Open Access

Role of stem cell-derived exosomes in cancer (Review)

  • Authors:
    • Junyi Wu
    • Zhen Qu
    • Zi‑Wei Fei
    • Jun‑Hua Wu
    • Chun‑Ping Jiang
  • View Affiliations

  • Published online on: March 7, 2017     https://doi.org/10.3892/ol.2017.5824
  • Pages: 2855-2866
  • Copyright: © Wu et al. 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 small, extracellular membrane-enclosed vesicles that contain a variety of molecules, including proteins, DNA, mRNA and non-coding RNA; these vesicles have been defined as new tools for intercellular communication between cells. Numerous types of cells, including stem cells, secrete exosomes into the extracellular environment, and are significant communicators in the tumor microenvironment. Stem cells are a unique cell population defined by their ability to indefinitely self-renew, differentiate into a variety of cell lines, and form clonal cell populations. Stem cells also secrete large amounts of exosomes, which have demonstrated great potential in a variety of diseases. Increasing evidence has revealed that the mechanism of interaction between stem cells and human tumor cells involves the exchange of biological material through exosomes. In this review, the latest developments in the role of stem cell‑derived exosomes in cancer are highlighted.

Introduction

Exosomes are small, lipid bilayer membrane vesicles of endocytic origin (30–100 nm in diameter). In recent years, there has been increasing interest in the relevance between biological and pathophysiological processes and these extracellular vesicles (EVs). Increasing evidence suggests that interactions between stem cells and human tumor cells involve the exchange of biological information via EVs, including exosomes (1). Stem cells secrete a large number of exosomes, which act as communicators in the tumor microenvironment and which play diverse roles in tumorigenesis, tumor angiogenesis and tumor metastases. However, the role of stem cell-derived exosomes in the pathophysiological processes of tumors has not been clarified until now. In this review, the recent findings with regard to the role of stem cell derived-exosomes in cancer are briefly summarized.

Biogenesis, contents and secretion of exosomes

Exosomes, which were first described in 1981, are derived from the internal vesicles of multivesicular bodies (MVBs) and consist of a lipid bilayer membrane surrounding a small amount of cytosol (2). Exosomes are secreted by all types of cells in culture and are observed in abundance in body fluids, including saliva, urine, blood and breast milk (3). In the present study, the current knowledge with regard to the biogenesis, contents and secretion of exosomes is summarized.

The formation of MVBs during exosome biogenesis is similar to the formation of MVBs during lysosome formation. First, the cell membrane is internalized to produce an endosome. Subsequently, the endosome forms inside a large number of small vesicles via invagination of portions of the endosome membrane. Such endosomes are called MVBs. Finally, exosomes are produced and packed with cytoplasmic contents, and the membrane of MVBs bulges inward and pinches off to create small membranous vesicles within the MVBs. The molecular mechanisms of exosome formation have been studied extensively; however, the exact mechanism of exosome packaging has not been fully clarified. Endosomal sorting complex required for transport (ESCRT)-dependent and ESCRT-independent signals have been suggested to be associated with the sorting of exosomes (4). ESCRT consists of four complexes and their associated proteins: ESCRT-0 identifies ubiquitinated proteins in the endosomal membrane, ESCRT-I and ESCRT-II direct endosomal membrane budding, and ESCRT-III facilitates separation from the endosomal membrane (5,6). Numerous studies have revealed that the ESCRT-0 protein hepatocyte growth factor-regulated tyrosine kinase substrate (HRS) is necessary for exosome formation (7). ESCRT-I members, including HeLa-CIITA, MCF-7 and tumor susceptibility gene 101 (TSG101), are also associated with another ESCRT-independent mechanism of exosome biogenesis. Research has demonstrated that ALG-2-interacting protein X (Alix), an ESCRT-III-associated protein, promotes exosome biogenesis and, thus, intraluminal budding of vesicles in endosomes. Two proteins that are degradation products of the ESCRT-III complex, namely vacuolar protein sorting-associated protein 4 (VPS4) and charged multivesicular body protein 4 (CHMP4), are also involved in exosome biogenesis (6). However, certain studies have demonstrated that ESCRT-independent signals were also involved in exosome biogenesis. These pathways may involve lipids including sphingosine-1-phosphate and four spin-enriched microdomains or heat shock proteins (6).

After exosomes are formed, they contain >14,000 biomolecules, including proteins, RNAs and DNA (8). In the late 1990s, the first ‘proteomic’ analyses of the protein composition of dendritic cell-derived exosomes were performed (9). Biochemically, exosomes contain common marker proteins [e.g., tetraspanins, including cluster of differentiation (CD) 9, CD10, CD26, CD53, CD63, CD81 and CD82], which are present in the exosomal membrane. Exosomes also contain Alix and TSG101, which are involved in the formation of MVBs. Two cytoplasmic heat shock proteins, Hsc70 and Hsp90, have also been observed in exosomes (3,10). mRNAs and microRNAs (miRNAs/miRs) were definitively identified in exosomes for the first time by Valadi et al (11). Thakur et al observed that exosomes from cancer cells contained double-stranded DNA that could reflect the mutational status of the originated cells (12). It has also been demonstrated that RNA carried in exosomes may be delivered to target cells and that the expression of genes in target cells is influenced by miRNAs contained in exosomes (13). In addition, mRNAs and miRNAs from different cells may be cell-type specific.

Exosomes may be secreted via the fusion of MVBs and the cell membrane, followed by the release of the contents of the MVBs (exosomes) into the extracellular environment. Alternatively, the contents of MVBs are degraded through lysosomes. There are numerous studies on the secretion of exosomes, and various proteins associated with this process. Rab2b, Rab5a, Rab7, Rab9a, Rab11, Rab27a, Rab27b and Rab35, members of the Rab family of small guanosine triphosphatase (GTPase) proteins, have been demonstrated to accurately regulate the secretion of exosomes (14). Soluble NSF-attachment protein receptor complexes are associated with the fusion of exosomes and the lipid bilayers (15). The accumulation of intracellular Ca2+ and intercellular pH has been observed to regulate the secretion of exosomes (16). In addition, heparanase overexpression promotes the secretion of exosomes (17). When exosomes are secreted, some of them are taken up by target cells localized near the cell of origin, while other exosomes are delivered to more distant sites through the blood or other biological fluids.

Uptake and functions of exosomes

In recent years, there has been increasing interest in intercellular communication via exosomes. A number of studies have attempted to determine the mechanism by which the cargo in exosomes is exchanged between exosomes and target cells. After exosomes are secreted, they may be taken up by the target cell via direct fusion with the plasma membrane, a receptor-ligand interaction, or endocytosis by phagocytosis (Fig. 1) (18,19). A number of biological molecules have significant roles in this process. Heat shock protein (HSP) 70, which is contained in exosomes, mediates the communication of cardioprotective signals to the heart and then activates a pathway downstream of toll-like receptor 4 (20). T-cell immunoglobulin- and mucin-domain-containing molecule, intercellular adhesion molecule 1 and heparan sulfate proteoglycans also influence the uptake of exosomes (16).

After exosomes are taken up by target cells, they play a vital role in cells. The primary function of exosomes in intercellular communication is the transfer of biologically active proteins, lipids and RNAs (21). Several studies have demonstrated that exosomes play crucial roles under normal and pathophysiological conditions, including lactation, the immune response, neuronal function and infectious diseases, as well as in the development and progression of liver disease, neurodegenerative diseases and cancer (22). Immune stimulation and tolerization are noted to be associated with exosomes; a previous study has suggested the potential use of exosomes in immunotherapy (23). Placental exosomes are involved in suppressive immunity during normal pregnancy (24). In addition, exosomes from human breast milk may contribute to the development of the infant immune system (23). The generation and progression of neurodegenerative diseases are also associated with exosomes. Exosomes transport proteins; thus, they may serve as a novel treatment approach or as new biomarkers in neurodegenerative diseases (24). In addition, exosomes have been promoted as specific therapeutic transporters for cardiovascular diseases (24), and are involved in the processes of infection biology, modulating the immune response and functioning as new acellular vaccines or infection biomarkers for infectious diseases (24). Additionally, exosomes are essential in the pathogenesis, diagnostics and therapeutics of liver diseases (24,25).

