International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
Cancer is a disease that poses threats to human health and life, and is characterized by aberrant cellular proliferation and the development of malignant tumors. According to data provided by the World Health Organization, millions of individuals worldwide are diagnosed with cancer annually (1,2). The incidence and mortality rates have exhibited an upward trend, and are subject to temporal and regional variations (3). Although advancements in oncology research have been made by scientists and clinicians in areas such as drug discovery, immunotherapy and radiotherapy, effective treatment of cancer is still facing great challenges, such as drug resistance, immune tolerance and severe side-effects (4–6). It is imperative to unravel the mechanisms underlying tumorigenesis and to develop novel intervention strategies.
Osteopontin (OPN) is a multifunctional glycophosphoprotein encoded by the secretory phosphoprotein 1 (SPP1) gene, which is expressed in various types of cells and tissues, including in the bone, dentin, cementum, hypertrophic cartilage, kidney, brain, bone-marrow-derived metrial gland cells, vascular tissues, cytotrophoblasts of the chorionic villus in the uterus and decidua, ganglia of the inner ear, brain cells, specialized epithelia found in mammary, salivary and sweat glands, bile and pancreatic ducts, distal renal tubules and the gut, as well as in activated macrophages and lymphocytes (7,8). OPN was first reported as a marker of transformation of epithelial cells in 1979 (9). Subsequently, more variants of OPN and their corresponding functions have been identified. For example, OPN splicing isoforms in solid tumors have been investigated in breast cancer, mesothelioma and lung cancer (10,11). As a secreted protein, OPN binds with its receptors, such as integrin or CD44, to exert functions, including modulating cell proliferation, adhesion, inflammatory responses, osteogenesis and wound healing (8). The role of OPN in tumorigenesis and development has attracted increasing attention (12,13). Abnormal OPN expression has been observed in different types of cancer such as brain, lung, kidney, liver, bladder and breast cancer (13,14). Diverse hypotheses have been proposed regarding the association between OPN and tumorigenesis; however, to the best of our knowledge, its precise molecular mechanisms remain to be illustrated. Given the pivotal role of OPN in tumorigenesis and progression, OPN-based interventions have been developed and applied in both basic research and clinical settings (15–17).
The present review, after briefly introducing the molecular structure, receptors and physiological functions of OPN, describes how OPN participates in cancer initiation, progression, metastasis and drug resistance. Furthermore, the present review summarizes current OPN-based tumor intervention strategies. The present review also provides perspectives on how to deepen the insights regarding the exact role of OPN in tumorigenesis in order to help develop novel strategies for tumor therapeutics. The present review broadens the knowledge regarding the pathophysiological role of OPN, so that novel OPN-based cancer treatment strategies may be proposed.
OPN was first reported as a transformation marker of malignant cells by Senger et al (9) in 1979. With the deepening of research, more definitions of OPN have been determined, including bone sialoprotein, SPP1 and early T-lymphocyte activation 1 (Eta-1) (8). Based on mRNA transcript variants, OPN can be translated into five subtypes, including OPN-a, OPN-b, OPN-c, OPN4 and OPN5, which can exist in secretory or intracellular forms (10). Therefore, OPN has multiple functions in pathophysiological conditions such as inflammation, cell survival, adhesion, migration, differentiation, apoptosis and bone matrix mineralization (18,19). Comprehensive understanding of the protein structure, receptors and functions of OPN is necessary and crucial in order to further reveal its mechanism involved in tumor genesis and development, and to develop therapeutic strategies.
OPN is encoded by the SPP1 gene located on chromosome 4q22.1 (19). The SPP1 gene contains seven exons, which undergo selective splicing to generate distinct variants. At present, seven exons of SPP1 have been reported. Exon 1 remains untranslated, whereas exons 2–7 contain coding sequences. Exon 2 encodes a signal peptide along with two amino acids constituting the mature protein; exon 3 encodes a serine phosphorylation motif; exon 4 encodes a proline-rich region and transglutaminase site; and exon 5 encodes an additional protein phosphorylation site. Notably, exons 6 and 7 account for >80% of the expressed OPN protein (19). Five isoforms of OPN have been reported, including OPN-a (comprising all exons and consisting of 314 amino acids), OPN-b (lacking exon 5 and comprising 300 amino acids), OPN-c (lacking exon 4 and comprising 287 amino acids), OPN4 (lacking exons 4 and 5, and consisting of 273 amino acids) and OPN5 (containing an additional exon, with a length of 327 amino acids) (10,20). Distinct splicing of the SPP1 gene has been implicated in cancer occurrence, progression and prognosis (19).
Human OPN contains ~314 amino acid residues, with a molecular weight ranging between 41 and 75 kDa, depending on different post-translational modifications (8,21). OPN contains several highly conserved domains, including the aspartate domain, arginine-glycine-aspartic acid (RGD) domain, serine-valine-valine-tyrosine-glutamate-leucine-arginine (SVVYGLR) domain, thrombin cleavage domain, calcium-binding domain and C-terminal heparin-binding domain (22). Given that OPN is a secreted protein, it is present not only in various tissues but also in biological fluids. The secreted OPN is subjected to various post-translational modifications, including glycosylation, phosphorylation and sulfuration, which endows OPN with diversified biological functions (8). Fig. 1 schematically illustrates the structure and domains of OPN.
OPN needs to bind with its receptors to exert its function. The well-characterized receptors of OPN include integrins and CD44 (23,24). Notably, under some conditions, OPN is subjected to protease cleavage to expose its specific domains, which exhibit binding affinity for potential receptors (25).
Integrins are transmembrane heterodimers that recognize various ligands, including OPN and other extracellular matrix proteins and cell surface proteins (26). The RGD sequence of OPN exhibits a strong binding affinity for integrins, including αvβ1, αvβ3, αvβ5, αvβ6, αvβ8, α8β1, α5β1 and αIIbβ3 (27,28). Another integrin-binding domain (SVVYGLR) can only be exposed after thrombin cleavage at Arg168-Ser169 (29). The exposed SVVYGLR interacts with integrin subtypes such as α9β1, α4β1 and α4β7 (25,28,30). Similarly, plasma proteases cleave OPN at Lys154-Ser155, producing an N-terminal OPN fragment that binds more strongly to α5β1 and αvβ3 integrins than the full-length OPN (25). MMP could cleave OPN at various sites such as Gly166-Leu167, Ala201-Tyr202 and Asp210-Leu211, producing a fragment that preferably binds with the α4β1 isoform of integrin (29).
CD44, also known as hyaluronic acid receptor, is expressed in various cell types such as osteocytes, endothelial cells, fibroblasts, epithelial cells and smooth muscle cells (31). The C-terminal fragment heparin-binding site of OPN can directly interact with CD44 variants CD44v3 and CD44v6 (30).
Therefore, OPN, with or without cleavage, interacts with integrins or CD44 to regulate multiple cellular processes, including cell attachment, migration, chemotaxis and immune modulation in diverse types of cells (24).
OPN is expressed in various types of cells and tissues. With the deepening of research, diverse functions of OPN in both physiological and pathological conditions have been revealed (32,33).
OPN is also known as Eta-1 and can trigger the immune response of macrophages, T cells and B cells (34). For example, OPN binds with integrin (αvβ3) of macrophages and reduces the production of anti-inflammatory IL-10, while its binding with CD44 promotes the secretion of proinflammatory IL-12 (23). OPN also influences the migration of immune cells (27,35). Wang and Denhardt (27) reported that OPN served as a type 1 T helper cell cytokine, contributing to mucosal defense against viral pathogens. OPN may regulate inflammatory reactions by stimulating the natural immune response of macrophages and neutrophils (27). OPN could also be secreted by activated T cells to aid in the recruitment and migration of immune cells, particularly macrophages, toward the site of inflammation, resulting in persistence and exacerbation of the inflammatory response (36).
OPN exerts pro-inflammatory effects by activating inflammatory signaling pathways, such as the NF-κB and STAT3 pathways, which leads to the secretion of cytokines, including TNF-α and CD16 (37). Additionally, OPN modulates immune cell functions by enhancing macrophage chemotaxis and phagocytic activity, promoting their polarization toward the pro-inflammatory M1 phenotype, and activating the immune responses of T cells, natural killer cells and dendritic cells (38,39). Furthermore, OPN stimulates tumor cells to secrete IL-17, thereby activating the JNK/c-Jun signaling pathway and contributing to immunosuppressive effects (40). OPN also serves a critical role in mediating immune escape mechanisms, such as enabling tumor cells to evade immune surveillance through the suppression of T cell activity (41). Development of single-cell RNA sequencing (scRNA-seq) has provided a better way to define the context-dependent functions of OPN. Ding et al (42) utilized scRNA-seq and revealed that CD163+ macrophages were enriched in lung tissues of patients with coronavirus disease 2019, and these cells highly expressed OPN. Wang et al (43) used scRNA-seq analysis and revealed that OPN expression was upregulated in menstrual blood cells from patients with endometriosis, which in synergy with inflammatory factors, such as IL-10 and IL-6, promoted local fibrotic processes.
