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
December-2025 Volume 56 Issue 6

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
December-2025 Volume 56 Issue 6

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

Periostin: A matricellular protein with a multifaceted role in tumorigenesis (Review)

  • Authors:
    • Yunjia Xian
    • Lufang Wang
  • View Affiliations / Copyright

    Affiliations: Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
    Copyright: © Xian et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
  • Article Number: 229
    |
    Published online on: October 21, 2025
       https://doi.org/10.3892/ijmm.2025.5670
  • 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

Extracellular matrix (ECM) proteins are key non‑cellular components of the tumor microenvironment and include structural and non‑structural proteins. POSTN belongs to a family of non‑structural matrix cell proteins first identified in bone and periodontal ligaments. POSTN exerts its multiple effects mainly through specific binding to other ECM proteins, growth factors and cytokines. Physiologically, POSTN is rarely expressed in adult tissues, except in the periosteum and some collagen‑rich connective tissues, such as periodontal ligaments and heart valves, but is highly expressed during inflammation, tissue repair, wound healing and malignant transformation. POSTN carries out an important role in tumor development. The present review summarizes the multiple roles of POSTN in tumorigenesis and development. First, the molecular structure of POSTN and its physiological and pathological expression, especially in known tumors is reviewed. Second, the manner in which POSTN affects tumor cells through relevant cancer hallmarks is discussed. Finally, the molecular mechanisms underlying the promotion of cancer by POSTN from the perspective of the TME is summarized.

Background

Cancer presents a considerable challenge in medical research due to its diversity and breadth. The proposal of cancer hallmarks attempts to convert the complexity of cancer into a set of provisional basic principles, which can help researchers understand the mechanisms underlying cancer development more comprehensively and logically. Cancer hallmarks are a set of capabilities acquired by human cells as they progress from a normal to a proliferative state, and are key to the malignant development of tumors (1). The tumor microenvironment (TME) is considered to carry out an important role in tumorigenesis and malignant progression. The TME is a dynamic environment that includes all non-cancerous and non-cellular components of a tumor, such as fibroblasts, immune cells, the extracellular matrix (ECM) and various secreted factors such as IL-6 and IFN-γ (2). Among these components (POSTN), a matricellular protein initially identified in bone and periodontal ligaments, has emerged as a multifunctional regulator in both physiological and pathological contexts (3). POSTN was initially discovered in 1993 (4), and numerous associated studies have since been conducted (5-11). Initially, POSTN was reported to be expressed minimally or not at all in the vast majority of mature tissues but, highly expressed in the context of disease states such as cancer, inflammation and fibrosis (12,13). Additionally, POSTN regulates the development and maturation of fibrous-rich embryonic tissues, such as heart valves and other periodontal ligaments (14,15).

In tumors, POSTN is a key player in tumor hallmarks such as proliferation, metastasis and immune evasion (12). POSTN achieves this through interactions with integrins and the activation of underlying downstream signaling pathways (1). Despite growing evidence associating POSTN with diverse cancer hallmarks, its isoform-specific functions, regulatory mechanisms within heterogeneous TME components and therapeutic potential remain incompletely characterized (16-22). An increasing number of preclinical and clinical studies have demonstrated the potential of POSTN in oncogenic mechanisms and as a biomarker (12,23-28). The present review primarily focuses on the latest research on the functional diversity of POSTN and its role in the tumor niche, which provides insights into therapeutic strategies.

Molecular structure of POSTN

Gene organization

Early studies of POSTN date back to 1993, when a gene was identified that encodes the 811 amino acid residue osteoblast-specific factor 2, later renamed as POSTN (4,29-31). The human POSTN gene, located on chromosome 13, spans 36,262 bp, contains 24 exons and encodes a protein of 836 amino acids (https://www.ncbi.nlm.nih.gov/gene/10631).

Alternative splicing

Selective splicing is a tightly regulated process that contributes to the high complexity of the multicellular eukaryotic transcriptome at the protein level. Various studies have revealed that in several species, there is alternative splicing of the POSTN gene in either insertion or deletion patterns (4,32-34), In humans, exons 17, 18, 19 and 21 of the POSTN gene are selectively spliced, resulting in a variety of isoforms. Moreover, other isoforms of the POSTN gene, including 10 variants of unknown function, have also been discovered in various types of cancer such as renal cell carcinoma and non-small cell lung cancer (32,35). Among the physiologically expressed POSTN isoforms, PN4 (POSTN lacking exons 17 and 21) is ubiquitously expressed and predominant (34). Several studies have discovered new isoforms and explored their possible roles (18,36-41). Additionally, POSTN3 containing exon 17 and 21 (but not containing exon 12) has been shown to promote the growth and metastasis of breast cancer tumors through alternative splicing (34). Similarly, although POSTN is a suppressor in bladder cancer, POSTN variant I, which is highly expressed in bladder cancer, does not exhibit a suppressive effect (42).

Structural domains of POSTN

POSTN is a disulfide-linked 90 kDa protein secreted by osteoblasts and osteoclast-like cell lines (8). The protein consists of six structural domains, including a cysteine-rich Egf-like module containing Mucin-like hormone receptor-like structural domain, four tandemly repeated FAS-1 structural domains and a hydrophilic C-terminal structural domain (4) (Fig. 1). The four-repeat fasciclin structural domain has homology to insect proteins, based on which the Fas1 structural domain characterizes it as a member of the fasciclin 1 family. The FAS-1 structural domain has been shown to interact with fibronectin (43), tenascin-C (44), bone morphogenetic protein-1 (45) and Cellular Communication Network factor 3 (46). As an ECM protein, POSTN relies largely on the FAS-1 structural domain to bind to integrins and thus mediate various signaling pathways (47). The C-terminal structural domain, conversely, lacks known functional domains and is intrinsically disordered (48).

Structure of POSTN. POSTN consists of
six structural domains, including EMI domain, four tandemly
repeated FAS-1 structural domains and a hydrophilic C-terminal
structural domain. N, N-terminal; CTD, C-terminal domain; POSTN,
periostin; EMI, Egf-like module containing Mucin-like hormone
receptor-like; FAS, Fasciclin.

Figure 1

Structure of POSTN. POSTN consists of six structural domains, including EMI domain, four tandemly repeated FAS-1 structural domains and a hydrophilic C-terminal structural domain. N, N-terminal; CTD, C-terminal domain; POSTN, periostin; EMI, Egf-like module containing Mucin-like hormone receptor-like; FAS, Fasciclin.

Expression of POSTN

Physiological expression of POSTN

POSTN is a matrix protein that is named after its preferential expression in mouse periosteum and periodontal ligaments. The expression of POSTN in these tissues is regulated by mechanical stresses (49). Physiologically expressed in collagen-rich connective tissues (such as, cardiac valves, tendons and lungs), POSTN becomes active during fetal development but is predominantly absent or minimally expressed in the majority of adult tissues. It regulates tissue maturation in embryonic structures such as the periosteum, periodontal ligaments, liver, kidneys and ureters (14,50). Notably, POSTN drives fetal cardiomyocyte lineage differentiation and heart valve maturation (15), positioning it as a potential biomarker for prenatal non-invasive screening of congenital heart defects. Additionally, elevated POSTN expression occurs in stem cell niches of adult organs, including the mammary gland, intestine, spleen, bone and skin, suggesting context-dependent roles in tissue homeostasis.

Pathological expression of POSTN

POSTN and its variants are differentially expressed and are upregulated in inflammation, sites of injury and tumors (18,51,52). POSTN contributes to pathological processes such as allergy, inflammation, tissue remodeling, regeneration, fibrosis and tumor progression. For example, POSTN is a valuable biomarker of type 2 inflammation (a specific type of immune response that is primarily mediated by type 2 helper T cells)as a downstream molecule of IL-4 and IL-13, which has been closely associated with the pathogenesis of asthma (53), chronic rhinosinusitis (54), allergic dermatitis (55) and pulmonary fibrosis (56). In addition, POSTN is also involved in the development of myocardial infarction through its response to TGF-β in inflammation and fibrosis in chronic liver and kidney disease (57), as well as through its interaction with other ECM proteins (for example, fibronectin, heparin and Tenascin-C) (58).

POSTN is expressed in several primary types of cancer, including breast, bladder, colon, pancreatic and ovarian cancer, as well as non-small cell lung and oral cancer (13,59) (Table I). For example, POSTN is expressed at low levels in normal breast tissue, but high expression of POSTN is often detected in breast cancer, with the average expression level of the periosteal protein being 20-fold higher compared with the baseline expression level defined by gene array data for normal breast tissue (60). POSTN is also abnormally elevated in metastatic cancer types, most notably liver metastases from colorectal cancer, which has been associated with a supportive fibrotic microenvironment promoted by both POSTN and TGF-β1 (61). However, the upregulation of POSTN expression in tumors is not universal. For example, POSTN is not expressed in hematologic malignancies such as leukemia and myeloma (62).

Table I

POSTN-related signaling pathways in malignant tumor progression.

Table I

POSTN-related signaling pathways in malignant tumor progression.

Authors, yearPathwayCancer typePOSTN functionAssociated cells(Refs.)
Ribatti et al, 2007 POSTN/α6β4Pancreatic cancerPromotes invasiveness and survivalCAFs(87)
Parker et al, 2020; Erkan et al, 2009; Halperin et al, 2022; Mouw et al, 2014; Boulter et al, 2021 POSTN/αvβ3, αvβ5Breast cancer, ovarian cancer, esophageal squamous cell carcinoma, Glioblastoma and gliomaRegulate adhesion, migration;TAM, CAFs(72,100,116,162,163)
Baril et al, 2007; Boulter et al, 2021; Xu et al, 2016POSTN/PI3K/AktNon-small-cell lung cancer, colon cancer and cholangiocarcinomaPromote cell proliferation, invasion and migrationCAFs(88,163,166)
Shao et al, 2004; Zhou et al, 2015; Ashley et al, 2017POSTN/TGF-βHepatocellular carcinoma, Ovarian cancer and colorectal cancerPromotes transformation and maintenance of CSCs; promotes metastasisMacrophage, CAFs(60,101,167)
Zhou et al, 2015; Halperin et al, 2022; Kiesler et al, 2024; Jamaluddin et al, 2018POSTN/NF-κBOvarian cancer, breast cancer, colorectal cancer and glioblastomaPromotes cancer metastasisM2 macrophage, CAFs(101,116,168,169)
Nielsen et al, 2016; Muscarella et al, 2021MAPK/ERKTriple-negative breast cancer, glioblastomaEnhance invasion, migration, stemness, growth; chemoresistanceCSCs(170,173)
Moniuszko et al, 2024POSTN/WntNeuroblastomaPromotes invasion, adhesion and growthCSCs(66)
Huang et al, 2024; Wang et al, 2022POSTN/FAKColorectal cancer, papillary thyroid tumorPromotes tumorigenesisCAFs(67,102)

[i] CAFs, cancer-associated fibroblasts; TAM, tumor-associated macrophage; CSCs, cancer stem cells; POSTN, periostin.

Role of POSTN in the hallmarks of cancer

Cancer development is a multistep process initiated by oncogenic mutations that confer clonal advantage, ultimately driven by genomic instability (63). This progression is marked by acquired hallmarks including sustained proliferation, evasion of growth suppression, resistance to apoptosis, replicative immortality, angiogenesis, invasion/metastasis, metabolic reprogramming and immune escape. Emerging features such as phenotypic plasticity, epigenetic dysregulation, microbiome interactions and senescence further define tumorigenesis (1). Notably, POSTN overexpression and its alternatively spliced isoforms (such as, in renal, lung, breast and gastrointestinal cancer) regulate multiple oncogenic processes (13,40,64).

Cell proliferation

Cell proliferation is a process regulated by the synergistic action of mitogenic growth-promoting and antiproliferative signals (65). POSTN has been shown to induce proliferation in a variety of tumors. For example, POSTN can interact with the classical Wnt signaling pathway to promote colorectal tumorigenesis (66) and proliferation in neuroblastoma (67). POSTN from tumor stromal fibroblasts promotes colorectal carcinogenesis by activating YAP/TAZ through the Integrin-focal adhesion kinase (FAK)-Src signaling pathway (19). POSTN also promotes gastric cancer proliferation studies revealed that POSTN enhances the proliferation of OCUM-2MLN and OCUM-12 diffuse gastric cancer cell lines in vitro through activation of ERK, intuitively, tumor growth in POSTN (−/−) mice is markedly slower compared with that in wild-type mice (68). POSTN-promoted proliferation of tumor cells is also exemplified in carcinomas such as the A549 lung carcinoma cell line (69), triple-negative breast carcinoma (17) and melanoma (70).

However, POSTN activity is not exclusively associated with tumor growth. Shao et al (60) established stable POSTN-overexpressing cell lines using three POSTN-deficient tumor models, demonstrating that POSTN overexpression did not alter proliferation rates under in vitro culture conditions but considerably enhanced invasive capacity.

Activating invasion and metastasis

Besides promoting cell proliferation, POSTN also carries out a key role in activating invasion and metastasis. Tumor invasion and metastasis are an inefficient process requiring the overcoming of several rate-limiting steps, the occurrence of which often portends malignancy and a poor prognosis (71). Due to the heterogeneity of tumor cells, tumor invasion and metastasis are difficult to treat and remain one of the major challenges in the field of tumor therapy (72).

