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

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

Full Size Image

Sign up for eToc alerts
Recommend to Library

Journals

International Journal of Molecular Medicine

International Journal of Molecular Medicine

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

International Journal of Oncology

International Journal of Oncology

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

Molecular Medicine Reports

Molecular Medicine Reports

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

Oncology Reports

Oncology Reports

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

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine

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

Oncology Letters

Oncology Letters

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

Biomedical Reports

Biomedical Reports

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

Molecular and Clinical Oncology

Molecular and Clinical Oncology

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

World Academy of Sciences Journal

World Academy of Sciences Journal

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

International Journal of Functional Nutrition

International Journal of Functional Nutrition

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

International Journal of Epigenetics

International Journal of Epigenetics

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

Medicine International

Medicine International

An International Open Access Journal Devoted to General Medicine.

Journal Cover
March-2026 Volume 57 Issue 3

Full Size Image

Sign up for eToc alerts
Recommend to Library

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

ZG16B: A key regulator of tumor progression and immune microenvironment modulation in cancer (Review)

  • Authors:
    • Xing-Ming Chen
    • Yu-Bo Liang
    • Jin-Xiang Zuo
    • Zi-Sheng Yang
    • Le-Yu Zhang
    • Xin-Yu Zhang
    • Ping Wan
    • Yang Ke
  • View Affiliations / Copyright

    Affiliations: Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, P.R. China, College of Clinical Oncology, Kunming Medical University, Kunming, Yunnan 650118, P.R. China, School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan 650500, P.R. China, Department of Gastroenterology, The First People's Hospital of Yunnan Province, Kunming, Yunnan 650032, P.R. China
    Copyright: © Chen et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
  • Article Number: 58
    |
    Published online on: January 9, 2026
       https://doi.org/10.3892/ijmm.2026.5729
  • 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

Zymogen granule protein 16B (ZG16B), also known as pancreatic adenocarcinoma upregulated factor, is a secretory lectin‑like glycoprotein that serves a crucial role in tumorigenesis and immune regulation. The present review summarizes the latest research progress on the molecular characteristics, biological functions, signaling pathway regulation and clinical importance of ZG16B. Structurally, ZG16B contains an N‑terminal hydrophobic signal peptide, a jacalin‑related lectin domain and a C‑terminal extension. Functionally, ZG16B promotes tumor cell proliferation, migration, invasion and angiogenesis, and increases vascular permeability by activating the Toll‑like receptor, C‑X‑C chemokine receptor type 4, β‑catenin and focal adhesion kinase signaling pathways. In the tumor microenvironment, ZG16B can modulate immune responses, enhance the immunosuppressive functions of myeloid‑derived suppressor cells and M2 macrophages, and also promote the maturation of dendritic cells. Clinically, ZG16B expression is upregulated in pancreatic cancer, ovarian cancer, colorectal cancer, gastric cancer and oral cancer, and its upregulation is associated with a worse prognosis in these malignancies. Several ZG16B‑specific therapeutic strategies, including monoclonal antibodies, RNA aptamers and trans‑splicing ribozymes, have shown preclinical efficacy against malignant tumors. Furthermore, a clinical trial is currently testing the efficacy and safety of PBP1510, a humanized ZG16B antibody, for the treatment of advanced pancreatic cancer. In conclusion, ZG16B may be considered a novel target for cancer diagnosis, prognosis and therapy.

Introduction

The development and progression of cancer is a complex, multi-step process driven by factors such as the aberrant expression of multiple genes, disruption of key signaling pathways and remodeling of the tumor microenvironment (TME) (1-7). A deeper understanding of the mechanisms underlying the key drivers of cancer is crucial for revealing the malignant nature of tumors, and developing novel diagnostic and therapeutic strategies (8-10).

Zymogen granule protein 16B (ZG16B), also known as pancreatic adenocarcinoma upregulated factor, was discovered and identified in 2009, and was revealed to be highly expressed in pancreatic ductal adenocarcinoma (11). ZG16B is a secretory lectin-like protein (12), capable of regulating intracellular signaling cascades in various cell types, including tumor cells, endothelial cells and immune cells. ZG16B is involved in key biological processes, such as tumor cell proliferation, migration, invasion, angiogenesis and immune suppression within the TME (13-16). As an oncogenic factor, ZG16B expression is upregulated in malignant tumors, such as pancreatic cancer, ovarian cancer, colorectal cancer and gastric cancer, and is closely associated with the tumorigenesis and progression of these types of cancer (17-19). Currently, drugs targeting ZG16B are under investigation in preclinical studies and clinical trials, with promising prospects for future clinical application (20-25).

The present review aims to integrate the existing research findings, and systematically elucidate the molecular characteristics, biological functions, signaling pathway regulatory networks, expression profiles and clinical relevance of ZG16B. In addition, new advancements in the roles of ZG16B in cancer diagnosis and prognosis, and its potential in targeted therapy, are described. The current review not only provides a new perspective for understanding the complex mechanisms underlying tumor malignant progression, but also lays the theoretical foundation for the development of ZG16B-based diagnostic and therapeutic strategies, which holds notable scientific and clinical value in improving patient prognosis.

Structural characteristics of ZG16B

The ZG16B gene is located on the short arm of chromosome 16 at region 13.3 in humans, an area that has several genes related to secretory functions, such as WFIKKN1, PRSS22, TPSB2, NTN3 and TSC2 (26,27). ZG16B consists of four exons (11), and is the human homolog of the mouse demilune cell and parotid protein 1 gene and the rat common salivary protein 1 gene (28,29).

The ZG16B protein is a secretory lectin-like protein that predominantly exists as a monomer in solution (11). The initially translated ZG16B precursor consists of 172 amino acids (aa) and contains three major regions: An N-terminal hydrophobic signal peptide (1-16 aa), a central jacalin-related lectin domain (17-149 aa), and a C-terminal extension (150-172 aa) (Fig. 1A) (12). After cleavage of the signal peptide, the mature protein comprises 156 aa. The theoretical molecular mass of ZG16B is ~17 kDa, whereas the secreted form exhibits an apparent molecular weight of ~25 kDa due to N-linked glycosylation (11). The N-terminal signal peptide directs ZG16B into the endoplasmic reticulum-Golgi secretory pathway, ensuring its proper secretion and transport (30).

Structure and functional domains of
ZG16B. (A) A schematic diagram of ZG16B, including the N-terminal
signal peptide (yellow, 1-16 aa), jacalin-related lectin domain
(pink, 17-149 aa) and C-terminal extension (turquoise, 150-172 aa).
The key post-translational modification sites include one
N-glycosylation site (black diamond, 161-165 aa), two
phosphorylation sites (blue triangles, 29-32 and 101-104 aa) and
three N-myristoylation sites (red triangles, 40-45, 66-71 and
113-119 aa). (B) Predicted crystal structure of ZG16B. The β-prism
fold (pink) includes three β-sheets, with an α-helix at the
N-terminus (yellow) and a C-terminal extension (turquoise). The
structure was modeled using Swiss-Model and PyMOL software. aa,
amino acid; ZG16B, zymogen granule protein 16B.

Figure 1

Structure and functional domains of ZG16B. (A) A schematic diagram of ZG16B, including the N-terminal signal peptide (yellow, 1-16 aa), jacalin-related lectin domain (pink, 17-149 aa) and C-terminal extension (turquoise, 150-172 aa). The key post-translational modification sites include one N-glycosylation site (black diamond, 161-165 aa), two phosphorylation sites (blue triangles, 29-32 and 101-104 aa) and three N-myristoylation sites (red triangles, 40-45, 66-71 and 113-119 aa). (B) Predicted crystal structure of ZG16B. The β-prism fold (pink) includes three β-sheets, with an α-helix at the N-terminus (yellow) and a C-terminal extension (turquoise). The structure was modeled using Swiss-Model and PyMOL software. aa, amino acid; ZG16B, zymogen granule protein 16B.

The conserved GXXXD motif at the carbohydrate-binding site is crucial for the carbohydrate-binding specificity of proteins, and replacing this motif with the QLLGIK sequence in ZG16B alters its carbohydrate-binding preference, abolishing its ability to bind monosaccharides (12,31). Surrounding this site, positively charged residues Lys87, Arg131, and Lys147 form a potential glycosaminoglycan-binding region (11). The lectin domain also harbors several potential post-translational modification (PTM) sites, which may be essential for protein function and stability (Fig. 1A); these PTM sites include two phosphorylation sites at aa 29-32 and 101-104, and three N-myristoylation sites at aa 40-45, 66-71 and 113-119 (11). The C-terminal extension contains a classical N-linked glycosylation motif (NXS/T; Fig. 1A).

For the present review, Swiss-Model (www.swissmodel.expasy.org) and PyMOL software (version 3.1; www.pymol.org) were used to build the protein structure model (Fig. 1B) (32,33). Within the central jacalin-related lectin domain, ZG16B adopts a characteristic β-prism fold composed of three β-sheets that form a triangular prism-like spatial conformation, with each β-sheet containing 3-4 β-strands (Fig. 1B) (11,12). This domain is responsible for recognizing glycoproteins and mediating cell-cell communication, host-pathogen interaction, cancer metastasis, embryonic development and tissue morphogenesis (11,12).

Evolutionarily, the ZG16B protein is highly conserved among primates. The aa sequence identity between humans and chimpanzees reaches 98% (with 100% similarity), and that between humans and rhesus monkeys is 92% (with 95% similarity) (11). By contrast, the conservation of the ZG16B protein is reduced in rodents with 31% identity and 45% similarity between humans and mice, suggesting that ZG16B is a rapidly evolving, primate-specific gene likely associated with specialized physiological functions in primates (11).

Paralogous homolog of ZG16B

ZG16 and ZG16B are paralogous homologs. ZG16 is located on the short arm of chromosome 16 at region 11.2 in humans, and encodes a secretory ZG16 protein that contains a jacalin-related lectin domain (12). However, structurally, ZG16 differs from ZG16B by having an additional short α-helix and a β-strand (12). Functionally, ZG16 exhibits various carbohydrate-binding abilities, primarily including chondroitin sulfate, heparan sulfate, and heparin (34-36). ZG16 has an important role in immune defense by recognizing specific glycan structures (37,38) and has anticancer effects (39,40). Additionally, ZG16 can regulate the channel switch on the membrane of zymogen granules in pancreatic acinar cells (41,42), participating in the concentration sorting and exocytosis of zymogen granules (43-46).

Biological functions of ZG16B

ZG16B can be secreted by tumor cells and can act on tumor cells through autocrine or paracrine mechanisms to promote their proliferation, migration and invasion. ZG16B can also affect vascular endothelial cells in the TME, enhancing angiogenesis and vascular permeability. Additionally, ZG16B can modulate immune cells within the TME, regulating T cell-mediated anti-tumor immunity (Fig. 2).

