Exosomal integrins in tumor progression, treatment and clinical prediction (Review)
- Authors:
- Published online on: November 13, 2024 https://doi.org/10.3892/ijo.2024.5706
- Article Number: 118
-
Copyright: © Shen et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
Abstract
Introduction
Integrins are important cell surface adhesion receptors that bind to both extracellular matrix (ECM) ligands and cell surface ligands (1). As pivotal signaling molecules, integrins mediate cell migration and adhesion (2,3). Previous reports have shown that integrins are highly expressed in a variety of cancer types including lung cancer, hepatocellular carcinoma, pancreatic cancer and head and neck squamous cell carcinomas, and serve crucial roles in nearly every stage of cancer progression, including the initiation of primary tumor formation, subsequent growth and the metastatic cascade (4-8). Integrin-mediated sensing, stiffening and remodeling of the tumor stroma are key steps in supporting tumor cell invasion, acquiring cancer stem cell (CSC) characteristics and developing drug resistance during cancer progression (9,10). In addition, integrins have emerged as attractive targets for both predicting cancer prognosis and devising therapeutic strategies (11). Several integrin inhibitors, which disrupt the interaction between integrins and their respective ligands, hold notable therapeutic promise (12).
Exosomes are extracellular vesicles (EVs) secreted by cells, which serve a crucial role in regulating intercellular transport. Specifically, exosomes influence the state of both adjacent and distant cells by delivering nucleic acids, proteins and lipids (13). Tumor-derived exosomes (TDEs) are particularly important in angiogenesis, immune system regulation and the remodeling of surrounding tissues, thereby supporting tumor progression and metastasis to organs (14). As intercellular messengers, exosomes reflect the physiological state of various tumor cells, thus serving as biomarkers for clinical diagnosis and evaluation (15). Integrins have been identified as important components of exosomes. Accumulating evidence indicates that exosomal integrins assist in exosome homing, signal transduction and the phenotypic transformation of recipient cells (16-18). Research on exosomes and integrins has been steadily advancing. However, there remains a lack of comprehensive reviews summarizing the role of exosomal integrins in tumor development. Exploring the role of exosomal integrins in tumors will enhance the understanding of tumor pathogenesis and help to identify new diagnostic and therapeutic strategies (10). In the present review, the roles of exosomal integrins in tumor migration, tumorigenesis [including epithelial-mesenchymal transition (EMT)], angiogenesis, formation of the pre-metastatic niche (PMN) and the development of tumor drug resistance were summarized. Additionally, the potential of exosomal integrins in tumor prediction and treatment were examined, which highlighted their prospects in improving the efficiency of these aspects.
Exosome biogenesis, composition and function
Cells release various types of EVs, including exosomes, microvesicles and apoptotic bodies. Exosomes are the smallest entities among EVs, with a diameter ranging 40-160 nm (average, ~100 nm), are secreted by almost all cell types and have been found in all biological fluids (19). The contents of exosomes vary and reflect the composition of the donor cells (20,21). Exosomes contain adhesion molecules, tetraspanins, major histocompatibility complex molecules, transmembrane proteins, cytosolic (such as heat shock proteins, cytoskeletal proteins and transporters) and nuclear proteins, lipids, nucleic acids (including DNA, mRNA and non-coding RNAs), amino acids and metabolites (such as bioactive lipids, glycosidases and nucleotides) (Fig. 1) (22,23).
The formation and secretion of exosomes is a complex process, which is typically divided into two pathways: The endosomal sorting complex required for transport (ESCRT)-dependent pathway and the ESCRT-independent pathway (24). The ESCRT system is a molecular machine that completes endosomal membrane invagination to form multivesicular bodies (MVBs) in eukaryotic cells. The ESCRT-dependent pathway involves a series of complexes such as ESCRT-0, -I, -II and -III (25). ESCRT-0 is responsible for the recognition of mono-ubiquitinated proteins. Then, ESCRT-I and -II bind to ESCRT-0 to induce endosomal membrane budding. Finally, ESCRT-Ⅲ aggregates at the bud neck to pinch off the membrane, thereby releasing intraluminal vesicles (ILVs) into the lumen to form the MVBs (24,26). This process includes three steps: i) Extracellular substances, such as lipids, proteins and metabolites, enter cells through the initial plasma membrane invagination to form early-sorting endosomes (ESEs); ii) ESEs mature into late-sorting endosomes and then form MVBs. MVBs contain ILVs regulated by the ESCRT complex, which are the precursors to exosomes; and iii) MVBs either fuse with the plasma membrane to release exosomes or merge with autophagosomes and are degraded in the lysosomes (Fig. 1) (23,27). Studies have shown that, after the components of the ESCRT complex are depleted, exosome production is not completely blocked and a small number of exosomes are still formed (28,29). This suggests that exosomes can form in a manner independent of the ESCRT pathway. In the ESCRT-independent mechanism, the release of exosomes is dependent on sphingomyelinase. This involves the hydrolysis process of sphingomyelins into ceramides via neutral sphingomyelinase, which promotes inward budding of vesicles (30).
Exosomes carry a variety of bioactive molecules that have key roles in physiological and pathological processes through precise and dynamic intercellular communication. These processes include immune responses and infections, metabolic diseases, cardiovascular diseases, neurodegenerative diseases and cancer (31,32). Particularly, exosomes have gained attention in the field of cancer biology. Exosomes alter the fate of both the recipient and exosome-releasing cells through autocrine and paracrine signaling pathways (33). TDEs affect tumor growth, metastasis and drug resistance by interacting with tumor and stromal cells (34). Tumor stromal cells are primarily cancer-associated fibroblasts (CAFs) and immune cells (35). Exosomes have been recognized as crucial mediators in regulating the extracellular communication and metabolic reprogramming between CAFs and cancer cells (36,37). Furthermore, TDEs remodel the distant microenvironment at metastatic sites via blood and lymphatic circulation (38). Accumulating evidence indicates that TDEs are associated with angiogenesis and ECM remodeling in the tumor microenvironment (TME) (39-41).
Structure and function of integrin and exosomal integrin
Integrins are cell adhesion molecules that serve as membrane protein complexes, linking the ECM to the cytoskeleton, and regulate various cell behaviors, such as adhesion, proliferation and apoptosis, by triggering signal transduction through their extracellular connections to the cytoskeleton. This regulation provides the impetus and direction for cell migration and invasion (42,43). Integrins are ubiquitous in mammals, chickens, zebrafish and lower eukaryotes (44). In mammals, integrins are composed of α and β subunits bound by non-covalent bonds. To date, 18 α subunits and 8 β subunits have been identified, forming 24 αβ heterodimeric integrins with different properties and tissue distributions based on the various combinations. Both the α and β subunits are type I transmembrane proteins, each containing ectodomains, transmembrane domains and cytoplasmic domains (45,46). The integrin ectodomains are responsible for interacting with integrin ligands of the ECM (47). Due to the different characteristics of the integrin-ligand combinations, integrins can be clustered into four classes: Arginine-glycine-aspartate (RGD)-binding integrins (including αvβ1, αvβ3, αvβ5, αvβ6, αvβ8, α5β1, α8β1 and αIIbβ3) (48), leukocyte cell-adhesion integrins (including α4β1, α9β1, αLβ2, αMβ2, αXβ2, αDβ2, α4β7 and αEβ7) (49), collagen-binding integrins (which recognize the GFOGER binding site, including α1β1, α2β1, α10β1 and α11β1) (50) and laminin-binding integrins (including α3β1, α6β1, α6β4 and α7β1) (Fig. 2) (51,52).
Integrin-mediated transmembrane signals can exert their role through conformational transition. These transitions strictly regulate the transformation of integrins from a low-affinity state to a high-affinity state (53). Proteins such as talin, kindlin and tetraspanin trigger integrins to adopt an active open conformation, while integrin cytoplasmic domain-associated protein-1 and SHARPIN stabilize and inactivate integrins (54). Integrin-mediated transmembrane signals are bidirectional. During 'outside-in' signaling, integrins exposed to exosomes recognize and bind to their specific ligands on the surface of the target cells, generating intracellular signals. These signals control a series of physiological functions, including regulating cell polarity, altering the cytoskeleton structure, as well as cell survival and proliferation (55). The affinity of cells to extracellular ligands is regulated by 'inside-out' signaling. As this affinity increases, the interaction between integrins and the ECM becomes strong enough to induce cells to migrate and the ECM to remodel (Fig. 2) (56).
Integrins are expressed on the surface of tumor cells. The expression of integrins αvβ3, αvβ5, α5β1, α6β4, α4β1 and αvβ6 on tumor cells may have an important role in the progression of lung, breast, prostate, pancreatic and colorectal cancers (4,57). The abnormal expression of integrins is involved in almost every stage of cancer development, from the formation of primary tumors to the establishment of metastatic niches (9). During tumor cell survival and proliferation, integrin β1 promotes the survival of cancer cells by activating different cell signaling proteins and enhances cell proliferation by phosphorylating focal adhesion kinase (FAK) (58). Additionally, integrin αvβ3 enhances the migration and invasion of non-small cell lung cancer (NSCLC) cells by triggering the FAK signaling pathway (59). A specific integrin subtype, αvβ6, is expressed in some types of malignant tumors, such as prostate, breast, colorectal and lung cancers, but not in normal epithelial cells or benign tumors (60). Furthermore, αvβ6 confers an invasive/metastatic phenotype to early colorectal cancer cells, promoting tumor metastasis and reducing patient survival (61). Integrins regulate the adhesion of tumor cells and the expression level of integrin is proportional to the adhesion (62). In the early stage of tumorigenesis, the expression of integrins is reduced, which is conducive to the growth and spread of tumors. Then, the tumor cells enter the blood circulation (63). Thereafter, the expression of integrin is increased, which is beneficial to the adhesion of tumor cells to the vascular endothelium. Specifically, integrins are the main adhesion molecule in the angiogenesis stage of tumorigenesis. Furthermore, integrins are expressed in the cavity and luminal surface of vascular endothelial cells, mediating endothelial cell migration and capillary lumen formation (64).
Exosomal integrins are a subset of integrins. Thus, cell membrane and exosomal integrins share similarities in that both are involved in cell-to-cell or cell-to-exosome communication. Currently, the understanding of the communication mechanism of exosomal integrins is still in its infancy. However, integrin function is considered to be regulated by talin in exosomes, as in other cellular systems (54). In addition to certain similarities in the communication mechanism, there are notable differences in the location and function of cell membrane and exosomal integrins (64,65). As aforementioned, cell membrane integrins mediate cell-ECM adhesion. In the occurrence, development and metastasis of tumors, this adhesion directly affects the physiological processes of tumor cell migration, proliferation and survival. By contrast, exosomal integrins are embedded in the membrane of exosomes, which are small EVs that promote intercellular communication. These exosomal integrins serve a crucial role in targeting specific cells or tissues to achieve long-term signal transduction in processes such as cancer metastasis and immune regulation (16,65). Exosomal integrins also serve an important role in directing the tissue distribution of exosomes, thereby supporting long-range cellular interactions (16,66). In addition, exosomal integrins are involved in multiple steps of tumor formation as they enhance cell adhesion and migration, participate in PMN formation, and they modulate angiogenesis (54). It has been reported that exosomal integrins also confer drug resistance to tumors and impair drug efficacy (67).
