1. Introduction
An estimated new cases of cancer incidences are 19.3 million and cancer related deaths are 10.0 million according to GLOBOCAN 2020 [
1]. Recent data showed that female breast cancer accounts for the second most commonly occurring cancer after lung globally. Tumor microenvironment (TME) consists of heterogenous cell subpopulations including infiltrating immune cells like tumor-associated macrophages (TAMs), dendritic cells (DCs), lymphocytes and other stromal cells like cancer-associated fibroblasts (CAFs), endothelial cells, pericytes, and extracellular matrix (ECM) [
2]. All these components form a complex network through cell-cell and cell-matrix interactions to shape the microenvironment conducive to tumor progression and metastasis. These cells enhance several hallmarks of cancer via secreting a rich array of cytokines and chemokines [
2].
Osteopontin (OPN), a sialic acid rich, non-collagenous, chemokine like, glycosylated, phospoprotein is expressed in various cell types such as osteoblasts, osteoclasts, epithelial cells, endothelial cells and immune and stromal cells [
3]. OPN has emerged as a pivotal mediator facilitating intracellular crosstalk within the breast TME. Due to variations in post-translational modifications (PTMs) and proteolytic cleavage, the molecular weight of OPN ranges from 44 to 75 kDa [
3]. OPN plays a crucial role in several normal physiological processes including vascularisation, bone remodelling and immue-regulation as well as pathological processes including tumor progression, metastasis, immunosuppression, angiogenesis and chemoresistance [
4,
5]. The overexpression of OPN has been linked to poor prognosis in various malignancies, including breast, lung cancer, glioblastoma, colorectal cancer, hepatocellular carcinoma, bladder cancer, melanoma, and acute myeloid leukemia [
6]. It also serves as diagnostic as well as prognostic biomarker in different cancers [
6].
In the present scenario, the diverse function attributes to OPN in tumor progression are directly linked to its structural features binding to specific integrins and CD44. Moreover, OPN- mediated signalling activates various oncogenic molecules and promotes tumor growth as well as metastasis [
3]. Interestingly, OPN also governs immunosuppression by activating macrophages and suppressing T-cell activation in various cancers [
7]. Single cell and spatial transcriptomics analyses, revealed a correlation between interactions of CAF and OPN+ TAM with the progression of cancer [
8].
This review comprehensively covers recent progress in the field of OPN with a focus on elucidating its structural features, immune modulatory properties, its role in regulating CAFs, influencing epithelial-mesenchymal transition (EMT), contributing to the enrichment of cancer stem cells (CSCs), promoting metastasis, fostering therapy resistance, and influencing metabolic regulation. The goal of this review is to provide the latest progress in elucidating the role of OPN in shaping the TME. We also discuss recent treatment strategies to target OPN and its receptors for the management of cancers using small molecules inhibitors, monoclonal antibodies and aptamer-based approaches.
3. OPN Expression in Various Cancers
OPN exhibits high levels of expression in a wide range of tumor types, such as breast, ovarian, cutaneous, head and neck, thyroid, lung, liver, esophageal, gastric, pancreatic, colorectal, kidney, prostate, bladder and glioblastoma [
15]. Tumor, stromal, and tumor- infiltrating myeloid cells express high levels of OPN within the heterogeneous TME [
6]. Clinical research has demonstrated a correlation between OPN expression in various tumor tissues, plasma, serum and has been shown to be correlated with an advanced tumor stage, grade, size, invasiveness, metastasis, poor survival rate of patients in a variety of human malignancies [
6]. Furthermore, the differential expression level of OPN isoforms vary depending upon the cancer types as shown in
Table 1. In general, OPN-a is expressed in various malignancies and is involved in promoting cancer progression, metastasis, angiogenesis, immunosuppression and drug resistance. In breast cancer, OPN-a and OPN-c are expressed at higher levels whereas high level of OPN-b is expressed in lung cancer and are associated with poor survival and relapse [
16]. However, the correlation of OPN expression with its clinical implications in various cancers remain to be explored.
