Open Access

Role of exosomes in cellular communication between tumor cells and the tumor microenvironment (Review)

  • Authors:
    • Qiao Li
  • View Affiliations

  • Published online on: May 31, 2022     https://doi.org/10.3892/ol.2022.13360
  • Article Number: 240
  • Copyright: © Li . This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Exosomes are nanovesicles secreted by almost all types of cells. They contain RNAs, microRNAs, proteins and other bioactive substances, and can be used as carriers and for communication between cells. They regulate numerous biological processes, such as tumor development, cell proliferation and resistance to chemotherapy. Exosome‑mediated communication between tumor cells and the tumor microenvironment (TME) is crucial in the initiation and progression of tumor development. The present review aims to summarize the role of exosomes in mediating the communication between tumor cells and the TME and to suggest the potential use of exosomes as targets for the development of novel therapeutic strategies against cancer.

Introduction

Exosomes were discovered by Johnstone et al in 1987 (1,2). The study found that sheep reticulocytes secrete exosomes to remove the transferrin receptor during their maturation (2). Exosomes are nanoscale extracellular vehicles with a diameter ranging between 50 and 150 nm (35) and are located in the density range of 1.13-1.19 g/ml after sucrose gradient centrifugation. Under an electron microscope, exosomes appear as cup-shaped vesicles (6). They are rich in Alix, tumor susceptibility gene 101, heat shock protein 70 (HSP70) (7), tetraspanins (CD63, CD81 and CD9) (7), cytoskeleton proteins, and major histocompatibility complex class II molecules (8). Exosomes contain specific components of the cells from which they originate (9). For example, exosomes deriving from tumor cells contain molecules that suggest their endoplasmic reticulum origin and cell components that can promote tumor development (10), including tyrosine kinase receptors, oncoproteins, phosphorylated proteins and microRNAs (miRNAs/miRs) (11). In addition, exosomes can be detected in a variety of body fluids, including blood, urine and breast milk (12). Therefore, the detection of tumor-related cell components in exosomes can be used as a method for the early diagnosis of some tumors (13).

The biogenesis process of exosomes distinguishes them from apoptotic bodies and other extracellular vesicles. Exosomes originate from the endosomal system and are formed from early endosomes: Inward budding of cellular membranes leads to formation of early endosomes (3,14,15). Early endosomes mature to late endosomes with multiple intraluminal vesicles (ILVs) and late endosomes form multivesicular bodies (MVBs). After the fusion of MVBs with the plasma cell membrane, exosomes are released from the cell into the extracellular environment, carrying the biological information of secretory cells and transmitting biological signals to recipient cells (3,14,15).

During the maturation of ILVs into MVBs, proteins are sorted. Internalized proteins in early endosomes are ubiquitinated and directed to late endosomes (3,14,15). Multiprotein complexes are involved in the sorting and ubiquitination of target proteins (3). The endosomal sorting complexes required for transport (ESCRT) complex regulates endosomal maturation (14). ESCRT complexes are composed of four complexes (ESCRT-0, -I, -II and -III) (14). Ubiquitylated proteins in the endosomal membrane are sequestered by the ESCRT-0 complex (15). The ESCRT-I and ESCRT-II complexes are responsible for the budding of the membrane. For instance, ESCRT induces MVB vesicles to sprout (15,16), while Rab proteins regulate intracellular vesicle transport (17). Furthermore, there are a variety of proteins involved in this process, including Rab GTPases (Rab11, Rab35, Rab27A and Rab 27b), which are involved in the formation of exosomes (17,18).

Exosomal nucleic acids

Exosomes can be secreted by reticulocytes (1), dendritic cells (19), macrophages (7) and epithelial cells (7). The double-layered membrane protects exosomal RNAs from being digested by RNA enzymes in the plasma or other body fluids (20). Therefore, exosomes can be used as tumor biomarkers for early diagnosis (20). For example, in gastric cancer, exosomal long noncoding RNA 152 can be used as a potential biological marker (21). Furthermore, miRNAs in circulating exosomes can be used as biomarkers for the screening and diagnosis of lung cancer (22) and several exosomal biomarkers have been reported in a prostate cancer study (23). Therefore, the identification of tumor exosomal RNAs has become a potential diagnostic tool.

Exosomes are known to contain a considerable number of functional messenger RNAs (mRNAs) and miRNAs (24). Exosomes from breast cancer, colorectal cancer and leukemia cells contain human telomerase reverse transcriptase (hTERT) mRNA (25). When these exosomes are absorbed by fibroblasts, they can promote the construction of a tumor metastatic environment by promoting the proliferation and survival of fibroblasts (25). Gutkin et al (25) reported that human tumor cells-derived exosomes can transfer hTERT transcriptional mRNA to telomerase-negative fibroblasts. The delivered mRNA can be translated into a protein, which renders the recipient cells telomerase-positive. This was the first study to report that exosomes can transfer the transcriptional mRNA of hTERT.

Exosomes from chronic lymphocytic leukemia (CLL) cells promote the proliferation of stromal cells by secreting miR-202-3p into human bone marrow cells (26). When CLL-exosomes, derived either from CLL cell culture supernatants or plasma from patients with CLL, were co-cultured with human stromal cells, the latter could accept these exosomes, resulting in the promotion of their proliferation associated with the induction of c-Fos expression (26). These exosomes are rich in small RNAs, particularly hsa-mir-202-3p, which can enhance the Hedgehog signal transduction pathway, thus promoting the proliferation of recipient cells (26). miR-9 transported by tumor cell-derived exosomes can promote endothelial cell migration and angiogenesis through the Janus kinase/signal transducer and activator of the transcription signaling pathway (27).