Exosomal integrins in tumorigenesis and metastasis
Exosomal integrins in the TME
The TME is comprised of primary tumor lesions and their surrounding cellular and non-cellular components (68). Key features of the TME include low oxygen and nutritional levels, as well as an acidic environment. Cancer cells become increasingly invasive in such conditions, affecting tumor development and metastasis (69). TDEs, carrying molecules such as oncoproteins, lipids and various types of RNA (such as microRNA, mRNA and long non-coding RNA), induce changes in the TME phenotype (70). The cellular components of the TME include stromal cells (such as CAFs, mesenchymal stromal cells and pericytes) and immune cells [such as T and B lymphocytes, natural killer cells and tumor-associated macrophages (TAMs)] (71,72). CAFs serve a notable role in altering tumor mechanisms and are considered the most effective cells for the deposition and remodeling of the TME (73). TDEs induce the differentiation of fibroblasts (36). Integrin β4-overexpressing triple-negative breast cancer cells transfer integrin β4 protein to CAFs via exosomes, promoting cancer progression (74). In the lung metastasis niche of hepatocellular carcinoma (HCC), highly metastatic HCC cells secrete exosomal microRNA (miR)-1247-3p, activating the β1-integrin-NF-κB signaling pathway and transforming fibroblasts into CAF (75). In addition, macrophages are abundant in the TME, including both M1 and M2 polarized macrophages. TAMs are considered to promote tumor invasion (76,77). Exosomal integrin αvβ3 secreted by M2-like macrophages triggers the FAK signaling pathways in recipient cells and confers migration and invasion capabilities to NSCLC cells (59). The non-cellular components of the TME mainly include the ECM, including collagen and fibronectin, in which the ECM provides a biological scaffold for mechanical support (77). Integrin signal transduction drives intracellular signaling pathways through the interaction between cells and the ECM. Integrins are critical for cell anchorage to the ECM. Fibronectin matrix assembly is an integrin-dependent process. Integrin α5β1 induce initial fibronectin fibrillogenesis by transmitting cytoskeleton-generated tension to extracellular fibronectin molecules (78). Integrins α1β1, α2β1, α10β1 and α11β1 can bind to collagen (79). In the basement membrane, α3β1, α6β1 and α6β4 integrins promote epithelial cell adhesion by recognizing the COOH-terminal globular domain of the laminin α subunit (80). Integrin αvβ3 and VEGF have synergistic signaling outputs during endothelial cell activation and angiogenesis, induced by the interaction of VEGF and ECM molecules (81). Based on the essential role of integrins in cells adhesion to the ECM, it could be suggested that tumor exosomal integrins serve a notable role in the TME by facilitating adhesion to adjacent cells, exosomes and the ECM. Abnormal adhesion functions can lead to diseases, including cancer progression (82). In summary, integrins, including exosomal integrins, affect the TME by regulating cell signaling pathways and interactions with surrounding matrices (Fig. 3A).
Exosomal integrins in EMT
Epithelium-derived tumor cells undergo a complex process termed EMT (83) to enhance their ability to disseminate from the original site, metastasize and invade other sites (84,85). EMT is a cellular developmental biology procedure that occurs during embryogenesis (83). Several key proteins, including E-cadherin, vimentin and N-cadherin, are involved in the EMT process (86). During EMT, epithelial cells lose their polarity and acquire mesenchymal characteristics by downregulating E-cadherin and upregulating vimentin and N-cadherin (87). Consequently, epithelial cells become invasive and attain stem cell-like properties (88). When hijacked by cancer cells, this process endows normal epithelial cells with malignant characteristics. CSCs, which possess stemness characteristics, have the initial capability for tumor metastasis and colonization. In fact, most cells maintain this ability through EMT (89). TDEs stimulate an EMT program through autocrine and paracrine signals within the tumor ecosystem (90). It has been demonstrated that diverse signaling pathways such as TGF-β, WNT, Notch and growth factor receptor tyrosine kinases induce the occurrence of EMT (91).
It has been shown that integrins serve an important role in regulating EMT and cancer stemness (89). Notably, integrins and TGF-β effectively synergistically induce the aberrant expression of EMT transcription factors, such as zinc finger E-box-binding homeobox 1 and snail2 (92,93). In a previous study, exosomes loaded with integrin β-like 1 (ITGBL1) from primary colorectal cancer (CRC) cells can convert fibroblasts in distal organs into CAFs by activating the TNFα-induced protein 3-mediated NF-κB signaling pathway. In this study, ITGBL1 overexpression enhanced the secretion of IL-6 and IL-8 from fibroblasts or stellate cells, thereby promoting the stemness and EMT of CRC cells (94). Additionally, CAF-derived IL-32 binds to integrin β3, thereby activating intracellular p38 MAPK signaling in breast cancer cells. This signaling increases the expression of EMT markers (including fibronectin, N-cadherin and vimentin) and promotes tumor cell invasion (95). These factors drive tumor cells to undergo EMT by various signaling pathways such as TGF-β, PI3K/AKT, MAPK and NF-κB (Fig. 3B) (52,96).
Exosomal integrins in tumor immunity
Previous studies have shown that TDEs have a pivotal role in regulating the TME by inducing immune suppressor cells and enabling cancer cells to evade immune effector cells (90,97). The antitumor ability of the human immune system is inhibited by TDEs through inflammatory signaling pathways (71). Exosomes secreted by cancer and immune cells deliver protein cargo similar to that of the primary tumor cells to specific tissues of the homing niche and alter the gene expression and molecular structure of the homing niche. Integrins regulate the tissue-specific homing pattern of exosomes (16). TDEs initiate immunosuppressive mechanisms at metastatic niches by triggering immune suppressor cells such as myeloid-derived suppressor cells, regulatory T cells (Tregs), tumor-associated neutrophils and TAMs (98).
Integrins on tumor cells are involved in the suppression of antitumor immunity throughout various stages of tumor formation and metastasis (99,100). Interferon (IFN)-γ-producing immune cells, mediated by α4β7, are recruited to CRC tissues where they exert an effective antitumor immune response (101). Immunologic targeting of integrin β4 significantly inhibited local tumor growth and metastases in both 4T1 mammary tumors and SCC7 head and neck squamous carcinoma models (102). Integrin αvβ3 regulates IFN-induced PD-L1 expression. In a mouse model, silencing αvβ3 expression reduced IFN-induced STAT1 phosphorylation, decreased PD-L1 expression and inhibited tumor growth (103). In addition, overexpression of integrin α2 increased the phosphorylation level of STAT3 in tumor cells to initiate PD-L1 transcription and thus upregulate PD-L1 expression (104). Integrin also regulates the activation of transforming growth factor-β (TGF-β) in immune cells, which may be another mechanism of tumor immune escape. In mouse melanoma and breast cancer models, Tregs (which express integrin αvβ8) are the predominant cell type that activate TGF-β produced by cancer cells (105). The activated TGF-β then protects the tumor from T cell attack by binding to and releasing αvβ8 on tumor cells or latent immune cells, thereby preventing T cell penetration into the tumor (106). The innate nature of integrins from cells to exosomes has implications for tumor immunity. Based on the inherent nature of exosomal integrins derived from tumor cell integrins, it is reasonable to speculate that exosomal integrins have similar tumor immune capabilities as tumor cell integrins. Indeed, chronic inflammation is known to contribute to cancer metastasis (107). Thus, integrin-guided preferential distribution of TDEs determines which specific organs may encounter TDE-mediated initiation of inflammation (108). Exosomal integrins not only target ECM proteins in distant tissues but are also delivered to the target cells themselves where they activate Src kinase signaling, leading to induction of the proinflammatory S-100 gene (16). The S100 proteins S100A8 and S100A9 are important mediators of various processes during chronic inflammation. The S100A8/9 proteins stimulate infiltration of inflammatory lesions by activated myeloid cells and are involved in leukocyte adhesion and migration (109). A tumor-bearing mouse model demonstrated that S100A8/A9 proteins participate in the activation and accumulation of MDSC cells during the induction of cancer T cell tolerance (110). S100A8/A9 proteins activate the NF-κB pathway in a positive feedback manner and ensure that the protein expression levels of S100A8/A9 are sufficient to maintain the immunosuppressive function of MDSC in the inflammatory tumor microenvironment (111). Therefore, the role of exosomal integrins in tumor immunosuppression underscores the potential of integrin-targeted immunotherapy (Fig. 3C).
Exosomal integrins in angiogenesis, vascular permeability and hematogenous
Tumor metastasis refers to the process by which tumor cells intravasate into blood vessels and lymphatic vessels from the primary tumor site. Tumor angiogenesis is a complex biological process involving several key steps, including local damage to the basement membrane in the tissue, endothelial cell migration activated by angiogenic factors and endothelial cell proliferation and stability. VEGF, fibroblast growth factor (FGF) and other angiogenic signals are key factors regulating the angiogenic process (112). Furthermore, exosomes derived from various human tumor cell lines or plasma are effective inducers of angiogenesis, especially under hypoxic conditions, by modulating endothelial cell properties to promote angiogenesis (113). Exosomes expressing tetraspanins can promote tumor growth by increasing angiogenesis. For instance, TDEs enriched with tetraspanin 8 and integrin α4 enhanced endothelial cell proliferation and angiogenesis in rat pancreatic cancer by upregulating angiogenesis-related genes through endothelial-exosomal interactions (114). With improved understanding of exosome heterogeneity, it is appealing to focus on the role of exosomal integrins in influencing endothelial metabolism and angiogenesis. The effect of exosomal integrins on the angiogenic potential of endothelial cells has also been demonstrated in prostate cancer (PrCa) progression. PrCa exosomes promote angiogenesis by transferring exosomal integrin αvβ6 to endothelial cells that do not typically express epithelial-specific integrin αvβ6. Exosomal integrin αvβ6 uptake is associated with an upregulation of the pro-angiogenic survivin levels and a downregulation of the angiogenic inhibitory phosphorylated STAT1 in endothelial cells (115). The increased expression of integrin αvβ3 during angiogenesis in lung, colon, pancreatic and breast cancer also suggests that integrins are involved in tumor angiogenesis (62).
In the TME, EMT endows endothelial cells and cancer cells with invasive capabilities, allowing cancer cells to traverse the matrix with the assistance of TAM-derived VEGFA. This leads to an abnormal increase in vascular permeability and the simultaneous intravasation of tumor cells (116). β1 integrins can affect vascular permeability, especially during inflammation, as the recruitment of circulating cells to inflamed tissues involves recognition of cell adhesion molecules (117). Specifically, integrin β1 can affect the ability of circulating cells to block, adhere and extravasate at sites of injury and vascular permeability (118). Integrin α4β1 serves a role in the homing of circulating progenitor cells to tumor neovascularization that expresses vascular cell adhesion protein 1 and cellular fibronectin (119). Additionally, integrin αvβ3 is required for angiogenesis induced by basic FGF or TNF-α, while αvβ5 is required for angiogenesis induced by VEGF, TGF-α or phorbol ester (120).
