BPG is committed to discovery and dissemination of knowledge
Review Open Access
Copyright ©The Author(s) 2026. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastrointest Oncol. Jan 15, 2026; 18(1): 114040
Published online Jan 15, 2026. doi: 10.4251/wjgo.v18.i1.114040
Tight junction proteins: Gatekeepers turned facilitators in the pathogenesis of gastric adenocarcinoma
Shobha Selvam, Balasubramaniyan Vairappan, Department of Biochemistry, Jawaharlal Institute of Postgraduate Medical Education and Research, Pondicherry 605006, India
ORCID number: Balasubramaniyan Vairappan (0000-0003-1708-4864).
Co-first authors: Shobha Selvam and Balasubramaniyan Vairappan.
Author contributions: Selvam S and Vairappan B contributed equally to this manuscript and are co-first authors. Selvam S and Vairappan B contributed to the manuscript writing and revisions.
Conflict-of-interest statement: All the authors report no relevant conflicts of interest for this article.
Open Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Balasubramaniyan Vairappan, PhD, Additional Professor, Department of Biochemistry, Jawaharlal Institute of Postgraduate Medical Education and Research, Dhanvantari Nagar, Pondicherry 605006, India. balasubramaniyan.v@jipmer.edu.in
Received: September 10, 2025
Revised: October 21, 2025
Accepted: November 24, 2025
Published online: January 15, 2026
Processing time: 124 Days and 8.6 Hours

Abstract

Gastric cancer (GC) is the fifth most prevalent malignancy worldwide and remains a leading cause of cancer-related mortality. Major risk factors for GC include Helicobacter pylori infection, increasing age, high dietary salt intake, and diets deficient in vegetables and fruits. Due to the often subtle and nonspecific early symptoms, coupled with the lack of routine screening programs, a significant proportion of GC cases are diagnosed at advanced stages. The etiology of GC is multifactorial, and diagnosis is confirmed histologically through endoscopic biopsy, followed by staging via computed tomography, positron emission tomography, staging laparoscopy, and endoscopic ultrasound. Treatment strategies typically involve a multidisciplinary approach including chemotherapy, surgical resection, radiotherapy, and emerging immunotherapeutic options. Despite advances in diagnostic and therapeutic modalities, the prognosis of advanced GC remains poor, with high rates of recurrence and metastasis. In recent years, increasing attention has been given to the role of tight junction (TJ) proteins in the pathogenesis and progression of GC. TJ proteins, critical components of epithelial barrier function, have been implicated in various stages of gastric carcinogenesis, from intestinal metaplasia to invasion and metastasis. Infection and inflammation, particularly due to Helicobacter pylori, disrupt TJ integrity, compromising the gastric mucosal barrier and facilitating neoplastic transformation. This review synthesizes current evidence from PubMed, EMBASE, Google Scholar, ScienceDirect, SpringerLink, and other reputable databases to provide a comprehensive overview of the involvement of TJ proteins in GC. By elucidating the molecular interplay between TJ dysregulation and gastric tumorigenesis, this work aims to highlight the potential of TJ proteins as novel diagnostic biomarkers and therapeutic targets in GC management.

Key Words: Claudins; Gastric cancer; Infection and inflammation; Occludin and zonula occludens; Tight junction

Core Tip: Tight junction (TJ) proteins play a pivotal role in preserving the integrity of the gastric epithelial barrier. Their dysregulation represents a critical early event linking chronic inflammation to gastric carcinogenesis. Inflammatory mediators induce TJ remodeling, thereby increasing mucosal permeability and promoting tumor initiation and progression. Emerging therapeutic strategies targeting TJ proteins either by pharmacologically restoring barrier integrity or modulating aberrant TJ signaling offer promising avenues to suppress inflammation, inhibit tumor growth and metastasis, and ultimately improve clinical outcomes in gastric cancer patients.



INTRODUCTION

Gastric cancer (GC) is the fifth most common malignancy and the third leading cause of cancer-related mortality worldwide, with a particularly high incidence in East Asia, where it accounts for approximately one in every twelve cancer-related deaths[1]. It is a highly heterogeneous disease, with over one million new cases estimated annually[2,3]. Globally, the lifetime risk of developing GC is approximately 1 in 54 for men and 1 in 126 for women, with men being twice as likely to be affected[2-4]. In the United States, the lifetime risk is about 1 in 95 for men and 1 in 154 for women. India falls into the low-incidence category but shows significant regional variation in disease occurrence[5]. According to GLOBOCAN 2018 data, East Asia, South America and Eastern Europe are the major hotspots for both the incidence and mortality of GC[6]. The disease often begins as precancerous changes in the gastric mucosa and may progress silently to advanced stages. GC presents with a wide range of symptoms, making early diagnosis difficult, and is often resistant to chemotherapy[2,3].

RISK FACTORS

Helicobacter pylori (H. pylori) infection is the most powerful risk factor for GC, promoting carcinogenesis through both direct and indirect mechanisms[2,3]. The cytotoxin-associated gene A protein, a virulence factor of H. pylori, interacts with host proteins that regulate cell growth, movement, and polarity. This interaction leads to morphological changes and increases the likelihood of epigenetic alterations that drive GC development[7]. Smoking is the second most significant risk factor for the onset of GC[2]. The toxins and carcinogens in cigarette fumes cause direct DNA damage, disrupt normal cell growth, and promote malignant transformation of gastric epithelial cells. Both former and current smoking contribute to carcinogenesis[8]. Dietary factors such as smoked and salted foods (pickled vegetables) also play a crucial role being the sources of nitrosamine (dietary carcinogen) that elevate GC risk[2]. Additionally, reactive oxygen species (ROS) generated by inflammation or poor diet cause DNA damage, leading to genetic mutations and cancer development[8]. Alcohol consumption is another contributing factor, its metabolite acetaldehyde, is a recognized human carcinogen that causes DNA damage by inhibiting DNA methylation and by interacting with retinoid metabolism[9]. It acts as a mucosal irritant, further increasing cancer risk[10]. A family history of GC, particularly in first-degree relatives and siblings, is associated with a higher risk, especially in individuals diagnosed before the age of 50[11]. Gastroesophageal reflux disease and obesity are specifically linked to cardia-type GC[12]. Radiation exposure has been associated with an increased risk of gastric lymphomas. Several other conditions are known to elevate the risk of GC, including blood group O, migrants from high- to low-incidence nations, chronic gastric ulcers, adenomatous gastric polyps, and prolonged exposure to H2-receptor blockers[3].

Gastric adenocarcinoma is the most common type of stomach cancer, originating from the glandular epithelium of the stomach lining. It typically arises in the mucosal layer and often begins as an ulcer or abnormal growth[13,14]. Gastric adenocarcinoma is histologically classified into two main subtypes: Intestinal type and diffuse type[15]. The diffuse type is characterized by a lack of cell cohesion, allowing individual cancer cells to infiltrate and thicken the stomach wall without forming a well-defined mass. This type tends to occur in younger patients, can involve any part of the stomach, and is associated with a poorer prognosis[13]. In contrast, the intestinal type features cohesive neoplastic cells that form gland-like tubular structures. These tumors are often ulcerative and typically develop in the antrum and along the lesser curvature of the stomach. Intestinal-type GC is frequently preceded by a well-defined precancerous sequence, including chronic gastritis, atrophy, intestinal metaplasia, and dysplasia[13].

DIAGNOSIS AND TREATMENT

The diagnosis of GC involves a comprehensive approach, beginning with a detailed clinical history and physical examination, followed by blood investigations and imaging techniques such as ultrasound, barium swallow, computed tomography scan and magnetic resonance imaging to invasive endoscopic biopsy. For accurate staging, advanced imaging modalities like positron emission tomography scans are utilized, followed by staging laparoscopy to assess the extent of disease spread. Early detection remains crucial for improving prognosis, therefore, individuals with risk factors or persistent symptoms should seek prompt evaluation by a gastroenterologist. An endoscopic gastric biopsy is definitive for diagnosing GC.

Although there are various treatment modalities like surgical (partial or total gastrectomy with or without lymph node dissection), chemotherapy, radiation therapy, and immunotherapy, the incidence of metastasis and resistance to therapy is still high. Despite the availability of these treatment modalities, metastatic disease and resistance to therapy remain significant challenges. As such, newer strategies like targeted drug therapy and monoclonal antibody-based treatments are being developed to improve therapeutic outcomes and reduce recurrence. In terms of prevention, dietary modifications play an important role. Increased consumption of fruits and vegetables has been consistently associated with a reduced risk of GC. Their anti-carcinogenic effects are largely attributed to the antioxidant properties of nutrients such as vitamin C and beta-carotene, which help neutralize free radicals and prevent DNA damage.

TIGHT JUNCTION PROTEINS

Tight junctions (TJs) are specialized protein complexes that seal the intercellular space between adjacent epithelial to endothelial cells, playing a critical role in maintaining selective permeability and cellular polarity[16,17]. They are primarily formed by interactions among proteins from three major families: Claudins, zonula occludens (ZO) proteins, and TJ-associated MARVEL proteins, including occludin[16-19]. The gastrointestinal (GI) epithelium provides a clear example of TJ structure and function. It is composed of a single layer of columnar epithelial cells, predominantly absorptive in nature, specialized for nutrient uptake from the intestinal lumen[17,19]. The TJ between these cells not only prevents the unregulated passage of ions, nutrients, and solutes through the paracellular space, but also act as molecular “fences” that maintain the distinction between the apical and basolateral domains of the plasma membrane, ensuring proper cell polarity and function[20,21]. Among TJ proteins, claudins serve as the structural backbone of TJ strands and are integral membrane proteins responsible for regulating paracellular permeability[22]. Under freeze-fracture electron microscopy, claudins appear as continuous, strand-like structures. In contrast, occludin and ZO-1 (a scaffolding protein) are more involved in the regulation, assembly, and signaling associated with TJ integrity. ZO-1 links transmembrane proteins like claudins and occludin to the actin cytoskeleton, facilitating TJ stability and signal transduction[23].