Aberrant OPN expression may induce dysregulation of the immune response, thereby impairing the normal recognition and response to self-antigens and foreign antigens, which contributes to the initiation and progression of autoimmune disorders (44). Improper expression of OPN is associated with the pathogenesis of various autoimmune diseases (such as multiple sclerosis, systemic lupus erythematosus, rheumatoid arthritis and atherosclerosis) and other inflammatory diseases (including cardiovascular diseases, chronic obstructive pulmonary disease, inflammatory bowel disease, liver diseases and asthma) (27).
OPN also participates in osteogenesis, regulating bone cell adhesion and osteoclast function, and promoting matrix mineralization. The strong affinity of OPN for hydroxyapatite leads to its accumulation in the bone (45–47). OPN exerts an effect on skeletal cell proliferation, differentiation and migration, while facilitating calcium phosphate mineralization (48). Given its crucial role in bone metabolism, aberrant OPN expression is associated with various types of bone diseases. In patient with osteolysis, OPN expression is downregulated, whereas in patients with fractures, OPN expression is increased (49).
OPN exhibits an important role in tissue repair and regeneration by facilitating cell migration, proliferation, neoangiogenesis and the formation of fibrous connective tissue (50). For example, OPN has been found to promote fibroblast proliferation during wound healing (50). Furthermore, Wen et al (51) found that, in a model of regenerating liver after partial hepatectomy, OPN promoted the inflammatory response and activated the IL-6/STAT3 pathway to facilitate the proliferation of hepatocytes and the repair of the liver. A previous study has demonstrated that OPN expression could induce vascular intimal thickening and promote neointimal formation following injury, suggesting a role in vascular remodeling (52). Upregulation of OPN leads to tissue fibrosis, including liver and kidney fibrosis, by facilitating the synthesis and deposition of extracellular matrix proteins (53).
OPN also participates in the regulation of cardiovascular function. OPN is upregulated in atherosclerosis and present in atherosclerotic plaques, suggesting its potential relevance (54). Furthermore, it has been demonstrated that OPN induces vascular calcification by facilitating mineral uptake and formation of apatite crystals (55). Speer et al (56) further revealed that phosphorylation of OPN could induce vascular calcification.
In summary, OPN serves diverse and important pathophysiological roles in a context-dependent manner. Elucidating the intricate structure and multifaceted functionality of OPN will consequently facilitate the development of OPN-based diagnostic and therapeutic strategies.
The role and mechanisms of OPN in tumor genesis and development have attracted increased attention (57–60). Increased OPN expression is observed in various malignancies, including breast cancer, leukemia, hepatocellular carcinoma (HCC), bladder cancer, colorectal cancer, melanoma, lung cancer, squamous cell carcinoma of the head and neck, and glioblastoma multiforme (13,61). Numerous studies have demonstrated that the upregulation of OPN is involved in tumorigenesis, chemotherapy resistance, angiogenesis and immunosuppression, and revealing how OPN participates in these processes is essential (62–67).
OPN is involved in cancer pathogenesis. One study has revealed high OPN expression in various types of cancer such as brain tumors, lung cancer, kidney cancer, liver cancer, bladder cancer, breast cancer, esophageal cancer, pancreatic cancer, gastric cancer, colorectal cancer and prostate cancer (13). The precise role and mechanism of OPN in cancer pathogenesis are not fully understood; however, the following aspects have been widely implicated in these.
Tumor cells exhibit anti-apoptosis capacities (68). Sun et al (69) demonstrated that OPN regulated Bcl-2 and Bax expression, thus enhancing the viability of tumor cells. Another group found that OPN could induce Bcl-2 expression and attenuate apoptosis (70). In addition, OPN facilitates tumor progression by regulating tumor surveillance mechanisms and inhibiting tumor cell apoptosis (71). Upregulated OPN interacts with integrin αvβ3 or CD44, facilitating complement factor H cell surface translocalization, and thus, complement-mediated cell death is suppressed (72). Zhou et al (73) found that OPN knockdown enhanced apoptosis and caused cell cycle arrest in leukemia stem cells.
Tumor metastasis is closely associated with the capacity of cell migration and invasion (72,74). Irby et al (75) reported that overexpressed OPN in SW480, HT29 and HCT116 cells bound with its receptors, including integrins (αvβ1, αvβ3 and αvβ5) and CD44, and enhanced colon cancer invasion. OPN has the ability to mediate the adhesion and interaction between tumor cells and the surrounding matrix, thereby augmenting the metastatic potential of tumor cells (76). Kale et al (77) found that OPN, after binding with integrin (α9β1), triggered cyclooxygenase-2 (COX-2) and prostaglandin E2 expression, and thus, promoted tumor growth and metastasis. OPN induces the expression and activation of MMP-2, MMP-3 and urokinase-type plasminogen activator in an integrin-dependent manner, which promotes extracellular matrix degradation and consequent metastasis (78). The capacity of OPN to induce cellular migration and extracellular matrix degradation contributes to the invasive and metastatic potential of cancer (78).
Angiogenesis is a feature of tumors, particularly solid tumors. Neovascularization enhances the supply of nutrients and oxygen, thus promoting tumor growth and metastasis (71,79). OPN is involved in tumor angiogenesis by facilitating the migration and proliferation of vascular endothelial cells, which promotes neovascularization (79). Gupta et al (80) reported that OPN activated MMP-9, which resulted in increased VEGF secretion and promoted the angiogenesis of prostate cancer, while OPN knockdown or αvβ3 mutation reversed these effects. Fukusada et al (81) reported that OPN induced VEGF expression and angiogenesis, which promoted pancreatic ductal adenocarcinoma progression.
Tumor cells are surrounded by the TME, which consists of extracellular matrix, stromal cells, blood vessels, immune cells, fibroblasts and secretory factors (82). A recent study has demonstrated that the interplay between tumor cells and the TME governs tumorigenesis, invasion, metastasis, chemotherapy resistance and the immune response, thereby driving tumor progression and invasion (83). OPN, as a component of the TME, could derive from tumor cells, endothelial cells, fibroblasts and immune cells. OPN exerts an influence on the formation and regulation of the TME (9,84,85). Wei et al (36) found that OPN promoted tumor progression by attracting macrophages and activating T cells in the glioblastoma TME. Butti et al (86) reported that OPN could remodel the TME immune suppression and promoted tumor progression by modulating the polarization of T helper cells, T-regulatory cells and tumor-associated macrophages. Rogers et al (87) reported that myofibroblastic cancer-associated fibroblasts in the TME promoted cancer stem cell proliferation and metastasis, whereas inhibition of OPN in a mouse breast cancer model resulted in diminished tumor stemness. Therefore, destruction of the TME by targeting OPN may effectively inhibit tumor progression.
OPN exhibits distinct effects across various cancer types. For example, OPN inhibits autophagy in colorectal cancer cells, induces breast cancer cell migration and increases angiogenesis in breast cancer, and leads to cell migration and invasion in lung adenocarcinoma (88,89). Further studies elucidating the precise involvement of OPN in cancer pathogenesis are required.
The essential role of OPN in tumorigenesis and development is well-established, and OPN exerts its effects by activating multiple intertwined signaling pathways such as the JNK, Ras/Raf/MEK/ERK, PI3K/Akt, Janus kinase (JAK)/STAT, NF-κB and TIAM Rac1 associated GEF 1/Rac1 pathways (14,57). These pathways regulate cell proliferation, adhesion, diffusion, migration, invasion and epithelial-mesenchymal transition (EMT) of tumor cells, as well as immunosuppression and drug resistance (1). The main pathways regulated by OPN that are involved in tumorigenesis and development are summarized and illustrated in Fig. 2 (90–99).
The JNK signaling pathway has been demonstrated to be activated in various types of cancer such as lymphoma, pancreatic cancer, hepatocellular carcinoma and childhood sarcoma (91). Messex et al (100) found that extraneously added OPN activated the JNK pathway, which accelerated cell proliferation and prostate cancer development. Insua-Rodríguez et al (101) demonstrated that JNK was activated in breast cancer xenograft tumor mouse models, which enhanced OPN expression, resulting in chemotherapy resistance and metastasis. Li et al (102) demonstrated that OPN bound to MB231 breast cancer cells and activated the JNK signaling pathway, which resulted in EMT initiation and cellular migration.