Hematogenous dissemination is often the primary mechanism of distant metastasis, leading to the implantation of tumor cells into distant organs through extravasation, ultimately forming micro and macro tumor metastases (121). Although primary tumors can shed millions of cells into the blood vessels every day, a very small number of circulating tumor cells (CTCs) eventually reach distant organs (122). The permeability of blood vessels increases the possibility of tumor cells and TDEs entering the blood. Millions of exosomes are secreted by primary tumors into the blood vessels every day, and are then transferred to specific organs via the vascular pathway by exosomal integrins (16) (Fig. 3D).
Exosomal integrins in organotropism and the establishment of PMNs
The combined systemic effects of tumor-secreted factors and tumor-shed extracellular vesicles induces a receptive tissue microenvironment from a distance. The formation of the microenvironment is initiated with local changes such as the induction of vascular permeability, remodeling of stroma and extracellular matrix, followed by systemic effects on the immune system. These microenvironments are termed PMNs. The presence of a PMN means that metastasis to specific organs is not random but predictable (123). Clinical cases have revealed that metastatic organ patterns follow particular rules, with certain tumors preferentially metastasizing and colonizing specific organs. Breast and prostate cancers preferentially metastasize to bone (124). However, colorectal and pancreatic cancers preferentially colonize the liver and lung (125). The mechanisms directing tumor cells to specific distant organs have long puzzled researchers, and the precise mechanisms remain largely unknown. However, Hoshino et al (16) confirmed that the key reason lies in exosomal integrins. The role of exosomes and exosomal integrins in tumor growth and metastasis has been emphasized. Exosomal integrins can be localized to specific organs. After target cells at the metastatic site ingest these exosomes, the PMN is established by activating Src phosphorylation and pro-inflammatory S-100 expression.
Studies have shown that PMN formation is a chronological event that precedes the arrival and colonization of tumor cells, effectively initiating the target site of metastasis. Soluble molecules secreted by the primary tumor have a crucial role in the formation of the PMN, promoting metastasis and even determining organ-specific sites of the metastasis (126,127). Hoshino et al (16) injected FM1-43 dye-labeled exosome isolated from organotropic human breast and pancreatic cancer cell lines, which predominantly metastasize to the lungs, into naive animals. Tumor FM1-43-labeled exosomes were then detected in pre-metastatic cells by electron microscopy, suggesting that tumor exosome uptake occurs at future metastatic sites. Tumor-derived secreted factors (TDSFs) and soluble molecular components, including EVs, secreted by primary tumors induce the mobilization and recruitment of multiple cell populations to secondary organ sites (123). Primary TDEs can promote the formation of the TME at secondary sites and guide bone marrow-derived dendritic cells (BMDCs) to form a pre-metastatic microenvironment. For instance, bone marrow-derived hematopoietic progenitor cells expressing VEGFR1 are mobilized and recruited to the PMN of the lungs (128). TDEs promote the formation of the PMN by inducing vascular remodeling, preparing for the arrival of CTCs, promoting the development of inflammation and recruiting BMDC (129). Exosomes from metastatic melanoma increase the metastasis of primary tumors by educating bone marrow progenitor cells via upregulation of the mesenchymal to epithelial transition factor receptor. In addition, melanoma-derived exosomes promote vascular leakage at the pre-metastatic site and reprogram bone marrow progenitor cells to adopt pro-angiogenic phenotypes (130). Macrophage inhibitory factor in pancreatic cancer cell exosomes induces the release of TGF-β, which in turn promotes the production of fibronectin. The deposition of fibronectin promotes the colonization of bone marrow-derived macrophages and neutrophils in liver metastasis (131).
Through quantitative mass spectrometry and western blotting analysis, it has been demonstrated that integrin α6, combined with integrins β4 and β1, is abundantly present in lung-tropic exosomes (16). Conversely, integrins β5 and αv are present in liver-derived exosomes and integrin β3 is predominant in brain-derived exosomes. Infrared imaging showed that integrins β4 and β5 are responsible for the specific uptake of exosomes by the liver and lung, respectively (16,132). In another study, integrin αvβ6 was encapsulated in exosomes isolated from PC3 and RWPE PrCa cell lines and was effectively transferred from donor cells to αvβ6-negative recipient cells, colonizing on their surfaces (133). PrCa is prone to distant metastasis, with bone metastasis being particularly significant and a primary cause of death in patients with PrCa (134). Among various integrins, αvβ3 has gained notable attention for its role in promoting bone metastasis through multiple regulatory mechanisms (135). Extracellular or membrane ligands (such as small integrin-binding ligand N-linked glycoproteins, connective tissue growth factor, cellular chemokines and ion channel proteins) combine with or activate αvβ3 to mediate PrCa bone metastasis. Moreover, the FAK, PI3K, ERK and αvβ3/RUNX2/RANKL intercellular signaling pathway is stimulated by αVβ3 and has been reported to be related to PrCa metastasis (136). The circulating exosomal integrin β3 level is associated with the survival rate and intracranial control after whole-brain radiotherapy in patients with brain metastasis from lung cancer, supporting the suggestion that exosomal integrin β3 mediates the pattern of brain metastasis (137). Additionally, integrins α3 and β1 are more abundant in urinary exosomes from patients with metastatic PrCa compared with those from individuals with benign prostatic hyperplasia or non-metastatic PrCa (138). In conclusion, exosomal integrins can be identified as determinants of metastatic organotropism (Fig. 3E).
Exosomal integrins in tumor drug resistance
Exosomes have a notable influence on drug resistance, which is induced through a variety of mechanisms. During chemotherapy, cancer cells cannot remove exosomes containing unfavorable biomolecules. However, drug-resistant cancer cells can load chemotherapy drugs into exosomes and expel them from tumor cells directly (139,140). Another mechanism involves exosomes carrying a drug-resistant phenotype from drug-resistant cancer cells to drug-sensitive cancer cells (141). Additionally, exosomes can regulate the transfer of functional proteins and/or miRNAs, contributing to drug resistance (142). The contents of exosomes can also cause immunosuppression, leading to drug resistance in HER2+ breast cancer (143,144). Therefore, the roles of exosomes and integrins in drug resistance across different cancer types are reviewed here (Table I).
In drug-resistant human ovarian cancer, cisplatin (CDDP) is encapsulated in exosomes and released within the secretory pathway (145). The ATP-binding cassette (ABC) transporter superfamily, including ABCB1 [also known as P-glycoprotein (P-gp)], ABCC1 (also known as multidrug resistance protein 1) and ABCG2 (also known as breast cancer resistance protein), function as multidrug resistance efflux transporters. These transporters, located on the exosomal membrane, actively pump anticancer drugs out of cells, resulting in chemoresistance (146). P-gp, a key anticancer pump transporter, plays a notable role in drug resistance by retaining drug concentrations in tumor cells below the therapeutic levels after chemotherapy, rendering the treatment ineffective. This process may be mediated by exosomal integrin, which facilitate the intercellular transfer of P-gp from multidrug-resistant cells to drug-sensitive cells (147). Suppression of αvβ6 downregulated the levels of MDR1 gene mRNA and P-gp. In particular, β6 shRNA-mediated silencing of the αvβ6 gene markedly decreased drug efflux ability (148). In addition, the delivery mechanisms of exosomes carrying nucleic acids and proteins are closely related to tumor drug resistance. For instance, the content of exosomes from paclitaxel-resistant ovarian cancer cells, namely miR-1246, induces chemoresistance by inhibiting 3′UTR caveolin-1, which directly suppresses the increase of p-gp expression (149).
The effect of adhesion crosstalk between tumor cells and stromal cells on the development of tumor drug resistance has been studied in detail. Cancer cells adhere to the ECM or stromal cells and can avoid being killed by radiotherapy and chemotherapy, which is known as cell adhesion-mediated drug resistance (CAM-DR) (150). CAM-DR is determined by the integrin-ECM interaction. For instance, integrin β1 is considered essential for radiotherapy resistance in human head and neck cancer (151) and mediates cell adhesion to the ECM. The role of integrins in tumor drug resistance is primarily related to integrin-mediated signaling pathways (152). For instance, the integrin β1/Src/AKT signaling pathway serves a key role in acquiring resistance to epidermal growth factor receptor-targeted anticancer drugs, such as gefitinib and erlotinib, in lung cancer (153). In human glioblastoma, resistance to temozolomide is mainly due to integrin α5β1 downregulating the p53 pathway (154). In breast cancer cells, integrin αvβ3 and its mediated FAK/PI3K/AKT signaling pathway are involved in CDDP resistance (155). Additionally, integrin α6 serves a key role in cancer drug resistance by regulating the MAPK/ERK and PI3K/AKT signaling pathways (156). Finally, integrin β4 and vinculin in exosomes are associated with taxane resistance in PrCa (157).
Exosomal integrins in the treatment and diagnosis of tumors
Exosomes are natural nano-biological delivery systems with properties of stability, biocompatibility (endogenous origin) and the ability to cross various physiological barriers. These features make exosomes promising carriers for delivering several drugs and biomacromolecules for cancer therapy (158). For instance, loading exosomes with paclitaxel has indicated the potential for delivering multiple chemotherapeutics to treat drug-resistant cancer (159). In cancer treatment, exosomes protect the integrity of nucleic acids and shield proteins from various enzymes and the immune system, making them excellent carriers for delivering these macromolecules in therapy. Kobayashi et al (160) demonstrated that loading miR-199a-3p into exosomes inhibited c-Met expression and reduced ovarian cancer cell proliferation, invasiveness and dissemination. Similarly, exosomes loaded with signal regulatory protein α (SIRPα) block the CD47 receptor on tumor cells more effectively when compared with ferritin-SIRPα, indicating that these exosomes have antitumor applications (161). In addition, since TDEs carry specific integrins on their surface, they ensure targeted delivery to specific organs and tissues. Exosome targeting was also accomplished by genetic modification of exosome donor cells (162). This characteristic enhances the accuracy and efficiency of delivering therapeutic contents through exosomes and exosomal integrins compared with other biological carriers (163). However, to meet the demands of large-scale clinical applications, the loading capacity and methods for exosomes require optimization.
The diagnosis of cancer is often invasive. However, non-invasive diagnostic methods in clinical oncology have emerged as feasible alternatives with the study of exosomes (164,165). Exosomes, which represent their source cells, contain biological information and are steadily secreted into body fluids, making them ideal specimens for liquid biopsy. More specifically, cancer biomarkers can be determined according to the characteristics of TDE contents (such as the proteins and nucleic acids) in blood, ascites or urine, as these exosomes contain information related to cancer progression (166). A study has shown that integrin β4 and vinculin levels in exosomes isolated from PC3 cells can serve as effective biomarkers for diagnosing PrCa associated with taxane resistance (157). The presence of integrin α2β1 in exosomes from tumor metastatic cells, but not in exosomes from non-cancerous WI-38 lung fibroblasts or epithelial MCF10A cells, indicates that this integrin can be regarded as a biomarker of metastasis (16).