Increasing evidence has suggested that exosomes have significant roles in tumor growth, progression, metastasis and drug resistance (16). Tumor-derived exosomes regulate the formation of new blood vessels, which support tumor angiogenesis, and exosomes have crucial roles in tumor cell proliferation (16). In addition, exosomes induce the formation of the pre-metastatic niche, which regulates tumor metastasis (26). Exosome-mediated mechanisms also clearly contribute to tumor cell drug resistance (26). Certain exosomes have a significant influence on the ability of tumors to evade immune surveillance; however, exosomes from different sources may enhance the immune response (26). Except for the roles of exosomes in the pathogenesis of cancer, there are numerous studies on the use of exosomes as a new tool for cancer diagnosis and therapeutics (Table I).

Table I.

Functions of exosomes from different sources.

Table I.

Functions of exosomes from different sources.

ConditionsOutcomeReference
Normal conditionsCell-cell communication21
Removal of unnecessary protein from cell22
Immune function23,24,26
Liver diseasesPathogenesis, diagnostics and therapeutics23,25
Neurodegenerative diseasesTransporting proteins, a novel treatment approach or new biomarkers24
Neurodegenerative diseasesNew acellular vaccines or infection biomarkers for infectious diseases24
Infectious diseasesNew acellular vaccines or infection biomarkers for infectious diseases24
CancerGrowth16
Angiogenesis16
Progression26
Metastasis26
Drug resistance26
Manipulation of their microenvironment26
Immune response26
Diagnosis and therapeutics biomarker16

In addition to the abovementioned studies, a number studies have examined the functions of exosomes (16,23,24,26); however, which specific class of molecules contained in exosomes influences the target cells remains unclear.

Stem cells

Stem cells are a class of pluripotent cells that self-renew and differentiate into a variety of cell types. In this review, we focus on mesenchymal stem cells (MSCs) and cancer stem cells (CSCs).

MSCs, which originate from almost all vascularized organs and tissues, exhibit migratory capabilities and regenerative potential (27). MSCs may be isolated from various sources, including umbilical cord (UC), bone marrow (BM), liver, adipose tissue, multiple dental tissues and induced pluripotent stem cells (28,29). MSCs express CD44, CD73, CD90 and CD105, but not CD45, CD34 and CD14; these cells are characterized by their ability to adhere to plastics under standard cell culture conditions (30). MSCs affect the surrounding microenvironment by secreting factors, including growth factors, in an autocrine and paracrine manner. These cells decrease inflammation, promote angiogenesis, support tissue repair and suppress immunity (31). Since MSCs have the unique ability to home to damaged and cancerous tissue, they are of great interest in regenerative medicine and cancer therapy (32). MSCs also have a significant role in tumors. Previous studies have demonstrated that human MSCs (hMSCs) promote tumor growth and angiogenesis, in addition to providing stromal support (33,34), through autocrine and paracrine signaling (35,36). Although there are numerous studies of MSCs, the underlying mechanism of the correlation between MSCs and tumors remains largely unexplored. In recent years, a number of studies have noted that the interaction between MSCs and human tumor cells is associated with the exchange of biological material via EVs, including exosomes from MSCs (3739). In the present review, MSC-derived exosomes are highlighted, which have a significant role in tumors, in order to realize the mechanism of this interaction.

During the past decade, CSCs have been increasingly identified in a number of malignancies. CSCs have stem-like characteristics and exhibit numerous features of embryonic or tissue stem cells (40). CSCs are associated with tumor initiation, metastasis, progression, invasion, recurrence and resistance to therapies. It is becoming widely accepted that CSCs play a central role in cancer cell biology; CSCs are essential for cancer initiation, formation and relapse. Previous studies have suggested that CSC-derived exosomes act as a vehicle to deliver genetic information and produce a favorable microenvironment for cancer development (4143). In the present review, the role of CSC-derived exosomes in cancer is summarized.

Roles of stem cell-derived exosomes in cancer

Previous studies have demonstrated that stem cells generate a number of exosomes that may act as paracrine mediators by exchanging genetic information (38,39). There are certain differences between stem cell (MSC and CSC)-derived exosomes and other sources of exosomes. CSC-derived exosomes contain several proteins, including TSG101, Rab GTPases, annexins and signal transduction molecules (e.g., 14-3-3, a heterotrimeric G protein, and Alix), that are potentially associated with their biogenesis, targeting and putative immunological function (44). In addition, MSC-derived exosomes are amenable to immortalization without compromising exosome production, and are not immunogenic. MSC-derived exosomes have intrinsic therapeutic properties that reduce tissue injury.

Tumor growth

Stem cell-derived exosomes have been noted to deliver gene regulatory information to target cells; this information regulates cell growth and angiogenesis by modulating a variety of cellular pathways. There have been a number studies into the use of stem cell-derived exosomes to promote tumor cell proliferation in order to analyze the effects of cellular interactions between stem cells and various cancer cells. In recent years, studies have demonstrated that MSCs have a significant role in regulating tumor growth and metastasis (40,45). EVs, including exosomes isolated from hMSCs, were first completely biochemically and molecularly analyzed by Vallabhaneni et al (46). These authors used co-injection xenograft assays to demonstrate that the exosomes secreted from hMSCs support breast cancer cell proliferation and metastasis. They also observed that hMSC-derived EVs contain a large amount of miR-21 and 34a, which are tumor-supportive miRNAs, and ~150 different proteins, most of which are known tumor supportive factors, including platelet-derived growth factor receptor-β, tissue inhibitor of metalloproteinase (TIMP)-1 and TIMP-2. The presence of bioactive lipids, including sphingomyelin, was verified in EVs, including exosomes, through lipidomic assays. Furthermore, metabolite assays identified the presence of lactic acid and glutamic acid in the EVs. In addition, Zhu et al revealed that MSC-derived exosomes enhanced vascular endothelial growth factor (VEGF) expression in tumor cells by activating the extracellular signal-regulated kinase 1/2 (ERK1/2) pathway, which promotes tumor growth (47). These authors were the first to demonstrate that exosomes from MSCs have a role in promoting tumor growth that is similar to that of the MSCs themselves, thus providing new insight into the actions of MSCs in tumor development and progression in vivo. Hernanda et al observed that exosomes (or microvesicles) secreted by MSCs may also be associated with tumor promotion (48). Subsequently, Yang et al demonstrated that the internalization of MSC-derived exosomes was involved in the acquisition of new tumor cell properties by altering cellular functionalities and providing the capability to re-organize the tumor microenvironment, which improves tumor growth (49). However, the effects of exosomes from different types of stem cells on cell proliferation may be completely different. Del Fattore et al demonstrated that BM- and UC-MSC-EVs (including exosomes) suppressed cell proliferation, while the opposite effect was observed with adipose tissue MSC-EVs (including exosomes) (50). In addition, microvesicles from human BM-derived MSCs inhibited tumor growth (51). Another experiment indicated that intra-tumoral injection of exosomes derived from miR-146-expressing MSCs significantly reduced glioma xenograft growth in a rat primary brain tumor model (52). miR-146 suppressed epidermal growth factor receptor (EGFR) expression through binding-targeting EGFR mRNA, and reduced in vitro growth, migration and invasion of cancer (53). These studies revealed that miRNAs may be packaged into MSC exosomes, delivered to target tumor cells in culture, and reduce glioma cells, which suggested that the export of specific therapeutic miRNAs into MSC exosomes represented a new treatment strategy for malignant glioma. Additionally, although exosomes derived from multiple myeloma BM-MSCs have been noted to improve multiple myeloma progression, normal BM MSC-derived exosomes reduce tumor promotion (54). Exosomes from human umbilical cord Wharton's jelly MSCs reduced bladder tumor cell growth in vitro and in vivo (55). Thus, the effects of different sources of MSC-derived exosomes are uncertain.