Biological functions of ZG16B. ZG16B
promotes tumor cell proliferation, migration and invasion through
autocrine and paracrine signaling. In vascular endothelial cells,
ZG16B activates the ERK, PI3K/AKT and eNOS pathways to enhance
angiogenesis and upregulates CXCR4 expression, increasing
SDF-1-induced angiogenesis. ZG16B also promotes the phosphorylation
of VE-cadherin, disrupting endothelial junctions and increasing
vascular permeability. On MDSCs, ZG16B binds TLR4 to activate the
MEK1/2, ERK and JNK pathways, promoting arginase, NO and ROS
production to suppress T-cell function. On monocytes, ZG16B binds
TLR2, inducing migration to the tumor and differentiation into M2
macrophages, which secrete IL-10 and arginase to inhibit T-cell
responses. In DCs, ZG16B activates the TLR4/MyD88 pathway,
increasing IL-12p70 and decreasing IL-10 expression, enhancing T
cell-mediated immunity. AP-1, activator protein-1; CXCR4, C-X-C
chemokine receptor type 4; DCs, dendritic cells; eNOS, endothelial
nitric oxide synthase; MDSCs, myeloid-derived suppressor cells;
MEK1, mitogen-activated protein kinase kinase 1; NO, nitric oxide;
ROS, reactive oxygen species; SDF-1, stromal cell-derived factor-1;
TLR, Toll-like receptor; ZG16B, zymogen granule protein 16B.

Figure 2

Biological functions of ZG16B. ZG16B promotes tumor cell proliferation, migration and invasion through autocrine and paracrine signaling. In vascular endothelial cells, ZG16B activates the ERK, PI3K/AKT and eNOS pathways to enhance angiogenesis and upregulates CXCR4 expression, increasing SDF-1-induced angiogenesis. ZG16B also promotes the phosphorylation of VE-cadherin, disrupting endothelial junctions and increasing vascular permeability. On MDSCs, ZG16B binds TLR4 to activate the MEK1/2, ERK and JNK pathways, promoting arginase, NO and ROS production to suppress T-cell function. On monocytes, ZG16B binds TLR2, inducing migration to the tumor and differentiation into M2 macrophages, which secrete IL-10 and arginase to inhibit T-cell responses. In DCs, ZG16B activates the TLR4/MyD88 pathway, increasing IL-12p70 and decreasing IL-10 expression, enhancing T cell-mediated immunity. AP-1, activator protein-1; CXCR4, C-X-C chemokine receptor type 4; DCs, dendritic cells; eNOS, endothelial nitric oxide synthase; MDSCs, myeloid-derived suppressor cells; MEK1, mitogen-activated protein kinase kinase 1; NO, nitric oxide; ROS, reactive oxygen species; SDF-1, stromal cell-derived factor-1; TLR, Toll-like receptor; ZG16B, zymogen granule protein 16B.

ZG16B promotes tumor cell proliferation, migration and invasion

ZG16B induces rapid proliferation of pancreatic cancer cells by specifically activating the AKT/GSK-3β signaling pathway to stabilize β-catenin (47). Furthermore, ZG16B can activate the ERK, JNK and AKT signaling pathways to upregulate the expression of C-X-C chemokine receptor type 4 (CXCR4), promoting the in vitro proliferation, migration and invasion of pancreatic cancer cells, as well as in vivo metastasis of pancreatic cancer (15). Moreover, ZG16B interacts with Toll-like receptor 4 (TLR4) to activate the TLR4/MyD88/NF-κB and TLR4/MEK/ERK signaling pathways, and enhances the migration and invasion of pancreatic cancer cells (14,16). Notably, the high expression of ZG16B is significantly associated with positive lymph node metastasis and worse prognosis in patients with pancreatic ductal adenocarcinoma (17).

ZG16B activates the PI3K/AKT1 pathway to enhance the proliferation and migration of breast cancer cells. Notably, ZG16B-induced proliferation, but not migration, of breast cancer cells depends on ERK1/2 phosphorylation and the presence of the estrogen receptor (48). In ovarian cancer, ZG16B promotes cell proliferation by activating the TLR4-mediated ERK, JNK and p38 signaling pathways, while it enhances migration, invasion and adhesion through the Src, ERK and AKT signaling pathways (18,49). Similarly, ZG16B enhances the proliferation, migration, invasion and adhesion of colorectal cancer cells by regulating the Wnt/β-catenin signaling pathway. Notably, silencing ZG16B suppresses these processes by inducing cell-cycle arrest at the G0/G1 phase and apoptosis (19,50). Furthermore, ZG16B promotes the proliferation and invasion of oral squamous cell carcinoma cells while inhibiting apoptosis. High expression of ZG16B is significantly associated with positive lymph node metastasis and worse prognosis in patients with oral squamous cell carcinoma (51).

Collectively, ZG16B expression is upregulated in various types of cancer, and promotes the proliferation, migration and invasion of different types of cancer cells by regulating multiple signaling pathways.

ZG16B promotes angiogenesis and vascular permeability

Tumor angiogenesis and vascular density are closely associated with tumor growth, maintenance and metastasis (52-58). ZG16B has been identified as a new endothelial cell activator, distinct from vascular endothelial growth factors, and its expression is markedly associated with tumor microvascular density (59). ZG16B can activate ERK and PI3K/AKT signaling to induce the expression of endothelial nitric oxide synthase (eNOS) in a time- and dose-dependent manner, promoting the proliferation, migration and tubular network formation of human umbilical vein endothelial cells (HUVECs) (59). ZG16B also enhances angiogenesis by upregulating CXCR4 expression in HUVECs to enhance the angiogenic activity of its ligand, stromal cell-derived factor-1 (SDF-1) (59,60).

ZG16B can also induce the Src-dependent phosphorylation of VE-cadherin, resulting in the disruption of cell-cell junctions between endothelial cells to increase vascular permeability (59,61). This suggests that ZG16B may serve as a potential biomarker for angiogenesis-related diseases, and a novel molecular target for the development of antitumor angiogenesis therapy (62).

Regulation of immune cells in the TME by ZG16B

The TME is important for tumorigenesis, progression and immune response (63-69), and contains stromal myeloid-derived suppressor cells (MDSCs), tumor-associated macrophages (TAMs) and dendritic cells (DCs) (70-72).

Regulation of MDSCs by ZG16B

MDSCs are a group of immature myeloid cells, including immature granulocytes, monocytes and DC precursors (73,74). In the TME, MDSCs can suppress T-cell activity, block antitumor immune responses and promote tumor progression (75-79). ZG16B secreted by tumor cells can enhance the migration of MDSCs by upregulating CXCR4 expression, inducing their recruitment to tumor tissues (13). Additionally, ZG16B interacts with TLR4 on the surface of MDSCs to activate the MEK1/2, ERK, and JNK signaling and the transcription factor activator protein-1 (AP-1). This promotes the production of immune suppressive factors such as arginase, nitric oxide and reactive oxygen species in tumor-infiltrating MDSCs, inducing oxidative damage to T cells and inhibiting CD8+ T-cell responses, exerting an immunosuppressive effect (13).

Regulation of TAMs by ZG16B

TAMs are a type of specialized macrophages present in the TME. Depending on the stimulating signals, mature macrophages can differentiate into M1 and M2 types (80-85). M1 macrophages have antitumor functions, whereas M2 macrophages exhibit protumor functions and become the predominant macrophages during tumor progression, which are associated with a poor prognosis (86-88). ZG16B in the TME, through binding to TLR2 on monocytes, recruits circulating monocytes into the TME, where ZG16B can induce the differentiation of monocytes into M2 macrophages (89). The ZG16B-induced M2 macrophages can secrete immune suppressive factors such as IL-10 and arginase, inhibiting the proliferation of CD4+ T cells and the activity of CD8+ T cells. This weakens T cell-mediated antitumor immune responses, allowing tumor cells to evade immune surveillance, and promoting tumor growth and progression (89).

Regulation of DCs by ZG16B

DCs are the professional antigen-presenting cells in the TME, and their maturation and activation status directly influence the strength of T-cell immune responses (78,90-93). ZG16B activates TLR4/MyD88 signaling in DCs, promoting their maturation, activation and migration. ZG16B can also upregulate IL-12p70 expression and downregulate IL-10 expression in DCs, favoring the activation and differentiation of naïve CD4+ T cells into T helper 1 cells, enhancing the levels of cytotoxic T lymphocyte immune activity, and mediating antitumor immunity (94). Furthermore, when DCs treated with recombinant ZG16B are pulsed with tumor-associated antigen peptides HPV16 E7 and OVA257-264, they can activate antigen peptide-specific CD8+ T cells and induce memory T-cell responses, leading to effective antitumor responses and long-term tumor suppression, prolonging the survival of tumor-bearing mice. This result suggests that ZG16B may enhance the antitumor efficacy of DC vaccines and serve as a potential novel adjuvant in humans (94).

Regulation of signaling pathways by ZG16B

ZG16B in the TME can act on signaling pathways such as the TLR, CXCR4, β-catenin and focal adhesion kinase (FAK) pathways, regulating the functions of tumor cells, endothelial cells and immune cells (Fig. 3). Additionally, the expression of ZG16B in tumor cells can be induced by TGF-β.

Signaling pathways regulated by
ZG16B. (A) TLR signaling pathway. ZG16B activates TLR2-JNK, p38 and
IKK-β/TPL-2/MEK/ERK signaling, enhancing AP-1-mediated cytokine
expression, and promoting angiogenesis and metastasis. Through
TLR4, ZG16B triggers the MyD88/NF-κB and MEK/ERK pathways to
enhance tumor cell migration and invasion. (B) CXCR4 signaling
pathway. ZG16B activates the ERK/p90RSK and JNK pathways,
upregulating CXCR4 gene transcription to increase tumor
motility and metastasis. ZG16B also stimulates ERK and
PI3K/AKT/eNOS-dependent angiogenesis and enhances
SDF-1/CXCR4-mediated angiogenic signaling. Through the CXCR4-TLR2
co-receptor, ZG16B activates cAMP/PKA to suppress NF-κB activity
and reduce TNF-α production, contributing to immune escape. (C)
β-catenin signaling pathway. ZG16B activates AKT, inhibiting
β-catenin degradation and promoting its nuclear accumulation, which
induces TCF/LEF-dependent CCND1 and JUN transcription to drive
proliferation. SIRT1 counteracts this effect by promoting β-catenin
degradation. (D) FAK signaling pathway. ZG16B promotes formation of
the FAK/Src complex at focal adhesions to enhance integrin-mediated
adhesion and F-actin assembly. This signaling also inhibits
caspase-3 activity, supporting anoikis resistance. AP-1, activator
protein-1; ATF2, activating transcription factor 2; CCND1, cyclin
D1; CK-1α, casein kinase-1α; CREB, cAMP-responsive element binding
protein; CXCR4, C-X-C chemokine receptor type 4; eNOS, endothelial
nitric oxide synthase; FAK, focal adhesion kinase; JUN, c-Jun; LEF,
lymphoid enhancer factor; MIF, macrophage migration inhibitory
factor; PKA, protein kinase A; p90RSK, p90 ribosomal s6 kinase;
SDF-1, stromal cell-derived factor-1; SIRT1, sirtuin 1; TCF, T-cell
factor; TLR, Toll-like receptor; TPL-2, tumor progression locus 2;
ZG16B, zymogen granule protein 16B.