In the gastric epithelium, TJs are vital for preserving mucosal barrier integrity, limiting the translocation of digestive enzymes, microbial toxins, and other harmful substances into the systemic circulation[24]. The coordinated interaction among TJ components (claudins, occludin, ZO proteins, and TJ-associated MARVEL proteins) is essential not only for maintaining epithelial and endothelial barrier integrity but also for modulating intracellular signaling pathways that influence cell proliferation, differentiation, and immune responses[25,26]. Various risk factors contributing to the initiation of GC and progression to advanced metastatic cancer stage are shown in Figure 1.

Figure 1
Figure 1 Factors contributing to the initiation and progression of gastric adenocarcinoma and metastasis. Oxidative stress, free radical induced injury, and Helicobacter pylori infection can trigger chronic inflammation, which in turn causes persistent DNA damage and activates multiple oncogenic pathways that drive carcinogenesis. Moreover, chronic inflammation and ulcer formation can disrupt the balance of gut microbiota, leading to dysbiosis and increased intestinal permeability (‘leaky gut’). This dysregulated microenvironment further promotes epithelial mesenchymal transition, thereby facilitating gastric cancer progression and metastasis. H. pylori: Helicobacter pylori.
ZO

ZO-1 was the first TJ protein identified. It is a cytosolic peripheral membrane protein belonging to the membrane-associated guanylate kinase-like family, functioning as a PDZ domain-containing scaffold[18,27]. Along with ZO-2 and ZO-3, ZO-1 anchors TJs to the actin cytoskeleton and links transmembrane proteins such as claudins, occludin, and junctional adhesion molecule-A (JAM-A) to cytoskeletal and signaling networks[28]. ZO-1 contains three PDZ domains in its N-terminal region: PDZ1 binds claudins (with affinity modulated by claudin phosphorylation), PDZ2 facilitates dimerization, and PDZ3 interacts with JAM-A[18,27,28]. Additional interaction domains SH3, U5, GUK, and an actin-binding region enable binding to other scaffold proteins like afadin and α-catenin. The PDZ3-SH3-GUK-U6 module also drives phase separation and polymerization of ZO proteins during TJ assembly. Functionally, ZO-1 is essential for epithelial barrier integrity, coordinating the structural assembly of TJs and participating in signaling pathways that influence gene expression and epithelial permeability[18,27,28].

The simultaneous deletion of ZO-1 and ZO-2 leads to highly permeable cell monolayers and a complete loss of TJ strands, indicating their redundant roles in TJ stabilization. ZO-1 also mediates communication between TJ components and the actin cytoskeleton[19,29]. Its actin-binding region interacts weakly and transiently with F-actin; both excessively strong or absent interactions can disrupt barrier integrity and increase TJ permeability[30]. Afadin interacts with ZO-1, JAM-A, nectin, and actin, playing a crucial role in initiating TJ formation and cell polarization by linking ZO-1 and JAM-A at adherens junctions. Cingulin and paracingulin form coiled-coil homodimers with a globular head and share an N-terminal ZO-1 interaction motif[19]. Cingulin binds multiple TJ proteins, including JAM-A, ZO-1, and ZO-2, and interacts strongly with both actin and microtubules an interaction regulated by AMP-activated protein kinase phosphorylation[25]. Both cingulin and paracingulin contribute to TJ regulation by suppressing Rho family GTPase activity, thereby downregulating claudin 2 expression and inhibiting cell proliferation[19,28].

TJs are essential for maintaining barrier function, cell polarity, and intracellular signaling pathways[18,27]. Disruption of TJs can result in the loss of cell polarity and an abnormal influx of growth factors, which may stimulate tumorigenic epithelial cells through autocrine and paracrine mechanisms[27-31]. ZO-1 is typically highly expressed in normal mucosa, as well as in carcinoma and metaplastic epithelium. However, its expression is significantly reduced in undifferentiated-type gastric adenocarcinomas, suggesting a correlation between ZO-1 downregulation and tumor progression[27,31].

OCCLUDIN

Occludin is an integral membrane protein of approximately 60 kDa, located in the TJs of epithelial and endothelial cells. It contains four transmembrane domains and, unlike many other TJ proteins, does not exhibit multiple isotypes across species[32]. Occludin plays a key role in maintaining TJ barrier integrity and defense functions. Interestingly, epithelial cells lacking occludin still form well-organized TJ strands, indicating that occludin may play a supportive rather than primary structural role in TJ formation[33,34]. It contributes to maintaining cell polarity and preventing paracellular leakage. Beyond its structural functions, occludin is involved in signaling pathways that regulate TJ assembly and function, as well as in cellular processes such as proliferation, differentiation, and migration[33,34]. Alterations in occludin expression or localization have been associated with disorders involving barrier dysfunction, including inflammatory bowel disease and cancer metastasis. Additionally, occludin enhances intracellular adhesion; its homotypic interactions help organize atypical protein kinase C-PAR3 and PALS1-associated TJ complexes at the leading edge during epithelial migration[35].

CLAUDINS

Claudins are essential transmembrane proteins of TJs, each with four transmembrane domains. The claudin gene family is expressed in a tissue-specific manner, with 24 members identified to date. In normal gastric mucosa, claudin 18 is typically expressed, whereas claudin 4 is not. However, oligonucleotide microarray studies have shown that claudin 4 is upregulated in GC, particularly in the intestinal-type subtype[26]. Elevated claudin 4 expression has been consistently observed in gastric intestinal-type adenocarcinoma (as summarized in Table 1). H. pylori infection has been shown to increase paracellular permeability by disrupting occludin, claudin-4, and claudin-5. Additionally, the transcription factor caudal-related homeobox transcription factor 2 plays a key role in regulating intestinal claudin expression. Notably, reduced claudin 4 expression correlates with higher recurrence rates and poorer survival outcomes in GC patients. Functionally, claudins form the backbone of TJ strands and contribute to paracellular ion selectivity. Certain claudins, such as those in kidney epithelial cells, form channels that selectively permit the passage of ions such as Mg2+. Mutations in these claudins can impair ion reabsorption, resulting in excessive urinary Mg2+ loss.

Table 1 Dysregulated tight junction proteins and their mechanistic associations with gastric adenocarcinoma.
No.
TJ proteins
Expression level in gastric carcinoma
Functional effect
Ref.
1ZO↓ ZO-1 and ZO-2, upregulated or downregulatedLoss of epithelial features. Induce EMT. Promote tumor invasion, metastasis and resistance to apoptosis[31]
2OccludinsDecreased expressionWnt/β-catenin pathway activation. β-catenin pathway accumulation in cytoplasm, translocation to nucleus. Uncontrolled cell proliferation[31]
3Claudin 1IncreasedIncreased claudin-1 → TJ hyperfunction → alters selective permeability of epithelium. Wnt/β-catenin pathway activation → uncontrolled cell proliferation, promotes EMT[37]
4Claudin 2IncreasedIncreased intestinal permeability, activation of oncogenic signaling pathway, disruption of normal epithelial architecture, weakened cell-cell adhesion → metastasis[37]
5Claudin 3IncreasedActivation of inflammatory pathways (JAK/STAT, NF-κB) → immune invasion, promotes angiogenesis[19]
6Claudin 4DecreasedInvasiveness, metastasis and tumor aggressiveness, poor survival[31]
IncreasedWnt/β-catenin pathway activation, PI3K/Akt signaling → enhances cell survival by promoting apoptosis and promotes EMT through TGF-β signaling. Promotes tumor-supportive microenvironment[37]
7Claudin 5IncreasedSuppresses apoptosis of gastric cells → uncontrolled proliferation of tumor cells[38]
8Claudin 6Increased Induces MMP-2 activation through claudin-1 membrane expression → promotes cell migration and invasiveness[39]
9Claudin 7IncreasedEMT → tumor invasion, aggression and metastasis[19,40]
10Claudin 12IncreasedThrough the EMT, regulates apoptosis[41]
11Claudin 18DecreasedSignaling cascade related to chemokines and Wnt signaling pathway[42,43]
12Claudin 23DecreasedRecruits claudin 3, 4 and mediates through it[44,45]
ROLE OF CLAUDIN IN GC

Dysregulation of claudin expression is frequently observed in tumorigenesis and is closely linked to cancer progression[36]. Claudin 1 expression has been linked to increased invasiveness and metastasis in various cancers, including gastric carcinoma. In hepatocellular carcinoma, claudin 1 promotes epithelial mesenchymal transition (EMT) by upregulating the transcription factors Slug and Zeb[37]. Claudin 4 overexpression has also been found to reduce apoptosis, supporting its role in tumor survival. It is highly expressed in metaplastic epithelium and gastric intestinal-type adenocarcinoma, while showing minimal expression in normal gastric epithelium[37]. Claudin 5 is overexpressed in GC due to its ability to suppress the gastric cells leading to uncontrolled proliferation of cancer cells[38]. Additionally, claudins facilitate tumor cell invasion through the activation of matrix metalloproteinases (MMPs). Specifically, claudin 1 activates MMP-2 via the protein kinase C signaling pathway, and claudin 6 has been shown to trigger MMP-2 activation through its interaction with claudin 1. MMPs not only degrade the extracellular matrix but also modify cell-cell and cell-matrix interactions, thereby promoting EMT and tumor progression. In gastric adenocarcinoma, claudin 1 interacts with extracellular pro-MMP-2 through its extracellular loops, leading to MMP-2 activation via MMP-14. Furthermore, claudin 4 is upregulated during early stages of gastric tumorigenesis, a change strongly associated with DNA hypomethylation[39]. Claudins 3, 4, 5, 12, 18, and 23 are widely expressed throughout the stomach. In particular, claudin 3, claudin 4, and claudin 7 are frequently upregulated in GC, whereas other claudins may be downregulated[40,41]. Unlike esophageal epithelium, the gastric epithelium is adapted to withstand acidic conditions due to its tight TJ network. However, H. pylori infection disrupts this barrier by raising the pH through urease activity, which leads to decreased transepithelial resistance, internalization of occludin, and phosphorylation of the myosin light chain. H. pylori significantly alters both the pore and leak pathways of TJs, contributing to changes in claudin expression that are commonly seen in GC.