The Ras/Raf/MEK/ERK signaling pathway serves a vital role in promoting cell proliferation, survival and metastasis in various cancer types (103). Yang et al (104) reported that knockdown of OPN in squalene synthase-overexpressing lung cancer cells inhibited their migration and invasion. Chernaya et al (105) found that, in thyroid cancer, activation of the RAS-RAF-MEK pathway led to upregulation of OPN and its receptors (integrin), which in turn increased the metastatic potential of tumor cells. Sun et al (106) adopted RNA interference (RNAi) to reduce OPN expression, which resulted in a decrease in the expression levels of MMP-2 and inhibition of the MEK/ERK pathway, eventually retarding hepatic carcinoma growth and metastasis.
The PI3K/AKT signaling pathway is aberrantly activated in various types of cancer (91). OPN has been found to activate the PI3K/AKT pathway by binding with CD44, consequently promoting cell proliferation and suppressing apoptosis (107). Zhang et al (108) reported that OPN bound with integrin αvβ3 and activated the PI3K/AKT pathway in breast cancer, while knockdown of OPN suppressed breast cancer metastasis. Furthermore, Fu et al (109) revealed that upregulation of OPN facilitated chemoresistance of non-small cell lung cancer (NSCLC), while silencing of OPN inhibited the phosphorylation of AKT and ERK, weakening gefitinib resistance in NSCLC cells.
The JAK/STAT signaling pathway is overactivated in cancer (110). OPN interacts with CD44 and triggers the activation of JAK protein kinase, which induces STAT phosphorylation and consequent enhanced hepatic carcinoma cell proliferation (111). Behera et al (112) demonstrated that OPN activated JAK2/STAT3 signaling and stimulated breast tumor growth in mice. JAK2 inhibitor (AG 490) administration could suppress OPN-induced STAT3 phosphorylation and promote tumor cell apoptosis (112).
The NF-κB signaling pathway has been found to be activated in cancer (104). OPN binding to integrin αvβ3 results in the translocation of NF-κB from the cytoplasm to the nucleus, and promotes tumorigenesis, proliferation, invasion and angiogenesis of HCC (113). Chen et al (114) demonstrated that specific small interfering RNA (siRNA) inhibition of OPN could effectively attenuate NF-κB levels, thereby inhibiting proliferation of esophageal squamous cell carcinoma cells and suppressing tumor formation and metastasis. Saurav et al (115) reported that inhibition of OPN and/or NF-κB signaling could enhance the efficacy of irinotecan, and the combination of OPN and/or an NF-κB inhibitor with irinotecan may enhance the therapeutic efficacy while reducing the development of drug resistance.
The p38 MAPK signaling pathway is a key signal transduction cascade that cells possess in response to environmental stimuli, and has attracted much attention as a promising target for cancer therapy (116,117). Yu et al (118) reported that OPN was highly expressed in head and neck squamous cell carcinoma (HNSCC), and OPN promoted the proliferation and invasion of HNSCC cells by activating the p38-MAPK signaling pathway. Huang et al (66) stated that OPN participated in the activation of the p38 MAPK signaling pathway, thereby promoting cell migration and invasion of colorectal cancer. OPN-conferred chemoresistance is also associated with the p38 MAPK signaling pathway. Pang et al (119) demonstrated that OPN mediated cyclophosphamide resistance in MDA-MB-231 tumor cells by activating p38 MAPK.
Cancer is threatening the life of millions of individuals worldwide, and its therapeutics still face great challenges (120). OPN possesses various functions in the context of cancer, including the enhancement of tumor progression and metastasis, inhibition of tumor cell apoptosis, regulation of the TME, and induction of chemotherapeutic drug resistance. OPN represents a promising therapeutic target for the improved treatment of cancer. RNAi, small-molecule inhibitors, aptamers targeting OPN, and blockade binding of OPN and its receptor are expected to be alternative methods for cancer therapy (14,89). Table I outlines available OPN-based therapeutic strategies.
siRNAs and short hairpin RNAs (shRNAs) can efficiently downregulate the expression of specific genes. Knockdown of OPN with siRNA represents a promising intervention for various types of cancer such as liver tumors, chronic myeloid leukemia, familial adenomatous polyposis and metastatic melanoma, and it is currently undergoing clinical trial (121). Ben-David-Naim et al (122) demonstrated that administration of nanoparticle-encapsulated OPN siRNA effectively reduced OPN expression and suppressed tumor growth in a mouse breast cancer model. Yang et al (123) reported that knockdown of OPN with specific siRNA or shRNA could inhibit breast tumor progression. These findings suggest that downregulation of OPN expression could inhibit tumor cell proliferation, migration, invasion, angiogenesis and other processes. Using siRNA technology to silence OPN offers a plethora of strategic opportunities to develop effective treatment modalities against cancer.
Given their compact size and efficient cell membrane permeability, small-molecule inhibitors targeting specific signaling pathways have attracted increasing attention in disease intervention (23,124). Small-molecule inhibitors toward OPN have been applied for cancer treatment. Sharma et al (125) demonstrated that inhibition of OPN expression with trichostatin A effectively prevented tumor growth and metastasis in a mouse xenograft model. Androstenedione (Andro), a diterpenoid compound isolated from androsacetum, inhibits OPN expression, resulting in cell cycle arrest and apoptosis of breast cancer cells, as well as tumor-endothelial cell interactions (126). Kumar et al (126) further demonstrated that Andro suppressed breast cancer cell proliferation by downregulating c-jun expression and inhibiting PI3K/Akt signaling activation. Parecoxib, a traditional COX-2 inhibitor, has been reported to retard the progression of colorectal cancer by suppressing OPN expression via blockade of the nuclear receptor subfamily 4 group A member 2 and Wnt signaling pathways (17). Matsuura et al (127) demonstrated that simvastatin, a high mobility group-CoA inhibitor, downregulated the expression levels of OPN and αvβ1, αvβ3, α5β1 and α5β3 integrin, accompanied by an enhancement in the invasion of ovarian cancer. Lv et al (128) found that combined administration of curcumin and basilic polysaccharides could effectively suppress cell proliferation and invasion by downregulating OPN and its receptor in ovarian cancer.
Aptamers, as short single-stranded deoxyribonucleic acid or ribonucleic acid molecules, have unique biometric capabilities. They are able to fold to form a three-dimensional conformation and precisely bind to a protein target (129). This process enables the aptamers to bind to specific targets with high selectivity, including proteins, peptides, small carbohydrate molecules and even living cells, showing great potential in biomedical research and diagnostic applications (130). Aptamers exhibit advantages such as high affinity for the target, biochemical stability and low immunogenicity, and could potentially be used as cancer therapeutics (130). Mi et al (15) demonstrated that the treatment of MDA-MB-231 breast cancer cells with OPN aptamers, OPN-R3, resulted in inhibition of cell adhesion, migration and invasion. The authors further demonstrated that OPN-R3 inactivated OPN-associated signaling pathways, including the PI3K, JNK1/2, Src and Akt pathways (15). Furthermore, in vivo, OPN-R3 administration prevented tumor growth in breast cancer xenotransplantation mouse models (131).
OPN needs to bind with its receptors to exert its roles in tumor genesis and progression. Therefore, disrupting the binding of OPN and its receptors represents an alternative strategy for tumor treatment. Rangaswami et al (90) reported that blocking antibodies against αvβ3-integrin effectively suppressed OPN-induced tumor growth and angiogenesis. Furthermore, αvβ3-blocking antibodies have been found to effectively inhibit OPN-mediated activator protein 1 activation in breast cancer cells (132). Ahmed et al (133) showed that administration of CD44 antibody disrupted the interaction between OPN and CD44, consequently restricting OPN-CD44 mediated tumor invasion and metastasis. Blocking the binding of OPN with its receptors offers a novel therapeutic strategy.
OPN needs to be cleaved by specific proteases to expose its receptor binding domain in order to exert its function. Therefore, blocking enzyme cleavage could render OPN inactive. Chiou et al (134) showed that follistatin-like protein 1 directly bound to the uncleaved form of OPN and inhibited the proteolytic activation of OPN, which blocked OPN-integrin/CD44-related signaling pathways and consequent lung cancer metastasis. Thrombin has been demonstrated to participate in OPN hydrolysis, which is essential for effective binding of OPN with its receptors (135). Peraramelli et al (136) reported that a thrombin inhibitor, dabigatran etexilate, retarded B16 melanoma growth by blocking OPN activation. Therefore, inhibiting OPN activation by blocking OPN signaling and proteolytic processing may be a feasible therapeutic approach to stop tumor progression. Current OPN-based interventions are summarized in Table I.