Epithelial-mesenchymal transition (EMT), initially identified during embryogenesis, is now recognized as a key driver of developmental processes, disease progression and tumor invasion (73). POSTN is involved in tumor invasion and metastasis by regulating EMT, the classical way of which is that POSTN binds to integrins and activates Akt/PKB and FAK-mediated signaling pathways (13). Studies demonstrate that stromal POSTN expression in ovarian cancer promotes tumor invasion and metastasis via PI3K/Akt pathway activation, mechanistically driving EMT (27,74,75). POSTN activates STAT3 signaling to modulate EMT markers such as caveolin-1 and angiogenesis-related genes such as HIF-1α and vascular endothelial growth factor-A (VEGF-A), with its knockdown shown to reverse glioblastoma stem cell (GSC) resistance to anti-VEGF-A therapy (76). POSTN has also been demonstrated to induce EMT in hepatoblastoma (77) and prostate cancer (78). Moreover, POSTN can inhibit specific targeted microRNAs (miRNAs) involved in the regulation of EMT. miRNAs are small endogenous RNAs that post-transcriptionally regulate gene expression and may target up to one-third of human mRNAs (79). Song et al (80) found that the POSTN gene was markedly upregulated in the gastric cancer cell line SGC-7901 treated with miR-148b mimic, which suggests the possible involvement of POSTN in the pathways related to the development and progression of gastric cancer. In lung cancer, POSTN promotes EMT through the MAPK/miR-381 axis (81).

POSTN promotes cell motility to promote tumor metastasis. It was demonstrated that secretion of POSTN by von Hippel-Lindau oncogene non-expressing (VHL-) cells enhanced the motility of VHL-WT cells and promoted vascular escape in clear cell renal cell carcinoma tumor cells (82). In a study on colorectal cancer, POSTN was found to activate FAK and AKT or STAT3, accelerating metastasis and activating migration of tumor and stromal cells through stromal remodeling capacity (83). These studies suggest that POSTN carries out an important role in the development of the TME and that increased expression of POSTN in the stroma is somewhat indicative of tumor stage and prognosis (84).

Interestingly, POSTN largely promotes tumor-invasive metastasis, yet it is regarded as a tumor suppressor in bladder cancer (85). This may be associated with the selective sharing of POSTN. Of the two splicing variations of POSTN (variants I and II) isolated from bladder cancer, Variant II has key domains that interact with matrix proteins such as integrins, inhibiting cancer cell invasion and metastasis. While Variant I, lacking certain exons, has a changed structure that weakens these interactions, losing its inhibitory function and potentially promoting cancer progression (42).

Inducing vasculature

Tumor angiogenesis enables nutrient and oxygen delivery to growing malignancies, with aberrant vascular features, such as disorganized architecture, sinusoidal vessel formation and functional heterogeneity, distinguishing tumor vasculature from normal tissues (86). VEGF is a central driver of angiogenesis, which binds tyrosine kinase receptors Flt-1 and Flk-1/KDR to initiate signaling. POSTN has been identified as a pro-angiogenic factor that facilitates tumor neovascularization via 'vascular co-option', exploiting pre-existing vessels in adjacent healthy tissues (87). When the tumor volume is >2 mm3, increased expression of POSTN enhances the resistance of cancer cells to hypoxic conditions (88). For instance, in colorectal cancer, POSTN activates the Akt/PKB pathway to increase endothelial cell survival and vascular proliferation, correlating with metastasis and serving as a prognostic marker (89). Similarly, ovarian cancer studies reveal that POSTN overexpression drives angiogenesis and metastatic spread (11,90-92). Mechanistically, POSTN upregulates Flk-1/KDR expression in endothelial cells via integrin αvβ3-FAK signaling, a pathway associated with breast cancer progression (22,60).

POSTN also promotes lymphangiogenesis, a key step in metastatic dissemination. Gillot et al (93) demonstrated that POSTN enhances VEGF-C-mediated lymphangiogenesis and primes lymph nodes for metastatic colonization. In head and neck squamous cell carcinoma (HNSCC), POSTN drives lymphatic remodeling through dual mechanisms: Indirect upregulation of VEGF-C mRNA and direct activation of Src/Akt pathways. This aligns with observed associations between POSTN-overexpressing xenografts and lymphatic involvement in HNSCC (94). Collectively, POSTN orchestrates tumor vascularization and lymphatic network expansion, fostering metastatic efficiency.

Evasion of apoptosis

Apoptosis is a regulated form of programmed cell death, which carries out dual roles in tumor biology. Its dysregulation contributes to tumor progression, therapy resistance and metabolic reprogramming by enabling uncontrolled proliferation or insufficient apoptosis (95). Apoptosis also interacts with tumor metabolism to collectively drive malignancy (96). Notably, apoptosis can paradoxically support tumor survival through microenvironmental interactions (97). Emerging concepts such as pan-apoptosis (combined apoptosis-necroptosis) further highlight the complexity of apoptosis regulation in cancer immunity (98).

ECM protein POSTN enhances tumor cell survival under stress conditions (hypoxia, nutrient deprivation). Mechanistically, POSTN activates β4 integrin/PI3K and Akt/PKB pathways to suppress apoptosis in pancreatic, colorectal and non-small cell lung cancer (88,89,99). In hypoxic pancreatic tumors, POSTN drives a pathological cycle: Hypoxia induces stellate cells to secrete POSTN, which sustains fibrosis and cellular viability while exacerbating hypoxia (100). This ECM-mediated adaptation enables tumor persistence across multiple cancer types by counteracting environmental stressors and death signals.

Avoiding immune destruction

Immunomodulation carries out an important role in tumor development and signaling between POSTN and immune cells can take place, thus carrying out a role in the immune escape of tumors. A study noted that GSCs in glioblastoma multiforme (GBM) secrete POSTN, which can recruit M2-type tumor-associated macrophages (TAMs) to support GBM growth (101). Similarly, POSTN can promote ovarian cancer metastasis by enhancing M2 macrophages and cancer-associated fibroblasts (CAFs) through integrin-mediated NF-κB and TGF-β2 signaling (74). Wang et al (102) discovered that POSTN+ CAFs can recruit secreted phosphoprotein 1 (SPP1)+ macrophages and trigger an increase in SPP1 expression via the IL-6/STAT3 signaling pathway, thereby enhancing therapeutic resistance in hepatocellular carcinoma (HCC). In addition, a recent study have shown that thyroid tumor cells and CAFs can promote tumor development through crosstalk between IL-4 and POSTN (103). The aforementioned studies may suggest new targets for cancer inhibition.

Cellular senescence

Cell senescence is a stress-induced stable growth arrest that has been regarded as a passive bystander in the cessation of proliferation. However, research over the past decade suggests that senescence also serves as the foundation for tissue development and function (104-107). For tumors, cellular senescence has two opposing functions. Firstly, senescence halts the cell cycle, thereby preventing the formation of malignant tumor cells and promoting tissue regeneration and repair (108). Secondly, senescent cells present in the TME can induce persistent inflammatory states and reprogram the TME (109) through the production of the senescence-associated secretory phenotype, thereby promoting tumor growth, metastasis and drug resistance (110). There is currently evidence suggesting an association between POSTN and cellular senescence, warranting further investigation. Zhu et al (111) experiments indicate that POSTN may promote nucleus pulposus cell senescence and ECM metabolism by activating the NF-κB and Wnt/β-catenin signaling pathways, carrying out a notable role in the progression of disc degeneration. Overexpression of POSTN simultaneously leads to fibrosis and senescence of cardiomyocytes (112). Similarly, POSTN has been revealed to promote kidney aging by participating in fibrosis and lipid metabolism (113). Huang et al (114) used RNA-Sequencing technology to examine gene expression changes in primary skin fibroblasts from healthy controls and patients with pyrroloquinoline quinone reductase 1 (PYCR1) mutations, and found that POSTN was one of the most downregulated candidate genes. To the best of our knowledge, although no published articles have yet described how POSTN influences cancer development through aging, the aforementioned discussions provide potential avenues for future research in this direction.

Epigenetic reprogramming

Epigenetic and metabolic reprogramming are closely related and mutually regulate each other, driving immune escape or hindering immune surveillance in certain cases, and carrying out an important role in tumor progression (115). CAFs are recognized as key components of the TME, which are stable at the genomic level but differ considerably from normal stromal precursors (116). The transformation of normal fibroblasts into CAFs involves extensive methylation changes, which can be induced by various factors such as TGFβ and pro-inflammatory cytokines such as leukemia inhibitory factor (117,118). The epigenetic modifications in CAFs not only activate and maintain their pro-invasive phenotype (118) but also enhances glycolysis in tumor metabolism. Becker et al (119) demonstrated that the epigenetic reprogramming of CAFs disrupts glucose metabolism and promotes breast cancer progression. Additionally, studies have elucidated the promotional role of the epigenetics of CAFs in prostate cancer (120) and bladder urothelial carcinoma (121). POSTN carries out an important role as a marker of CAFs in this process. Han et al (122) reported that POSTN is one of the DNA methylation biomarkers of nasopharyngeal carcinoma and patients with high POSTN expression have shorter overall survival times. The methylation of POSTN is also associated with the molecular mechanisms of important pathways associated with glioblastoma formation and patient overall survival (123).

POSTN is a multifunctional, pleiotropic molecule in cancer, with its effects exhibiting high context-dependence. It serves as a potent 'oncogenic factor' while also potentially assuming the complex role of a 'potential collaborator' under specific conditions, with the underlying core being the dynamic changes in TME (124,125). First, cancer of different tissue origins and molecular subtypes exhibit considerable differences in their TME. POSTN can be secreted by various cell types (such as, tumor cells and CAFs) and its effects may depend on the source cells as well as the specific receptors and downstream signaling pathways with which it interacts. POSTN typically promotes immune suppression; however, in certain contexts (such as specific immunotherapy combinations), the immune cells it recruits may unexpectedly alter the balance of the TME, potentially producing effects favorable to therapeutic response. This is not a direct 'beneficial' effect but an indirect outcome of specific contextual factors (126). Additionally, different splice variants of POSTN can have opposed effects, as demonstrated in bladder cancer (85).

Role of POSTN in the TME

Cancer is driven by the accumulation of mutations in cancerous cells but is not exclusively genetically determined. The majority of cancer types (90-95%) occur in close association with the environmental exposures and lifestyle choices of an individual, while genetic factors account for only a small proportion (5-10%) of all cancer cases (127).

TME refers to all the non-cancerous cells and non-cellular components in a tumor, which can be regarded as a special ecological niche where tumor cells are located. The cellular components include fibroblasts, adipocytes, endothelial cells, infiltrating immune cells such as lymphocytes and dendritic cells; while the non-cellular components include ECM and soluble components such as hormones, growth factors or cytokines (12). These components were previously considered 'innocent bystanders', but several studies have revealed that constant, reciprocal and dynamic interactions between tumor cells and the TME (2,128) are key in the cancer process (129) (Fig. 2). Current evidence suggests that POSTN activates key downstream signaling pathways by interacting with its receptor integrin, remodels the ECM by interacting with other ECM proteins [such as type I collagen, fibronectin, tenascin C and thrombospondin 1 (TSP-10)] (44) and remodels TME by cross-talking with other signaling molecules and recruiting inflammatory and immune cells (3).

Roles of POSTN in the development of
malignant cancer. POSTN is a key driver of tumor progression and
metastasis. It promotes cancer stem cell proliferation via the
POSTN/TGF-β pathway and induces angiogenesis through VEGFR2. POSTN
recruits stromal cells to form a fibrotic, immunosuppressive tumor
microenvironment that supports tumor growth, protects disseminated
tumor cells, and facilitates invasion. Furthermore, POSTN
contributes to the formation of pre-metastatic niches and bone
marrow colonization, enabling DTC dormancy and outgrowth. MDSC,
myeloid-derived suppressor cells; HSCs, hematopoietic stem cells;
DTC, disseminated tumor cell; EV, extracellular vesicle; CAFs,
cancer-associated fibroblasts; NK, natural killer; POSTN,
periostin; PMN, pre-metastatic niche.

Figure 2

Roles of POSTN in the development of malignant cancer. POSTN is a key driver of tumor progression and metastasis. It promotes cancer stem cell proliferation via the POSTN/TGF-β pathway and induces angiogenesis through VEGFR2. POSTN recruits stromal cells to form a fibrotic, immunosuppressive tumor microenvironment that supports tumor growth, protects disseminated tumor cells, and facilitates invasion. Furthermore, POSTN contributes to the formation of pre-metastatic niches and bone marrow colonization, enabling DTC dormancy and outgrowth. MDSC, myeloid-derived suppressor cells; HSCs, hematopoietic stem cells; DTC, disseminated tumor cell; EV, extracellular vesicle; CAFs, cancer-associated fibroblasts; NK, natural killer; POSTN, periostin; PMN, pre-metastatic niche.

POSTN in the metastatic niche

Tumor metastasis is an inefficient process where the majority of disseminated cells die, with only rare cells successfully colonizing distant sites. Emerging evidence implicates cancer stem cells (CSCs) as key drivers of metastasis, facilitated by interactions with the metastatic niche (130-133). Stromal fibroblast-derived POSTN, a component of normal stem cell niches, mediates CSC-niche crosstalk, as demonstrated by Malanchi et al (134). In metastatic tissues, POSTN is primarily secreted by CAFs, which are activated by paracrine or autocrine signals to coordinate metastasis through direct interactions with tumor cells and stromal components (135). For example, in colorectal cancer, POSTN overexpression in hepatic metastatic niches promotes tumor cell survival, angiogenesis and metastasis via the Akt/PKB pathway (89). Similarly, cervical squamous cell carcinoma studies reveal that POSTN+ CAFs disrupt lymphatic endothelial barriers by activating integrin-FAK/Src-VE-calmodulin signaling, facilitating dissemination (136-138). In ovarian cancer, POSTN drives metastasis through autocrine NF-κB/TGF-β2 activation in tumor cells and by recruiting M2 macrophages and CAFs. Stromal POSTN also enhances metastatic infiltration in skin and head-neck squamous carcinomas (74). Furthermore, POSTN can synergize with TNC, for example, POSTN may aggregate Wnt for stem cells, while TNC may enhance the response of these cells to Wnt and Notch (139) are 'two sides of the same metastatic ecological niche'.

