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

    I agree
Search articles by DOI, keyword, author or affiliation
Search
Advanced Search
presentation
International Journal of Oncology
Join Editorial Board Propose a Special Issue
Print ISSN: 1019-6439 Online ISSN: 1791-2423
Journal Cover
February-2026 Volume 68 Issue 2

Full Size Image

Cover Legend PDF

Sign up for eToc alerts
Recommend to Library

Journals

International Journal of Molecular Medicine

International Journal of Molecular Medicine

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

International Journal of Oncology

International Journal of Oncology

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

Molecular Medicine Reports

Molecular Medicine Reports

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

Oncology Reports

Oncology Reports

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

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine

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

Oncology Letters

Oncology Letters

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

Biomedical Reports

Biomedical Reports

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

Molecular and Clinical Oncology

Molecular and Clinical Oncology

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

World Academy of Sciences Journal

World Academy of Sciences Journal

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

International Journal of Functional Nutrition

International Journal of Functional Nutrition

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

International Journal of Epigenetics

International Journal of Epigenetics

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

Medicine International

Medicine International

An International Open Access Journal Devoted to General Medicine.

Journal Cover
February-2026 Volume 68 Issue 2

Full Size Image

Cover Legend PDF

Sign up for eToc alerts
Recommend to Library

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

  • Supplementary Files
    • Supplementary_Data.pdf
Review Open Access

Roles of interleukins in spasmolytic polypeptide‑expressing metaplasia (Review)

  • Authors:
    • Jiale Ma
    • Tiancheng Zhan
    • Xinyuan Zhang
    • Wang Gao
    • Shuangmei Zhao
    • Huizhen Li
  • View Affiliations / Copyright

    Affiliations: Department of Gastroenterology, The Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300250, P.R China, Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, P.R. China
    Copyright: © Ma et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
  • Article Number: 17
    |
    Published online on: December 3, 2025
       https://doi.org/10.3892/ijo.2025.5830
  • Expand metrics +
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Metrics: Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )
Cited By (CrossRef): 0 citations Loading Articles...

This article is mentioned in:



Abstract

Gastric cancer (GC) is a major global health burden, ranking fifth in incidence and third in cancer‑related mortality. By 2040, there are expected to be ~1.8 million new cases and 1.3 million fatalities associated with GC. Spasmolytic polypeptide‑expressing metaplasia (SPEM) is a central component of gastric precancerous lesions, which remodels the gastric mucosa in response to injury through a lineage of mucus‑secreting cells. Interleukins (ILs) are the communication means for innate and adaptive immune cells as well as non‑immune cells and tissues. Their complex network regulation contributes to the development of SPEM and is a key driver in the transformation of SPEM to GC. The present review systematically described the IL‑related mechanisms underlying the formation and progression of SPEM and categorizes the roles of different ILs by family. In addition, the molecular association between IL dynamics and SPEM following Helicobacter pylori infection is explored, and various SPEM experimental model characteristics and IL‑based therapeutic strategy advances and limitations are discussed. The clinical translation of IL‑targeted therapies is limited, but the development of therapies that target pathogenesis specifically and the enhancement of IL therapy combinations with other therapeutic options may improve efficacy and reduce side effects. Increased understanding of the causes of SPEM and the mechanisms underlying GC may open up new avenues for early detection and targeted therapy.

Introduction

Gastric cancer (GC) continues to have a high global burden, ranking third in terms of cancer-related mortality and fifth in terms of incidence (1,2). Annually, ~660,000 individuals die from GC, despite improvements in early detection and treatment methods (3); by 2040, it is estimated to increase to ~1.8 million new cases and 1.3 million fatalities (4). GC follows a multistage progression pattern driven by chronic inflammation, including atrophy, metaplasia, dysplasia and invasive carcinoma, termed the 'Correa pathway' proposed by Correa et al (5) in 1975. Current research is improving the understanding of the molecular mechanisms underlying the progression of precancerous lesions through inflammation-mediated epigenetic changes, such as microbiota dysbiosis, while therapeutic studies are investigating novel approaches to target particular pathways, such as glycosylation, in order to prevent metastasis (6,7). When a terminally differentiated cell type that is not typically found at a specific anatomical location replaces pre-existing terminally differentiated cells, this is termed metaplasia (8). In most cases, the metaplasia lineage is characterized by mucus secretion. A key characteristic of the metaplastic lineage is the remodeling of the gastric mucosa by mucus-secreting cell cohorts in response to damage (9), and mucus production is typically the distinguishing characteristic of this lineage. In the gastric mucosa, two main forms of metaplasia have been identified: i) Intestinal metaplasia (IM); and ii) spasmolytic polypeptide-expressing metaplasia (SPEM).

SPEM, also termed pseudopyloric metaplasia or mucous metaplasia, is a regenerative lesion that presents histologically as Brunner's glands or deep proventricular glands. It is distinguished by the expression of Trefoil factor 2 (TFF2), Griffonia simplicifolia lectin II (GS II) and mucin (MUC) 6 (10). SPEM formation is frequently accompanied by abnormal gastric pit hyperplasia, macrophage infiltration and glandular structure disarray. Chronic damage (such as inflammation or NSAID use) causes the gastric epithelium to ablate, which leads to the formation of SPEM during regeneration. Similar to intestinal cup cells (MUC5AC, MUC6 and MUC2), these metaplasia cells typically exhibit a distinct mucus-secreting phenotype, altering the defensive and digestive characteristics of the gastric mucosa (10,11). SPEM and IM are two distinct lineages of metaplasia cells. However, SPEM may precede IM and promote carcinogenesis via a variety of mechanisms (12). According to independent clinical studies carried out in the US, Japan and Iceland, SPEM is found in >90% of GC resection specimens, is linked to >50% of early GCs and is frequently found in the paracarcinoma or dysplasia region (13-15). Furthermore, animal models have confirmed that SPEM is an important intermediate stage in gastric precancerous lesions (16-18).

Interleukins (ILs) are potent secreted regulators of various cell types and activities. By targeting and influencing various cell signaling pathways, they aid in the development and progression of inflammatory diseases and various types of cancer. ILs also regulate processes such as cell death, proliferation, differentiation and migration, and are involved in cellular communication during homeostasis and disease (19-22). IL production and function are central regulators of SPEM development. Research has verified that the development of SPEM is closely associated with chronic inflammation and involves an imbalance in the network of pro-inflammatory/anti-inflammatory ILs, such as IL-1β, IL-10, IL-17A and IL-33. ILs bind to high-affinity receptors on the cell membrane and activate intracellular signaling pathways, including NF-κB, MAPK and JAK-STAT, resulting in gene transcription changes. This in turn regulates local cell proliferation, differentiation and reprogramming (23-26) and induces or alters specific cellular functions (27-29). The IL network may disturb the normal homeostasis of gastric mucosal cells by controlling local inflammation, cellular infiltration and multi-pathway interactions. This could lead to the development of SPEM or be a major factor in the transformation of SPEM into GC (30).

The present review summarized recent findings on IL-related mechanisms involved in SPEM formation and progression. Furthermore, the roles of various ILs in SPEM and the relationship between IL changes and SPEM after Helicobacter pylori (H. pylori) infection are described. In addition, the present review offered a comprehensive summary of the features of the different SPEM experimental models that are employed in IL studies. Finally, by describing the progress and limitations of ILs for therapeutic use, novel directions for future investigation are discussed.

Overview of SPEM

The mucosal barrier, made up of closely knit epithelial cells and a thick layer of mucus, protects the gastric epithelium from harmful agents such as ingested food, bacteria, gastric acid and digestive enzymes, stopping the stomach contents from penetrating into the underlying tissues (31). Pepsinogen-producing chief cells, mucus-producing surface foveolar cells, neck cells and acid-producing parietal cells make up the majority of the gastric mucosal barrier (32). The chief cell is a differentiated cell lineage, and a subset of it serves as 'reserve' stem cells in the gastric mucosa. During gastric injury, parietal cells become deficient due to a variety of pathogenic factors, resulting in a compensatory proliferative response of gastric stem and progenitor cells, as well as metaplasia with zymogenic chief cells that can be recruited back into the cell cycle. Metaplasia in the stomach, which results in the structural change of its glands, is considered to be an 'adaptive' process that reacts to a range of endogenous or exogenous aggressors, including pH, hormones, chemicals and microbiota changes (33). SPEM is a type of metaplasia that occurs during the healing of gastric mucosal damage. This is due to the metaplasia cells expressing spasmolytic polypeptide, also termed TFF2. Fully differentiated gastric chief cells can be reprogrammed into mucin-secreting metaplasia cells, and the presence of mature chief cells in the corpus is the source of SPEM cells via transdifferentiation. These metaplasia changes take place concurrently with or in response to parietal cell death and inflammation (34). SPEM cells are histologically localized at the base of metaplasia glands following injury, and their specificity was determined by the co-expression of molecular markers including GS II lectin, TFF2, Muc6, CD44v9 and AQP5 (35). The formation of metaplasia lineage cells, proliferation of neck cells and amplification of the small foveolar/tuft cell lineage in the gastric mucosa are characteristic pathological alterations (36,37). Additionally, excessive immune activation in the stomach can damage epithelial cells and induce the development of SPEM (38). Acute SPEM may be a healing mechanism, but when it persists in an environment of chronic inflammation, chronic SPEM is strongly associated with the development of gastric adenocarcinoma (34,38). It has been recorded that >95% of GCs are adenocarcinomas, and according to Laurén's classification, they can be divided into two types: i) Diffuse; and ii) intestinal (39). SPEM is present in almost all GC types, and research on animal models with acute oxyntic atrophy, transgenic and knockout (KO) genes, chronic infection and genetic modification indicates that SPEM is a key precancerous intermediate in the malignant transformation of gastric mucositis (9,40). Although SPEM is partially reversible, intestinal SPEM and/or IM are considered precancerous lesions that may be irreversible in the presence of chronic inflammatory stimuli (41,42). Dysplasia progression is an inevitable stage of cancer development (39). The susceptibility of gastric precancerous lesions to alterations in the expression of numerous ILs suggests that immune cells and different ILs implicated in the progression of metaplasia may be important in predicting the risk of carcinogenesis (27,39,43,44). Thus, additional identification and clarification of ILs associated with the SPEM response will greatly enhance the present understanding of gastric carcinogenesis.

IL family and SPEM-associated IL signaling

ILs are a subset of cytokines and a family of signaling proteins that are essential for the regulation and coordination of immune system responses. The family consists of small proteins secreted by different cells to facilitate cellular communication and interaction (45). At least 40 ILs have been identified, a number of which can be classified into separate families or superfamilies (46). Typically, each IL interacts with a different receptor or group of closely related receptors expressed on the target cell, and when bound, the intracellular structural domain of the receptor is activated, triggering an intracellular signaling cascade (Fig. 1). Numerous ILs are especially important for the onset and course of SPEM, and their pleiotropic effects depend on a number of factors, including dose dependency, receptor heterogeneity, cellular source diversity and signaling pathway complexity (21). In the present review, the cytokine signals associated with SPEM were systematically categorized (Fig. 2), including the IL-1 superfamily, IL-2 family, IL-6 family, IL-10 family, IL-12 family, IL-17 family and other ILs, as well as cytokines closely related to ILs (Table I).

IL family surface receptors and
intracellular signaling. The IL-2 interleukin family receptors for
IL-2, IL-15, IL-7, IL-9, IL-21 and IL-4 share a common γc subunit.
IL-2 and IL-15 also share IL-2Rβ. All γc cytokines activate JAK1
and JAK3. They phosphorylate various STAT proteins downstream by
activating the JAK/STAT signaling pathway. The IL-2 receptor has
forms with low, intermediate and high affinity and activates
JAK-STAT, PI3K and MAPK signaling pathways upon its engagement in
the specific receptor complexes. There are three forms of IL-4
receptors involving the IL-4Rα subunit, γc chain, IL-13Rα1 subunit
and the IL-2Rα subunit, which activate the PI3K and MAPK pathways.
IL-4 activates intracellular pathways to produce phosphorylated
STAT6, ERK and AKT. IL-6 has classical and trans-signaling
properties and activates multiple intracellular pathways, including
JAK1-STAT3, RAS-MAPK and PI3K-AKT. IL-1 family receptors are
heterodimers consisting of ligand-sensing and signaling monomers.
Cellular activation by stimulation of the polymerization of the
IRAK4-MyD88 complex leads to the recruitment of IRAK1/2, further
recruitment of TRAF6 and activation of MAPK/NIK, and finally
activation of the IKK complex, NF-κB, AP-1 and IRFs. IL-33/ST2
activates NF-κB, PI3K/AKT, MAPKs and ERK1/2. The six members of the
IL-17 family form homodimers and heterodimers that recruit the
ubiquitin ligase Act1 to the SEFIR structural domain, triggering
the downstream TRAF6-TAK1-NF-κB pathway, as well as activating the
C/EBP and MAPK pathways. IL, interleukin; IRFs, interferon
regulatory factors; γc, γ-chain; IRAK4, IL-1 receptor-associated
kinase; MyD88, myeloid differentiation primary response 88; TRAF6,
tumor necrosis factor receptor-associated factor 6; NIK,
NF-κB-inducing kinase; AP-1, activator protein 1; SEFIR, similar
expression to fibroblast growth factor genes/IL-17 receptors; TAK1,
TGF-β-activated kinase 1; C/EBP, CCAAT/enhancer-binding
protein.

Figure 1

IL family surface receptors and intracellular signaling. The IL-2 interleukin family receptors for IL-2, IL-15, IL-7, IL-9, IL-21 and IL-4 share a common γc subunit. IL-2 and IL-15 also share IL-2Rβ. All γc cytokines activate JAK1 and JAK3. They phosphorylate various STAT proteins downstream by activating the JAK/STAT signaling pathway. The IL-2 receptor has forms with low, intermediate and high affinity and activates JAK-STAT, PI3K and MAPK signaling pathways upon its engagement in the specific receptor complexes. There are three forms of IL-4 receptors involving the IL-4Rα subunit, γc chain, IL-13Rα1 subunit and the IL-2Rα subunit, which activate the PI3K and MAPK pathways. IL-4 activates intracellular pathways to produce phosphorylated STAT6, ERK and AKT. IL-6 has classical and trans-signaling properties and activates multiple intracellular pathways, including JAK1-STAT3, RAS-MAPK and PI3K-AKT. IL-1 family receptors are heterodimers consisting of ligand-sensing and signaling monomers. Cellular activation by stimulation of the polymerization of the IRAK4-MyD88 complex leads to the recruitment of IRAK1/2, further recruitment of TRAF6 and activation of MAPK/NIK, and finally activation of the IKK complex, NF-κB, AP-1 and IRFs. IL-33/ST2 activates NF-κB, PI3K/AKT, MAPKs and ERK1/2. The six members of the IL-17 family form homodimers and heterodimers that recruit the ubiquitin ligase Act1 to the SEFIR structural domain, triggering the downstream TRAF6-TAK1-NF-κB pathway, as well as activating the C/EBP and MAPK pathways. IL, interleukin; IRFs, interferon regulatory factors; γc, γ-chain; IRAK4, IL-1 receptor-associated kinase; MyD88, myeloid differentiation primary response 88; TRAF6, tumor necrosis factor receptor-associated factor 6; NIK, NF-κB-inducing kinase; AP-1, activator protein 1; SEFIR, similar expression to fibroblast growth factor genes/IL-17 receptors; TAK1, TGF-β-activated kinase 1; C/EBP, CCAAT/enhancer-binding protein.

ILs in the progression of SPEM. IL-18
promotes the Th1 response to induce chronic persistent inflammation
and mediates IFN-γ to generate gastritis. IL-2 induces TNF-α and
IFN-γ to promote SPEM, and high IL-2 inhibits gastric
acid-associated chronic inflammation. IL-10 inhibits gastric
tissues from entering the SPEM state. IL-1β inhibits gastric acid
secretion and induces gastric mucosal atrophy that directly
promotes SPEM development, stimulates proliferation of gastric
epithelial cells and promotes Helicobacter pylori damage to
the parietal cells, leading to gastric carcinogenesis. IL-6
triggers alterations in the SPEM spectrum, and disruption of the
pathway may favor the development of gastric adenomas. IL-11
triggers the progression of spontaneous atrophic gastritis to
locally advanced epithelial hyperplasia but does not initiate
cancer. The chronic inflammatory state caused by IL-8 may drive
SPEM gastric cancer early stage and pre-cancer progression. IL-33
activates the ST2 signaling axis to drive a Th2-type immune
response and facilitates IL-13-induced transdifferentiation of
chief cells into SPEM cells. IL-13 also promotes the maturation and
stabilization of SPEM. IL-4 suppresses inflammation by decreasing
IFN-γ. By reducing immune cells' secretion of IL-17A, IL-27
considerably lessens the severity of gastritis and SPEM. At the
same time, it signals directly to gastric epithelial cells to slow
the development of gastric metaplasia. IL-17A directly promotes the
development of chronic atrophic gastritis and SPEM and promotes
cancer progression. IL, interleukin; SPEM, spasmolytic
polypeptide-expressing metaplasia.