Although OPN is a promising target for cancer therapy, there are still some limitations in practical applications. Gene interference technologies such as RNAi have poor stability and low delivery efficiency, making it difficult to continuously and specifically inhibit OPN expression in vivo (137). Small-molecule inhibitors have several drawbacks, including a short half-life, poor oral bioavailability, drug resistance, multiple side effects and high production costs. Off-target effects occur when a molecule in the body binds non-specifically to molecules other than its intended target, which may lead to unexpected biological effects or adverse reactions (138). Blocking the interaction between OPN and receptors is also challenging because OPN can bind to multiple receptors, and single blocking has limited effects, and may also trigger unknown biological effects. Complete inactivation of OPN may affect its normal physiological functions and cause side effects. Despite the limitations of current strategies, each approach has its unique advantages and applicable scenarios (23). In practical applications, appropriate strategies should be selected based on specific diseases and treatment requirements, and their limitations should be overcome through technological innovation and optimization. For example, the therapeutic efficacy and safety can be effectively improved by optimizing aptamer design, improving delivery systems and enhancing the bioavailability of small-molecule inhibitors (89).
The combination of multiple therapeutic strategies could provide more comprehensive and effective treatments, and a representative combination is OPN inhibitors together with immune checkpoint intervention. Lu et al (139) found that administration of the WD repeat domain 5 protein (WDR5), an inhibitor of OPN, suppressed the growth of orthotopic pancreatic tumors in mice. Furthermore, WDR5 enhanced the efficacy of anti-programmed cell death protein 1 immunotherapy in inhibiting pancreatic tumor growth in mice (139). Zhu et al (140) reported that OPN expression in HCC was positively associated with the programmed death-ligand 1 levels, whereas OPN knockout enhanced the antitumor function of T-helper 1 cells. Future research should focus on mechanism verification and optimization of multi-target combination strategies to achieve more precise antitumor treatment.
OPN, as a secretory protein, is aberrantly expressed in various types of cancer and exhibits a crucial role in both the pathogenesis and progression of cancer. OPN exerts an oncogenesis-promoting effect by stimulating cell proliferation, antagonizing apoptosis, promoting angiogenesis, suppressing immunity and inducing drug resistance. Given its pivotal role, OPN has been regarded as one of the targets for cancer treatment. OPN-based cancer therapeutics based on gene interference, inhibitors and immunoregulation have displayed great potential in research and clinical settings. However, a few aspects deserve to be considered to broaden OPN-based cancer interventions. Firstly, the molecular pathways through which OPN participates in tumorigenesis could be revealed by multiple-omics analysis. Secondly, the dynamic changes of OPNs during cancer development and treatment should be precisely monitored. Thirdly, OPN-based therapeutics should be combined with other treatment strategies. In summary, by elucidating the various roles of OPN in cancer genesis and development, the present review could deepen the understanding of cancer biology and help develop a more effective cancer treatment strategy.
The authors would like to thank Mr. Peng Ye (Shanxi Medical University, Taiyuan, Shanxi 030000, China) and Ms. Shangyu Xie (Shanxi Medical University, Taiyuan, Shanxi 030000, China) for their drafting guidance in preparing the manuscript.
The present study was supported by the Doctoral Fund of Shanxi Medical University (grant no. XD1805), the Doctoral Provincial Research Fund of Shanxi Medical University (grant no. SD1806), the Research Fund of Shanxi Province Health Commission (grant no. 2021060), the Shanxi Basic Research Program for Young Scientists (grant no. 202203021212366) and the Basic Research Plan Project of Shanxi Province (grant no. 20210302123265).
Not applicable.
CZ proposed the concept and edited the draft. WX and ZB wrote original draft. FF, LL and LC reviewed and finalized the draft. Data authentication is not applicable. All authors have read and approved the final version of the manuscript.
Not applicable.
Not applicable.
The authors declare that they have no competing interests.
|
Kariya Y and Kariya Y: Osteopontin in cancer: Mechanisms and therapeutic targets. Int J Transl Med. 2:419–447. 2022. | |
|
Liu X, Fang J, Huang S, Wu X, Xie X, Wang J, Liu F, Zhang M, Peng Z and Hu N: Tumor-on-a-chip: From bioinspired design to biomedical application. Microsyst Nanoeng. 7:502021. View Article : Google Scholar : PubMed/NCBI | |
|
Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A and Bray F: Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 71:209–249. 2021.PubMed/NCBI | |
|
Yang M, Cui M, Sun Y, Liu S and Jiang W: Mechanisms, combination therapy, and biomarkers in cancer immunotherapy resistance. Cell Commun Signa. 22:3382024. View Article : Google Scholar : PubMed/NCBI | |
|
Toledo B, Deiana C, Scianò F, Brandi G, Marchal JA, Perán M and Giovannetti E: Treatment resistance in pancreatic and biliary tract cancer: Molecular and clinical pharmacology perspectives. Expert Rev Clin Pharmacol. 17:323–347. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang Z, Bao C, Jiang L, Wang S, Wang K, Lu C and Fang H: When cancer drug resistance meets metabolomics (bulk, single-cell and/or spatial): Progress, potential, and perspective. Front Oncol. 12:10542332022. View Article : Google Scholar : PubMed/NCBI | |
|
Yim A, Smith C and Brown AM: Osteopontin/secreted phosphoprotein-1 harnesses glial-, immune-, and neuronal cell ligand-receptor interactions to sense and regulate acute and chronic neuroinflammation. Immunol Rev. 311:224–233. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Sodek J, Ganss B and McKee MD: Osteopontin. Crit Rev Oral Biol Med. 11:279–303. 2000. View Article : Google Scholar : PubMed/NCBI | |
|
Senger DR, Wirth DF and Hynes RO: Transformed mammalian cells secrete specific proteins and phosphoproteins. Cell. 16:885–893. 1979. View Article : Google Scholar : PubMed/NCBI | |
|
Bastos ACSF, Blunck CB, Emerenciano M and Gimba ERP: Osteopontin and their roles in hematological malignancies: Splice variants on the new avenues. Cancer Lett. 408:138–143. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Gimba ER and Tilli TM: Human osteopontin splicing isoforms: Known roles, potential clinical applications and activated signaling pathways. Cancer Lett. 331:11–17. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Jia Q, Ouyang Y, Yang Y, Yao S, Chen X and Hu Z: Osteopontin: A novel therapeutic target for respiratory diseases. Lung. 202:25–39. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao H, Chen Q, Alam A, Cui J, Suen KC, Soo AP, Eguchi S, Gu J and Ma D: The role of osteopontin in the progression of solid organ tumour. Cell Death Dis. 9:3562018. View Article : Google Scholar : PubMed/NCBI | |
|
Panda VK, Mishra B, Nath AN, Butti R, Yadav AS, Malhotra D, Khanra S, Mahapatra S, Mishra P, Swain B, et al: Osteopontin: A key multifaceted regulator in tumor progression and immunomodulation. Biomedicines. 12:15272024. View Article : Google Scholar : PubMed/NCBI | |
|
Mi Z, Guo H, Russell MB, Liu Y, Sullenger BA and Kuo PC: RNA aptamer blockade of osteopontin inhibits growth and metastasis of MDA-MB231 breast cancer cells. Mol Ther. 17:153–161. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Kumar V, Behera R, Lohite K, Karnik S and Kundu GC: p38 kinase is crucial for osteopontin-induced furin expression that supports cervical cancer progression. Cancer Res. 70:10381–10391. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Zagani R, Hamzaoui N, Cacheux W, de Reyniès A, Terris B, Chaussade S, Romagnolo B, Perret C and Lamarque D: Cyclooxygenase-2 inhibitors down-regulate osteopontin and Nr4A2-new therapeutic targets for colorectal cancers. Gastroenterology. 137:1358–1366.e1-e3. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Standal T, Borset M and Sundan A: Role of osteopontin in adhesion, migration, cell survival and bone remodeling. Exp Oncol. 26:179–184. 2004.PubMed/NCBI | |