Figure 3

Signaling pathways regulated by ZG16B. (A) TLR signaling pathway. ZG16B activates TLR2-JNK, p38 and IKK-β/TPL-2/MEK/ERK signaling, enhancing AP-1-mediated cytokine expression, and promoting angiogenesis and metastasis. Through TLR4, ZG16B triggers the MyD88/NF-κB and MEK/ERK pathways to enhance tumor cell migration and invasion. (B) CXCR4 signaling pathway. ZG16B activates the ERK/p90RSK and JNK pathways, upregulating CXCR4 gene transcription to increase tumor motility and metastasis. ZG16B also stimulates ERK and PI3K/AKT/eNOS-dependent angiogenesis and enhances SDF-1/CXCR4-mediated angiogenic signaling. Through the CXCR4-TLR2 co-receptor, ZG16B activates cAMP/PKA to suppress NF-κB activity and reduce TNF-α production, contributing to immune escape. (C) β-catenin signaling pathway. ZG16B activates AKT, inhibiting β-catenin degradation and promoting its nuclear accumulation, which induces TCF/LEF-dependent CCND1 and JUN transcription to drive proliferation. SIRT1 counteracts this effect by promoting β-catenin degradation. (D) FAK signaling pathway. ZG16B promotes formation of the FAK/Src complex at focal adhesions to enhance integrin-mediated adhesion and F-actin assembly. This signaling also inhibits caspase-3 activity, supporting anoikis resistance. AP-1, activator protein-1; ATF2, activating transcription factor 2; CCND1, cyclin D1; CK-1α, casein kinase-1α; CREB, cAMP-responsive element binding protein; CXCR4, C-X-C chemokine receptor type 4; eNOS, endothelial nitric oxide synthase; FAK, focal adhesion kinase; JUN, c-Jun; LEF, lymphoid enhancer factor; MIF, macrophage migration inhibitory factor; PKA, protein kinase A; p90RSK, p90 ribosomal s6 kinase; SDF-1, stromal cell-derived factor-1; SIRT1, sirtuin 1; TCF, T-cell factor; TLR, Toll-like receptor; TPL-2, tumor progression locus 2; ZG16B, zymogen granule protein 16B.

ZG16B and the TLR signaling pathway

TLRs are key components of the innate immune system. As a type of pattern recognition receptor, they can recognize pathogen-associated molecular patterns and damage-associated molecular patterns, initiating both innate and adaptive immune responses (95-97). Additionally, TLR signaling regulates tumor cell proliferation, migration and invasion, and contributes to the process of immune evasion (98-100).

ZG16B can act as an endogenous ligand for TLR2 and TLR4 on the cell surface to activate the TLR2-mediated ERK, JNK and p38 signaling in human acute monocytic leukemia THP-1 cells (31). Similarly, ZG16B activates IKK-β and tumor progression locus 2/MEK/ERK signaling in 293T cells through exogenously expressed TLR2 (31). As a result, ZG16B increases AP-1 expression to promote the secretion of cytokines, such as RANTES, macrophage migration inhibitory factor and IL-1RA, which favor tumor progression, metastasis and angiogenesis (31). However, ZG16B does not activate TLR2 or TLR4-mediated NF-κB signaling in THP-1 and 293T cells (31). By contrast, ZG16B can induce the migration and invasion of pancreatic cancer cells by activating the TLR4/MyD88/NF-κB signaling, independent of the TLR4/TRIF pathway (16). Additionally, through TLR4, ZG16B activates MEK/ERK signaling to promote the migration and invasion of pancreatic cancer cells (14).

ZG16B and the CXCR4 signaling pathway

The CXCR4 signaling pathway is crucial for organ-specific metastasis in various types of malignancies. The increased expression of CXCR4 by ZG16B markedly enhances the migration and invasion of tumor cells (101-103).

In pancreatic cancer cells, ZG16B can induce rapid and transient activation of the ERK, JNK and PI3K/AKT signaling pathways in an autocrine manner. Activated ERK can activate p90 ribosomal s6 kinase, and downstream transcription factors cAMP-responsive element binding protein and ELK-1; activated JNK can activate the transcription factors c-Jun (JUN), activating transcription factor 2 and ELK-1. These transcription factors synergistically regulate the expression of CXCR4 to promote the motility and metastasis of pancreatic cancer cells (15). In addition, ZG16B enhances the responses of endothelial cells to SDF-1, the CXCR4 ligand, by activating the ERK and PI3K/AKT/eNOS signaling pathways, and upregulating CXCR4 expression, thus enhancing angiogenesis. This creates a TME that favors tumor progression and metastasis (59,60). In THP-1 cells, ZG16B binds to the CXCR4/TLR2 receptor complex to activate cAMP and cAMP-dependent protein kinase A (PKA). Subsequently, PKA inhibits the TLR2-mediated NF-κB activation, reducing TNF-α production and contributing to the immune evasion of tumor cells (31).

ZG16B and the β-catenin signaling pathway

The Wnt/β-catenin signaling is aberrantly activated in various types of cancer and is crucial for biological processes, such as tumor cell proliferation, differentiation and apoptosis resistance (104-108). In resting non-tumor cells, β-catenin binds to a destruction complex consisting of APC, Axin, casein kinase-1α (CK-1α) and GSK-3β. CK-1α and GSK-3β phosphorylate β-catenin at the Ser45 and Ser33/37/Thr41 sites, respectively, leading to β-catenin ubiquitination and proteasomal degradation to maintain β-catenin levels at a low baseline (109).

In pancreatic cancer cells, ZG16B can activate the AKT/GSK-3β pathway, leading to slight phosphorylation of β-catenin at Ser33/37/Thr41 and Ser675, while greatly reducing phosphorylation at Ser45, thus stabilizing and activating β-catenin (47). In the nucleus, β-catenin forms a complex with the T-cell factor/lymphoid enhancer factor family, activating the expression of β-catenin-targeted genes, such as CCND1 and JUN, to promote the rapid proliferation of pancreatic cancer cells (47). The deacetylase sirtuin 1 (SIRT1) can mediate the proteasomal degradation of β-catenin and inhibit the expression of the target gene CCND1, thereby suppressing the ZG16B-mediated pancreatic cancer cell proliferation, independent of SIRT1 nuclear translocation (110).

ZG16B and the FAK signaling pathway

FAK is a non-receptor tyrosine kinase usually present at cell focal adhesion sites. FAK is important for tumor cell adhesion, proliferation, migration, and regulating the interactions between tumor cells and the extracellular matrix. FAK is a marker of tumor progression and an indicator of poor prognosis (111-113).

In pancreatic cancer cells, ZG16B activates FAK signaling, which recruits Src to the focal adhesion sites, forming a stable FAK/Src complex; this can phosphorylate the scaffold protein paxillin to promote the formation of F-actin. This process increases the focal adhesion density and enhances tumor cell adhesion to extracellular matrix molecules, such as fibronectin, collagen I and vitronectin (114). Additionally, ZG16B activates the FAK/Src signaling to inhibit the activity of the apoptosis-related protein caspase-3, thereby enhancing the resistance of pancreatic cancer cells to anoikis, and promoting the progression and metastasis of pancreatic cancer (114,115).

TGF-β signaling pathway promotes ZG16B expression

TGF-β is a pleiotropic cytokine, which activates Smad signaling through its receptors; this is important in regulating processes such as immune regulation and wound healing (116,117). The TGF-β/Smad signaling pathway can promote TME remodeling by inducing the epithelial-mesenchymal transition (EMT), regulating angiogenesis, immune suppression, activating fibroblasts, extracellular matrix remodeling and inflammation modulation, ultimately promoting malignant tumor progression (118-120).

In pancreatic cancer cells, TGF-β binds to the TGF-β receptor 2 to activate the phosphorylation of Smad2/3, and facilitate the formation of a complex of Smad2/3 and Smad4 in the cytoplasm. Subsequently, this complex translocates into the nucleus and binds to the Smad-binding element in the ZG16B promoter, enhancing ZG16B expression (14). TGF-β/Smad-mediated ZG16B expression promotes the expression of transcription factors SNAI1 and ZEB1 via the MEK/ERK signaling pathway, upregulating mesenchymal vimentin and α-SMA expression, while downregulating epithelial E-cadherin and ZO-1 expression, inducing the EMT process in pancreatic cancer (14).

Relevance of ZG16B to tumors and its translational potential

Clinical relevance and prognostic importance of ZG16B in tumors

Physiologically, ZG16B is expressed at a low level in most non-tumor tissues, but it is highly expressed in the placenta (11). Additionally, ZG16B expression is markedly upregulated in various types of tumor tissues, including pancreatic cancer, colorectal cancer, ovarian cancer and gastric cancer (11). ZG16B is crucial for the pathogenesis of multiple types of cancer and disease, and ZG16B has broad activities and clinical potential.

ZG16B expression is low or undetectable in non-tumor pancreatic tissues, whereas its expression is notably upregulated in pancreatic cancer cells at the tumor margins. High ZG16B expression is associated with cancer progression and cachexia (15,17,121). Similarly, ZG16B is highly expressed in colorectal cancer (19,50), ovarian cancer (18,122) and cervical cancer (123,124), and its high expression is an independent prognostic risk factor, markedly associated with tumor stage progression, shorter overall survival, disease-free survival and progression-free survival. Furthermore, ZG16B is highly expressed in prostate cancer (125,126) and oral squamous cell carcinoma (51,127), supporting malignant behaviors. By contrast, high ZG16B expression in breast cancer is associated with a favorable prognosis (128). This may be explained by the fact that ZG16B is co-expressed with several tumor-suppressive genes, such as SPINT1, TFAP2A, FOXA1 and FAM96B in breast cancer; its high expression predominantly occurs in hormone receptor-positive breast cancers, which are intrinsically associated with better outcomes; and its upregulation is mainly driven by promoter demethylation rather than oncogenic mutations (128). Functionally, ZG16B can act as a marker of hormone receptor-related differentiation or luminal epithelial differentiation, but not a driver factor, or an indicator of tumor aggressiveness in breast cancer (128). This clinical association suggests that the function of ZG16B may vary among different types of cancer.

Additionally, ZG16B appears to participate in the physiological processes and pathogenesis of several non-tumor diseases. Notably, ZG16B is highly expressed in reflex tears (129) and amniotic fluid (130), constituting a part of their specific protein components. In the oral cavity, through its lectin structure, ZG16B binds to the cell wall lipopolysaccharides of the commensal bacterium Streptococcus vestibularis and interacts with salivary mucin 7. This inhibits the growth of these microorganisms through mucin-assisted clearance mechanisms, contributing to the maintenance of oral micro-ecological balance (131). ZG16B also binds to the lipoteichoic acid of the oral bacterium Enterococcus faecalis, inhibiting activation of the TLR2/NF-κB pathway in macrophages, alleviating inflammation and regulating immune responses to oral infections (132). ZG16B expression is downregulated in the oral salivary glands and saliva of patients with chronic graft-vs.-host disease, which may be related to salivary gland dysfunction (133). Furthermore, ZG16B has potential value as a biomarker or therapeutic target in diseases such as atherosclerosis (decreased expression in urine), acute coronary syndrome (decreased expression in urine) (134), dry eye syndrome (decreased expression in tear fluid) (135), multiple sclerosis (decreased expression in tear fluid) (136), Parkinson's disease (elevated expression in brain tissue) (137) and idiopathic pulmonary fibrosis (elevated expression in lung tissue) (138).

Overall, ZG16B shows promise as a biomarker for both diagnosis and prognosis. Its high expression in pancreatic, colorectal and ovarian cancers supports its potential for use in early diagnosis. Elevated ZG16B levels are also associated with worse survival in patients with colorectal or ovarian cancer, whereas they are linked to a more favorable prognosis in patients with hormone receptor-positive breast cancer. These findings highlight the value of ZG16B in cancer diagnosis and prognosis, with potential for tailored therapies.

Notably, while most current research focuses on the tissue-level expression of ZG16B and its relevance to cancer, clinical evidence supporting the detection of circulating ZG16B for early diagnosis or prognostic prediction in patient cohorts remains lacking (19). As a secreted protein with carbohydrate-binding sites and glycosaminoglycan-binding capabilities, ZG16B possesses the biological potential to be released into blood and other body fluids, making it a promising candidate for liquid biopsy (139-144). Future prospective cohort studies are required to validate critical parameters such as sensitivity, specificity and area under the curve for testing circulating ZG16B in patients with cancer, and to compare its performance with existing biomarkers.