Figure 2

ILs in the progression of SPEM. IL-18 promotes the Th1 response to induce chronic persistent inflammation and mediates IFN-γ to generate gastritis. IL-2 induces TNF-α and IFN-γ to promote SPEM, and high IL-2 inhibits gastric acid-associated chronic inflammation. IL-10 inhibits gastric tissues from entering the SPEM state. IL-1β inhibits gastric acid secretion and induces gastric mucosal atrophy that directly promotes SPEM development, stimulates proliferation of gastric epithelial cells and promotes Helicobacter pylori damage to the parietal cells, leading to gastric carcinogenesis. IL-6 triggers alterations in the SPEM spectrum, and disruption of the pathway may favor the development of gastric adenomas. IL-11 triggers the progression of spontaneous atrophic gastritis to locally advanced epithelial hyperplasia but does not initiate cancer. The chronic inflammatory state caused by IL-8 may drive SPEM gastric cancer early stage and pre-cancer progression. IL-33 activates the ST2 signaling axis to drive a Th2-type immune response and facilitates IL-13-induced transdifferentiation of chief cells into SPEM cells. IL-13 also promotes the maturation and stabilization of SPEM. IL-4 suppresses inflammation by decreasing IFN-γ. By reducing immune cells' secretion of IL-17A, IL-27 considerably lessens the severity of gastritis and SPEM. At the same time, it signals directly to gastric epithelial cells to slow the development of gastric metaplasia. IL-17A directly promotes the development of chronic atrophic gastritis and SPEM and promotes cancer progression. IL, interleukin; SPEM, spasmolytic polypeptide-expressing metaplasia.

Table I

SPEM-related cytokine signaling.

Table I

SPEM-related cytokine signaling.

First author/s, yearCytokinesReceptorTargeted cellsMeasurement of SPEMRelated signaling pathwaysPromotes or suppresses SPEMMouse modelMethods(Refs.)
El-Zaatari et al, 2013IL-1β-Epithelial mucous cellsHistopathology scoring (0-3) and biomarkers to determine lineagesIL-6/pSTAT3PromoteC57BL/6 and Gli1+/LacZImmunofluorescence, RT-qPCR, flow cytometry and western blot(31)
Lee C et al, 2017IL-10IL10Rα and IL10RβEpithelial cellsCalculated the number of PCs and determine lineage using biomarkers-UnknownTamoxifen-induced C57BL/6H&E staining, immunofluorescence, RNA-seq and multiplex immunoassay(25)
Buzzelli JN et al, 2019IL-11IL-11RαEpithelial cellsHistopathology scoring (0-5) and biomarkers to determine lineages gp130-JAK-STAT3PromoteK19-IL11TgRT-qPCR, immunoblotting, ELISA, immunofluorescence, immunohistochemistry, flow cytometry and AB-PAS staining(104)
Howlett M et al, 2012IL-11IL-11RαEpithelial cellsHistological examination and biomarkers to determine lineagesIL-11/STAT3PromoteWild-type C57BL/6 and HKβ−/− Immunohistochemistry, immunoblotting, RT-qPCR and PAS Staining(103)
Howlett M et al, 2009IL-11IL-11RαEpithelial cellsHistological examination and biomarkers to determine lineages IL-11/STAT3/ERK1/2Promote gp130757F/F, IL-11R−/−, HK−/−, K19-C2mE and K19-Wnt/C2mEH&E/alcian blue/periodic acid-Schiff staining, qPCR, immunohistochemistry and immunoblotting(105)
Petersen CP et al, 2018IL-13IL13Rα1Chief cellsBiomarkers to determine lineagesIL-33/ST2 axis and IL-13/IL13rα1PromoteL635-treated C57BL/6FACS analysis, immunofluorescence, RNA-seq, RT-qPCR(63)
Noto CN et al, 2022IL-13IL4RαEpithelial cellsHistopathology scoring (0-4) and biomarkers to determine organoid lineagesIL-33/ST2 axis and IL-13/IL4RαPromoteGastric organoids, Tx A23 xIfng−/−, Tx A23 andTx A23 x Il13-YfpH&E/toluidine blue staining, immunofluorescence, RNA-seq, flow cytometry and RT-qPCR(144)
Meyer AR et al, 2020IL-13-Chief cellsHistological examination and biomarkers to determine lineagesIL-33/ILC2s/IL-13 axisPromoteWild-type C57BL/6 and IL-13-tdTomato reporterELISA, immunostaining, flow cytometry and FACS analysis(36)
Busada JT et al, 2021IL-13--Biomarkers to determine lineagesIL-33/ILC2s/IL-13 axisPromoteAdrenalectomy and castration C57BL/6JqRT-PCR, H&E staining, flow cytometry and scRNA-seq(38)
Contreras-Panta EW et al, 2024IL-13-SPEM cellsHistological examination and biomarkers to determine lineagesIL-13/STAT6PromoteIL-13-tdTomato reporterFlow cytometry, scRNA-seq, RT-qPCR, H&E staining, immunofluorescence and immunoblotting(39)
O'Keefe-RN et al, 2023IL-13IL13RαILC2s, Tuft cells and stem cellsBiomarkers to determine lineagesILC2-tuft cell circuitPromoteBAC(Dclk1: CreERT2), CreERT2-negative Ros 26DTA (TCWT)Flow cytometry, scRNA-seq, RT-qPCR and immunofluorescence(138)
Miska J et al, 2018IL-13IL4RαEpithelial and immune cellsHistopathology scoring (0-4) and biomarkers to determine organoid lineages IL-4/IL-13-independent hits and Tet3-mediated epigenetic dysregulationPromoteCTLA4KD IL4/IL13° and CTLA4KD IL4/IL13+Genetic ablation, flow cytometry, RT-qPCR, H&E staining and immunofluorescence(149)
Bockerstett KA et al, 2018IL-17AIL17RAPCsHistopathology scoring (0-4) and biomarkers to determine lineagesIL-17A/IL17RAPromoteTxA23 and BALB/cH&E staining, immunofluorescence and flow cytometry(34)
Bockerstett KA et al, 2020IL-27IL27RACD4+ T cellsHistopathology scoring (0-4) and biomarkers to determine lineagesIL-27/IL27RASuppressTxA23 and TxA23 x Ebi3−/−Flow cytometry, scRNA-seq, qRT-PCR, H&E staining and immunofluorescence(43)
Petersen CP et al, 2018IL-33ST2ILC2s, M2 macrophagesBiomarkers to determine lineagesIL-33/ST2 axisPromoteL635-treated C57BL/6FACS analysis, immunofluorescence, RNA-seq, RT-qPCR(63)
Jeong H et al, 2021IL-33ST2Immune cellsHistological examination and biomarkers to determine lineagesIL-33/ST2 axis, NF-κBPromoteWfdc2−/− and C57BL/6FACS analysis, immunofluorescence, H&E staining, RNA-seq and RT-qPCR(186)
Buzzelli JN et al, 2015IL-33ST2Immune regulatory cellsBiomarkers to determine lineagesERK1/2PromoteC57Bl/6 wild-typeFlow cytometry, RT-qPCR, immunofluorescence, immunohistochemistry and immunosorbent assay(60)
De Salvo C et al, 2021IL-33ST2L/IL-1R4Immune cells, M2 macrophagesHistopathology scoring and biomarkers to determine lineagesIL-33/ST2 axisPromoteSAMP x Il33−/− and C57Bl/6J wild-typeH&E/staining, AB-PAS blue staining, RT-qPCR, western blot, BrdU staining, immunohistochemistry and flow cytometry(62)
Zeng X et al, 2023IL-33ST2Immune cellsHistological examinationROS/NRF2-HO-1/NFкB axisPromote GRIM-19−/−H&E staining, RT-qPCR, western blots, cytokine ELISA plate array, immunohistochemistry and flow cytometry(28)
Li W et al, 2024IL-33ST2ILC2sHistological examination and biomarkers to determine lineagesIL-33/ILC2s/IL-13 axisPromoteRAG1 knockout and wild type C57BL/6 miceFlow cytometry, immunofluorescence and immunohistochemical staining(72)

[i] IL, interleukin; SPEM, spasmolytic polypeptide-expressing metaplasia; PC, parietal cells; gp130, glycoprotein 130; pSTAT3, phosphorylated STAT 3; RT-qPCR, reverse transcription-quantitative PCR; AB-PAS, Alcian blue-periodic acid Schiff; RNA-seq, RNA sequencing; scRNA-seq, single cell RNA-seq; BrdU, 5-bromo-2'-deoxyuridine; GRIM-19, gene-related to melanoma-19.

IL-1 super family
IL-1

IL-1 is a polypeptide synthesized and secreted by lymphocytes and monocyte macrophages and has various functions, such as participation in immune response, thermogenesis and mediation of inflammation (47). Within a 430 kilobase region, the IL-1 gene cluster on chromosome 2q comprises three related genes, IL-1A, IL-IB and IL-1RN, which encode IL-1α, IL-1β and IL-1 receptor antagonist (IL-1Ra) (27). IL-1α and IL-1β are alarm cytokines (also termed alarm proteins), both of which initiate and amplify local inflammation via the IL-1 receptor (IL-1R) (48). Additionally, IL-1β directly stimulates the development of SPEM by strongly suppressing the secretion of gastric acid and causing the atrophy of the gastric mucosa. When pathogen-associated molecular patterns (PAMPs) or damage-associated molecular patterns activate the pattern recognition receptor, IL-1β is produced as an inactive precursor (pro-IL-1β). The cleavage of pro-IL-1β into its active form by caspase-1 proteolysis requires the activation of inflammasomes, which are then released extracellularly (47). Inflammasome-mediated caspase-1 activation triggers both IL-1β release and pyroptosis, affecting inflammation progression. In addition, IL-1β stimulates the proliferation of gastric epithelial cells, which may further contribute to gastric carcinogenesis (49). Prolonged and persistent acid inhibition by IL-1β may facilitate H. pylori transfer from the gastric antrum to the corpus, endangering acid-producing parietal cells and promoting SPEM (50). IL-1Ra can also combine with the IL-1β receptor, thereby affecting the action of IL-1β.

IL-18

IL-18, a member of the IL-1 superfamily, is a pleiotropic pro-inflammatory cytokine that is important in gastric mucosal inflammation and immunomodulation (27,51). IL-18 signaling begins with a ligand binding to the receptor IL-18Rα (also termed IL-1R-related protein). When IL-18 binds to IL-18Rα, IL-18Rβ/IL-18RAcP is recruited into the complex and activates downstream signaling pathways such as NF-κB. IL-18 can dominate the gastric mucosal response to H. pylori infection by promoting T helper (Th) 1 responses and inducing chronic persistent inflammatory changes (52). Initially, it was referred to as an IFN-inducing factor that could stimulate the production of IFN-γ by splenocytes, hepatic lymphocytes and Th1 cells, as well as increase the cytotoxicity of natural killer (NK) cells and stimulate the production of IFN-γ in conjunction with IL-12 (53). The production of IFN-γ causes the death of gastric epithelial cells that express the IFN-γ receptor and is directly linked to the pathological process of SPEM (54). Although the link between IFN-γ and subsequent neoplasia remains uncertain, studies have shown that IFN-γ is crucial during the SPEM stage of precancerous lesions (55). Current studies have typically focused on IL-18-related gene polymorphisms. Research has demonstrated that IL-18RAP gene polymorphisms serve a notable role in the development of gastric precancerous lesions by controlling the IFN-γ production pathway, and may be linked to precancerous lesions or susceptibility to GC in GC high-risk populations (51,53). However, according to a different study, there was no meaningful association between the progression of gastric precancerous lesions and IL-18 promoter polymorphisms (56). Therefore, there is a lack of direct evidence that IL-18 serves a key role in gastric precancerous lesions and SPEM progression.

IL-33

IL-33, a member of the IL-1 superfamily, is a cytokine involved in tissue homeostasis, pathogenic infections, inflammation, allergies and the type 2 immune response. Through its receptor ST2 (ST2L; also termed T1/ST2, IL1RL1, T1, Der4, Fit1 or IL-33R), IL-33 forms a heterodimeric complex with IL-1 receptor accessory protein (IL-1R3). IL-33 is extensively found on the surfaces of Th2 cells, group 2 innate lymphocytes (ILC2s), mast cells and macrophages (57,58). IL-33 serves as an intranuclear transcriptional regulator, an extracellular alarmin and an immune regulation mediator. Under healthy conditions, IL-33 translocates to the nucleus and is involved in regulating gene expression (59). When necrosis or cell damage compromises the epithelial cell barrier, cells release IL-33, which also acts as an alarm protein (60,61). The immune defense and repair systems are coordinated by extracellular IL-33, which also initiates adaptive immune responses. Increased extracellular IL-33 activates expanded tissue ILC2 and Treg and promotes the recruitment and survival of immune cells (such as eosinophils, AAM, Th2 cells and basophils). These cells and signals feed back to the tissue to promote remodeling and limit inflammation. In the gastric mucosa, IL-33 is mainly secreted by damaged epithelial cells, a subset of mucous foveolar epithelial cells and M2-type macrophages (60,62,63). When there is acute inflammation and a loss of gastric mucosal parietal cells, macrophages infiltrate and gastric pit epithelial cells release IL-33 to synchronize immune defense and repair processes. IL-33 also promotes epithelial proliferation through activation of the MAPK pathway (ERK1/2, JNK and p38) (64). The IL-33/IL-13 axis is critical for gastric metaplasia and gastric epithelial repair, and several studies have linked reduced IL-33 in TFF2-deficient mice to defective metaplasia development and delayed downstream gastric epithelial repair (60,65,66). Following parietal cell loss, IL-33 stimulates the ST2 signaling axis, triggers a Th2-type immune response, promotes the release of cytokines such as IL-13 and causes the transdifferentiation of gastric chief cells into SPEM cells (63). IL-33- or ST2-KO mice are unable to form SPEM after L635-induced acute injury, suggesting that appropriate ILs are required to promote transdifferentiation as part of the gastric epithelial repair process (63,67). IL-33 also enhances ILC2 activity by upregulating GATA3 signaling, drives the gastric mucosal Th2 response and synergistically promotes goblet cell proliferation with IL-25 (68). Furthermore, IL-33 activates ILC2 in combination with IL-25 released by Tuft cells, increasing type II cytokines such as IL-4, IL-5, IL-9 and IL-13 (57,63,69). Among these cytokines, IL-13 and IL-4 mediate M2 macrophage activation through the co-receptor IL-4Rα (70). M2 macrophages and eosinophils in turn produce IL-13 and IL-33, self-sustaining the feed-forward cycle and stimulating mast cell activity (71,72). The resulting downstream release of IL-13 facilitates the progression of SPEM. A series of IL-33-mediated signaling events that promote SPEM progression are centrally involved, and persistent inflammation of the gastric mucosa can further lead to additional IM and carcinogenesis (68). Long-term and high levels of IL-33 cause M2 macrophage polarization, eosinophil proliferation and severe infiltration into the stomach mucosa, resulting in the maintenance of Th2-driven chronic inflammation (73). This increases the risk of cancer and promotes metaplasia to transform into GC (60,62,64,68). Given that IL-33 stimulates epithelial proliferation and metaplasia whilst also inducing and maintaining chronic Th2-driven inflammation, it is a key mediator of SPEM/intestinal SPEM manifestations.

IL-2 family (common γ-chain)
IL-2

IL-2 is a pro- and anti-inflammatory cytokine produced primarily by activated T cells that regulates immune response and anti-tumor immunity. The IL-2 receptor consists of three subunits: i) IL-2Rα (CD25); ii) IL-2Rβ (CD122); and iii) common γ-chain (γc; CD132). The IL-2Rαβγc heterotrimer is constitutively expressed on Tregs and ILC2s and has the highest affinity for IL-2. In the context of chronic or acute inflammation, mucosal cells proliferate at an accelerated rate to promote the repair of damaged epithelium (74). H. pylori-associated gastritis is closely related to IL-2, and the DMP-777 model shows that parietal cell deficiency is sufficient to trigger SPEM, but inflammation is critical for further differentiation and the development of SPEM (75,76). Studies into the inflammatory component in mice have shown that the progression of SPEM to developmental abnormalities requires a Th1-dominated inflammatory response (77,78). Th1 cells characteristically secrete IFN-γ and IL-2 (79). Th1 cells in H. pylori-infected individuals produce large amounts of IL-2, which induces T cell differentiation and immune memory (80,81). IL-2 is considered to provide a pathological basis for gastric carcinogenesis and SPEM by suppressing the secretion of gastric acid and modifying Th1 immune responses. A previous study found that the Th1-specific cytokines IL-2 and IFN-γ directly inhibit acid secretion in mice, which can lead to gastric atrophy and promote the development of precursor lesions to GC (82). However, there is no direct experimental evidence that indicates that IL-2 is directly related to SPEM differentiation and maturation. Instead, IL-2 primarily mitigates tissue damage by reducing gastric acid secretion and thus acid-related chronic inflammation (83). In summary, IL-2 serves an important role in SPEM progression by regulating the intensity of inflammation and the immune microenvironment, but its specific signaling pathways and interactions with other cytokines still need further exploration.