|
Briones-Orta MA, Avendaño-Vázquez SE, Aparicio-Bautista DI, Coombes JD, Weber GF and Syn WK: Osteopontin splice variants and polymorphisms in cancer progression and prognosis. Biochim Biophys Acta Rev Cancer. 1868:93–108.A. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Hao C, Cui Y, Owen S, Li W, Cheng S and Jiang WG: Human osteopontin: Potential clinical applications in cancer (review). Int J Mol Med. 39:1327–1337. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
O'Regan A and Berman JS: Osteopontin: A key cytokine in cell-mediated and granulomatous inflammation. Int J Exp Pathol. 81:373–390. 2000. View Article : Google Scholar : PubMed/NCBI | |
|
Mirzaei A, Mohammadi S, Ghaffari SH, Yaghmaie M, Vaezi M, Alimoghaddam K and Ghavamzadeh A: Osteopontin b and c splice isoforms in leukemias and solid tumors: Angiogenesis alongside chemoresistance. Asian Pac J Cancer Prev. 19:615–623. 2018.PubMed/NCBI | |
|
Castello LM, Raineri D, Salmi L, Clemente N, Vaschetto R, Quaglia M, Garzaro M, Gentilli S, Navalesi P, Cantaluppi V, et al: Osteopontin at the crossroads of inflammation and tumor progression. Mediators Inflamm. 2017:40490982017. View Article : Google Scholar : PubMed/NCBI | |
|
Lamort AS, Giopanou I, Psallidas I and Stathopoulos GT: Osteopontin as a link between inflammation and cancer: The thorax in the spotlight. Cells. 8:8152019. View Article : Google Scholar : PubMed/NCBI | |
|
Schytte GN, Christensen B, Bregenov I, Kjøge K, Scavenius C, Petersen SV, Enghild JJ and Sørensen ES: FAM20C phosphorylation of the RGDSVVYGLR motif in osteopontin inhibits interaction with the αvβ3 integrin. J Cell Biochem. 121:4809–4818. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Clark EA and Brugge JS: Integrins and signal transduction pathways: The road taken. Science. 268:233–239. 1995. View Article : Google Scholar : PubMed/NCBI | |
|
Wang KX and Denhardt DT: Osteopontin: Role in immune regulation and stress responses. Cytokine Growth Factor Rev. 19:333–345. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Yokosaki Y, Matsuura N, Sasaki T, Murakami I, Schneider H, Higashiyama S, Saitoh Y, Yamakido M, Taooka Y and Sheppard D: The integrin alpha(9)beta(1) binds to a novel recognition sequence (SVVYGLR) in the thrombin-cleaved amino-terminal fragment of osteopontin. J Biol Chem. 274:36328–36334. 1999. View Article : Google Scholar : PubMed/NCBI | |
|
Ito K, Kon S, Nakayama Y, Kurotaki D, Saito Y, Kanayama M, Kimura C, Diao H, Morimoto J, Matsui Y and Uede T: The differential amino acid requirement within osteopontin in alpha4 and alpha9 integrin-mediated cell binding and migration. Matrix Biol. 28:11–19. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Maeda N and Maenaka K: The roles of matricellular proteins in oncogenic virus-induced cancers and their potential utilities as therapeutic targets. Int J Mol Sci. 17:21982017. View Article : Google Scholar | |
|
Denhardt DT and Guo X: Osteopontin: A protein with diverse functions. FASEB J. 7:1475–1482. 1993. View Article : Google Scholar : PubMed/NCBI | |
|
Kazanecki CC, Uzwiak DJ and Denhardt DT: Control of osteopontin signaling and function by post-translational phosphorylation and protein folding. J Cell Biochem. 102:912–924. 2007. View Article : Google Scholar : PubMed/NCBI | |
|
Bellahcène A, Castronovo V, Ogbureke KUE, Fisher LW and Fedarko NS: Small integrin-binding ligand N-linked glycoproteins (SIBLINGs): Multifunctional proteins in cancer. Nat Rev Cancer. 8:212–226. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Dalal S, Zha Q, Daniels CR, Steagall RJ, Joyner WL, Gadeau AP, Singh M and Singh K: Osteopontin stimulates apoptosis in adult cardiac myocytes via the involvement of CD44 receptors, mitochondrial death pathway, and endoplasmic reticulum stress. Am J Physiol Heart Circ Physiol. 306:H1182–H1191. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Miyazaki K, Okada Y, Yamanaka O, Kitano A, Ikeda K, Kon S, Uede T, Rittling SR, Denhardt DT, Kao WW and Saika S: Corneal wound healing in an osteopontin-deficient mouse. Invest Ophth Vis Sci. 49:1367–1375. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Wei J, Marisetty A, Schrand B, Gabrusiewicz K, Hashimoto Y, Ott M, Grami Z, Kong LY, Ling X, Caruso H, et al: Osteopontin mediates glioblastoma-associated macrophage infiltration and is a potential therapeutic target. J Clin Invest. 129:137–149. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Sun C, Rahman MSU, Enkhjargal B, Peng J, Zhou K, Xie Z, Wu L, Zhang T, Zhu Q, Tang J, et al: Osteopontin modulates microglial activation states and attenuates inflammatory responses after subarachnoid hemorrhage in rats. Exp Neurol. 371:1145852024. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao Y, Huang Z, Gao L, Ma H and Chang R: Osteopontin/SPP1: A potential mediator between immune cells and vascular calcification. Front Immunol. 15:13955962024. View Article : Google Scholar : PubMed/NCBI | |
|
Tang S, Hu H, Li M, Zhang K, Wu Q, Liu X, Wu L, Yu B and Chen X: OPN promotes pro-inflammatory cytokine expression via ERK/JNK pathway and M1 macrophage polarization in rosacea. Front Immunol. 14:12859512024. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao L, Wang Z, Tan Y, Ma J, Huang W, Zhang X, Jin C, Zhang T, Liu W and Yang YG: IL-17A/CEBPβ/OPN/LYVE-1 axis inhibits anti-tumor immunity by promoting tumor-associated tissue-resident macrophages. Cell Rep. 43:1150392024. View Article : Google Scholar : PubMed/NCBI | |
|
He Z, Shen S, Yi Y, Ren L, Tao H, Wang F and Jia Y: CD31 promotes diffuse large B-cell lymphoma metastasis by upregulating OPN through the AKT pathway and inhibiting CD8+ T cells through the mTOR pathway. Am J Transl Res. 15:2656–2675. 2023.PubMed/NCBI | |
|
Ding W, Deng S, Wang Z, Chen X, Xu Z, Zhong J, Wu X, Lin R, Ye S, Li C and Li H: CD163+ macrophages drive rapid pulmonary fibrosis via osteopontin secretion. Int Immunopharmacol. 161:1149762025. View Article : Google Scholar : PubMed/NCBI | |
|
Wang H, Gan Z, Wang Y, Hu D, Zhang L, Ye F and Duan P: A noninvasive menstrual Blood-based diagnostic platform for endometriosis using digital droplet Enzyme-linked immunosorbent assay and Single-cell RNA sequencing. Research (Wash DC). 8:06522025.PubMed/NCBI | |
|
Crawford HC, Matrisian LM and Liaw L: Distinct roles of osteopontin in host defense activity and tumor survival during squamous cell carcinoma progression in vivo. Cancer Res. 58:5206–5215. 1998.PubMed/NCBI | |
|
Icer MA and Gezmen-Karadag M: The multiple functions and mechanisms of osteopontin. Clin Biochem. 59:17–24. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Steitz SA, Speer MY, McKee MD, Liaw L, Almeida M, Yang H and Giachelli CM: Osteopontin inhibits mineral deposition and promotes regression of ectopic calcification. Am J Pathol. 161:2035–2046. 2002. View Article : Google Scholar : PubMed/NCBI | |
|
Giachelli CM and Steitz S: Osteopontin: A versatile regulator of inflammation and biomineralization. Matrix Biol. 19:615–622. 2000. View Article : Google Scholar : PubMed/NCBI | |
|
McKee MD, Pedraza CE and Kaartinen MT: Osteopontin and wound healing in bone. Cells Tissues Organs. 194:313–319. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Ishijima M, Rittling SR, Yamashita T, Tsuji K, Kurosawa H, Nifuji A, Denhardt DT and Noda M: Enhancement of osteoclastic bone resorption and suppression of osteoblastic bone formation in response to reduced mechanical stress do not occur in the absence of osteopontin. J Exp Med. 193:399–404. 2001. View Article : Google Scholar : PubMed/NCBI | |
|