Integrins have are promising, yet challenging, targets for the treatment of cancer. The specific expression of integrins in TDEs allows them to be used for disease monitoring, predicting patient survival and potentially distinguishing between cancer types and stages (167-169). For instance, integrin αvβ6, which is not expressed in normal adult epithelial cells but is present in cancer cells, can be utilized for the diagnosis and treatment of certain cancers such as ovarian, pancreatic, esophageal, bile duct, oral and cervical cancers (170). In mouse models, heavy lead peptides combined with αvβ6 have been used for non-invasive imaging, highlighting the potential of αvβ6 as a promising biomarker (171). Additionally, a patent states that the monoclonal antibody, 10D5, which specifically binds to β6, can be used to treat cancer (172). Invasive diagnostic techniques for multiple brain metastasis are often impractical in clinical settings, making it attractive to evaluate circulating EVs and related integrins as biomarkers. Experts isolated and quantified exosomal integrins of 75 patients with lung cancer with brain metastasis and analyzed the association of exosomal integrins with clinical factors, survival and intracranial or extracranial failure. Accordingly, it was proposed that integrin β3 may serve as a potential biomarker for the development of brain metastasis (137). Furthermore, the differential expression of exosomal integrins α6, αv and β1 is associated with the tumor stage of various epithelial cancers such as colon, lung, ovarian and prostate cancers (169).
Integrins are cell adhesion and signaling proteins present on the surface of various cell subsets, making them potential therapeutic targets. Numerous integrins involved in tumor progression have been studied as attractive therapeutic targets for cancer therapy (173,174). A study has shown that integrin antagonists, which currently include monoclonal antibodies, RGD peptide analogues and non-RGD antagonists, inhibit tumor growth by affecting tumor cells and tumor-associated host cells. For instance, integrin αvβ3 and αvβ5 inhibitors, such as cilengitide, have demonstrated this capability (132). Cilengitide can reduce the expression levels of integrin genes and inhibit the proliferation of tumor cells (175). In addition, cilengitide can activate αvβ3 and αvβ5 integrins of the FAK/Paxillin/AKT signaling pathway to combat chemotherapy resistance in glioblastoma (176). At present and to the best of our knowledge, there are seven drugs targeting integrins on the market: Abciximab, eptifibatide, tirofiban, natalizumab, vidolizumab, lifitegrast and carotegrast. However, drugs specifically targeting exosomal integrins have not yet been officially introduced to the market (52). The application of exosomes and integrins in cancer treatment and diagnosis is summarized in Table II.
Challenges and prospectives
The effectiveness of bodily fluid biopsy relies on advanced techniques for the isolation and characterization of exosomal integrins. Despite significant efforts in developing liquid biopsy methods and tumor biomarkers in oncology, only a few have progressed to the clinical stage. This is because clinical studies using liquid biopsy are often reliant on integrin content and heterogeneity, thereby rendering them more expensive and time-consuming compared with other common clinical testing techniques (177). To promote exosomal integrins as biomarkers to a broader population, more portable, efficient and accurate characterization techniques are needed (178). Standardization in the selection, separation, characterization, storage, management and quality control of exosomal integrins is crucial for their clinical application in tumor diagnosis and treatment. Most existing studies use tumor cell integrins as markers, but these are often affected by the complexity of tumors. Using specific integrins from TDEs in the humoral circulation as markers could reduce this interference.
Over the past 30 years, research on exosomal integrins as therapeutic targets has gained significant attention. However, most drugs have failed in phase III clinical trials, highlighting the challenges in translating experimental findings into clinical therapies (179). The pharmacodynamics and complex physiology of integrins contribute to the ongoing problems of toxicity and poor efficacy in drugs that have reached the market (180). Current treatment strategies mainly focus on interfering with integrin-ligand interactions. However, the class-specific nature of integrin-targeted therapy presents another challenge. Since the same integrin subunit can form different integrin heterodimers, the accuracy of targeted therapies is affected. Additionally, other molecules can interfere with targeted drugs. For instance, abciximab binds to both glycoprotein IIb/IIIa and integrin αvβ3 with a similar affinity, suggesting it may act as an antagonist of both GPIIb/IIIa and αvβ3 (181). Such complexities complicate the use of preclinical experimental data in developing effective therapies.
The heterodimeric structure of integrins enables them to specifically recognize amino-acid motifs. The binding of ligands to integrins can affect the allosteric states of integrins, thereby altering the movement of EVs. This suggests that exosomal integrins may act as sensors of the molecular environment. To effectively utilize exosomal integrins in cancer therapy, it is essential to fully understand their mechanisms of action in tumor development. Additionally, combining targeted drugs that act on ligands and integrins or their downstream effectors may offer a promising approach. This direction could lead to the development of new therapeutic strategies.
Availability of data and materials
Not applicable.
Authors' contributions
RX and XZ made significant contributions to the conception and design of the manuscript. YS, LS, and SW were responsible for the acquisition, analysis and interpretation of data. RX and YS undertook the editing, drafting and writing of the manuscript. Data authentication is not applicable. All authors have read and approved the final version of the 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.
Acknowledgments
Not applicable.
Funding
The present work was supported by the Research Fund of Anhui Institute of Translational Medicine (grant no. 2023zhyx-C90) and the Basic and Clinical Cooperative Research and Promotion Program of Anhui Medical University (grant no. 2023xkjT046).
References
Moreno-Layseca P, Icha J, Hamidi H and Ivaska J: Integrin trafficking in cells and tissues. Nat Cell Biol. 21:122–132. 2019. View Article : Google Scholar : PubMed/NCBI | |
Calderwood DA, Campbell ID and Critchley DR: Talins and kindlins: Partners in integrin-mediated adhesion. Nat Rev Mol Cell Biol. 14:503–517. 2013. View Article : Google Scholar : PubMed/NCBI | |
Grigoryeva ES, Tashireva LA, Savelieva OE, Zavyalova MV, Popova NO, Kuznetsov GA, Andryuhova ES and Perelmuter VM: The association of integrins β3, β4, and αVβ5 on exosomes, CTCs and tumor cells with localization of distant metastasis in breast cancer patients. Int J Mol Sci. 24:29292023. View Article : Google Scholar | |
Liu F, Wu Q, Dong Z and Liu K: Integrins in cancer: Emerging mechanisms and therapeutic opportunities. Pharmacol Ther. 247:1084582023. View Article : Google Scholar : PubMed/NCBI | |
Mammadova-Bach E, Zigrino P, Brucker C, Bourdon C, Freund M, De Arcangelis A, Abrams SI, Orend G, Gachet C and Mangin PH: Platelet integrin α6β1 controls lung metastasis through direct binding to cancer cell-derived ADAM9. JCI Insight. 1:e882452016. View Article : Google Scholar | |
Sun F, Wang J, Sun Q, Li F, Gao H, Xu L, Zhang J, Sun X, Tian Y, Zhao Q, et al: Interleukin-8 promotes integrin β3 upregulation and cell invasion through PI3K/Akt pathway in hepatocellular carcinoma. J Exp Clin Cancer Res. 38:4492019. View Article : Google Scholar | |
Reader CS, Vallath S, Steele CW, Haider S, Brentnall A, Desai A, Moore KM, Jamieson NB, Chang D, Bailey P, et al: The integrin αvβ6 drives pancreatic cancer through diverse mechanisms and represents an effective target for therapy. J Pathol. 249:332–342. 2019. View Article : Google Scholar : PubMed/NCBI | |
Gopal S, Veracini L, Grall D, Butori C, Schaub S, Audebert S, Camoin L, Baudelet E, Radwanska A, Beghelli-de la Forest Divonne S, et al: Fibronectin-guided migration of carcinoma collectives. Nat Commun. 8:141052017. View Article : Google Scholar : PubMed/NCBI | |
Hamidi H and Ivaska J: Every step of the way: Integrins in cancer progression and metastasis. Nat Rev Cancer. 18:533–548. 2018. View Article : Google Scholar : PubMed/NCBI | |
Raab-Westphal S, Marshall JF and Goodman SL: Integrins as therapeutic targets: Successes and cancers. Cancers (Basel). 9:1102017. View Article : Google Scholar : PubMed/NCBI | |
Hamidi H, Pietilä M and Ivaska J: The complexity of integrins in cancer and new scopes for therapeutic targeting. Br J Cancer. 115:1017–1023. 2016. View Article : Google Scholar : PubMed/NCBI | |
Slack RJ, Macdonald SJF, Roper JA, Jenkins RG and Hatley RJD: Emerging therapeutic opportunities for integrin inhibitors. Nat Rev Drug Discov. 21:60–78. 2022. View Article : Google Scholar | |
Pegtel DM and Gould SJ: Exosomes. Annu Rev Biochem. 88:487–514. 2019. View Article : Google Scholar : PubMed/NCBI | |
Kalluri R: The biology and function of exosomes in cancer. J Clin Invest. 126:1208–1215. 2016. View Article : Google Scholar : PubMed/NCBI | |