IL-4

IL-4 is an important immunomodulatory cytokine involved in the cascade transduction of several cytokines. Its signaling is initiated through two heterodimeric receptor complexes: i) Type I (IL-4Rα/γc) specifically bound to IL-4; and ii) type II (IL-4Rα/IL-13Rα1) bound to both IL-4 and IL-13, activating STAT6, JNK or IRS2 signaling pathways that drive macrophage polarization towards the alternative activation phenotype (M2a) (70,84). Activated JAK phosphorylates IL-4Rα tyrosine residues, creating binding sites for STAT6, which dimerizes and translocates to the nucleus to activate gene transcription (85). IL-4 also maintains immune homeostasis by balancing M1/M2 macrophage polarization via STAT1/3 and STAT6 synergistic regulation (84,86). In gastric mucosal inflammation, IL-4 acts as an anti-inflammatory factor to inhibit inflammation and atrophy by reducing IFN-γ. It inhibits the secretion of pro-inflammatory cytokines, such as IL-1, IL-6 and TNF-α, and regulates lymphocyte differentiation and the tumor microenvironment (84). Due to the structural similarity of the surface receptors, in a variety of diseases IL-4 acts predominantly in conjunction with another type II cytokine, IL-13 (84). IL-4 and IL-13 activate macrophages by alternating, a process that is also an important step in the progression of SPEM (70,86-88). IL-4 also serves a central role in the Th2 cell phenotype, with studies revealing that sequential activation of STAT5 and STAT6 enhances the subsequent response to IL-4, ultimately leading to Th2 T cell differentiation (89,90). Studies have shown that IL-4 also inhibits the growth of GC cells, and a notable increase in serum IL-4 levels has also been reported in patients with GC (85,91,92). IL-4 is considered to be associated with the repair of epithelial damage in the early stages, and the mechanism of its association with cancer progression in the late stages has not yet been demonstrated. Furthermore, a study demonstrated that IL-4 notably increased M2 macrophage differentiation in a diabetic mouse model, indicating that it may serve a similar role in gastric mucosal injury repair (57).

IL-6 family
IL-6

IL-6, a pleiotropic cytokine, is secreted by immune cells (such as monocytes, macrophages and lymphocytes) and non-immune cells (such as endothelial cells, intestinal epithelial cells, tumor-associated fibroblasts and tumor cells) (93-95). IL-6 regulates inflammatory mediators and endocrine responses, and serves as a link between innate and adaptive systems in host defense mechanisms (96). IL-6 signaling necessitates binding to the specific receptor IL-6R or IL-11R, which then binds to the common signal transduction receptor subunit glycoprotein 130 (gp130), causing its recruitment and homodimerization, eventually leading to the activation of the JAK/STAT and SHP2/Ras/MAPK/ERK1-2/activator protein 1 (AP-1) signaling cascades (97-99). In gastric mucosal lesions, IL-6 regulates gastric mucosal homeostasis by regulating TFF1 and mucosal proliferation, inflammation, angiogenesis and apoptosis-related mediators, and its signaling regulates gastric mucosal homeostasis by balancing SHP2/Ras/ERK and STAT3. Elevated IL-6 levels were noted in chronic inflammatory conditions, and the downregulation of TFF1 tumor suppressor activity was caused by the absence of ERK/AP-1 signaling downstream of gp130. This, in turn, results in the activation of constitutive and oncogenic STAT3, which inhibits apoptotic processes and promotes cellular proliferation and inflammatory responses. These factors ultimately lead to the development of mucosal atrophy, metaplasia, TFF2-associated changes in the SPEM lineage, aberrant cell proliferation and the progression to dysplasia and submucosal invasion (98). When IL-6 family signaling pathways are interfered with, increased STAT3 signaling may promote the growth of gastric adenomas, while increased SHP2/ERK2 signaling may result in chronic mucosal inflammation (100). Therapeutic strategies targeting the IL-6/JAK/STAT3 axis, such as the JAK inhibitor WP1066, have been shown to inhibit STAT3 phosphorylation and reduce pro-inflammatory factors (IL-6, IL-11 and IL-1β) and growth factor expression, thereby blocking tumor proliferation and inducing apoptosis (101). Nonetheless, there is insufficient evidence to support a causal role for IL-6 in cancer progression, implying that this IL may serve a protective rather than pathogenic role in the stomach (102).

IL-11

IL-11 belongs to the IL-6 family and is a pleiotropic cytokine. It is a key mediator of inflammation in the gastrointestinal tract, influencing the activity of numerous immune cell populations such as macrophages, mast cells, dendritic cells (DCs) and lymphocytes (103). IL-11 initiates signal transduction by binding to IL-11 receptor α, which recruits the signal transduction receptor gp130. By extending stem cell survival, decreasing apoptosis and promoting mitosis, IL-11 can impact epithelial homeostasis and is crucial for gastric injury, mucosal repair and cancer progression (104). Chronic IL-11 elevation causes severe fundus damage, including inflammation, metaplasia, loss of chief and parietal cells and increased proliferation, all of which are closely linked to chronic atrophic gastritis in humans (103). IL-11 has been shown to mediate the hyperactivation of STAT3 by gp130 in a gp130F/F mouse model of gastric tumorigenesis (105,106). Overexpression of IL-11 in the stomach caused by mutations in the gp130-JAK-STAT3 pathway can cause spontaneous atrophic gastritis to progress to locally advanced epithelial hyperplasia, but not dysplasia or cancer (104). Furthermore, alongside IL-11-induced gastric atrophy, there is a modest influx of polymorphonuclear cells, accompanied by elevated expression of IL-1β and IL-33 (103). IL-33 modulates Th1/Th2 cytokine balance in epithelial cells (107), whereas IL-1β regulates the expression of HK-ATPaseα subunit 64 and suppresses gastrin-dependent acid secretion (108,109). Notably, IL-11 can directly activate GC cells, thereby driving the development of an invasive phenotype (110), and is upregulated in both mouse and human GC as well as preneoplastic mucosa (111).

IL-10 family

IL-10 is an important anti-inflammatory cytokine and serves a key role in immunomodulation by inhibiting the synthesis of pro-inflammatory cytokines such as IL-1β, IL-6, IL-12 and TNF-α (112-114). IL-10 activity is mediated by heterodimeric IL-10 receptors (IL-10R1 and IL-10R2) (112). By preventing macrophage-mediated inflammatory responses and limiting pro-inflammatory signaling pathways such as NF-κB, IL-10 facilitates tissue repair (112,113). In a tamoxifen-induced SPEM model of gastric mucosa, IL-10 expression was notably reduced 3 days after administration and coincided with a histological loss of ~90% of parietal cells in the gastric mucosa. IL-10 levels were restored after 10 and 21 days, accompanied by histological normalization, suggesting that it may be involved in mucosal repair by modulating parietal cell function (25). Deficits in IL-10 can also result in the development of precancerous microenvironments and chronic inflammation (42). In addition, intracellular IL-10 levels are significantly elevated in patients with advanced GC (115). IL-10 present in tumor-associated macrophages creates an immune-evasive microenvironment (116), and elevated IL-10 levels are associated with GC metastasis and poor prognosis (117). The regulatory mechanism of IL-10 on SPEM-related carcinogenesis is still being investigated (42); however, IL-10 is regarded as a key target for intervention in gastric metaplasia and carcinogenesis (25,118).

IL-12 family

IL-27, a heterodimeric cytokine composed of p28 and EBI3, has a receptor consisting of IL-27Rα and gp130 and is widely expressed in immune cells such as T cells (43). Studies have shown that IL-27 serves a key regulatory role in chronic gastritis and gastric mucosal metaplasia. IL-27-deficient mice progress more rapidly during gastric carcinogenesis, as evidenced by earlier and faster development of severe gastric atrophy, metaplasia and inflammatory infiltrates, whereas exogenous IL-27 treatment notably reduces the severity of gastritis, atrophy and SPEM (43,119). IL-27 protective effects are primarily twofold: i) Reduces immune cell secretion of IL-17A; and ii) Transmits signals directly to gastric epithelial cells, slowing the progression of gastric metaplasia and possibly promoting reparative metaplasia (120). Rapid disease progression and notable variations in the degree of STAT signaling pathway activation in gastric epithelial cells are linked to increased Th17 cell proportion and pro-inflammatory cytokine levels (IL-6, G-CSF, MIP-2 and IL-5) in IL-27-deficient models (119,121,122). Notably, IL-27 largely controls metaplasia linked to chronic inflammation and has no discernible impact on metaplasia brought on by acute injury (122). Based on this evidence, IL-27 may serve a crucial protective role in preventing gastric carcinogenesis and SPEM by regulating immune cell activity and epithelial cell response.

IL-17 family

The IL-17 cytokine family consists of six members [IL-17A, IL-17B, IL-17C, IL-17D, IL-17E (IL-25) and IL-17F] (123-125). IL-17A (also commonly referred to as IL-17) has been extensively studied and serves a crucial role in host defense against microbes and in the development of inflammatory diseases (126-130). IL-17 is derived from a wide range of sources, including adaptive immune cells (such as CD8+ T cells and γδ T cells) and innate immune cells or myeloid cells (NK cells, macrophages, DCs, neutrophils and eosinophils) (126,131-135). The receptor for IL-17A consists of two protein monomers: i) IL-17 receptor A; and ii) IL-17 receptor C. The IL-17 receptor complex is expressed on numerous cell types, including various types of epithelial cells (34). IL-17A promotes inflammation by upregulating cellular adhesion molecules such as vascular cell adhesion molecule-1 and intercellular adhesion molecule-1, activating downstream signaling pathways (including NF-κB, MAPKs and JAK2/STAT3), and stimulating the production of cytokines and chemokines (including IL-1, IL-6 and monocyte chemotactic protein-1) (123,136). IL-17A is a primary cause of mural cell atrophy and metaplasia in chronic atrophic gastritis. Overproduction of IL-17A in the setting of chronic inflammation directly causes parietal cell apoptosis and accelerates the process of gastric mucosal atrophy and metaplasia, whilst neutralization of IL-17A successfully inhibits the aforementioned pathological alterations (34,137). Patients with gastro-IM have increased IL-17 levels, indicating that a prolonged Th17 response may occur prior to cancer formation. The gastric epithelium may undergo physiological and morphological changes as a result of a persistent inflammatory state or immune response in the gastric region. This increases the risk of tumor transformation due to atrophy and decreased gastric acid levels (72). By upregulating the matrix metalloproteinases MMP2/MMP9, disrupting the extracellular matrix and activating the JAK2/STAT3 signaling pathway, IL-17A promotes the formation of the tumor microenvironment by increasing the migration and invasion of cancer cells (132). According to clinical evidence, patients with GC had significantly higher serum IL-17A levels, which may indicate that the protein could be used as a diagnostic marker (133). In summary, cytokine networks control the proinflammatory and procarcinogenic effects of IL-17A, which is pleiotropic in gastric mucosal inflammation, metaplasia and carcinogenesis.

IL-25

IL-25, also termed IL-17E, is a member of the IL-17 cytokine family. IL-25 is an important regulator of the type 2 immune response, and its increased expression is closely related to the metaplasia microenvironment after parietal cell loss (138). Its receptor is expressed on the surface of ILC2 cells, and when the gastric mucosa is severely injured, surface mucus cells produce IL-33 and IL-25, which stimulate ILC2 to secrete IL-13 together (Fig. 3). IL-13 is an important driver of the reprogramming of chief cells into SPEM cells and the development of pyloric metaplasia. Cluster cells further activate ILC2 by secreting IL-25, forming a signaling loop (138,139). Notably, IL-25 also indirectly triggers epithelial cell proliferation by inducing IL-13 production in ILC2 (139). The IL-25/ILC2 axis is implicated in the development of metaplasia, but it also facilitates the malignant progression of metaplasia to tumor cells. According to the aforementioned findings, inhibiting IL-25 signaling or ablation of tuft cells or ILC2 has notable therapeutic potential in both the early (gastric metaplasia) and advanced stages of the tumor trajectory.

IL-33, IL-25, IL-13 and IL-17 are
associated with the onset and progression of gastric epithelial
metaplasia as SPEM. Epithelial injury and parietal cell apoptosis
lead to the release of IL-33, which causes macrophage polarization
toward M2. IL-25 released by tuft cells along with IL-33 also
contributes to the potential induction of ILC2 activation and
proliferation. M2 macrophages and eosinophils in turn produce IL-13
and more IL-33. B cells, Th2 cells and mast cells also produce
IL-13. The development of SPEM is aided by the downstream release
of IL-13 and IL-17, which are produced by various T cells and NK
cells. IL, interleukin; SPEM, spasmolytic polypeptide-expressing
metaplasia; NK, natural killer; Th, T helper cell.

Figure 3

IL-33, IL-25, IL-13 and IL-17 are associated with the onset and progression of gastric epithelial metaplasia as SPEM. Epithelial injury and parietal cell apoptosis lead to the release of IL-33, which causes macrophage polarization toward M2. IL-25 released by tuft cells along with IL-33 also contributes to the potential induction of ILC2 activation and proliferation. M2 macrophages and eosinophils in turn produce IL-13 and more IL-33. B cells, Th2 cells and mast cells also produce IL-13. The development of SPEM is aided by the downstream release of IL-13 and IL-17, which are produced by various T cells and NK cells. IL, interleukin; SPEM, spasmolytic polypeptide-expressing metaplasia; NK, natural killer; Th, T helper cell.

Other ILs
IL-8

IL-8, also termed CXCL8, belongs to the CXC chemokine family. By binding to the G protein-coupled receptors CXCR1 and CXCR2, it activates downstream signaling pathways that contribute to angiogenesis, the inflammatory response and the growth of tumors (140). Its expression is influenced by inflammatory factors (such as TNF-α and IL-1β), environmental stress (including hypoxia and chemotherapy) and hormones (including androgens and dexamethasone), and is associated with a poor cancer prognosis (140-142). IL-8 was first identified for its neutrophil chemotactic and degranulation effects, but its pro-inflammatory properties and link to tumor metastasis are especially important in GC (143). The chronic inflammatory state caused by IL-8 may accelerate the progression of early pre-cancerous lesions in GC, but no study has directly demonstrated a link between IL-8 and SPEM.

IL-13

IL-13 is the central cytokine of the Th2 immune response, mediating local tissue effects in gastritis such as chemokine secretion, goblet cell proliferation, mucus production and smooth muscle changes (84). There are six immune cell types that secrete IL-13, and in autoimmune gastritis models, mast cells produce the majority of them. They also penetrate the gastric mucosa during chronic inflammation (138,144,145). Another key source of IL-13 is ILC2s, which are also found in inflammatory stomach tissue. IL4Rα, IL13Rα1 and IL13Rα2 are three heterodimeric receptors that deliver IL-13 signaling (146,147). The process involves JAK signaling transducers (including JAK1 and JAK3) and the activator of transcription (STAT) pathway (63,144). The most important of these is STAT6, where IL-13 induces STAT6 phosphorylation and gene transcriptional regulation to propel chief cell transdifferentiation toward the SPEM phenotype (39,144,147). The gastric epithelium expresses IL-4Rα, and IL-13 affects epithelial cells directly, which leads to the expansion of mucus neck cells and the development of SPEM. Furthermore, IL-13 can directly control chief cells through IL-13Rα. In gastric metaplasia, its importance is mainly associated with mucus hypersecretion, a specific feature of SPEM. IL-13 is a core effector molecule of the IL-33/ILC2s/IL-13 axis (Fig. 4), and after chemically-induced acute parietal cell loss, ILC2 is the major source of IL-13. In conjunction with amphiphysin and IL-4, IL-13 promotes the recruitment of eosinophils and the activation of M2-type macrophages, as well as the induction and growth of SPEM (36,39). By contrast, ILC2 depletion or ablation notably inhibits metaplasia induction, cell proliferation and macrophage infiltration (36,38). Mice lacking the IL-33 receptor (ST2) are unable to develop SPEM following L635 induction. However, exogenous recombinant IL-13 aids in the redevelopment of SPEM, and neutralizing IL-13 notably reverses the metaplasia in gastritis mice (62,144). This suggests that IL-13 serves an important role in the pathogenesis of SPEM, with IL-33 acting only as its upstream inducer (63,148).

IL-33/ILC2s/IL-13 axis regulates the
development of spasmolytic polypeptide-expressing metaplasia. When
mucosal damage or parietal cell loss occurs, IL-33 is released and
interacts with the IL-33 receptor (ST2) on type II innate
lymphocytes (ILC2s). IL-13 signaling downstream of IL-33 promotes
SPEM formation and the selective activation of macrophages to M2.
Activated macrophages generate IL-33, which may further increase
IL-13 release and aid in chief cell transdifferentiation into SPEM
cells. IL, interleukin; SPEM, spasmolytic polypeptide-expressing
metaplasia.