Based on the aforementioned studies, it is known that exosomes secreted from MSCs may result in cell-to-cell transfer of mRNA, miRNA and proteins (56). However, the exact roles and mechanisms of MSC-produced exosomes in tumor biology remain largely elusive.

Cancer stem cell-derived exosomes also have a notable influence on tumor growth. It is known that human gliomas have a population of stem cells with a tumor-supporting ability; these cells are called glioma-associated stem cells (GASCs). Bourkoula et al demonstrated that exosomes derived from GASCs support tumor growth and have a tumor-supporting phenotype (42). In addition, EVs, including exosomes from glioblastoma-derived CSCs, regulate tumor growth through chloride intracellular channel-1 (CLIC1) (43).

Thus, exosomes released from stem cells affect tumor growth (Table II); however, further studies of stem cell-derived exosomes are required.

Table II.

Overview of functions of stem cell-derived exosomes in cancer.

Table II.

Overview of functions of stem cell-derived exosomes in cancer.

Cancer typeDonor cellsExosomal cargoTarget cellsPathway involvedApplication or outcomeReference
Breast cancerhMSCsmiR-21, miR34a, ~150 different proteins, sphingomyelinMCF-7 breast cancer cellsTumors co-injected with EVs exhibited higher angiogenesisSupported breast cancer cell proliferation46
Human gastric carcinomaMSCUnclearSGC-7901 cellsVEGF ERK1/2 pathwayPromoted tumor growth47
Breast cancer and carcinoma of the ovaryMSCMMP-2 proteinSCCOHT-1 cells, MCF-7 breast cancer cellsAltered cellular functionalitiesImproved tumor growth and provided capability to re-organize tumor microenvironment49
Malignant glialtumorsUmbilical cord MSCUnclearU87MG glioblastoma cellsCell-cell communicationDecreased cell proliferation50
Malignant glialtumorsAdipose tissue MSCUnclearU87MG glioblastoma cellsCell communicationImproved tumor growth50
GliomamiR-146-expressing MSCsmiR-146Glioma cellsmiR-146 bound EGFR mRNA and suppressed EGFR expressionReduced glioma xenograft growth52
Multiple myelomaMultiple myeloma BM-MSCLower miR-15a levels and higher content levels of CCL2Multiple myeloma cellsLower miR-15a expression in MM vs. normal BM-MSC-derived exosomesMM BM-MSC-derived exosomes promoted MM tumor growth54
Multiple myelomaNormal BM-MSCSuppressor miRNAs and normal level of CCL2Multiple myeloma cellsTransfer of tumor suppressor miRNAs (including miR-15a)Normal BM-MSC-derived exosomes inhibited growth of MM cells54
Bladder tumorhWJMSCsUnclearT24 cellsDownregulated phosphorylation of Akt protein kinase and upregulated cleaved caspase 3Anti-proliferation and pro-apoptosis55
GliomaGASCUnclearGlioblastoma cell linesUnclearSupported tumor growth42
GlioblastomaGBM-derived CSCsCLIC1GBM cell linesStimulated cell growth through CLIC1Novel regulator of GBM growth43
Renal cancerCD105-positive cancer stem cellsProangiogenic mRNAs and miRNAsHUVECsmRNAs, including VEGF, FGF, angiopoietin1, ephrin A3, MMP-2 and MMP-9, and functional miRNAs, including miR-200c, miR-92 and miR-141, contribute to tumor angiogenesisContributed to triggering angiogenic switch and coordinating metastatic diffusion39
Breast cancer cellsMSCsmiR-164T1 cellsDownregulated VEGF expressionSuppressed angiogenesis59
Gastric cancerMSCmiR-221HGC-27Regulated miR-221 expressionPromoted proliferation and migration65
Breast cancerAdipose MSCGenes associated with cell migration upregulatedBreast cancer cell line MCF-7Wnt signaling pathwayPromoted migration and proliferation of breast cancer cell line MCF-766
Breast cancerDCIS stem-like cellsmiR-140 is downregulated in exosomesBreast cancer cellsDownregulation of miR-140 removed inhibition of Wnt, Sox2 and Sox9 stem cell regulatory pathwaysLead to higher cancer stem cell populations and breast cancer progression67
Breast cancerDCIS stem-like cellsmiR-21, miR-140 and miR-29aMDA-MB-231 cellsmiRNAs signaled through nearby immune cells via interaction with toll-like receptors to upregulate secretion of TNF and IL-6 secretionEnhanced migratory capacity68
GliomaMSCsmiR-124 and miR-145Glioma cells U87 and A172miR-124 and miR-145 mimics decreased luciferase activity of respected reporter target genes, SCP-1 and Sox2Decreased migration of glioma cells and self-renewal of GSCs69
Breast cancerBM-MSCsmiR-23bBM2 cellsIncreased miR-23b and decreased MARCKS expressionPromoted breast cancer cell dormancy in a metastatic niche70
OsteosarcomaBM-MSCsmiR-143Human osteosarcoma cell line 143BDelivered miR-143 to target cellsReduced migration of osteosarcoma cells71

[i] hMSCs, human mesenchymal stem cells; miR/miRNA, microRNA; EV, extracellular vesicles; MSCs, mesenchymal stem/stroma cells; VEGF, vascular endothelial growth factor; ERK, extracellular signal-regulated kinase; MMP, matrix metalloproteinase; EGFR, epidermal growth factor receptor; BM, bone marrow; CCL2, chemokine (C-C motif) ligand 2; MM, multiple myeloma; hWJMSCs, human Wharton's jelly mesenchymal stem cells; GASC, glioma-associated stem cells; GBM, glioblastoma; CSCs, cancer stem cells; CLIC1, chloride intracellular channel-1; CD, cluster of differentiation; HUVECs, human umbilical vein endothelial cells; FGF, fibroblast growth factor; DCIS, ductal carcinoma in situ; Sox, sex-determining region Y-box 2; TNF, tumor necrosis factor; IL-6, interleukin 6; SCP-1, small carboxy-terminal domain phosphatase 1; GSCs, glioma stem cells; MARCKS, myristoylated alanine-rich C-kinase substrate.

Tumor angiogenesis

The formation of new blood vessels is necessary for tumor growth and development, and plays a significant role in tumor progression and metastasis. The process of angiogenesis is extremely complex; it generally includes vascular endothelial substrate degradation, vascular endothelial cell migration and endothelial cell proliferation, as well as the formation of vascular pipeline branches and a new basement membrane. Exosomes contain abundant angiogenic factors that regulate tumor angiogenesis. For example, MSC-derived exosomes promote tumor angiogenesis by increasing VEGF expression in tumor cells and activating ERK1/2 and p38 mitogen-activated protein kinase pathways (47). Other studies have demonstrated that placental MSC exosomes promote vascular network formation and improve microvascular endothelial cell migration in a concentration- and oxygen-dependent manner (57). In addition, EVs released by adipose mesenchymal stem cells (ASCs) may contribute to ASC-induced angiogenesis (58). In cancer stem cells, exosomes derived only from CD105-positive cancer stem cells conferred an activated angiogenic phenotype to normal human endothelial cells, stimulating their growth and vessel formation (39). The results defined a specific source of cancer stem cell-derived MVs that contribute to triggering the angiogenic switch and coordinating metastatic diffusion during tumor progression (39). The effects of exosomes from different types of stem cells on tumor angiogenesis are similar to the effects of stem cell-derived exosomes on tumor growth. However, their effect may be completely different. Lee et al observed that MSC-derived exosomes suppressed angiogenesis by transferring anti-angiogenic molecules and serving as a significant mediator of cell-to-cell communication within the tumor microenvironment (59). These authors noted that MSC-derived exosomes inhibited tumor growth and angiogenesis in breast cancer by downregulating the expression of VEGF, which is a pro-angiogenic factor that is frequently overexpressed in cancer (60). miR-16, which is contained in MSC-derived exosomes, reduces the VEGF expression level in 4T1 cells. It has been demonstrated that miR-16 controls VEGF expression (6163). In summary, exosomes are significantly associated with tumor angiogenesis.