The gastric mucosa has more extensive and deeper TJ strands compared to the esophagus, conferring lower water and solute permeability and resistance to gastric acid. Disruptions in claudin expression compromise this defense and contribute to GC development. The stomach-specific claudin 18 is notably downregulated in atrophic gastritis and gastric carcinoma, and its loss is associated with poor prognosis. Claudin 18 plays a crucial role in maintaining cell lineage differentiation and the structural integrity of the gastric mucosa[42,43]. Claudin 23 is decreased in GC, it mediates its action by recruiting claudin 3 and 4[44,45].

INFLAMMATION AND GC

Inflammation represents the body’s natural defense response to injury, infection, or harmful stimuli, involving immune cells, blood vessels, and molecular mediators[46]. Acute inflammation protects the body by eliminating harmful agents and promoting tissue repair[46,47]. However, when inflammation becomes persistent or chronic, it contributes to various pathologies, including cancer. Pro-inflammatory cytokines such as tumor necrosis factor-alpha (TNF-α) and interleukins (ILs) can alter the expression and localization of TJ proteins, compromising the gastric epithelial barrier and increasing susceptibility to malignant transformation[47,48].

H. pylori infection is a well-known risk factor for GC[49]. The bacterium induces chronic gastric inflammation, leading to gastritis and progressive mucosal damage[50]. The persistent inflammation owing to H. pylori infection can disrupt the function and structure of TJs, leading to increased gastric permeability[51]. This allows harmful substances to penetrate the gastric mucosa, contributing to tissue damage and promoting carcinogenesis.

IL-1β

IL-1β is a potent pro-inflammatory cytokine markedly upregulated during H. pylori infection. It plays a central role in sustaining gastric mucosal inflammation and has been strongly associated with an increased risk of GC. IL-1β suppresses gastric acid secretion, creating a favorable environment for H. pylori persistence and chronic inflammation[52]. Beyond immune activation, IL-1β drives gastric carcinogenesis by promoting epithelial proliferation, angiogenesis, and tumor progression[53]. Mechanistically, IL-1β disrupts epithelial barrier integrity by activating the mitogen-activated protein kinase kinase-1-nuclear factor kappa B (NF-κB) signaling cascade[54]. NF-κB activation induces myosin light chain kinase expression, which alters cytoskeletal dynamics and TJ protein organization, ultimately leading to TJ disassembly and loss of barrier function[55]. Genetic polymorphisms in the IL-1β gene, including 511T and 31C variants, are linked to elevated IL-1β production and increased susceptibility to gastric adenocarcinoma, particularly in H. pylori-infected individuals[56]. Moreover, IL-1β promotes carcinogenesis through epigenetic mechanisms, notably aberrant DNA methylation, further underscoring its pivotal role in gastric tumorigenesis[57].

TNF-α

TNF-α is a central mediator of chronic inflammation and a key contributor to gastric carcinogenesis[58]. Secreted mainly by activated macrophages and immune cells, TNF-α fosters a tumor-promoting microenvironment[59]. It activates multiple signaling pathways, including NF-κB and mitogen-activated protein kinases, which regulate genes involved in cell proliferation, angiogenesis, and inhibition of apoptosis[60]. These processes enable gastric epithelial cells to evade growth control, resist cell death, and acquire malignant properties.

During H. pylori infection, TNF-α expression is upregulated as part of the host immune response, perpetuating chronic mucosal inflammation. Persistent TNF-α exposure damages the gastric epithelium, increases ROS generation, and promotes EMT, a key step in invasion and metastasis[61]. Elevated TNF-α levels correlate with aggressive histological subtypes, advanced tumor stages, and poor prognosis[61]. Genetic polymorphisms in TNF-α have also been linked to heightened GC risk in specific populations[61]. Mechanistically, TNF-α enhances GC aggressiveness by increasing cytoplasmic claudin 1 expression, which promotes invasion and cell survival through NF-κB and MMP-7-dependent pathways. In contrast, other TJ proteins such as claudin 4, occludin, and ZO-1 are downregulated in advanced GC, reflecting TJ disruption as a hallmark of tumor progression, although direct regulation of these proteins by TNF-α remains unclear[61,62].

IL-6

IL-6 is a multifunctional cytokine that plays a crucial role in GC progression[62]. Elevated IL-6 levels are consistently detected in the serum and tumor microenvironment of GC patients and are associated with poor prognosis[63]. IL-6 promotes tumor progression primarily through activation of the Janus kinase (JAK)/signal transducer and activator of transcription 3 (STAT3) signaling pathway, which enhances cancer cell proliferation, survival, invasion, and EMT[63]. IL-6 also contributes to epithelial barrier dysfunction by modulating TJ protein expression. Studies in GI epithelial models demonstrate that IL-6 upregulates claudin 2 via the c-Jun N-terminal kinase and phosphatidylinositol 3-kinase/extracellular signal-regulated kinase pathways, increasing paracellular permeability[64]. Although this mechanism is best characterized in the intestinal epithelium, it is likely relevant to the gastric mucosa, particularly under chronic inflammatory conditions such as H. pylori infection. TJ disruption compromises mucosal integrity, facilitates microbial translocation, and perpetuates inflammation, creating a self-sustaining cycle that promotes carcinogenesis. Clinically, IL-6 expression is significantly higher in gastric tumor tissues than in adjacent non-tumor or normal gastric samples. Elevated intra-tumoral and circulating IL-6 levels correlate with advanced disease stage, reduced patient survival, and decline following effective therapy[64,65]. Within the tumor microenvironment, cancer-associated fibroblasts (CAFs) secrete IL-6, which further enhances GC cell growth and invasion through the JAK2/STAT3 pathway[66]. Experimental IL-6 knockout in murine models markedly suppresses gastric tumor formation, underscoring its pivotal role in inflammation-driven gastric carcinogenesis[67].

IL-8

IL-8, also known as C-X-C motif chemokine ligand 8, is a pro-inflammatory chemokine that plays a crucial role in the recruitment of immune cells particularly neutrophils to sites of inflammation[68]. It is frequently upregulated in response to H. pylori infection and is significantly implicated in the progression of GC[69]. IL-8 promotes tumor growth by stimulating angiogenesis, enhancing the supply of oxygen and nutrients to cancerous tissues[69,70]. Additionally, IL-8 recruits neutrophils which release ROS and proteolytic enzymes that can cause DNA damage, facilitating tumor initiation and progression. Beyond its pro-inflammatory role, IL-8 enhances GC cell migration and invasion, contributing directly to metastatic potential[70]. A key pathway for metastasis in GC is via tumor-draining lymph nodes, with lymph node metastasis being a well-established predictor of poor prognosis. The tumor microenvironment comprising CAFs, immune and inflammatory cells, and the extracellular matrix plays a pivotal role in shaping metastatic behavior[71]. CAFs in particular have been identified as a major source of soluble IL-8, contributing to a pro-tumorigenic microenvironment[71].

Recent findings have shown that elevated IL-8 levels can induce programmed death-1 (PD-1) expression on CD8+ T cells, leading to local immunosuppression within both the primary tumor and tumor-draining lymph nodes[72]. Mechanistically, IL-8 activates the JAK2/STAT3 signaling pathway to upregulate PD-1 and simultaneously inhibits PD-1 degradation by downregulating Fbxo38, an E3 ubiquitin ligase involved in PD-1 ubiquitination[71,72]. This dual action fosters an immunosuppressive niche that facilitates immune evasion and promotes lymph node metastasis. Clinically, high levels of soluble IL-8 are closely associated with increased lymph node metastasis and are indicative of a poorer overall prognosis in GC patients. In endothelial cells, IL-8 was shown to enhance permeability by reducing both mRNA and protein levels of occludin, claudin 5, and ZO-1 in a manner dependent on concentration and exposure time. Additionally, it caused redistribution of TJ proteins where occludin shifted from the membrane into the cytoplasm, while membrane-associated ZO-1 was diminished[73]. IL-8 exhibits a complex, context-dependent role in GC. While patient tumor tissues often show reduced claudin 4 expression, elevated IL-8 levels in advanced disease paradoxically upregulate claudin 4. This alteration is closely linked to the induction of EMT, mediated by increased Snail and Twist through extracellular signal-regulated kinase/mitogen-activated protein kinases pathway activation, ultimately emphasizing the significant contribution of IL-8 to GC progression[73].

Oxidative stress

Oxidative stress plays a pivotal role in the initiation and progression of GC by inducing DNA damage, sustaining chronic inflammation, activating oncogenic signaling pathways, and enhancing tumor cell survival[74]. A major source of oxidative stress in the gastric environment is H. pylori infection, a well-established risk factor for GC[75]. H. pylori stimulates the host immune response through neutrophil and macrophage activation, leading to excessive production of reactive oxygen and nitrogen species[75,76]. Persistent generation of these reactive species causes oxidative damage to lipids, proteins, and nucleic acids, contributing to chronic gastritis, intestinal metaplasia, and malignant transformation of gastric epithelial cells. Additionally, ROS activate redox-sensitive transcription factors such as NF-κB and activating protein-1, amplifying pro-inflammatory cytokine expression and perpetuating a cycle of inflammation and oxidative injury[77]. Oxidative stress also disrupts the gastric epithelial barrier by impairing TJ proteins. In gastric epithelial cells, hydrogen peroxide decreases claudin 3 expression and transepithelial electrical resistance, indicating increased permeability. These effects are reversed by the antioxidant rebamipide, demonstrating a direct oxidative stress mediated mechanism of TJ disruption and barrier dysfunction[77].