POSTN in the CSC niche

CSCs or tumor-initiating cells, drive tumor heterogeneity through self-renewal, clonal initiation and long-term repopulation capabilities. The CSC niche preserves these properties, shields CSCs from immune surveillance and facilitates metastasis (132). As highlighted by Malanchi et al (134), CD90+CD24+ breast CSCs from primary tumors metastasize to the lungs and activate CAFs to secrete POSTN. This stromal POSTN recruits Wnt ligands into CSCs, amplifying Wnt signaling to fuel CSC expansion and metastatic colonization. In basal-like breast carcinoma, POSTN maintains CSC traits and mesenchymal phenotypes by regulating NF-κB-dependent cytokines (IL4 and IL8) via ERK signaling, thereby promoting metastasis (140). Similarly, ovarian cancer studies show that POSTN enhances CSC stemness, increasing invasiveness, therapy resistance and recurrence (20,27,74,90,141). In colon cancer, high Wnt activity defines CSC stemness (142) and POSTN interacts with Wnt ligands (Wnt1/Wnt3A) in colon CSCs, suggesting its role in reinforcing stemness (143). Further supporting this paradigm, Chen et al (144) identified a POSTN/TGF-β1 positive feedback loop in HCC. Through analysis of 110 HCC patient tissues and xenograft models, this study demonstrated that POSTN/TGF-β1 activates AP-2α transcription to sustain HCC CSC stemness and accelerate malignancy.

POSTN in the perivascular niche

Perivascular niche is key for maintaining both hematopoietic stem cells and CSCs, serving as a dynamic hub in cancer evolution (83). This niche context-dependently regulates tumor dormancy, metastatic spread, stemness and immune evasion, shaping tumor progression (145). Tumor cells exploit this niche to invade blood vessels (extravasation), a process involving endothelial cell contractility (via myosin) and mechanical interactions with the subendothelial matrix (146). However, disseminated tumor cells (DTCs) often face suppression in secondary organs, with only a subset surviving or entering dormancy. DTC 'awakening' represents a key rate-limiting step for metastasis. Stable vasculature suppresses proliferation through stromal TSP-1, while sprouting neo vessels exhibit pro-metastatic properties: Their endothelial cells upregulate POSTN, tenascin-C, fibronectin and TGF-β1, creating a microenvironment that drives tumor reactivation (3,147-149).

The perivascular niche also modulates immunity by facilitating immune cell infiltration. Perivascular TAMs promote angiogenesis, immunosuppression and tumor survival (150). POSTN recruits TAMs to remodel this niche, as seen in GBM, where GSCs secrete POSTN to attract and polarize TAMs toward the pro-tumor M2 subtype. This POSTN-mediated crosstalk sustains GSC-TAM clustering in perivascular regions, fostering therapy resistance and recurrence (101,151,152). These dual roles, structural support for metastasis and immunomodulation, highlight the perivascular niche as a promising therapeutic target.

POSTN in the pre-metastatic niche (PMN)

PMN, consisting of distinct resident cell types, ECM components and infiltrating cell populations, is a microenvironment induced by tumors in distant tissues that can provide a conducive environment for the proliferation of incoming tumor cells (153). It is currently considered that distant sites can be reprogrammed into the PMN by extracellular vesicles (EVs) carrying tumor-secreted factors. Potential mechanisms include enhanced angiogenesis and vascular remodeling, stromal remodeling, inhibitory effects of EVs on immune cells and alteration of the metabolic milieu. Formation of PMN is an integrative process that extends from the local to the systemic level (146,154,155).

POSTN can be induced by tumor products that reach distant tissues first and participate in pre-metastatic ecological niche formation (134). A study using lung fibroblasts isolated from the lung tissues of C5BL/6 J mice revealed that CAF EVs induced pre-metastatic ecotone formation in the SACC lungs of mice and demonstrated more robust matrix remodeling than SACC EVs. This may be due to POSTN activation of the TGF-β signaling pathway in lung fibroblasts (156). Previous studies reveal multifaceted roles of POSTN in pre-metastatic niche formation: Tumor-derived EVs deliver lncRNA MALAT1 to mediate M2 macrophage polarization via the POSTN/Hippo/YAP axis in triple-negative breast cancer (157), while colorectal cancer EVs carrying ITGBL1 reprogram fibroblasts to prime metastatic sites (158). POSTN facilitates immunosuppression by engaging myeloid-derived suppressor cells in tumor immune evasion (159) and promotes niche establishment through trauma-induced secretion in melanoma (160). Exosome-mediated POSTN transfer modifies distant tissue microenvironments (99), with elevated exosomal POSTN levels in thyroid cancer patients directly accelerating progression via niche remodeling (161).

POSTN in the fibrotic niche

Formation of a fibrotic microenvironment is associated with a series of events, including tissue injury, inflammation, myofibroblast differentiation and ultimately the excessive deposition of ECM components. The ECM comprises fibrous proteins and glycoproteins, forming a dynamic 3D structure that regulates cell adhesion, migration, tissue repair and disease processes through mechanotransduction (162,163). POSTN promotes fibrosis in pathological and cancer contexts, by interacting with soluble factors to induce myofibroblast differentiation (61,164). Activated myofibroblasts secrete a large amount of ECM components, primarily collagen, elastin and glycosaminoglycans (165).

The formation of fibrotic niches is key for tumor metastasis and the growth of tumors characterized by fibroproliferation. POSTN is one of the markers of fibrotic niches and it primarily functions through interactions with key signaling pathways such as TGF-β1 (61) and Wnt/β-catenin (166). Elevated POSTN levels in melanoma lung metastases associate with bleomycin-induced pulmonary fibrosis progression (167). In uterine fibroids, POSTN acts as both a biomarker and a therapeutic target by stimulating collagen overproduction and myometrial cell transformation (168,169). Additionally, metastasis-associated macrophages activate hepatic stellate cells via granulin secretion, generating POSTN-producing myofibroblasts that remodel liver stroma for tumor growth (170).

POSTN in the bone marrow microenvironment

Bone is a frequent metastatic site for solid tumors (for example, breast, prostate and lung cancer) (171,172). Bone marrow, a semi-solid tissue within bone cavities, facilitates hematopoiesis via hematopoietic stem cells and comprises cellular components such as mesenchymal stromal cells, osteoblasts and immune cells, as well as non-cellular elements including cytokines and growth factors (173,174). This microenvironment protects DTCs during colonization, dormancy and growth, and contributes to hematologic malignancies such as leukemia and myeloma (175,176).

POSTN is associated with bone metastasis. In breast cancer, BMSC-derived POSTN promotes bone metastasis, with murine models showing an 8-fold POSTN increase at metastasis sites compared with controls (177,178). Contrastingly, C-terminal intact periosteal protein (iPTN) levels decrease in breast cancer bone metastases due to histone K cleavage, suggesting iPTN as a potential biomarker for bone recurrence detection (179). POSTN also drives HNSCC progression via BMSC-induced PI3K/Akt/mTOR pathway activation (180). In multiple myeloma, elevated POSTN levels are associated with advanced disease severity, such as International Staging System stage 3 (181) and with dysregulation of bone metabolism, including an inverse relationship with bone formation markers and a positive correlation with bone resorption markers (182). Additionally, POSTN facilitates prostate cancer bone metastasis via integrin-mediated interactions with osteoblasts and tumor cells (183) and supports B-cell Acute Lymphoblastic Leukemia cell proliferation in the bone marrow niche (184).

Clinical translation of POSTN: Biomarker value and therapeutic targeting prospects

Although extensive basic research has confirmed that POSTN carries a central role in various pathological processes (185-187), its successful translation from a basic molecular marker to a clinically useful tool remains challenging. Currently, the practical application value of POSTN as a clinical biomarker is still under investigation, and there remains a notable gap between its potential applications and clinical practice.

POSTN as a diagnostic and prognostic biomarker

Since the pathological expression levels of POSTN are markedly higher when compared with normal expression levels (64,188), POSTN has potential as a diagnostic and prognostic biomarker for diseases. Since the upregulation of POSTN is not specific to any particular disease, this may limit its application as a standalone diagnostic biomarker. Currently, research is focused on combining POSTN with other biomarkers to develop more precise diagnostic and prognostic prediction models. A study demonstrated that the combination of plasma POSTN and CA19.9 considerably improved the sensitivity, specificity and AUC for diagnosing cholangiocarcinoma compared with using a single marker, reaching 87 and 91%, and 0.94, respectively (plasma POSTN: 78 and 85%, and 0.86; CA19.9: 67 and 90%, and 0.86) (189). Jia et al (25) demonstrated that, to distinguish healthy controls from locally advanced BCa, POSTN exhibited the highest AUC (AUCPOSTN=0.72, AUCCA153=0.57 and AUCCEA=0.62), and the AUC of CA153 and CEA were markedly improved when used in combination with POSTN. Furthermore, multiple studies have demonstrated that elevated serum POSTN levels are associated with worse overall survival and progression-free survival (7,190-193). In summary, the potential role of POSTN as a diagnostic and prognostic biomarker holds clinical value.

Subtype-specific strategies

With the advent of the era of precision medicine, accurate identification of subtypes has become increasingly important. In various types of tumors, the 'POSTN+' subtype is typically associated with features such as stromal proliferation, EM and TM characteristics (17,102,126). Patients with this subtype often exhibit greater invasiveness, increased metastasis risk and worse response rates to conventional chemotherapy. By contrast, the 'POSTN-' subtype is associated with relatively improved prognosis. Zhang et al (141) revealed that the stroma markedly contributes to the molecular classification of the mesenchymal subtype in high-grade serous ovarian cancer, with POSTN being one of the genes predominantly expressed in the stroma. In osteosarcoma, POSTN-positive expression is notably associated with histological subtype (P=0.000), Enneking staging (P=0.027) and tumor size (P=0.009) (194).

In invasive basal cell carcinoma (BBC), the two upregulated genes POSTN and WISP1 were validated at the protein level as being associated with invasive BCC (195). This subtype classification lays the foundation for developing precision strategies. A molecular typing kit based on POSTN expression levels could be developed to identify high-risk patient groups, guiding more intensive follow-up and early intervention. In terms of treatment, it provides a basis for achieving stratified therapy. Patients with high POSTN expression in this subtype may be the optimal candidate population for targeted therapies targeting the POSTN pathway itself or its downstream effector molecules or for immunotherapies targeting the immunosuppressive microenvironment.

Additionally, new CAF subtypes can be defined based on POSTN expression levels. Wang et al (102) recently found that high infiltration of POSTN+ CAFs and SPP1+ macrophages is associated with resistance to immunotherapy. For example, a study identified five CAF subtypes in gastric cancer, revealing that the CAF_0 subtype was highly associated with poor prognosis, with POSTN being the most notable marker gene for this subtype (196). These findings suggest that targeting specific CAF subpopulations may have potential benefits for improving clinical responses to immunotherapy.

Therapeutic clinical outlook

Targeted therapy for POSTN has progressed from the proof-of-concept stage to practical application. In preclinical studies, various targeted strategies have shown great promise. For example, PNX-001 (a humanized anti-POSTN monoclonal antibody) effectively blocks the interaction between POSTN and integrin in preclinical models, inhibits tumor growth and metastasis, and enhances the delivery of chemotherapy drugs (197). Another neutralizing monoclonal antibody targeting POSTN (MZ-1) has been shown to have a considerable growth-inhibitory effect on a subgroup of invasive ovarian tumors that overexpress POSTN (198). PNDA-3 (human osteopontin-3) is a DNA aptamer that selectively binds to the FAS-1 domain of POSTN, disrupting its interaction with its cell surface receptors αvβ3 and αvβ5 integrins, thereby markedly reducing primary tumor growth and distant metastasis (199,200).

Due to the increasing number of POSTN-related diseases, several clinical trials are currently underway. However, therapies targeting POSTN in the field of oncology remain at a relatively early stage, with very limited publicly available clinical data. Key future challenges include: Determining the optimal treatment window, selecting appropriate biomarkers to screen for patient populations likely to respond and exploring combination strategies with other therapies (such as immune checkpoint inhibitors, anti-angiogenic drugs) to overcome the complexity of the TME.

Future challenges and possible solutions

Despite considerable progress in POSTN research, there remain notable knowledge gaps and technical limitations in this field, which hinder its clinical translation process. First, there are conflicting data in the existing literature. In certain types of cancer, high POSTN expression is associated with poor prognosis [such as, breast cancer (13,128,130,139), glioblastoma (101,123), ovarian cancer (20,27,74,91,141) and liver cancer (13,62,71,89,193), but in other types of cancer (201,202), such as estrogen receptor-negative (ER-) or progesterone receptor-negative (PR-) phenotypes of breast cancer (203), its expression levels show only a modest association with clinical outcomes (203-205). These inconsistencies may stem from methodological heterogeneity, differences in patient cohorts and variations in the cellular origin and tissue localization of POSTN within the TME. Typically, POSTN is present in the ECM, while the main presence of POSTN in tumor tissues is in the cancer matrix (206). Additionally, current detection methods struggle to distinguish between different POSTN splice variants, which may possess entirely different or even opposing biological functions, potentially contributing to the data inconsistencies (34). Current research also faces notable technical limitations. The majority of studies rely on static measurements from tissue biopsies, which cannot capture the dynamic changes in POSTN expression or its real-time functional role in treatment response and disease progression. More importantly, complete inhibition of POSTN may interfere with important physiological functions, leading to side effects such as impaired wound healing, abnormal bone fibrosis or cardiac dysfunction (64). Although the role of periosteal proteins in promoting metastasis and chemotherapy resistance has been well established, it remains unclear how targeting periosteal proteins interacts with existing treatment modalities, such as chemotherapy, immunotherapy or radiotherapy, within the TME.

