gastritis (CAG) and intestinal metaplasia (IM) are recognized as precancerous conditions contributing to GC development. Qilianxiaopi formula (QLXP), a traditional Chinese medicine (TCM), has demonstrated significant [...] Read more.
Background: Gastric cancer (GC) is a leading cause of mortality worldwide, particularly in China. Chronic atrophic gastritis (CAG) and intestinal metaplasia (IM) are recognized as precancerous conditions contributing to GC development. Qilianxiaopi formula (QLXP), a traditional Chinese medicine (TCM), has demonstrated significant therapeutic effects on CAG and IM; however, its underlying mechanisms remain poorly understood. Methods: This study utilized chromatography-mass spectrometry to identify the major compounds in QLXP. Network pharmacology was used to predict the associated targets of these components. Thermal proteome profiling (TPP) pinpointed the potential binding proteins of QLXP, which were validated by bioinformatic analyses. Bio-layer interferometry (BLI) was used to analyze the interactions between QLXP and its key target proteins, thereby determining their binding components. Molecular docking predicted the binding modes between the components and proteins. Results: ADAM17 was identified as a key binding protein for QLXP. Further investigation revealed that QLXP inhibits the enzymatic activity of ADAM17, thereby reducing the secretion of the pro-inflammatory cytokine TNF-α, contributing to the anti-inflammatory properties of QLXP. BLI confirmed direct and reversible binding interactions between QLXP and ADAM17. Narirutin, isolated from the ADAM17 binding fraction, displayed the highest affinity for QLXP. Conclusions: This study highlights ADAM17 as a key molecular target of QLXP and narirutin as its principal binding component. The integrated approach combining chromatography-mass spectrometry, network pharmacology, TPP, BLI, and molecular docking provides a robust framework for elucidating the mechanisms of action of TCM. Full article
(This article belongs to the Special Issue Therapeutic Potential of Natural Products in Internal Diseases)
Show Figures

Graphical abstract

11 pages, 1823 KB  
Article
Characteristics and Neoplastic Progression in Barrett’s Esophagus: A Large Population-Based Study from Iceland
by Ken Namikawa, Melkorka Sverrisdottir, Hilmar Freyr Fridgeirsson, Hjalti Dagur Hjaltason, Helgi Kristinn Sigmundsson, Jon Gunnlaugur Jonasson, Einar Stefan Bjornsson and Magnus Konradsson
Diagnostics 2025, 15(6), 684; https://doi.org/10.3390/diagnostics15060684 - 11 Mar 2025
Viewed by 1474
Abstract
Background: Barrett’s esophagus (BE) is a known precursor to esophageal adenocarcinoma (EAC). However, reports on incidence and progression-to-neoplasm rates have been very variable and conflicting. The aims of the study were to evaluate the characteristics of BE and its progression to neoplasm in [...] Read more.
Background: Barrett’s esophagus (BE) is a known precursor to esophageal adenocarcinoma (EAC). However, reports on incidence and progression-to-neoplasm rates have been very variable and conflicting. The aims of the study were to evaluate the characteristics of BE and its progression to neoplasm in a large homogeneous population. Methods: This was a retrospective population-based study with patients identified from 11 institutions through the databases in two centralized pathology laboratories. Demographics and relevant clinicopathological features were obtained from medical records among patients with a pathologically confirmed BE by the presence of intestinal metaplasia between 2003 and 2022. Results: A total of 1388 patients were identified with BE: 948 were men (69%); the median age at diagnosis was 62 years (IQR, 53–72). The ratio of long-segment BE to short-segment BE was significantly higher in patients ≥ 60 years (1.15, 284/248) than those ≤ 60 years (0.77, 205/265) (p = 0.0025). At BE diagnosis, 9.4% had neoplasms: LGD (n = 65), HGD (n = 16), and EAC (n = 49). Among 1258 non-dysplastic BE (NDBE) patients, 4.6% developed a neoplasm—LGD (n = 35), HGD (n = 8), and EAC (n = 15)—with a median observation-period of 5 years (IQR, 3–7). Overall, 160 cases with neoplasms were diagnosed in this BE cohort; 130 (74%) were present at initial BE diagnosis, and 58 (26%) progressed to neoplasms from NDBE. Conclusions: The ratio of long-segment BE was found to be significantly higher in patients ≥ 60 years. Around 9% of the patients were diagnosed as harboring a neoplasm concomitantly with BE, accounting for approximately 74% of all neoplasms. After a median follow-up of 5 years, about 5% of BE showed dysplastic or malignant progression. Full article
Show Figures