Targeting ZG16B in tumor prevention and therapy

Targeting ZG16B has potential for tumor prevention and therapy in different types of cancer. In a mouse model susceptible to breast cancer, treatment with both the chemopreventive agents bexarotene and carvedilol has been reported to suppress ZG16B expression, reducing the proliferation and migration of non-malignant breast cells (48). Similarly, treatment with anti-ZG16B antibody, together with docetaxel displays strong antitumor effects in an ovarian cancer mouse model (49). Furthermore, inhibiting ZG16B expression in pancreatic cancer stem cells can reduce the expression levels of multidrug resistant protein 5 and ribonucleotide reductase M2, thus increasing the sensitivity of these cells to gemcitabine and 5-fluorouracil in a pancreatic cancer mouse model (145,146). Additionally, ZG16B can upregulate the expression of type I interferon-α receptor in pancreatic cancer cells to activate downstream TYK2 and STAT1, which increases their resistance to oncolytic virus H-1 infection, suggesting the important role of targeting ZG16B in oncolytic virotherapy (147).

Currently, there are various therapeutic strategies for targeting ZG16B. Kim et al (22) designed an RNA aptamer (P12FR2) that specifically binds to the ZG16B protein, and effectively inhibited pancreatic cancer growth. Kim et al (20) also developed a trans-splicing ribozyme (TSR) targeting ZG16B mRNA to selectively kill ZG16B+ tumor cells. Additionally, Kim et al (21) designed a humanized monoclonal antibody against ZG16B (PMAb83), which effectively inhibited the proliferation and invasiveness of pancreatic cancer cells, tumor angiogenesis, and suppressed tumor growth and metastasis.

A multicenter, open-label Phase I/IIa clinical trial is currently testing the efficacy and safety of humanized anti-ZG16B for patients with advanced pancreatic cancer (NCT05141149, ClinicalTrials.gov) (23-25). The study population consists of adults with locally advanced or metastatic pancreatic cancer who have relapsed in response to multiple lines of chemotherapy. These patients receive PBP1510, a novel humanized IgG1 monoclonal antibody against ZG16B, either as monotherapy or in combination with gemcitabine. If successful, this therapeutic strategy may offer new treatment options for patients with pancreatic cancer in the future.

In summary, targeting ZG16B offers promising therapeutic strategies across multiple types of cancer. Preclinical studies have demonstrated that inhibiting ZG16B expression in cancer models may reduce tumor cell proliferation, migration and resistance to chemotherapy. Therapeutic approaches include RNA aptamers, TSRs and monoclonal antibodies, such as PMAb83, which have potential to inhibit tumor growth, metastasis and angiogenesis. Additionally, the ongoing clinical trial of PBP1510, a monoclonal antibody against ZG16B for patients with pancreatic cancer, holds potential for providing new treatment options for patients with advanced cancer.

Future research directions for ZG16B

ZG16B is a promising regulator in tumor progression and immune responses, but several challenges remain. The lack of high-resolution structural data, unclear role of PTMs and unidentified receptors hinder a full understanding of its molecular mechanisms. Its multifunctional roles and varied expression across diseases further complicate its therapeutic targeting. Overcoming these challenges will require advanced structural studies, molecular profiling and disease-specific models to fully explore its therapeutic potential.

Structural and functional studies

Future research should clarify the carbohydrate-binding specificity of the ZG16B lectin domain and determine how PTMs, such as phosphorylation, N-myristoylation and glycosylation, affect its structural stability and downstream signaling. Integrating high-resolution structural approaches with site-directed mutagenesis or PTM-deficient ZG16B mutants will enable to precisely map its modification-dependent functional motifs. These studies will reveal how ZG16B orchestrates tumor progression and immune modulation at the molecular level.

Molecular regulatory network studies

Defining the upstream and downstream regulatory networks of ZG16B remains essential for understanding its pleiotropic roles across different types of cancer. Particular attention should be paid to transcriptional and post-transcriptional regulation by noncoding RNAs (such as microRNAs and long noncoding RNAs) as well as modulation by major oncogenic pathways, such as NF-κB, MAPK and PI3K/AKT. Parallel efforts to identify and validate ZG16B receptors, including integrins, TLR family members or as-yet-uncharacterized surface proteins, will clarify how extracellular ZG16B is converted into cellular responses. CRISPR-based regulatory screening and multi-omics profiling may accelerate the discovery of ZG16B-centered signaling hubs and refine the understanding of its functional connectivity in the TME.

Drug design

A deeper understanding of ZG16B-receptor interactions will lay the foundation for developing ZG16B-targeted precision therapies. High-resolution structural studies should map critical binding interfaces to support rational design of small-molecule inhibitors using computational docking and molecular dynamics modeling. Concurrently, developing monoclonal antibodies or biologics that disrupt ZG16B or its receptor engagement may provide new options for modulating immune suppression or tumor progression. Rigorous evaluation of these therapeutic strategies in in vitro and in vivo models, particularly in tumors with high ZG16B expression, will be crucial for determining feasibility and guiding translational development.

Exploration in non-tumor diseases

ZG16B expression dysregulation in cardiovascular, neurological and autoimmune diseases suggests that its biological relevance extends far beyond malignancy. Investigating its roles in chronic inflammation, endothelial activation or immune dysregulation using disease-specific models, such as atherosclerosis, neuroinflammation or autoimmune liver disease, may reveal conserved mechanisms shared with cancer biology. These studies have the potential not only to define ZG16B as a cross-disease biomarker but also to provide therapeutic opportunities in non-oncological conditions where ZG16B-driven pathways remain largely unexplored.

Conclusion

The present review comprehensively summarizes the research progress on the oncogenic roles of ZG16B. As a secretory protein, ZG16B can activate multiple signaling pathways, including TLR, CXCR4, β-catenin and FAK, to promote tumor cell proliferation, migration, invasion and angiogenesis. ZG16B is crucial for tumor development and metastasis by reshaping the TME and regulating immune cell functions, including MDSCs, TAMs and DCs. ZG16B is highly expressed in various types of cancer, and also has potential value for non-invasive tumor diagnosis and prognosis. Furthermore, treatment strategies targeting ZG16B have potential to serve as new therapeutic options for multiple types of cancer.

Availability of data and materials

Not applicable.

Authors' contributions

PW and YK conceived and designed the study. XMC, YBL, JXZ, ZSY, LYZ and XYZ contributed equally to literature review, interpretation, synthesis, and writing the first draft of the manuscript and preparing the figures. PW and YK edited the manuscript and were responsible for critical revisions. Data authentication is not applicable. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgements

Not applicable.

Funding

This work was supported by grants from the National Natural Science Foundation of China (grant no. 82460461), the Medical Subject Leader of Yunnan Province (General Surgery) (grant no. D-2024029), the Yunnan Fundamental Research Project for Excellent Young Scholars (grant no. 202401AW070003), the Young and Mid-aged Academic and Technical Leader Reserve Talent Program of Yunnan Province (grant no. 202205AC160063), the Beijing Bethune Charitable Foundation (grant no. STLKY0089), the Top Talent Project of Kunming Medical University-Yang Ke to Yang Ke, and the Graduate Research Project of the Yunnan Provincial Department of Education Scientific Research Fund (grant no. 2025Y0385) to Yu-Bo Liang.

References

1 

Xiang J, Li Y, Mei S, Ou Z, Wang L, Ke Y and Li Z: Novel diagnostic and therapeutic strategies based on PANoptosis for hepatocellular carcinoma. Cancer Biol Med. 22:928–939. 2025.PubMed/NCBI

2 

Li J, Wang QB, Liang YB, Chen XM, Luo WL, Li YK, Chen X, Lu QY and Ke Y: Tumor-associated lymphatic vessel density is a reliable biomarker for prognosis of esophageal cancer after radical resection: A systemic review and meta-analysis. Front Immunol. 15:14534822024. View Article : Google Scholar : PubMed/NCBI

3 

Li J, Liang YB, Wang QB, Li YK, Chen XM, Luo WL, Lakang Y, Yang ZS, Wang Y, Li ZW and Ke Y: Tumor-associated lymphatic vessel density is a postoperative prognostic biomarker of hepatobiliary cancers: A systematic review and meta-analysis. Front Immunol. 15:15199992025. View Article : Google Scholar : PubMed/NCBI

4 

Xia F, Yi Q, Xu Z, Zhou Z, Tang H, Zhang K and Yan Y: Microbial modulation as a game changer: Boosting immunotherapy efficacy in breast cancer. Semin Cancer Biol. 117:152–167. 2025. View Article : Google Scholar : PubMed/NCBI

5 

Xu Z, Zhou H, Li T, Yi Q, Thakur A, Zhang K, Ma X, Qin JJ and Yan Y: Application of biomimetic nanovaccines in cancer immunotherapy: A useful strategy to help combat immunotherapy resistance. Drug Resist Updat. 75:1010982024. View Article : Google Scholar : PubMed/NCBI

6 

Wang R, Wang C, Lu L, Yuan F and He F: Baicalin and baicalein in modulating tumor microenvironment for cancer treatment: A comprehensive review with future perspectives. Pharmacol Res. 199:1070322024. View Article : Google Scholar

7 

Du M, Sun L, Guo J and Lv H: Macrophages and tumor-associated macrophages in the senescent microenvironment: From immunosuppressive TME to targeted tumor therapy. Pharmacol Res. 204:1071982024. View Article : Google Scholar : PubMed/NCBI

8 

Xu M, Li Y, Liu Y, Chang J, Zhou C, Weng W, Sun H, Tan C, Wang X, Wang X, et al: The development and implementation of pathological parameters and molecular testing impact prognosis of colorectal adenocarcinoma. J Natl Cancer Cent. 4:74–85. 2024.PubMed/NCBI

9 

Wang Y, Nong J, Lu B, Gao Y, Hu M, Chen C, Zhang L, Tan J, Yang X, Lin PP, et al: Disseminated tumor cells in bone marrow as predictive classifiers for small cell lung cancer patients. J Natl Cancer Cent. 4:335–345. 2024.PubMed/NCBI

10 

Li N, Song K, Chen H and Dai M: Advance and challenge of DNA methylation as cancer biomarkers for risk stratification, screening and early detection. J Natl Cancer Cent. 5:108–112. 2025.PubMed/NCBI

11 

Kim SA, Lee Y, Jung DE, Park KH, Park JY, Gang J, Jeon SB, Park EC, Kim YG, Lee B, et al: Pancreatic adenocarcinoma up-regulated factor (PAUF), a novel up-regulated secretory protein in pancreatic ductal adenocarcinoma. Cancer Sci. 100:828–836. 2009. View Article : Google Scholar : PubMed/NCBI

12 

Kanagawa M, Satoh T, Ikeda A, Nakano Y, Yagi H, Kato K, Kojima-Aikawa K and Yamaguchi Y: Crystal structures of human secretory proteins ZG16p and ZG16b reveal a Jacalin-related β-prism fold. Biochem Biophys Res Commun. 404:201–205. 2011. View Article : Google Scholar

13 

Song J, Lee J, Kim J, Jo S, Kim YJ, Baek JE, Kwon ES, Lee KP, Yang S, Kwon KS, et al: Pancreatic adenocarcinoma up-regulated factor (PAUF) enhances the accumulation and functional activity of myeloid-derived suppressor cells (MDSCs) in pancreatic cancer. Oncotarget. 7:51840–51853. 2016. View Article : Google Scholar : PubMed/NCBI