ROLE OF GASTRIC MICROBIOTA AND DYSREGULATED TJ PROTEINS IN THE DEVELOPMENT OF GC

Under normal conditions, TJs prevent the unregulated entry of gastric and intestinal microbes, their toxins, and antigens into the underlying mucosa. By preserving this selective barrier, TJ proteins maintain immune balance permitting only limited and controlled exposure of microbial components to immune cells (via dendritic cells and specialized epithelial mechanisms) without provoking persistent inflammation. Occludin and claudins determine the sealing and pore-forming properties of the junctions, thereby controlling the size and type of molecules that pass between epithelial cells. ZO-1 connects these proteins to the actin cytoskeleton, providing structural stability and integrating barrier regulation with innate immune signaling. Thus, intact TJ proteins compartmentalize gastric microbiota at the luminal surface, fostering beneficial host-microbe interactions while preventing overgrowth, dysbiosis, or aberrant immune activation. Conversely, disruption of TJ proteins leads to barrier leakage, enabling microbial products such as lipopolysaccharide (LPS) and peptidoglycan to reach deeper tissue layers, where they activate Toll-like receptors, inflammasomes, and cytokine pathways. This cascade drives chronic inflammation and contributes to the progression toward gastric carcinogenesis[78,79].

The gut microbiota plays a fundamental role in maintaining GI health by modulating immune responses, regulating intestinal permeability, and preserving epithelial barrier integrity[80]. It consists of trillions of microorganisms including bacteria, fungi, viruses, and archaea that contribute significantly to the maintenance of TJs[81]. Certain beneficial bacteria, particularly those that produce short-chain fatty acids like butyrate, enhance TJ function[82]. Butyrate specifically strengthens the epithelial barrier by upregulating TJ protein expression and increasing mucus production, which further protects the gut lining[82]. A healthy microbiota also helps modulate local immune responses, thereby preventing chronic inflammation. In contrast, dysbiosis an imbalance in the microbial community can disrupt TJs, increase intestinal permeability, and promote inflammation. Pathogenic or opportunistic bacteria may produce substances that degrade TJ proteins or activate inflammatory signaling pathways, compromising the epithelial barrier[83].

LPS, produced by Gram-negative bacteria, is a key contributor to TJ disruption and inflammation. LPS activates immune cells and induces inflammatory cytokine production, further impairing barrier function[84]. When TJs are compromised, larger molecules such as bacterial endotoxins and undigested food particles can translocate across the gut lining into the systemic circulation a phenomenon often referred to as “leaky gut syndrome” which may trigger systemic inflammation and immune activation[84].

In the gastric environment, reduced acid secretion due to H. pylori infection facilitates the colonization of additional bacterial species. For instance, the genus Lactobacillus is found in higher abundance in GC patients[85]. Microbes from the Lactobacillus and Lactococcus genera produce lactic acid, which may fuel tumor progression by serving as an energy source for cancer cell proliferation and promoting angiogenesis. While these microbes may also play a regulatory role in inflammation, their abundance often decreases during inflammatory conditions[86].

Interestingly, the phylum Nitrospirae has been detected in all GC patients but is absent in individuals with chronic gastritis. Members of this phylum are involved in nitrate and nitrite metabolism[87]. As nitrate consumption is a known risk factor for GC, these bacteria may contribute to the generation of carcinogenic N-nitroso compounds. Several bacterial genera typically found in the oral cavity such as Fusobacterium, Veillonella, Leptotrichia, Haemophilus, and Campylobacter have been detected in higher relative abundance in GC patients. Additionally, Propionibacterium acnes, commonly found on the skin, is often overrepresented in gastric tumor tissues. It is hypothesized that short-chain fatty acids produced by this bacterium may contribute to lymphocytic gastritis[88-90]. The role of inflammation and altered gut microbiota in causing GC is schematically shown in Figure 2.

Figure 2
Figure 2 Schematic illustration of the interplay among tight junctions, inflammation, gut microbiota, and gastric cancer progression. Helicobacter pylori infection triggers gastric inflammation by activating cytokine, nuclear factor kappa B, and signal transducer and activator of transcription 3 mediated inflammatory cascades, leading to tight junction disruption and increased intestinal permeability. The resulting leaky gut permits translocation of microbial toxins and inflammatory mediators, which promote epithelial mesenchymal transition and drive gastric carcinogenesis. IL: Interleukin; TNF: Tumor necrosis factor; NF-κB: Nuclear factor kappa B; STAT3: Signal transducer and activator of transcription 3; H. pylori: Helicobacter pylori; TJ: Tight junction; EMT: Epithelial mesenchymal transition.
TREATMENT APPROACHES IN GC PATIENTS TARGETING TJ PROTEINS

Although several stage-specific treatment modalities are well established such as the FLOT regimen based chemotherapy and gastrectomy with or without lymphadenectomy for locally advanced resectable tumors it is increasingly important to explore novel molecular targets to overcome chemotherapy resistance[91]. TJ proteins have emerged as promising therapeutic targets in this context. Ongoing clinical trials are currently investigating agents directed against TJ proteins, including claudin 4, claudin 8, and claudin 18.2, to evaluate their efficacy and safety in the treatment of GC. Targeting these proteins may therefore represent a novel strategy to overcome treatment resistance and improve patient outcomes. Several ongoing clinical trials are exploring monoclonal antibodies, antibody-drug conjugates (ADCs), and chimeric antigen receptor (CAR) T-cell therapies directed against TJ proteins such as claudin 4, claudin 8, and claudin 18.2. Among these, claudin 18.2 has emerged as the most promising target, with multiple agents such as zolbetuximab, CMG901, and CAR T-cell therapy CT041[92] showing encouraging results in early- and late-phase trials (as listed in Table 2). These therapies have demonstrated manageable safety profiles and promising antitumor activity, highlighting the therapeutic potential of TJ protein-directed approaches. These TJ protein-targeted therapies represent a rapidly evolving field that may complement or even redefine the current treatment paradigm for GC, particularly in patients who fail to respond adequately to standard chemotherapy-based regimens.

Table 2 Claudin based immunotherapy for gastric cancer patients.
No.
Targeted TJ proteins
Therapeutic agent(s)
Development stages in GC
Effect in GC patients
Ref.
1Claudin 18.2 mAbCMG901Phase IDemonstrated a manageable safety profile and promising antitumor activity[93]
2Claudin 18.2 mAbIBI343Phase IDemonstrated a manageable safety profile with low gastrointestinal adverse events and showed promising efficacy[94]
3Claudin 18.2 mAbCAR T-cell therapy, CT041Phase IIDemonstrated significant improvement in progression-free survival, with a manageable safety profile[95]
4Claudin 18.2 mAbZolbetuximab plus mFOLFOX6Phase IIISignificantly prolonged progression-free survival and overall survival compared with placebo[101]
5Claudin 18.2 mAbZolbetuximab (IMAB362) combined with EOXPhase IIProvided longer progression-free survival and overall survival compared with EOX alone. The primary endpoint was progression-free survival, while overall survival was assessed as a secondary endpoint[102]
6Claudin 18.2 mAbZolbetuximabPhase IAntitumor activity of IMAB362, both as a single agent and in combination with standard therapeutics, was observed[103]
7Claudin 18.2 mAbClaudiximabPhase IIDemonstrated significant efficacy and a favorable safety profile[104]
CLAUDIN 18.2 TARGETED THERAPIES IN GC

CMG901 is a first-in-class ADC comprising a humanized anti-claudin 18.2 monoclonal antibody linked to the microtubule-disrupting agent monomethyl auristatin E. It has been evaluated in patients with gastric and gastroesophageal junction (GEJ) cancers. CMG901 was administered intravenously every three weeks at doses ranging from 0.3-2.4 mg/kg during dose escalation and 2.2-3 mg/kg during dose expansion[93]. The primary endpoints included assessment of dose-limiting toxicities (DLTs) and overall safety. Pancreatitis was identified as a serious DLT at 2.2 mg/kg, establishing the maximum tolerated dose. Other common adverse events included vomiting, reduced appetite, proteinuria, and fatigue. Hematologic toxicities such as leukopenia, neutropenia, lymphopenia, thrombocytopenia, and anemia were also observed. GI symptoms, including nausea, vomiting, and diarrhea, were generally mild and manageable compared with conventional chemotherapy[93].

IBI343 is an ADC composed of a fully humanized monoclonal antibody targeting claudin 18.2, conjugated to the topoisomerase I inhibitor exatecan via site-specific glyco-conjugation using a cleavable linker, yielding a drug-to-antibody ratio of 4. In clinical studies, the recommended phase II dose was 6 mg/kg administered every three weeks. This regimen achieved an objective response and a median progression-free survival of 5.5 months in patients with gastric or GEJ adenocarcinoma exhibiting high claudin 18.2 expression[94]. The treatment demonstrated favorable tolerability and a manageable safety profile. DLTs were observed at 10 mg/kg, consisting of grade 4 myelosuppression and neutropenia, accompanied by grade 3 febrile neutropenia. Severe GI adverse events (grade ≥ 3) were uncommon. Overall, IBI343 monotherapy showed encouraging antitumor activity with acceptable safety. Combination strategies particularly with immunotherapy are under investigation to further enhance therapeutic efficacy. Phase III, multicenter, randomized trials are ongoing to validate its role in claudin 18.2 positive gastric and other solid tumors[94].