Figure 4

IL-33/ILC2s/IL-13 axis regulates the development of spasmolytic polypeptide-expressing metaplasia. When mucosal damage or parietal cell loss occurs, IL-33 is released and interacts with the IL-33 receptor (ST2) on type II innate lymphocytes (ILC2s). IL-13 signaling downstream of IL-33 promotes SPEM formation and the selective activation of macrophages to M2. Activated macrophages generate IL-33, which may further increase IL-13 release and aid in chief cell transdifferentiation into SPEM cells. IL, interleukin; SPEM, spasmolytic polypeptide-expressing metaplasia.

Existing research on the role of IL-13 in the progression of SPEM to malignancy takes two opposing perspectives. A previous study suggested that IL-13 mainly promotes the maturation and stabilization of SPEM. By activating the IL-13/STAT6 axis, it causes STAT6 phosphorylation in SPEM cells and boosts mucin production, resulting in a mature and proliferative SPEM cell phenotype (39). Another study reported the potential pro-cancer mechanism of IL-13; ILC2-derived IL-13 promotes the growth and secretion of IL-25 by gastric mucosal tufted cells, which in turn triggers ILC2 activity. This creates an IL-13/IL-25 feedback loop and speeds up the development of cancer from SPEM (138). Simultaneously, TNF+ Tregs release IL-13, which activates STAT3, directly promoting the malignant biologicals of cancer cells (149). In addition, the increase of the Th2 response and IL-4/IL-13 in chronic inflammatory environments can trigger Tet3-mediated epigenetic disorders. This process causes changes in gene expression programs, which initiate epithelial transformation and differentiation of precancerous cells (149). In conclusion, interventions targeting IL-13 may be expected to alleviate gastrointestinal tumor precursors and possibly reverse the development of gastric metaplasia.

IL-32

IL-32 was first discovered in 1992 in NK cells activated by IL-2 (150). Due to its then unknown function, it was termed the N-terminal and four kringle structural domain of the hepatocyte growth factor protein (NK4) (151,152). Of the nine IL-32 isoforms that have been identified, IL-32α is the most widely expressed, IL-32γ has the highest biological activity and IL-32β has the highest genetic homology to humans (153). IL-32 is mainly expressed by NK cells, monocytes, epithelial cell lines and T cells. The structure of IL-32 is unique in that it lacks any specific cell surface receptors involved in extracellular signaling and is hypothesized to exert its effects through a number of intracellular molecular interactions (154). The expression of IL-32 is highly increased in the gastric mucosa of patients with GC and is rare in healthy gastric mucosa, but the increased levels of IL-32 are mainly confined to the cytoplasm of gastric epithelial cells in patients with gastritis and GC (152). Internal infection causes the synthesis and secretion of various pro-inflammatory and anti-inflammatory cytokines, resulting in the formation of a tumor microenvironment in GC cells. These microenvironments are constantly involved in the synthesis of IL-8, which has a biological effect by triggering the synthesis of IL-32 (154). It not only promotes the development of inflammation, but also induces the expression of a number of potent inflammatory cytokines. Further studies are needed to investigate the different pathways regulated by IL-32 and its role in SPEM.

ILs and H. pylori infection SPEM

H. pylori is a spiral, flagellated, microaerophilic, gram-negative bacillus that colonizes the gastric mucosa and is the leading cause of chronic gastritis, peptic ulcers and cancer (75) Although most H. pylori live freely in the mucous layer, a small portion can adhere to mucosal epithelial cells (125). Its interaction with the epithelial cells of the gastric body triggers an immune response that leads to the production of several cytokines and the establishment of chronic inflammation (68). In this process, ILs induce the inflammatory process by recruiting neutrophils, lymphocytes, macrophages and DCs (155). As a major carcinogen, H. pylori is widely recognized to be associated with mucosal barrier disruption (156). During H. pylori infection, the gastric epithelium enlarges the mucous cell compartment at the expense of parietal cells and chief cells, thereby reducing acid secretion (13,31). H. pylori-dependent activation of different signaling cascades induces upregulation of pro-inflammatory ILs and induces morphological rearrangement of epithelial cells, leading to chronic gastritis, which progresses from atrophy to antispasmodic polypeptide expression and IM, dysplasia and ultimately to cancer (125). Patients with H. pylori infection have a much higher prevalence of SPEM compared with patients without the infection, and 6 months after H. pylori eradication therapy, SPEM declines as inflammation improves (157).

Following H. pylori infection, chronic inflammation progressively worsens and alters the dynamics of ILs (Fig. 5). H. pylori cytotoxins (VacA, CagA, CagL and GGT) and PAMPs (flagella and lipopolysaccharide) in the gastric mucosa activate antigen-presenting cells. These cells in turn activate Th1 and Th17 cells through the production of IL-12 and IL-23, respectively (68). The IL-2 and IFN-γ released by Th1 cells promote B-cell differentiation and the secretion of anti-H. pylori IgG, while the IL-32 generated by epithelial cells causes inflammation and chemokines (68). Cytotoxin-associated antigen A (CagA) is commonly acknowledged as the primary virulence determinant of H. pylori among the aforementioned virulence factors. It is delivered to the epithelial cytoplasm through the type IV secretion system and regulates multiple signaling pathways to exert its pathogenic effects. IL-6 expression is strongly linked to H. pylori infection, and CagA+ H. pylori stimulates transcriptional activator-3 and signaling transducer by altering the gp130 signaling switch, causing aberrant activation that leads to aberrant proliferation and gastric epithelial cell metaplasia (100). IL-18 and IL-1β activate Treg cells in CagA+ H. pylori-infected individuals, modifying the immune response and resulting in a persistent infection. Bacterial density and the virulence factor have a direct relationship with IL-8 expression levels. In vitro studies have demonstrated that CagA and/or cytotoxin-associated gene pathogenicity island activate the NF-κB pathway, leading to increased IL-8 or IL-32 levels and anti-apoptotic responses. The gastric sinusoidal mucosa of patients with CagA+ H. pylori produces increased levels IL-8 and IL-1α, which attracts inflammatory cells locally and promotes mucosal damage and chronic inflammation (79,143,158). In summary, the intricate relationship between virulence factors and host immune cells ultimately triggers transcription factors and inflammatory pathways, which in turn increases the synthesis of additional cytokines.

Dynamics of H. pylori
infection and ILs. After infection, IL-8 expression activates and
recruits neutrophils to the gastric mucosa to establish an innate
immune response against H. pylori infection. Dendritic cell
secretion produces IL-12 and IL-23 to activate Th1 and Th17 cells,
respectively and Th1 cells release IL-2 and IFN-γ, which stimulate
B-cell differentiation and anti-H. pylori IgG secretion.
Synergistic IL-17A is increased by IL-23, and when combined with
IL-22, it causes IL-8 secretion and intensifies the inflammatory
response. The virulence factor CagA triggers upregulation of IL-8
or IL-32. Treg cells are activated by IL-18 and IL-1β to modulate
the immune response and lead to persistent infection. Infection
leading to massive production of IL-1β, IL-6 and IL-11 may
exacerbate mucosal damage, inhibit gastric acid secretion and
promote chronic inflammation. DC-produced IL-10 and TGF-β inhibit
host immune response through Treg differentiation. Below right is
the structure of H. pylori. IL, interleukin; SPEM,
spasmolytic polypeptide-expressing metaplasia; Th, T helper cell;
Treg, T regulatory cell; CagA, Cytotoxin-associated antigen A; DC,
dendritic cell; Helicobacter pylori, H. pylori; VacA,
vacuolating cytotoxin A.

Figure 5

Dynamics of H. pylori infection and ILs. After infection, IL-8 expression activates and recruits neutrophils to the gastric mucosa to establish an innate immune response against H. pylori infection. Dendritic cell secretion produces IL-12 and IL-23 to activate Th1 and Th17 cells, respectively and Th1 cells release IL-2 and IFN-γ, which stimulate B-cell differentiation and anti-H. pylori IgG secretion. Synergistic IL-17A is increased by IL-23, and when combined with IL-22, it causes IL-8 secretion and intensifies the inflammatory response. The virulence factor CagA triggers upregulation of IL-8 or IL-32. Treg cells are activated by IL-18 and IL-1β to modulate the immune response and lead to persistent infection. Infection leading to massive production of IL-1β, IL-6 and IL-11 may exacerbate mucosal damage, inhibit gastric acid secretion and promote chronic inflammation. DC-produced IL-10 and TGF-β inhibit host immune response through Treg differentiation. Below right is the structure of H. pylori. IL, interleukin; SPEM, spasmolytic polypeptide-expressing metaplasia; Th, T helper cell; Treg, T regulatory cell; CagA, Cytotoxin-associated antigen A; DC, dendritic cell; Helicobacter pylori, H. pylori; VacA, vacuolating cytotoxin A.

Overproduction of IL-1β during H. pylori gastric infection can lead to mucosal damage, chronic inflammation and increased risk of tumorigenesis by inhibiting gastric acid secretion, inducing GC-related gene methylation and promoting angiogenesis (48,159). IL-2 gene polymorphisms are associated with an increased risk of developing GC, and they may also promote gastric mucosal atrophy by increasing inflammatory responses (75). Notably, high IL-2 production may enhance the immune response to H. pylori eradication, reducing gastric mucosal inflammation. In the gastric mucosa of patients infected with H. pylori, studies have revealed a marked upregulation of IL-6 and IL-11 expression. This is associated with persistent phosphorylation of STAT3 and ERK1/2, which leads to abnormal proliferation of the gastric epithelium and the development of SPEM (100,160,161). IL-8 is considered to be one of the most important cytokines associated with the immune response and inflammatory process against H. pylori. Increased expression of IL-8 was detected in patients with H. pylori infection and gastric diseases (27). Infection can cause gastric mucosal epithelial cells to secrete IL-8 by activating EPK and MAPK chemoattracting inflammatory cytokines to the local gastric mucosa and promoting H. pylori adhesion to the mucosa, eventually leading to chronic inflammation and continuous circulation (143). H. pylori-infected gastric stromal cells control the secretion of IL-23 by DCs and monocytes, which in turn stimulates the expression of IL-17 and Th17 cell differentiation (162). In addition, bacterial infection induces the transformation of naive T cells or tissue-resident T cells in gastric tissue into Th17 cells, which recruit inflammatory cells by secreting IL-17A (163). By triggering the ERK 1/2 MAP kinase pathway, IL-17 can also trigger the release of IL-8, which chemoattracts neutrophil infiltration and intensifies the inflammatory response (125). In addition, IL-17A can also synergize with IL-22 to enhance neutrophil recruitment and further aggravate chronic inflammation after H. pylori infection (163). Treg cells secrete TGF-β1 and IL-10 to inhibit the Th17-mediated inflammatory response, and the imbalance between the two impacts gastric inflammation (114,162). IL-10 is also secreted by gastric mucosal tissue and protects the mucosal barrier by regulating mucosal homeostasis and preventing excessive inflammatory reactions caused by H. pylori (25,112). IL-17A inhibits Th1 differentiation and inhibits H. pylori-induced gastric mucosal damage (136). However, it also activates NF-κB signaling and increases IFN-γ levels in the gastric mucosa, which promotes cancer (163). By activating the NF-κB pathway, infection raises the expression of pro-inflammatory cytokines (IL-1β, IL-6, IL-8 and TNF-α), which increases the risk of damaged DNA and tumorigenesis (164). Chronic inflammation and immune responses caused by infection recruit M2 macrophages, which release IL-33, accelerating the progression of SPEM to malignancy (60,156). These mechanisms suggest that H. pylori disrupts the IL network via multiple pathways, leading to chronic inflammation, SPEM and malignant transformation.

SPEM models in IL research

Animal models and gastric organoids are the two most common types of SPEM models used in IL research. There are three types of animal models: i) Drug-induced models; ii) transgenic mouse models; and iii) H. pylori infection models (Table II). Specific drugs cause SPEM by directly inducing parietal cell death, making it an ideal model for studying specific stages of the metaplasia process due to its rapid and precise action. The drugs commonly used to induce SPEM are DMP-777, L635 and tamoxifen. As a leukocyte elastase protease inhibitor, DMP-777 was administered for 10-14 days to induce gastric oxidative atrophy and metaplasia, accompanied by mild inflammation (165). Its analog L635 rapidly induces parietal cell loss in an inflammatory environment, and treatment for 3 days can trigger inflammatory infiltration and proliferative SPEM (advanced SPEM). Alterations in its lineage are strikingly similar to those seen following 6-12 months of H. pylori infection (166-168). Tamoxifen, as a chemotherapeutic drug, can rapidly induce SPEM formation through a selective estrogen receptor-independent pathway within 3 days following oral or intraperitoneal injection (169). The drug causes proton leakage by damaging cells rich in proton pumps and mitochondria (such as parietal cells), reducing mitochondrial phosphorylation efficiency and interfering with intracellular pH homeostasis. Its resulting loss of parietal cells can be reversed by the proton pump inhibitor omeprazole, confirming that its action is dependent on active acid secretion (170). Therefore, tamoxifen-induced SPEM is considered a reversible precursor of precancerous lesions (42,165,169,170). In the autoimmune transgenic TxA23 mouse model, CD4+ T cells cause chronic gastritis by targeting H+/K+ ATPase and inducing apoptosis of gastric parietal cells. This mimics a number of aspects of human atrophic gastritis and metaplasia, including chronic inflammation and parietal cell atrophy, mucus neck cell proliferation, SPEM and, eventually, gastric intraepithelial tumors (34). Focal SPEM lesions were observed in 2-month-old mice, while extensive and severe SPEM lesions were observed in 12-month-old mice. SPEM was also highly proliferated in H. felis-infected mice. A total of 6 months after H. pylori infection, mice showed acid secretion atrophy and TFF2-expressing metaplasia (9). The detection methods for mouse model establishment include histological analysis and marker detection. Histological analysis identified the morphological features (such as focal lesions and glandular structural abnormalities) and inflammation of the metaplastic glands by H&E staining. When inflammation occurs, different types of metaplastic glands can form in nearby focal lesions, creating a complex glandular assembly made up of a variety of intestinal or gastric cell lineages that co-positively test for different cell lineage markers. Cell lineage markers, such as HE4, TFF2, MUC6, CD44v9 of SPEM and TFF3, MUC2, tail-type homeobox 1/2 or α-defense 5 of IM, were used to identify the lineage. Together with the identification of associated ILs, Ki67, CD44, MUC1, MUC5AC, p53, VEGF and other biomarkers, it can be used to assess the malignant potential of SPEM (74).

Table II

Mouse models of SPEM induced by different approaches of IL research.

Table II

Mouse models of SPEM induced by different approaches of IL research.

Model typeILMouse typeMethodDrugDosageFrequencyTime of administrationSPEMP/I SPEMIMInflammatory responseInvasive glandsMarkers(Refs.)
DMP-777-inducedIL-13C57BL/6Intraperitoneal injectionDMP-777350 mg/kgOnce daily1,3,7 and 14 daysYesNoNoNoNoGS II, CD44v9, p120(160)
IL-13C57BL/6 and IL13KOOral gavageDMP-777350 mg/kgOnce daily8 daysYesNoNoNoNoGS II, CD44v9, GIF(59)
IL-33C57BL/6 and Wfdc2−/−Oral inoculationDMP-777350 mg/kgOnce daily7 or 14 daysYesNoNoNoNoGS II, CD44v9, GIF(159)
L-635-inducedIL-13C57BL/6Oral gavageL-635350 mg/kgOnce daily3 daysYesYesNoYesNoGS II, GIF,(36)
IL-13IL-13-td and TomatoOral gavageL-635350 mg/kgOnce daily3 daysYesYesNoYesNoGS II, AQP5, CD44v9TFF2(39)
IL-13C57BL/6 and IL13KOOral gavageL-635350 mg/kgOnce daily3 daysYesYesNoYesNoGS II, CD44v9, GIF(59)
IL-13C57BL/6Orally injectionL-635350 mg/kgOnce daily2-4 daysYesYesNoYesNoGS II, CD44v9, p120(160)
IL-13C57BL/6Intraperitoneal injectionL-635350 mg/kgOnce daily1, 2, and 3 daysYesYesNoYesNoGS II, CD44v9, p120(160)
IL-33C57BL/6 and IL33KO and ST2KOOral gavageL-635350 mg/kgOnce daily3 daysYesYesNoYesNoGS II, CD44v9, GIF(59)
IL-33C57BL/6 and Wfdc2−/−Oral inoculationL-635350 mg/kgOnce daily3 daysYesYesNoYesNoGS II, CD44v9, GIF(159)
IL-33C57BL/6Oral gavageL-635350 mg/kgOnce daily3 daysYesYesNoYesNoGIF, GS II(68)
Tamoxifen-inducedIL-10C57BL/6Intraperitoneal injectionTamoxifen3 mg/20 gOnce daily3 daysYesNoNoNoNoGS II, VEGF-β(25)
IL-13 gp130F/FIntraperitoneal injectionTamoxifen50 mg/kgTwo doses3 days apartYesNoNoYesNoGS II, GIF, TFF2(121)
IL-13 gp130F/FIntraperitoneal injectionTamoxifen250 mg/kgOnce daily3 daysYesNoNoYesNoGS II, GIF, TFF2(121)
IL-25 gp130F/FIntraperitoneal injectionTamoxifen50 mg/kgTwo doses3 days apartYesNoNoYesNoGS II, GIF, TFF2(121)
IL-25 gp130F/FIntraperitoneal injectionTamoxifen250 mg/kgOnce daily3 daysYesNoNoYesNoGS II, GIF, TFF2(121)
IL-27TxA23 x Ebi3−/−Intraperitoneal injectionTamoxifen5 mg/20 gOnce daily3 daysYesNoNoYesNoGS II, VEGF-β, CD44v9(105)
IL-33C57BL/6 and Wfdc2−/−Intraperitoneal injectionTamoxifen250 mg/kgOnce daily3 daysYesNoNoYesNoGS II, CD44v9, GIF(159)
Transgenic and autoimmuneIL-13TxA23Transgenic---4 monthsYesYesNoYesNoGS II, GKN3(125)
IL-17ATxA23Transgenic---2-12 monthsYesNoNoYesNoVEGF-β, CD44v9(125)
IL-27TxA23Transgenic---2-12 monthsYesNoNoYesNoGS II, VEGF-β, CD44v9(105)
TransgenicIL-33SAMP1/YitFcTransgenic---4 weeksYesNoNoYesNoGS II, CD44v9, GIF(58)
IL-33PC-specific GRIM-19 KOTransgenic---2-10 weeksYesNoNoYesNoGS II, GIF(28)
SurgicalIL-13C57BL/6JAdrenalectomy and castration---2 monthsYesNoNoYesNoGS II, CD44v9(38)
Helicobacter infectionIL-1βC57BL/6 and Gli1−/−Helicobacter felis (CS1 strain); oral gavageBrucella broth with H. felis108 H. felis cells in 100 mlonce daily3 daysYesNoNoYesYesGS II(31)

[i] IL, interleukin; SPEM, spasmolytic polypeptide-expressing metaplasia; P/I SPEM proliferative/intestinalized SPEM; IM, intestinal metaplasia; gp130, glycoprotein 130; GS II, Griffonia simplicifolia lectin II; KO, knockout; GIF, gastric intrinsic factor; AQP5, aquaporin 5; TFF2, trefoil factor 2; p120, p120 catenin; GKN3, gastrokine 3; GRIM-19, gene-related to melanoma-19; PC, parietal cell; IL13KO, IL-13 knockout; Wfdc2−/−, whey acidic protein four-disulfide core domain 2 knockout; IL33KO, IL-33 knockout; ST2KO, suppression of tumorigenicity 2 knockout.