Zhou Y, Zhang Y, Gong H, Luo S and Cui Y: The role of exosomes and their applications in cancer. Int J Mol Sci. 22:122042021. View Article : Google Scholar : PubMed/NCBI | |
Hoshino A, Costa-Silva B, Shen TL, Rodrigues G, Hashimoto A, Tesic Mark M, Molina H, Kohsaka S, Di Giannatale A, Ceder S, et al: Tumour exosome integrins determine organotropic metastasis. Nature. 527:329–335. 2015. View Article : Google Scholar : PubMed/NCBI | |
Kalappurakkal JM, Anilkumar AA, Patra C, van Zanten TS, Sheetz MP and Mayor S: Integrin mechano-chemical signaling generates plasma membrane nanodomains that promote cell spreading. Cell. 177:1738–1756.e23. 2019. View Article : Google Scholar : PubMed/NCBI | |
Lu H, Bowler N, Harshyne LA, Craig Hooper D, Krishn SR, Kurtoglu S, Fedele C, Liu Q, Tang HY, Kossenkov AV, et al: Exosomal αvβ6 integrin is required for monocyte M2 polarization in prostate cancer. Matrix Biol. 70:20–35. 2018. View Article : Google Scholar : PubMed/NCBI | |
Welsh JA, Goberdhan DCI, O'Driscoll L, Buzas EI, Blenkiron C, Bussolati B, Cai H, Di Vizio D, Driedonks TAP, Erdbrügger U, et al: Minimal information for studies of extracellular vesicles (MISEV2023): From basic to advanced approaches. J Extracell Vesicles. 13:e124042024. View Article : Google Scholar : PubMed/NCBI | |
Février B and Raposo G: Exosomes: Endosomal-derived vesicles shipping extracellular messages. Curr Opin Cell Biol. 16:415–421. 2004. View Article : Google Scholar : PubMed/NCBI | |
Valadi H, Ekström K, Bossios A, Sjöstrand M, Lee JJ and Lötvall JO: Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol. 9:654–659. 2007. View Article : Google Scholar : PubMed/NCBI | |
Doyle L and Wang M: Overview of extracellular vesicles, their origin, composition, purpose, and methods for exosome isolation and analysis. Cells. 8:7272019. View Article : Google Scholar : PubMed/NCBI | |
Kalluri R and LeBleu VS: The biology, function, and biomedical applications of exosomes. Science. 367:eaau69772020. View Article : Google Scholar : PubMed/NCBI | |
Mashouri L, Yousefi H, Aref AR, Ahadi AM, Molaei F and Alahari SK: Exosomes: Composition, biogenesis, and mechanisms in cancer metastasis and drug resistance. Mol Cancer. 18:752019. View Article : Google Scholar : PubMed/NCBI | |
Hurley JH: ESCRT complexes and the biogenesis of multivesicular bodies. Curr Opin Cell Biol. 20:4–11. 2008. View Article : Google Scholar : PubMed/NCBI | |
Mathieu M, Martin-Jaular L, Lavieu G and Théry C: Specificities of secretion and uptake of exosomes and other extracellular vesicles for cell-to-cell communication. Nat Cell Biol. 21:9–17. 2019. View Article : Google Scholar : PubMed/NCBI | |
Tricarico C, Clancy J and D'Souza-Schorey C: Biology and biogenesis of shed microvesicles. Small GTPases. 8:220–232. 2017. View Article : Google Scholar : | |
Stuffers S, Sem Wegner C, Stenmark H and Brech A: Multivesicular endosome biogenesis in the absence of ESCRTs. Traffic. 10:925–937. 2009. View Article : Google Scholar : PubMed/NCBI | |
Theos AC, Truschel ST, Tenza D, Hurbain I, Harper DC, Berson JF, Thomas PC, Raposo G and Marks MS: A novel pathway for sorting to intralumenal vesicles of multivesicular endosomes involved in organelle morphogenesis. Dev Cell. 10:343–354. 2006. View Article : Google Scholar : PubMed/NCBI | |
Yang K, Fu W, Deng M, Li X, Wu M and Wang Y: The sphingolipids change in exosomes from cancer patients and association between exosome release and sphingolipids level based on a pseudotargeted lipidomics method. Anal Chim Acta. 1305:3425272024. View Article : Google Scholar : PubMed/NCBI | |
Guay C and Regazzi R: Exosomes as new players in metabolic organ cross-talk. Diabetes Obes Metab. 19(Suppl 1): S137–S146. 2017. View Article : Google Scholar | |
Quek C and Hill AF: The role of extracellular vesicles in neurodegenerative diseases. Biochem Biophys Res Commun. 483:1178–1186. 2017. View Article : Google Scholar | |
Li Y, Chen ZK, Duan X, Zhang HJ, Xiao BL, Wang KM and Chen G: Targeted inhibition of tumor-derived exosomes as a novel therapeutic option for cancer. Exp Mol Med. 54:1379–1389. 2022. View Article : Google Scholar : PubMed/NCBI | |
Ciardiello C, Cavallini L, Spinelli C, Yang J, Reis-Sobreiro M, de Candia P, Minciacchi VR and Di Vizio D: Focus on extracellular vesicles: New frontiers of cell-to-cell communication in cancer. Int J Mol Sci. 17:1752016. View Article : Google Scholar : PubMed/NCBI | |
Whiteside TL: Immune modulation of T-cell and NK (natural killer) cell activities by TEXs (tumour-derived exosomes). Biochem Soc Trans. 41:245–251. 2013. View Article : Google Scholar : PubMed/NCBI | |
Webber J, Steadman R, Mason MD, Tabi Z and Clayton A: Cancer exosomes trigger fibroblast to myofibroblast differentiation. Cancer Res. 70:9621–9630. 2010. View Article : Google Scholar : PubMed/NCBI | |
Rai A, Greening DW, Chen M, Xu R, Ji H and Simpson RJ: Exosomes derived from human primary and metastatic colorectal cancer cells contribute to functional heterogeneity of activated fibroblasts by reprogramming their proteome. Proteomics. 19:e18001482019. View Article : Google Scholar | |
Cueni LN and Detmar M: The lymphatic system in health and disease. Lymphat Res Biol. 6:109–122. 2008. View Article : Google Scholar : PubMed/NCBI | |
Yang E, Wang X, Gong Z, Yu M, Wu H and Zhang D: Exosome-mediated metabolic reprogramming: The emerging role in tumor microenvironment remodeling and its influence on cancer progression. Signal Transduct Target Ther. 5:2422020. View Article : Google Scholar : PubMed/NCBI | |
Ludwig N and Whiteside TL: Potential roles of tumor-derived exosomes in angiogenesis. Expert Opin Ther Targets. 22:409–417. 2018. View Article : Google Scholar : PubMed/NCBI | |
Wu B, Liu DA, Guan L, Myint PK, Chin L, Dang H, Xu Y, Ren J, Li T, Yu Z, et al: Stiff matrix induces exosome secretion to promote tumour growth. Nat Cell Biol. 25:415–424. 2023. View Article : Google Scholar : PubMed/NCBI | |
Hynes RO: The emergence of integrins: A personal and historical perspective. Matrix Biol. 23:333–340. 2004. View Article : Google Scholar : PubMed/NCBI | |
Campbell ID and Humphries MJ: Integrin structure, activation, and interactions. Cold Spring Harb Perspect Biol. 3:a0049942011. View Article : Google Scholar : PubMed/NCBI | |
Takada Y, Ye X and Simon S: The integrins. Genome Biol. 8:2152007. View Article : Google Scholar : PubMed/NCBI | |
Van Der Flier A and Sonnenberg A: Function and interactions of integrins. Cell Tissue Res. 305:285–298. 2001. View Article : Google Scholar : PubMed/NCBI | |
Zheng Y and Leftheris K: Insights into protein-ligand interactions in integrin complexes: Advances in structure determinations. J Med Chem. 63:5675–5696. 2020. View Article : Google Scholar : PubMed/NCBI | |
Humphries JD, Byron A and Humphries MJ: Integrin ligands at a glance. J Cell Sci. 119:3901–3903. 2006. View Article : Google Scholar : PubMed/NCBI | |
Sun CC, Qu XJ and Gao ZH: Arginine-glycine-aspartate-binding integrins as therapeutic and diagnostic targets. Am J Ther. 23:e198–e207. 2016. View Article : Google Scholar | |
Mitroulis I, Alexaki VI, Kourtzelis I, Ziogas A, Hajishengallis G and Chavakis T: Leukocyte integrins: Role in leukocyte recruitment and as therapeutic targets in inflammatory disease. Pharmacol Ther. 147:123–135. 2015. View Article : Google Scholar | |
Zeltz C and Gullberg D: The integrin-collagen connection-a glue for tissue repair? J Cell Sci. 129:653–664. 2016. View Article : Google Scholar : PubMed/NCBI | |
Aumailley M: The laminin family. Cell Adhes Migr. 7:48–55. 2013. View Article : Google Scholar | |
Pang X, He X, Qiu Z, Zhang H, Xie R, Liu Z, Gu Y, Zhao N, Xiang Q and Cui Y: Targeting integrin pathways: Mechanisms and advances in therapy. Signal Transduct Target Ther. 8:12023. View Article : Google Scholar : PubMed/NCBI | |
Anthis NJ and Campbell ID: The tail of integrin activation. Trends Biochem Sci. 36:191–198. 2011. View Article : Google Scholar : PubMed/NCBI | |
Soe ZY, Park EJ and Shimaoka M: Integrin regulation in immunological and cancerous cells and exosomes. Int J Mol Sci. 22:21932021. View Article : Google Scholar : PubMed/NCBI | |
Shattil SJ, Kim C and Ginsberg MH: The final steps of integrin activation: The end game. Nat Rev Mol Cell Biol. 11:288–300. 2010. View Article : Google Scholar : PubMed/NCBI | |
Ginsberg MH, Partridge A and Shattil SJ: Integrin regulation. Curr Opin Cell Biol. 17:509–516. 2005. View Article : Google Scholar : PubMed/NCBI | |
Ganguly KK, Pal S, Moulik S and Chatterjee A: Integrins and metastasis. Cell Adhes Migr. 7:251–261. 2013. View Article : Google Scholar | |
Zhang L, Qu J, Qi Y, Duan Y, Huang YW, Zhou Z, Li P, Yao J, Huang B, Zhang S and Yu D: EZH2 engages TGFβ signaling to promote breast cancer bone metastasis via integrin β1-FAK activation. Nat Commun. 13:25432022. View Article : Google Scholar | |
Huang L, Wang F, Wang X, Su C, Wu S, Yang C, Luo M, Zhang J and Fu L: M2-like macrophage-derived exosomes facilitate metastasis in non-small-cell lung cancer by delivering integrin αVβ3. MedComm (2020). 4:e1912022. View Article : Google Scholar | |
Hazelbag S, Kenter GG, Gorter A, Dreef EJ, Koopman LA, Violette SM, Weinreb PH and Fleuren GJ: Overexpression of the alpha v beta 6 integrin in cervical squamous cell carcinoma is a prognostic factor for decreased survival. J Pathol. 212:316–324. 2007. View Article : Google Scholar : PubMed/NCBI | |
Cantor DI, Cheruku HR, Nice EC and Baker MS: Integrin αvβ6 sets the stage for colorectal cancer metastasis. Cancer Metastasis Rev. 34:715–734. 2015. View Article : Google Scholar : PubMed/NCBI | |