Miragliotta V, Pirone A, Donadio E, Abramo F, Ricciardi MP and Theoret CL: Osteopontin expression in healing wounds of horses and in human keloids. Equine Vet J. 48:72–77. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Wen Y, Feng D, Wu H, Liu W, Li H, Wang F, Xia Q, Gao WQ and Kong X: Defective initiation of liver regeneration in osteopontin-deficient mice after partial hepatectomy due to insufficient activation of IL-6/Stat3 pathway. Int J Biol Sci. 11:1236–1247. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Ström A, Franzén A, Wängnerud C, Knutsson AK, Heinegård D and Hultgårdh-Nilsson A: Altered vascular remodeling in osteopontin-deficient atherosclerotic mice. J Vasc Res. 41:314–322. 2004. View Article : Google Scholar : PubMed/NCBI | |
|
Khuri FR, Shin DM, Glisson BS, Lippman SM and Hong WK: Treatment of patients with recurrent or metastatic squamous cell carcinoma of the head and neck: Current status and future directions. Semin Oncol. 27:25–33. 2000.PubMed/NCBI | |
|
Peng Y, Feng W, Huang H, Chen Y and Yang SY: Macrophage-targeting antisenescence nanomedicine enables in-situ NO induction for gaseous and antioxidative atherosclerosis intervention. Bioact Mater. 48:294–312. 2025.PubMed/NCBI | |
|
Lomashvili KA, Cobbs S, Hennigar RA, Hardcastle KI and O'Neill WC: Phosphate-induced vascular calcification: Role of pyrophosphate and osteopontin. J Am Soc Nephrol. 15:1392–1401. 2004. View Article : Google Scholar : PubMed/NCBI | |
|
Speer MY, McKee MD, Guldberg RE, Liaw L, Yang HY, Tung E, Karsenty G and Giachelli CM: Inactivation of the osteopontin gene enhances vascular calcification of matrix Gla Protein-deficient mice: Evidence for osteopontin as an inducible inhibitor of vascular calcification in vivo. J Exp Med. 196:1047–1055. 2002. View Article : Google Scholar : PubMed/NCBI | |
|
Bandopadhyay M, Bulbule A, Butti R, Chakraborty G, Ghorpade P, Ghosh P, Gorain M, Kale S, Kumar D, Kumar S, et al: Osteopontin as a therapeutic target for cancer. Expert Opin Ther Targets. 18:883–895. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Chambers AF, Wilson SM, Kerkvliet N, O'Malley FP, Harris JF and Casson AG: Osteopontin expression in lung cancer. Lung Cancer. 15:311–323. 1996. View Article : Google Scholar : PubMed/NCBI | |
|
Gotoh M, Sakamoto M, Kanetaka K, Chuuma M and Hirohashi S: Overexpression of osteopontin in hepatocellular carcinoma. Pathol Int. 52:19–24. 2002. View Article : Google Scholar : PubMed/NCBI | |
|
Cook AC, Tuck AB, McCarthy S, Turner JG, Irby RB, Bloom GC, Yeatman TJ and Chambers AF: Osteopontin induces multiple changes in gene expression that reflect the six ‘hallmarks of cancer’ in a model of breast cancer progression. Mol Carcinog. 43:225–236. 2005. View Article : Google Scholar : PubMed/NCBI | |
|
Moorman HR, Poschel D, Klement JD, Lu C, Redd PS and Liu K: Osteopontin: A key regulator of tumor progression and immunomodulation. Cancers (Basel). 12:33792020. View Article : Google Scholar : PubMed/NCBI | |
|
Kariya Y, Oyama M, Kariya Y and Hashimoto Y: Phosphorylated osteopontin secreted from cancer cells induces cancer cell motility. Biomolecules. 11:13232021. View Article : Google Scholar : PubMed/NCBI | |
|
Klement JD, Paschall AV, Redd PS, Ibrahim ML, Lu C, Yang D, Celis E, Abrams SI, Ozato K and Liu K: An osteopontin/CD44 immune checkpoint controls CD8+ T cell activation and tumor immune evasion. J Clin Invest. 128:5549–5560. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Cao J, Li J, Sun L, Qin T, Xiao Y, Chen K, Qian W, Duan W, Lei J, Ma J, et al: Hypoxia-driven paracrine osteopontin/integrin αvβ3 signaling promotes pancreatic cancer cell epithelial-mesenchymal transition and cancer stem cell-like properties by modulating forkhead box protein M1. Mol Oncol. 13:228–245. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Qian J, LeSavage BL, Hubka KM, Ma C, Natarajan S, Eggold JT, Xiao Y, Fuh KC, Krishnan V, Enejder A, et al: Cancer-associated mesothelial cells promote ovarian cancer chemoresistance through paracrine osteopontin signaling. J Clin Invest. 131:e1461862021. View Article : Google Scholar : PubMed/NCBI | |
|
Huang R, Quan Y, Chen JH, Wang TF, Xu M, Ye M, Yuan H, Zhang CJ, Liu XJ and Min ZJ: Osteopontin promotes cell migration and invasion, and inhibits apoptosis and autophagy in colorectal cancer by activating the p38 MAPK signaling pathway. Cell Physiol Biochem. 41:1851–1864. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Ahmed M, Sottnik JL, Dancik GM, Sahu D, Hansel DE, Theodorescu D and Schwartz MA: An osteopontin/CD44 Axis in RhoGDI2-Mediated metastasis suppression. Cancer Cell. 30:432–443. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Fresno Vara JA, Casado E, de Castro J, Cejas P, Belda-Iniesta C and González-Barón M: PI3K/akt signalling pathway and cancer. Cancer Treat Rev. 30:193–204. 2004. View Article : Google Scholar : PubMed/NCBI | |
|
Sun C, Enkhjargal B, Reis C, Zhou KR, Xie ZY, Wu LY, Zhang TY, Zhu QQ, Tang JP, Jiang XD and Zhang JH: Osteopontin attenuates early brain injury through regulating autophagy-apoptosis interaction after subarachnoid hemorrhage in rats. Cns Neurosci Ther. 25:1162–1172. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Wei R, Wong JPC and Kwok HF: Osteopontin-a promising biomarker for cancer therapy. J Cancer. 8:2173–2183. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao J, Dong L, Lu B, Wu G, Xu D, Chen J, Li K, Tong X, Dai J, Yao S, et al: Down-regulation of osteopontin suppresses growth and metastasis of hepatocellular carcinoma via induction of apoptosis. Gastroenterology. 135:956–968. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Fedarko NS, Fohr B, Robey PG, Young MF and Fisher LW: Factor H binding to bone sialoprotein and osteopontin enables tumor cell evasion of complement-mediated attack. J Biol Chem. 275:16666–16672. 2000. View Article : Google Scholar : PubMed/NCBI | |
|
Zhou J, Chen X, Zhou P, Sun X, Chen Y, Li M, Chu Y, Zhou J, Hu X, Luo Y, et al: Osteopontin is required for the maintenance of leukemia stem cells in acute myeloid leukemia. Biochem Biophys Res Commun. 600:29–34. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Chen Q, Shou P, Zhang L, Xu C, Zheng C, Han Y, Li W, Huang Y, Zhang X, Shao C, et al: An osteopontin-integrin interaction plays a critical role in directing adipogenesis and osteogenesis by mesenchymal stem cells. Stem Cells. 32:327–337. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Irby RB, Mccarthy SM and Yeatman TJ: Osteopontin regulates multiple functions contributing to human colon cancer development and progression. Clin Exp Metastas. 21:515–523. 2004. View Article : Google Scholar : PubMed/NCBI | |
|
Denhardt DT, Mistretta D, Chambers AF, Krishna S, Porter JF, Raghuram S and Rittling SR: Transcriptional regulation of osteopontin and the metastatic phenotype: Evidence for a Ras-activated enhancer in the human OPN promoter. Clin Exp Metastas. 20:77–84. 2003. View Article : Google Scholar : PubMed/NCBI | |
|
Kale S, Raja R, Thorat D, Soundararajan G, Patil TV and Kundu GC: Osteopontin signaling upregulates cyclooxygenase-2 expression in tumor-associated macrophages leading to enhanced angiogenesis and melanoma growth via α9β1 integrin. Oncogene. 33:2295–2306. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Mi Z, Guo H, Wai PY, Gao C and Kuo PC: Integrin-linked kinase regulates osteopontin-dependent MMP-2 and uPA expression to convey metastatic function in murine mammary epithelial cancer cells. Carcinogenesis. 27:1134–1145. 2006. View Article : Google Scholar : PubMed/NCBI | |
|
Wai PY and Kuo PC: The role of osteopontin in tumor metastasis. J Surg Res. 121:228–241. 2004. View Article : Google Scholar : PubMed/NCBI | |
|