14 

Lee M, Ham H, Lee J, Lee ES, Chung CH, Kong DH, Park JR and Lee DK: TGF-β-induced PAUF plays a pivotal role in the migration and invasion of human pancreatic ductal adenocarcinoma cell line panc-1. Int J Mol Sci. 25:114202024. View Article : Google Scholar

15 

Lee Y, Kim SJ, Park HD, Park EH, Huang SM, Jeon SB, Kim JM, Lim DS and Koh SS: PAUF functions in the metastasis of human pancreatic cancer cells and upregulates CXCR4 expression. Oncogene. 29:56–67. 2010. View Article : Google Scholar

16 

Youn SE, Jiang F, Won HY, Hong DE, Kang TH, Park YY and Koh SS: PAUF induces migration of human pancreatic cancer cells exclusively via the TLR4/MyD88/NF-ĸB signaling pathway. Int J Mol Sci. 23:114142022. View Article : Google Scholar

17 

Kim JH, Na HY, Jung K, Jang D, Youn Y, Kim DH, Han HD and Hwang JH: Quantitative immunohistochemistry analysis of pancreatic adenocarcinoma upregulated factor expression in pancreatic cancer and its prognostic significance. World J Gastrointest Oncol. 17:1090552025. View Article : Google Scholar : PubMed/NCBI

18 

Choi CH, Kang TH, Song JS, Kim YS, Chung EJ, Ylaya K, Kim S, Koh SS, Chung JY, Kim JH, et al: Elevated expression of pancreatic adenocarcinoma upregulated factor (PAUF) is associated with poor prognosis and chemoresistance in epithelial ovarian cancer. Sci Rep. 8:121612018. View Article : Google Scholar : PubMed/NCBI

19 

Barderas R, Mendes M, Torres S, Bartolome RA, Lopez-Lucendo M, Villar-Vazquez R, Pelaez-Garcia A, Fuente E, Bonilla F and Casal JI: In-depth characterization of the secretome of colorectal cancer metastatic cells identifies key proteins in cell adhesion, migration, and invasion. Mol Cell Proteomics. 12:1602–1620. 2013. View Article : Google Scholar : PubMed/NCBI

20 

Kim YH, Moon JY, Kim EO, Lee SJ, Kang SH, Kim SK, Heo K, Lee Y, Kim H, Kim KT, et al: Efficient targeting and tumor retardation effect of pancreatic adenocarcinoma up-regulated factor (PAUF)-specific RNA replacement in pancreatic cancer mouse model. Cancer Lett. 344:223–231. 2014. View Article : Google Scholar

21 

Kim SJ, Chang S, Lee Y, Kim NY, Hwang Y, Min HJ, Yoo KS, Park EH, Kim S, Chung YH, et al: A PAUF-neutralizing antibody targets both carcinoma and endothelial cells to impede pancreatic tumor progression and metastasis. Biochem Biophys Res Commun. 454:144–150. 2014. View Article : Google Scholar : PubMed/NCBI

22 

Kim YH, Sung HJ, Kim S, Kim EO, Lee JW, Moon JY, Choi K, Jung JE, Lee Y, Koh SS, et al: An RNA aptamer that specifically binds pancreatic adenocarcinoma up-regulated factor inhibits migration and growth of pancreatic cancer cells. Cancer Lett. 313:76–83. 2011. View Article : Google Scholar : PubMed/NCBI

23 

King D, Tai D, Feliú J, Kim J, Mandakhalikar KD, Lim ML, Pradhan S and Park YY: First-in-human phase 1/2a study of PBP1510 (anti-PAUF mAb) in advanced/metastatic pancreatic adenocarcinoma: Safety results from early combination cohorts. J Clin Oncol. 43:e163632025. View Article : Google Scholar

24 

Feliu J, Ghanem I, King D, Park YY, Kim J, Pradhan S, Ho J, Loh WQ and Mandakhalikar KD: PBP1510, a novel monoclonal antibody targeting pancreatic adenocarcinoma upregulated factor (PAUF): Phase 1/2a monotherapy and combination with gemcitabine in patients with advanced/metastatic pancreatic cancer. J Clin Oncol. 42:e162962024. View Article : Google Scholar

25 

Mandakhalikar KD, Koh SS, Jeong SY, Moshinsky D, Feyaerts P, Karuna R, Kim J, Jaison L, Pradhan S, Kim YJ and Park J: First-in-class monoclonal antibody (mAb) PBP1510 targeting pancreatic adenocarcinoma upregulated factor (PAUF) for pancreatic cancer (PC) treatment: Preclinical perspectives. J Clin Oncol. 40:e162742022. View Article : Google Scholar

26 

Zhang H, Yngvadottir B, Andreou A, Cole Y, Woodward ER, Whitworth J and Maher ER: Clinical and genetic features of multiple primary tumours cohorts with a renal cell carcinoma: Implications for molecular genetic investigations. Int J Cancer. 157:2532–2543. 2025. View Article : Google Scholar : PubMed/NCBI

27 

Turner JA, Van Gulick RJ, Robinson WA, Mughal T, Tobin RP, MacBeth ML, Holman B, Classon A, Bagby SM, Yacob BW, et al: Expanding the landscape of oncogenic drivers and treatment options in acral and mucosal melanomas by targeted genomic profiling. Int J Cancer. 155:1792–1807. 2024. View Article : Google Scholar : PubMed/NCBI

28 

Mullins JJ, Mullins LJ, Dunbar DR, Brammar WJ, Gross KW and Morley SD: Identification of a human ortholog of the mouse Dcpp gene locus, encoding a novel member of the CSP-1/Dcpp salivary protein family. Physiol Genomics. 28:129–140. 2006. View Article : Google Scholar : PubMed/NCBI

29 

Song H, Song J, Kim YJ, Jeong HH, Min HJ and Koh SS: DCPP1 is the mouse ortholog of human PAUF that possesses functional analogy in pancreatic cancer. Biochem Biophys Res Commun. 493:1498–1503. 2017. View Article : Google Scholar : PubMed/NCBI

30 

Zhang S, He Z, Wang H and Zhai J: Signal peptides: From molecular mechanisms to applications in protein and vaccine engineering. Biomolecules. 15:8972025. View Article : Google Scholar : PubMed/NCBI

31 

Park HD, Lee Y, Oh YK, Jung JG, Park YW, Myung K, Kim KH, Koh SS and Lim DS: Pancreatic adenocarcinoma upregulated factor promotes metastasis by regulating TLR/CXCR4 activation. Oncogene. 30:201–211. 2011. View Article : Google Scholar

32 

Cheng M, Guan Y, Xin X, Yi X and Liu Y: Targeting p38 MAPK signaling pathway: Quercetin as a novel therapy for TMJ synovitis. Int J Mol Med. 57:212026.

33 

Waterhouse A, Bertoni M, Bienert S, Studer G, Tauriello G, Gumienny R, Heer FT, de Beer TAP, Rempfer C, Bordoli L, et al: SWISS-MODEL: Homology modelling of protein structures and complexes. Nucleic Acids Res. 46:W296–W303. 2018. View Article : Google Scholar : PubMed/NCBI

34 

Kumazawa-Inoue K, Mimura T, Hosokawa-Tamiya S, Nakano Y, Dohmae N, Kinoshita-Toyoda A, Toyoda H and Kojima-Aikawa K: ZG16p, an animal homolog of beta-prism fold plant lectins, interacts with heparan sulfate proteoglycans in pancreatic zymogen granules. Glycobiology. 22:258–266. 2012. View Article : Google Scholar

35 

Kleene R, Dartsch H and Kern HF: The secretory lectin ZG16p mediates sorting of enzyme proteins to the zymogen granule membrane in pancreatic acinar cells. Eur J Cell Biol. 78:79–90. 1999. View Article : Google Scholar : PubMed/NCBI

36 

Kanagawa M, Liu Y, Hanashima S, Ikeda A, Chai W, Nakano Y, Kojima-Aikawa K, Feizi T and Yamaguchi Y: Structural basis for multiple sugar recognition of Jacalin-related human ZG16p lectin. J Biol Chem. 289:16954–16965. 2014. View Article : Google Scholar : PubMed/NCBI

37 

Tateno H, Yabe R, Sato T, Shibazaki A, Shikanai T, Gonoi T, Narimatsu H and Hirabayashi J: Human ZG16p recognizes pathogenic fungi through non-self polyvalent mannose in the digestive system. Glycobiology. 22:210–220. 2012. View Article : Google Scholar

38 

Hanashima S, Gotze S, Liu Y, Ikeda A, Kojima-Aikawa K, Taniguchi N, Varon Silva D, Feizi T, Seeberger PH and Yamaguchi Y: Defining the interaction of human soluble lectin ZG16p and mycobacterial phosphatidylinositol mannosides. Chembiochem. 16:1502–1511. 2015. View Article : Google Scholar : PubMed/NCBI

39 

Mito A, Nakano Y, Saitoh T, Gouraud SSS, Yamaguchi Y, Sato T, Sasaki N and Kojima-Aikawa K: Lectin ZG16p inhibits proliferation of human colorectal cancer cells via its carbohydrate-binding sites. Glycobiology. 28:21–31. 2018. View Article : Google Scholar

40 

Mito A, Kumazawa-Inoue K and Kojima-Aikawa K: ZG16p, an animal homologue of plant beta-prism fold lectins: Purification methods of natural and recombinant ZG16p and inhibition assay of cancer cell growth using ZG16p. Methods Mol Biol. 2132:339–347. 2020. View Article : Google Scholar

41 

Thevenod F, Braun M, Roussa E and Fuller CM: Molecular characterisation of pancreatic zymogen granule ion channel and regulator proteins involved in exocytosis. J Korean Med Sci. 15(Suppl 1): S51–52. 2000. View Article : Google Scholar : PubMed/NCBI

42 

Braun M and Thevenod F: Photoaffinity labeling and purification of ZG-16p, a high-affinity dihydropyridine binding protein of rat pancreatic zymogen granule membranes that regulates a K(+)-selective conductance. Mol Pharmacol. 57:308–316. 2000. View Article : Google Scholar : PubMed/NCBI

43 

Kalus I, Hodel A, Koch A, Kleene R, Edwardson JM and Schrader M: Interaction of syncollin with GP-2, the major membrane protein of pancreatic zymogen granules, and association with lipid microdomains. Biochem J. 362:433–442. 2002. View Article : Google Scholar : PubMed/NCBI

44 

Cronshagen U, Voland P and Kern HF: cDNA cloning and characterization of a novel 16 kDa protein located in zymogen granules of rat pancreas and goblet cells of the gut. Eur J Cell Biol. 65:366–377. 1994.PubMed/NCBI

45 

Schmidt K, Schrader M, Kern HF and Kleene R: Regulated apical secretion of zymogens in rat pancreas. Involvement of the glycosylphosphatidylinositol-anchored glycoprotein GP-2, the lectin ZG16p, and cholesterol-glycosphingolipid-enriched microdomains. J Biol Chem. 276:14315–14323. 2001. View Article : Google Scholar : PubMed/NCBI

46 

Schmidt K, Dartsch H, Linder D, Kern HF and Kleene R: A submembranous matrix of proteoglycans on zymogen granule membranes is involved in granule formation in rat pancreatic acinar cells. J Cell Sci. 113(Pt 12): 2233–2242. 2000. View Article : Google Scholar : PubMed/NCBI