Tumor metastases

Exosomes released from stem cells also contribute to tumor metastasis. A number of the key steps in tumor invasion and metastasis are associated with MSCs, including facilitating epithelial-mesenchymal transition and the induction of stem-like properties that allow cancer stem cells to increase their survivability through the circulation (64). A number of studies have examined the role of CSC- and MSC-derived exosomes in metastasis, tumor reseeding (self-seeding) and the formation of a pre-metastatic niche. Gastric cancer (GC) MSC-derived exosomes were observed to deliver miR-221 to HGC-27 cells, which facilitated the proliferation and migration of these cells (65). In addition, MSC-derived exosomes promoted Wnt signaling pathway activation to facilitate the migration and proliferation of the breast cancer cell line MCF-7 (66). The Wnt signaling pathway is characterized by the nuclear accumulation of β-catenin, which is involved in not only embryonic development but also tumor development (66). As for CSCs, Wolfson et al noted that exosomes derived from ductal carcinoma in situ (DCIS) stem-like cells contained lower levels of miR-140 compared with exosomes derived from a DCIS whole cell population, which could improve tumor growth and metastases (67). Dysregulation of miR-140 has an important role in regulating the transition of DCIS to invasive ductal carcinoma (IDC) (67). As the tumor grade increases, miR-140 is progressively downregulated and plays a significant role in the stem cell regulatory pathways. Downregulation of miR-140 leads to higher CSC populations and breast cancer progression by removing tumor suppressive pathways (67). Other studies have characterized the exosomal exchange of miRNAs between DCIS stem-like cells and target cells; exosomes from DCIS stem-like cells were observed to enhance the migratory capacity via several miRNAs, including miR-140, miR-29a and miR-21, which are differentially expressed in the exosomes (68). These findings suggest that CSC-derived exosomes may improve tumor metastases.

However, there are certain contrary findings. Lee et al demonstrated that MSC-derived exosomes transferred specific miRNA mimics in a gap junction-dependent and contact-independent manner. miR-124 and miR-145 mimics vitally decrease the luciferase activity of their respective reporter target genes, including small carboxy-terminal domain phosphatase 1 (SCP-1) and sex-determining region Y-box 2 (Sox2), and decrease the migration of glioma cells and the self-renewal of glioma stem cells (69). Additionally, Ono et al noted that breast cancer BM-MSC-derived exosomes have more varied miRNAs than adult fibroblast-derived exosomes. BM-MSC-derived exosomes overexpress miR-23b, which induces a dormant phenotype through suppressing a target gene, myristoylated alanine-rich C-kinase substrate, which encodes a protein that promotes cell cycling and motility (70). These findings suggest that exosomal transfer of miRNAs from the BM may promote breast cancer cell dormancy in a metastatic niche. Another study has also presented the same result; these authors observed that the delivery of miR-143 via MSC-derived exosomes significantly reduced the migration of osteosarcoma cells (71).

Taken together, these findings reveal that stem cell-derived exosome-mediated intercellular communication may be an essential mechanism for tumor metastasis.

Tumor therapy

Personalized medicine is used to identify patient- and tumor-specific factors that are useful for the identification of therapeutic options and the prognostic stratification of patients in order to maximize effectiveness and minimize treatment-associated toxicity (72). To achieve this goal, exosomes from stem cells, possibly MSCs, may provide a new approach to personalized medicine. Over the past few years, the potential of using MSCs in regenerative medicine has received increasing attention, and the use of MSCs in anticancer therapy has been extensively studied. A previous study suggested that MCS-derived exosomes may have a key role in not only regenerative medicine to repair damaged tissue but also tumor therapy (73). MSCs may communicate with cancer cells via gap junctional intercellular communication and via the secretion of exosomes (74,75). Chen and Lim revealed that intercellular communication between MSCs and tumor cells could be promoted by secreting microparticles, including exosomes, through the exosomal transfer of miRNAs (76). Additionally, in recent years, miRNAs have emerged as potential anti-cancer agents. Although miRNAs are an effective therapeutic method for cancer, the effectiveness of this approach requires targeted delivery. Exosomes work as delivery vehicles for nucleic acids or drugs; this novel approach has gained increasing interest due to the effective biocompatibility and biodistribution of exosomes. In addition, stem cells have an infinite capacity for reproducible production of exosomes as drug delivery vehicles, which may be essential for tumor therapy. Lim et al observed that exosomes transmit miRNAs from BM stroma to breast cancer cells in tumor cell quiescence. This study revealed that the transfer of miRNAs from BM stroma to breast cancer cells through exosomes may have a significant role in the dormancy of BM metastases through exosomes (77). Munoz et al revealed that MSC-derived exosomes could deliver anti-miR-9, which blocks miR-9, to glioblastoma multiforme (GBM) cells. Anti-miR-9 was involved in the expression of the drug efflux transporter P-glycoprotein, reversed the expression of the multidrug transporter, and sensitized the GBM cells to temozolomide, which increases cell death and caspase activity (78). These data demonstrated a potential role for MSCs in the functional delivery of synthetic anti-miR-9 to reverse the chemoresistance of GBM cells. Boelens et al revealed that stromal cell-derived exosomes stimulated the pattern recognition receptor retinoic acid-inducible gene I to activate signal transducer and activator of transcription 1-dependent antiviral signaling and activate NOTCH3 in breast cancer cells to expand breast cancer cell subpopulations that are adept at resisting therapy and reinitiating tumor growth. These findings suggest a possible novel use of MSCs in the development of new ‘biotech drugs’ with increased efficacy and homing capacity (79). In addition, miR-122-modified adipose tissue-derived MSCs secrete exosomes to increase chemosensitivity (80). MSC-derived exosomes effectively silence the polo-like kinase 1 (PLK-1) gene by transporting PLK-1 small interfering RNA to bladder cancer cells (81). Pascucci et al were the first to demonstrate that MSCs act as a factory to develop drugs with a higher cell-target specificity through packaging and delivering active drugs, and suggested the possibility of using MSCs (82). These authors noted that paclitaxel (PTX) is incorporated by MSCs and released in exosomes. PTX is delivered to target cells and inhibits tumor growth through a simple procedure of exposing the cells to an extremely high concentration of PTX. Fuhrmann et al demonstrated that EVs, including exosomes, loaded with hydrophilic porphyrins induced a stronger phototoxic effect than free drugs in a cancer cell model; this approach may significantly improve cellular uptake and the therapeutic effect of phototoxic porphyrins in vitro (83). Significantly, these methods are simple and directly applicable to other drugs and vesicles, thus providing a new approach to cancer therapy.

EVs from other stem cells also have a potential role in tumor therapy. Fonsato et al demonstrated that EVs derived from human adult liver stem cells may inhibit HepG2 hepatoma and primary hepatocellular carcinoma cell growth and survival (Table III) (84). In summary, stem cell-derived exosomes have a significant role in anti-cancer treatment (Fig. 2).