To overcome these challenges and drive further development in this field, future research should establish unified POSTN detection standards and conduct large-scale, multicenter prospective cohort studies to systematically assess the predictive and prognostic value of POSTN in different disease stages and treatment settings. Additionally, novel dynamic imaging technologies, such as molecular imaging probes capable of non-invasively and real-time monitoring of POSTN expression and functional activity within the body, should be actively developed. In terms of treatment strategies, the focus should be on precision interventions, including the design of inhibitors that can specifically block pathological protein interactions (such as integrin binding) without affecting physiological functions, the development of drugs targeting disease-specific isoforms and the exploration of prodrug strategies based on TME activation to minimize off-target risks to normal tissues. Finally, any clinical translation research must comprehensively assess the potential long-term effects of inhibiting POSTN on the regenerative and reparative functions of key organs such as bones, skin and the heart during the preclinical stage to ensure treatment safety. By adopting these recommendations, future research will be able to more reliably evaluate the clinical efficacy of POSTN and design safer, more effective targeted treatment strategies, ultimately bridging the gap between basic research and clinical application.

Concluding remarks

As an important matricellular protein, POSTN carries out multifaceted roles in both physiological and pathological processes, including embryonic development, allergic inflammation, fibrotic diseases and tumorigenesis (12,64). Increasing evidence indicates that POSTN can regulate various cancer hallmarks by interacting with integrins and modulating key signaling pathways such as Wnt/β-catenin, PI3K/Akt and TGF-β (27,49,51,61,74,111,166,168,180,188,207). The present review focuses on the promoting effects of POSTN on tumor growth, invasion-metastasis, angiogenesis, anti-apoptosis and immune evasion. Considering the increasingly recognized role of TME, POSTN also exerts considerable effects on niche regulation, particularly in the maintenance of CSCs, remodeling of the ECM and immune-suppressive reprogramming (102,135,140,144,208).

These findings suggest a promising therapeutic and prognostic potential for targeting POSTN in tumor management. In terms of cancer treatment, the main approaches include blocking the POSTN signaling pathway and using POSTN inhibitors. Studies have shown that Let-7f reduces angiogenesis mimicry in glioma cells by inhibiting POSTN (209,210). DNA aptamers targeting POSTN have been revealed to effectively inhibit the growth and metastasis of breast and gastric cancer in mouse models (209). POSTN can also be combined with immune checkpoint inhibitors to enhance the therapeutic efficacy of colorectal tumors (211). Moreover, selecting appropriate POSTN isoforms is important. For instance, POSTN-203 may be a more promising target for glioma because it is associated with low patient survival and contains multiple predicted human leukocyte antigen-restricted epitopes (40). POSTN can also serve as a prognostic biomarker for gastrointestinal malignancies, hepatobiliary and pancreatic cancer (212), breast cancer, ovarian cancer (20) and genitourinary cancer (59).

Key knowledge gaps still exist, including the functional heterogeneity of POSTN isoforms, the detailed mechanisms of its molecular interactions and the potential off-target effects of POSTN-targeted therapies. In summary, future research should prioritize addressing these challenges to advance POSTN-based diagnostics and therapeutics and ultimately improve the clinical outcomes of cancer and associated pathologies.

Availability of data and materials

Not applicable.

Authors' contributions

YX have made substantial contributions to the conception of the present review, wrote the main manuscript text, prepared Figs. 1 and 2, and reviewed the manuscript. LW made substantial contributions to the conception of the present review and reviewed the manuscript. Data authentication 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

The present work was supported by the Open Research Fund of Hubei Province Key Laboratory of Precision Radiation Oncology (grant no. 2024ZLJZFL003) and the Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education (grant no. 2024MZFS010), which are awarded to Dr Lufang Wang, Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China.

References

1 

Hanahan D: Hallmarks of cancer: New Dimensions. Cancer Discov. 12:31–46. 2022. View Article : Google Scholar : PubMed/NCBI

2 

Xiao Y and Yu D: Tumor microenvironment as a therapeutic target in cancer. Pharmacol Ther. 221:1077532021. View Article : Google Scholar :

3 

Liu AY, Zheng H and Ouyang G: Periostin, a multifunctional matricellular protein in inflammatory and tumor microenvironments. Matrix Biol. 37:150–156. 2014. View Article : Google Scholar : PubMed/NCBI

4 

Takeshita S, Kikuno R, Tezuka K and Amann E: Osteoblast-specific factor 2: Cloning of a putative bone adhesion protein with homology with the insect protein fasciclin I. Biochem J. 294:271–278. 1993. View Article : Google Scholar : PubMed/NCBI

5 

Horiuchi K, Amizuka N, Takeshita S, Takamatsu H, Katsuura M, Ozawa H, Toyama Y, Bonewald LF and Kudo A: Identification and characterization of a novel protein, periostin, with restricted expression to periosteum and periodontal ligament and increased expression by transforming growth factor beta. J Bone Miner Res. 14:1239–1249. 1999. View Article : Google Scholar : PubMed/NCBI

6 

Sasaki H, Dai M, Auclair D, Fukai I, Kiriyama M, Yamakawa Y, Fujii Y and Chen LB: Serum level of the periostin, a homologue of an insect cell adhesion molecule, as a prognostic marker in nonsmall cell lung carcinomas. Cancer. 92:843–848. 2001. View Article : Google Scholar : PubMed/NCBI

7 

Sasaki H, Dai M, Auclair D, Kaji M, Fukai I, Kiriyama M, Yamakawa Y, Fujii Y and Chen LB: Serum level of the periostin, a homologue of an insect cell adhesion molecule, in thymoma patients. Cancer Lett. 172:37–42. 2001. View Article : Google Scholar : PubMed/NCBI

8 

Borg TK and Markwald R: Periostin: More than just an adhesion molecule. Circ Res. 101:230–231. 2007. View Article : Google Scholar : PubMed/NCBI

9 

Ruan K, Bao S and Ouyang G: The multifaceted role of periostin in tumorigenesis. Cell Mol Life Sci. 66:2219–2230. 2009. View Article : Google Scholar : PubMed/NCBI

10 

Kudo A: Periostin in fibrillogenesis for tissue regeneration: Periostin actions inside and outside the cell. Cell Mol Life Sci. 68:3201–3207. 2011. View Article : Google Scholar : PubMed/NCBI

11 

Zhu M, Fejzo MS, Anderson L, Dering J, Ginther C, Ramos L, Gasson JC, Karlan BY and Slamon DJ: Periostin promotes ovarian cancer angiogenesis and metastasis. Gynecol Oncol. 119:337–344. 2010. View Article : Google Scholar : PubMed/NCBI

12 

González-González L and Alonso J: Periostin: A matricellular protein with multiple functions in cancer development and progression. Front Oncol. 8:2252018. View Article : Google Scholar : PubMed/NCBI

13 

Morra L and Moch H: Periostin expression and Epithelial-mesenchymal transition in cancer: A review and an update. Virchows Arch. 459:465–475. 2011. View Article : Google Scholar : PubMed/NCBI

14 

Norris RA, Moreno-Rodriguez RA, Sugi Y, Hoffman S, Amos J, Hart MM, Potts JD, Goodwin RL and Markwald RR: Periostin regulates atrioventricular valve maturation. Dev Biol. 316:200–213. 2008. View Article : Google Scholar : PubMed/NCBI

15 

Sorocos K, Kostoulias X, Cullen-McEwen L, Hart AH, Bertram JF and Caruana G: Expression patterns and roles of periostin during kidney and ureter development. J Urol. 186:1537–1544. 2011. View Article : Google Scholar : PubMed/NCBI

16 

Sun D, Lu J, Tian H, Li H, Chen X, Hua F, Yang W, Yu J and Chen D: The impact of POSTN on tumor cell behavior and the tumor microenvironment in lung adenocarcinoma. Int Immunopharmacol. 145:1137132025. View Article : Google Scholar

17 

Lin S, Zhou M, Cheng L, Shuai Z, Zhao M, Jie R, Wan Q, Peng F and Ding S: Exploring the association of POSTN+ cancer-associated fibroblasts with Triple-negative breast cancer. Int J Biol Macromol. 268:1315602024. View Article : Google Scholar

18 

Kanemoto Y, Sanada F, Shibata K, Tsunetoshi Y, Katsuragi N, Koibuchi N, Yoshinami T, Yamamoto K, Morishita R, Taniyama Y and Shimazu K: Expression of periostin alternative splicing variants in normal tissue and breast cancer. Biomolecules. 14:10932024. View Article : Google Scholar : PubMed/NCBI

19 

Ma H, Wang J, Zhao X, Wu T, Huang Z, Chen D, Liu Y and Ouyang G: Periostin promotes colorectal tumorigenesis through integrin-FAK-Src Pathway-mediated YAP/TAZ activation. Cell Rep. 30:793–806.e6. 2020. View Article : Google Scholar : PubMed/NCBI

20 

Kujawa KA, Zembala-Nożyńska E, Cortez AJ, Kujawa T, Kupryjańczyk J and Lisowska KM: Fibronectin and periostin as prognostic markers in ovarian cancer. Cells. 9:1492020. View Article : Google Scholar : PubMed/NCBI

21 

Pickup MW, Mouw JK and Weaver VM: The extracellular matrix modulates the hallmarks of cancer. EMBO Rep. 15:1243–1253. 2014. View Article : Google Scholar : PubMed/NCBI

22 

Försti A, Jin Q, Altieri A, Johansson R, Wagner K, Enquist K, Grzybowska E, Pamula J, Pekala W, Hallmans G, et al: Polymorphisms in the KDR and POSTN genes: Association with breast cancer susceptibility and prognosis. Breast Cancer Res Treat. 101:83–93. 2007. View Article : Google Scholar

23 

Szefler S, Corren J, Silverberg JI, Okragly A, Sun Z, Natalie CR, Zitnik R, Siu K and Blauvelt A: Lebrikizumab decreases type 2 inflammatory biomarker levels in patients with asthma: Data from randomized phase 3 trials (LAVOLTA I and II). Immunotherapy. 16:1211–1216. 2024. View Article : Google Scholar

24 

Gan C, Li M, Lu Y, Peng G, Li W, Wang H, Peng Y, Hu Q, Wei W, Wang F, et al: SPOCK1 and POSTN are valuable prognostic biomarkers and correlate with tumor immune infiltrates in colorectal cancer. BMC Gastroenterol. 23:42023. View Article : Google Scholar : PubMed/NCBI

25 

Jia L, Li G, Ma N, Zhang A, Zhou Y, Ren L and Dong D: Soluble POSTN is a novel biomarker complementing CA153 and CEA for breast cancer diagnosis and metastasis prediction. BMC Cancer. 22:7602022. View Article : Google Scholar : PubMed/NCBI

26 

Alzobaidi N, Rehman S, Naqvi M, Gulati K and Ray A: Periostin: A potential biomarker and therapeutic target in pulmonary diseases. J Pharm Pharm Sci. 25:137–148. 2022. View Article : Google Scholar : PubMed/NCBI

27 

Yue H, Li W, Chen R, Wang J, Lu X and Li J: Stromal POSTN induced by TGF-β1 facilitates the migration and invasion of ovarian cancer. Gynecol Oncol. 160:530–538. 2021. View Article : Google Scholar

28 

Olsson L, Hammarström ML, Israelsson A, Lindmark G and Hammarström S: Allocating colorectal cancer patients to different risk categories by using a Five-biomarker mRNA combination in lymph node analysis. PLoS One. 15:e02290072020. View Article : Google Scholar : PubMed/NCBI

29 

Sugiura T, Takamatsu H, Kudo A and Amann E: Expression and characterization of murine osteoblast-specific factor 2 (OSF-2) in a baculovirus expression system. Protein Expr Purif. 6:305–311. 1995. View Article : Google Scholar : PubMed/NCBI

30 

Ducy P, Geoffroy V and Karsenty G: Study of Osteoblast-specific expression of one mouse osteocalcin gene: Characterization of the factor binding to OSE2. Connect Tissue Res. 35:7–14. 1996. View Article : Google Scholar : PubMed/NCBI

31 

Ulstrup JC, Jeansson S, Wiker HG and Harboe M: Relationship of secretion pattern and MPB70 homology with osteoblast-specific factor 2 to osteitis following Mycobacterium bovis BCG vaccination. Infect Immun. 63:672–675. 1995. View Article : Google Scholar : PubMed/NCBI

32 

Hoersch S and Andrade-Navarro MA: Periostin shows increased evolutionary plasticity in its alternatively spliced region. BMC Evol Biol. 10:302010. View Article : Google Scholar : PubMed/NCBI

33 

Nakama T, Yoshida S, Ishikawa K, Kobayashi Y, Abe T, Kiyonari H, Shioi G, Katsuragi N, Ishibashi T, Morishita R and Taniyama Y: Different roles played by periostin splice variants in retinal neovascularization. Exp Eye Res. 153:133–140. 2016. View Article : Google Scholar : PubMed/NCBI

34 

Ikeda-Iwabu Y, Taniyama Y, Katsuragi N, Sanada F, Koibuchi N, Shibata K, Shimazu K, Rakugi H and Morishita R: Periostin short fragment with Exon 17 via aberrant alternative splicing is required for breast cancer growth and metastasis. Cells. 10:8922021. View Article : Google Scholar : PubMed/NCBI

35 

De Oliveira Macena Y, Cezar MEN, Lira CBF, De Oliveira LBDM, Almeida TN, Costa ADAV, De Araujo BMD, de Almeida D Junior, Dantas HM, De Mélo EC, et al: The roles of periostin derived from Cancer-associated fibroblasts in tumor progression and treatment response. Cancer Metastasis Rev. 44:112024. View Article : Google Scholar : PubMed/NCBI

36 

Morra L, Rechsteiner M, Casagrande S, Duc Luu V, Santimaria R, Diener PA, Sulser T, Kristiansen G, Schraml P, Moch H and Soltermann A: Relevance of periostin splice variants in renal cell carcinoma. Am J Pathol. 179:1513–1521. 2011. View Article : Google Scholar : PubMed/NCBI

37 

Kuebart T, Oezel L, Gürsoy B, Maus U, Windolf J, Bittersohl B and Grotheer V: Periostin splice variant expression in human osteoblasts from osteoporotic patients and its effects on Interleukin-6 and osteoprotegerin. Int J Mol Sci. 26:9322025. View Article : Google Scholar : PubMed/NCBI

38 

Litvin J, Selim AH, Montgomery MO, Lehmann K, Rico MC, Devlin H, Bednarik DP and Safadi FF: Expression and function of Periostin-isoforms in bone. J Cell Biochem. 92:1044–1061. 2004. View Article : Google Scholar : PubMed/NCBI

39 

Viloria K and Hill NJ: Embracing the complexity of matricellular proteins: The functional and clinical significance of splice variation. Biomol Concepts. 7:117–132. 2016. View Article : Google Scholar : PubMed/NCBI

40 

Xiong Z, Sneiderman CT, Kuminkoski CR, Reinheimer J, Schwegman L, Sever RE, Habib A, Hu B, Agnihotri S, Rajasundaram D, et al: Transcript-targeted antigen mapping reveals the potential of POSTN splicing junction epitopes in glioblastoma immunotherapy. Genes Immun. 26:190–199. 2025. View Article : Google Scholar : PubMed/NCBI

41 

Wenhua S, Tsunematsu T, Umeda M, Tawara H, Fujiwara N, Mouri Y, Arakaki R, Ishimaru N and Kudo Y: Cancer Cell-derived novel periostin isoform promotes invasion in head and neck squamous cell carcinoma. Cancer Med. 12:8510–8525. 2023. View Article : Google Scholar : PubMed/NCBI

42 

Kim CJ, Isono T, Tambe Y, Chano T, Okabe H, Okada Y and Inoue H: Role of alternative splicing of periostin in human bladder carcinogenesis. Int J Oncol. 32:161–169. 2008.