CT041 is a claudin 18.2-specific CAR T-cell therapy generated from autologous T cells engineered to express a receptor targeting the claudin 18.2 antigen[95]. Preclinical studies demonstrated potent antitumor activity against claudin 18.2-positive GC cell lines and patient-derived xenograft models. Based on phase I safety evaluations, the data safety monitoring committee recommended a dose of 2.5 × 108 CAR-T cells for expansion cohorts[95]. The most common adverse events were hematologic, related primarily to preconditioning, including leukopenia, neutropenia, anemia, and thrombocytopenia. Notably, no grade ≥ 3 cytokine release syndrome was reported. Tumor regression occurred in most treated patients, with CAR-T cell expansion correlating with reductions in circulating tumor DNA. Collectively, clinical trials of CT041 in advanced gastric and other GI cancers have shown robust antitumor activity with manageable safety profiles[96,97]. Ongoing studies continue to explore next-generation claudin 18.2-targeted approaches, including bispecific antibodies such as IBI389 and universal “modular” CAR-T platforms like IBI345, which aim to broaden applicability and enhance therapeutic outcomes.

More recently, zolbetuximab, a monoclonal antibody selectively targeting claudin 18.2, has emerged as a promising targeted therapy for advanced GC. In phase III trials, the addition of zolbetuximab to chemotherapy significantly improved overall survival compared with chemotherapy alone in HER2-negative patients with claudin 18.2 positive unresectable gastric or GEJ adenocarcinoma[98]. Beyond zolbetuximab, several claudin 18.2-directed immunotherapies including monoclonal antibodies, bispecific antibodies, CAR T-cell therapies, and ADCs are under active clinical development[99,100].

CONCLUSION

TJ proteins are crucial for maintaining gastric mucosal integrity, and their dysregulation is strongly linked to the initiation and progression of GC. Aberrant TJ protein expression increases epithelial permeability, drives chronic inflammation, and promotes tumor growth, invasion, and metastasis. These findings underscore the role of TJ proteins not only in gastric carcinogenesis but also as potential biomarkers and therapeutic targets. While claudin 18.2 based therapies have already entered clinical application, further research is needed to elucidate the mechanisms underlying dysregulation of other TJ proteins. Such efforts may facilitate the development of novel diagnostic tools and precision therapies that improve survival and quality of life in GC patients.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Oncology

Country of origin: India

Peer-review report’s classification

Scientific Quality: Grade B, Grade C

Novelty: Grade C, Grade C

Creativity or Innovation: Grade C, Grade C

Scientific Significance: Grade B, Grade C

P-Reviewer: Shirsat P, MD, Consultant, United States; Sitkin S, MD, PhD, Associate Professor, Head, Russia S-Editor: Wang JJ L-Editor: Webster JR P-Editor: Zhang L