Role of exosomes in tumor metastasis

Increasing evidence shows that exosomes serve an important role in intercellular communication (28,29). Exosomes transfer proteins, mRNAs and miRNAs to receptive cells, resulting in the regulation of a variety of functions, including immunoregulation, matrix remodeling, growth factor delivery and oncoprotein transfer (28,3032). The tumor microenvironment (TME) is composed of tumor-associated fibroblasts, osteoblasts and immune cells. The TME can promote the proliferation of tumor cells and confers resistance to chemotherapy (33). Exosomes can regulate multiple biological behaviors of tumor cells, as shown in Fig. 1. Furthermore, exosomal miRNAs, mRNA, proteins and other functional molecules are accepted by the stromal cells in the TME where they exert regulatory effects (34).

Exosomes from the breast cancer microenvironment can promote breast cancer cell metastasis through the Wnt plane cell polarity (PCP) signal (35). During breast cancer cell metastasis, exosomes can promote tumor metastasis via the Wnt PCP pathway, which is different from the classical Wnt signaling pathway (35). Hood et al (30) found that exosomes secreted by melanoma cells prepare sentinel lymph nodes for tumor cell metastasis as tumor cells are more likely to metastasize to a site with abundant melanoma exosomes. Bone marrow progenitor cells are permanently ‘educated’ by exosomes from highly invasive melanoma cells through the receptor tyrosine kinase, MET, to promote the invasion and metastasis of primary tumor cells (36). The receptor tyrosine kinase MET transforms bone marrow precursor cells into an angiogenesis-promoting phenotype, which expresses c-kit, receptor tyrosine kinase EGF-like domains 2 and Met (36). The inhibition of exosomal Met expression can weaken the metastasis that is promoted by bone marrow progenitor cells (36).

Exosomes derived from lung cancer cells can promote epithelial-to-mesenchymal transition (EMT) (37). When serum-derived exosomes from patients with advanced lung cancer are used to treat human bronchial epithelium, they can promote the EMT of the recipient cells (37). Exosomes from pancreatic cancer cells can induce the formation of the metastasis-promoting microenvironment in the liver (38). Exosomes derived from gastric cancer cells can carry EGFR and regulate the microenvironment of the liver to promote the metastasis of gastric cancer cells (39). Exosomes isolated from the serum of patients with gastric cancer contain EGFR, while the exosomes derived from the serum of normal individuals do not (39). This indicates that the levels of the EGFR oncoprotein in the exosomes from the serum of patients with gastric cancer are associated with gastric cancer metastatic stages. The exosomes carrying the EGFR from the gastric cancer can be taken up by liver cells, which will further activate liver hepatocyte growth factor and prepare the premetastatic ‘soil’ for the gastric cancer (39).

Role of exosomes in drug resistance

Exosomes derived from tumor cells can promote the resistance of tumor cells to chemotherapeutic drugs via multiple mechanisms, such as carrying multidrug resistance (MDR) proteins or by secreting chemotherapeutic drugs (40). For example, exosomes can transfer drug resistance by secreting MDR proteins into recipient cells (41). Exosomes derived from 5T33 bone marrow stromal cells promote the proliferation of multiple myeloma cells and induce their resistance to bortezomib (42). Exosomes can mediate drug resistance by transporting drug resistance proteins. In prostate cancer, docetaxel resistance is associated with an increased secretion of exosomes (43). Exosomes secreted from docetaxel-resistant prostate cancer cells can confer docetaxel resistance to docetaxel-sensitive prostate cancer cells by transporting P-glycoprotein in the exosomes (43). Exosomes can have countertherapeutic effects by binding to chemotherapy drugs. In breast cancer, HER-2-overexpressing exosomes can resist the therapeutic effect of trastuzumab. Exosomes secreted by HER2-overexpressing SK-BR-3 and br-474 breast cancer cells can bind to trastuzumab. In the early stage of breast cancer, the binding level of exosomes isolated from the patients is lower than that of patients at late stages of the disease (44). In ovarian cancer cisplatin-resistant cells, cisplatin can promote the development of drug resistance through exosomes (45). When the cells were treated with cisplatin, the exosomes from cisplatin-resistant ovarian cancer cells contained 2.6 times more cisplatin compared with those from cisplatin-sensitive ovarian cancer cells (45). The levels of the cisplatin transporters, multidrug resistance-associated protein 2, ATPase copper transporting α and ATPase copper transporting β, were also higher in the exosomes from the resistant cells compared with those from the sensitive cells (45). Exosomes can induce drug resistance of tumor cells via the efflux of chemotherapeutic drugs. Exosomes released from human ovarian carcinoma cells can export cisplatin via exosomes (45). Exosomes can also mediate chemotherapy resistance through priming cancer stem cells (CSCs) (46). Hu et al (46) revealed that cancer-associated fibroblast (CAF)-derived exosomes promote the chemoresistance of colorectal cancer cells by priming CSCs. Drug-sensitive cells can be transformed to drug resistant by acquiring exosomes from chemoresistant cells. In breast cancer, drug resistance can be transferred by glutathione S-transferase P1 (GSTP1)-containing exosomes. GSTP1 can conjugate with glutathione and detoxify chemotherapy drugs (47). Chemosensitive breast cancer cells exhibit increased resistance to anticancer drugs after acquiring the exosomes from chemoresistant breast cancer cells (47). Additionally, the expression levels of GSTP1 in exosomes are associated with the clinical outcomes of patients with breast cancer (47).