8. Conclusion and Future Perspective
OPN emerges as a pivotal regulator in the intricate interplay between cancer cells and the TME, exerting influence across various facets of cancer progression. The abundance of OPN within the TME is responsible for modulating the fate of tumor and stromal cells but yet its clinical relevance remains to be a major limiting factor. Our earlier reports have dissected several OPN- modulated fundamental signalling pathways in breast and other cancers [
3,
11,
52,
81]. Furthermore, its multifaceted role encompasses angiogenesis, CSCs, bone metastasis, cancer cell metabolism, and modulation of the TME, involving interactions with CAFs, adipocytes, osteoclasts and immune cells. In addition, OPN intricately regulates cancer cell metabolism, particularly in glucose metabolism, influencing glucose transporters and contributing to the Warburg effect [
98]. Moreover, OPN plays a central role by affecting the bone microenvironment, promoting bone metastasis, and contributing to the formation of premetastatic niches [
85]. Thus, OPN acts as a potential therapeutic target for mitigating osteoclast development and controlling skeletal metastases.
In TME, the influence of OPN spans interactions with CAFs, adipocytes and osteoclasts. OPN-reprogrammed CAFs contribute to proinflammatory states, angiogenesis, and EMT, emphasizing its role in cancer progression [
52]. OPN significantly impacts immunomodulation within the TME, influencing macrophage polarization towards a tumor-promoting phenotype (M2 type) and contributing to an immunosuppressive microenvironment. Moreover, OPN is implicated in immune evasion mechanisms, including the inhibition of T-cell activation and the regulation of immune checkpoint molecules such as PD-L1. CAF and TAM interaction are majorly regulated by OPN and its associated signalling events causing upregulation of various tumor events. For example, scRNA-seq revealed that OPN mediates the interaction between OPN-PTGER4 and OPN-CD44 stimulating the crosstalk between HCC cells and macrophages [
162]. Similarly, single-cell transcriptome sequencing identified three TAMs subgroups: C1Q+, FCN1+ and OPN (SPP1)+ TAMs whereas OPN (SPP1)+ TAMs modulate TIME via interaction with CAFs [
163]. Zhang et al reported that OPN+ TAMs are tightly associated with CAFs and endothelial cells in modulating TME [
164]. However, the complex molecular mechanism of OPN mediated TAM subset enrichment, regulation of metabolic switch and intricate crosstalk between CAF-TAM is poorly elucidated. Although, multiple studies have demonstrated the function of CAFs in regulation of TAM, comprehending the influence of TAM in controlling CAF phenotypes warrant further investigation.
Using single-cell and spatial transcriptomics, immunofluorescent labelling, and other methods, have advanced our understanding the role of OPN in inflamed and immune-excluded TME. However, its role in immune-desert needs further exploration [
129]. Detailed mechanistic studies on OPN-regulated networks and core regulatory transcription factors governing OPN expression in tumor and immune cells are warranted.
Considering the central role of OPN in cancer progression, targeted therapies aimed at inhibiting OPN expression or its downstream signalling pathways hold promise. However, current interventions lack efficacy in both preclinical and clinical trials, necessitating further development of small molecules or antibodies to neutralize the effect of OPN. Stratifying breast cancer patient based on expression levels of OPN and associated signalling pathways could tailor treatment strategies and identify the patient subgroups more responsive to OPN-targeted therapies. Understanding the immunomodulatory role of OPN suggests exploring combination therapies involving OPN inhibition and immunotherapeutic agents, potentially enhancing the effectiveness of immune checkpoint inhibitors and promoting antitumor immune responses.
In summary, the diverse function of OPN in breast and other cancers underscore its significance as a potential therapeutic target and diagnostic marker. Thus, the continued research on role of OPN and its associated signalling networks will unravel new insights in developing innovative therapeutic strategies for precision medicine, ultimately improving patient outcomes in various cancers including breast.
Figure 1.