43 

Kii I, Nishiyama T and Kudo A: Periostin promotes secretion of fibronectin from the endoplasmic reticulum. Biochem Biophys Res Commun. 470:888–893. 2016. View Article : Google Scholar : PubMed/NCBI

44 

Kii I and Ito H: Periostin and its interacting proteins in the construction of extracellular architectures. Cell Mol Life Sci. 74:4269–4277. 2017. View Article : Google Scholar : PubMed/NCBI

45 

Maruhashi T, Kii I, Saito M and Kudo A: Interaction between periostin and BMP-1 promotes proteolytic activation of lysyl oxidase. J Biol Chem. 285:13294–13303. 2010. View Article : Google Scholar : PubMed/NCBI

46 

Takayama I, Tanabe H, Nishiyama T, Ito H, Amizuka N, Li M, Katsube KI, Kii I and Kudo A: Periostin is required for matricellular localization of CCN3 in periodontal ligament of mice. J Cell Commun Signal. 11:5–13. 2017. View Article : Google Scholar :

47 

Orecchia P, Conte R, Balza E, Castellani P, Borsi L, Zardi L, Mingari MC and Carnemolla B: Identification of a novel cell binding site of periostin involved in tumour growth. Eur J Cancer. 47:2221–2229. 2011. View Article : Google Scholar : PubMed/NCBI

48 

Rusbjerg-Weberskov CE, Johansen ML, Nowak JS, Otzen DE, Pedersen JS, Enghild JJ and Nielsen NS: Periostin C-Terminal is intrinsically disordered and interacts with 143 proteins in an in vitro epidermal model of atopic dermatitis. Biochemistry. 62:2803–2815. 2023. View Article : Google Scholar : PubMed/NCBI

49 

Wang Z, Lin M, Pan Y, Liu Y, Yang C, Wu J, Wang Y, Yan B, Zhou J, Chen R and Liu C: Periostin+ myeloid cells improved long bone regeneration in a mechanosensitive manner. Bone Res. 12:592024. View Article : Google Scholar :

50 

Norris RA, Borg TK, Butcher JT, Baudino TA, Banerjee I and Markwald RR: Neonatal and adult cardiovascular pathophysiological remodeling and repair: Developmental role of periostin. Ann N Y Acad Sci. 1123:30–40. 2008. View Article : Google Scholar : PubMed/NCBI

51 

Yang L, Guo T, Chen Y and Bian K: The multiple roles of periostin in Non-neoplastic disease. Cells. 12:502022. View Article : Google Scholar

52 

Walker JT, McLeod K, Kim S, Conway SJ and Hamilton DW: Periostin as a multifunctional modulator of the wound healing response. Cell Tissue Res. 365:453–465. 2016. View Article : Google Scholar : PubMed/NCBI

53 

Matsumoto H: Serum periostin: A novel biomarker for asthma management. Allergol Int. 63:153–160. 2014. View Article : Google Scholar : PubMed/NCBI

54 

Danielides G, Lygeros S, Kanakis M and Naxakis S: Periostin as a biomarker in chronic rhinosinusitis: A contemporary systematic review. Int Forum Allergy Rhinol. 12:1535–1550. 2022. View Article : Google Scholar : PubMed/NCBI

55 

Maintz L, Welchowski T, Herrmann N, Brauer J, Traidl-Hoffmann C, Havenith R, Müller S, Rhyner C, Dreher A, Schmid M, et al: IL-13, periostin and dipeptidyl-peptidase-4 reveal endotype-phenotype associations in atopic dermatitis. Allergy. Jan 17–2023. View Article : Google Scholar : Epub ahead of print. View Article : Google Scholar

56 

Ono J, Takai M, Kamei A, Azuma Y and Izuhara K: Pathological roles and clinical usefulness of periostin in type 2 inflammation and pulmonary fibrosis. Biomolecules. 11:10842021. View Article : Google Scholar : PubMed/NCBI

57 

Mael-Ainin M, Abed A, Conway SJ, Dussaule JC and Chatziantoniou C: Inhibition of periostin expression protects against the development of renal inflammation and fibrosis. J Am Soc Nephrol. 25:1724–1736. 2014. View Article : Google Scholar : PubMed/NCBI

58 

Qiao B, Liu X, Wang B and Wei S: The role of periostin in cardiac fibrosis. Heart Fail Rev. 29:191–206. 2024. View Article : Google Scholar

59 

Nuzzo PV, Buzzatti G, Ricci F, Rubagotti A, Argellati F, Zinoli L and Boccardo F: Periostin: A novel prognostic and therapeutic target for genitourinary cancer? Clin Genitourin Cancer. 12:301–311. 2014. View Article : Google Scholar : PubMed/NCBI

60 

Shao R, Bao S, Bai X, Blanchette C, Anderson RM, Dang T, Gishizky ML, Marks JR and Wang XF: Acquired expression of periostin by human breast cancers promotes tumor angiogenesis through up-regulation of vascular endothelial growth factor receptor 2 expression. Mol Cell Biol. 24:3992–4003. 2004. View Article : Google Scholar : PubMed/NCBI

61 

Liu B, Wu T, Lin B, Liu X, Liu Y, Song G, Fan C and Ouyang G: Periostin-TGF-β feedforward loop contributes to tumour-stroma crosstalk in liver metastatic outgrowth of colorectal cancer. Br J Cancer. 130:358–368. 2024. View Article : Google Scholar

62 

Tilman G, Mattiussi M, Brasseur F, van Baren N and Decottignies A: Human periostin gene expression in normal tissues, tumors and melanoma: Evidences for periostin production by both stromal and melanoma cells. Mol Cancer. 6:802007. View Article : Google Scholar : PubMed/NCBI

63 

Zhang S, Xiao X, Yi Y, Wang X, Zhu L, Shen Y, Lin D and Wu C: Tumor initiation and early tumorigenesis: Molecular mechanisms and interventional targets. Signal Transduct Target Ther. 9:1492024. View Article : Google Scholar : PubMed/NCBI

64 

Dorafshan S, Razmi M, Safaei S, Gentilin E, Madjd Z and Ghods R: Periostin: Biology and function in cancer. Cancer Cell Int. 22:3152022. View Article : Google Scholar : PubMed/NCBI

65 

Pavlova NN, Zhu J and Thompson CB: The hallmarks of cancer metabolism: Still emerging. Cell Metab. 34:355–377. 2022. View Article : Google Scholar : PubMed/NCBI

66 

Moniuszko T, Wincewicz A, Koda M, Domysławska I and Sulkowski S: Role of periostin in esophageal, gastric and colon cancer. Oncol Lett. 12:783–787. 2016. View Article : Google Scholar : PubMed/NCBI

67 

Huang M, Fang W, Farrel A, Li L, Chronopoulos A, Nasholm N, Cheng B, Zheng T, Yoda H, Barata MJ, et al: ALK upregulates POSTN and WNT signaling to drive neuroblastoma. Cell Rep. 43:1139272024. View Article : Google Scholar : PubMed/NCBI

68 

Kikuchi Y, Kunita A, Iwata C, Komura D, Nishiyama T, Shimazu K, Takeshita K, Shibahara J, Kii I, Morishita Y, et al: The niche component periostin is produced by cancer-associated fibroblasts, supporting growth of gastric cancer through ERK activation. Am J Pathol. 184:859–870. 2014. View Article : Google Scholar : PubMed/NCBI

69 

Hong L, Sun H, Lv X, Yang D, Zhang J and Shi Y: Expression of periostin in the serum of NSCLC and its function on proliferation and migration of human lung adenocarcinoma cell line (A549) in vitro. Mol Biol Rep. 37:2285–2293. 2010. View Article : Google Scholar

70 

Kotobuki Y, Yang L, Serada S, Tanemura A, Yang F, Nomura S, Kudo A, Izuhara K, Murota H, Fujimoto M, et al: Periostin accelerates human malignant melanoma progression by modifying the melanoma microenvironment. Pigment Cell Melanoma Res. 27:630–639. 2014. View Article : Google Scholar : PubMed/NCBI

71 

Chambers AF, Groom AC and MacDonald IC: Dissemination and growth of cancer cells in metastatic sites. Nat Rev Cancer. 2:563–572. 2002. View Article : Google Scholar : PubMed/NCBI

72 

Parker TM, Henriques V, Beltran A, Nakshatri H and Gogna R: Cell competition and tumor heterogeneity. Semin Cancer Biol. 63:1–10. 2020. View Article : Google Scholar

73 

Huang Y, Hong W and Wei X: The molecular mechanisms and therapeutic strategies of EMT in tumor progression and metastasis. J Hematol Oncol. 15:1292022. View Article : Google Scholar : PubMed/NCBI

74 

Lin SC, Liao YC, Chen PM, Yang YY, Wang YH, Tung SL, Chuang CM, Sung YW, Jang TH, Chuang SE, et al: Periostin promotes ovarian cancer metastasis by enhancing M2 macrophages and cancer-associated fibroblasts via integrin-mediated NF-κB and TGF-β2 signaling. J Biomed Sci. 29:1092022. View Article : Google Scholar

75 

Cheon DJ, Tong Y, Sim MS, Dering J, Berel D, Cui X, Lester J, Beach JA, Tighiouart M, Walts AE, et al: A Collagen-remodeling gene signature regulated by TGF-β signaling is associated with metastasis and poor survival in serous ovarian cancer. Clin Cancer Res. 20:711–723. 2014. View Article : Google Scholar

76 

Park SY, Piao Y, Jeong KJ, Dong J and de Groot JF: Periostin (POSTN) Regulates Tumor Resistance to Antiangiogenic Therapy in Glioma Models. Mol Cancer Ther. 15:2187–2197. 2016. View Article : Google Scholar : PubMed/NCBI

77 

Chen L, Tian X, Gong W, Sun B, Li G, Liu D, Guo P, He Y, Chen Z, Xia Y, et al: Periostin mediates epithelial-mesenchymal transition through the MAPK/ERK pathway in hepatoblastoma. Cancer Biol Med. 16:89–100. 2019. View Article : Google Scholar : PubMed/NCBI

78 

Tian Y, Choi CH, Li QK, Rahmatpanah FB, Chen X, Kim SR, Veltri R, Chia D, Zhang Z, Mercola D and Zhang H: Overexpression of periostin in stroma positively associated with aggressive prostate cancer. PLoS One. 10:e01215022015. View Article : Google Scholar : PubMed/NCBI

79 

Pauley KM and Chan EK: MicroRNAs and their emerging roles in immunology. Ann N Y Acad Sci. 1143:226–239. 2008. View Article : Google Scholar : PubMed/NCBI

80 

Song Y, Sun J, Xu Y, Liu J, Gao P, Chen X, Zhao J and Wang Z: Microarray analysis of long non-coding RNAs related to microRNA-148b in gastric cancer. Neoplasma. 64:199–208. 2017. View Article : Google Scholar : PubMed/NCBI

81 

Hu WW, Chen PC, Chen JM, Wu YM, Liu PY, Lu CH, Lin YF, Tang CH and Chao CC: Periostin promotes epithelial-mesenchymal transition via the MAPK/miR-381 axis in lung cancer. Oncotarget. 8:62248–62260. 2017. View Article : Google Scholar : PubMed/NCBI

82 

Hu J, Tan P, Ishihara M, Bayley NA, Schokrpur S, Reynoso JG, Zhang Y, Lim RJ, Dumitras C, Yang L, et al: Tumor heterogeneity in VHL drives metastasis in clear cell renal cell carcinoma. Signal Transduct Target Ther. 8:1552023. View Article : Google Scholar : PubMed/NCBI

83 

Ueki A, Komura M, Koshino A, Wang C, Nagao K, Homochi M, Tsukada Y, Ebi M, Ogasawara N, Tsuzuki T, et al: Stromal POSTN enhances motility of both cancer and stromal cells and predicts poor survival in colorectal cancer. Cancers (Basel). 15:6062023. View Article : Google Scholar : PubMed/NCBI

84 

Miyako S, Koma YI, Nakanishi T, Tsukamoto S, Yamanaka K, Ishihara N, Azumi Y, Urakami S, Shimizu M, Kodama T, et al: Periostin in Cancer-associated fibroblasts promotes esophageal squamous cell carcinoma progression by enhancing cancer and stromal cell migration. Am J Pathol. 194:828–848. 2024. View Article : Google Scholar : PubMed/NCBI