References
1.  Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, Bray F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin. 2021;71:209-249.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 75126]  [Cited by in RCA: 67569]  [Article Influence: 13513.8]  [Reference Citation Analysis (185)]
2.  Smyth EC, Nilsson M, Grabsch HI, van Grieken NC, Lordick F. Gastric cancer. Lancet. 2020;396:635-648.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1150]  [Cited by in RCA: 3199]  [Article Influence: 533.2]  [Reference Citation Analysis (5)]
3.  Thrift AP, El-Serag HB. Burden of Gastric Cancer. Clin Gastroenterol Hepatol. 2020;18:534-542.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 432]  [Cited by in RCA: 1057]  [Article Influence: 176.2]  [Reference Citation Analysis (7)]
4.  Thrift AP, Wenker TN, El-Serag HB. Global burden of gastric cancer: epidemiological trends, risk factors, screening and prevention. Nat Rev Clin Oncol. 2023;20:338-349.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 445]  [Reference Citation Analysis (1)]
5.  Servarayan Murugesan C, Manickavasagam K, Chandramohan A, Jebaraj A, Jameel ARA, Jain MS, Venkataraman J. Gastric cancer in India: epidemiology and standard of treatment. Updates Surg. 2018;70:233-239.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 17]  [Cited by in RCA: 28]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
6.  Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68:394-424.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 53206]  [Cited by in RCA: 56469]  [Article Influence: 7058.6]  [Reference Citation Analysis (132)]
7.  Liu Z, Zhang D, Chen S. Unveiling the gastric microbiota: implications for gastric carcinogenesis, immune responses, and clinical prospects. J Exp Clin Cancer Res. 2024;43:118.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 22]  [Reference Citation Analysis (0)]
8.  Poorolajal J, Moradi L, Mohammadi Y, Cheraghi Z, Gohari-Ensaf F. Risk factors for stomach cancer: a systematic review and meta-analysis. Epidemiol Health. 2020;42:e2020004.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 180]  [Cited by in RCA: 182]  [Article Influence: 30.3]  [Reference Citation Analysis (0)]
9.  Fanfarillo F, Caronti B, Lucarelli M, Francati S, Tarani L, Ceccanti M, Piccioni MG, Verdone L, Caserta M, Venditti S, Ferraguti G, Fiore M. Alcohol Consumption and Breast and Ovarian Cancer Development: Molecular Pathways and Mechanisms. Curr Issues Mol Biol. 2024;46:14438-14452.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
10.  Yang S, Lee J, Choi IJ, Kim YW, Ryu KW, Sung J, Kim J. Effects of alcohol consumption, ALDH2 rs671 polymorphism, and Helicobacter pylori infection on the gastric cancer risk in a Korean population. Oncotarget. 2017;8:6630-6641.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 20]  [Cited by in RCA: 24]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
11.  Song M, Camargo MC, Weinstein SJ, Best AF, Männistö S, Albanes D, Rabkin CS. Family history of cancer in first-degree relatives and risk of gastric cancer and its precursors in a Western population. Gastric Cancer. 2018;21:729-737.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 18]  [Cited by in RCA: 28]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
12.  Duan F, Song C, Shi J, Wang P, Ye H, Dai L, Zhang J, Wang K. Identification and epidemiological evaluation of gastric cancer risk factors: based on a field synopsis and meta-analysis in Chinese population. Aging (Albany NY). 2021;13:21451-21469.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 14]  [Cited by in RCA: 14]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
13.  Charles L, Elangovan A, Nisha Y, Jafa E, Kate V, Selvarajan S, Kayal S, Ganesh RN, Dubashi B, Penumadu P, Ganesan P. Clinicopathological features and survival outcomes for gastric adenocarcinoma: Real-world single-center data. Indian J Gastroenterol. 2024;43:1209-1219.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
14.  Hu YL, Pang W, Huang Y, Zhang Y, Zhang CJ. The Gastric Microbiome Is Perturbed in Advanced Gastric Adenocarcinoma Identified Through Shotgun Metagenomics. Front Cell Infect Microbiol. 2018;8:433.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 56]  [Cited by in RCA: 94]  [Article Influence: 11.8]  [Reference Citation Analysis (0)]
15.  Cao B, Hu J, Li H, Liu X, Rong C, Li S, He X, Zheng X, Liu K, Wang C, Guo W, Wu X. Preoperative prediction of the Lauren classification in gastric cancer using automated nnU-Net and radiomics: a multicenter study. Insights Imaging. 2025;16:48.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
16.  Tsukita S, Furuse M, Itoh M. Multifunctional strands in tight junctions. Nat Rev Mol Cell Biol. 2001;2:285-293.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1851]  [Cited by in RCA: 1926]  [Article Influence: 77.0]  [Reference Citation Analysis (0)]
17.  Turner JR. Intestinal mucosal barrier function in health and disease. Nat Rev Immunol. 2009;9:799-809.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 2121]  [Cited by in RCA: 2806]  [Article Influence: 165.1]  [Reference Citation Analysis (0)]
18.  Kuo WT, Odenwald MA, Turner JR, Zuo L. Tight junction proteins occludin and ZO-1 as regulators of epithelial proliferation and survival. Ann N Y Acad Sci. 2022;1514:21-33.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 368]  [Cited by in RCA: 324]  [Article Influence: 81.0]  [Reference Citation Analysis (0)]
19.  Moonwiriyakit A, Pathomthongtaweechai N, Steinhagen PR, Chantawichitwong P, Satianrapapong W, Pongkorpsakol P. Tight junctions: from molecules to gastrointestinal diseases. Tissue Barriers. 2023;11:2077620.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 59]  [Reference Citation Analysis (0)]
20.  Shin K, Fogg VC, Margolis B. Tight junctions and cell polarity. Annu Rev Cell Dev Biol. 2006;22:207-235.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 531]  [Cited by in RCA: 584]  [Article Influence: 29.2]  [Reference Citation Analysis (2)]
21.  Steed E, Balda MS, Matter K. Dynamics and functions of tight junctions. Trends Cell Biol. 2010;20:142-149.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 278]  [Cited by in RCA: 311]  [Article Influence: 19.4]  [Reference Citation Analysis (0)]
22.  Tsukita S, Tanaka H, Tamura A. The Claudins: From Tight Junctions to Biological Systems. Trends Biochem Sci. 2019;44:141-152.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 156]  [Cited by in RCA: 322]  [Article Influence: 46.0]  [Reference Citation Analysis (0)]
23.  Chelakkot C, Ghim J, Ryu SH. Mechanisms regulating intestinal barrier integrity and its pathological implications. Exp Mol Med. 2018;50:1-9.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 484]  [Cited by in RCA: 1275]  [Article Influence: 159.4]  [Reference Citation Analysis (1)]
24.  Guinane CM, Cotter PD. Role of the gut microbiota in health and chronic gastrointestinal disease: understanding a hidden metabolic organ. Therap Adv Gastroenterol. 2013;6:295-308.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 499]  [Cited by in RCA: 582]  [Article Influence: 44.8]  [Reference Citation Analysis (0)]
25.  Díaz-Coránguez M, Liu X, Antonetti DA. Tight Junctions in Cell Proliferation. Int J Mol Sci. 2019;20:5972.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 54]  [Cited by in RCA: 73]  [Article Influence: 10.4]  [Reference Citation Analysis (0)]
26.  Hashimoto I, Oshima T. Claudins and Gastric Cancer: An Overview. Cancers (Basel). 2022;14:290.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 22]  [Cited by in RCA: 101]  [Article Influence: 25.3]  [Reference Citation Analysis (0)]
27.  Ram AK, Vairappan B. Role of zonula occludens in gastrointestinal and liver cancers. World J Clin Cases. 2022;10:3647-3661.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in CrossRef: 7]  [Cited by in RCA: 6]  [Article Influence: 1.5]  [Reference Citation Analysis (3)]
28.  Otani T, Furuse M. Tight Junction Structure and Function Revisited. Trends Cell Biol. 2020;30:805-817.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 108]  [Cited by in RCA: 446]  [Article Influence: 74.3]  [Reference Citation Analysis (2)]
29.  Otani T, Nguyen TP, Tokuda S, Sugihara K, Sugawara T, Furuse K, Miura T, Ebnet K, Furuse M. Claudins and JAM-A coordinately regulate tight junction formation and epithelial polarity. J Cell Biol. 2019;218:3372-3396.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 129]  [Cited by in RCA: 167]  [Article Influence: 23.9]  [Reference Citation Analysis (0)]
30.  Belardi B, Hamkins-Indik T, Harris AR, Kim J, Xu K, Fletcher DA. A Weak Link with Actin Organizes Tight Junctions to Control Epithelial Permeability. Dev Cell. 2020;54:792-804.e7.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 22]  [Cited by in RCA: 62]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
31.  Ohtani S, Terashima M, Satoh J, Soeta N, Saze Z, Kashimura S, Ohsuka F, Hoshino Y, Kogure M, Gotoh M. Expression of tight-junction-associated proteins in human gastric cancer: downregulation of claudin-4 correlates with tumor aggressiveness and survival. Gastric Cancer. 2009;12:43-51.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 60]  [Cited by in RCA: 61]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
32.  Du D, Xu F, Yu L, Zhang C, Lu X, Yuan H, Huang Q, Zhang F, Bao H, Jia L, Wu X, Zhu X, Zhang X, Zhang Z, Chen Z. The tight junction protein, occludin, regulates the directional migration of epithelial cells. Dev Cell. 2010;18:52-63.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 117]  [Cited by in RCA: 136]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
33.  Furuse M, Fujimoto K, Sato N, Hirase T, Tsukita S, Tsukita S. Overexpression of occludin, a tight junction-associated integral membrane protein, induces the formation of intracellular multilamellar bodies bearing tight junction-like structures. J Cell Sci. 1996;109 (Pt 2):429-435.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 140]  [Cited by in RCA: 130]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
34.  McCarthy KM, Skare IB, Stankewich MC, Furuse M, Tsukita S, Rogers RA, Lynch RD, Schneeberger EE. Occludin is a functional component of the tight junction. J Cell Sci. 1996;109 (Pt 9):2287-2298.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 473]  [Cited by in RCA: 454]  [Article Influence: 15.1]  [Reference Citation Analysis (1)]
35.  Feldman GJ, Mullin JM, Ryan MP. Occludin: structure, function and regulation. Adv Drug Deliv Rev. 2005;57:883-917.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 281]  [Cited by in RCA: 378]  [Article Influence: 18.0]  [Reference Citation Analysis (0)]
36.  Yang L, Sun X, Meng X. Differences in the expression profiles of claudin proteins in human gastric carcinoma compared with nonneoplastic mucosa. Mol Med Rep. 2018;18:1271-1278.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 8]  [Cited by in RCA: 14]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
37.  Nehme Z, Roehlen N, Dhawan P, Baumert TF. Tight Junction Protein Signaling and Cancer Biology. Cells. 2023;12:243.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 29]  [Cited by in RCA: 53]  [Article Influence: 17.7]  [Reference Citation Analysis (0)]
38.  Han L, Cui DJ, Huang B, Yang Q, Huang T, Lin GY, Chen SJ. CLDN5 identified as a biomarker for metastasis and immune infiltration in gastric cancer via pan-cancer analysis. Aging (Albany NY). 2023;15:5032-5051.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