Li ZH, Zhou Y, Ding YX, Guo QL and Zhao L: Roles of integrin in tumor development and the target inhibitors. Chin J Nat Med. 17:241–251. 2019.PubMed/NCBI | |
Lawrence R, Watters M, Davies CR, Pantel K and Lu YJ: Circulating tumour cells for early detection of clinically relevant cancer. Nat Rev Clin Oncol. 20:487–500. 2023. View Article : Google Scholar : PubMed/NCBI | |
Silva R, D'Amico G, Hodivala-Dilke KM and Reynolds LE: Integrins: The keys to unlocking angiogenesis. Arterioscler Thromb Vasc Biol. 28:1703–1713. 2008. View Article : Google Scholar : PubMed/NCBI | |
Shimaoka M, Kawamoto E, Gaowa A, Okamoto T and Park EJ: Connexins and integrins in exosomes. Cancers (Basel). 11:1062019. View Article : Google Scholar : PubMed/NCBI | |
Grigoryeva ES, Savelieva OE, Popova NO, Cherdyntseva NV and Perelmuter VM: Do tumor exosome integrins alone determine organotropic metastasis? Mol Biol Rep. 47:8145–8157. 2020. View Article : Google Scholar : PubMed/NCBI | |
Domenis R, Marino M, Cifù A, Scardino G, Curcio F and Fabris M: Circulating exosomes express α4β7 integrin and compete with CD4+ T cells for the binding to Vedolizumab. PLoS One. 15:e02423422020. View Article : Google Scholar | |
Maman S and Witz IP: A history of exploring cancer in context. Nat Rev Cancer. 18:359–376. 2018. View Article : Google Scholar : PubMed/NCBI | |
Ma Z, Wang LZ, Cheng JT, Lam WST, Ma X, Xiang X, Wong AL, Goh BC, Gong Q, Sethi G and Wang L: Targeting hypoxia-inducible factor-1-mediated metastasis for cancer therapy. Antioxid Redox Signal. 34:1484–1497. 2021. View Article : Google Scholar | |
Xu R, Rai A, Chen M, Suwakulsiri W, Greening DW and Simpson RJ: Extracellular vesicles in cancer-implications for future improvements in cancer care. Nat Rev Clin Oncol. 15:617–638. 2018. View Article : Google Scholar : PubMed/NCBI | |
Li K, Chen Y, Li A, Tan C and Liu X: Exosomes play roles in sequential processes of tumor metastasis. Int J Cancer. 144:1486–1495. 2019. View Article : Google Scholar | |
Roma-Rodrigues C, Mendes R, Baptista PV and Fernandes AR: Targeting tumor microenvironment for cancer therapy. Int J Mol Sci. 20:8402019. View Article : Google Scholar : PubMed/NCBI | |
Sahai E, Astsaturov I, Cukierman E, DeNardo DG, Egeblad M, Evans RM, Fearon D, Greten FR, Hingorani SR, Hunter T, et al: A framework for advancing our understanding of cancer-associated fibroblasts. Nat Rev Cancer. 20:174–186. 2020. View Article : Google Scholar : PubMed/NCBI | |
Sung JS, Kang CW, Kang S, Jang Y, Chae YC, Kim BG and Cho NH: ITGB4-mediated metabolic reprogramming of cancer-associated fibroblasts. Oncogene. 39:664–676. 2020. View Article : Google Scholar | |
Fang T, Lv H, Lv G, Li T, Wang C, Han Q, Yu L, Su B, Guo L, Huang S, et al: Tumor-derived exosomal miR-1247-3p induces cancer-associated fibroblast activation to foster lung metastasis of liver cancer. Nat Commun. 9:1912018. View Article : Google Scholar : PubMed/NCBI | |
Grivennikov SI, Greten FR and Karin M: Immunity, inflammation, and cancer. Cell. 140:883–899. 2010. View Article : Google Scholar : PubMed/NCBI | |
Xiao Y and Yu D: Tumor microenvironment as a therapeutic target in cancer. Pharmacol Ther. 221:1077532021. View Article : Google Scholar : | |
Pankov R, Cukierman E, Katz BZ, Matsumoto K, Lin DC, Lin S, Hahn C and Yamada KM: Integrin dynamics and matrix assembly: Tensin-dependent translocation of alpha(5)beta(1) integrins promotes early fibronectin fibrillogenesis. J Cell Biol. 148:1075–1090. 2000. View Article : Google Scholar : PubMed/NCBI | |
Bachmann M, Kukkurainen S, Hytönen VP and Wehrle-Haller B: Cell adhesion by integrins. Physiol Rev. 99:1655–1699. 2019. View Article : Google Scholar : PubMed/NCBI | |
Yazlovitskaya EM, Viquez OM, Tu T, De Arcangelis A, Georges-Labouesse E, Sonnenberg A, Pozzi A and Zent R: The laminin binding α3 and α6 integrins cooperate to promote epithelial cell adhesion and growth. Matrix Biol. 77:101–116. 2019. View Article : Google Scholar | |
Somanath PR, Malinin NL and Byzova TV: Cooperation between integrin alphavbeta3 and VEGFR2 in angiogenesis. Angiogenesis. 12:177–185. 2009. View Article : Google Scholar : PubMed/NCBI | |
Kanchanawong P and Calderwood DA: Organization, dynamics and mechanoregulation of integrin-mediated cell-ECM adhesions. Nat Rev Mol Cell Biol. 24:142–161. 2023. View Article : Google Scholar | |
Nieto MA, Huang RYJ, Jackson RA and Thiery JP: EMT: 2016. Cell. 166:21–45. 2016. View Article : Google Scholar : PubMed/NCBI | |
Liao TT and Yang MH: Revisiting epithelial-mesenchymal transition in cancer metastasis: The connection between epithelial plasticity and stemness. Mol Oncol. 11:792–804. 2017. View Article : Google Scholar : PubMed/NCBI | |
Liao TT and Yang MH: Hybrid epithelial/mesenchymal state in cancer metastasis: Clinical significance and regulatory mechanisms. Cells. 9:6232020. View Article : Google Scholar : PubMed/NCBI | |
Lu W and Kang Y: Epithelial-mesenchymal plasticity in cancer progression and metastasis. Dev Cell. 49:361–374. 2019. View Article : Google Scholar : PubMed/NCBI | |
Grünert S, Jechlinger M and Beug H: Diverse cellular and molecular mechanisms contribute to epithelial plasticity and metastasis. Nat Rev Mol Cell Biol. 4:657–665. 2003. View Article : Google Scholar : PubMed/NCBI | |
Lambert AW, Pattabiraman DR and Weinberg RA: Emerging biological principles of metastasis. Cell. 168:670–691. 2017. View Article : Google Scholar : PubMed/NCBI | |
Tanabe S, Quader S, Cabral H and Ono R: Interplay of EMT and CSC in cancer and the potential therapeutic strategies. Front Pharmacol. 11:9042020. View Article : Google Scholar : PubMed/NCBI | |
Syn N, Wang L, Sethi G, Thiery JP and Goh BC: Exosome-mediated metastasis: From epithelial-mesenchymal transition to escape from immunosurveillance. Trends Pharmacol Sci. 37:606–617. 2016. View Article : Google Scholar : PubMed/NCBI | |
Yeung KT and Yang J: Epithelial-mesenchymal transition in tumor metastasis. Mol Oncol. 11:28–39. 2017. View Article : Google Scholar : PubMed/NCBI | |
Mamuya FA and Duncan MK: aV integrins and TGF-β-induced EMT: A circle of regulation. J Cell Mol Med. 16:445–455. 2012. View Article : Google Scholar : | |
Caramel J, Papadogeorgakis E, Hill L, Browne GJ, Richard G, Wierinckx A, Saldanha G, Osborne J, Hutchinson P, Tse G, et al: A switch in the expression of embryonic EMT-inducers drives the development of malignant melanoma. Cancer Cell. 24:466–480. 2013. View Article : Google Scholar : PubMed/NCBI | |
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 | |
Wen S, Hou Y, Fu L, Xi L, Yang D, Zhao M, Qin Y, Sun K, Teng Y and Liu M: Cancer-associated fibroblast (CAF)-derived IL32 promotes breast cancer cell invasion and metastasis via integrin β3-p38 MAPK signalling. Cancer Lett. 442:320–332. 2019. View Article : Google Scholar | |
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 | |
Hao Q, Wu Y, Wu Y, Wang P and Vadgama JV: Tumor-derived exosomes in tumor-induced immune suppression. Int J Mol Sci. 23:14612022. View Article : Google Scholar : PubMed/NCBI | |
Zhao L, Ma X and Yu J: Exosomes and organ-specific metastasis. Mol Ther Methods Clin Dev. 22:133–147. 2021. View Article : Google Scholar : PubMed/NCBI | |
Chen Y, Yang S, Tavormina J, Tampe D, Zeisberg M, Wang H, Mahadevan KK, Wu CJ, Sugimoto H, Chang CC, et al: Oncogenic collagen I homotrimers from cancer cells bind to α3β1 integrin and impact tumor microbiome and immunity to promote pancreatic cancer. Cancer Cell. 40:818–834.e9. 2022. View Article : Google Scholar | |
Genduso S, Freytag V, Schetler D, Kirchner L, Schiecke A, Maar H, Wicklein D, Gebauer F, Bröker K, Stürken C, et al: Tumor cell integrin β4 and tumor stroma E-/P-selectin cooperatively regulate tumor growth in vivo. J Hematol Oncol. 16:232023. View Article : Google Scholar | |
Zhang Y, Xie R, Zhang H, Zheng Y, Lin C, Yang L, Huang M, Li M, Song F, Lu L, et al: Integrin β7 inhibits colorectal cancer pathogenesis via maintaining antitumor immunity. Cancer Immunol Res. 9:967–980. 2021. View Article : Google Scholar : PubMed/NCBI | |
Ruan S, Lin M, Zhu Y, Lum LG, Thakur A, Jin R, Shao W, Zhang Y, Hu Y, Huang S, et al: Integrin β4-targeted cancer immunotherapies inhibit tumor growth and decrease metastasis. Cancer Res. 80:771–783. 2020. View Article : Google Scholar | |
Vannini A, Leoni V, Barboni C, Sanapo M, Zaghini A, Malatesta P, Campadelli-Fiume G and Gianni T: αvβ3-integrin regulates PD-L1 expression and is involved in cancer immune evasion. Proc Natl Acad Sci USA. 116:20141–20150. 2019. View Article : Google Scholar | |
Ren D, Zhao J, Sun Y, Li D, Meng Z, Wang B, Fan P, Liu Z, Jin X and Wu H: Overexpressed ITGA2 promotes malignant tumor aggression by up-regulating PD-L1 expression through the activation of the STAT3 signaling pathway. J Exp Clin Cancer Res. 38:4852019. View Article : Google Scholar : PubMed/NCBI | |
Lainé A, Labiad O, Hernandez-Vargas H, This S, Sanlaville A, Léon S, Dalle S, Sheppard D, Travis MA, Paidassi H and Marie JC: Regulatory T cells promote cancer immune-escape through integrin αvβ8-mediated TGF-β activation. Nat Commun. 12:62282021. View Article : Google Scholar | |
Takasaka N, Seed RI, Cormier A, Bondesson AJ, Lou J, Elattma A, Ito S, Yanagisawa H, Hashimoto M, Ma R, et al: Integrin αvβ8-expressing tumor cells evade host immunity by regulating TGF-β activation in immune cells. JCI Insight. 3:e1225912018. View Article : Google Scholar | |