Structural domains of full-length OPN and its receptors. N-terminus of OPN consists of poly-D region, calcium binding domain, and ELVTDFPTDLPAT sequence motif which interacts with α4β1 integrin. The central region consists of RGD domain, binds with other integrins such as αvβ3, αvβ1, αvβ5, αvβ6, αvβ8, α5β1 and α8β1; the SVVYGLR sequence binds to α9β1, α4β1 and α4β7 integrins. The C-terminal region includes another calcium- binding domain, MMP-cleavage site, and heparin-binding domain which facilitates the interaction of OPN with CD44. Adopted from Mirzaei et al, Asian Pacific J of Cancer Prevention, 19 (3), 2018 and Bandopadhyay et al, EOTT, 18 (8), 2014 with modifications.
Figure 1.
Structural domains of full-length OPN and its receptors. N-terminus of OPN consists of poly-D region, calcium binding domain, and ELVTDFPTDLPAT sequence motif which interacts with α4β1 integrin. The central region consists of RGD domain, binds with other integrins such as αvβ3, αvβ1, αvβ5, αvβ6, αvβ8, α5β1 and α8β1; the SVVYGLR sequence binds to α9β1, α4β1 and α4β7 integrins. The C-terminal region includes another calcium- binding domain, MMP-cleavage site, and heparin-binding domain which facilitates the interaction of OPN with CD44. Adopted from Mirzaei et al, Asian Pacific J of Cancer Prevention, 19 (3), 2018 and Bandopadhyay et al, EOTT, 18 (8), 2014 with modifications.
Figure 2.
Schematic illustration of the OPN gene and its splice variants. Alternative splicing of OPN transcript results in five splice variants, which are denoted as OPN- a, OPN-b, OPN-c, OPN-4, and OPN-5. OPN-a is a full-length variant that consists of 7 exons; OPN-b lacks exon 5; while in OPN-c exon 4 is absent. In OPN-4, both exon 4 and exon 5 are missing; whereas OPN-5 is the longest variant which consists of an additional exon, generated from a portion of intron 3. Additionally, four new sub-variants of OPN-5 (OPN-5b, OPN-5c, OPN- 5d, OPN-5e) have been identified. OPN-5a is the same as OPN-5; OPN-5b has the extra shortened exon while in OPN-5c has additional 9 base pairs in the 3’ region of extra exon. In OPN-5d, there is deletion of exon 5 with the addition of 9 base pairs in the 3’ region of the extra exon. OPN-5e lacks exon 5. Adopted from Bastos et al, Intl J of Mol Sci, 24, 2023 with modification.
Figure 2.
Schematic illustration of the OPN gene and its splice variants. Alternative splicing of OPN transcript results in five splice variants, which are denoted as OPN- a, OPN-b, OPN-c, OPN-4, and OPN-5. OPN-a is a full-length variant that consists of 7 exons; OPN-b lacks exon 5; while in OPN-c exon 4 is absent. In OPN-4, both exon 4 and exon 5 are missing; whereas OPN-5 is the longest variant which consists of an additional exon, generated from a portion of intron 3. Additionally, four new sub-variants of OPN-5 (OPN-5b, OPN-5c, OPN- 5d, OPN-5e) have been identified. OPN-5a is the same as OPN-5; OPN-5b has the extra shortened exon while in OPN-5c has additional 9 base pairs in the 3’ region of extra exon. In OPN-5d, there is deletion of exon 5 with the addition of 9 base pairs in the 3’ region of the extra exon. OPN-5e lacks exon 5. Adopted from Bastos et al, Intl J of Mol Sci, 24, 2023 with modification.
Figure 3.
Role of OPN in the regulation of various signalling pathways. OPN through its interaction with αvβ3, α4β1, α3β2, and α9β1 integrins, and CD44 receptor transduces multiple signalling pathways and their cross-talks such as FAK/MEK/ERK, PKCγ. /PKC/PI3K/Akt/mTOR, NIK/IBα/NFκB, JAK/STAT3, PI3K/Akt/β-catenin, NFκB/HIF1α/BMI1, c-Src/EGFR/MEK/ERK and MAPK pathways. These signalling cascades induce the activation of various tumor-promoting genes such as VEGF, MMPs, and COX-2, thereby inducing the tumor growth at the primary sites, angiogenesis, metastases at the distance sites, ECM remodeling, immune suppression, stemness, immune evasion, chemoresistance, migration, and survival. Adopted from Han et al, Oncology Letters, 17, 2019 and Ahmed et al EOTT, 15 (9), 2011 with modifications.