Table III.

Exosomes in tumor therapy.

Table III.

Exosomes in tumor therapy.

Cancer typeDonor cellsExosomal cargoTarget cellsApplication or outcomeReference
GlioblastomaMSCsAnti-miR-9Resistant GBM cellsIncrease tumor death78
Breast cancerStromal cellsNon-coding RNA and proteinsBreast cancer cells (1833)Biotech drugs regulate therapy resistance79
Hepatocellular carcinomamiR-122-modified adipose tissue-derived MSCsmiR-122HepG2Increase chemosensitivity80
Bladder cancerMSCsPLK-1 siRNABladder cancer cell lines Pancreatic cellSelective gene silencing of PLK-181
Pancreatic cancerMSC with PTXPTXline sr4987Deliver drugs82
Breast cancerhMSCsHydrophilic porphyrinsMDA-MB231 breast cancer (MDA) cellsDeliver drugs83
Hepatocellular carcinomaLiver stem cellsAntitumor miRNAs, including miR-451, miR-223 and miR-31HepG2Inhibit growth and survival84

[i] MSCs, mesenchymal stem cells; miR/miRNA, microRNA; siRNA, small interfering RNA; GBM, glioblastoma; PLK-1, polo-like kinase 1; PTX, paclitaxel; hMSCs, human mesenchymal stem cells.

Tumor biomarkers

Stem cell-derived exosomes provide an enriched source of biomarkers as they contain bioactive molecules that reflect the pathological state of the origin cells. Recently, CSCs have been used in cancer diagnostics and treatment. Exosomes released from rotenone-treated prostate and breast CSCs have specific biomolecular characteristics, including the expression of several exosomal markers, including CD9, CD63, CD81, Alix and TSG101. Thus, the release of exosomal markers may be highly relevant for biological activity and may be used as potential targets (44). In addition, GC-MSC-derived exosomes contain miR-221, which is a potential new biomarker for tumor diagnosis (65).

Taken together, these findings suggest that the constituents of exosomes deliver information from cell to cell (delivering proteins and nucleic acids), and may be used as a biomarker for targeted cancer therapy. There are a number of studies on the use of MSCs in regenerative medicine and anti-cancer treatments (8587). Over the past few years, these studies have raised high expectations. However, safety concerns and tight regulations hamper their practical use in clinical settings. The use of stem cell-derived exosomes may have numerous advantages compared with cell-based approaches, and may improve the safety of MSCs in tumor therapy. However, further experiments are required to demonstrate the safety and feasibility of stem cell-derived exosomes.

Conclusion

In conclusion, studies have demonstrated that exosomes derived from various cell types, including stem cells, may act as mediators of cell-to-cell communication. The role of stem cell-derived exosomes in tumor development has been intensively studied due to the influence of exosomes on tumors and the significant therapeutic potential of stem cells. MSC- and CSC-derived exosomes contain different protein and RNA profiles compared with their donor cells. Exosomes transfer their molecular contents, including numerous types of special proteins and RNA, between cells, thus influencing tumor behavior and proliferation. In addition, exosomes carry a number of the desirable attributes of a synthetic liposome vehicle, including the capacity to carry hydrophobic drugs, and are effective and safe drug delivery vehicles. In the present review, several aspects of stem cell-derived exosome biology in cancer, which has the ability to communicate with surrounding and distant cells, are discussed. MSC- and CSC-derived exosomes have a significant role in synergistically influencing cancer development, metastasis, progression and drug resistance. Although a number of studies have examined the role of stem cell-derived exosomes in cancer (4649), the exact mechanisms of the effects of stem cell-derived exosomes on cancer have been largely unexplored and untested. The utility of exosomes as a delivery vehicle is also unclear. Additional studies of stem cell-derived exosomes are required to determine their roles in the pathogenesis of cancer and to provide a new tool for cancer diagnosis and therapeutics.

Acknowledgements

This study was supported by the National Basic Research Program of China (973 Program, 2013CB834100), the National Natural Science Foundation of China (no. 81572393, 81472820, 81172143, 11334004 and 81421091), the Key Project supported by Medical Science and Technology Development Foundation, Nanjing Municipality Health Bureau (no. ZKX15020 and ZKX 12011), the Natural Science Foundation of Jiangsu Province (no. BK20141324), the Jiangsu Province Innovation for Ph.D. Candidates (no. KYLX_0058), the Scientific Research Foundation of the Graduate School of Nanjing University (no. 2013CL14), and the Jiangsu Special Program for Clinical Medical Science and Technology (BL2014054).

Glossary

Abbreviations

Abbreviations:

MVBs

multivesicular bodies

MSCs

mesenchymal stroma/stem cells

CSCs

cancer stem cells

ESCRT

endosomal sorting complex required for transport

GTPase

guanosine triphosphatase

EVs

extracellular vesicles

VEGF

vascular endothelial growth factor

CLIC1

chloride intracellular channel-1

ERK

extracellular signal-regulated kinase

References

1 

Lopatina T, Gai C, Deregibus MC, Kholia S and Camussi G: Cross talk between cancer and mesenchymal stem cells through extracellular vesicles carrying nucleic acids. Front Oncol. 6:1252016. View Article : Google Scholar : PubMed/NCBI

2 

Trams EG, Lauter CJ, Salem N Jr and Heine U: Exfoliation of membrane ecto-enzymes in the form of micro-vesicles. Biochim Biophys Acta. 645:63–70. 1981. View Article : Google Scholar : PubMed/NCBI

3 

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 : PubMed/NCBI

4 

Trajkovic K, Hsu C, Chiantia S, Rajendran L, Wenzel D, Wieland F, Schwille P, Brügger B and Simons M: Ceramide triggers budding of exosome vesicles into multivesicular endosomes. Science. 319:1244–1247. 2008. View Article : Google Scholar : PubMed/NCBI

5 

Brinton LT, Sloane HS, Kester M and Kelly KA: Formation and role of exosomes in cancer. Cell Mol Life Sci. 72:659–671. 2015. View Article : Google Scholar : PubMed/NCBI

6 

Kowal J, Tkach M and Théry C: Biogenesis and secretion of exosomes. Curr Opin Cell Biol. 29:116–125. 2014. View Article : Google Scholar : PubMed/NCBI

7 

Colombo M, Moita C, van Niel G, Kowal J, Vigneron J, Benaroch P, Manel N, Moita LF, Théry C and Raposo G: Analysis of ESCRT functions in exosome biogenesis, composition and secretion highlights the heterogeneity of extracellular vesicles. J Cell Sci. 126:5553–5565. 2013. View Article : Google Scholar : PubMed/NCBI

8 

Mathivanan S, Fahner CJ, Reid GE and Simpson RJ: ExoCarta 2012: Database of exosomal proteins, RNA and lipids. Nucleic Acids Res. 40:(Database Issue). D1241–D1244. 2012. View Article : Google Scholar : PubMed/NCBI

9 

Théry C, Regnault A, Garin J, Wolfers J, Zitvogel L, Ricciardi-Castagnoli P, Raposo G and Amigorena S: Molecular characterization of dendritic cell-derived exosomes. Selective accumulation of the heat shock protein hsc73. J Cell Biol. 147:599–610. 1999. View Article : Google Scholar : PubMed/NCBI

10 

Bang C and Thum T: Exosomes: New players in cell-cell communication. Int J Biochem Cell Biol. 44:2060–2064. 2012. View Article : Google Scholar : PubMed/NCBI

11 

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

12 

Thakur BK, Zhang H, Becker A, Matei I, Huang Y, Costa-Silva B, Zheng Y, Hoshino A, Brazier H, Xiang J, et al: Double-stranded DNA in exosomes: A novel biomarker in cancer detection. Cell Res. 24:766–769. 2014. View Article : Google Scholar : PubMed/NCBI