Balkwill F and Mantovani A: Inflammation and cancer: Back to virchow? Lancet. 357:539–545. 2001. View Article : Google Scholar : PubMed/NCBI | |
Myint PK, Park EJ, Gaowa A, Kawamoto E and Shimaoka M: Targeted remodeling of breast cancer and immune cell homing niches by exosomal integrins. Diagn Pathol. 15:382020. View Article : Google Scholar : PubMed/NCBI | |
Lukanidin E and Sleeman JP: Building the niche: The role of the S100 proteins in metastatic growth. Semin Cancer Biol. 22:216–225. 2012. View Article : Google Scholar : PubMed/NCBI | |
Nagaraj S, Gupta K, Pisarev V, Kinarsky L, Sherman S, Kang L, Herber DL, Schneck J and Gabrilovich DI: Altered recognition of antigen is a mechanism of CD8+ T cell tolerance in cancer. Nat Med. 13:828–835. 2007. View Article : Google Scholar : PubMed/NCBI | |
Ichikawa M, Williams R, Wang L, Vogl T and Srikrishna G: S100A8/A9 activate key genes and pathways in colon tumor progression. Mol Cancer Res. 9:133–148. 2011. View Article : Google Scholar : PubMed/NCBI | |
Nishida N, Yano H, Nishida T, Kamura T and Kojiro M: Angiogenesis in cancer. Vasc Health Risk Manag. 2:213–219. 2006. View Article : Google Scholar | |
Sharghi-Namini S, Tan E, Ong LLS, Ge R and Asada HH: Dll4-containing exosomes induce capillary sprout retraction in a 3D microenvironment. Sci Rep. 4:40312014. View Article : Google Scholar : PubMed/NCBI | |
Nazarenko I, Rana S, Baumann A, McAlear J, Hellwig A, Trendelenburg M, Lochnit G, Preissner KT and Zöller M: Cell surface tetraspanin Tspan8 contributes to molecular pathways of exosome-induced endothelial cell activation. Cancer Res. 70:1668–1678. 2010. View Article : Google Scholar : PubMed/NCBI | |
Krishn SR, Singh A, Bowler N, Duffy AN, Friedman A, Fedele C, Kurtoglu S, Tripathi SK, Wang K, Hawkins A, et al: Prostate cancer sheds the αvβ3 integrin in vivo through exosomes. Matrix Biol. 77:41–57. 2019. View Article : Google Scholar | |
Harney AS, Arwert EN, Entenberg D, Wang Y, Guo P, Qian BZ, Oktay MH, Pollard JW, Jones JG and Condeelis JS: Real-time imaging reveals local, transient vascular permeability and tumor cell intravasation stimulated by Tie2Hi macrophage-derived VEGFA. Cancer Discov. 5:932–943. 2015. View Article : Google Scholar : PubMed/NCBI | |
Sixt M, Bauer M, Lämmermann T and Fässler R: Beta1 integrins: Zip codes and signaling relay for blood cells. Curr Opin Cell Biol. 18:482–490. 2006. View Article : Google Scholar : PubMed/NCBI | |
Sun L, Guo S, Xie Y and Yao Y: The characteristics and the multiple functions of integrin β1 in human cancers. J Transl Med. 21:7872023. View Article : Google Scholar | |
Jin H, Su J, Garmy-Susini B, Kleeman J and Varner J: Integrin alpha4beta1 promotes monocyte trafficking and angiogenesis in tumors. Cancer Res. 66:2146–2152. 2006. View Article : Google Scholar : PubMed/NCBI | |
Weis SM: Evaluating integrin function in models of angiogenesis and vascular permeability. Methods Enzymol. 426:505–528. 2007. View Article : Google Scholar : PubMed/NCBI | |
Sobierajska K, Ciszewski WM, Sacewicz-Hofman I and Niewiarowska J: Endothelial cells in the tumor microenvironment. Adv Exp Med Biol. 1234:71–86. 2020. View Article : Google Scholar : PubMed/NCBI | |
Labelle M and Hynes RO: The initial hours of metastasis: The importance of cooperative host-tumor cell interactions during hematogenous dissemination. Cancer Discov. 2:1091–1099. 2012. View Article : Google Scholar : PubMed/NCBI | |
Peinado H, Lavotshkin S and Lyden D: The secreted factors responsible for pre-metastatic niche formation: Old sayings and new thoughts. Semin Cancer Biol. 21:139–146. 2011. View Article : Google Scholar : PubMed/NCBI | |
Coleman RE, Croucher PI, Padhani AR, Clézardin P, Chow E, Fallon M, Guise T, Colangeli S, Capanna R and Costa L: Bone metastases. Nat Rev Dis Primer. 6:832020. View Article : Google Scholar | |
Nguyen DX, Bos PD and Massagué J: Metastasis: From dissemination to organ-specific colonization. Nat Rev Cancer. 9:274–284. 2009. View Article : Google Scholar : PubMed/NCBI | |
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 | |
Sceneay J, Smyth MJ and Möller A: The pre-metastatic niche: Finding common ground. Cancer Metastasis Rev. 32:449–464. 2013. View Article : Google Scholar : PubMed/NCBI | |
Kaplan RN, Riba RD, Zacharoulis S, Bramley AH, Vincent L, Costa C, MacDonald DD, Jin DK, Shido K, Kerns SA, et al: VEGFR1-positive haematopoietic bone marrow progenitors initiate the pre-metastatic niche. Nature. 438:820–827. 2005. View Article : Google Scholar : PubMed/NCBI | |
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 | |
Peinado H, Alečković M, Lavotshkin S, Matei I, Costa-Silva B, Moreno-Bueno G, Hergueta-Redondo M, Williams C, García-Santos G, Ghajar C, et al: Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat Med. 18:883–891. 2012. View Article : Google Scholar : PubMed/NCBI | |
Costa-Silva B, Aiello NM, Ocean AJ, Singh S, Zhang H, Thakur BK, Becker A, Hoshino A, Mark MT, Molina H, et al: Pancreatic cancer exosomes initiate pre-metastatic niche formation in the liver. Nat Cell Biol. 17:816–826. 2015. View Article : Google Scholar : PubMed/NCBI | |
Desgrosellier JS and Cheresh DA: Integrins in cancer: Biological implications and therapeutic opportunities. Nat Rev Cancer. 10:9–22. 2010. View Article : Google Scholar | |
Fedele C, Singh A, Zerlanko BJ, Iozzo RV and Languino LR: The αvβ6 integrin is transferred intercellularly via exosomes. J Biol Chem. 290:4545–4551. 2015. View Article : Google Scholar : PubMed/NCBI | |
Zhang X: Interactions between cancer cells and bone microenvironment promote bone metastasis in prostate cancer. Cancer Commun (Lond). 39:762019. View Article : Google Scholar : PubMed/NCBI | |
Geng X, Chang B and Shan J: Role and correlation of exosomes and integrins in bone metastasis of prostate cancer. Andrologia. 54:e145502022. View Article : Google Scholar : PubMed/NCBI | |
Tang L, Xu M, Zhang L, Qu L and Liu X: Role of αVβ3 in prostate cancer: Metastasis initiator and important therapeutic target. Onco Targets Ther. 13:7411–7422. 2020. View Article : Google Scholar : | |
Chen GY, Cheng JCH, Chen YF, Yang JCH and Hsu FM: Circulating exosomal integrin β3 is associated with intracranial failure and survival in lung cancer patients receiving cranial irradiation for brain metastases: A prospective observational study. Cancers (Basel). 13:3802021. View Article : Google Scholar | |
Bijnsdorp IV, Geldof AA, Lavaei M, Piersma SR, Van Moorselaar RJA and Jimenez CR: Exosomal ITGA3 interferes with non-cancerous prostate cell functions and is increased in urine exosomes of metastatic prostate cancer patients. J Extracell Vesicles. 2: View Article : Google Scholar : 2013.PubMed/NCBI | |
Dallavalle S, Dobričić V, Lazzarato L, Gazzano E, Machuqueiro M, Pajeva I, Tsakovska I, Zidar N and Fruttero R: Improvement of conventional anti-cancer drugs as new tools against multidrug resistant tumors. Drug Resist Updat. 50:1006822020. View Article : Google Scholar : PubMed/NCBI | |
Sousa D, Lima RT and Vasconcelos MH: Intercellular transfer of cancer drug resistance traits by extracellular vesicles. Trends Mol Med. 21:595–608. 2015. View Article : Google Scholar : PubMed/NCBI | |
Lv MM, Zhu XY, Chen WX, Zhong SL, Hu Q, Ma TF, Zhang J, Chen L, Tang JH and Zhao JH: Exosomes mediate drug resistance transfer in MCF-7 breast cancer cells and a probable mechanism is delivery of P-glycoprotein. Tumor Biol. 35:10773–10779. 2014. View Article : Google Scholar | |
Wang B, Zhang Y, Ye M, Wu J, Ma L and Chen H: Cisplatin-resistant MDA-MB-231 cell-derived exosomes increase the resistance of recipient cells in an exosomal miR-423-5p-dependent manner. Curr Drug Metab. 20:804–814. 2019. View Article : Google Scholar : PubMed/NCBI | |
Dong X, Bai X, Ni J, Zhang H, Duan W, Graham P and Li Y: Exosomes and breast cancer drug resistance. Cell Death Dis. 11:9872020. View Article : Google Scholar : PubMed/NCBI | |
Martinez VG, O'Neill S, Salimu J, Breslin S, Clayton A, Crown J and O'Driscoll L: Resistance to HER2-targeted anti-cancer drugs is associated with immune evasion in cancer cells and their derived extracellular vesicles. Oncoimmunology. 6:e13625302017. View Article : Google Scholar : PubMed/NCBI | |
Safaei R, Larson BJ, Cheng TC, Gibson MA, Otani S, Naerdemann W and Howell SB: Abnormal lysosomal trafficking and enhanced exosomal export of cisplatin in drug-resistant human ovarian carcinoma cells. Mol Cancer Ther. 4:1595–1604. 2005. View Article : Google Scholar : PubMed/NCBI | |
Goler-Baron V and Assaraf YG: Structure and function of ABCG2-rich extracellular vesicles mediating multidrug resistance. PLoS One. 6:e160072011. View Article : Google Scholar : PubMed/NCBI | |
Mollazadeh S, Sahebkar A, Hadizadeh F, Behravan J and Arabzadeh S: Structural and functional aspects of P-glycoprotein and its inhibitors. Life Sci. 214:118–123. 2018. View Article : Google Scholar : PubMed/NCBI | |
Zhang YH, Gao ZF, Dong GH, Li X, Wu Y, Li G, Wang AL, Li HL and Yin DL: Suppression of αvβ6 downregulates P-glycoprotein and sensitizes multidrug-resistant breast cancer cells to anticancer drugs. Neoplasma. 67:379–388. 2020. View Article : Google Scholar : PubMed/NCBI | |
Lopes-Rodrigues V, Seca H, Sousa D, Sousa E, Lima RT and Vasconcelos MH: The network of P-glycoprotein and microRNAs interactions. Int J Cancer. 135:253–263. 2014. View Article : Google Scholar | |
Damiano JS, Cress AE, Hazlehurst LA, Shtil AA and Dalton WS: Cell adhesion mediated drug resistance (CAM-DR): Role of integrins and resistance to apoptosis in human myeloma cell lines. Blood. 93:1658–1667. 1999. View Article : Google Scholar : PubMed/NCBI | |