47 

Cho IR, Koh SS, Min HJ, Kim SJ, Lee Y, Park EH, Ratakorn S, Jhun BH, Oh S, Johnston RN and Chung YH: Pancreatic adenocarcinoma up-regulated factor (PAUF) enhances the expression of β-catenin, leading to a rapid proliferation of pancreatic cells. Exp Mol Med. 43:82–90. 2011. View Article : Google Scholar : PubMed/NCBI

48 

Lengyel M, Molnar A, Nagy T, Jdeed S, Garai I, Horvath Z and Uray IP: Zymogen granule protein 16B (ZG16B) is a druggable epigenetic target to modulate the mammary extracellular matrix. Cancer Sci. 116:81–94. 2025. View Article : Google Scholar :

49 

Kim YJ, Jiang F, Park J, Jeong HH, Baek JE, Hong SM, Jeong SY and Koh SS: PAUF as a target for treatment of high PAUF-expressing ovarian cancer. Front Pharmacol. 13:8906142022. View Article : Google Scholar : PubMed/NCBI

50 

Liu PF, Wu YY, Hu Y, Wang L, He SB, Zhu YB and Zhu XG: Silencing of pancreatic adenocarcinoma upregulated factor by RNA interference inhibits the malignant phenotypes of human colorectal cancer cells. Oncol Rep. 30:213–220. 2013. View Article : Google Scholar : PubMed/NCBI

51 

Sasahira T, Kurihara M, Nishiguchi Y, Nakashima C, Kirita T and Kuniyasu H: Pancreatic adenocarcinoma up-regulated factor has oncogenic functions in oral squamous cell carcinoma. Histopathology. 70:539–548. 2017. View Article : Google Scholar

52 

Wang P and Kong G: Comprehensive analysis of angiogenesis and ferroptosis genes for predicting the survival outcome and immunotherapy response of hepatocellular carcinoma. J Hepatocell Carcinoma. 11:1845–1859. 2024. View Article : Google Scholar : PubMed/NCBI

53 

Hong X, Hu D, Zhou WJ, Wang XD, Huang LH, Huang TA, Guan YW, Qian J and Ding WB: ALBI grade analyses of TACE combined with anti-angiogenesis therapies plus PD-1 inhibitors versus anti-angiogenesis therapies plus PD-1 inhibitors in advanced HCC. J Hepatocell Carcinoma. 11:2505–2514. 2024. View Article : Google Scholar : PubMed/NCBI

54 

Li YK, Wu S, Wu YS, Zhang WH, Wang Y, Li YH, Kang Q, Huang SQ, Zheng K, Jiang GM, et al: Portal venous and hepatic arterial coefficients predict post-hepatectomy overall and recurrence-free survival in patients with hepatocellular carcinoma: A retrospective study. J Hepatocell Carcinoma. 11:1389–1402. 2024. View Article : Google Scholar : PubMed/NCBI

55 

Wang QB, Luo WL, Li YK, Li J, Yang ZS, Zhao K, Lakang Y, Liang YB, Chen XM, Zuo JX, et al: Tumor compression of the hepatic or portal vein predicts the presence of microvascular invasion and satellite nodules in hepatocellular carcinoma: A retrospective study. J Hepatocell Carcinoma. 12:2055–2067. 2025. View Article : Google Scholar : PubMed/NCBI

56 

Zhu Y, Hu Y, Yang C, Huang S, Wen J, Huang W and Xiao S: Progress of angiogenesis signal pathway and antiangiogenic drugs in nasopharyngeal carcinoma. Curr Mol Pharmacol. 17:e187614292909332024. View Article : Google Scholar : PubMed/NCBI

57 

Liu Y, Liu Y, Sun X, Wang Y, Du C and Bai J: Morphologically transformable peptide nanocarriers coloaded with doxorubicin and curcumin inhibit the growth and metastasis of hepatocellular carcinoma. Mater Today Bio. 24:1009032023. View Article : Google Scholar : PubMed/NCBI

58 

Luo WL, Wang QB, Li YK, Liang YB, Li J, Chen XM, Lakang Y, Yang ZS, Zuo JX, Wang W, et al: Impact of middle hepatic vein resection during hemihepatectomy on surgical outcomes and long-term prognosis in hepatocellular carcinoma: A retrospective study. J Hepatocell Carcinoma. 12:2681–2692. 2025. View Article : Google Scholar : PubMed/NCBI

59 

Kim SJ, Lee Y, Kim NY, Hwang Y, Hwang B, Min JK and Koh SS: Pancreatic adenocarcinoma upregulated factor, a novel endothelial activator, promotes angiogenesis and vascular permeability. Oncogene. 32:3638–3647. 2013. View Article : Google Scholar

60 

Bao S, Darvishi M, H Amin A, Al-Haideri MT, Patra I, Kashikova K, Ahmad I, Alsaikhan F, Al-Qaim ZH, Al-Gazally ME, et al: CXC chemokine receptor 4 (CXCR4) blockade in cancer treatment. J Cancer Res Clin Oncol. 149:7945–7968. 2023. View Article : Google Scholar : PubMed/NCBI

61 

Wakasugi R, Suzuki K and Kaneko-Kawano T: Molecular mechanisms regulating vascular endothelial permeability. Int J Mol Sci. 25:64152024. View Article : Google Scholar : PubMed/NCBI

62 

Wang QB, Li J, Zhang ZJ, Li YK, Liang YB, Chen XM, Luo WL, Lakang Y, Yang ZS, Liu GY, et al: The effectiveness and safety of therapies for hepatocellular carcinoma with tumor thrombus in the hepatic vein, inferior vena cave and/or right atrium: A systematic review and single-arm meta-analysis. Expert Rev Anticancer Ther. 25:561–570. 2025. View Article : Google Scholar : PubMed/NCBI

63 

Racacho KJ, Shiau YP, Villa R, Mahri S, Tang M, Lin TY and Li Y: The tumor immune microenvironment: Implications for cancer immunotherapy, treatment strategies, and monitoring approaches. Front Immunol. 16:16218122025. View Article : Google Scholar : PubMed/NCBI

64 

Liang X, Guo J, Wang X, Luo B, Fu R, Chen H, Yang Y, Jin Z, Lin C, Zang A, et al: Overexpression of ornithine decarboxylase 1 mediates the immune-deserted microenvironment and poor prognosis in diffuse large B-cell lymphoma. J Natl Cancer Cent. 5:57–74. 2024.

65 

Xu W, Lu J, Zhang H and Ye D: Decoding the tumor microenvironment: Insights into immunotherapy and beyond. J Natl Cancer Cent. 5:426–428. 2025.PubMed/NCBI

66 

Wang X, Wang C, Liu W, Thakur A, Zhang K, Xu Z and Li J: The roles of ultrasound-responsive nanomaterials in enhancing cancer immunotherapy. Pharmacol Res. 221:1079752025. View Article : Google Scholar : PubMed/NCBI

67 

Jiang Q, He J, Zhang H, Chi H, Shi Y and Xu X: Recent advances in the development of tumor microenvironment-activatable nanomotors for deep tumor penetration. Mater Today Bio. 27:1011192024. View Article : Google Scholar : PubMed/NCBI

68 

Bao X, Sun M, Meng L, Zhang H, Yi X and Zhang P: Applications of pyroptosis activators in tumor immunotherapy. Mater Today Bio. 28:1011912024. View Article : Google Scholar : PubMed/NCBI

69 

Wang J, Chen Z, Liu W, Xu Z, Liu H, Li Y and Yan Y: Harnessing plant-derived natural compounds to target ferroptosis, pyroptosis, immune modulation and renin-angiotensin system in renal cell carcinoma. J Renin Angiotensin Aldosterone Syst. 26:147032032513863092025. View Article : Google Scholar

70 

Wang D, Zhang Z, Yang L, Zhao L, Liu Z and Lou C: PD-1 inhibitors combined with tyrosine kinase inhibitors with or without hepatic artery infusion chemotherapy for the first-line treatment of HBV-related advanced hepatocellular carcinoma: A retrospective study. J Hepatocell Carcinoma. 11:1157–1170. 2024. View Article : Google Scholar : PubMed/NCBI

71 

Chen H, Li J, Cao D and Tang H: Construction of a prognostic model for hepatocellular carcinoma based on macrophage polarization-related genes. J Hepatocell Carcinoma. 11:857–878. 2024. View Article : Google Scholar : PubMed/NCBI

72 

Wang Z, Ge Q, Mo R, Lu J, Tian X, Anwaier A, Ye S, Zhou S, Guo W, Cai C, et al: Spatial and maturity heterogeneity of tertiary lymphoid structures shapes immune microenvironment and progression in prostate cancer. J Natl Cancer Cent. 5:501–514. 2025.PubMed/NCBI

73 

Li J, Liang YB, Wang QB, Luo WL, Chen XM, Lakang Y, Yang ZS, Zuo JX, Li YK, Li ZW, et al: Rechallenge with immune checkpoint inhibitors in patients with hepatocellular carcinoma: A narrative review. Liver Cancer. Oct 31–2025.Epub ahead of print. View Article : Google Scholar

74 

Kudo M: Fluorine-18 fluorodeoxyglucose positron emission tomography: A potential imaging biomarker for predicting response to combination immunotherapy in hepatocellular carcinoma. Liver Cancer. 14:511–517. 2025. View Article : Google Scholar : PubMed/NCBI

75 

Zhao Q, Wei T, Ma R, Fu Y, Yang R, Su Y, Yu Y, Li B and Li Y: Progress on immuno-microenvironment and immune-related therapies in patients with pseudomyxoma peritonei. Cancer Biol Med. 21:586–605. 2024.PubMed/NCBI

76 

Zhang W, Wang S, Zhang H, Meng Y, Jiao S, An L and Zhou Z: Modeling human gastric cancers in immunocompetent mice. Cancer Biol Med. 21:553–570. 2024.PubMed/NCBI

77 

Aoki T, Kudo M, Ueshima K, Morita M, Chishina H, Takita M, Hagiwara S, Ida H, Minami Y, Tsurusaki M and Nishida N: Incidence of hyper progressive disease in combination immunotherapy and anti-programmed cell death protein 1/programmed death-ligand 1 monotherapy for unresectable hepatocellular carcinoma. Liver Cancer. 13:56–69. 2024. View Article : Google Scholar : PubMed/NCBI

78 

Aoki T, Nishida N, Kurebayashi Y, Sakai K, Morita M, Chishina H, Takita M, Hagiwara S, Ida H, Ueshima K, et al: Two distinct characteristics of immune microenvironment in human hepatocellular carcinoma with Wnt/β-catenin mutations. Liver Cancer. 13:285–305. 2023. View Article : Google Scholar

79 

Lin Y, Ruze R, Zhang R, Tuergan T, Wang M, Tulahong A, Zhu D, Yuan Z, Jiang T, Aji T and Shao Y: Immunometabolic targets in CD8(+) T cells within the tumor microenvironment of hepatocellular carcinoma. Liver Cancer. 14:474–496. 2024. View Article : Google Scholar

80 

Luo M, Zhao F, Cheng H, Su M and Wang Y: Macrophage polarization: An important role in inflammatory diseases. Front Immunol. 15:13529462024. View Article : Google Scholar : PubMed/NCBI

81 

Yang L, Zhang Y and Yang L: Adenosine signaling in tumor-associated macrophages and targeting adenosine signaling for cancer therapy. Cancer Biol Med. 21:995–1011. 2024.PubMed/NCBI