13 

Ismail N, Wang Y, Dakhlallah D, Moldovan L, Agarwal K, Batte K, Shah P, Wisler J, Eubank TD, Tridandapani S, et al: Macrophage microvesicles induce macrophage differentiation and miR-223 transfer. Blood. 121:984–995. 2013. View Article : Google Scholar : PubMed/NCBI

14 

Tolmachova T, Anders R, Stinchcombe J, Bossi G, Griffiths GM, Huxley C and Seabra MC: A general role for Rab27a in secretory cells. Mol Biol Cell. 15:332–344. 2004. View Article : Google Scholar : PubMed/NCBI

15 

Zylbersztejn K and Galli T: Vesicular traffic in cell navigation. FEBS J. 278:4497–4505. 2011. View Article : Google Scholar : PubMed/NCBI

16 

Zhang X, Yuan X, Shi H, Wu L, Qian H and Xu W: Exosomes in cancer: Small particle, big player. J Hematol Oncol. 8:832015. View Article : Google Scholar : PubMed/NCBI

17 

Thompson CA, Purushothaman A, Ramani VC, Vlodavsky I and Sanderson RD: Heparanase regulates secretion, composition, and function of tumor cell-derived exosomes. J Biol Chem. 288:10093–10099. 2013. View Article : Google Scholar : PubMed/NCBI

18 

Gruenberg J and van der Goot FG: Mechanisms of pathogen entry through the endosomal compartments. Nat Rev Mol Cell Biol. 7:495–504. 2006. View Article : Google Scholar : PubMed/NCBI

19 

Yu X, Harris SL and Levine AJ: The regulation of exosome secretion: A novel function of the p53 protein. Cancer Res. 66:4795–4801. 2006. View Article : Google Scholar : PubMed/NCBI

20 

Vicencio JM, Yellon DM, Sivaraman V, Das D, Boi-Doku C, Arjun S, Zheng Y, Riquelme JA, Kearney J, Sharma V, et al: Plasma exosomes protect the myocardium from ischemia-reperfusion injury. J Am Coll Cardiol. 65:1525–1536. 2015. View Article : Google Scholar : PubMed/NCBI

21 

Meckes DG Jr and Raab-Traub N: Microvesicles and viral infection. J Virol. 85:12844–12854. 2011. View Article : Google Scholar : PubMed/NCBI

22 

Admyre C, Johansson SM, Qazi KR, Filén JJ, Lahesmaa R, Norman M, Neve EP, Scheynius A and Gabrielsson S: Exosomes with immune modulatory features are present in human breast milk. J Immunol. 179:1969–1978. 2007. View Article : Google Scholar : PubMed/NCBI

23 

Robbins PD and Morelli AE: Regulation of immune responses by extracellular vesicles. Nat Rev Immunol. 14:195–208. 2014. View Article : Google Scholar : PubMed/NCBI

24 

De Toro J, Herschlik L, Waldner C and Mongini C: Emerging roles of exosomes in normal and pathological conditions: New insights for diagnosis and therapeutic applications. Front Immunol. 6:2032015. View Article : Google Scholar : PubMed/NCBI

25 

Masyuk AI, Masyuk TV and Larusso NF: Exosomes in the pathogenesis, diagnostics and therapeutics of liver diseases. J Hepatol. 59:621–625. 2013. View Article : Google Scholar : PubMed/NCBI

26 

Miller IV and Grunewald TG: Tumour-derived exosomes: Tiny envelopes for big stories. Biol Cell. 107:287–305. 2015. View Article : Google Scholar : PubMed/NCBI

27 

Caplan AI: Mesenchymal stem cells. J Orthop Res. 9:641–650. 1991. View Article : Google Scholar : PubMed/NCBI

28 

Bieback K and Klüter H: Mesenchymal stromal cells from umbilical cord blood. Curr Stem Cell Res Ther. 2:310–323. 2007. View Article : Google Scholar : PubMed/NCBI

29 

Lu T, Hu P, Su X, Li C, Ma Y and Guan W: Isolation and characterization of mesenchymal stem cells derived from fetal bovine liver. Cell Tissue Bank. 15:439–450. 2014. View Article : Google Scholar : PubMed/NCBI

30 

Natunen S, Lampinen M, Suila H, Ritamo I, Pitkänen V, Nairn AV, Räbinä J, Laitinen S, Moremen KW, Reutter W and Valmu L: Metabolic glycoengineering of mesenchymal stromal cells with N-propanoylmannosamine. Glycobiology. 23:1004–1012. 2013. View Article : Google Scholar : PubMed/NCBI

31 

Park JS, Suryaprakash S, Lao YH and Leong KW: Engineering mesenchymal stem cells for regenerative medicine and drug delivery. Methods. 84:3–16. 2015. View Article : Google Scholar : PubMed/NCBI

32 

Baglio SR, Pegtel DM and Baldini N: Mesenchymal stem cell secreted vesicles provide novel opportunities in (stem) cell-free therapy. Front Physiol. 3:3592012. View Article : Google Scholar : PubMed/NCBI

33 

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 : PubMed/NCBI

34 

Huang WH, Chang MC, Tsai KS, Hung MC, Chen HL and Hung SC: Mesenchymal stem cells promote growth and angiogenesis of tumors in mice. Oncogene. 32:4343–4354. 2013. View Article : Google Scholar : PubMed/NCBI

35 

Karnoub AE, Dash AB, Vo AP, Sullivan A, Brooks MW, Bell GW, Richardson AL, Polyak K, Tubo R and Weinberg RA: Mesenchymal stem cells within tumour stroma promote breast cancer metastasis. Nature. 449:557–563. 2007. View Article : Google Scholar : PubMed/NCBI

36 

Ljujic B, Milovanovic M, Volarevic V, Murray B, Bugarski D, Przyborski S, Arsenijevic N, Lukic ML and Stojkovic M: Human mesenchymal stem cells creating an immunosuppressive environment and promote breast cancer in mice. Sci Rep. 3:22982013. View Article : Google Scholar : PubMed/NCBI

37 

Akyurekli C, Le Y, Richardson RB, Fergusson D, Tay J and Allan DS: A systematic review of preclinical studies on the therapeutic potential of mesenchymal stromal cell-derived microvesicles. Stem Cell Rev. 11:150–160. 2015. View Article : Google Scholar : PubMed/NCBI

38 

Ratajczak J, Miekus K, Kucia M, Zhang J, Reca R, Dvorak P and Ratajczak MZ: Embryonic stem cell-derived microvesicles reprogram hematopoietic progenitors: Evidence for horizontal transfer of mRNA and protein delivery. Leukemia. 20:847–856. 2006. View Article : Google Scholar : PubMed/NCBI

39 

Grange C, Tapparo M, Collino F, Vitillo L, Damasco C, Deregibus MC, Tetta C, Bussolati B and Camussi G: Microvesicles released from human renal cancer stem cells stimulate angiogenesis and formation of lung premetastatic niche. Cancer Res. 71:5346–5356. 2011. View Article : Google Scholar : PubMed/NCBI

40 

Takebe N, Miele L, Harris PJ, Jeong W, Bando H, Kahn M, Yang SX and Ivy SP: Targeting notch, hedgehog, and Wnt pathways in cancer stem cells: Clinical update. Nat Rev Clin Oncol. 12:445–464. 2015. View Article : Google Scholar : PubMed/NCBI

41 

Hannafon BN and Ding WQ: Cancer stem cells and exosome signaling. Stem Cell Investig. 2:112015.PubMed/NCBI