Seguin L, Desgrosellier JS, Weis SM and Cheresh DA: Integrins and cancer: Regulators of cancer stemness, metastasis, and drug resistance. Trends Cell Biol. 25:234–240. 2015. View Article : Google Scholar : PubMed/NCBI | |
Cooper J and Giancotti FG: Integrin signaling in cancer: Mechanotransduction, stemness, epithelial plasticity, and therapeutic resistance. Cancer Cell. 35:347–367. 2019. View Article : Google Scholar : PubMed/NCBI | |
Kanda R, Kawahara A, Watari K, Murakami Y, Sonoda K, Maeda M, Fujita H, Kage M, Uramoto H, Costa C, et al: Erlotinib resistance in lung cancer cells mediated by integrin β1/Src/Akt-driven bypass signaling. Cancer Res. 73:6243–6253. 2013. View Article : Google Scholar : PubMed/NCBI | |
Janouskova H, Maglott A, Leger DY, Bossert C, Noulet F, Guerin E, Guenot D, Pinel S, Chastagner P, Plenat F, et al: Integrin α5β1 plays a critical role in resistance to temozolomide by interfering with the p53 pathway in high-grade glioma. Cancer Res. 72:3463–3470. 2012. View Article : Google Scholar : PubMed/NCBI | |
Luo J, Yao JF, Deng XF, Zheng XD, Jia M, Wang YQ, Huang Y and Zhu JH: 14, 15-EET induces breast cancer cell EMT and cisplatin resistance by up-regulating integrin αvβ3 and activating FAK/PI3K/AKT signaling. J Exp Clin Cancer Res. 37:232018. View Article : Google Scholar | |
Khademi R, Malekzadeh H, Bahrami S, Saki N, Khademi R and Villa-Diaz LG: Regulation and functions of α6-integrin (CD49f) in cancer biology. Cancers (Basel). 15:34662023. View Article : Google Scholar | |
Kawakami K, Fujita Y, Kato T, Mizutani K, Kameyama K, Tsumoto H, Miura Y, Deguchi T and Ito M: Integrin β4 and vinculin contained in exosomes are potential markers for progression of prostate cancer associated with taxane-resistance. Int J Oncol. 47:384–390. 2015. View Article : Google Scholar : PubMed/NCBI | |
Tenchov R, Sasso JM, Wang X, Liaw WS, Chen CA and Zhou QA: Exosomes-nature's lipid nanoparticles, a rising star in drug delivery and diagnostics. ACS Nano. 16:17802–17846. 2022. View Article : Google Scholar : PubMed/NCBI | |
Kim MS, Haney MJ, Zhao Y, Mahajan V, Deygen I, Klyachko NL, Inskoe E, Piroyan A, Sokolsky M, Okolie O, et al: Development of exosome-encapsulated paclitaxel to overcome MDR in cancer cells. Nanomedicine. 12:655–664. 2016. View Article : Google Scholar | |
Kobayashi M, Sawada K, Miyamoto M, Shimizu A, Yamamoto M, Kinose Y, Nakamura K, Kawano M, Kodama M, Hashimoto K and Kimura T: Exploring the potential of engineered exosomes as delivery systems for tumor-suppressor microRNA replacement therapy in ovarian cancer. Biochem Biophys Res Commun. 527:153–161. 2020. View Article : Google Scholar : PubMed/NCBI | |
Cho E, Nam GH, Hong Y, Kim YK, Kim DH, Yang Y and Kim IS: Comparison of exosomes and ferritin protein nanocages for the delivery of membrane protein therapeutics. J Controlled Release. 279:326–335. 2018. View Article : Google Scholar | |
Alvarez-Erviti L, Seow Y, Yin H, Betts C, Lakhal S and Wood MJA: Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes. Nat Biotechnol. 29:341–345. 2011. View Article : Google Scholar : PubMed/NCBI | |
Gaurav I, Thakur A, Iyaswamy A, Wang X, Chen X and Yang Z: Factors affecting extracellular vesicles based drug delivery systems. Molecules. 26:15442021. View Article : Google Scholar : PubMed/NCBI | |
Li MY, Liu LZ and Dong M: Progress on pivotal role and application of exosome in lung cancer carcinogenesis, diagnosis, therapy and prognosis. Mol Cancer. 20:222021. View Article : Google Scholar : PubMed/NCBI | |
Zhao L, Wang H, Fu J, Wu X, Liang XY, Liu XY, Wu X, Cao LL, Xu ZY and Dong M: Microfluidic-based exosome isolation and highly sensitive aptamer exosome membrane protein detection for lung cancer diagnosis. Biosens Bioelectron. 214:1144872022. View Article : Google Scholar : PubMed/NCBI | |
Zhang L and Yu D: Exosomes in cancer development, metastasis, and immunity. Biochim Biophys Acta Rev Cancer. 1871:455–468. 2019. View Article : Google Scholar : PubMed/NCBI | |
Yang H, Xu Z, Peng Y, Wang J and Xiang Y: Integrin β4 as a potential diagnostic and therapeutic tumor marker. Biomolecules. 11:11972021. View Article : Google Scholar | |
Valdembri D and Serini G: The roles of integrins in cancer. Fac Rev. 10:452021. View Article : Google Scholar : PubMed/NCBI | |
Hurwitz SN and Meckes DG Jr: Extracellular vesicle integrins distinguish unique cancers. Proteomes. 7:142019. View Article : Google Scholar : PubMed/NCBI | |
Zhang Z, Wang Z, Liu T, Tang J, Liu Y, Gou T, Chen K, Wang L, Zhang J, Yang Y and Zhang H: Exploring the role of ITGB6: Fibrosis, cancer, and other diseases. Apoptosis. 29:570–585. 2024. View Article : Google Scholar | |
Bandyopadhyay A and Raghavan S: Defining the role of integrin αvβ6 in cancer. Curr Drug Targets. 10:645–652. 2009. View Article : Google Scholar : PubMed/NCBI | |
Hausner SH, DiCara D, Marik J, Marshall JF and Sutcliffe JL: Use of a peptide derived from foot-and-mouth disease virus for the noninvasive imaging of human cancer: Generation and evaluation of 4-[18F]fluorobenzoyl A20FMDV2 for in vivo imaging of integrin alphavbeta6 expression with positron emission tomography. Cancer Res. 67:7833–7840. 2007. View Article : Google Scholar : PubMed/NCBI | |
Su L, Chen Y, Huang C, Wu S, Wang X, Zhao X, Xu Q, Sun R, Kong X, Jiang X, et al: Targeting Src reactivates pyroptosis to reverse chemoresistance in lung and pancreatic cancer models. Sci Transl Med. 15:eabl78952023. View Article : Google Scholar : PubMed/NCBI | |
Chen JR, Zhao JT and Xie ZZ: Integrin-mediated cancer progression as a specific target in clinical therapy. Biomed Pharmacother. 155:1137452022. View Article : Google Scholar : PubMed/NCBI | |
Zhang L, Gülses A, Purcz N, Weimer J, Wiltfang J and Açil Y: A comparative assessment of the effects of integrin inhibitor cilengitide on primary culture of head and neck squamous cell carcinoma (HNSCC) and HNSCC cell lines. Clin Transl Oncol. 21:1052–1060. 2019. View Article : Google Scholar : PubMed/NCBI | |
Yu Q, Xiao W, Sun S, Sohrabi A, Liang J and Seidlits SK: Extracellular matrix proteins confer cell adhesion-mediated drug resistance through integrin αv in glioblastoma cells. Front Cell Dev Biol. 9:6165802021. View Article : Google Scholar | |
Ignatiadis M, Sledge GW and Jeffrey SS: Liquid biopsy enters the clinic-implementation issues and future challenges. Nat Rev Clin Oncol. 18:297–312. 2021. View Article : Google Scholar : PubMed/NCBI | |
Yu W, Hurley J, Roberts D, Chakrabortty SK, Enderle D, Noerholm M, Breakefield XO and Skog JK: Exosome-based liquid biopsies in cancer: Opportunities and challenges. Ann Oncol. 32:466–477. 2021. View Article : Google Scholar : PubMed/NCBI | |
Bergonzini C, Kroese K, Zweemer AJM and Danen EHJ: Targeting integrins for cancer therapy-disappointments and opportunities. Front Cell Dev Biol. 10:8638502022. View Article : Google Scholar | |
Cox D: How not to discover a drug-integrins. Expert Opin Drug Discov. 16:197–211. 2021. View Article : Google Scholar | |
Tam SH, Sassoli PM, Jordan RE and Nakada MT: Abciximab (ReoPro, chimeric 7E3 Fab) demonstrates equivalent affinity and functional blockade of glycoprotein IIb/IIIa and alpha(v)beta3 integrins. Circulation. 98:1085–1091. 1998. View Article : Google Scholar : PubMed/NCBI | |
Corcoran C, Rani S, O'Brien K, O'Neill A, Prencipe M, Sheikh R, Webb G, McDermott R, Watson W, Crown J and O'Driscoll L: Docetaxel-resistance in prostate cancer: Evaluating associated phenotypic changes and potential for resistance transfer via exosomes. PLoS One. 7:e509992012. View Article : Google Scholar : PubMed/NCBI | |
Xiao X, Yu S, Li S, Wu J, Ma R, Cao H, Zhu Y and Feng J: Exosomes: Decreased sensitivity of lung cancer A549 cells to cisplatin. PLoS One. 9:e895342014. View Article : Google Scholar : PubMed/NCBI | |
Hazlehurst LA, Argilagos RF and Dalton WS: Beta1 integrin mediated adhesion increases Bim protein degradation and contributes to drug resistance in leukaemia cells. Br J Haematol. 136:269–275. 2007. View Article : Google Scholar : PubMed/NCBI | |
Wei H, Chen J, Wang S, Fu F, Zhu X, Wu C, Liu Z, Zhong G and Lin J: A nanodrug consisting of doxorubicin and exosome derived from mesenchymal stem cells for osteosarcoma treatment in vitro. Int J Nanomedicine. 14:8603–8610. 2019. View Article : Google Scholar : PubMed/NCBI | |
Tian Y, Li S, Song J, Ji T, Zhu M, Anderson GJ, Wei J and Nie G: A doxorubicin delivery platform using engineered natural membrane vesicle exosomes for targeted tumor therapy. Biomaterials. 35:2383–2390. 2014. View Article : Google Scholar | |
Chen K and Chen X: Integrin targeted delivery of chemotherapeutics. Theranostics. 1:189–200. 2011. View Article : Google Scholar : PubMed/NCBI | |
Albelda SM, Mette SA, Elder DE, Stewart R, Damjanovich L, Herlyn M and Buck CA: Integrin distribution in malignant melanoma: Association of the beta 3 subunit with tumor progression. Cancer Res. 50:6757–6764. 1990.PubMed/NCBI | |
Choi H, Choi Y, Yim HY, Mirzaaghasi A, Yoo JK and Choi C: Biodistribution of exosomes and engineering strategies for targeted delivery of therapeutic exosomes. Tissue Eng Regen Med. 18:499–511. 2021. View Article : Google Scholar : PubMed/NCBI | |
Gingras MC, Roussel E, Bruner JM, Branch CD and Moser RP: Comparison of cell adhesion molecule expression between glioblastoma multiforme and autologous normal brain tissue. J Neuroimmunol. 57:143–153. 1995. View Article : Google Scholar : PubMed/NCBI |