Role of exosomes in the TME

Tumor-related fibroblasts are the most important cells in the TME (48). There are a number of theories regarding the formation of tumor fibroblasts, such as the differentiation from myofibroblasts to fibroblasts, the transdifferentiation of epithelial cells and endothelial cells to fibroblasts via EMT, or the transdifferentiation of mesenchymal stem cells (MSCs) to fibroblasts (49). Exosomes can transfer miR-9 to human breast fibroblasts and transform them into tumor-related fibroblasts (50). Tumor-derived exosomes can increase transforming growth factor-β (TGF-β) expression and signals in mesenchymal cells (50). For example, exosomes from gastric, ovarian and breast cancer can transform MSCs into fibroblast-like cells, which is manifested by the increase in the levels of α-smooth muscle actin (α-SMA) and Smad2 phosphorylation (5153). Furthermore, exosomes derived from gastric cancer cells can transform MSCs derived from cord blood into tumor-related fibroblasts (53). These studies demonstrated that exosomes promote tumor stromal cells to acquire tumor-promoting properties (5153).

As the soil of tumor cell proliferation, the TME regulates the proliferation, metastasis and vascular growth of tumor cells (54). The TME is rich in mesenchymal cells, fibroblasts, endothelial cells, extracellular matrix components, CAFs, and immune cells, including T lymphocytes, B lymphocytes, dendritic cells, macrophages and neutrophils (48). Exosomes can also promote tumor cell proliferation and angiogenesis (4). Vascular endothelial growth factor (VEGF) (55), fibroblast growth factor, TGF (56), platelet-derived growth factor and IL-8 can be used as exosomal proteins to promote the angiogenesis of target cells (57). The exosome-mediated interaction between chronic myeloid leukemia cells and human bone marrow stromal cells can promote the survival of IL-8-dependent leukemia cells (57). Chronic myeloid leukemia cells secrete exosomes to stimulate the secretion of IL-8 from bone marrow stromal cells, resulting in the regulation of angiogenesis and lymphocyte survival (57). Zhu et al (58) reported that exosomes from human bone marrow MSCs can promote the expression of tumor VEGF and activate ERK1/2 in vivo, enhancing tumor cell proliferation.

Exosomes from breast cancer cells can transform adipose tissue-derived MSCs into fibroblast-like cells via the Smad signaling pathway (51). This process is associated with increased expression levels of α-SMA, tumor-promoting factors TGF receptors I and II, stromal cell-derived factor 1 (SDF-1), VEGF, C-C motif chemokine ligand 5, and transglutaminases (51). Exosomes derived from ovarian cancer cells can also induce adipose tissue-derived MSCs to obtain cells with functional characteristics of fibroblasts (52). During this process, the expression levels of α-SMA, tumor-promoting factor SDF-1 and TGF-β in the adipose tissue-derived MSCs also increase, correlating to the increase in the expression levels of TGF-β receptor I and II (52).

Exosomes from the phosphorous cell carcinoma TME can increase the activity of the TGF-β signaling pathway in squamous cell carcinoma (59). TGF-β can activate Smad2 and Smad3 signaling pathways and regulate gene transcription by binding to the TGF-β receptor (59). The exosomes from tumor stromal fibroblasts contain TGF-β type II receptor (TβRII), a receptor component of TGF-β (59). The transferred receptor component increases the TβRII and Smad2 phosphorylation levels in squamous cell carcinoma cells (59).

Exosomes secreted by CLL cells can induce stromal cells to transform into tumor-associated fibroblasts (57). Paggetti et al (57) revealed that when PKH67 fluorescence-labeled CLL cells were co-cultured with MSCs and endothelial cells from the bone marrow, the exosomes from the CLL cells were absorbed by the stromal cells. This event was also associated with the transfer of miRNAs and proteins into these recipient cells, resulting in the activation of their inflammatory signals, such as the NF-κB signaling pathway, the transformation of chronic lymphocytic leukemia stromal cells into tumor-related fibroblasts, the upregulation of the expression levels of genes encoding cytokines and chemokines (C-X-C motif chemokine ligand 1, IL6, IL34 and C-C motif chemokine ligand 2) and migration-related factors (intercellular adhesion molecule 1 and MMP1), and the induction of the proliferation and migration of these stromal cells (57). This study not only provided valuable evidence for the regulatory effect of exosomes on the TME but also indicated that tumor cells act on tumor stromal cells and induce the formation of tumor-related fibroblasts (57). Webber et al (60) revealed that TGF-β delivery by tumor exosomes was sufficient to transform fibroblasts into myofibroblasts. Myofibroblasts are activated during the process of wound healing and express α-SMA. Myofibroblasts can be activated in the stroma of solid cancer and support the metastasis of cancer cells (61,62).

EMT regulated by exosomes

EMT is one of the key mechanisms for malignant cells to achieve metastasis (63). When tumor cells undergo EMT, the cells lose epithelial cell traits and acquire a mesenchymal phenotype, especially the hummingbird phenotype (63). Cancer cells that underwent EMT would exhibit downregulated E-cadherin expression and exhibit upregulated expression levels of N-cadherin, twist, snail and vimentin (63). Tumor cells acquire a migratory ability, invade tissues and vessels, and are transported by the circulation, which facilitates the remote colonization of other tissues (63). Exosomes can have robust impacts in the activation of the EMT machinery (64).

Exosomes can deliver oncogenes to the receipt cells to promote EMT and stem cell traits (65). For example, latent membrane protein 1 can be transported to the receipt cells by exosomes (65). Exosomes can also deliver miRNAs to recipient cells (66). For instance, exosomes can deliver miRNA to recipient cells to promote liver cancer EMT and metastasis (66). CAFs can enhance the EMT of oral cancer cells via delivery of miR-34a-5p by exosomes; miR-34a-5p in the exosome will activate EMT by the AKT/GSK-3β/β-catenin signaling pathway (67). By taking up the exosomal miRNA that was secreted in the tumor environment, the oral cancer cells increase their metastatic ability.