Figure 1

20 pages, 3177 KB  
Article
The Status of SOX2 Expression in Gastric Cancers with Induction of CDX2 Defines Groups with Different Genomic Landscapes
by Ioannis A. Voutsadakis
Genes 2025, 16(3), 279; https://doi.org/10.3390/genes16030279 - 26 Feb 2025
Viewed by 1030
Abstract
Background: Gastric adenocarcinoma is a highly lethal neoplasm with a short survival especially when metastatic. Few effective treatments are available for the control of the disease and palliation of patients with metastatic gastric cancer. Although progress has been made in the elucidation of [...] Read more.
Background: Gastric adenocarcinoma is a highly lethal neoplasm with a short survival especially when metastatic. Few effective treatments are available for the control of the disease and palliation of patients with metastatic gastric cancer. Although progress has been made in the elucidation of molecular pathways invoked in gastric carcinogenesis, this knowledge has not yet led to major breakthroughs, in contrast to several other types of cancer. The role of stem cell transcription factors SOX2 and CDX2 is of particular interest in the pathogenesis of gastric cancer. Methods: The cohort of gastric adenocarcinomas from The Cancer Genome Atlas (TCGA) was interrogated and two groups of gastric cancers, with CDX2 induction and SOX2 suppression on the one hand and with CDX2 induction and SOX2 maintained expression on the other hand were retained. The induction of expression of the two transcription factors was defined as a mRNA expression z score compared with normal samples above zero. The two groups were compared for clinical-pathologic and genomic differences. Results: Among gastric cancers with up-regulated CDX2 mRNA, cancers with suppressed SOX2 mRNA were slightly more numerous (55.9%) than those with a maintained SOX2 expression. The SOX2 suppressed group had a higher prevalence of MSI high cancers (30.9% versus 10%) and of cases with high tumor mutation burden (35% versus 12.4%) than cancers with a SOX2 maintained expression, which presented more frequently high Chromosomal Instability (CIN). The group with SOX2 suppression had higher rates of mutations in many gastric cancer-associated genes such as epigenetic modifiers ARID1A, KMT2D, KMT2C, and KMT2B, as well as higher rates of mutations in genes encoding for receptor tyrosine kinases ERBB4 and FGFR1. On the other hand, TP53 mutations and amplifications in MYC, ERBB2, and CCNE1 were more common in the group with a maintained expression of SOX2, approaching significance for MYC. Conclusions: Notable differences are present in the genomic landscape of CDX2-induced gastric cancer depending on the level of expression of SOX2 mRNA. Despite this, SOX2 mRNA expression levels were not prognostic. Full article
(This article belongs to the Section Human Genomics and Genetic Diseases)
Show Figures