Gupta A, Zhou C and Chellaiah M: Osteopontin and MMP9: Associations with VEGF expression/secretion and angiogenesis in PC3 prostate cancer cells. Cancers (Basel). 5:617–638. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Fukusada S, Shimura T, Natsume M, Nishigaki R, Okuda Y, Iwasaki H, Sugimura N, Kitagawa M, Katano T, Tanaka M, et al: Osteopontin secreted from obese adipocytes enhances angiogenesis and promotes progression of pancreatic ductal adenocarcinoma in obesity. Cell Oncol. 45:229–244. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Wei R, Liu S, Zhang S, Min L and Zhu S: Cellular and extracellular components in tumor microenvironment and their application in early diagnosis of cancers. Anal Cell Pathol (Amst). 2020:62837962020.PubMed/NCBI | |
|
Tan Y, Zhao L, Yang YG and Liu W: The role of osteopontin in tumor progression through tumor-associated macrophages. Front Oncol. 12:9532832022. View Article : Google Scholar : PubMed/NCBI | |
|
Shao L, Zhang B, Wang L, Wu L, Kan Q and Fan K: MMP-9-cleaved osteopontin isoform mediates tumor immune escape by inducing expansion of myeloid-derived suppressor cells. Biochem Biophys Res Commun. 493:1478–1484. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Kariya Y, Oyama M, Hashimoto Y, Gu J and Kariya Y: β4-integrin/PI3K signaling promotes tumor progression through the galectin-3-N-glycan complex. Mol Cancer Res. 16:1024–1034. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Butti R, Kumar TVS, Nimma R, Banerjee P, Kundu IG and Kundu GC: Osteopontin signaling in shaping tumor microenvironment conducive to malignant progression. Adv Exp Med Biol. 1329:419–441. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Rogers MP, Kothari A, Read M, Kuo PC and Mi Z: Maintaining myofibroblastic-like cancer-associated fibroblasts by cancer stemness signal transduction feedback loop. Cureus. 14:e293542022.PubMed/NCBI | |
|
Leung LL, Myles T and Morser J: Thrombin cleavage of osteopontin and the host anti-tumor immune response. Cancers. 15:34802023. View Article : Google Scholar : PubMed/NCBI | |
|
Yan Z, Hu X, Tang B and Deng F: Role of osteopontin in cancer development and treatment. Heliyon. 9:e210552023. View Article : Google Scholar : PubMed/NCBI | |
|
Rangaswami H, Bulbule A and Kundu GC: Osteopontin: Role in cell signaling and cancer progression. Trends Cell Biol. 16:79–87. 2006. View Article : Google Scholar : PubMed/NCBI | |
|
Abdelrahman KS, Hassan HA, Abdel-Aziz SA, Marzouk AA, Narumi A, Konno H and Abdel-Aziz M: JNK signaling as a target for anticancer therapy. Pharmacol Rep. 73:405–434. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Shi L and Wang X: Role of osteopontin in lung cancer evolution and heterogeneity. Semin Cell Dev Biol. 64:40–47. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Iida T, Wagatsuma K, Hirayama D and Nakase H: Is osteopontin a friend or foe of cell apoptosis in inflammatory gastrointestinal and liver diseases? Int J Mol Sci. 19:72017. View Article : Google Scholar : PubMed/NCBI | |
|
Katoh M: Multi-layered prevention and treatment of chronic inflammation, organ fibrosis and cancer associated with canonical WNT/β-catenin signaling activation (review). Int J Mol Med. 42:713–725. 2018.PubMed/NCBI | |
|
Bastos ACSDF, Gomes AVP, Silva GR, Emerenciano M, Ferreira LB and Gimba ERP: The intracellular and secreted sides of osteopontin and their putative physiopathological roles. Int J Mol Sci. 24:29422023. View Article : Google Scholar : PubMed/NCBI | |
|
Berge G, Pettersen S, Grotterød I, Bettum IJ, Boye K and Mælandsmo GM: Osteopontin-an important downstream effector of S100A4-mediated invasion and metastasis. Int J Cancer. 129:780–790. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Amilca-Seba K, Sabbah M, Larsen AK and Denis JA: Osteopontin as a regulator of colorectal cancer progression and its clinical applications. Cancers. 13:37932021. View Article : Google Scholar : PubMed/NCBI | |
|
Hao C, Lane J and Jiang WG: Osteopontin and cancer: Insights into its role in drug resistance. Biomedicines. 11:1972023. View Article : Google Scholar : PubMed/NCBI | |
|
Moldogazieva NT, Mokhosoev IM, Zavadskiy SP and Terentiev AA: Proteomic profiling and artificial intelligence for hepatocellular carcinoma translational medicine. Biomedicines. 9:1592021. View Article : Google Scholar : PubMed/NCBI | |
|
Messex JK, Byrd CJ, Thomas MU and Liou GY: Macrophages cytokine Spp1 increases growth of prostate intraepithelial neoplasia to promote prostate tumor progression. Int J Mol Sci. 23:42472022. View Article : Google Scholar : PubMed/NCBI | |
|
Insua-Rodríguez J, Pein M, Hongu T, Meier J, Descot A, Lowy CM, De Braekeleer E, Sinn HP, Spaich S, Sütterlin M, et al: Stress signaling in breast cancer cells induces matrix components that promote chemoresistant metastasis. EMBO Mol Med. 10:e90032018. View Article : Google Scholar : PubMed/NCBI | |
|
Li NY, Weber CE, Wai PY, Cuevas BD, Zhang J, Kuo PC and Mi Z: An MAPK-dependent pathway induces epithelial-mesenchymal transition via twist activation in human breast cancer cell lines. Surgery. 154:404–410. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
McCubrey JA, Steelman LS, Chappell WH, Abrams SL, Montalto G, Cervello M, Nicoletti F, Fagone P, Malaponte G, Mazzarino MC, et al: Mutations and deregulation of ras/raf/MEK/ERK and PI3K/PTEN/akt/mTOR cascades which alter therapy response. Oncotarget. 3:954–987. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Yang YF, Chang YC, Jan YH, Yang CJ, Huang MS and Hsiao M: Squalene synthase promotes the invasion of lung cancer cells via the osteopontin/ERK pathway. Oncogenesis. 9:782020. View Article : Google Scholar : PubMed/NCBI | |
|
Chernaya G, Mikhno N, Khabalova T, Svyatchenko S, Mostovich L, Shevchenko S and Gulyaeva L: The expression profile of integrin receptors and osteopontin in thyroid malignancies varies depending on the tumor progression rate and presence of BRAF V600E mutation. Surg Oncol. 27:702–708. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Sun BS, Dong QZ, Ye QH, Sun HJ, Jia HL, Zhu XQ, Liu DY, Chen J, Xue Q, Zhou HJ, et al: Lentiviral-mediated miRNA against osteopontin suppresses tumor growth and metastasis of human hepatocellular carcinoma. Hepatology. 48:1834–1842. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Lin YH and Yang-Yen HF: The osteopontin-CD44 survival signal involves activation of the phosphatidylinositol 3-kinase/akt signaling pathway. J Biol Chem. 276:46024–46030. 2001. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang H, Guo M, Chen JH, Wang Z, Du XF, Liu PX and Li WH: Osteopontin knockdown inhibits αv,β3 integrin-induced cell migration and invasion and promotes apoptosis of breast cancer cells by inducing autophagy and inactivating the PI3K/akt/mTOR pathway. Cell Physiol Biochem. 33:991–1002. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Fu Y, Zhang Y, Lei Z, Liu T, Cai T, Wang A, Du W, Zeng Y, Zhu J, Liu Z and Huang JA: Abnormally activated OPN/integrin αVβ3/FAK signalling is responsible for EGFR-TKI resistance in EGFR mutant Non-small-cell lung cancer. J Hematol Oncol. 13:1692020. View Article : Google Scholar : PubMed/NCBI | |
|
Hui T, Sørensen ES and Rittling SR: Osteopontin binding to the alpha 4 integrin requires highest affinity integrin conformation, but is independent of Post-translational modifications of osteopontin. Matrix Biol. 41:19–25. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Wu Q, Li L, Miao C, Hasnat M, Sun L, Jiang Z and Zhang L: Osteopontin promotes hepatocellular carcinoma progression through inducing JAK2/STAT3/NOX1-mediated ROS production. Cell Death Dis. 13:3412022. View Article : Google Scholar : PubMed/NCBI | |
|
Behera R, Kumar V, Lohite K, Karnik S and Kundu GC: Activation of JAK2/STAT3 signaling by osteopontin promotes tumor growth in human breast cancer cells. Carcinogenesis. 31:192–200. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Cao L, Fan X, Jing W, Liang Y, Chen R, Liu Y, Zhu M, Jia R, Wang H, Zhang X, et al: Osteopontin promotes a cancer stem cell-like phenotype in hepatocellular carcinoma cells via an integrin-NF-κB-HIF-1α pathway. Oncotarget. 6:6627–6640. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Chen B, Liang S, Guo H, Xu L, Li J and Peng J: OPN promotes cell proliferation and invasion through NF-κB in human esophageal squamous cell carcinoma. Genet Res (Camb). 2022:31548272022. View Article : Google Scholar : PubMed/NCBI | |