Hypoxic bone marrow MSC-derived exosomal miRNAs promote metastasis of lung cancer cells via a STAT3-induced EMT (68). Zhang et al (68) reported that exosomes derived from hypoxic bone marrow MSCs can be taken up by lung cancer cells, which activates STAT3 signaling and induces EMT in recipient cells, These plasma exosomes containing miRNA in the patients with lung cancer also showed a diagnostic value (68).

Cancer-derived exosomes can transfer miRNAs to macrophages to induce their M2 polarization and activate EMT in recipient cells (69). In colon cancer, exosomes derived from colon cancer cells can promote the polarization of macrophages into an M2 phenotype (69). Colorectal cancer cells deliver exosomes to macrophages and promote the upregulation of miRNA-106b-5p (miR-106b) in macrophages (69). This process further activates the PI3Kγ/AKT/mTOR signaling cascade in macrophages, prompting them to polarize into an M2 phenotype (69). The polarized M2 macrophages will further promote the EMT and metastasis of colorectal cancer cells (69). CSCs of clear cell renal cell carcinoma (CCRCC) contain integrin CD103, which can be transported to cancer cells by activating EMT through PTEN targeting (70). Bioactive miR-19b-3p is transported to recipient cells by cancer stem cell-derived exosomes, where miR-19b-3p activates CCRCC EMT by targeting PTEN (70). miRNAs are small non-coding single-stranded RNA molecules with a length of 22 nucleotides; miRNAs can regulate gene expression at the post-transcriptional level (71). Sun et al (72) reported that exosomes derived from colorectal cancer cells express high levels of miR-335-5p, which is transported to colorectal cells to promote EMT and migration of the recipient cells by the RAS p21 protein activator 1.

Immunoregulatory roles of exosomes

Exosomes from the TME can serve immunoregulatory roles in the communication of cancer cells (73) Exosomes can circumvent immune defenses to cancer cells through multiple mechanisms (74). Exosomes can promote an immunosuppressive environment through expanding the pool of CD4+/CD25+ regulatory T cells (75). They can also inhibit the proliferation of immune cells (53). For example, tumor-derived exosomes can induce the apoptosis of tumor activated CD8+ T lymphocytes (76). Exosomes derived from dendritic cells can directly present antigen to T cells (77,78). Exosomes derived from dendritic cells contain functional major histocompatibility complex (MHC)-peptide complexes, which can be presented on their surface and transferred to recipient cells (79). Exosomes carrying MHC-peptide complexes can be captured, processed and presented by antigen-presenting cells (APCs) (79).

Tumor antigens can be transferred by exosomes to antigen presenting cells, including macrophages and dendritic cells, after phagocytosis of exosomes by APCs and release of the exosomal antigens into the cytoplasm of APCs (80). For example, HSPs (HSP70-80) can be transferred to dendritic cells by exosomes and induces CD8+ T cell activation (81).

Exosomal antigen presentation to T cells can also occur indirectly. For instance, ‘cross-dressing’ involves the delivery of MHC-peptide complexes to antigen presenting cells through the fusion of antigen-presenting cells with exosomes derived from dendritic cells (82). This study has demonstrated the activation of CD4+ T cells in vivo following injection of exosomes carrying antigens (82). With the presence of MHC class II-negative but T cell costimulatory molecules-positive dendritic cells, antigen-bearing exosomes can activate T cells in vitro (82). This indicates that peptide-MHC complexes can be exchanged by exosomes, and presented to T cells to initiate an adaptive immune response (82).

Conclusion

The interaction between tumor cells and the TME is mediated by exosomes. The role of tumor cells and the TME is not a one-way information transfer but a two-way information exchange (29). Exosomes as communicators between cancer cells and the tumor environment are shown in Fig. 2. The targeted killing of tumor cells is not sufficient for the treatment of tumors. The TME is vital in tumor occurrence, and thus, has gained attention. Exosomes are messengers between tumor cells and the TME. Therefore, exosomes can be used as tools for the development of novel therapeutic strategies (83).

At present, researchers can trace exosomes, for example through the use of labeling tetraspanins, including CD63, CD9 and CD82, which can also help visualize the interaction between exosomes and tumor stromal cells. Researchers can use a technique based on the Cre recombinase-locus of X (cross)-over in P1 system to label exosome transportation by immunofluorescent signals (84). When the exosomes are released by cells expressing the Cre recombinase, they are absorbed by Cre reporter cells, and the latter can display fluorescence signals. Based on this technique, researchers can track exosomes in vivo and in vitro (84).

The interaction between exosomes and the TME regulates all aspects of tumor development, including the promotion of tumor cell proliferation, the enhancement of stromal cell transformation, the induction of tolerance of tumor cells to chemotherapeutic drugs and the promotion of distant metastasis (54). Participation of exosomes in all aspects of tumor biological mechanisms makes them good targets for antitumor treatment. Blocking the production and release of exosomes and their uptake by receptor cells may be an alternative treatment strategy for the development of an antitumor treatment that targets exosomes.

Acknowledgements

Not applicable.

Funding

Funding: No funding was received.

Availability of data and materials

Not applicable.

Authors' contributions

QL wrote, read and approved the final manuscript. Data authentication is not applicable.

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.