42 

Bourkoula E, Mangoni D, Ius T, Pucer A, Isola M, Musiello D, Marzinotto S, Toffoletto B, Sorrentino M, Palma A, et al: Glioma-associated stem cells: A novel class of tumor-supporting cells able to predict prognosis of human low-grade gliomas. Stem Cells. 32:1239–1253. 2014. View Article : Google Scholar : PubMed/NCBI

43 

Setti M, Osti D, Richichi C, Ortensi B, Del Bene M, Fornasari L, Beznoussenko G, Mironov A, Rappa G, Cuomo A, et al: Extracellular vesicle-mediated transfer of CLIC1 protein is a novel mechanism for the regulation of glioblastoma growth. Oncotarget. 6:31413–31427. 2015.PubMed/NCBI

44 

Kumar D, Gupta D, Shankar S and Srivastava RK: Biomolecular characterization of exosomes released from cancer stem cells: Possible implications for biomarker and treatment of cancer. Oncotarget. 6:3280–3291. 2015. View Article : Google Scholar : PubMed/NCBI

45 

Kahlert C and Kalluri R: Exosomes in tumor microenvironment influence cancer progression and metastasis. J Mol Med (Berl). 91:431–437. 2013. View Article : Google Scholar : PubMed/NCBI

46 

Vallabhaneni KC, Penfornis P, Dhule S, Guillonneau F, Adams KV, Mo YY, Xu R, Liu Y, Watabe K, Vemuri MC and Pochampally R: Extracellular vesicles from bone marrow mesenchymal stem/stromal cells transport tumor regulatory microRNA, proteins, and metabolites. Oncotarget. 6:4953–4967. 2015. View Article : Google Scholar : PubMed/NCBI

47 

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 : PubMed/NCBI

48 

Hernanda PY, Pedroza-Gonzalez A, van der Laan LJ, Bröker ME, Hoogduijn MJ, Ijzermans JN, Bruno MJ, Janssen HL, Peppelenbosch MP and Pan Q: Tumor promotion through the mesenchymal stem cell compartment in human hepatocellular carcinoma. Carcinogenesis. 34:2330–2340. 2013. View Article : Google Scholar : PubMed/NCBI

49 

Yang Y, Bucan V, Baehre H, von der Ohe J, Otte A and Hass R: Acquisition of new tumor cell properties by MSC-derived exosomes. Int J Oncol. 47:244–252. 2015.PubMed/NCBI

50 

Del Fattore A, Luciano R, Saracino R, Battafarano G, Rizzo C, Pascucci L, Alessandri G, Pessina A, Perrotta A, Fierabracci A and Muraca M: Differential effects of extracellular vesicles secreted by mesenchymal stem cells from different sources on glioblastoma cells. Expert Opin Biol Ther. 15:495–504. 2015. View Article : Google Scholar : PubMed/NCBI

51 

Bruno S, Collino F, Deregibus MC, Grange C, Tetta C and Camussi G: Microvesicles derived from human bone marrow mesenchymal stem cells inhibit tumor growth. Stem Cells Dev. 22:758–771. 2013. View Article : Google Scholar : PubMed/NCBI

52 

Katakowski M, Buller B, Zheng X, Lu Y, Rogers T, Osobamiro O, Shu W, Jiang F and Chopp M: Exosomes from marrow stromal cells expressing miR-146b inhibit glioma growth. Cancer Lett. 335:201–204. 2013. View Article : Google Scholar : PubMed/NCBI

53 

Katakowski M, Zheng X, Jiang F, Rogers T, Szalad A and Chopp M: MiR-146b-5p suppresses EGFR expression and reduces in vitro migration and invasion of glioma. Cancer Invest. 28:1024–1030. 2010. View Article : Google Scholar : PubMed/NCBI

54 

Roccaro AM, Sacco A, Maiso P, Azab AK, Tai YT, Reagan M, Azab F, Flores LM, Campigotto F, Weller E, et al: BM mesenchymal stromal cell-derived exosomes facilitate multiple myeloma progression. J Clin Invest. 123:1542–1555. 2013. View Article : Google Scholar : PubMed/NCBI

55 

Wu S, Ju GQ, Du T, Zhu YJ and Liu GH: Microvesicles derived from human umbilical cord Wharton's jelly mesenchymal stem cells attenuate bladder tumor cell growth in vitro and in vivo. PLoS One. 8:e613662013. View Article : Google Scholar : PubMed/NCBI

56 

Biancone L, Bruno S, Deregibus MC, Tetta C and Camussi G: Therapeutic potential of mesenchymal stem cell-derived microvesicles. Nephrol Dial Transplant. 27:3037–3042. 2012. View Article : Google Scholar : PubMed/NCBI

57 

Salomon C, Ryan J, Sobrevia L, Kobayashi M, Ashman K, Mitchell M and Rice GE: Exosomal signaling during hypoxia mediates microvascular endothelial cell migration and vasculogenesis. PLoS One. 8:e684512013. View Article : Google Scholar : PubMed/NCBI

58 

Lopatina T, Bruno S, Tetta C, Kalinina N, Porta M and Camussi G: Platelet-derived growth factor regulates the secretion of extracellular vesicles by adipose mesenchymal stem cells and enhances their angiogenic potential. Cell Commun Signal. 12:262014. View Article : Google Scholar : PubMed/NCBI

59 

Lee JK, Park SR, Jung BK, Jeon YK, Lee YS, Kim MK, Kim YG, Jang JY and Kim CW: Exosomes derived from mesenchymal stem cells suppress angiogenesis by down-regulating VEGF expression in breast cancer cells. PLoS One. 8:e842562013. View Article : Google Scholar : PubMed/NCBI

60 

Borgström P, Hillan KJ, Sriramarao P and Ferrara N: Complete inhibition of angiogenesis and growth of microtumors by anti-vascular endothelial growth factor neutralizing antibody: Novel concepts of angiostatic therapy from intravital videomicroscopy. Cancer Res. 56:4032–4039. 1996.PubMed/NCBI

61 

Chamorro-Jorganes A, Araldi E, Penalva LO, Sandhu D, Fernández-Hernando C and Suárez Y: MicroRNA-16 and microRNA-424 regulate cell-autonomous angiogenic functions in endothelial cells via targeting vascular endothelial growth factor receptor-2 and fibroblast growth factor receptor-1. Arterioscler Thromb Vasc Biol. 31:2595–2606. 2011. View Article : Google Scholar : PubMed/NCBI

62 

Dejean E, Renalier MH, Foisseau M, Agirre X, Joseph N, de Paiva GR, Al Saati T, Soulier J, Desjobert C, Lamant L, et al: Hypoxia-microRNA-16 downregulation induces VEGF expression in anaplastic lymphoma kinase (ALK)-positive anaplastic large-cell lymphomas. Leukemia. 25:1882–1890. 2011. View Article : Google Scholar : PubMed/NCBI

63 

Hua Z, Lv Q, Ye W, Wong CK, Cai G, Gu D, Ji Y, Zhao C, Wang J, Yang BB and Zhang Y: MiRNA-directed regulation of VEGF and other angiogenic factors under hypoxia. PLoS One. 1:e1162006. View Article : Google Scholar : PubMed/NCBI

64 

Chang AI, Schwertschkow AH, Nolta JA and Wu J: Involvement of mesenchymal stem cells in cancer progression and metastases. Curr Cancer Drug Targets. 15:88–98. 2015. View Article : Google Scholar : PubMed/NCBI

65 

Wang M, Zhao C, Shi H, Zhang B, Zhang L, Zhang X, Wang S, Wu X, Yang T, Huang F, et al: Deregulated microRNAs in gastric cancer tissue-derived mesenchymal stem cells: Novel biomarkers and a mechanism for gastric cancer. Br J Cancer. 110:1199–1210. 2014. View Article : Google Scholar : PubMed/NCBI