Gastric organoids, also referred to as gastroids, are 3-dimensional spheroids made by researchers from glands that have separated from the stomach corpus mucosa. These gastroids allowed the examination of the direct effects of ILs on gastric epithelial cells (144). Gastric glands were isolated from the target mice, and Matrigel was used to cultivate whole gastric glands. They were placed in 24- or 48-well plates, and cultured gastric organoids were treated with recombinant IL (Table SI). Typical images were captured of organoids aged 10 days following a 7-day IL treatment (34,39,138), and microphotographs were used to examine changes in gastroids. Organoid diameter, relative organoid diameter, organoid size, survival rate, Muc6 expression level, AQP5/MUC6 double positive rate, structural departure from normal tissue, height deviation from normal tissue, growth arrest, mortality and structural collapse were among the statistical indicators. This model is a crucial tool for studying the onset and progression of SPEM as it closely resembles the cellular makeup and behavior of the normal organism, making it easier to add ILs from exogenous sources.

Therapeutic implications and future prospects

At present, SPEM detection in clinical practice has not yet been popularized, but there is a growing interest in the importance of metaplasia in precancerous lesions. Endoscopy, tissue biopsy and biomarker detection are the most effective diagnostic methods for gastric precancerous lesions (74). To lower the risk of inflammatory cancer transformation and enhance the quality of life of patients, the recommended management strategies include routine gastroscopy monitoring, H. pylori eradication, lifestyle modifications and medication interventions. The therapeutic potential of ILs has gained interest from basic and translational cancer researchers. A growing number of ongoing clinical trials have highlighted their value as therapeutic agents and targets (171-175). Among these, IL-13 and its receptor were found to be promising targets for treatment in order to prevent and/or reverse the development of metaplasia in atrophic gastritis (144). IL-13 and IL-4Ra neutralizing antibodies (lebrikizumab, dupilumab) have been used in clinical settings, and antagonizing the IL-33/IL-13 signaling pathway has become a prospective treatment paradigm to reduce the progression of metaplasia and tumor transformation (144,176). By controlling the cytokine environment and CD4+ T cell infiltration, systemic IL-27 administration prevents gastric injury and aids in the recovery of acid glands (43). Non-small cell lung cancer, myelodysplastic syndrome and chronic myeloid leukemia have been treated with IL-1 neutralization strategies (such as canakinumab and anakinra) and the IL-6 antibody siltuximab (177,178). Furthermore, IL-2 monotherapy has shown early success in treating melanoma, renal cell carcinoma and gastrointestinal tract cancer, and may be useful in reducing the risk of tumor transformation and metaplasia (179). Innovative IL-12 therapies (such as oncolytic viruses, nanoparticles and CAR-T cells) can enhance the therapeutic potential of gastrointestinal cancer through precise delivery (180,181).

Current clinical trials related to GC focus on innovative applications of engineered ILs (Table SII). The safety of Bempegaldesleukin, a polyethylene glycolized non-α IL-2 variant, which extends half-life and reduces toxicity by blocking the IL-2Rα subunit binding site via a PEG residue, has been validated (ClinicalTrials.gov Identifier: NCT02983045, https://clinicaltrials.gov/ct2/show/NCT02983045). The comparable drug THOR707 (SAR444245) enhances anti-tumor activity through selective activation of intermediate affinity IL-2R (ClinicalTrials.gov Identifier: NCT04009681, https://clinicaltrials.gov/ct2/show/NCT04009681). In addition, ALKS 4230, which uses an IL-2/IL-2Rα circular replacement structure to optimize receptor targeting, is in phase I/II studies (ClinicalTrials.gov Identifier: NCT03861793, https://clinicaltrials.gov/ct2/show/NCT03861793). VG161 is a recombinant human IL-12/15/PD-L1B triple factor tumor lysing HSV-1 injection that induces systemic anti-tumor immunity via local cytokine release (ClinicalTrials.gov Identifier: NCT06008925, https://clinicaltrials.gov/ct2/show/NCT06008925). Recombinant human nsIL12 tumor solubilizing adenovirus injection (BioTTT001) in combination with SOX chemotherapy and teraplizumab is being evaluated for synergistic efficacy against peritoneal metastasis of GC (ClinicalTrials.gov Identifier: NCT06283121, https://clinicaltrials.gov/ct2/show/NCT06283121). The engineered modification breaks through the limitations of traditional IL therapy and provides a novel direction for GC immunotherapy.

However, IL therapy is challenged by its short half-life and narrow therapeutic window, leading to suboptimal efficacy and unpredictable side effects (21). Common adverse reactions such as influenza-like symptoms (including fever, chills and fatigue) are short but notably affect the quality of life of patients. Severe side effects included capillary leak syndrome, hypotension, fluid retention, hepatotoxicity (liver function damage and elevated enzyme levels), bone marrow suppression (increased risk of anemia, infection and bleeding) and cardiovascular complications (arrhythmia and fluid excess) (45). IL therapy is difficult to develop and is not considered suitable as a standalone treatment for clinical conditions. The pleiotropic nature of ILs contributes to the aforementioned issues by causing dose-limiting toxicity or inadequate efficacy (46,182). Future research must elucidate the mechanism of IL-mediated SPEM in order to develop novel therapies and investigate IL modification strategies, such as targeted delivery mechanisms, PEGylation, fusion construction, affinity regulation and synergy with other immunotumor drugs (177,183-185). The emergence of more effective immunotherapy and improved understanding of the tumor microenvironment provide novel methods for the treatment of cancer using the IL network (74,137). Furthermore, patients with advanced disease are currently included in the majority of trials, which may not be the optimal setting for IL-based therapies. Future research should focus on identifying and treating gastric metaplasia with a tendency to become cancerous (28,41,186). Specific directions may include: i) Developing more personalized IL modification strategies for patients with early-stage SPEM, ii) designing combination trials of IL therapy in patients with high-risk gastric metaplasia to halt malignant progression, and iii) exploring the efficacy of IL-targeted therapies in well-defined SPEM cohorts with targeted design. Further research to target early precancerous lesions, combined with tumor microenvironment regulation, deepen the understanding of the pathogenesis of SPEM and provide direction for targeted therapy to alleviate the progression of GC is warranted.

Conclusion

The present review described the role of various ILs and their expression products in the formation and progression of SPEM. IL effects are complex and multifactorial. It was summarized that certain ILs, such as IL-2, IL-4, IL-6, IL-8, IL-18 and IL-32, have not yet been studied to confirm a direct relationship with SPEM but are closely related to IM and GC development, which require further investigation. M2 macrophages promote SPEM progression in the context of inflammation and parietal cell atrophy, and the establishment of a chronic inflammatory environment through pro-inflammatory IL recruitment is also a potential direction for research. Studies have focused on the role that these ILs may serve in GC-associated with angiogenesis, metastasis and chemotherapy resistance, which are key factors influencing carcinogenesis and tumor progression, quality and patient survival. Although there are still a number of challenges to overcome in the development of IL- or IL-targeted therapies, the development of biologics and small-molecule inhibitors that selectively target pathogenesis could improve therapeutic efficacy and minimize side effects. With the discovery of specific markers, the understanding of SPEM origin and cancer progression will be improved, which may contribute to the simple and accurate diagnosis of early developmental abnormalities. These results provide a research direction for the pathogenesis of SPEM and open up new avenues for future diagnosis and treatment.

Supplementary Data

Availability of data and materials

Not applicable.

Authors' contributions

Conceptualization, literature investigation, writing the original draft, writing, review and editing, and visualization were performed by TZ, MJ and XZ. WG, SZ and HL made substantial contributions to writing, reviewing and editing. HL and MJ were responsible for conceptualization (substantial contribution to the design of the work), supervision and project administration. Data authentication is not applicable. All authors read and approved the final manuscript, and agree to be accountable for all aspects of the work.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgements

Not applicable.

Funding

The present work was supported by grants from the National Natural Science Foundation of China (grant no. 82274442) and the Tianjin Municipal Education Commission (grant no. 2024KJ043).

References

1 

Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA and Jemal A: Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 68:394–424. 2018.PubMed/NCBI

2 

Smyth EC, Nilsson M, Grabsch HI, van Grieken NC and Lordick F: Gastric cancer. Lancet. 396:635–648. 2020. View Article : Google Scholar : PubMed/NCBI

3 

Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I and Jemal A: Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 74:229–263. 2024.PubMed/NCBI

4 

Morgan E, Arnold M, Camargo MC, Gini A, Kunzmann AT, Matsuda T, Meheus F, Verhoeven RHA, Vignat J, Laversanne M, et al: The current and future incidence and mortality of gastric cancer in 185 countries, 2020-40: A population-based modelling study. EClinicalMedicine. 47:1014042022. View Article : Google Scholar : PubMed/NCBI

5 

Correa P, Haenszel W, Cuello C, Tannenbaum S and Archer M: A model for gastric cancer epidemiology. Lancet. 2:58–60. 1975. View Article : Google Scholar : PubMed/NCBI

6 

Yan L, Li W, Chen F, Wang J, Chen J, Chen Y and Ye W: Inflammation as a mediator of microbiome Dysbiosis-associated DNA methylation changes in gastric premalignant lesions. Phenomics. 3:496–501. 2023. View Article : Google Scholar : PubMed/NCBI

7 

Huang N, He HW, He YY, Gu W, Xu MJ and Liu L: Xiaotan Sanjie recipe, a compound Chinese herbal medicine, inhibits gastric cancer metastasis by regulating GnT-V-mediated E-cadherin glycosylation. J Integr Med. 21:561–574. 2023. View Article : Google Scholar : PubMed/NCBI

8 

Giroux V and Rustgi AK: Metaplasia: Tissue injury adaptation and a precursor to the dysplasia-cancer sequence. Nat Rev Cancer. 17:594–604. 2017. View Article : Google Scholar : PubMed/NCBI

9 

Petersen CP, Mills JC and Goldenring JR: Murine models of gastric corpus preneoplasia. Cell Mol Gastroenterol Hepatol. 3:11–26. 2017. View Article : Google Scholar : PubMed/NCBI

10 

Zhang M, Hu S, Min M, Ni Y, Lu Z, Sun X, Wu J, Liu B, Ying X and Liu Y: Dissecting transcriptional heterogeneity in primary gastric adenocarcinoma by single cell RNA sequencing. Gut. 70:464–475. 2021. View Article : Google Scholar

11 

Muthupalani S, Ge Z, Joy J, Feng Y, Dobey C, Cho HY, Langenbach R, Wang TC, Hagen SJ and Fox JG: Muc5ac null mice are predisposed to spontaneous gastric antro-pyloric hyperplasia and adenomas coupled with attenuated H. pylori-induced corpus mucous metaplasia. Lab Invest. 99:1887–1905. 2019. View Article : Google Scholar : PubMed/NCBI

12 

Goldenring JR: Spasmolytic polypeptide-expressing metaplasia (SPEM) cell lineages can be an origin of gastric cancer. J Pathol. 260:109–111. 2023. View Article : Google Scholar : PubMed/NCBI

13 

Schmidt PH, Lee JR, Joshi V, Playford RJ, Poulsom R, Wright NA and Goldenring JR: Identification of a metaplastic cell lineage associated with human gastric adenocarcinoma. Lab Invest. 79:639–646. 1999.PubMed/NCBI

14 

Halldórsdóttir AM, Sigurdardóttrir M, Jónasson JG, Oddsdóttir M, Magnússon J, Lee JR and Goldenring JR: Spasmolytic polypeptide-expressing metaplasia (SPEM) associated with gastric cancer in Iceland. Dig Dis Sci. 48:431–441. 2003. View Article : Google Scholar : PubMed/NCBI

15 

Yamaguchi H, Goldenring JR, Kaminishi M and Lee JR: Identification of spasmolytic polypeptide expressing metaplasia (SPEM) in remnant gastric cancer and surveillance postgastrectomy biopsies. Dig Dis Sci. 47:573–578. 2002. View Article : Google Scholar : PubMed/NCBI

16 

Katz JP, Perreault N, Goldstein BG, Actman L, McNally SR, Silberg DG, Furth EE and Kaestner KH: Loss of Klf4 in mice causes altered proliferation and differentiation and precancerous changes in the adult stomach. Gastroenterology. 128:935–945. 2005. View Article : Google Scholar : PubMed/NCBI

17 

Oshima H, Matsunaga A, Fujimura T, Tsukamoto T, Taketo MM and Oshima M: Carcinogenesis in mouse stomach by simultaneous activation of the Wnt signaling and prostaglandin E2 pathway. Gastroenterology. 131:1086–1095. 2006. View Article : Google Scholar : PubMed/NCBI

18 

Goldenring JR, Nam KT, Wang TC, Mills JC and Wright NA: Spasmolytic polypeptide-expressing metaplasia and intestinal metaplasia: Time for reevaluation of metaplasias and the origins of gastric cancer. Gastroenterology. 138:2207–2210.e1. 2010. View Article : Google Scholar : PubMed/NCBI

19 

Reyes ME, Pulgar V, Vivallo C, Ili CG, Mora-Lagos B and Brebi P: Epigenetic modulation of cytokine expression in gastric cancer: Influence on angiogenesis, metastasis and chemoresistance. Front Immunol. 15:13475302024. View Article : Google Scholar : PubMed/NCBI

20 

Bockerstett KA and DiPaolo RJ: Regulation of gastric carcinogenesis by inflammatory cytokines. Cell Mol Gastroenterol Hepatol. 4:47–53. 2017. View Article : Google Scholar : PubMed/NCBI

21 

Briukhovetska D, Dörr J, Endres S, Libby P, Dinarello CA and Kobold S: Interleukins in cancer: From biology to therapy. Nat Rev Cancer. 21:481–499. 2021. View Article : Google Scholar : PubMed/NCBI

22 

Dranoff G: Cytokines in cancer pathogenesis and cancer therapy. Nat Rev Cancer. 4:11–22. 2004. View Article : Google Scholar : PubMed/NCBI

23 

Yasmin R, Siraj S, Hassan A, Khan AR, Abbasi R and Ahmad N: Epigenetic regulation of inflammatory cytokines and associated genes in human malignancies. Mediators Inflamm. 2015:2017032015. View Article : Google Scholar : PubMed/NCBI

24 

Oshima M, Oshima H, Matsunaga A and Taketo MM: Hyperplastic gastric tumors with spasmolytic polypeptide-expressing metaplasia caused by tumor necrosis factor-alpha-dependent inflammation in cyclooxygenase-2/microsomal prostaglandin E synthase-1 transgenic mice. Cancer Res. 65:9147–9151. 2005. View Article : Google Scholar : PubMed/NCBI