|
Saurav S, Karfa S, Vu T, Liu Z, Datta A, Manne U, Samuel T and Datta PK: Overcoming irinotecan resistance by targeting its downstream signaling pathways in colon cancer. Cancers (Basel). 16:34912024. View Article : Google Scholar : PubMed/NCBI | |
|
Graessmann M, Berg B, Fuchs B, Klein A and Graessmann A: Chemotherapy resistance of mouse WAP-SVT/t breast cancer cells is mediated by osteopontin, inhibiting apoptosis downstream of caspase-3. Oncogene. 26:2840–2850. 2007. View Article : Google Scholar : PubMed/NCBI | |
|
Yong HY, Koh MS and Moon A: The p38 MAPK inhibitors for the treatment of inflammatory diseases and cancer. Expert Opin Investig Drugs. 18:1893–1905. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Yu X, Du Y, Liang S, Zhang N, Jing S, Sui L, Kong Y, Dong M and Kong H: OPN up-regulated proliferation and invasion of head and neck squamous cell carcinoma through the p38MAPK signaling pathway. Oral Surg Oral Med Oral Pathol Oral Radiol. 136:70–79. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Pang H, Cai L, Yang Y, Chen X, Sui G and Zhao C: Knockdown of osteopontin chemosensitizes MDA-MB-231 cells to cyclophosphamide by enhancing apoptosis through activating p38 MAPK pathway. Cancer Biother Radiopharm. 26:165–173. 2011.PubMed/NCBI | |
|
Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I and Jemal A: Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 74:229–263. 2024.PubMed/NCBI | |
|
Davidson BL and McCray PB: Current prospects for RNA interference-based therapies. Nat Rev Genet. 12:329–340. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Ben-David-Naim M, Dagan A, Grad E, Aizik G, Nordling-David MM, Morss Clyne A, Granot Z and Golomb G: Targeted siRNA nanoparticles for mammary carcinoma therapy. Cancers (Basel). 11:4422019. View Article : Google Scholar : PubMed/NCBI | |
|
Yang L, Zhao W, Zuo WS, Wei L, Song XR, Wang XW, Zheng G and Zheng MZ: Silencing of osteopontin promotes the radiosensitivity of breast cancer cells by reducing the expression of hypoxia inducible factor 1 and vascular endothelial growth factor. Chin Med J (Engl). 125:293–299. 2012.PubMed/NCBI | |
|
Wang F, Fu K, Wang Y, Pan C, Wang X, Liu Z, Yang C, Zheng Y, Li X, Lu Y, et al: Small-molecule agents for cancer immunotherapy. Acta Pharm Sin B. 14:905–952. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Sharma P, Kumar S and Kundu GC: Transcriptional regulation of human osteopontin promoter by histone deacetylase inhibitor, trichostatin A in cervical cancer cells. Mol Cancer. 9:1782010. View Article : Google Scholar : PubMed/NCBI | |
|
Kumar S, Patil HS, Sharma P, Kumar D, Dasari S, Puranik VG, Thulasiram HV and Kundu GC: Andrographolide inhibits osteopontin expression and breast tumor growth through down regulation of PI3 kinase/akt signaling pathway. Curr Mol Med. 12:952–966. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Matsuura M, Suzuki T and Saito T: Osteopontin is a new target molecule for ovarian clear cell carcinoma therapy. Cancer Sci. 101:1828–1833. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Lv J, Shao Q, Wang H, Shi H, Wang T, Gao W, Song B, Zheng G, Kong B and Qu X: Effects and mechanisms of curcumin and basil polysaccharide on the invasion of SKOV3 cells and dendritic cells. Mol Med Rep. 8:1580–1586. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Nimjee SM, White RR, Becker RC and Sullenger BA: Aptamers as therapeutics. Annu Rev Pharmacol Toxicol. 57:61–79. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
He S, Du Y, Tao H and Duan H: Advances in aptamer-mediated targeted delivery system for cancer treatment. Int J Biol Macromol. 238:1241732023. View Article : Google Scholar : PubMed/NCBI | |
|
Talbot LJ, Mi Z, Bhattacharya SD, Kim V, Guo H and Kuo PC: Pharmacokinetic characterization of an RNA aptamer against osteopontin and demonstration of in vivo efficacy in reversing growth of human breast cancer cells. Surgery. 150:224–230. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Ahmed M and Kundu GC: Osteopontin selectively regulates p70S6K/mTOR phosphorylation leading to NF-kappaB dependent AP-1-mediated ICAM-1 expression in breast cancer cells. Mol Cancer. 9:1012010. View Article : Google Scholar : PubMed/NCBI | |
|
Ahmed M, Behera R, Chakraborty G, Jain S, Kumar V, Sharma P, Bulbule A, Kale S, Kumar S, Mishra R, et al: Osteopontin: A potentially important therapeutic target in cancer. Expert Opin Ther Targets. 15:1113–1126. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Chiou J, Chang YC, Tsai HF, Lin YF, Huang MS, Yang CJ and Hsiao M: Follistatin-like protein 1 inhibits lung cancer metastasis by preventing proteolytic activation of osteopontin. Cancer Res. 79:6113–6125. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Salih E, Ashkar S, Gerstenfeld LC and Glimcher MJ: Identification of the phosphorylated sites of metabolically 32P-labeled osteopontin from cultured chicken osteoblasts. J Biol Chem. 272:13966–13973. 1997. View Article : Google Scholar : PubMed/NCBI | |
|
Peraramelli S, Zhou Q, Zhou Q, Wanko B, Zhao L, Nishimura T, Leung TH, Mizuno S, Ito M, Myles T, et al: Thrombin cleavage of osteopontin initiates osteopontin's tumor-promoting activity. J Thromb Haemost. 20:1256–1270. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Zou Y, Sun X, Wang Y, Yan C, Liu Y, Li J, Zhang D, Zheng M, Chung RS and Shi B: Single siRNA nanocapsules for effective siRNA brain delivery and glioblastoma treatment. Adv Mater. 32:e20004162020. View Article : Google Scholar : PubMed/NCBI | |
|
Bansal I, Pandey AK and Ruwali M: Small-molecule inhibitors of kinases in breast cancer therapy: Recent advances, opportunities, and challenges. Front Pharmacol. 14:12445972023. View Article : Google Scholar : PubMed/NCBI | |
|
Lu C, Liu Z, Klement JD, Yang D, Merting AD, Poschel D, Albers T, Waller JL, Shi H and Liu K: WDR5-H3K4me3 epigenetic axis regulates OPN expression to compensate PD-L1 function to promote pancreatic cancer immune escape. J Immunother Cancer. 9:e0026242021. View Article : Google Scholar : PubMed/NCBI | |
|
Zhu Y, Yang J, Xu D, Gao XM, Zhang Z, Hsu JL, Li CW, Lim SO, Sheng YY, Zhang Y, et al: Disruption of tumour-associated macrophage trafficking by the osteopontin-induced colony-stimulating factor-1 signalling sensitises hepatocellular carcinoma to anti-PD-L1 blockade. Gut. 68:1653–1666. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Saleh S, Thompson DE, McConkey J, Murray P and Moorehead RA: Osteopontin regulates proliferation, apoptosis, and migration of murine Claudin-low mammary tumor cells. BMC Cancer. 16:3592016. View Article : Google Scholar : PubMed/NCBI | |
|
Cho W-Y, Hong SH, Singh B, Islam MA, Lee S, Lee AY, Gankhuyag N, Kim JE, Yu KN, Kim KH, et al: Suppression of tumor growth in lung cancer xenograft model mice by poly(sorbitol-co-PEI)-mediated delivery of osteopontin siRNA. Eur J Pharm Biopharm. 94:450–462. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Bhattacharya SD, Garrison J, Guo H, Mi Z, Markovic J, Kim VM and Kuo PC: Micro-RNA-181a regulates osteopontin-dependent metastatic function in hepatocellular cancer cell lines. Surgery. 148:291–297. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Mason CK, McFarlane S, Johnston PG, Crowe P, Erwin PJ, Domostoj MM, Campbell FC, Manaviazar S, Hale KJ, El-Tanani M, et al: Agelastatin A: A novel inhibitor of osteopontin-mediated adhesion, invasion, and colony formation. Mol Cancer Ther. 7:548–558. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Dai J, Li B, Shi J, Peng L, Zhang D, Qian W, Hou S, Zhao L, Gao J, Cao Z, et al: A humanized Anti-osteopontin antibody inhibits breast cancer growth and metastasis in vivo. Cancer Immunol Immunother. 59:355–366. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Teramoto H, Castellone MD, Malek RL, Letwin N, Frank B, Gutkind JS and Lee NH: Autocrine activation of an osteopontin-CD44-Rac pathway enhances invasion and transformation by H-RasV12. Oncogene. 24:489–501. 2005. View Article : Google Scholar : PubMed/NCBI |