82 

Nishida N and Kudo M: Genetic/Epigenetic alteration and tumor immune microenvironment in intrahepatic cholangiocarcinoma: Transforming the immune microenvironment with molecular-targeted agents. Liver Cancer. 13:136–149. 2023. View Article : Google Scholar

83 

Xu Z, Zhou Z, Yang X, Thakur A, Han N, Li HT, Li LG, Hu J, Li TF and Yan Y: Determining M2 macrophages content for the anti-tumor effects of metal-organic framework-encapsulated pazopanib nanoparticles in breast cancer. J Nanobiotechnology. 22:4292024. View Article : Google Scholar : PubMed/NCBI

84 

Zhang J, Wang L, Guo H, Kong S, Li W, He Q, Ding L and Yang B: The role of Tim-3 blockade in the tumor immune microenvironment beyond T cells. Pharmacol Res. 209:1074582024. View Article : Google Scholar : PubMed/NCBI

85 

Lv Q, Zhang Y, Gao W, Wang J, Hu Y, Yang H, Xie Y, Lv Y, Zhang H, Wu D, et al: CSF1R inhibition reprograms tumor-associated macrophages to potentiate anti-PD-1 therapy efficacy against colorectal cancer. Pharmacol Res. 202:1071262024. View Article : Google Scholar : PubMed/NCBI

86 

Wang X, Chen J and Jia G: From dichotomy to diversity: Deciphering the multifaceted roles of tumor-associated macrophages in cancer progression and therapy. Cancer Biol Med. 21:132–138. 2023.PubMed/NCBI

87 

Chen J, Li H, Zhuo J, Lin Z, Hu Z, He C, Wu X, Jin Y, Lin Z, Su R, et al: Impact of immunosuppressants on tumor pulmonary metastasis: New insight into transplantation for hepatocellular carcinoma. Cancer Biol Med. 21:1033–1049. 2024. View Article : Google Scholar : PubMed/NCBI

88 

Dong Y, Khan L and Yao Y: Immunological features of EGFR-mutant non-small cell lung cancer and clinical practice: A narrative review. J Natl Cancer Cent. 4:289–298. 2024.PubMed/NCBI

89 

Kim YJ, Nanda SS, Jiang F, Pyo SY, Han JY, Koh SS and Kang TH: Pancreatic adenocarcinoma up-regulated factor (PAUF) transforms human monocytes into alternative M2 macrophages with immunosuppressive action. Int J Mol Sci. 25:115452024. View Article : Google Scholar : PubMed/NCBI

90 

Zhang Y, Ji S, Miao G, Du S, Wang H, Yang X, Li A, Lu Y, Wang X and Zhao X: The current role of dendritic cells in the progression and treatment of colorectal cancer. Cancer Biol Med. 21:769–783. 2024.PubMed/NCBI

91 

Kudo M: Challenges in adjuvant immunotherapy after resection or ablation for hepatocellular carcinoma at high-risk of recurrence. Liver Cancer. 13:573–578. 2024. View Article : Google Scholar : PubMed/NCBI

92 

Tang X, Zhang J, Sui D, Xu Z, Yang Q, Wang T, Li X, Liu X, Deng Y and Song Y: Durable protective efficiency provide by mRNA vaccines require robust immune memory to antigens and weak immune memory to lipid nanoparticles. Mater Today Bio. 25:1009882024. View Article : Google Scholar : PubMed/NCBI

93 

Chen X, Yang Z and Li M: Molecular regulatory mechanisms and diagnostic potential of dendritic cell-derived exosomes in liver transplantation: From immune tolerance induction to translational challenges. Front Immunol. 16:16579562025. View Article : Google Scholar : PubMed/NCBI

94 

Kang TH, Kim YS, Kim S, Yang B, Lee JJ, Lee HJ, Lee J, Jung ID, Han HD, Lee SH, et al: Pancreatic adenocarcinoma upregulated factor serves as adjuvant by activating dendritic cells through stimulation of TLR4. Oncotarget. 6:27751–27762. 2015. View Article : Google Scholar : PubMed/NCBI

95 

Liao Y and Yang H: Metabolic regulation of innate immunity in cancer immunotherapy. Cancer Biol Med. 20:898–902. 2024. View Article : Google Scholar : PubMed/NCBI

96 

Xi W, Wu W, Zhou L, Zhang Q, Yang S, Huang L, Lu Y, Wang J, Chi X and Kang Y: Multifunctional nanoparticles confers both multiple inflammatory mediators scavenging and macrophage polarization for sepsis therapy. Mater Today Bio. 30:1014212025. View Article : Google Scholar : PubMed/NCBI

97 

Qu Z, Guo Z, Yang C, Guan X, Cheng M, Wang P and Xu D: Role of toll-like receptors in pulmonary immunity: Mechanisms and therapeutic implications. Front Immunol. 16:16826492025. View Article : Google Scholar : PubMed/NCBI

98 

Yang Y, Cui H, Li D, Chen L, Liu Y, Zhou C, Li L, Feng M, Chen X, Cao Y and Gao Y: S100A8 promotes tumor progression by inducing phenotypic polarization of microglia through the TLR4/IL-10 signaling pathway in glioma. J Natl Cancer Cent. 4:369–381. 2024.PubMed/NCBI

99 

Wang T, Liu C, Hu X, Yang N and Qiu C: Senescent macrophages in cancer: Roles in tumor progression and treatment opportunities. Cancer Biol Med. 22:439–459. 2025.PubMed/NCBI

100 

Fang Z, Ding X, Huang H, Jiang H, Jiang J and Zheng X: Revolutionizing tumor immunotherapy: Unleashing the power of progenitor exhausted T cells. Cancer Biol Med. 21:499–512. 2024.PubMed/NCBI

101 

Bauckneht M and Filippi L: Pentixather: Paving the way for radioligand therapy in oncohematology. Expert Rev Anticancer Ther. 24:205–209. 2024. View Article : Google Scholar : PubMed/NCBI

102 

Gupta T, Mani S, Chatterjee A, Dasgupta A, Epari S and Chinnaswamy G: Risk-stratification for treatment de-intensification in WNT-pathway medulloblastoma: Finding the optimal balance between survival and quality of survivorship. Expert Rev Anticancer Ther. 24:589–598. 2024. View Article : Google Scholar : PubMed/NCBI

103 

Li SR, Wu ZZ, Yu HJ and Sun ZJ: Targeting erythroid progenitor cells for cancer immunotherapy. Int J Cancer. 155:1928–1938. 2024. View Article : Google Scholar : PubMed/NCBI

104 

Liang Y, Xie Y, Dang Z, Li M, Yu L, Wang X, Wang P and Yang Z: Yiqi Liangxue Jiedu prescription inhibited the canonical wnt pathway to prevent hepatocellular precancerous lesions. J Hepatocell Carcinoma. 11:2293–2308. 2024. View Article : Google Scholar : PubMed/NCBI

105 

Wang J, Gao W, Yu H, Xu Y, Bai C, Cong Q and Zhu Y: Research progress on the role of epigenetic methylation modification in hepatocellular carcinoma. J Hepatocell Carcinoma. 11:1143–1156. 2024. View Article : Google Scholar : PubMed/NCBI

106 

Tyraskis A, Zen Y, Strautnieks S, Cook R, Foskett P, De Vito C, Deheragoda M, Quaglia A, Heaton N, Davenport M and Thompson RJ: High frequency of CTNNB1 variants associated with benign and malignant liver tumors in patients with congenital porto-systemic shunts. Liver Cancer. 14:408–419. 2024. View Article : Google Scholar

107 

Higuchi Y, Nguyen C, Chimge NO, Ouyang C, Teo JL and Kahn M: E7386 is not a specific CBP/β-catenin antagonist. Curr Mol Pharmacol. 17:e2905232174092024.

108 

Xue W, Zhu B, Zhao K, Huang Q, Luo H, Shou Y, Huang Z and Guo H: Targeting LRP6: A new strategy for cancer therapy. Pharmacol Res. 204:1072002024. View Article : Google Scholar : PubMed/NCBI

109 

Zhu X, Trehan R and Xie C: Primary liver cancer organoids and their application to research and therapy. J Natl Cancer Cent. 4:195–202. 2024.PubMed/NCBI

110 

Cho IR, Koh SS, Malilas W, Srisuttee R, Moon J, Choi YW, Horio Y, Oh S and Chung YH: SIRT1 inhibits proliferation of pancreatic cancer cells expressing pancreatic adenocarcinoma up-regulated factor (PAUF), a novel oncogene, by suppression of β-catenin. Biochem Biophys Res Commun. 423:270–275. 2012. View Article : Google Scholar : PubMed/NCBI

111 

Du F, Xie Y, Wu S, Ji M, Dong B and Zhu C: Expression and targeted application of claudins family in hepatobiliary and pancreatic diseases. J Hepatocell Carcinoma. 11:1801–1821. 2024. View Article : Google Scholar : PubMed/NCBI

112 

Xia J, Zhang Z, Huang Y, Wang Y and Liu G: Regulation of neutrophil extracellular traps in cancer. Int J Cancer. 154:773–785. 2024. View Article : Google Scholar

113 

Toshida K, Itoh S, Iseda N, Tanaka S, Nakazono K, Tomiyama T, Yoshiya S, Toshima T, Harada N, Kohashi K, et al: The impact of TP53-induced glycolysis and apoptosis regulator on prognosis in hepatocellular carcinoma: Association with tumor microenvironment and ferroptosis. Liver Cancer. 14:36–57. 2024.

114 

Lee YS, Kim SJ, Min HJ, Jo JY, Park EH and Koh SS: PAUF promotes adhesiveness of pancreatic cancer cells by modulating focal adhesion kinase. Exp Mol Med. 43:291–297. 2011. View Article : Google Scholar : PubMed/NCBI

115 

Wang Y, Cheng S, Fleishman JS, Chen J, Tang H, Chen ZS, Chen W and Ding M: Targeting anoikis resistance as a strategy for cancer therapy. Drug Resist Updat. 75:1010992024. View Article : Google Scholar : PubMed/NCBI

116 

Deng Z, Fan T, Xiao C, Tian H, Zheng Y, Li C and He J: TGF-β signaling in health, disease, and therapeutics. Signal Transduct Target Ther. 9:612024. View Article : Google Scholar

117 

Elkoshi Z: TGF-β, IL-1β, IL-6 levels and TGF-β/Smad pathway reactivity regulate the link between allergic diseases, cancer risk, and metabolic dysregulations. Front Immunol. 15:13717532024. View Article : Google Scholar

118 

Du YQ, Yuan B, Ye YX, Zhou FL, Liu H, Huang JJ and Wei YF: Plumbagin regulates snail to inhibit hepatocellular carcinoma epithelial-mesenchymal transition in vivo and in vitro. J Hepatocell Carcinoma. 11:565–580. 2024. View Article : Google Scholar : PubMed/NCBI

119 

Qiu X, Dong L, Wang K, Zhong X, Xu H, Xu S, Guo H, Wei X, Chen W and Xu X: Development and validation of a novel nomogram integrated with hypoxic and lactate metabolic characteristics for prognosis prediction in hepatocellular carcinoma. J Hepatocell Carcinoma. 11:241–255. 2024. View Article : Google Scholar : PubMed/NCBI

120 

Wang P, Ke B and Ma G: Drug-tolerant persister cancer cells. J Natl Cancer Cent. 4:1–5. 2023.