25 

Lee C, Lee H, Hwang SY, Moon CM and Hong SN: IL-10 plays a pivotal role in Tamoxifen-induced spasmolytic Polypeptide-expressing metaplasia in gastric mucosa. Gut Liver. 11:789–797. 2017. View Article : Google Scholar : PubMed/NCBI

26 

Demitrack ES, Gifford GB, Keeley TM, Horita N, Todisco A, Turgeon DK, Siebel CW and Samuelson LC: NOTCH1 and NOTCH2 regulate epithelial cell proliferation in mouse and human gastric corpus. Am J Physiol Gastrointest Liver Physiol. 312:G133–G144. 2017. View Article : Google Scholar :

27 

Negovan A, Iancu M, Fülöp E and Bănescu C: Helicobacter pylori and cytokine gene variants as predictors of premalignant gastric lesions. World J Gastroenterol. 25:4105–4124. 2019. View Article : Google Scholar : PubMed/NCBI

28 

Zeng X, Yang M, Ye T, Feng J, Xu X, Yang H, Wang X, Bao L, Li R, Xue B, et al: Mitochondrial GRIM-19 loss in parietal cells promotes spasmolytic polypeptide-expressing metaplasia through NLR family pyrin domain-containing 3 (NLRP3)-mediated IL-33 activation via a reactive oxygen species (ROS)-NRF2-Heme oxygenase-1(HO-1)-NF-кB axis. Free Radic Biol Med. 202:46–61. 2023. View Article : Google Scholar : PubMed/NCBI

29 

Berraondo P, Sanmamed MF, Ochoa MC, Etxeberria I, Aznar MA, Pérez-Gracia JL, Rodríguez-Ruiz ME, Ponz-Sarvise M, Castañón E and Melero I: Cytokines in clinical cancer immunotherapy. Br J Cancer. 120:6–15. 2019. View Article : Google Scholar :

30 

Kureshi CT and Dougan SK: Cytokines in cancer. Cancer Cell. 43:15–35. 2025. View Article : Google Scholar :

31 

El-Zaatari M, Kao JY, Tessier A, Bai L, Hayes MM, Fontaine C, Eaton KA and Merchant JL: Gli1 deletion prevents Helicobacter-induced gastric metaplasia and expansion of myeloid cell subsets. PLoS One. 8:e589352013. View Article : Google Scholar : PubMed/NCBI

32 

Goldenring JR, Nam KT and Mills JC: The origin of pre-neoplastic metaplasia in the stomach: Chief cells emerge from the Mist. Exp Cell Res. 317:2759–2764. 2011. View Article : Google Scholar : PubMed/NCBI

33 

Kang WQ, Rathinavelu S, Samuelson LC and Merchant JL: Interferon gamma induction of gastric mucous neck cell hypertrophy. Lab Invest. 85:702–715. 2005. View Article : Google Scholar : PubMed/NCBI

34 

Bockerstett KA, Osaki LH, Petersen CP, Cai CW, Wong CF, Nguyen TM, Ford EL, Hoft DF, Mills JC, Goldenring JR and DiPaolo RJ: Interleukin-17A promotes parietal cell atrophy by inducing apoptosis. Cell Mol Gastroenterol Hepatol. 5:678–690.e1. 2018. View Article : Google Scholar : PubMed/NCBI

35 

Caldwell B, Meyer AR, Weis JA, Engevik AC and Choi E: Chief cell plasticity is the origin of metaplasia following acute injury in the stomach mucosa. Gut. 71:1068–1077. 2022. View Article : Google Scholar

36 

Meyer AR, Engevik AC, Madorsky T, Belmont E, Stier MT, Norlander AE, Pilkinton MA, McDonnell WJ, Weis JA, Jang B, et al: Group 2 innate lymphoid cells coordinate damage response in the stomach. Gastroenterology. 159:2077–2091.e8. 2020. View Article : Google Scholar : PubMed/NCBI

37 

He L, Zhang X, Zhang S, Wang Y, Hu W, Li J, Liu Y, Liao Y, Peng X, Li J, et al: H. pylori-facilitated TERT/Wnt/β-Catenin triggers spasmolytic Polypeptide-expressing metaplasia and oxyntic atrophy. Adv Sci (Weinh). 12:e24012272025. View Article : Google Scholar

38 

Busada JT, Peterson KN, Khadka S, Xu X, Oakley RH, Cook DN and Cidlowski JA: Glucocorticoids and androgens protect from gastric metaplasia by suppressing group 2 innate lymphoid cell activation. Gastroenterology. 161:637–652.e4. 2021. View Article : Google Scholar : PubMed/NCBI

39 

Contreras-Panta EW, Lee SH, Won Y, Norlander AE, Simmons AJ, Peebles RS Jr, Lau KS, Choi E and Goldenring JR: Interleukin 13 promotes maturation and proliferation in metaplastic gastroids. Cell Mol Gastroenterol Hepatol. 18:1013662024. View Article : Google Scholar : PubMed/NCBI

40 

Goldenring JR and Nomura S: Differentiation of the gastric mucosa III. Animal models of oxyntic atrophy and metaplasia. Am J Physiol Gastrointest Liver Physiol. 291:G999–G1004. 2006. View Article : Google Scholar : PubMed/NCBI

41 

Lee SH, Jang B, Min J, Contreras-Panta EW, Presentation KS, Delgado AG, Piazuelo MB, Choi E and Goldenring JR: Up-regulation of aquaporin 5 defines spasmolytic Polypeptide-expressing metaplasia and progression to incomplete intestinal metaplasia. Cell Mol Gastroenterol Hepatol. 13:199–217. 2022. View Article : Google Scholar

42 

Park DJ and Kim SE: The Role of IL-10 in gastric spasmolytic Polypeptide-expressing Metaplasia-related carcinogenesis. Gut Liver. 11:741–742. 2017. View Article : Google Scholar : PubMed/NCBI

43 

Bockerstett KA, Petersen CP, Noto CN, Kuehm LM, Wong CF, Ford EL, Teague RM, Mills JC, Goldenring JR and DiPaolo RJ: Interleukin 27 protects from gastric atrophy and metaplasia during chronic autoimmune gastritis. Cell Mol Gastroenterol Hepatol. 10:561–579. 2020. View Article : Google Scholar : PubMed/NCBI

44 

de Brito BB, da Silva FAF and de Melo FF: Role of polymorphisms in genes that encode cytokines and Helicobacter pylori virulence factors in gastric carcinogenesis. World J Clin Oncol. 9:83–89. 2018. View Article : Google Scholar : PubMed/NCBI

45 

Saleh RO, Jasim SA, Kadhum WR, Hjazi A, Faraz A, Abid MK, Yumashev A, Alawadi A, Aiad IAZ and Alsalamy A: Exploring the detailed role of interleukins in cancer: A comprehensive review of literature. Pathol Res Pract. 257:1552842024. View Article : Google Scholar : PubMed/NCBI

46 

Propper DJ and Balkwill FR: Harnessing cytokines and chemokines for cancer therapy. Nat Rev Clin Oncol. 19:237–253. 2022. View Article : Google Scholar : PubMed/NCBI

47 

Lamkanfi M and Dixit VM: Mechanisms and functions of inflammasomes. Cell. 157:1013–1022. 2014. View Article : Google Scholar : PubMed/NCBI

48 

Santos JC, Ladeira MS, Pedrazzoli J Jr and Ribeiro ML: Relationship of IL-1 and TNF-α polymorphisms with Helicobacter pylori in gastric diseases in a Brazilian population. Braz J Med Biol Res. 45:811–817. 2012. View Article : Google Scholar : PubMed/NCBI

49 

Starzyńska T, Ferenc K, Wex T, Kähne T, Lubiński J, Lawniczak M, Marlicz K and Malfertheiner P: The association between the interleukin-1 polymorphisms and gastric cancer risk depends on the family history of gastric carcinoma in the study population. Am J Gastroenterol. 101:248–254. 2006. View Article : Google Scholar

50 

Xue H, Lin B, Ni P, Xu H and Huang G: Interleukin-1B and interleukin-1 RN polymorphisms and gastric carcinoma risk: A meta-analysis. J Gastroenterol Hepatol. 25:1604–1617. 2010. View Article : Google Scholar : PubMed/NCBI

51 

Wang YM, Li ZX, Tang FB, Zhang Y, Zhou T, Zhang L, Ma JL, You WC and Pan KF: Association of genetic polymorphisms of interleukins with gastric cancer and precancerous gastric lesions in a high-risk Chinese population. Tumour Biol. 37:2233–2242. 2016. View Article : Google Scholar

52 

Myung DS, Lee WS, Park YL, Kim N, Oh HH, Kim MY, Oak CY, Chung CY, Park HC, Kim JS, et al: Association between interleukin-18 gene polymorphism and Helicobacter pylori infection in the Korean population. Sci Rep. 5:115352015. View Article : Google Scholar : PubMed/NCBI

53 

Cheung H, Chen NJ, Cao Z, Ono N, Ohashi PS and Yeh WC: Accessory protein-like is essential for IL-18-mediated signaling. J Immunol. 174:5351–5357. 2005. View Article : Google Scholar : PubMed/NCBI

54 

Osaki LH, Bockerstett KA, Wong CF, Ford EL, Madison BB, DiPaolo RJ and Mills JC: Interferon-γ directly induces gastric epithelial cell death and is required for progression to metaplasia. J Pathol. 247:513–523. 2019. View Article : Google Scholar :

55 

Syu LJ, El-Zaatari M, Eaton KA, Liu Z, Tetarbe M, Keeley TM, Pero J, Ferris J, Wilbert D, Kaatz A, et al: Transgenic expression of interferon-γ in mouse stomach leads to inflammation, metaplasia, and dysplasia. Am J Pathol. 181:2114–2125. 2012. View Article : Google Scholar : PubMed/NCBI

56 

Leung WK, Chan MC, To KF, Man EP, Ng EK, Chu ES, Lau JY, Lin SR and Sung JJ: H. pylori genotypes and cytokine gene polymorphisms influence the development of gastric intestinal metaplasia in a Chinese population. Am J Gastroenterol. 101:714–720. 2006. View Article : Google Scholar : PubMed/NCBI

57 

He R, Yin H, Yuan B, Liu T, Luo L, Huang P, Dai L and Zeng K: IL-33 improves wound healing through enhanced M2 macrophage polarization in diabetic mice. Mol Immunol. 90:42–49. 2017. View Article : Google Scholar : PubMed/NCBI

58 

Schmitz J, Owyang A, Oldham E, Song Y, Murphy E, McClanahan TK, Zurawski G, Moshrefi M, Qin J, Li X, et al: IL-33, an interleukin-1-like cytokine that signals via the IL-1 receptor-related protein ST2 and induces T helper type 2-associated cytokines. Immunity. 23:479–490. 2005. View Article : Google Scholar : PubMed/NCBI

59 

Carriere V, Roussel L, Ortega N, Lacorre DA, Americh L, Aguilar L, Bouche G and Girard JP: IL-33, the IL-1-like cytokine ligand for ST2 receptor, is a chromatin-associated nuclear factor in vivo. Proc Natl Acad Sci USA. 104:282–287. 2007. View Article : Google Scholar :

60 

Buzzelli JN, Chalinor HV, Pavlic DI, Sutton P, Menheniott TR, Giraud AS and Judd LM: IL33 is a stomach alarmin that initiates a skewed Th2 response to injury and infection. Cell Mol Gastroenterol Hepatol. 1:203–221.e3. 2015. View Article : Google Scholar : PubMed/NCBI

61 

Schwartz C, O'Grady K, Lavelle EC and Fallon PG: Interleukin 33: An innate alarm for adaptive responses beyond Th2 immunity-emerging roles in obesity, intestinal inflammation, and cancer. Eur J Immunol. 46:1091–1100. 2016. View Article : Google Scholar : PubMed/NCBI

62 

De Salvo C, Pastorelli L, Petersen CP, Buttò LF, Buela KA, Omenetti S, Locovei SA, Ray S, Friedman HR, Duijser J, et al: Interleukin 33 triggers early Eosinophil-dependent events leading to metaplasia in a chronic model of Gastritis-prone mice. Gastroenterology. 160:302–316.e7. 2021. View Article : Google Scholar

63 

Petersen CP, Meyer AR, De Salvo C, Choi E, Schlegel C, Petersen A, Engevik AC, Prasad N, Levy SE, Peebles RS, et al: A signalling cascade of IL-33 to IL-13 regulates metaplasia in the mouse stomach. Gut. 67:805–817. 2018. View Article : Google Scholar

64 

Huang N, Cui X, Li W, Zhang C, Liu L and Li J: IL-33/ST2 promotes the malignant progression of gastric cancer via the MAPK pathway. Mol Med Rep. 23:3612021. View Article : Google Scholar :

65 

Engevik AC, Feng R, Choi E, White S, Bertaux-Skeirik N, Li J, Mahe MM, Aihara E, Yang L, DiPasquale B, et al: The development of spasmolytic polypeptide/TFF2-expressing metaplasia (SPEM) during gastric repair is absent in the aged stomach. Cell Mol Gastroenterol Hepatol. 2:605–624. 2016. View Article : Google Scholar : PubMed/NCBI

66 

Farrell JJ, Taupin D, Koh TJ, Chen D, Zhao CM, Podolsky DK and Wang TC: TFF2/SP-deficient mice show decreased gastric proliferation, increased acid secretion, and increased susceptibility to NSAID injury. J Clin Invest. 109:193–204. 2002. View Article : Google Scholar : PubMed/NCBI

67 

Nozaki K, Ogawa M, Williams JA, Lafleur BJ, Ng V, Drapkin RI, Mills JC, Konieczny SF, Nomura S and Goldenring JR: A molecular signature of gastric metaplasia arising in response to acute parietal cell loss. Gastroenterology. 134:511–522. 2008. View Article : Google Scholar : PubMed/NCBI

68 

Privitera G, Williams JJ and De Salvo C: The importance of Th2 immune responses in mediating the progression of Gastritis-associated metaplasia to gastric cancer. Cancers (Basel). 16:5222024. View Article : Google Scholar : PubMed/NCBI

69 

Meyer AR and Goldenring JR: Injury, repair, inflammation and metaplasia in the stomach. J Physiol. 596:3861–3867. 2018. View Article : Google Scholar : PubMed/NCBI

70 

Gordon S: Alternative activation of macrophages. Nat Rev Immunol. 3:23–35. 2003. View Article : Google Scholar : PubMed/NCBI

71 

Bernink JH, Germar K and Spits H: The role of ILC2 in pathology of type 2 inflammatory diseases. Curr Opin Immunol. 31:115–120. 2014. View Article : Google Scholar : PubMed/NCBI

72 

Li W, Huang X, Han X, Zhang J, Gao L and Chen H: IL-17A in gastric carcinogenesis: Good or bad? Front Immunol. 15:15012932024. View Article : Google Scholar : PubMed/NCBI

73 

Pisani LF, Teani I, Vecchi M and Pastorelli L: Interleukin-33: Friend or foe in gastrointestinal tract cancers? Cells. 12:14812023. View Article : Google Scholar : PubMed/NCBI

74 

Ye Q, Zhu Y, Ma Y, Wang Z and Xu G: Emerging role of spasmolytic polypeptide-expressing metaplasia in gastric cancer. J Gastrointest Oncol. 15:2673–2683. 2024. View Article : Google Scholar

75 

Melchiades JL, Zabaglia LM, Sallas ML, Orcini WA, Chen E, Smith MAC, Payão SLM and Rasmussen LT: Polymorphisms and haplotypes of the interleukin 2 gene are associated with an increased risk of gastric cancer. The possible involvement of Helicobacter pylori. Cytokine. 96:203–207. 2017. View Article : Google Scholar : PubMed/NCBI

76 

Shin WG, Jang JS, Kim HS, Kim SJ, Kim KH, Jang MK, Lee JH, Kim HJ and Kim HY: Polymorphisms of interleukin-1 and interleukin-2 genes in patients with gastric cancer in Korea. J Gastroenterol Hepatol. 23:1567–1573. 2008. View Article : Google Scholar : PubMed/NCBI

77 

Mohammadi M, Czinn S, Redline R and Nedrud J: Helicobacter-specific cell-mediated immune responses display a predominant Th1 phenotype and promote a delayed-type hypersensitivity response in the stomachs of mice. J Immunol. 156:4729–4738. 1996. View Article : Google Scholar : PubMed/NCBI

78 

Roth KA, Kapadia SB, Martin SM and Lorenz RG: Cellular immune responses are essential for the development of Helicobacter felis-associated gastric pathology. J Immunol. 163:1490–1497. 1999. View Article : Google Scholar : PubMed/NCBI

79 

Yang E, Chua W, Ng W and Roberts TL: Peripheral cytokine levels as a prognostic indicator in gastric cancer: A review of existing literature. Biomedicines. 9:19162021. View Article : Google Scholar : PubMed/NCBI

80 

Bevington SL, Keane P, Soley JK, Tauch S, Gajdasik DW, Fiancette R, Matei-Rascu V, Willis CM, Withers DR and Cockerill PN: IL-2/IL-7-inducible factors pioneer the path to T cell differentiation in advance of lineage-defining factors. EMBO J. 39:e1052202020. View Article : Google Scholar : PubMed/NCBI