85 

Kim CJ, Sakamoto K, Tambe Y and Inoue H: Opposite regulation of epithelial-to-mesenchymal transition and cell invasiveness by periostin between prostate and bladder cancer cells. Int J Oncol. 38:1759–1766. 2011.PubMed/NCBI

86 

Ribatti D, Nico B, Crivellato E and Vacca A: The structure of the vascular network of tumors. Cancer Lett. 248:18–23. 2007. View Article : Google Scholar

87 

Ribatti D, Vacca A and Dammacco F: New non-angiogenesis dependent pathways for tumour growth. Eur J Cancer. 39:1835–1841. 2003. View Article : Google Scholar : PubMed/NCBI

88 

Baril P, Gangeswaran R, Mahon PC, Caulee K, Kocher HM, Harada T, Zhu M, Kalthoff H, Crnogorac-Jurcevic T and Lemoine NR: Periostin promotes invasiveness and resistance of pancreatic cancer cells to hypoxia-induced cell death: Role of the beta4 integrin and the PI3k pathway. Oncogene. 26:2082–2094. 2007. View Article : Google Scholar

89 

Bao S, Ouyang G, Bai X, Huang Z, Ma C, Liu M, Shao R, Anderson RM, Rich JN and Wang XF: Periostin potently promotes metastatic growth of colon cancer by augmenting cell survival via the Akt/PKB pathway. Cancer Cell. 5:329–339. 2004. View Article : Google Scholar : PubMed/NCBI

90 

Huang Z, Byrd O, Tan S, Hu K, Knight B, Lo G, Taylor L, Wu Y, Berchuck A and Murphy SK: Periostin facilitates ovarian cancer recurrence by enhancing cancer stemness. Sci Rep. 13:213822023. View Article : Google Scholar : PubMed/NCBI

91 

Mariani A, Wang C, Oberg AL, Riska SM, Torres M, Kumka J, Multinu F, Sagar G, Roy D and Jung DB: Genes associated with bowel metastases in ovarian cancer. Gynecol Oncol. 154:495–504. 2019. View Article : Google Scholar : PubMed/NCBI

92 

Lozneanu L, Caruntu ID, Amalinei C, Moscalu M, Gafton B, Marinca MV, Rusu A, Balan R and Giusca SE: Periostin in ovarian carcinoma: From heterogeneity to prognostic value. Folia Histochem Cytobiol. 61:1–16. 2023. View Article : Google Scholar : PubMed/NCBI

93 

Gillot L, Lebeau A, Baudin L, Pottier C, Louis T, Durré T, Longuespée R, Mazzucchelli G, Nizet C, Blacher S, et al: Periostin in lymph node pre-metastatic niches governs lymphatic endothelial cell functions and metastatic colonization. Cell Mol Life Sci. 79:2952022. View Article : Google Scholar : PubMed/NCBI

94 

Kudo Y, Iizuka S, Yoshida M, Nguyen PT, Siriwardena SB, Tsunematsu T, Ohbayashi M, Ando T, Hatakeyama D, Shibata T, et al: Periostin directly and indirectly promotes tumor lymphangiogenesis of head and neck cancer. PLoS One. 7:e444882012. View Article : Google Scholar : PubMed/NCBI

95 

Pistritto G, Trisciuoglio D, Ceci C, Garufi A and D'Orazi G: Apoptosis as anticancer mechanism: Function and dysfunction of its modulators and targeted therapeutic strategies. Aging (Albany NY). 8:603–619. 2016. View Article : Google Scholar : PubMed/NCBI

96 

Vaghari-Tabari M, Ferns GA, Qujeq D, Andevari AN, Sabahi Z and Moein S: Signaling, metabolism, and cancer: An important relationship for therapeutic intervention. J Cell Physiol. 236:5512–5532. 2021. View Article : Google Scholar : PubMed/NCBI

97 

Morana O, Wood W and Gregory CD: The apoptosis paradox in cancer. Int J Mol Sci. 23:13282022. View Article : Google Scholar : PubMed/NCBI

98 

Gao L, Shay C and Teng Y: Cell death shapes cancer immunity: Spotlighting PANoptosis. J Exp Clin Cancer Res. 43:1682024. View Article : Google Scholar : PubMed/NCBI

99 

Ouyang G, Liu M, Ruan K, Song G, Mao Y and Bao S: Upregulated expression of periostin by hypoxia in non-small-cell lung cancer cells promotes cell survival via the Akt/PKB pathway. Cancer Lett. 281:213–219. 2009. View Article : Google Scholar : PubMed/NCBI

100 

Erkan M, Reiser-Erkan C, Michalski CW, Deucker S, Sauliunaite D, Streit S, Esposito I, Friess H and Kleeff J: Cancer-stellate cell interactions perpetuate the Hypoxia-fibrosis cycle in pancreatic ductal adenocarcinoma. Neoplasia. 11:497–508. 2009. View Article : Google Scholar : PubMed/NCBI

101 

Zhou W, Ke SQ, Huang Z, Flavahan W, Fang X, Paul J, Wu L, Sloan AE, McLendon RE, Li X, et al: Periostin secreted by glioblastoma stem cells recruits M2 Tumour-associated macrophages and promotes malignant growth. Nat Cell Biol. 17:170–182. 2015. View Article : Google Scholar : PubMed/NCBI

102 

Wang H, Liang Y, Liu Z, Zhang R, Chao J, Wang M, Liu M, Qiao L, Xuan Z, Zhao H and Lu L: POSTN+ cancer-associated fibroblasts determine the efficacy of immunotherapy in hepatocellular carcinoma. J Immunother Cancer. 12:e0087212024. View Article : Google Scholar :

103 

Jin X, Deng Q, Ye S, Liu S, Fu Y, Liu Y, Wu G, Ouyang G and Wu T: Cancer-associated fibroblast-derived periostin promotes papillary thyroid tumor growth through integrin-FAK-STAT3 signaling. Theranostics. 14:3014–3028. 2024. View Article : Google Scholar : PubMed/NCBI

104 

Muñoz-Espín D and Serrano M: Cellular senescence: From physiology to pathology. Nat Rev Mol Cell Biol. 15:482–496. 2014. View Article : Google Scholar : PubMed/NCBI

105 

McHugh D, Durán I and Gil J: Senescence as a therapeutic target in cancer and age-related diseases. Nat Rev Drug Discov. 24:57–71. 2025. View Article : Google Scholar

106 

Hernandez-Segura A, Nehme J and Demaria M: Hallmarks of cellular senescence. Trends Cell Biol. 28:436–453. 2018. View Article : Google Scholar : PubMed/NCBI

107 

Calcinotto A, Kohli J, Zagato E, Pellegrini L, Demaria M and Alimonti A: Cellular senescence: Aging, Cancer, and Injury. Physiol Rev. 99:1047–1078. 2019. View Article : Google Scholar : PubMed/NCBI

108 

de Magalhães JP: Cellular senescence in normal physiology. Science. 384:1300–1301. 2024. View Article : Google Scholar : PubMed/NCBI

109 

Fane M and Weeraratna AT: How the ageing microenvironment influences tumour progression. Nat Rev Cancer. 20:89–106. 2020. View Article : Google Scholar :

110 

D'Ambrosio M and Gil J: Reshaping of the tumor microenvironment by cellular senescence: An opportunity for senotherapies. Dev Cell. 58:1007–1021. 2023. View Article : Google Scholar : PubMed/NCBI

111 

Zhu D, Chen S, Sheng P, Wang Z, Li Y and Kang X: POSTN promotes nucleus pulposus cell senescence and extracellular matrix metabolism via activing Wnt/β-catenin and NF-κB signal pathway in intervertebral disc degeneration. Cell Signal. 121:1112772024. View Article : Google Scholar

112 

Li Q, Liu X and Wei J: Ageing related periostin expression increase from cardiac fibroblasts promotes cardiomyocytes senescent. Biochem Biophys Res Commun. 452:497–502. 2014. View Article : Google Scholar : PubMed/NCBI

113 

An JN, Kim H, Kim EN, Cho A, Cho Y, Choi YW, Kim JH, Yang SH, Choi BS, Lim CS, et al: Effects of periostin deficiency on kidney aging and lipid metabolism. Aging (Albany NY). 13:22649–22665. 2021. View Article : Google Scholar : PubMed/NCBI

114 

Huang YW, Chiang MF, Ho CS, Hung PL, Hsu MH, Lee TH, Chu LJ, Liu H, Tang P and Victor Ng W: A transcriptome study of progeroid neurocutaneous syndrome reveals POSTN As a new element in proline metabolic disorder. Aging Dis. 9:1043–1057. 2018. View Article : Google Scholar : PubMed/NCBI

115 

Sun L, Zhang H and Gao P: Metabolic reprogramming and epigenetic modifications on the path to cancer. Protein Cell. 13:877–919. 2022. View Article : Google Scholar :

116 

Halperin C, Hey J, Weichenhan D, Stein Y, Mayer S, Lutsik P, Plass C and Scherz-Shouval R: Global DNA methylation analysis of Cancer-associated fibroblasts reveals extensive epigenetic rewiring linked with RUNX1 upregulation in breast cancer stroma. Cancer Res. 82:4139–4152. 2022. View Article : Google Scholar : PubMed/NCBI

117 

Kehrberg RJ, Bhyravbhatla N, Batra SK and Kumar S: Epigenetic regulation of cancer-associated fibroblast heterogeneity. Biochim Biophys Acta Rev Cancer. 1878:1889012023. View Article : Google Scholar : PubMed/NCBI

118 

Albrengues J, Bertero T, Grasset E, Bonan S, Maiel M, Bourget I, Philippe C, Herraiz Serrano C, Benamar S, Croce O, et al: Epigenetic switch drives the conversion of fibroblasts into proinvasive cancer-associated fibroblasts. Nat Commun. 6:102042015. View Article : Google Scholar : PubMed/NCBI

119 

Becker LM, O'Connell JT, Vo AP, Cain MP, Tampe D, Bizarro L, Sugimoto H, McGow AK, Asara JM, Lovisa S, et al: Epigenetic reprogramming of Cancer-associated fibroblasts deregulates glucose metabolism and facilitates progression of breast cancer. Cell Rep. 31:1077012020. View Article : Google Scholar : PubMed/NCBI

120 

Mishra R, Haldar S, Placencio V, Madhav A, Rohena-Rivera K, Agarwal P, Duong F, Angara B, Tripathi M, Liu Z, et al: Stromal epigenetic alterations drive metabolic and neuroendocrine prostate cancer reprogramming. J Clin Invest. 128:4472–4484. 2018. View Article : Google Scholar : PubMed/NCBI

121 

Yang M, Wang B, Hou W, Zeng H, He W, Zhang XK, Yan D, Yu H, Huang L, Pei L, et al: NAD+ metabolism enzyme NNMT in cancer-associated fibroblasts drives tumor progression and resistance to immunotherapy by modulating macrophages in urothelial bladder cancer. J Immunother Cancer. 12:e0092812024. View Article : Google Scholar :

122 

Han B, Yang X, Zhang P, Zhang Y, Tu Y, He Z, Li Y, Yuan J, Dong Y, Hosseini DK, et al: DNA methylation biomarkers for nasopharyngeal carcinoma. PLoS One. 15:e02305242020. View Article : Google Scholar : PubMed/NCBI

123 

Tang Y, Qing C, Wang J and Zeng Z: DNA Methylation-based diagnostic and prognostic biomarkers for glioblastoma. Cell Transplant. 29:9636897209332412020. View Article : Google Scholar : PubMed/NCBI

124 

Nabi-Afjadi M, Farzam F, Soltani S, Golestani N, Asl SS, Aziziyan F, Zalpoor H, Ghasemi F, Dabirmanesh B and Khajeh K: The association between periostin and tumor microenvironment: A promising cancer prognostic biomarker and therapeutic target to combat tumor progression and chemoresistance. Cancer Cell Int. 25:3202025. View Article : Google Scholar : PubMed/NCBI

125 

Gerarduzzi C, Hartmann U, Leask A and Drobetsky E: The matrix revolution: Matricellular proteins and restructuring of the cancer microenvironment. Cancer Res. 80:2705–2717. 2020. View Article : Google Scholar : PubMed/NCBI

126 

Li Z, Pai R, Gupta S, Currenti J, Guo W, Di Bartolomeo A, Feng H, Zhang Z, Li Z, Liu L, et al: Presence of Onco-fetal neighborhoods in hepatocellular carcinoma is associated with relapse and response to immunotherapy. Nat Cancer. 5:167–186. 2024. View Article : Google Scholar : PubMed/NCBI

127 

Anand P, Kunnumakkara AB, Sundaram C, Harikumar KB, Tharakan ST, Lai OS, Sung B and Aggarwal BB: Cancer is a preventable disease that requires major lifestyle changes. Pharm Res. 25:2097–2116. 2008. View Article : Google Scholar : PubMed/NCBI

128 

Lu P, Weaver VM and Werb Z: The extracellular matrix: A dynamic niche in cancer progression. J Cell Biol. 196:395–406. 2012. View Article : Google Scholar : PubMed/NCBI

129 

Kudo A and Kii I: Periostin function in communication with extracellular matrices. J Cell Commun Signal. 12:301–308. 2018. View Article : Google Scholar :

130 

Wang Z and Ouyang G: Periostin: A bridge between cancer stem cells and their metastatic niche. Cell Stem Cell. 10:111–112. 2012. View Article : Google Scholar : PubMed/NCBI

131 

Celià-Terrassa T and Jolly MK: Cancer stem cells and Epithelial-to-mesenchymal transition in cancer metastasis. Cold Spring Harb Perspect Med. 10:a0369052020. View Article : Google Scholar

132 

Plaks V, Kong N and Werb Z: The cancer stem cell niche: How essential is the niche in regulating stemness of tumor cells? Cell Stem Cell. 16:225–238. 2015. View Article : Google Scholar : PubMed/NCBI

133 

Babaei G, Aziz SG and Jaghi NZZ: EMT cancer stem cells and autophagy; The three main axes of metastasis. Biomed Pharmacother. 133:1109092021. View Article : Google Scholar