39.  Torres-Martínez AC, Gallardo-Vera JF, Lara-Holguin AN, Montaño LF, Rendón-Huerta EP. Claudin-6 enhances cell invasiveness through claudin-1 in AGS human adenocarcinoma gastric cancer cells. Exp Cell Res. 2017;350:226-235.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 19]  [Cited by in RCA: 29]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
40.  Wu Z, Shi J, Song Y, Zhao J, Sun J, Chen X, Gao P, Wang Z. Claudin-7 (CLDN7) is overexpressed in gastric cancer and promotes gastric cancer cell proliferation, invasion and maintains mesenchymal state. Neoplasma. 2018;65:349-359.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 10]  [Cited by in RCA: 16]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
41.  Kolchakova D, Moten D, Batsalova T, Dzhambazov B. Tight Junction Protein Claudin-12 Is Involved in Cell Migration during Metastasis. Biomolecules. 2021;11:636.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 4]  [Cited by in RCA: 14]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
42.  Mathias-Machado MC, de Jesus VHF, Jácome A, Donadio MD, Aruquipa MPS, Fogacci J, Cunha RG, da Silva LM, Peixoto RD. Claudin 18.2 as a New Biomarker in Gastric Cancer-What Should We Know? Cancers (Basel). 2024;16:679.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 29]  [Reference Citation Analysis (0)]
43.  Ogawa H, Abe H, Yagi K, Seto Y, Ushiku T. Claudin-18 status and its correlation with HER2 and PD-L1 expression in gastric cancer with peritoneal dissemination. Gastric Cancer. 2024;27:802-810.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 16]  [Reference Citation Analysis (0)]
44.  Katoh M, Katoh M. CLDN23 gene, frequently down-regulated in intestinal-type gastric cancer, is a novel member of CLAUDIN gene family. Int J Mol Med. 2003;11:683-689.  [PubMed]  [DOI]
45.  Raya-Sandino A, Lozada-Soto KM, Rajagopal N, Garcia-Hernandez V, Luissint AC, Brazil JC, Cui G, Koval M, Parkos CA, Nangia S, Nusrat A. Claudin-23 reshapes epithelial tight junction architecture to regulate barrier function. Nat Commun. 2023;14:6214.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 16]  [Cited by in RCA: 43]  [Article Influence: 14.3]  [Reference Citation Analysis (0)]
46.  Xie Y, Liu F, Wu Y, Zhu Y, Jiang Y, Wu Q, Dong Z, Liu K. Inflammation in cancer: therapeutic opportunities from new insights. Mol Cancer. 2025;24:51.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 2]  [Cited by in RCA: 41]  [Article Influence: 41.0]  [Reference Citation Analysis (0)]
47.  Chen L, Deng H, Cui H, Fang J, Zuo Z, Deng J, Li Y, Wang X, Zhao L. Inflammatory responses and inflammation-associated diseases in organs. Oncotarget. 2018;9:7204-7218.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 2209]  [Cited by in RCA: 3091]  [Article Influence: 386.4]  [Reference Citation Analysis (0)]
48.  Akdis M, Aab A, Altunbulakli C, Azkur K, Costa RA, Crameri R, Duan S, Eiwegger T, Eljaszewicz A, Ferstl R, Frei R, Garbani M, Globinska A, Hess L, Huitema C, Kubo T, Komlosi Z, Konieczna P, Kovacs N, Kucuksezer UC, Meyer N, Morita H, Olzhausen J, O'Mahony L, Pezer M, Prati M, Rebane A, Rhyner C, Rinaldi A, Sokolowska M, Stanic B, Sugita K, Treis A, van de Veen W, Wanke K, Wawrzyniak M, Wawrzyniak P, Wirz OF, Zakzuk JS, Akdis CA. Interleukins (from IL-1 to IL-38), interferons, transforming growth factor β, and TNF-α: Receptors, functions, and roles in diseases. J Allergy Clin Immunol. 2016;138:984-1010.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 419]  [Cited by in RCA: 645]  [Article Influence: 64.5]  [Reference Citation Analysis (0)]
49.  Wei YF, Li X, Zhao MR, Liu S, Min L, Zhu ST, Zhang ST, Xie SA. Helicobacter pylori disrupts gastric mucosal homeostasis by stimulating macrophages to secrete CCL3. Cell Commun Signal. 2024;22:263.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 9]  [Cited by in RCA: 15]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
50.  He Z, Zhou Y, Liu J, Li N, Fan H. The intersection of Helicobacter pylori and gastric cancer: signaling pathways and molecular mechanisms. Front Cell Infect Microbiol. 2025;15:1601501.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
51.  Wroblewski LE, Shen L, Ogden S, Romero-Gallo J, Lapierre LA, Israel DA, Turner JR, Peek RM Jr. Helicobacter pylori dysregulation of gastric epithelial tight junctions by urease-mediated myosin II activation. Gastroenterology. 2009;136:236-246.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 163]  [Cited by in RCA: 154]  [Article Influence: 9.1]  [Reference Citation Analysis (0)]
52.  Hong JB, Zuo W, Wang AJ, Lu NH. Helicobacter pylori Infection Synergistic with IL-1β Gene Polymorphisms Potentially Contributes to the Carcinogenesis of Gastric Cancer. Int J Med Sci. 2016;13:298-303.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 23]  [Cited by in RCA: 37]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
53.  Balkwill F, Mantovani A. Inflammation and cancer: back to Virchow? Lancet. 2001;357:539-545.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 5245]  [Cited by in RCA: 5830]  [Article Influence: 233.2]  [Reference Citation Analysis (0)]
54.  Al-Sadi R, Ye D, Said HM, Ma TY. IL-1beta-induced increase in intestinal epithelial tight junction permeability is mediated by MEKK-1 activation of canonical NF-kappaB pathway. Am J Pathol. 2010;177:2310-2322.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 153]  [Cited by in RCA: 172]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
55.  Zheng W, Zhang S, Zhang S, Min L, Wang Y, Xie J, Hou Y, Tian X, Cheng J, Liu K, Xu D, Yu X, Liu Z, Lv Y, Liang N, Zhang J, Liu F, Tian Y. The relationship between tumor necrosis factor-α polymorphisms and gastric cancer risk: An updated meta-analysis. Biomed Rep. 2017;7:133-142.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 12]  [Cited by in RCA: 18]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
56.  O'Hara AM, Bhattacharyya A, Bai J, Mifflin RC, Ernst PB, Mitra S, Crowe SE. Tumor necrosis factor (TNF)-alpha-induced IL-8 expression in gastric epithelial cells: role of reactive oxygen species and AP endonuclease-1/redox factor (Ref)-1. Cytokine. 2009;46:359-369.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 46]  [Cited by in RCA: 49]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
57.  Poritz LS, Garver KI, Tilberg AF, Koltun WA. Tumor necrosis factor alpha disrupts tight junction assembly. J Surg Res. 2004;116:14-18.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 68]  [Cited by in RCA: 70]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
58.  Gu H, Huang T, Shen Y, Liu Y, Zhou F, Jin Y, Sattar H, Wei Y. Reactive Oxygen Species-Mediated Tumor Microenvironment Transformation: The Mechanism of Radioresistant Gastric Cancer. Oxid Med Cell Longev. 2018;2018:5801209.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 29]  [Cited by in RCA: 55]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
59.  Xu T, Kong Z, Zhao H. Relationship Between Tumor Necrosis Factor-α rs361525 Polymorphism and Gastric Cancer Risk: A Meta-Analysis. Front Physiol. 2018;9:469.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 9]  [Cited by in RCA: 14]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
60.  Chaithongyot S, Jantaree P, Sokolova O, Naumann M. NF-κB in Gastric Cancer Development and Therapy. Biomedicines. 2021;9:870.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 8]  [Cited by in RCA: 32]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
61.  Chen G, Tang N, Wang C, Xiao L, Yu M, Zhao L, Cai H, Han L, Xie C, Zhang Y. TNF-α-inducing protein of Helicobacter pylori induces epithelial-mesenchymal transition (EMT) in gastric cancer cells through activation of IL-6/STAT3 signaling pathway. Biochem Biophys Res Commun. 2017;484:311-317.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 28]  [Cited by in RCA: 33]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
62.  Wang L, Xiao S, Zheng Y, Gao Z, Fan F. Impact of interaction between interleukin-6 gene polymorphism and Helicobacter pylori infection on susceptibility to gastric cancer. Eur J Cancer Prev. 2024;33:136-140.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 2]  [Cited by in RCA: 2]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
63.  Wu X, Tao P, Zhou Q, Li J, Yu Z, Wang X, Li J, Li C, Yan M, Zhu Z, Liu B, Su L. IL-6 secreted by cancer-associated fibroblasts promotes epithelial-mesenchymal transition and metastasis of gastric cancer via JAK2/STAT3 signaling pathway. Oncotarget. 2017;8:20741-20750.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 153]  [Cited by in RCA: 286]  [Article Influence: 31.8]  [Reference Citation Analysis (0)]
64.  Suzuki T, Yoshinaga N, Tanabe S. Interleukin-6 (IL-6) regulates claudin-2 expression and tight junction permeability in intestinal epithelium. J Biol Chem. 2011;286:31263-31271.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 336]  [Cited by in RCA: 435]  [Article Influence: 29.0]  [Reference Citation Analysis (0)]
65.  Wu CW, Wang SR, Chao MF, Wu TC, Lui WY, P'eng FK, Chi CW. Serum interleukin-6 levels reflect disease status of gastric cancer. Am J Gastroenterol. 1996;91:1417-1422.  [PubMed]  [DOI]
66.  Yu B, de Vos D, Guo X, Peng S, Xie W, Peppelenbosch MP, Fu Y, Fuhler GM. IL-6 facilitates cross-talk between epithelial cells and tumor- associated macrophages in Helicobacter pylori-linked gastric carcinogenesis. Neoplasia. 2024;50:100981.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 12]  [Reference Citation Analysis (0)]
67.  Kinoshita H, Hirata Y, Nakagawa H, Sakamoto K, Hayakawa Y, Takahashi R, Nakata W, Sakitani K, Serizawa T, Hikiba Y, Akanuma M, Shibata W, Maeda S, Koike K. Interleukin-6 mediates epithelial-stromal interactions and promotes gastric tumorigenesis. PLoS One. 2013;8:e60914.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 54]  [Cited by in RCA: 73]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
68.  Li X, Xie G, Zhai J, He Y, Wang T, Wang Y, Shen L. Association of serum Interleukin-8 level with lymph node metastasis and tumor recurrence in gastric cancer. Front Oncol. 2022;12:975269.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Reference Citation Analysis (0)]
69.  Lee KE, Khoi PN, Xia Y, Park JS, Joo YE, Kim KK, Choi SY, Jung YD. Helicobacter pylori and interleukin-8 in gastric cancer. World J Gastroenterol. 2013;19:8192-8202.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in CrossRef: 90]  [Cited by in RCA: 95]  [Article Influence: 7.3]  [Reference Citation Analysis (1)]
70.  Li X, Zhai J, Shen Y, Zhang T, Wang Y, He Y, You Q, Shen L. Tumor-derived IL-8 facilitates lymph node metastasis of gastric cancer via PD-1 up-regulation in CD8(+) T cells. Cancer Immunol Immunother. 2022;71:3057-3070.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 1]  [Cited by in RCA: 41]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
71.  Zhai J, Shen J, Xie G, Wu J, He M, Gao L, Zhang Y, Yao X, Shen L. Cancer-associated fibroblasts-derived IL-8 mediates resistance to cisplatin in human gastric cancer. Cancer Lett. 2019;454:37-43.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 88]  [Cited by in RCA: 210]  [Article Influence: 30.0]  [Reference Citation Analysis (0)]
72.  Meier C, Brieger A. The role of IL-8 in cancer development and its impact on immunotherapy resistance. Eur J Cancer. 2025;218:115267.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 17]  [Reference Citation Analysis (0)]