Figure 3.
Role of OPN in the regulation of various signalling pathways. OPN through its interaction with αvβ3, α4β1, α3β2, and α9β1 integrins, and CD44 receptor transduces multiple signalling pathways and their cross-talks such as FAK/MEK/ERK, PKCγ. /PKC/PI3K/Akt/mTOR, NIK/IBα/NFκB, JAK/STAT3, PI3K/Akt/β-catenin, NFκB/HIF1α/BMI1, c-Src/EGFR/MEK/ERK and MAPK pathways. These signalling cascades induce the activation of various tumor-promoting genes such as VEGF, MMPs, and COX-2, thereby inducing the tumor growth at the primary sites, angiogenesis, metastases at the distance sites, ECM remodeling, immune suppression, stemness, immune evasion, chemoresistance, migration, and survival. Adopted from Han et al, Oncology Letters, 17, 2019 and Ahmed et al EOTT, 15 (9), 2011 with modifications.
Figure 4.
Diagrammatic representation of multifaceted function of OPN in various tumors. OPN regulates the epithelial to mesenchymal transition (EMT) resulting in the loss of tight junctions, thereby enhancing metastasis, with the high expression of vimentin, slug, snail and fibronectin. Under hypoxic conditions, OPN induces activation of PI3K leading to phosphorylation of Akt, thereby upregulating VEGF dependent angiogenesis. OPN is responsible for metabolic function by activating HIF1α, under hypoxic conditions which further aids in the glycolytic process with high expression of VEGF, PDK1, LDHA, iNOS, GLUT1 and GLUT3. OPN further regulates CSC enrichment by activating a cascade of signalling pathways involving PI3K/Akt/mTOR, Hedgehog, MAPK, Wnt/β-catenin, JAK/STAT and Notch signalling. OPN- activated signalling pathways result in drug resistance. The interaction between OPN, αvβ3, and CD44 results in the activation of PI3K/Akt, FAK/MEK/ERK, EGFR, and Wnt/NFκB signalling cascades thereby aiding therapeutic resistance.
Figure 4.
Diagrammatic representation of multifaceted function of OPN in various tumors. OPN regulates the epithelial to mesenchymal transition (EMT) resulting in the loss of tight junctions, thereby enhancing metastasis, with the high expression of vimentin, slug, snail and fibronectin. Under hypoxic conditions, OPN induces activation of PI3K leading to phosphorylation of Akt, thereby upregulating VEGF dependent angiogenesis. OPN is responsible for metabolic function by activating HIF1α, under hypoxic conditions which further aids in the glycolytic process with high expression of VEGF, PDK1, LDHA, iNOS, GLUT1 and GLUT3. OPN further regulates CSC enrichment by activating a cascade of signalling pathways involving PI3K/Akt/mTOR, Hedgehog, MAPK, Wnt/β-catenin, JAK/STAT and Notch signalling. OPN- activated signalling pathways result in drug resistance. The interaction between OPN, αvβ3, and CD44 results in the activation of PI3K/Akt, FAK/MEK/ERK, EGFR, and Wnt/NFκB signalling cascades thereby aiding therapeutic resistance.
Figure 5.
Myofibroblast differentiation of resident fibroblasts and MSCs by tumor derived OPN. Tumor-derived OPN is involved in the transition of resident fibroblasts and MSCs into myofibroblast or CAFs. CAF-derived factors induce ECM deposition, EMT, angiogenesis, CSC enrichment, metabolic reprogramming, and tumor survival, resulting in the enhancement of tumor progression.
Figure 5.
Myofibroblast differentiation of resident fibroblasts and MSCs by tumor derived OPN. Tumor-derived OPN is involved in the transition of resident fibroblasts and MSCs into myofibroblast or CAFs. CAF-derived factors induce ECM deposition, EMT, angiogenesis, CSC enrichment, metabolic reprogramming, and tumor survival, resulting in the enhancement of tumor progression.