References

1 

Johnstone RM, Adam M, Hammond JR, Orr L and Turbide C: Vesicle formation during reticulocyte maturation. Association of plasma membrane activities with released vesicles (exosomes). J Biol Chem. 262:9412–9420. 1987. View Article : Google Scholar : PubMed/NCBI

2 

Johnstone RM: Revisiting the road to the discovery of exosomes. Blood Cells Mol Dis. 34:214–219. 2005. View Article : Google Scholar : PubMed/NCBI

3 

Colombo M, Raposo G and Théry C: Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu Rev Cell Dev Biol. 30:255–289. 2014. View Article : Google Scholar

4 

Kalluri R: The biology and function of exosomes in cancer. J Clin Invest. 126:1208–1215. 2016. View Article : Google Scholar

5 

Kahlert C, Melo SA, Protopopov A, Tang J, Seth S, Koch M, Zhang J, Weitz J, Chin L, Futreal A and Kalluri R: Identification of double-stranded genomic DNA spanning all chromosomes with mutated KRAS and p53 DNA in the serum exosomes of patients with pancreatic cancer. J Biol Chem. 289:3869–3875. 2014. View Article : Google Scholar

6 

Escola JM, Kleijmeer MJ, Stoorvogel W, Griffith JM, Yoshie O and Geuze HJ: Selective enrichment of tetraspan proteins on the internal vesicles of multivesicular endosomes and on exosomes secreted by human B-lymphocytes. J Biol Chem. 273:20121–20127. 1998. View Article : Google Scholar : PubMed/NCBI

7 

Vlassov AV, Magdaleno S, Setterquist R and Conrad R: Exosomes: Current knowledge of their composition, biological functions, and diagnostic and therapeutic potentials. Biochim Biophys Acta. 1820:940–948. 2012. View Article : Google Scholar

8 

Bobrie A, Colombo M, Krumeich S, Raposo G and Théry C: Diverse subpopulations of vesicles secreted by different intracellular mechanisms are present in exosome preparations obtained by differential ultracentrifugation. J Extracell Vesicles. 1:2012. View Article : Google Scholar : PubMed/NCBI

9 

Théry C, Boussac M, Véron P, Ricciardi-Castagnoli P, Raposo G, Garin J and Amigorena S: Proteomic analysis of dendritic cell-derived exosomes: A secreted subcellular compartment distinct from apoptotic vesicles. J Immunol. 166:7309–7318. 2001. View Article : Google Scholar

10 

Chen H, Chengalvala V, Hu H and Sun D: Tumor-derived exosomes: Nanovesicles made by cancer cells to promote cancer metastasis. Acta Pharm Sin B. 11:2136–2149. 2021. View Article : Google Scholar : PubMed/NCBI

11 

Raulf N, Lucarelli P, Thavaraj S, Brown S, Vicencio JM, Sauter T and Tavassoli M: Annexin A1 regulates EGFR activity and alters EGFR-containing tumour-derived exosomes in head and neck cancers. Eur J Cancer. 102:52–68. 2018. View Article : Google Scholar : PubMed/NCBI

12 

Dai J, Su Y, Zhong S, Cong L, Liu B, Yang J, Tao Y, He Z, Chen C and Jiang Y: Exosomes: Key players in cancer and potential therapeutic strategy. Signal Transduct Target Ther. 5:1452020. View Article : Google Scholar : PubMed/NCBI

13 

Sun W, Luo JD, Jiang H and Duan DD: Tumor exosomes: A double-edged sword in cancer therapy. Acta Pharmacol Sin. 39:534–541. 2018. View Article : Google Scholar : PubMed/NCBI

14 

Wollert T and Hurley JH: Molecular mechanism of multivesicular body biogenesis by ESCRT complexes. Nature. 464:864–869. 2010. View Article : Google Scholar : PubMed/NCBI

15 

Raiborg C and Stenmark H: The ESCRT machinery in endosomal sorting of ubiquitylated membrane proteins. Nature. 458:445–452. 2009. View Article : Google Scholar : PubMed/NCBI

16 

Michelet X, Djeddi A and Legouis R: Developmental and cellular functions of the ESCRT machinery in pluricellular organisms. Biol Cell. 102:191–202. 2010. View Article : Google Scholar

17 

Bhuin T and Roy JK: Rab proteins: The key regulators of intracellular vesicle transport. Exp Cell Res. 328:1–19. 2014. View Article : Google Scholar

18 

Ostrowski M, Carmo NB, Krumeich S, Fanget I, Raposo G, Savina A, Moita CF, Schauer K, Hume AN, Freitas RP, et al: Rab27a and Rab27b control different steps of the exosome secretion pathway. Nat Cell Biol. 12:19–30. 1–13. 2010. View Article : Google Scholar

19 

Chaput N, Flament C, Viaud S, Taieb J, Roux S, Spatz A, André F, LePecq JB, Boussac M, Garin J, et al: Dendritic cell derived-exosomes: Biology and clinical implementations. J Leukoc Biol. 80:471–478. 2006. View Article : Google Scholar

20 

Li W, Li C, Zhou T, Liu X, Liu X, Li X and Chen D: Role of exosomal proteins in cancer diagnosis. Mol Cancer. 16:1452017. View Article : Google Scholar : PubMed/NCBI

21 

Li Q, Shao Y, Zhang X, Zheng T, Miao M, Qin L, Wang B, Ye G, Xiao B and Guo J: Plasma long noncoding RNA protected by exosomes as a potential stable biomarker for gastric cancer. Tumour Biol. 36:2007–2012. 2015. View Article : Google Scholar

22 

Smolarz M and Widlak P: Serum exosomes and their miRNA load-a potential biomarker of lung cancer. Cancers (Basel). 13:13732021. View Article : Google Scholar : PubMed/NCBI

23 

Lorenc T, Klimczyk K, Michalczewska I, Słomka M, Kubiak-Tomaszewska G and Olejarz W: Exosomes in prostate cancer diagnosis, prognosis and therapy. Int J Mol Sci. 21:21182020. View Article : Google Scholar