134 

Malanchi I, Santamaria-Martínez A, Susanto E, Peng H, Lehr HA, Delaloye JF and Huelsken J: Interactions between cancer stem cells and their niche govern metastatic colonization. Nature. 481:85–89. 2011. View Article : Google Scholar : PubMed/NCBI

135 

Costa A, Kieffer Y, Scholer-Dahirel A, Pelon F, Bourachot B, Cardon M, Sirven P, Magagna I, Fuhrmann L, Bernard C, et al: Fibroblast heterogeneity and immunosuppressive environment in human breast cancer. Cancer Cell. 33:463–479.e10. 2018. View Article : Google Scholar : PubMed/NCBI

136 

Wei WF, Chen XJ, Liang LJ, Yu L, Wu XG, Zhou CF, Wang ZC, Fan LS, Hu Z, Liang L and Wang W: Periostin+ cancer-associated fibroblasts promote lymph node metastasis by impairing the lymphatic endothelial barriers in cervical squamous cell carcinoma. Mol Oncol. 15:210–227. 2021. View Article : Google Scholar

137 

Bhattacharjee S, Hamberger F, Ravichandra A, Miller M, Nair A, Affo S, Filliol A, Chin L, Savage TM, Yin D, et al: Tumor restriction by type I collagen opposes Tumor-promoting effects of Cancer-associated fibroblasts. J Clin Invest. 131:e1469872021. View Article : Google Scholar : PubMed/NCBI

138 

Han CC, Liu TY and Yin R: Biomarkers for Cancer-associated fibroblasts. Biomark Res. 8:642020. View Article : Google Scholar : PubMed/NCBI

139 

Oskarsson T and Massagué J: Extracellular matrix players in metastatic niches. EMBO J. 31:254–256. 2012. View Article : Google Scholar :

140 

Lambert AW, Wong CK, Ozturk S, Papageorgis P, Raghunathan R, Alekseyev Y, Gower AC, Reinhard BM, Abdolmaleky HM and Thiagalingam S: Tumor Cell-derived periostin regulates cytokines that maintain breast cancer stem cells. Mol Cancer Res. 14:103–113. 2016. View Article : Google Scholar :

141 

Zhang Q, Wang C and Cliby WA: Cancer-associated stroma significantly contributes to the mesenchymal subtype signature of serous ovarian cancer. Gynecol Oncol. 152:368–374. 2019. View Article : Google Scholar :

142 

Vermeulen L, De Sousa EMF, van der Heijden M, Cameron K, de Jong JH, Borovski T, Tuynman JB, Todaro M, Merz C, Rodermond H, et al: Wnt activity defines colon cancer stem cells and is regulated by the microenvironment. Nat Cell Biol. 12:468–476. 2010. View Article : Google Scholar : PubMed/NCBI

143 

Li P, Yang R and Gao WQ: Contributions of Epithelial-mesenchymal transition and cancer stem cells to the development of castration resistance of prostate cancer. Mol Cancer. 13:552014. View Article : Google Scholar : PubMed/NCBI

144 

Chen G, Wang Y, Zhao X, Xie XZ, Zhao JG, Deng T, Chen ZY, Chen HB, Tong YF, Yang Z, et al: A positive feedback loop between Periostin and TGFβ1 induces and maintains the stemness of hepatocellular carcinoma cells via AP-2α activation. J Exp Clin Cancer Res. 40:2182021. View Article : Google Scholar

145 

Nowosad A, Marine JC and Karras P: Perivascular niches: Critical hubs in cancer evolution. Trends Cancer. 9:897–910. 2023. View Article : Google Scholar : PubMed/NCBI

146 

Liu Z, Chen J, Ren Y, Liu S, Ba Y, Zuo A, Luo P, Cheng Q, Xu H and Han X: Multi-stage mechanisms of tumor metastasis and therapeutic strategies. Signal Transduct Target Ther. 9:2702024. View Article : Google Scholar : PubMed/NCBI

147 

Erez N: Cancer: Angiogenic awakening. Nature. 500:37–38. 2013. View Article : Google Scholar : PubMed/NCBI

148 

Weis SM and Cheresh DA: A wake-up call for hibernating tumour cells. Nat Cell Biol. 15:721–723. 2013. View Article : Google Scholar : PubMed/NCBI

149 

Ghajar CM, Peinado H, Mori H, Matei IR, Evason KJ, Brazier H, Almeida D, Koller A, Hajjar KA, Stainier DY, et al: The perivascular niche regulates breast tumour dormancy. Nat Cell Biol. 15:807–817. 2013. View Article : Google Scholar : PubMed/NCBI

150 

Lewis CE, Harney AS and Pollard JW: The multifaceted role of perivascular macrophages in tumors. Cancer Cell. 30:3652016. View Article : Google Scholar : PubMed/NCBI

151 

Pietras A, Katz AM, Ekström EJ, Wee B, Halliday JJ, Pitter KL, Werbeck JL, Amankulor NM, Huse JT and Holland EC: Osteopontin-CD44 signaling in the glioma perivascular niche enhances cancer stem cell phenotypes and promotes aggressive tumor growth. Cell Stem Cell. 14:357–369. 2014. View Article : Google Scholar : PubMed/NCBI

152 

Shi Y, Ping YF, Zhang X and Bian XW: Hostile takeover: Glioma stem cells recruit TAMs to support tumor progression. Cell Stem Cell. 16:219–220. 2015. View Article : Google Scholar : PubMed/NCBI

153 

Liu Y and Cao X: Characteristics and Significance of the Pre-metastatic Niche. Cancer Cell. 30:668–681. 2016. View Article : Google Scholar : PubMed/NCBI

154 

Li Y, Zheng Y, Tan X, Du Y, Wei Y and Liu S: Extracellular vesicle-mediated pre-metastatic niche formation via altering host microenvironments. Front Immunol. 15:13673732024. View Article : Google Scholar : PubMed/NCBI

155 

Peinado H, Zhang H, Matei IR, Costa-Silva B, Hoshino A, Rodrigues G, Psaila B, Kaplan RN, Bromberg JF, Kang Y, et al: Pre-metastatic niches: Organ-specific homes for metastases. Nat Rev Cancer. 17:302–317. 2017. View Article : Google Scholar : PubMed/NCBI

156 

Kong J, Tian H, Zhang F, Zhang Z, Li J, Liu X, Li X, Liu J, Li X, Jin D, et al: Extracellular vesicles of carcinoma-associated fibroblasts creates a pre-metastatic niche in the lung through activating fibroblasts. Mol Cancer. 18:1752019. View Article : Google Scholar : PubMed/NCBI

157 

Wang X, Jian Q, Zhang Z, Gu J, Wang X and Wang Y: Effect of tumor-derived extracellular vesicle-shuttled lncRNA MALAT1 on proliferation, invasion and metastasis of triple-negative breast cancer by regulating macrophage M2 polarization via the POSTN/Hippo/YAP axis. Transl Oncol. 49:1020762024. View Article : Google Scholar : PubMed/NCBI

158 

Ji Q, Zhou L, Sui H, Yang L, Wu X, Song Q, Jia R, Li R, Sun J, Wang Z, et al: Primary tumors release ITGBL1-rich extracellular vesicles to promote distal metastatic tumor growth through fibroblast-niche formation. Nat Commun. 11:12112020. View Article : Google Scholar : PubMed/NCBI

159 

Pitt JM, Marabelle A, Eggermont A, Soria JC, Kroemer G and Zitvogel L: Targeting the tumor microenvironment: Removing obstruction to anticancer immune responses and immunotherapy. Ann Oncol. 27:1482–1492. 2016. View Article : Google Scholar : PubMed/NCBI

160 

Fukuda K, Sugihara E, Ohta S, Izuhara K, Funakoshi T, Amagai M and Saya H: Periostin is a key niche component for wound metastasis of melanoma. PLoS One. 10:e01297042015. View Article : Google Scholar : PubMed/NCBI

161 

Wang Y, Li Q, Yang X, Guo H, Ren T, Zhang T, Ghadakpour P and Ren F: Exosome-mediated communication in thyroid cancer: Implications for prognosis and therapeutic targets. Biochem Genet. 63:2673–2697. 2025. View Article : Google Scholar

162 

Mouw JK, Ou G and Weaver VM: Extracellular matrix assembly: A multiscale deconstruction. Nat Rev Mol Cell Biol. 15:771–785. 2014. View Article : Google Scholar : PubMed/NCBI

163 

Boulter L, Bullock E, Mabruk Z and Brunton VG: The fibrotic and immune microenvironments as targetable drivers of metastasis. Br J Cancer. 124:27–36. 2021. View Article : Google Scholar :

164 

Sonnenberg-Riethmacher E, Miehe M and Riethmacher D: Periostin in allergy and inflammation. Front Immunol. 12:7221702021. View Article : Google Scholar : PubMed/NCBI

165 

Tomasek JJ, Gabbiani G, Hinz B, Chaponnier C and Brown RA: Myofibroblasts and mechano-regulation of connective tissue remodelling. Nat Rev Mol Cell Biol. 3:349–363. 2002. View Article : Google Scholar : PubMed/NCBI

166 

Xu X, Chang W, Yuan J, Han X, Tan X, Ding Y, Luo Y, Cai H, Liu Y, Gao X, et al: Periostin expression in intra-tumoral stromal cells is prognostic and predictive for colorectal carcinoma via creating a cancer-supportive niche. Oncotarget. 7:798–813. 2016. View Article : Google Scholar :

167 

Ashley SL, Wilke CA, Kim KK and Moore BB: Periostin regulates fibrocyte function to promote myofibroblast differentiation and lung fibrosis. Mucosal Immunol. 10:341–351. 2017. View Article : Google Scholar :

168 

Kiesler ZG, Hunter MI, Balboula AZ and Patterson AL: Periostin's role in uterine leiomyoma development: A Mini-review on the potential periostin poses as a pharmacological intervention for uterine leiomyoma. Arch Gynecol Obstet. 309:1825–1831. 2024. View Article : Google Scholar : PubMed/NCBI

169 

Jamaluddin MFB, Ko YA, Kumar M, Brown Y, Bajwa P, Nagendra PB, Skerrett-Byrne DA, Hondermarck H, Baker MA, Dun MD, et al: Proteomic profiling of human uterine fibroids reveals upregulation of the extracellular matrix protein periostin. Endocrinology. 159:1106–1118. 2018. View Article : Google Scholar

170 

Nielsen SR, Quaranta V, Linford A, Emeagi P, Rainer C, Santos A, Ireland L, Sakai T, Sakai K, Kim YS, et al: Macrophage-secreted granulin supports pancreatic cancer metastasis by inducing liver fibrosis. Nat Cell Biol. 18:549–560. 2016. View Article : Google Scholar : PubMed/NCBI

171 

Clézardin P, Coleman R, Puppo M, Ottewell P, Bonnelye E, Paycha F, Confavreux CB and Holen I: Bone metastasis: Mechanisms, therapies, and biomarkers. Physiol Rev. 101:797–855. 2021. View Article : Google Scholar

172 

Jackett KN, Browne AT, Aber ER, Clements M and Kaplan RN: How the bone microenvironment shapes the pre-metastatic niche and metastasis. Nat Cancer. 5:1800–1814. 2024. View Article : Google Scholar : PubMed/NCBI

173 

Muscarella AM, Aguirre S, Hao X, Waldvogel SM and Zhang XH: Exploiting bone niches: Progression of disseminated tumor cells to metastasis. J Clin Invest. 131:e1437642021. View Article : Google Scholar : PubMed/NCBI

174 

Baccin C, Al-Sabah J, Velten L, Helbling PM, Grünschläger F, Hernández-Malmierca P, Nombela-Arrieta C, Steinmetz LM, Trumpp A and Haas S: Combined single-cell and spatial transcriptomics reveal the molecular, cellular and spatial bone marrow niche organization. Nat Cell Biol. 22:38–48. 2020. View Article : Google Scholar

175 

Chen F, Han Y and Kang Y: Bone marrow niches in the regulation of bone metastasis. Br J Cancer. 124:1912–1920. 2021. View Article : Google Scholar : PubMed/NCBI

176 

Shafat MS, Gnaneswaran B, Bowles KM and Rushworth SA: The bone marrow microenvironment-Home of the leukemic blasts. Blood Rev. 31:277–286. 2017. View Article : Google Scholar : PubMed/NCBI

177 

Weilbaecher KN, Guise TA and McCauley LK: Cancer to bone: A fatal attraction. Nat Rev Cancer. 11:411–425. 2011. View Article : Google Scholar : PubMed/NCBI

178 

Contié S, Voorzanger-Rousselot N, Litvin J, Clézardin P and Garnero P: Increased expression and serum levels of the stromal cell-secreted protein periostin in breast cancer bone metastases. Int J Cancer. 128:352–360. 2011. View Article : Google Scholar

179 

Gineyts E, Bonnet N, Bertholon C, Millet M, Pagnon-Minot A, Borel O, Geraci S, Bonnelye E, Croset M, Suhail A, et al: The C-terminal intact forms of periostin (iPTN) are surrogate markers for osteolytic lesions in experimental breast cancer bone metastasis. Calcif Tissue Int. 103:567–580. 2018. View Article : Google Scholar : PubMed/NCBI

180 

Liu C, Feng X, Wang B, Wang X, Wang C, Yu M, Cao G and Wang H: Bone marrow mesenchymal stem cells promote head and neck cancer progression through Periostin-mediated phosphoinositide 3-kinase/Akt/mammalian target of rapamycin. Cancer Sci. 109:688–698. 2018. View Article : Google Scholar :

181 

Ling W, Johnson SK, Mehdi SJ, Alapat DV, Bauer M, Zangari M, Schinke C, Thanendrarajan S, van Rhee F and Yaccoby S: EDNRA-expressing mesenchymal cells are expanded in myeloma interstitial bone marrow and associated with disease progression. Cancers (Basel). 15:45192023. View Article : Google Scholar : PubMed/NCBI