Figure 6.
Model depicting the role of OPN in tumor immune microenvironment (TIME): A. Role of OPN in shaping immunosuppressive TME: Tumor-derived OPN activates stromal cells by trans-differentiation of fibroblasts to myofibroblasts resulting in expansion of tumor. OPN- regulated PD-1/PDL1 interaction inhibits T-cell activation. Further, tumor cells induce lower expression of IRF8, resulting in overexpression of OPN thus leading to T-cell suppression. OPN via the NF-κB pathway, upregulates PD-L1 expression aiding in immune therapy escape. OPN induces the polarization of macrophages and the recruitment of monocytes. OPN activates TAM leading to angiogenesis, metastasis and enrichment of cancer stem cells via upregulation of various tumor-promoting factors like MMP-9, N-Cadherin, Vimentin, ICAM-1, COX-2, PGE-2, VEGF, Sox-2, Oct-3/4, Nanog and ALDH. The polarity ratio of CXCL9 and OPN (SPP1) determines the anti- and pro-tumorigenic properties of TAMs. B. Involvement of OPN in tumor immunity continuum: OPN is primarily associated in inflamed and immune excluded tumors whereas its role in immune dessert tumor is remains elusive. C. Schematic representations to identify OPN regulated immune cell heterogeneity in cancer: OPN regulated immune modulatory genes may be identified in TIME by CRISPR technology in breast cancer using scRNA seq. based platform.
Figure 6.
Model depicting the role of OPN in tumor immune microenvironment (TIME): A. Role of OPN in shaping immunosuppressive TME: Tumor-derived OPN activates stromal cells by trans-differentiation of fibroblasts to myofibroblasts resulting in expansion of tumor. OPN- regulated PD-1/PDL1 interaction inhibits T-cell activation. Further, tumor cells induce lower expression of IRF8, resulting in overexpression of OPN thus leading to T-cell suppression. OPN via the NF-κB pathway, upregulates PD-L1 expression aiding in immune therapy escape. OPN induces the polarization of macrophages and the recruitment of monocytes. OPN activates TAM leading to angiogenesis, metastasis and enrichment of cancer stem cells via upregulation of various tumor-promoting factors like MMP-9, N-Cadherin, Vimentin, ICAM-1, COX-2, PGE-2, VEGF, Sox-2, Oct-3/4, Nanog and ALDH. The polarity ratio of CXCL9 and OPN (SPP1) determines the anti- and pro-tumorigenic properties of TAMs. B. Involvement of OPN in tumor immunity continuum: OPN is primarily associated in inflamed and immune excluded tumors whereas its role in immune dessert tumor is remains elusive. C. Schematic representations to identify OPN regulated immune cell heterogeneity in cancer: OPN regulated immune modulatory genes may be identified in TIME by CRISPR technology in breast cancer using scRNA seq. based platform.
Figure 7.
OPN targeted novel therapeutic strategies. Small molecule inhibitors (andrographolide, curcumin, etc), OPN-specific siRNA, miRNA, shRNA, OPN aptamers, synthetic RGD peptides, OPN neutralizing antibodies, CD44 or integrin blocking antibodies, have been used recently as therapeutic approach to target OPN-integrin/CD44 axis which leads to downregulation of various oncogenic molecules and suppression of tumor progression by disrupting OPN regulated various signalling pathways in cancers.
Figure 7.
OPN targeted novel therapeutic strategies. Small molecule inhibitors (andrographolide, curcumin, etc), OPN-specific siRNA, miRNA, shRNA, OPN aptamers, synthetic RGD peptides, OPN neutralizing antibodies, CD44 or integrin blocking antibodies, have been used recently as therapeutic approach to target OPN-integrin/CD44 axis which leads to downregulation of various oncogenic molecules and suppression of tumor progression by disrupting OPN regulated various signalling pathways in cancers.
Table 1.
Expression of OPN in various cancers.
Table 1.
Expression of OPN in various cancers.
Table 2.
OPN as early prognostic & diagnostic biomarker in various cancers.
Table 2.
OPN as early prognostic & diagnostic biomarker in various cancers.