24 

Zhang J, Li S, Li L, Li M, Guo C, Yao J and Mi S: Exosome and exosomal microRNA: Trafficking, sorting, and function. Genomics Proteomics Bioinformatics. 13:17–24. 2015. View Article : Google Scholar : PubMed/NCBI

25 

Gutkin A, Uziel O, Beery E, Nordenberg J, Pinchasi M, Goldvaser H, Henick S, Goldberg M and Lahav M: Tumor cells derived exosomes contain hTERT mRNA and transform nonmalignant fibroblasts into telomerase positive cells. Oncotarget. 7:59173–59188. 2016. View Article : Google Scholar

26 

Farahani M, Rubbi C, Liu L, Slupsky JR and Kalakonda N: CLL exosomes modulate the transcriptome and behaviour of recipient stromal cells and are selectively enriched in miR-202-3p. PLoS One. 10:e01414292015. View Article : Google Scholar : PubMed/NCBI

27 

Zhuang G, Wu X, Jiang Z, Kasman I, Yao J, Guan Y, Oeh J, Modrusan Z, Bais C, Sampath D and Ferrara N: Tumour-secreted miR-9 promotes endothelial cell migration and angiogenesis by activating the JAK-STAT pathway. EMBO J. 31:3513–3523. 2012. View Article : Google Scholar : PubMed/NCBI

28 

Mathivanan S, Ji H and Simpson RJ: Exosomes: Extracellular organelles important in intercellular communication. J Proteomics. 73:1907–1920. 2010. View Article : Google Scholar : PubMed/NCBI

29 

Kharaziha P, Ceder S, Li Q and Panaretakis T: Tumor cell-derived exosomes: A message in a bottle. Biochim Biophys Acta. 1826:103–111. 2012.

30 

Hood JL, San RS and Wickline SA: Exosomes released by melanoma cells prepare sentinel lymph nodes for tumor metastasis. Cancer Res. 71:3792–3801. 2011. View Article : Google Scholar

31 

Luga V and Wrana JL: Tumor-stroma interaction: Revealing fibroblast-secreted exosomes as potent regulators of Wnt-planar cell polarity signaling in cancer metastasis. Cancer Res. 73:6843–6847. 2013. View Article : Google Scholar : PubMed/NCBI

32 

Nogués L, Benito-Martin A, Hergueta-Redondo M and Peinado H: The influence of tumour-derived extracellular vesicles on local and distal metastatic dissemination. Mol Aspects Med. 60:15–26. 2018. View Article : Google Scholar

33 

Hanahan D and Weinberg RA: Hallmarks of cancer: The next generation. Cell. 144:646–674. 2011. View Article : Google Scholar

34 

Maia J, Caja S, Strano Moraes MC, Couto N and Costa-Silva B: Exosome-based cell-cell communication in the tumor microenvironment. Front Cell Dev Biol. 6:182018. View Article : Google Scholar

35 

Luga V, Zhang L, Viloria-Petit AM, Ogunjimi AA, Inanlou MR, Chiu E, Buchanan M, Hosein AN, Basik M and Wrana JL: Exosomes mediate stromal mobilization of autocrine Wnt-PCP signaling in breast cancer cell migration. Cell. 151:1542–1556. 2012. View Article : Google Scholar

36 

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

37 

Rahman MA, Barger JF, Lovat F, Gao M, Otterson GA and Nana-Sinkam P: Lung cancer exosomes as drivers of epithelial mesenchymal transition. Oncotarget. 7:54852–54866. 2016. View Article : Google Scholar

38 

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

39 

Zhang H, Deng T, Liu R, Bai M, Zhou L, Wang X, Li S, Wang X, Yang H, Li J, et al: Exosome-delivered EGFR regulates liver microenvironment to promote gastric cancer liver metastasis. Nat Commun. 8:150162017. View Article : Google Scholar : PubMed/NCBI

40 

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

41 

Torreggiani E, Roncuzzi L, Perut F, Zini N and Baldini N: Multimodal transfer of MDR by exosomes in human osteosarcoma. Int J Oncol. 49:189–196. 2016. View Article : Google Scholar

42 

Wang J, Hendrix A, Hernot S, Lemaire M, De Bruyne E, Van Valckenborgh E, Lahoutte T, De Wever O, Vanderkerken K and Menu E: Bone marrow stromal cell-derived exosomes as communicators in drug resistance in multiple myeloma cells. Blood. 124:555–566. 2014. View Article : Google Scholar : PubMed/NCBI

43 

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

44 

Ciravolo V, Huber V, Ghedini GC, Venturelli E, Bianchi F, Campiglio M, Morelli D, Villa A, Della Mina P, Menard S, et al: Potential role of HER2-overexpressing exosomes in countering trastuzumab-based therapy. J Cell Physiol. 227:658–567. 2012. View Article : Google Scholar

45 

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

46 

Hu Y, Yan C, Mu L, Huang K, Li X, Tao D, Wu Y and Qin J: Fibroblast-derived exosomes contribute to chemoresistance through priming cancer stem cells in colorectal cancer. PLoS One. 10:e01256252015. View Article : Google Scholar

47 

Yang SJ, Wang DD, Li J, Xu HZ, Shen HY, Chen X, Zhou SY, Zhong SL, Zhao JH and Tang JH: Predictive role of GSTP1-containing exosomes in chemotherapy-resistant breast cancer. Gene. 623:5–14. 2017. View Article : Google Scholar : PubMed/NCBI

48 

Kim J and Bae JS: Tumor-associated macrophages and neutrophils in tumor microenvironment. Mediators Inflamm. 2016:60581472016. View Article : Google Scholar : PubMed/NCBI

49 

Xouri G and Christian S: Origin and function of tumor stroma fibroblasts. Semin Cell Dev Biol. 21:40–46. 2010. View Article : Google Scholar

50 

Baroni S, Romero-Cordoba S, Plantamura I, Dugo M, D'Ippolito E, Cataldo A, Cosentino G, Angeloni V, Rossini A, Daidone MG and Iorio MV: Exosome-mediated delivery of miR-9 induces cancer-associated fibroblast-like properties in human breast fibroblasts. Cell Death Dis. 7:e23122016. View Article : Google Scholar : PubMed/NCBI

51 

Cho JA, Park H, Lim EH and Lee KW: Exosomes from breast cancer cells can convert adipose tissue-derived mesenchymal stem cells into myofibroblast-like cells. Int J Oncol. 40:130–138. 2012.