182 

Terpos E, Christoulas D, Kastritis E, Bagratuni T, Gavriatopoulou M, Roussou M, Papatheodorou A, Eleutherakis-Papaiakovou E, Kanellias N, Liakou C, et al: High levels of periostin correlate with increased fracture rate, diffuse MRI pattern, abnormal bone remodeling and advanced disease stage in patients with newly diagnosed symptomatic multiple myeloma. Blood Cancer J. 6:e4822016. View Article : Google Scholar : PubMed/NCBI

183 

Sun CY, Mi YY, Ge SY, Hu QF, Xu K, Guo YJ, Tan YF, Zhang Y, Zhong F and Xia GW: Tumor- and Osteoblast-derived periostin in prostate cancer bone metastases. Front Oncol. 11:7957122021. View Article : Google Scholar

184 

Ma Z, Zhao X, Deng M, Huang Z, Wang J, Wu Y, Cui D, Liu Y, Liu R and Ouyang G: Bone marrow mesenchymal stromal cell-derived periostin promotes B-ALL progression by modulating CCL2 in leukemia cells. Cell Rep. 26:1533–1543.e4. 2019. View Article : Google Scholar : PubMed/NCBI

185 

Izuhara K, Nunomura S, Nanri Y, Ogawa M, Ono J, Mitamura Y and Yoshihara T: Periostin in inflammation and allergy. Cell Mol Life Sci. 74:4293–4303. 2017. View Article : Google Scholar : PubMed/NCBI

186 

Kii I: Practical application of periostin as a biomarker for pathological conditions. Adv Exp Med Biol. 1132:195–204. 2019. View Article : Google Scholar : PubMed/NCBI

187 

Conway SJ, Izuhara K, Kudo Y, Litvin J, Markwald R, Ouyang G, Arron JR, Holweg CT and Kudo A: The role of periostin in tissue remodeling across health and disease. Cell Mol Life Sci. 71:1279–1288. 2014. View Article : Google Scholar :

188 

Lu S, Peng L, Ma F, Chai J, Hua Y, Yang W and Zhang Z: Increased expression of POSTN predicts poor prognosis: A potential therapeutic target for gastric cancer. J Gastrointest Surg. 27:233–249. 2023. View Article : Google Scholar

189 

Chu LJ, Tsai CJ, Chien CC, Hsiao YC, Yu JS, Tsai JY and Yeh TS: Plasma POSTN derived from bile proteome is a promising biomarker for cholangiocarcinoma with efficacy comparable and complementary to CA19.9. United European Gastroenterol J. 13:1155–1170. 2025. View Article : Google Scholar : PubMed/NCBI

190 

Rachner TD, Göbel A, Hoffmann O, Erdmann K, Kasimir-Bauer S, Breining D, Kimmig R, Hofbauer LC and Bittner AK: High serum levels of periostin are associated with a poor survival in breast cancer. Breast Cancer Res Treat. 180:515–524. 2020. View Article : Google Scholar : PubMed/NCBI

191 

Dong D, Zhang L, Jia L, Ji W, Wang Z, Ren L, Niu R and Zhou Y: Identification of serum periostin as a potential diagnostic and prognostic marker for colorectal cancer. Clin Lab. 64:973–981. 2018. View Article : Google Scholar : PubMed/NCBI

192 

Lv Y, Wang W, Jia WD, Sun QK, Huang M, Zhou HC, Xia HH, Liu WB, Chen H, Sun SN and Xu GL: High preoparative levels of serum periostin are associated with poor prognosis in patients with hepatocellular carcinoma after hepatectomy. Eur J Surg Oncol. 39:1129–1135. 2013. View Article : Google Scholar : PubMed/NCBI

193 

Fujimoto K, Kawaguchi T, Nakashima O, Ono J, Ohta S, Kawaguchi A, Tonan T, Ohshima K, Yano H, Hayabuchi N, et al: Periostin, a matrix protein, has potential as a novel serodiagnostic marker for cholangiocarcinoma. Oncol Rep. 25:1211–1216. 2011. View Article : Google Scholar : PubMed/NCBI

194 

Hu F, Shang XF, Wang W, Jiang W, Fang C, Tan D and Zhou HC: High-level expression of periostin is significantly correlated with tumour angiogenesis and poor prognosis in osteosarcoma. Int J Exp Pathol. 97:86–92. 2016. View Article : Google Scholar : PubMed/NCBI

195 

Villani R, Murigneux V, Alexis J, Sim SL, Wagels M, Saunders N, Soyer HP, Parmentier L, Nikolaev S, Fink JL, et al: Subtype-specific analyses reveal infiltrative basal cell carcinomas are highly interactive with their environment. J Invest Dermatol. 141:2380–2390. 2021. View Article : Google Scholar : PubMed/NCBI

196 

Zhao Z, Mak TK, Shi Y, Li K, Huo M and Zhang C: Integrative analysis of Cancer-associated fibroblast signature in gastric cancer. Heliyon. 9:e192172023. View Article : Google Scholar : PubMed/NCBI

197 

Kyutoku M, Taniyama Y, Katsuragi N, Shimizu H, Kunugiza Y, Iekushi K, Koibuchi N, Sanada F, Oshita Y and Morishita R: Role of periostin in cancer progression and metastasis: Inhibition of breast cancer progression and metastasis by anti-periostin antibody in a murine model. Int J Mol Med. 28:181–186. 2011.PubMed/NCBI

198 

Zhu M, Saxton RE, Ramos L, Chang DD, Karlan BY, Gasson JC and Slamon DJ: Neutralizing monoclonal antibody to periostin inhibits ovarian tumor growth and metastasis. Mol Cancer Ther. 10:1500–1508. 2011. View Article : Google Scholar : PubMed/NCBI

199 

Lee YJ, Kim IS, Park SA, Kim Y, Lee JE, Noh DY, Kim KT, Ryu SH and Suh PG: Periostin-binding DNA aptamer inhibits breast cancer growth and metastasis. Mol Ther. 21:1004–1013. 2013. View Article : Google Scholar : PubMed/NCBI

200 

Liu GX, Xi HQ, Sun XY and Wei B: Role of periostin and its antagonist PNDA-3 in gastric cancer metastasis. World J Gastroenterol. 21:2605–2613. 2015. View Article : Google Scholar : PubMed/NCBI

201 

Halin Bergström S, Semenas J, Nordstrand A, Thysell E, Wänman J, Crnalic S, Widmark A, Thellenberg-Karlsson C, Andersson P, Gidlund S, et al: Morphological heterogeneities in prostate cancer bone metastases are related to molecular subtypes and prognosis. Clin Exp Metastasis. 49:422025.

202 

Zeng L, Li L, Liao X, Zhang L, Yin C, Chen X and Sun J: Population-based high-dimensional analyses identify multiple intrinsic characters for cancer vaccines against lung squamous cell carcinoma. Med Oncol. 41:422024. View Article : Google Scholar : PubMed/NCBI

203 

Ratajczak-Wielgomas K, Grzegrzolka J, Piotrowska A, Matkowski R, Wojnar A, Rys J, Ugorski M and Dziegiel P: Expression of periostin in breast cancer cells. Int J Oncol. 51:1300–1310. 2017. View Article : Google Scholar : PubMed/NCBI

204 

Yang T, Deng Z, Pan Z, Qian Y, Yao W and Wang J: Prognostic value of periostin in multiple solid cancers: A systematic review with Meta-analysis. J Cell Physiol. 235:2800–2808. 2020. View Article : Google Scholar

205 

Li M, Ma G, Li X and Zhou Q: The prognostic value of periostin expression in Non-small cell lung cancer: A Meta-analysis. Med Sci Monit. 28:e9368982022. View Article : Google Scholar : PubMed/NCBI

206 

Morra L, Rechsteiner M, Casagrande S, von Teichman A, Schraml P, Moch H and Soltermann A: Characterization of periostin isoform pattern in non-small cell lung cancer. Lung Cancer. 76:183–190. 2012. View Article : Google Scholar

207 

Utispan K, Sonongbua J, Thuwajit P, Chau-In S, Pairojkul C, Wongkham S and Thuwajit C: Periostin activates integrin α5β1 through a PI3K/AKT-dependent pathway in invasion of cholangiocarcinoma. Int J Oncol. 41:1110–1118. 2012. View Article : Google Scholar : PubMed/NCBI

208 

Wu Y, Li S, Yu H, Zhang S, Yan L, Guan X, Xu W, Wang Z, Lv A, Tian X, et al: Integrative Single-cell and spatial transcriptomics analysis reveals ECM-remodeling Cancer-associated Fibroblast-derived POSTN as a key mediator in pancreatic ductal adenocarcinoma progression. Int J Biol Sci. 21:3573–3596. 2025. View Article : Google Scholar : PubMed/NCBI

209 

Xue H, Gao X, Xu S, Zhang J, Guo X, Yan S, Li T, Guo X, Liu Q and Li G: MicroRNA-Let-7f reduces the vasculogenic mimicry of human glioma cells by regulating periostin-dependent migration. Oncol Rep. 35:1771–1777. 2016. View Article : Google Scholar : PubMed/NCBI

210 

Yan S, Han X, Xue H, Zhang P, Guo X, Li T, Guo X, Yuan G, Deng L and Li G: Let-7f inhibits glioma cell proliferation, migration, and invasion by targeting periostin. J Cell Biochem. 116:1680–1692. 2015. View Article : Google Scholar : PubMed/NCBI

211 

Wei T, Wang K, Liu S, Fang Y, Hong Z, Liu Y, Zhang H, Yang C, Ouyang G and Wu T: Periostin deficiency reduces PD-1+ tumor-associated macrophage infiltration and enhances anti-PD-1 efficacy in colorectal cancer. Cell Rep. 42:1120902023. View Article : Google Scholar

212 

Zhang W, Wu FH, Wang J, Wei HZ, Wang TW and Zhang YC: Periostin: A predictable molecule to prognosis and chemotherapy responses of gastrointestinal and hepato-biliary-pancreatic malignant tumors? Neoplasma. 69:491–503. 2022. View Article : Google Scholar : PubMed/NCBI

213 

Ishibashi Y, Mochizuki S, Horiuchi K, Tsujimoto H, Kouzu K, Kishi Y, Okada Y and Ueno H: Periostin derived from cancer-associated fibroblasts promotes esophageal squamous cell carcinoma progression via ADAM17 activation. Biochim Biophys Acta Mol Basis Dis. 1869:1666692023. View Article : Google Scholar : PubMed/NCBI

214 

Mikheev AM, Mikheeva SA, Trister AD, Tokita MJ, Emerson SN, Parada CA, Born DE, Carnemolla B, Frankel S, Kim DH, et al: Periostin is a novel therapeutic target that predicts and regulates glioma malignancy. Neuro Oncol. 17:372–382. 2015. View Article : Google Scholar :

215 

Chen G, Nakamura I, Dhanasekaran R, Iguchi E, Tolosa EJ, Romecin PA, Vera RE, Almada LL, Miamen AG, Chaiteerakij R, et al: Transcriptional induction of periostin by a sulfatase 2-TGFβ1-SMAD signaling axis mediates tumor angiogenesis in hepatocellular carcinoma. Cancer Res. 77:632–645. 2017. View Article : Google Scholar

216 

Shang Y, Liang Y, Zhang B, Wu W, Peng Y, Wang J, Zhang M and Niu C: Periostin-mediated activation of NF-κB signaling promotes tumor progression and chemoresistance in glioblastoma. Sci Rep. 15:139552025. View Article : Google Scholar

217 

Wu J, Li J, Xu H, Qiu N, Huang X and Li H: Periostin drives extracellular matrix degradation, stemness, and chemoresistance by activating the MAPK/ERK signaling pathway in triple-negative breast cancer cells. Lipids Health Dis. 22:1532023. View Article : Google Scholar : PubMed/NCBI

218 

Wang Y, Wang B, Cao W and Xu X: PTX3 activates POSTN and promotes the progression of glioblastoma via the MAPK/ERK signalling axis. Biochem Biophys Res Commun. 703:1496652024. View Article : Google Scholar : PubMed/NCBI

Related Articles

  • Abstract
  • View
  • Download
  • Twitter
Copy and paste a formatted citation
Spandidos Publications style
Xian Y and Wang L: Periostin: A matricellular protein with a multifaceted role in tumorigenesis (Review). Int J Mol Med 56: 229, 2025.
APA
Xian, Y., & Wang, L. (2025). Periostin: A matricellular protein with a multifaceted role in tumorigenesis (Review). International Journal of Molecular Medicine, 56, 229. https://doi.org/10.3892/ijmm.2025.5670
MLA
Xian, Y., Wang, L."Periostin: A matricellular protein with a multifaceted role in tumorigenesis (Review)". International Journal of Molecular Medicine 56.6 (2025): 229.
Chicago
Xian, Y., Wang, L."Periostin: A matricellular protein with a multifaceted role in tumorigenesis (Review)". International Journal of Molecular Medicine 56, no. 6 (2025): 229. https://doi.org/10.3892/ijmm.2025.5670
Copy and paste a formatted citation
x
Spandidos Publications style
Xian Y and Wang L: Periostin: A matricellular protein with a multifaceted role in tumorigenesis (Review). Int J Mol Med 56: 229, 2025.
APA
Xian, Y., & Wang, L. (2025). Periostin: A matricellular protein with a multifaceted role in tumorigenesis (Review). International Journal of Molecular Medicine, 56, 229. https://doi.org/10.3892/ijmm.2025.5670
MLA
Xian, Y., Wang, L."Periostin: A matricellular protein with a multifaceted role in tumorigenesis (Review)". International Journal of Molecular Medicine 56.6 (2025): 229.
Chicago
Xian, Y., Wang, L."Periostin: A matricellular protein with a multifaceted role in tumorigenesis (Review)". International Journal of Molecular Medicine 56, no. 6 (2025): 229. https://doi.org/10.3892/ijmm.2025.5670
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