66 

Lin R, Wang S and Zhao RC: Exosomes from human adipose-derived mesenchymal stem cells promote migration through Wnt signaling pathway in a breast cancer cell model. Mol Cell Biochem. 383:13–20. 2013. View Article : Google Scholar : PubMed/NCBI

67 

Wolfson B, Eades G and Zhou Q: Roles of microRNA-140 in stem cell-associated early stage breast cancer. World J Stem Cells. 6:591–597. 2014. View Article : Google Scholar : PubMed/NCBI

68 

Li Q, Eades G, Yao Y, Zhang Y and Zhou Q: Characterization of a stem-like subpopulation in basal-like ductal carcinoma in situ (DCIS) lesions. J Biol Chem. 289:1303–1312. 2014. View Article : Google Scholar : PubMed/NCBI

69 

Lee HK, Finniss S, Cazacu S, Bucris E, Ziv-Av A, Xiang C, Bobbitt K, Rempel SA, Hasselbach L, Mikkelsen T, et al: Mesenchymal stem cells deliver synthetic microRNA mimics to glioma cells and glioma stem cells and inhibit their cell migration and self-renewal. Oncotarget. 4:346–361. 2013. View Article : Google Scholar : PubMed/NCBI

70 

Ono M, Kosaka N, Tominaga N, Yoshioka Y, Takeshita F, Takahashi RU, Yoshida M, Tsuda H, Tamura K and Ochiya T: Exosomes from bone marrow mesenchymal stem cells contain a microRNA that promotes dormancy in metastatic breast cancer cells. Sci Signal. 7:ra632014. View Article : Google Scholar : PubMed/NCBI

71 

Shimbo K, Miyaki S, Ishitobi H, Kato Y, Kubo T, Shimose S and Ochi M: Exosome-formed synthetic microRNA-143 is transferred to osteosarcoma cells and inhibits their migration. Biochem Biophys Res Commun. 445:381–387. 2014. View Article : Google Scholar : PubMed/NCBI

72 

Hamburg MA and Collins FS: The path to personalized medicine. N Engl J Med. 363:301–304. 2010. View Article : Google Scholar : PubMed/NCBI

73 

Dai S, Wei D, Wu Z, Zhou X, Wei X, Huang H and Li G: Phase I clinical trial of autologous ascites-derived exosomes combined with GM-CSF for colorectal cancer. Mol Ther. 16:782–790. 2008. View Article : Google Scholar : PubMed/NCBI

74 

Greco SJ and Rameshwar P: Mesenchymal stem cells in drug/gene delivery: Implications for cell therapy. Ther Deliv. 3:997–1004. 2012. View Article : Google Scholar : PubMed/NCBI

75 

Matuskova M, Hlubinova K, Pastorakova A, Hunakova L, Altanerova V, Altaner C and Kucerova L: HSV-tk expressing mesenchymal stem cells exert bystander effect on human glioblastoma cells. Cancer Lett. 290:58–67. 2010. View Article : Google Scholar : PubMed/NCBI

76 

Chen TS and Lim SK: Measurement of precursor miRNA in exosomes from human ESC-derived mesenchymal stem cells. Methods Mol Biol. 1024:69–86. 2013. View Article : Google Scholar : PubMed/NCBI

77 

Lim PK, Bliss SA, Patel SA, Taborga M, Dave MA, Gregory LA, Greco SJ, Bryan M, Patel PS and Rameshwar P: Gap junction-mediated import of microRNA from bone marrow stromal cells can elicit cell cycle quiescence in breast cancer cells. Cancer Res. 71:1550–1560. 2011. View Article : Google Scholar : PubMed/NCBI

78 

Munoz JL, Bliss SA, Greco SJ, Ramkissoon SH, Ligon KL and Rameshwar P: Delivery of functional anti-miR-9 by mesenchymal stem cell-derived exosomes to glioblastoma multiforme cells conferred chemosensitivity. Mol Ther Nucleic Acids. 2:e1262013. View Article : Google Scholar : PubMed/NCBI

79 

Boelens MC, Wu TJ, Nabet BY, Xu B, Qiu Y, Yoon T, Azzam DJ, Twyman-Saint Victor C, Wiemann BZ, Ishwaran H, et al: Exosome transfer from stromal to breast cancer cells regulates therapy resistance pathways. Cell. 159:499–513. 2014. View Article : Google Scholar : PubMed/NCBI

80 

Lou G, Song X, Yang F, Wu S, Wang J, Chen Z and Liu Y: Exosomes derived from miR-122-modified adipose tissue-derived MSCs increase chemosensitivity of hepatocellular carcinoma. J Hematol Oncol. 8:1222015. View Article : Google Scholar : PubMed/NCBI

81 

Greco KA, Franzen CA, Foreman KE, Flanigan RC, Kuo PC and Gupta GN: PLK-1 silencing in bladder cancer by siRNA delivered with exosomes. Urology. 91:241.e1–e7. 2016. View Article : Google Scholar

82 

Pascucci L, Coccè V, Bonomi A, Ami D, Ceccarelli P, Ciusani E, Viganò L, Locatelli A, Sisto F, Doglia SM, et al: Paclitaxel is incorporated by mesenchymal stromal cells and released in exosomes that inhibit in vitro tumor growth: A new approach for drug delivery. J Control Release. 192:262–270. 2014. View Article : Google Scholar : PubMed/NCBI

83 

Fuhrmann G, Serio A, Mazo M, Nair R and Stevens MM: Active loading into extracellular vesicles significantly improves the cellular uptake and photodynamic effect of porphyrins. J Control Release. 205:35–44. 2015. View Article : Google Scholar : PubMed/NCBI

84 

Fonsato V, Collino F, Herrera MB, Cavallari C, Deregibus MC, Cisterna B, Bruno S, Romagnoli R, Salizzoni M, Tetta C and Camussi G: Human liver stem cell-derived microvesicles inhibit hepatoma growth in SCID mice by delivering antitumor microRNAs. Stem Cells. 30:1985–1998. 2012. View Article : Google Scholar : PubMed/NCBI

85 

Gjorgieva D, Zaidman N and Bosnakovski D: Mesenchymal stem cells for anti-cancer drug delivery. Recent Pat Anticancer Drug Discov. 8:310–318. 2013. View Article : Google Scholar : PubMed/NCBI

86 

Toh WS, Lai RC, Hui JH and Lim SK: MSC exosome as a cell-free MSC therapy for cartilage regeneration: Implications for osteoarthritis treatment. Semin Cell Dev Biol. Nov 18–2016.(Epub ahead of print). View Article : Google Scholar : PubMed/NCBI

87 

Sherman LS, Shaker M, Mariotti V and Rameshwar P: Mesenchymal stromal/stem cells in drug therapy: New perspective. Cytotherapy. 19:19–27. 2017. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2017
Volume 13 Issue 5

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
Wu J, Qu Z, Fei ZW, Wu JH and Jiang CP: Role of stem cell-derived exosomes in cancer (Review). Oncol Lett 13: 2855-2866, 2017
APA
Wu, J., Qu, Z., Fei, Z., Wu, J., & Jiang, C. (2017). Role of stem cell-derived exosomes in cancer (Review). Oncology Letters, 13, 2855-2866. https://doi.org/10.3892/ol.2017.5824
MLA
Wu, J., Qu, Z., Fei, Z., Wu, J., Jiang, C."Role of stem cell-derived exosomes in cancer (Review)". Oncology Letters 13.5 (2017): 2855-2866.
Chicago
Wu, J., Qu, Z., Fei, Z., Wu, J., Jiang, C."Role of stem cell-derived exosomes in cancer (Review)". Oncology Letters 13, no. 5 (2017): 2855-2866. https://doi.org/10.3892/ol.2017.5824