52 

Cho JA, Park H, Lim EH, Kim KH, Choi JS, Lee JH, Shin JW and Lee KW: Exosomes from ovarian cancer cells induce adipose tissue-derived mesenchymal stem cells to acquire the physical and functional characteristics of tumor-supporting myofibroblasts. Gynecol Oncol. 123:379–386. 2011. View Article : Google Scholar

53 

Gu J, Qian H, Shen L, Zhang X, Zhu W, Huang L, Yan Y, Mao F, Zhao C, Shi Y and Xu W: Gastric cancer exosomes trigger differentiation of umbilical cord derived mesenchymal stem cells to carcinoma-associated fibroblasts through TGF-β/Smad pathway. PLoS One. 7:e524652012. View Article : Google Scholar : PubMed/NCBI

54 

Milane L, Singh A, Mattheolabakis G, Suresh M and Amiji MM: Exosome mediated communication within the tumor microenvironment. J Control Release. 219:278–294. 2015. View Article : Google Scholar : PubMed/NCBI

55 

Han Y, Ren J, Bai Y, Pei X and Han Y: Exosomes from hypoxia-treated human adipose-derived mesenchymal stem cells enhance angiogenesis through VEGF/VEGF-R. Int J Biochem Cell Biol. 109:59–68. 2019. View Article : Google Scholar

56 

Li Z, Zeng C, Nong Q, Long F, Liu J, Mu Z, Chen B, Wu D and Wu H: Exosomal leucine-rich-alpha2-glycoprotein 1 derived from non-small-cell lung cancer cells promotes angiogenesis via TGF-β signal pathway. Mol Ther Oncolytics. 14:313–322. 2019. View Article : Google Scholar

57 

Paggetti J, Haderk F, Seiffert M, Janji B, Distler U, Ammerlaan W, Kim YJ, Adam J, Lichter P, Solary E, et al: Exosomes released by chronic lymphocytic leukemia cells induce the transition of stromal cells into cancer-associated fibroblasts. Blood. 126:1106–1117. 2015. View Article : Google Scholar : PubMed/NCBI

58 

Zhu W, Huang L, Li Y, Zhang X, Gu J, Yan Y, Xu X, Wang M, Qian H and Xu W: Exosomes derived from human bone marrow mesenchymal stem cells promote tumor growth in vivo. Cancer Lett. 315:28–37. 2012. View Article : Google Scholar

59 

Languino LR, Singh A, Prisco M, Inman GJ, Luginbuhl A, Curry JM and South AP: Exosome-mediated transfer from the tumor microenvironment increases TGFβ signaling in squamous cell carcinoma. Am J Transl Res. 8:2432–2437. 2016.PubMed/NCBI

60 

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

61 

Olumi AF, Grossfeld GD, Hayward SW, Carroll PR, Tlsty TD and Cunha GR: Carcinoma-associated fibroblasts direct tumor progression of initiated human prostatic epithelium. Cancer Res. 59:5002–5011. 1999.PubMed/NCBI

62 

Orimo A, Gupta PB, Sgroi DC, Arenzana-Seisdedos F, Delaunay T, Naeem R, Carey VJ, Richardson AL and Weinberg RA: Stromal fibroblasts present in invasive human breast carcinomas promote tumor growth and angiogenesis through elevated SDF-1/CXCL12 secretion. Cell. 121:335–348. 2005. View Article : Google Scholar

63 

Bakir B, Chiarella AM, Pitarresi JR and Rustgi AK: EMT, MET, plasticity, and tumor metastasis. Trends Cell Biol. 30:764–776. 2020. View Article : Google Scholar

64 

Kim H, Lee S, Shin E, Seong KM, Jin YW, Youn H and Youn B: The emerging roles of exosomes as EMT regulators in cancer. Cells. 9:8612020. View Article : Google Scholar

65 

Yoshizaki T, Kondo S, Wakisaka N, Murono S, Endo K, Sugimoto H, Nakanishi S, Tsuji A and Ito M: Pathogenic role of Epstein-Barr virus latent membrane protein-1 in the development of nasopharyngeal carcinoma. Cancer Lett. 337:1–7. 2013. View Article : Google Scholar

66 

Lin Q, Zhou CR, Bai MJ, Zhu D, Chen JW, Wang HF, Li MA, Wu C, Li ZR and Huang MS: Exosome-mediated miRNA delivery promotes liver cancer EMT and metastasis. Am J Transl Res. 12:1080–1095. 2020.PubMed/NCBI

67 

Li YY, Tao YW, Gao S, Li P, Zheng JM, Zhang SE, Liang J and Zhang Y: Cancer-associated fibroblasts contribute to oral cancer cells proliferation and metastasis via exosome-mediated paracrine miR-34a-5p. EbioMedicine. 36:209–220. 2018. View Article : Google Scholar : PubMed/NCBI

68 

Zhang X, Sai B, Wang F, Wang L, Wang Y, Zheng L, Li G, Tang J and Xiang J: Hypoxic BMSC-derived exosomal miRNAs promote metastasis of lung cancer cells via STAT3-induced EMT. Mol Cance. 18:402019. View Article : Google Scholar