Figure 1

17 pages, 2930 KB  
Article
Predictors of the Development of Gastric Cancer in Post-Helicobacter pylori-Eradication Patients Followed Up for More than 10 Years: A Histological, Serological, and Endoscopic Study
by Kazuhiro Mizukami, Masaaki Kodama, Yuka Hirashita, Masahide Fukuda, Sotaro Ozaka, Koshiro Tsutsumi, Ryota Sagami, Kensuke Fukuda, Ryo Ogawa and Kazunari Murakami
Cancers 2025, 17(3), 552; https://doi.org/10.3390/cancers17030552 - 6 Feb 2025
Cited by 2 | Viewed by 1403
Abstract
Background/Objectives: Although Helicobacter pylori (H. pylori) eradication therapy is important for preventing gastric cancer (GC), the occurrence of GC after H. pylori eradication remains a problem. In this study, the aim was to identify risk factors for GC after H. pylori [...] Read more.
Background/Objectives: Although Helicobacter pylori (H. pylori) eradication therapy is important for preventing gastric cancer (GC), the occurrence of GC after H. pylori eradication remains a problem. In this study, the aim was to identify risk factors for GC after H. pylori eradication by comparing long-term histological, endoscopic, and serological evaluations of patients with and without GC. Methods: Patients who underwent H. pylori eradication therapy at Oita University Hospital between June 1997 and August 2013 and were followed for at least 3 years with long-term endoscopy, histology, and serum biochemical tests were included, and the GC (215 cases) and non-GC (11 cases) groups were compared. Results: The GC group was older than the non-GC group at the time of eradication, had lower serum pepsinogen I/II levels, had severe endoscopic atrophic changes, had higher activity at the antrum, and inflammation and intestinal metaplasia (IM) at the corpus on updated Sydney system scoring. On long-term follow-up after eradication, the GC group had a wider range of endoscopic mucosal atrophy and a lower serum pepsinogen I/II ratio at any time point. Conclusions: Endoscopic mucosal atrophy and the serum pepsinogen I/II ratio are useful predictors of GC in patients post H. pylori eradication at any time point. Full article
Show Figures

Figure 1

14 pages, 2412 KB  
Article
Gastric Microbiota Associated with Gastric Precancerous Lesions in Helicobacter pylori-Negative Patients
by Han-Na Kim, Min-Jeong Kim, Jonathan P. Jacobs and Hyo-Joon Yang
Microorganisms 2025, 13(1), 81; https://doi.org/10.3390/microorganisms13010081 - 3 Jan 2025
Cited by 1 | Viewed by 1777
Abstract
Studies on the gastric microbiota associated with gastric precancerous lesions remain limited. This study aimed to profile the gastric mucosal microbiota in patients with Helicobacter pylori-negative precancerous lesions. Gastric mucosal samples were obtained from 67 H. pylori-negative patients, including those with [...] Read more.
Studies on the gastric microbiota associated with gastric precancerous lesions remain limited. This study aimed to profile the gastric mucosal microbiota in patients with Helicobacter pylori-negative precancerous lesions. Gastric mucosal samples were obtained from 67 H. pylori-negative patients, including those with chronic gastritis (CG), intestinal metaplasia (IM), and dysplasia. The V3–V4 region of the 16S rRNA gene was sequenced and analyzed. No significant difference was observed in the alpha or beta diversity of the gastric microbiota among the groups. However, a taxonomic analysis revealed a significant enrichment of Lautropia mirabilis and the depletion of Limosilactobacillus reuteri, Solobacxterium moorei, Haemophilus haemolyticus, and Duncaniella dubosii in the IM and dysplasia groups compared to those in the CG group. Prevotella jejuni and the genus Parvimonas were enriched in the IM group. A predictive functional analysis revealed enrichment of the ornithine degradation pathway in the IM and dysplasia groups, suggesting its role in persistent gastric mucosal inflammation associated with gastric precancerous lesions. The gastric microbiota associated with H. pylori-negative gastric precancerous lesions showed an increased abundance of oral microbes linked to gastric cancer and a reduction in anti-inflammatory bacteria. These alterations might contribute to chronic gastric mucosal inflammation, promoting carcinogenesis in the absence of H. pylori infection. Full article
(This article belongs to the Special Issue Correlations Between the Gastrointestinal Microbiome and Diseases)
Show Figures

Figure 1

Back to TopTop