81 

Bamford KB, Fan X, Crowe SE, Leary JF, Gourley WK, Luthra GK, Brooks EG, Graham DY, Reyes VE and Ernst PB: Lymphocytes in the human gastric mucosa during Helicobacter pylori have a T helper cell 1 phenotype. Gastroenterology. 114:482–492. 1998. View Article : Google Scholar : PubMed/NCBI

82 

Padol IT and Hunt RH: Effect of Th1 cytokines on acid secretion in pharmacologically characterised mouse gastric glands. Gut. 53:1075–1081. 2004. View Article : Google Scholar : PubMed/NCBI

83 

Togawa S, Joh T, Itoh M, Katsuda N, Ito H, Matsuo K, Tajima K and Hamajima N: Interleukin-2 gene polymorphisms associated with increased risk of gastric atrophy from Helicobacter pylori infection. Helicobacter. 10:172–178. 2005. View Article : Google Scholar : PubMed/NCBI

84 

Van Dyken SJ and Locksley RM: Interleukin-4- and interleukin-13-mediated alternatively activated macrophages: Roles in homeostasis and disease. Annu Rev Immunol. 31:317–343. 2013. View Article : Google Scholar : PubMed/NCBI

85 

Pan K, Li Q, Guo Z and Li Z: Healing action of Interleukin-4 (IL-4) in acute and chronic inflammatory conditions: Mechanisms and therapeutic strategies. Pharmacol Ther. 265:1087602025. View Article : Google Scholar

86 

Labonte AC, Tosello-Trampont AC and Hahn YS: The role of macrophage polarization in infectious and inflammatory diseases. Mol Cells. 37:275–285. 2014. View Article : Google Scholar : PubMed/NCBI

87 

Liu YC, Zou XB, Chai YF and Yao YM: Macrophage polarization in inflammatory diseases. Int J Biol Sci. 10:520–529. 2014. View Article : Google Scholar : PubMed/NCBI

88 

Petersen CP, Weis VG, Nam KT, Sousa JF, Fingleton B and Goldenring JR: Macrophages promote progression of spasmolytic polypeptide-expressing metaplasia after acute loss of parietal cells. Gastroenterology. 146:1727–1738.e8. 2014. View Article : Google Scholar : PubMed/NCBI

89 

Yoshimoto T: The hunt for the source of primary Interleukin-4: How we discovered that natural killer T cells and basophils determine T helper type 2 cell differentiation in vivo. Front Immunol. 9:7162018. View Article : Google Scholar : PubMed/NCBI

90 

Zhou JY, Alvarez CA and Cobb BA: Integration of IL-2 and IL-4 signals coordinates divergent regulatory T cell responses and drives therapeutic efficacy. Elife. 10:e574172021. View Article : Google Scholar : PubMed/NCBI

91 

Zhang Z, Chen Z, Que Z, Fang Z, Zhu H and Tian J: Chinese medicines and natural medicine as immunotherapeutic agents for gastric cancer: Recent advances. Cancer Rep (Hoboken). 7:e21342024.

92 

Yang F, Shaibu Z, Liu Q and Zhu W: Cytokine profiles as predictive biomarkers for treatment outcomes in advanced gastric cancer patients undergoing PD-1 blockade immunochemotherapy: A meta-analysis. Clin Exp Med. 25:1362025. View Article : Google Scholar : PubMed/NCBI

93 

Ham IH, Oh HJ, Jin H, Bae CA, Jeon SM, Choi KS, Son SY, Han SU, Brekken RA, Lee D and Hur H: Targeting interleukin-6 as a strategy to overcome Stroma-induced resistance to chemotherapy in gastric cancer. Mol Cancer. 18:682019. View Article : Google Scholar : PubMed/NCBI

94 

Wu X, Tao P, Zhou Q, Li J, Yu Z, Wang X, Li J, Li C, Yan M, Zhu Z, et al: IL-6 secreted by cancer-associated fibroblasts promotes epithelial-mesenchymal transition and metastasis of gastric cancer via JAK2/STAT3 signaling pathway. Oncotarget. 8:20741–20750. 2017. View Article : Google Scholar : PubMed/NCBI

95 

Matsuo K, Oka M, Murase K, Soda H, Isomoto H, Takeshima F, Mizuta Y, Murata I and Kohno S: Expression of interleukin 6 and its receptor in human gastric and colorectal cancers. J Int Med Res. 31:69–75. 2003. View Article : Google Scholar : PubMed/NCBI

96 

Leja M, Wex T and Malfertheiner P: Markers for gastric cancer premalignant lesions: Where do we go? Dig Dis. 30:268–276. 2012. View Article : Google Scholar : PubMed/NCBI

97 

Johnson DE, O'Keefe RA and Grandis JR: Targeting the IL-6/JAK/STAT3 signalling axis in cancer. Nat Rev Clin Oncol. 15:234–248. 2018. View Article : Google Scholar : PubMed/NCBI

98 

Giraud AS, Jackson C, Menheniott TR and Judd LM: Differentiation of the gastric mucosa IV. Role of trefoil peptides and IL-6 cytokine family signaling in gastric homeostasis. Am J Physiol Gastrointest Liver Physiol. 292:G1–G5. 2007. View Article : Google Scholar

99 

Kamimura D, Ishihara K and Hirano T: IL-6 signal transduction and its physiological roles: The signal orchestration model. Rev Physiol Biochem Pharmacol. 149:1–38. 2003.PubMed/NCBI

100 

Lee IO, Kim JH, Choi YJ, Pillinger MH, Kim SY, Blaser MJ and Lee YC: Helicobacter pylori CagA phosphorylation status determines the gp130-activated SHP2/ERK and JAK/STAT signal transduction pathways in gastric epithelial cells. J Biol Chem. 285:16042–16050. 2010. View Article : Google Scholar : PubMed/NCBI

101 

Judd LM, Menheniott TR, Ling H, Jackson CB, Howlett M, Kalantzis A, Priebe W and Giraud AS: Inhibition of the JAK2/STAT3 pathway reduces gastric cancer growth in vitro and in vivo. PLoS One. 9:e959932014. View Article : Google Scholar : PubMed/NCBI

102 

Hwang IR, Hsu PI, Peterson LE, Gutierrez O, Kim JG, Graham DY and Yamaoka Y: Interleukin-6 genetic polymorphisms are not related to Helicobacter pylori-associated gastroduodenal diseases. Helicobacter. 8:142–148. 2003. View Article : Google Scholar : PubMed/NCBI

103 

Howlett M, Chalinor HV, Buzzelli JN, Nguyen N, van Driel IR, Bell KM, Fox JG, Dimitriadis E, Menheniott TR, Giraud AS and Judd LM: IL-11 is a parietal cell cytokine that induces atrophic gastritis. Gut. 61:1398–1409. 2012. View Article : Google Scholar

104 

Buzzelli JN, O'Connor L, Scurr M, Chung Nien Chin S, Catubig A, Ng GZ, Oshima M, Oshima H, Giraud AS, Sutton P, et al: Overexpression of IL-11 promotes premalignant gastric epithelial hyperplasia in isolation from germline gp130-JAK-STAT driver mutations. Am J Physiol Gastrointest Liver Physiol. 316:G251–G262. 2019. View Article : Google Scholar

105 

Howlett M, Giraud AS, Lescesen H, Jackson CB, Kalantzis A, Van Driel IR, Robb L, Van der Hoek M, Ernst M, Minamoto T, et al: The interleukin-6 family cytokine interleukin-11 regulates homeostatic epithelial cell turnover and promotes gastric tumor development. Gastroenterology. 136:967–977. 2009. View Article : Google Scholar : PubMed/NCBI

106 

Tebbutt NC, Giraud AS, Inglese M, Jenkins B, Waring P, Clay FJ, Malki S, Alderman BM, Grail D, Hollande F, et al: Reciprocal regulation of gastrointestinal homeostasis by SHP2 and STAT-mediated trefoil gene activation in gp130 mutant mice. Nature medicine. 8:1089–1097. 2002. View Article : Google Scholar : PubMed/NCBI

107 

Pastorelli L, Garg RR, Hoang SB, Spina L, Mattioli B, Scarpa M, Fiocchi C, Vecchi M and Pizarro TT: Epithelial-derived IL-33 and its receptor ST2 are dysregulated in ulcerative colitis and in experimental Th1/Th2 driven enteritis. Proc Natl Acad Sci USA. 107:8017–8022. 2010. View Article : Google Scholar : PubMed/NCBI

108 

Saha A, Hammond CE, Gooz M and Smolka AJ: The role of Sp1 in IL-1beta and H. pylori-mediated regulation of H,K-ATPase gene transcription. Am J Physiol Gastrointest Liver Physiol. 295:G977–G986. 2008. View Article : Google Scholar : PubMed/NCBI

109 

Wallace JL, Cucala M, Mugridge K and Parente L: Secretagogue-specific effects of interleukin-1 on gastric acid secretion. Am J Physiol. 261:G559–G564. 1991.PubMed/NCBI

110 

Ham IH, Lee D and Hur H: Role of Cancer-associated fibroblast in gastric cancer progression and resistance to treatments. J Oncol. 2019:62707842019. View Article : Google Scholar : PubMed/NCBI

111 

Howlett M, Menheniott TR, Judd LM and Giraud AS: Cytokine signalling via gp130 in gastric cancer. Biochim Biophys Acta. 1793:1623–1633. 2009. View Article : Google Scholar : PubMed/NCBI

112 

Iyer SS and Cheng G: Role of interleukin 10 transcriptional regulation in inflammation and autoimmune disease. Crit Rev Immunol. 32:23–63. 2012. View Article : Google Scholar : PubMed/NCBI

113 

Qu X, Tang Y and Hua S: Immunological approaches towards cancer and inflammation: A cross talk. Front Immunol. 9:5632018. View Article : Google Scholar : PubMed/NCBI

114 

Gao L, Weck MN, Michel A, Pawlita M and Brenner H: Association between chronic atrophic gastritis and serum antibodies to 15 Helicobacter pylori proteins measured by multiplex serology. Cancer Res. 69:2973–2980. 2009. View Article : Google Scholar : PubMed/NCBI

115 

Sugai H, Kono K, Takahashi A, Ichihara F, Kawaida H, Fujii H and Matsumoto Y: Characteristic alteration of monocytes with increased intracellular IL-10 and IL-12 in patients with advanced-stage gastric cancer. J Surg Res. 116:277–287. 2004. View Article : Google Scholar : PubMed/NCBI

116 

Zhang H, Li R, Cao Y, Gu Y, Lin C, Liu X, Lv K, He X, Fang H, Jin K, et al: Poor clinical outcomes and immunoevasive contexture in intratumoral IL-10-Producing macrophages enriched gastric cancer patients. Ann Surg. 275:e626–e635. 2022. View Article : Google Scholar

117 

Park HS, Kwon WS, Park S, Jo E, Lim SJ, Lee CK, Lee JB, Jung M, Kim HS, Beom SH, et al: Comprehensive immune profiling and immune-monitoring using body fluid of patients with metastatic gastric cancer. J Immunother Cancer. 7:2682019. View Article : Google Scholar : PubMed/NCBI

118 

Yarmohammadi R, Najafi K, Noroozbeygi M, Didehvar K, Rastin A, Ataei F, Atashzar MR and Shushtari SS: The Role of IL-6, IL-10 and CRP in gastrointestinal cancers. Cell Biol Int. 49:1061–1078. 2025. View Article : Google Scholar : PubMed/NCBI

119 

Nguyen TLM, Khurana SS, Sagartz JE, Mills JC and DiPaolo R: EBI3 (IL-27/IL-35) regulates the progression from atrophic gastritis to gastric cancer by Regulating IL-17 production in the gastric mucosa. Gastroenterology. 146(Suppl 1): S64–S65. 2014. View Article : Google Scholar

120 

Bockerstett KA, Nguyen TLM, Wong CF, Ford EL and DiPaolo RJ: IL-27 Regulates gastric metaplasia through effects on both immune and epithelial cells. Gastroenterology. 152(Suppl 1): S55–S56. 2017. View Article : Google Scholar

121 

Bockerstett K, Petersen C, Ford E, Goldenring JR and DiPaolo R: IL-17A promotes the progression from gastritis to gastric cancer. Gastroenterology. 150(Suppl 1): S110. 2016. View Article : Google Scholar

122 

Bockerstett KA, Petersen C, Noto CN, Kuehm L, Wong CF, Ford EL, Mills J, Goldenring JR and DiPaolo RJ: Interleukin-27-producing immune cells in the gastric mucosa protect the stomach from inflammation-induced atrophy and metaplasia. Gastroenterology. 156(Suppl 1): S186. 2019. View Article : Google Scholar

123 

Gu C, Wu L and Li X: IL-17 family: Cytokines, receptors and signaling. Cytokine. 64:477–485. 2013. View Article : Google Scholar : PubMed/NCBI

124 

Zhao J, Chen X, Herjan T and Li X: The role of interleukin-17 in tumor development and progression. J Exp Med. 217:e201902972020. View Article : Google Scholar :

125 

Kabir S: The role of interleukin-17 in the Helicobacter pylori induced infection and immunity. Helicobacter. 16:1–8. 2011. View Article : Google Scholar : PubMed/NCBI

126 

Meng XY, Zhou CH, Ma J, Jiang C and Ji P: Expression of interleukin-17 and its clinical significance in gastric cancer patients. Med Oncol. 29:3024–3028. 2012. View Article : Google Scholar : PubMed/NCBI

127 

Brackman LC, Jung MS, Green EH, Joshi N, Revetta FL, McClain MS, Markham NO, Piazuelo MB and Scott Algood HM: IL-17 signaling protects against Helicobacter pylori-induced gastric cancer. Gut microbes. 16:24304212024. View Article : Google Scholar : PubMed/NCBI

128 

Tangye SG and Puel A: The Th17/IL-17 Axis and host defense against fungal infections. J Allergy Clin Immunol Pract. 11:1624–1634. 2023. View Article : Google Scholar : PubMed/NCBI

129 

Kabil AK, Liu LT, Xu C, Nayyar N, González L, Chopra S, Brassard J, Beaulieu MJ, Li Y, Damji A, et al: Microbial dysbiosis sculpts a systemic ILC3/IL-17 axis governing lung inflammatory responses and central hematopoiesis. Mucosal Immunol. 18:1139–1158. 2025. View Article : Google Scholar : PubMed/NCBI

130 

Zhang J and Shen M: The Role of IL-17 in systemic autoinflammatory diseases: Mechanisms and therapeutic perspectives. Clin Rev Allergy Immunol. 68:272025. View Article : Google Scholar : PubMed/NCBI

131 

Zhong F, Cui D, Tao H, Du H and Xing C: IL-17A-producing T cells and associated cytokines are involved in the progression of gastric cancer. Oncol Rep. 34:2365–2374. 2015. View Article : Google Scholar : PubMed/NCBI

132 

Zhang J, Li S, Zhao Y, Ma P, Cao Y, Liu C, Zhang X, Wang W, Chen L and Li Y: Cancer-associated fibroblasts promote the migration and invasion of gastric cancer cells via activating IL-17a/JAK2/STAT3 signaling. Ann Transl Med. 8:8772020. View Article : Google Scholar : PubMed/NCBI

133 

Karabulut M, Usul Afsar C, Serimez M and Karabulut S: Serum IL-17 levels can be diagnostic for gastric cancer. J BUON. 24:1601–1609. 2019.PubMed/NCBI

134 

Park H, Li Z, Yang XO, Chang SH, Nurieva R, Wang YH, Wang Y, Hood L, Zhu Z, Tian Q and Dong C: A distinct lineage of CD4 T cells regulates tissue inflammation by producing interleukin 17. Nat Immunol. 6:1133–1141. 2005. View Article : Google Scholar : PubMed/NCBI

135 

Onishi RM and Gaffen SL: Interleukin-17 and its target genes: Mechanisms of interleukin-17 function in disease. Immunology. 129:311–321. 2010. View Article : Google Scholar : PubMed/NCBI

136 

Otani K, Watanabe T, Tanigawa T, Okazaki H, Yamagami H, Watanabe K, Tominaga K, Fujiwara Y, Oshitani N and Arakawa T: Anti-inflammatory effects of IL-17A on Helicobacter pylori-induced gastritis. Biochem Biophys Res Commun. 382:252–258. 2009. View Article : Google Scholar : PubMed/NCBI

137 

Chong Y, Yu D, Lu Z and Nie F: Role and research progress of spasmolytic polypeptide-expressing metaplasia in gastric cancer (Review). Int J Oncol. 64:332024. View Article : Google Scholar :

138 

O'Keefe RN, Carli ALE, Baloyan D, Chisanga D, Shi W, Afshar-Sterle S, Eissmann MF, Poh AR, Pal B, Seillet C, et al: A tuft cell-ILC2 signaling circuit provides therapeutic targets to inhibit gastric metaplasia and tumor development. Nat Commun. 14:68722023. View Article : Google Scholar