69 

Yang C, Dou R, Wei C, Liu K, Shi D, Zhang C, Liu Q, Wang S and Xiong B: Tumor-derived exosomal microRNA-106b-5p activates EMT-cancer cell and M2-subtype TAM interaction to facilitate CRC metastasis. Mol Ther. 29:2088–2107. 2021. View Article : Google Scholar : PubMed/NCBI

70 

Wang L, Yang G, Zhao D, Wang J, Bai Y, Peng Q, Wang H, Fang R, Chen G, Wang Z, et al: CD103-positive CSC exosome promotes EMT of clear cell renal cell carcinoma: Role of remote MiR-19b-3p. Mol Cancer. 18:862019. View Article : Google Scholar : PubMed/NCBI

71 

Mishra S, Yadav T and Rani V: Exploring miRNA based approaches in cancer diagnostics and therapeutics. Crit Rev Oncol Hematol. 98:12–23. 2016. View Article : Google Scholar

72 

Sun X, Lin F, Sun W, Zhu W, Fang D, Luo L, Li S, Zhang W and Jiang L: Exosome-transmitted miRNA-335-5p promotes colorectal cancer invasion and metastasis by facilitating EMT via targeting RASA1. Mol Ther Nucleic Acids. 24:164–174. 2021. View Article : Google Scholar : PubMed/NCBI

73 

Olejarz W, Dominiak A, Żołnierzak A, Kubiak-Tomaszewska G and Lorenc T: Tumor-derived exosomes in immunosuppression and immunotherapy. J Immunol Res. 2020:62724982020. View Article : Google Scholar : PubMed/NCBI

74 

Graner MW, Schnell S and Olin MR: Tumor-derived exosomes, microRNAs, and cancer immune suppression. Semin Immunopathol. 40:505–515. 2018. View Article : Google Scholar

75 

Szajnik M, Czystowska M, Szczepanski MJ, Mandapathil M and Whiteside TL: Tumor-derived microvesicles induce, expand and up-regulate biological activities of human regulatory T cells (Treg). PLoS One. 5:e114692010. View Article : Google Scholar : PubMed/NCBI

76 

Wieckowski EU, Visus C, Szajnik M, Szczepanski MJ, Storkus WJ and Whiteside TL: Tumor-derived microvesicles promote regulatory T cell expansion and induce apoptosis in tumor-reactive activated CD8+ T lymphocytes. J Immunol. 183:3720–3730. 2009. View Article : Google Scholar

77 

Pitt JM, André F, Amigorena S, Soria JC, Eggermont A, Kroemer G and Zitvogel L: Dendritic cell-derived exosomes for cancer therapy. J Clin Invest. 126:1224–1232. 2016. View Article : Google Scholar

78 

Tkach M, Kowal J, Zucchetti AE, Enserink L, Jouve M, Lankar D, Saitakis M, Martin-Jaular L and Théry C: Qualitative differences in T-cell activation by dendritic cell-derived extracellular vesicle subtypes. EMBO J. 36:3012–3028. 2017. View Article : Google Scholar : PubMed/NCBI

79 

Montecalvo A, Shufesky WJ, Stolz DB, Sullivan MG, Wang Z, Divito SJ, Papworth GD, Watkins SC, Robbins PD, Larregina AT and Morelli AE: Exosomes as a short-range mechanism to spread alloantigen between dendritic cells during T cell allorecognition. J Immunol. 180:3081–3090. 2008. View Article : Google Scholar

80 

Whiteside TL: Exosomes and tumor-mediated immune suppression. J Clin Invest. 126:1216–1223. 2016. View Article : Google Scholar

81 

Wolfers J, Lozier A, Raposo G, Regnault A, Théry C, Masurier C, Flament C, Pouzieux S, Faure F, Tursz T, et al: Tumor-derived exosomes are a source of shared tumor rejection antigens for CTL cross-priming. Nat Med. 7:297–303. 2001. View Article : Google Scholar : PubMed/NCBI

82 

Théry C, Duban L, Segura E, Véron P, Lantz O and Amigorena S: Indirect activation of naïve CD4+ T cells by dendritic cell-derived exosomes. Nat Immunol. 3:1156–1162. 2002. View Article : Google Scholar

83 

Xu Z, Zeng S, Gong Z and Yan Y: Exosome-based immunotherapy: A promising approach for cancer treatment. Mol Cancer. 19:1602020. View Article : Google Scholar : PubMed/NCBI

84 

Mohr M, Tosun S, Arnold WH, Edenhofer F, Zänker KS and Dittmar T: Quantification of cell fusion events human breast cancer cells and breast epithelial cells using a Cre-LoxP-based double fluorescence reporter system. Cell Mol Life Sci. 72:3769–3782. 2015. View Article : Google Scholar

Related Articles

Journal Cover

July-2022
Volume 24 Issue 1

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Li Q: Role of exosomes in cellular communication between tumor cells and the tumor microenvironment (Review). Oncol Lett 24: 240, 2022
APA
Li, Q. (2022). Role of exosomes in cellular communication between tumor cells and the tumor microenvironment (Review). Oncology Letters, 24, 240. https://doi.org/10.3892/ol.2022.13360
MLA
Li, Q."Role of exosomes in cellular communication between tumor cells and the tumor microenvironment (Review)". Oncology Letters 24.1 (2022): 240.
Chicago
Li, Q."Role of exosomes in cellular communication between tumor cells and the tumor microenvironment (Review)". Oncology Letters 24, no. 1 (2022): 240. https://doi.org/10.3892/ol.2022.13360