121 

Yoo W, Choi H, Son YH, Lee J, Jo S, Jung D, Kim YJ, Koh SS, Yang YR, Kwon ES, et al: Pancreatic cancer induces muscle wasting by promoting the release of pancreatic adenocarcinoma upregulated factor. Exp Mol Med. 53:432–445. 2021. View Article : Google Scholar : PubMed/NCBI

122 

Kim SK, Song SY, Kim S, Cho NH, Yim GW, Kim SW, Kim YT and Nam EJ: Association of pancreatic adenocarcinoma up-regulated factor expression in ovarian mucinous adenocarcinoma with poor prognosis. Int J Clin Exp Pathol. 7:5103–5110. 2014.PubMed/NCBI

123 

Kim J, Chung JY, Kim TJ, Lee JW, Kim BG, Bae DS, Choi CH and Hewitt SM: Genomic network-based analysis reveals pancreatic adenocarcinoma up-regulating factor-related prognostic markers in cervical carcinoma. Front Oncol. 8:4652018. View Article : Google Scholar : PubMed/NCBI

124 

Choi CH, Chung JY, Park HS, Jun M, Lee YY, Kim BG and Hewitt SM: Pancreatic adenocarcinoma up-regulated factor expression is associated with disease-specific survival in cervical cancer patients. Hum Pathol. 46:884–893. 2015. View Article : Google Scholar : PubMed/NCBI

125 

Zhang T, Wang Y, Dong Y, Liu L, Han Y, Wang H, Wei Q, Xia P, Ma W and Li L: Identification of novel diagnostic biomarkers in prostate adenocarcinoma based on the stromal-immune score and analysis of the WGCNA and ceRNA network. Dis Markers. 2022:19091962022.PubMed/NCBI

126 

Jin HJ, Jung S, DebRoy AR and Davuluri RV: Identification and validation of regulatory SNPs that modulate transcription factor chromatin binding and gene expression in prostate cancer. Oncotarget. 7:54616–54626. 2016. View Article : Google Scholar : PubMed/NCBI

127 

Chen W, Liao L, Lai H, Yi X and Wang D: Identification of core biomarkers associated with pathogenesis and prognostic outcomes of laryngeal squamous-cell cancer using bioinformatics analysis. Eur Arch Otorhinolaryngol. 277:1397–1408. 2020. View Article : Google Scholar : PubMed/NCBI

128 

Lu H, Shi C, Liu X, Liang C, Yang C, Wan X, Li L and Liu Y: Identification of ZG16B as a prognostic biomarker in breast cancer. Open Med (Wars). 16:1–13. 2020. View Article : Google Scholar : PubMed/NCBI

129 

Perumal N, Funke S, Wolters D, Pfeiffer N and Grus FH: Characterization of human reflex tear proteome reveals high expression of lacrimal proline-rich protein 4 (PRR4). Proteomics. 15:3370–3381. 2015. View Article : Google Scholar : PubMed/NCBI

130 

Wang Y, Luo H, Che G, Li Y, Gao J, Yang Q, Zhou B, Gao L, Wang T, Liang Y, et al: Placental protein 14 as a potential biomarker for diagnosis of preterm premature rupture of membranes. Mol Med Rep. 18:113–122. 2018.PubMed/NCBI

131 

Ghosh S, Ahearn CP, Isabella CR, Marando VM, Dodge GJ, Bartlett H, McPherson RL, Dugan AE, Jain S, Neznanova L, et al: Human oral lectin ZG16B acts as a cell wall polysaccharide probe to decode host-microbe interactions with oral commensals. Proc Natl Acad Sci USA. 120:e22163041202023. View Article : Google Scholar : PubMed/NCBI

132 

Baik JE, Choe HI, Hong SW, Kang SS, Ahn KB, Cho K, Yun CH and Han SH: Human salivary proteins with affinity to lipoteichoic acid of Enterococcus faecalis. Mol Immunol. 77:52–59. 2016. View Article : Google Scholar : PubMed/NCBI

133 

Costa-da-Silva AC, Aure MH, Dodge J, Martin D, Dhamala S, Cho M, Rose JJ, Bassim CW, Ambatipudi K, Hakim FT, et al: Salivary ZG16B expression loss follows exocrine gland dysfunction related to oral chronic graft-versus-host disease. iScience. 25:1035922021. View Article : Google Scholar

134 

Martin-Lorenzo M, Zubiri I, Maroto AS, Gonzalez-Calero L, Posada-Ayala M, de la Cuesta F, Mourino-Alvarez L, Lopez-Almodovar LF, Calvo-Bonacho E, Ruilope LM, et al: KLK1 and ZG16B proteins and arginine-proline metabolism identified as novel targets to monitor atherosclerosis, acute coronary syndrome and recovery. Metabolomics. 11:1056–1067. 2015. View Article : Google Scholar : PubMed/NCBI

135 

Perumal N, Funke S, Pfeiffer N and Grus FH: Proteomics analysis of human tears from aqueous-deficient and evaporative dry eye patients. Sci Rep. 6:296292016. View Article : Google Scholar : PubMed/NCBI

136 

Salvisberg C, Tajouri N, Hainard A, Burkhard PR, Lalive PH and Turck N: Exploring the human tear fluid: Discovery of new biomarkers in multiple sclerosis. Proteomics Clin Appl. 8:185–194. 2014. View Article : Google Scholar : PubMed/NCBI

137 

Sun Y, Ye L, Zheng Y and Yang Z: Identification of crucial genes associated with Parkinson's disease using microarray data. Mol Med Rep. 17:3775–3782. 2018.

138 

Hu W and Xu Y: Transcriptomics in idiopathic pulmonary fibrosis unveiled: A new perspective from differentially expressed genes to therapeutic targets. Front Immunol. 15:13751712024. View Article : Google Scholar : PubMed/NCBI

139 

Kunimune Y, Suehiro Y, Saeki I, Yamauchi Y, Tanabe N, Matsumoto T, Higaki S, Fujii I, Suzuki C, Okayama N, et al: Combination assay of methylated HOXA1 with tumor markers shows high sensitivity for detection of early-stage hepatocellular carcinoma. Liver Cancer. 13:487–497. 2024. View Article : Google Scholar : PubMed/NCBI

140 

Zezulinski D, Hoteit MA, Kaplan DE, Simeone A, Zhan T, Doria C, Ahmed FY, Roberts LR, Block TM and Sayeed A: Detection of circulating mRNA variants in hepatocellular carcinoma patients using targeted RNAseq. Liver Cancer. 14:555–586. 2025. View Article : Google Scholar : PubMed/NCBI

141 

Garcia-Silva S, Marchetti D and Gallardo M: Editorial: Liquid biopsies in hematological malignancies. Front Immunol. 15:14403942024. View Article : Google Scholar : PubMed/NCBI

142 

Li M, Zhang Y, Yu D, Yu Y and Ma W: Immunotherapy biomarkers in brain metastases: Insights into tumor microenvironment dynamics. Front Immunol. 16:16002612025. View Article : Google Scholar : PubMed/NCBI

143 

Zhang S, Zhao H, Wang K, Li L, Pan Q, Lu M and Zhang X: Tracing the history of clinical practice of liquid biopsy: A bibliometric analysis. Front Immunol. 16:15747362025. View Article : Google Scholar : PubMed/NCBI

144 

Sato K, Toh S, Murakami T, Nakano T, Hongo T, Matsuo M, Hashimoto K, Sugasawa M, Yamazaki K, Ueki Y, et al: Nationwide multi-centric prospective study for the identification of biomarkers to predict the treatment responses of nivolumab through comprehensive analyses of pretreatment plasma exosome mRNAs from head and neck cancer patients (BIONEXT study). Front Immunol. 15:14644192025. View Article : Google Scholar : PubMed/NCBI

145 

Cho JH, Kim SA, Park SB, Kim HM and Song SY: Suppression of pancreatic adenocarcinoma upregulated factor (PAUF) increases the sensitivity of pancreatic cancer to gemcitabine and 5FU, and inhibits the formation of pancreatic cancer stem like cells. Oncotarget. 8:76398–76407. 2017. View Article : Google Scholar : PubMed/NCBI

146 

Gao CC, Xu XL, Li F, Gong BG, Liu S, Cui YQ, Sun HC, Xu PY, Zheng YM and Jiang H: Silencing pancreatic adenocarcinoma upregulated factor (PAUF) increases the sensitivity of pancreatic cancer cells to gemcitabine. Tumour Biol. 37:7555–7564. 2016. View Article : Google Scholar

147 

Kaowinn S, Cho IR, Moon J, Jun SW, Kim CS, Kang HY, Kim M, Koh SS and Chung YH: Pancreatic adenocarcinoma upregulated factor (PAUF) confers resistance to pancreatic cancer cells against oncolytic parvovirus H-1 infection through IFNA receptor-mediated signaling. Biochem Biophys Res Commun. 459:313–318. 2015. View Article : Google Scholar : PubMed/NCBI

Related Articles

  • Abstract
  • View
  • Download
  • Twitter
Copy and paste a formatted citation
Spandidos Publications style
Chen X, Liang Y, Zuo J, Yang Z, Zhang L, Zhang X, Wan P and Ke Y: ZG16B: A key regulator of tumor progression and immune microenvironment modulation in cancer (Review). Int J Mol Med 57: 58, 2026.
APA
Chen, X., Liang, Y., Zuo, J., Yang, Z., Zhang, L., Zhang, X. ... Ke, Y. (2026). ZG16B: A key regulator of tumor progression and immune microenvironment modulation in cancer (Review). International Journal of Molecular Medicine, 57, 58. https://doi.org/10.3892/ijmm.2026.5729
MLA
Chen, X., Liang, Y., Zuo, J., Yang, Z., Zhang, L., Zhang, X., Wan, P., Ke, Y."ZG16B: A key regulator of tumor progression and immune microenvironment modulation in cancer (Review)". International Journal of Molecular Medicine 57.3 (2026): 58.
Chicago
Chen, X., Liang, Y., Zuo, J., Yang, Z., Zhang, L., Zhang, X., Wan, P., Ke, Y."ZG16B: A key regulator of tumor progression and immune microenvironment modulation in cancer (Review)". International Journal of Molecular Medicine 57, no. 3 (2026): 58. https://doi.org/10.3892/ijmm.2026.5729
Copy and paste a formatted citation
x
Spandidos Publications style
Chen X, Liang Y, Zuo J, Yang Z, Zhang L, Zhang X, Wan P and Ke Y: ZG16B: A key regulator of tumor progression and immune microenvironment modulation in cancer (Review). Int J Mol Med 57: 58, 2026.
APA
Chen, X., Liang, Y., Zuo, J., Yang, Z., Zhang, L., Zhang, X. ... Ke, Y. (2026). ZG16B: A key regulator of tumor progression and immune microenvironment modulation in cancer (Review). International Journal of Molecular Medicine, 57, 58. https://doi.org/10.3892/ijmm.2026.5729
MLA
Chen, X., Liang, Y., Zuo, J., Yang, Z., Zhang, L., Zhang, X., Wan, P., Ke, Y."ZG16B: A key regulator of tumor progression and immune microenvironment modulation in cancer (Review)". International Journal of Molecular Medicine 57.3 (2026): 58.
Chicago
Chen, X., Liang, Y., Zuo, J., Yang, Z., Zhang, L., Zhang, X., Wan, P., Ke, Y."ZG16B: A key regulator of tumor progression and immune microenvironment modulation in cancer (Review)". International Journal of Molecular Medicine 57, no. 3 (2026): 58. https://doi.org/10.3892/ijmm.2026.5729
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