73.  Yu H, Huang X, Ma Y, Gao M, Wang O, Gao T, Shen Y, Liu X. Interleukin-8 regulates endothelial permeability by down-regulation of tight junction but not dependent on integrins induced focal adhesions. Int J Biol Sci. 2013;9:966-979.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 74]  [Cited by in RCA: 122]  [Article Influence: 9.4]  [Reference Citation Analysis (10)]
74.  Liu Y, Shi Y, Han R, Liu C, Qin X, Li P, Gu R. Signaling pathways of oxidative stress response: the potential therapeutic targets in gastric cancer. Front Immunol. 2023;14:1139589.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 34]  [Reference Citation Analysis (0)]
75.  Han L, Shu X, Wang J. Helicobacter pylori-Mediated Oxidative Stress and Gastric Diseases: A Review. Front Microbiol. 2022;13:811258.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 5]  [Cited by in RCA: 50]  [Article Influence: 12.5]  [Reference Citation Analysis (0)]
76.  Pratas A, Malhão B, Palma R, Mendonça P, Cervantes R, Marques-Ramos A. Effects of apigenin on gastric cancer cells. Biomed Pharmacother. 2024;172:116251.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 10]  [Reference Citation Analysis (0)]
77.  Hashimoto K, Oshima T, Tomita T, Kim Y, Matsumoto T, Joh T, Miwa H. Oxidative stress induces gastric epithelial permeability through claudin-3. Biochem Biophys Res Commun. 2008;376:154-157.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 66]  [Cited by in RCA: 75]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
78.  Clemente JC, Ursell LK, Parfrey LW, Knight R. The impact of the gut microbiota on human health: an integrative view. Cell. 2012;148:1258-1270.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 2236]  [Cited by in RCA: 2628]  [Article Influence: 187.7]  [Reference Citation Analysis (0)]
79.  Kåhrström CT, Pariente N, Weiss U. Intestinal microbiota in health and disease. Nature. 2016;535:47.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 96]  [Cited by in RCA: 160]  [Article Influence: 16.0]  [Reference Citation Analysis (0)]
80.  Stewart OA, Wu F, Chen Y. The role of gastric microbiota in gastric cancer. Gut Microbes. 2020;11:1220-1230.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 54]  [Cited by in RCA: 132]  [Article Influence: 22.0]  [Reference Citation Analysis (0)]
81.  Park YH, Kim N. Review of atrophic gastritis and intestinal metaplasia as a premalignant lesion of gastric cancer. J Cancer Prev. 2015;20:25-40.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 164]  [Cited by in RCA: 221]  [Article Influence: 20.1]  [Reference Citation Analysis (0)]
82.  Peng L, Li ZR, Green RS, Holzman IR, Lin J. Butyrate enhances the intestinal barrier by facilitating tight junction assembly via activation of AMP-activated protein kinase in Caco-2 cell monolayers. J Nutr. 2009;139:1619-1625.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1443]  [Cited by in RCA: 1422]  [Article Influence: 83.6]  [Reference Citation Analysis (0)]
83.  Sheh A, Fox JG. The role of the gastrointestinal microbiome in Helicobacter pylori pathogenesis. Gut Microbes. 2013;4:505-531.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 127]  [Cited by in RCA: 157]  [Article Influence: 12.1]  [Reference Citation Analysis (0)]
84.  Stephens M, von der Weid PY. Lipopolysaccharides modulate intestinal epithelial permeability and inflammation in a species-specific manner. Gut Microbes. 2020;11:421-432.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 53]  [Cited by in RCA: 207]  [Article Influence: 34.5]  [Reference Citation Analysis (0)]
85.  Mattarelli P, Brandi G, Calabrese C, Fornari F, Prati GM, Biavati B, Sgorbati B. Occurrence of Bifidobacteriaceae in human hypochlorhydria stomach. Microb Ecol Health Dis. 2014;25.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 9]  [Cited by in RCA: 13]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
86.  Li S, Zhu S, Yu J. The role of gut microbiota and metabolites in cancer chemotherapy. J Adv Res. 2024;64:223-235.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 3]  [Cited by in RCA: 39]  [Article Influence: 19.5]  [Reference Citation Analysis (0)]
87.  Bik EM, Eckburg PB, Gill SR, Nelson KE, Purdom EA, Francois F, Perez-Perez G, Blaser MJ, Relman DA. Molecular analysis of the bacterial microbiota in the human stomach. Proc Natl Acad Sci U S A. 2006;103:732-737.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 730]  [Cited by in RCA: 790]  [Article Influence: 39.5]  [Reference Citation Analysis (0)]
88.  Aviles-Jimenez F, Vazquez-Jimenez F, Medrano-Guzman R, Mantilla A, Torres J. Stomach microbiota composition varies between patients with non-atrophic gastritis and patients with intestinal type of gastric cancer. Sci Rep. 2014;4:4202.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 179]  [Cited by in RCA: 270]  [Article Influence: 22.5]  [Reference Citation Analysis (0)]
89.  Dicksved J, Lindberg M, Rosenquist M, Enroth H, Jansson JK, Engstrand L. Molecular characterization of the stomach microbiota in patients with gastric cancer and in controls. J Med Microbiol. 2009;58:509-516.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 211]  [Cited by in RCA: 245]  [Article Influence: 14.4]  [Reference Citation Analysis (0)]
90.  Wang L, Zhou J, Xin Y, Geng C, Tian Z, Yu X, Dong Q. Bacterial overgrowth and diversification of microbiota in gastric cancer. Eur J Gastroenterol Hepatol. 2016;28:261-266.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 171]  [Cited by in RCA: 163]  [Article Influence: 16.3]  [Reference Citation Analysis (0)]
91.  Poniewierska-Baran A, Plewa P, Żabicka Z, Pawlik A. Claudin18.2 as a Promising Therapeutic Target in Gastric Cancer. Cells. 2025;14:1285.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 1]  [Cited by in RCA: 1]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
92.  Kubota Y, Shitara K. Zolbetuximab for Claudin18.2-positive gastric or gastroesophageal junction cancer. Ther Adv Med Oncol. 2024;16:17588359231217967.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 23]  [Reference Citation Analysis (0)]
93.  Ruan DY, Liu FR, Wei XL, Luo SX, Zhuang ZX, Wang ZN, Liu FN, Zhang YQ, Yang JW, Chen ZD, Wang YS, Wang JY, Liang XH, Wu XJ, Zheng YL, Liu J, Shi X, Kumar R, Liu W, Chen B, Zhang DS, Xu RH. Claudin 18.2-targeting antibody-drug conjugate CMG901 in patients with advanced gastric or gastro-oesophageal junction cancer (KYM901): a multicentre, open-label, single-arm, phase 1 trial. Lancet Oncol. 2025;26:227-238.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 8]  [Cited by in RCA: 23]  [Article Influence: 23.0]  [Reference Citation Analysis (0)]
94.  Liu J, Yang J, Sun Y, Gong J, Yue J, Pan Y, Sun M, Song R, Xiao X, Tazbirkova A, Ruan J, Liu Z, Liu Z, Li Z, Sheng L, Qin Y, Ying J, Yu X, Zhang J, Mou Y, Lu C, Chen P, Li S, Li J, Qu X, Deng T, Du J, Zhou A, Li E, Yuan X, Liang X, Yu W, Morris M, Luo Y, Zhao X, Guo Y, Zhou H, Shen L. CLDN18.2-targeting antibody-drug conjugate IBI343 in advanced gastric or gastroesophageal junction adenocarcinoma: a phase 1 trial. Nat Med. 2025;31:3028-3036.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 2]  [Cited by in RCA: 7]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
95.  Qi C, Liu C, Peng Z, Zhang Y, Wei J, Qiu W, Zhang X, Pan H, Niu Z, Qiu M, Qin Y, Fang W, Ye F, Li N, Liu T, Liu A, Zhang X, Hu C, Zhang J, Cui J, Lin X, Wang S, Zhang J, Lin T, Qu X, Yuan X, Gong J, Li J, Gao W, Gai L, Wang Y, Yuan D, Li Z, Shen L. Claudin-18 isoform 2-specific CAR T-cell therapy (satri-cel) versus treatment of physician's choice for previously treated advanced gastric or gastro-oesophageal junction cancer (CT041-ST-01): a randomised, open-label, phase 2 trial. Lancet. 2025;405:2049-2060.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 24]  [Cited by in RCA: 34]  [Article Influence: 34.0]  [Reference Citation Analysis (0)]
96.  Botta GP, Becerra CR, Jin Z, Kim DW, Zhao D, Lenz H, Ma H, Ween A, Acha P, Li Z, Yoon HH. Multicenter phase Ib trial in the U.S. of salvage CT041 CLDN18.2-specific chimeric antigen receptor T-cell therapy for patients with advanced gastric and pancreatic adenocarcinoma. J Clin Oncol. 2022;40:2538.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
97.  Xu H, Li W, Lv H, Gu D, Wei X, Dai H. Tandem CAR-T cells targeting CLDN18.2 and NKG2DL for treatment of gastric cancer. J Clin Oncol. 2022;40:4030.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
98.  Klempner SJ, Lee KW, Shitara K, Metges JP, Lonardi S, Ilson DH, Fazio N, Kim TY, Bai LY, Moran D, Yang J, Arozullah A, Park JW, Raizer JJ, Bang YJ, Shah MA. ILUSTRO: Phase II Multicohort Trial of Zolbetuximab in Patients with Advanced or Metastatic Claudin 18.2-Positive Gastric or Gastroesophageal Junction Adenocarcinoma. Clin Cancer Res. 2023;29:3882-3891.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 37]  [Cited by in RCA: 51]  [Article Influence: 17.0]  [Reference Citation Analysis (0)]
99.  Li J, Pan H, Liu T, Xu N, Zhang Y, Qin Y, Shi J, Liao D, Shen L, Luo S, Pan Y, Zhao W, Zheng Y, Zhuang R, Mao C, Ma Y, Wang H, Li Z. A multicenter, phase 1 study of AB011, a recombinant humanized anti-CLDN18.2 monoclonal antibody, as monotherapy and combined with capecitabine and oxaliplatin (CAPOX) in patients with advanced solid tumors. J Clin Oncol. 2023;41:391.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
100.  Teng F, Gu Y, Chai H, Guo H, Li H, Wu X, Yao X, Xu F, Shi L, Yan Z, Zi X, Dai Z, Liao T, Zheng L, Fan F, Li Z, Yang J, Qian X. Abstract 5183: The preclinical characterization of TST001, a novel humanized anti-claudin18.2 mAb with enhanced binding affinity and anti-tumor activity. Cancer Res. 2020;80:5183.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 6]  [Reference Citation Analysis (0)]
101.  Shitara K, Lordick F, Bang YJ, Enzinger P, Ilson D, Shah MA, Van Cutsem E, Xu RH, Aprile G, Xu J, Chao J, Pazo-Cid R, Kang YK, Yang J, Moran D, Bhattacharya P, Arozullah A, Park JW, Oh M, Ajani JA. Zolbetuximab plus mFOLFOX6 in patients with CLDN18.2-positive, HER2-negative, untreated, locally advanced unresectable or metastatic gastric or gastro-oesophageal junction adenocarcinoma (SPOTLIGHT): a multicentre, randomised, double-blind, phase 3 trial. Lancet. 2023;401:1655-1668.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 346]  [Cited by in RCA: 433]  [Article Influence: 144.3]  [Reference Citation Analysis (0)]
102.  Sahin U, Türeci Ö, Manikhas G, Lordick F, Rusyn A, Vynnychenko I, Dudov A, Bazin I, Bondarenko I, Melichar B, Dhaene K, Wiechen K, Huber C, Maurus D, Arozullah A, Park JW, Schuler M, Al-Batran SE. FAST: a randomised phase II study of zolbetuximab (IMAB362) plus EOX versus EOX alone for first-line treatment of advanced CLDN18.2-positive gastric and gastro-oesophageal adenocarcinoma. Ann Oncol. 2021;32:609-619.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 54]  [Cited by in RCA: 287]  [Article Influence: 57.4]  [Reference Citation Analysis (0)]
103.  Sahin U, Schuler M, Richly H, Bauer S, Krilova A, Dechow T, Jerling M, Utsch M, Rohde C, Dhaene K, Huber C, Türeci Ö. A phase I dose-escalation study of IMAB362 (Zolbetuximab) in patients with advanced gastric and gastro-oesophageal junction cancer. Eur J Cancer. 2018;100:17-26.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 32]  [Cited by in RCA: 122]  [Article Influence: 15.3]  [Reference Citation Analysis (0)]
104.  Singh P, Toom S, Huang Y. Anti-claudin 18.2 antibody as new targeted therapy for advanced gastric cancer. J Hematol Oncol. 2017;10:105.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 67]  [Cited by in RCA: 160]  [Article Influence: 17.8]  [Reference Citation Analysis (0)]