139 

von Moltke J, Ji M, Liang HE and Locksley RM: Tuft-cell-derived IL-25 regulates an intestinal ILC2-epithelial response circuit. Nature. 529:221–225. 2016. View Article : Google Scholar

140 

Waugh DJ and Wilson C: The interleukin-8 pathway in cancer. Clin Cancer Res. 14:6735–6741. 2008. View Article : Google Scholar : PubMed/NCBI

141 

Brat DJ, Bellail AC and Van Meir EG: The role of interleukin-8 and its receptors in gliomagenesis and tumoral angiogenesis. Neuro Oncol. 7:122–133. 2005. View Article : Google Scholar : PubMed/NCBI

142 

He R, Li X, Zhang S, Liu Y, Xue Q, Luo Y, Yu B, Li X and Liu Z: Dexamethasone inhibits IL-8 via glycolysis and mitochondria-related pathway to regulate inflammatory pain. BMC Anesthesiol. 23:3172023. View Article : Google Scholar : PubMed/NCBI

143 

Lee KE, Khoi PN, Xia Y, Park JS, Joo YE, Kim KK, Choi SY and Jung YD: Helicobacter pylori and interleukin-8 in gastric cancer. World J Gastroenterol. 19:8192–8202. 2013. View Article : Google Scholar : PubMed/NCBI

144 

Noto CN, Hoft SG, Bockerstett KA, Jackson NM, Ford EL, Vest LS and DiPaolo RJ: IL13 acts directly on gastric epithelial cells to promote metaplasia development during chronic gastritis. Cell Mol Gastroenterol Hepatol. 13:623–642. 2022. View Article : Google Scholar :

145 

Zhao R, Cao G, Zhang B, Wei L, Zhang X, Jin M, He B, Zhang B, He Z and Bie Q: TNF+ regulatory T cells regulate the stemness of gastric cancer cells through the IL13/STAT3 pathway. Front Oncol. 13:11629382023. View Article : Google Scholar : PubMed/NCBI

146 

Grünig G, Warnock M, Wakil AE, Venkayya R, Brombacher F, Rennick DM, Sheppard D, Mohrs M, Donaldson DD, Locksley RM and Corry DB: Requirement for IL-13 independently of IL-4 in experimental asthma. Science. 282:2261–2263. 1998. View Article : Google Scholar : PubMed/NCBI

147 

Seyfizadeh N, Seyfizadeh N, Gharibi T and Babaloo Z: Interleukin-13 as an important cytokine: A review on its roles in some human diseases. Acta Microbiol Immunol Hung. 62:341–378. 2015. View Article : Google Scholar : PubMed/NCBI

148 

Bockerstett K, Lewis SA, Wolf KJ, Noto CN, Jackson NM, Ford EL, Ahn TH and DiPaolo RJ: Single-cell transcriptional analyses of spasmolytic polypeptide-expressing metaplasia arising from acute drug injury and chronic inflammation in the stomach. Gut. 69:1027–1038. 2020. View Article : Google Scholar

149 

Miska J, Lui JB, Toomer KH, Devarajan P, Cai X, Houghton J, Lopez DM, Abreu MT, Wang G and Chen Z: Initiation of inflammatory tumorigenesis by CTLA4 insufficiency due to type 2 cytokines. J Exp Med. 215:841–858. 2018. View Article : Google Scholar : PubMed/NCBI

150 

Dahl CA, Schall RP, He HL and Cairns JS: Identification of a novel gene expressed in activated natural killer cells and T cells. J Immunol. 148:597–603. 1992. View Article : Google Scholar : PubMed/NCBI

151 

Meng D, Dong H, Wang C, Zang R and Wang J: Role of interleukin-32 in cancer progression (review). Oncol Lett. 27:542024. View Article : Google Scholar

152 

Han S and Yang Y: Interleukin-32: Frenemy in cancer? BMB Rep. 52:165–174. 2019. View Article : Google Scholar : PubMed/NCBI

153 

Fung KY, Nguyen PM and Putoczki TL: Emerging roles for Interleukin-18 in the gastrointestinal tumor microenvironment. Adv Exp Med Biol. 1240:59–72. 2020. View Article : Google Scholar : PubMed/NCBI

154 

Khawar MB, Abbasi MH and Sheikh N: IL-32: A novel pluripotent inflammatory interleukin, towards gastric inflammation, gastric cancer, and chronic rhino sinusitis. Mediators Inflamm. 2016:84137682016. View Article : Google Scholar : PubMed/NCBI

155 

Bagheri V, Memar B, Momtazi AA, Sahebkar A, Gholamin M and Abbaszadegan MR: Cytokine networks and their association with Helicobacter pylori infection in gastric carcinoma. J Cell Physiol. 233:2791–2803. 2018. View Article : Google Scholar

156 

Li ML, Hong XX, Zhang WJ, Liang YZ, Cai TT, Xu YF, Pan HF, Kang JY, Guo SJ and Li HW: Helicobacter pylori plays a key role in gastric adenocarcinoma induced by spasmolytic polypeptide-expressing metaplasia. World J Clin Cases. 11:3714–3724. 2023. View Article : Google Scholar : PubMed/NCBI

157 

Kuo HY, Chang WL, Yeh YC, Cheng HC, Tsai YC, Wu CT, Lin SH, Yang HB, Lu CC and Sheu BS: Spasmolytic polypeptide-expressing metaplasia associated with higher expressions of miR-21, 155, and 223 can be regressed by Helicobacter pylori eradication in the gastric cancer familial relatives. Helicobacter. 24:e125782019. View Article : Google Scholar : PubMed/NCBI

158 

Yamaoka Y, Kodama T, Kita M, Imanishi J, Kashima K and Graham DY: Relation between clinical presentation, Helicobacter pylori density, interleukin 1beta and 8 production, and cagA status. Gut. 45:804–811. 1999. View Article : Google Scholar : PubMed/NCBI

159 

El-Omar EM, Carrington M, Chow WH, McColl KE, Bream JH, Young HA, Herrera J, Lissowska J, Yuan CC, Rothman N, et al: Interleukin-1 polymorphisms associated with increased risk of gastric cancer. Nature. 404:398–402. 2000. View Article : Google Scholar : PubMed/NCBI

160 

Jackson CB, Judd LM, Menheniott TR, Kronborg I, Dow C, Yeomans ND, Boussioutas A, Robb L and Giraud AS: Augmented gp130-mediated cytokine signalling accompanies human gastric cancer progression. J Pathol. 213:140–151. 2007. View Article : Google Scholar : PubMed/NCBI

161 

Yamaoka Y, Kita M, Kodama T, Sawai N and Imanishi J: Helicobacter pylori cagA gene and expression of cytokine messenger RNA in gastric mucosa. Gastroenterology. 110:1744–1752. 1996. View Article : Google Scholar : PubMed/NCBI

162 

Balkwill F: Tumour necrosis factor and cancer. Nat Rev Cancer. 9:361–371. 2009. View Article : Google Scholar : PubMed/NCBI

163 

Adamsson J, Ottsjö LS, Lundin SB, Svennerholm AM and Raghavan S: Gastric expression of IL-17A and IFNγ in Helicobacter pylori infected individuals is related to symptoms. Cytokine. 99:30–34. 2017. View Article : Google Scholar : PubMed/NCBI

164 

Wang F, Meng W, Wang B and Qiao L: Helicobacter pylori-induced gastric inflammation and gastric cancer. Cancer Lett. 345:196–202. 2014. View Article : Google Scholar

165 

Goldenring JR, Ray GS, Coffey RJ, Meunier PC, Haley PJ, Barnes TB and Car BD: Reversible drug-induced oxyntic atrophy in rats. Gastroenterology. 118:1080–1093. 2000. View Article : Google Scholar : PubMed/NCBI

166 

Nam KT, Lee HJ, Sousa JF, Weis VG, O'Neal RL, Finke PE, Romero-Gallo J, Shi G, Mills JC, Peek RM Jr, et al: Mature chief cells are cryptic progenitors for metaplasia in the stomach. Gastroenterology. 139:2028–2037.e9. 2010. View Article : Google Scholar : PubMed/NCBI

167 

Weis VG, Sousa JF, LaFleur BJ, Nam KT, Weis JA, Finke PE, Ameen NA, Fox JG and Goldenring JR: Heterogeneity in mouse spasmolytic polypeptide-expressing metaplasia lineages identifies markers of metaplastic progression. Gut. 62:1270–1279. 2013. View Article : Google Scholar

168 

Nomura S, Yamaguchi H, Ogawa M, Wang TC, Lee JR and Goldenring JR: Alterations in gastric mucosal lineages induced by acute oxyntic atrophy in wild-type and gastrin-deficient mice. Am J Physiol Gastrointest Liver Physiol. 288:G362–G375. 2005. View Article : Google Scholar : PubMed/NCBI

169 

Cardoso CM, Custódio JB, Almeida LM and Moreno AJ: Mechanisms of the deleterious effects of tamoxifen on mitochondrial respiration rate and phosphorylation efficiency. Toxicol Appl Pharmacol. 176:145–152. 2001. View Article : Google Scholar : PubMed/NCBI

170 

Huh WJ, Khurana SS, Geahlen JH, Kohli K, Waller RA and Mills JC: Tamoxifen induces rapid, reversible atrophy, and metaplasia in mouse stomach. Gastroenterology. 142:21–24.e27. 2012. View Article : Google Scholar

171 

Kudo-Saito C, Imazeki H, Nagashima K, Shoji H, Tsugaru K, Takahashi N, Kawakami T, Amanuma Y, Wakatsuki T, Okano N, et al: IL33-ST2 axis is a predictive biomarker for anti-PD1 therapeutic efficacy in advanced gastric cancer. J Transl Med. 23:11252025. View Article : Google Scholar : PubMed/NCBI

172 

Leidner R, Conlon K, McNeel DG, Wang-Gillam A, Gupta S, Wesolowski R, Chaudhari M, Hassounah N, Lee JB, Ho Lee L, et al: First-in-human phase I/Ib study of NIZ985, a recombinant heterodimer of IL-15 and IL-15Rα, as a single agent and in combination with spartalizumab in patients with advanced and metastatic solid tumors. J Immunother Cancer. 11:e0077252023. View Article : Google Scholar

173 

Lordick F, Thuss-Patience P, Bitzer M, Maurus D, Sahin U and Türeci Ö: Immunological effects and activity of multiple doses of zolbetuximab in combination with zoledronic acid and interleukin-2 in a phase 1 study in patients with advanced gastric and gastroesophageal junction cancer. J Cancer Res Clin Oncol. 149:5937–5950. 2023. View Article : Google Scholar : PubMed/NCBI

174 

Gooderham M, Ameen M, Hong HC, Katoh N, Langley RG, Peris K, Reich K, Silvestre JF, Simpson E, Werfel T, et al: Efficacy of up to 4 years of tralokinumab in adults with moderate-to-severe atopic dermatitis. J Eur Acad Dermatol Venereol. Nov 11–2025. View Article : Google Scholar : Epub ahead of print.

175 

Song EJ, Ehst B, Glick B, Lewitt GM, Rich P, Ezra N, Bagel J, Anschutz T, Bialik B, Duan C, et al: Efficacy and safety of risankizumab in genital or scalp psoriasis in the UnlIMMited phase 4 randomized clinical trial at week 16. Dermatol Ther (Heidelb). Oct 25–2025. View Article : Google Scholar : Epub ahead of print.

176 

Yu S, Yang M, Lim KM, Cho Y, Kim H, Lee K, Jeong SH, Coffey RJ, Goldenring JR and Nam KT: Expression of LRIG1, a negative regulator of EGFR, is dynamically altered during different stages of gastric carcinogenesis. Am J Pathol. 188:2912–2923. 2018. View Article : Google Scholar : PubMed/NCBI

177 

Dickerson LK, Carter JA, Kohli K and Pillarisetty VG: Emerging interleukin targets in the tumour microenvironment: Implications for the treatment of gastrointestinal tumours. Gut. 72:1592–1606. 2023. View Article : Google Scholar : PubMed/NCBI

178 

Mohamed AH, Ahmed AT, Al Abdulmonem W, Bokov DO, Shafie A, Al-Hetty HRAK, Hsu CY, Alissa M, Nazir S, Jamali MC and Mudhafar M: Interleukin-6 serves as a critical factor in various cancer progression and therapy. Med Oncol. 41:1822024. View Article : Google Scholar : PubMed/NCBI

179 

Tanigawa K and Redmond WL: Current landscape and future prospects of interleukin-2 receptor (IL-2R) agonists in cancer immunotherapy. Oncoimmunology. 14:24526542025. View Article : Google Scholar : PubMed/NCBI

180 

Dong C, Tan D, Sun H, Li Z, Zhang L, Zheng Y, Liu S, Zhang Y and He Q: Interleukin-12 delivery strategies and advances in tumor immunotherapy. Curr Issues Mol Biol. 46:11548–11579. 2024. View Article : Google Scholar : PubMed/NCBI

181 

Xu Y, Sun X and Tong Y: Interleukin-12 in multimodal tumor therapies for induction of anti-tumor immunity. Discov Oncol. 15:1702024. View Article : Google Scholar : PubMed/NCBI

182 

Lee HM, Lee HJ and Chang JE: Inflammatory cytokine: An attractive target for cancer treatment. Biomedicines. 10:21162022. View Article : Google Scholar : PubMed/NCBI

183 

Yi M, Li T, Niu M, Zhang H, Wu Y, Wu K and Dai Z: Targeting cytokine and chemokine signaling pathways for cancer therapy. Signal Transduct Target Ther. 9:1762024. View Article : Google Scholar : PubMed/NCBI

184 

Holder PG, Lim SA, Huang CS, Sharma P, Dagdas YS, Bulutoglu B and Sockolosky JT: Engineering interferons and interleukins for cancer immunotherapy. Adv Drug Deliv Rev. 182:1141122022. View Article : Google Scholar : PubMed/NCBI

185 

Tomasovic LM, Liu K, VanDyke D, Fabilane CS and Spangler JB: Molecular engineering of interleukin-2 for enhanced therapeutic activity in autoimmune diseases. BioDrugs. 38:227–248. 2024. View Article : Google Scholar :

186 

Jeong H, Lee B, Kim KH, Cho SY, Cho Y, Park J, Lee Y, Oh Y, Hwang BR, Jang AR, et al: WFDC2 Promotes spasmolytic Polypeptide-Expressing metaplasia through the Up-Regulation of IL33 in response to injury. Gastroenterology. 161:953–967.e15. 2021. View Article : Google Scholar

Related Articles

  • Abstract
  • View
  • Download
  • Twitter
Copy and paste a formatted citation
Spandidos Publications style
Ma J, Zhan T, Zhang X, Gao W, Zhao S and Li H: Roles of interleukins in spasmolytic polypeptide‑expressing metaplasia (Review). Int J Oncol 68: 17, 2026.
APA
Ma, J., Zhan, T., Zhang, X., Gao, W., Zhao, S., & Li, H. (2026). Roles of interleukins in spasmolytic polypeptide‑expressing metaplasia (Review). International Journal of Oncology, 68, 17. https://doi.org/10.3892/ijo.2025.5830
MLA
Ma, J., Zhan, T., Zhang, X., Gao, W., Zhao, S., Li, H."Roles of interleukins in spasmolytic polypeptide‑expressing metaplasia (Review)". International Journal of Oncology 68.2 (2026): 17.
Chicago
Ma, J., Zhan, T., Zhang, X., Gao, W., Zhao, S., Li, H."Roles of interleukins in spasmolytic polypeptide‑expressing metaplasia (Review)". International Journal of Oncology 68, no. 2 (2026): 17. https://doi.org/10.3892/ijo.2025.5830
Copy and paste a formatted citation
x
Spandidos Publications style
Ma J, Zhan T, Zhang X, Gao W, Zhao S and Li H: Roles of interleukins in spasmolytic polypeptide‑expressing metaplasia (Review). Int J Oncol 68: 17, 2026.
APA
Ma, J., Zhan, T., Zhang, X., Gao, W., Zhao, S., & Li, H. (2026). Roles of interleukins in spasmolytic polypeptide‑expressing metaplasia (Review). International Journal of Oncology, 68, 17. https://doi.org/10.3892/ijo.2025.5830
MLA
Ma, J., Zhan, T., Zhang, X., Gao, W., Zhao, S., Li, H."Roles of interleukins in spasmolytic polypeptide‑expressing metaplasia (Review)". International Journal of Oncology 68.2 (2026): 17.
Chicago
Ma, J., Zhan, T., Zhang, X., Gao, W., Zhao, S., Li, H."Roles of interleukins in spasmolytic polypeptide‑expressing metaplasia (Review)". International Journal of Oncology 68, no. 2 (2026): 17. https://doi.org/10.3892/ijo.2025.5830
Follow us
  • Twitter
  • LinkedIn
  • Facebook
About
  • Spandidos Publications
  • Careers
  • Cookie Policy
  • Privacy Policy
How can we help?
  • Help
  • Live Chat
  • Contact
  • Email to our Support Team