Skip to main content

Exosomes in cancer: small particle, big player

Abstract

Exosomes have emerged as a novel mode of intercellular communication. Exosomes can shuttle bioactive molecules including proteins, DNA, mRNA, as well as non-coding RNAs from one cell to another, leading to the exchange of genetic information and reprogramming of the recipient cells. Increasing evidence suggests that tumor cells release excessive amount of exosomes, which may influence tumor initiation, growth, progression, metastasis, and drug resistance. In addition, exosomes transfer message from tumor cells to immune cells and stromal cells, contributing to the escape from immune surveillance and the formation of tumor niche. In this review, we highlight the recent advances in the biology of exosomes as cancer communicasomes. We review the multifaceted roles of exosomes, the small secreted particles, in communicating with other cells within tumor microenvironment. Given that exosomes are cell type specific, stable, and accessible from body fluids, exosomes may provide promising biomarkers for cancer diagnosis and represent new targets for cancer therapy.

Introduction

Exosomes are small, lipid bilayer membrane vesicles of endocytic origin. Exosomes can be defined by several common characteristics, including size (50–100 nm in diameter), density (1.13–1.19 g/ml), morphology (“cup” or “dish” shaped in transmission electron microscopy), and certain enriched protein markers (tetraspanins, TSG101, Hsp70). Initially discovered as the garbage bags for removal of unwanted material from cells, the role of exosomes in immune response is gradually recognized as they function in antigen presentation. More recently, the researchers reveal that exosomes contain proteins and nucleic acids that are functional when transferred into recipient cells. Exosomes have been shown to act as shuttles between cells by transmitting signals (referred to as communicasomes). In this review, we highlight the recent advances in the roles of exosomes in cancer with an emphasis on the potential of exosomes as diagnosis biomarker and therapy target.

Biogenesis, release, and uptake of exosomes

Exosome formation is a fine-tuned process which includes four stages: initiation, endocytosis, multivesicular bodies (MVBs) formation, and exosome secretion [1]. Multivesicular bodies (MVBs) are endocytic structures formed by the budding of an endosomal membrane into the lumen of the compartment. After vesicular accumulation, the MVBs are either sorted for cargo degradation in the lysosome or released into the extracellular space as exosomes by fusing with the plasma membrane (Fig. 1). The mechanisms underlying the sorting of cargo into the intraluminal vesicles (ILVs) are not yet fully elucidated. Both endosomal sorting complex required for transport (ESCRT)-dependent and independent signals have been suggested to determine the sorting of exosomes [2]. The formation of exosomes has been shown to be controlled by the syndecan heparan sulfate proteoglycans and their cytoplasmic adaptor syntenin [3].

Fig. 1
figure 1

Biogenesis, release, structure, and uptake of exosomes. Exosomes are produced from the multivesicular bodies (MVBs) (also known as late endosomes). The membrane of the MVBs bulges inward to form exosomes. During this process, proteins (e.g., receptor, cytoplasmic proteins, tetraspanin), nucleic acids (e.g., DNA, mRNA, miRNA), and lipids (e.g., cholesterol, ceramide) are packed into exosomes in a cell type-dependent manner. MVBs fuse with the cellular membrane to release exosomes into the extracellular space. Several mechanisms have been suggested to mediate the uptake of exosomes, including a exosome fusion with the cellular membrane of the recipient cell, leading to the release of the exosomal cargo into the cytoplasm, b juxtracrine signaling through receptor-ligand interactions, c and endocytosis by phagocytosis

The Rab guanosine triphosphatases (GTPases) have been found to critically regulate exosome secretion. Ostrowski et al. have identified that Rab27a/b affects the size and localization of MVBs [4]. Hsu et al. suggest that Rab3 regulates MVBs docking to tethering at the plasma membrane [5]. The accumulation of intracellular Ca2+ results in increased exosome secretion [6]. In addition, intracellular and intercellular pH has been shown to affect exosome release. When the microenvironmental pH is low, exosome secretion and uptake by recipient cells increases [7]. There is evidence that oncogenes and tumor suppressors regulate exosome secretion in cancer [8]. Yu et al. demonstrate that p53-regulated protein tumor suppressor-activated pathway 6 (TSAP6) induces exosome secretion under stressed conditions [9, 10]. Heparanase is an enzyme with elevated level in cancer. Overexpression of heparanase promotes exosome secretion [11]. Intriguingly, exosomes from normal mammary epithelial cells inhibit exosome secretion by breast cancer cells, implicating a feedback control to maintain dynamic equilibrium [12].

Exosomes transfer information to the target cells through three main ways: (1) receptor-ligand interaction; (2) direct fusion with plasma membrane; (3) endocytosis by phagocytosis (Fig. 1). Although the specific receptors that mediate the uptake of exosomes have not been found, there are several proteins that may act as potential receptors for exosome uptake, such as Tim1/4 for B cells [13] and ICAM-1 for APCs [14]. The uptake of exosomes by direct plasma membrane fusion mode has not been well studied. Melanoma cells could take up exosomes by fusion and low pH facilitates this process [15]. Phagocytosis is an efficient way of exosome uptake. Phagocytic cells have a greater uptake of exosomes than non-phagocytic cells [16]. The uptake of exosomes by recipient cells is energy dependent [17]. Heparan sulfate proteoglycans (HSPGs) function as internalizing receptors of cancer cell-derived exosomes. Enzymatic depletion of cell-surface HSPG or pharmacological inhibition of endogenous proteoglycan biosynthesis significantly attenuates exosome uptake [18].

Structure and contents of exosomes

Exosomes consist of a lipid bilayer membrane surrounding a small cytosol (Fig. 1). The structured lipids not only mold the exosomes but are also involved in exosome function. In addition to lipids, nucleic acids and proteins have also been detected in exosomes. Thakur et al. demonstrate that double-stranded DNA is present in exosomes from cancer cells and reflects the mutational status of the originated cells [19]. Valadi et al. demonstrate that exosomes contain mRNA and miRNA [20]. Exosome-carried RNA can shuttle between cells and thus is called “exosomal shuttle RNA” (esRNA). The protein composition of tumor cell-derived exosomes has been well characterized for a number of cancers by using different proteomic methods. The most common proteins, mRNA, and miRNAs found in exosomes have been deposited in ExoCarta (www.exocarta.org). To date, 4563 proteins, 1639 mRNAs, and 764 miRNAs have been identified in exosomes from different species and tissues by independent examinations. The exosomal contents vary between different physiological and pathological conditions and original cell types. Moreover, the composition of exosomes can be distinct from the originated cells due to the selective sorting of the cargo into exosomes.

Isolation, detection, and analysis of exosomes

Exosomes have been isolated and characterized from distinct cells under normal and stressed conditions. At present, the most commonly used methods for exosome isolation include ultracentrifugation, combined with sucrose gradient, and the immune-bead isolation (e.g., magnetic activated cell sorting; MACS). There are many commercial kits available for the extraction of exosomes. Transmission electron microscopy (TEM), Western blot, and FACS are frequently used to characterize the isolated exosomes based on their biochemical properties (e.g., morphology, size, exosomal markers). There is a lack of the accurate method to determine the concentration of exosomes. The researchers have to rely on inaccurate measurements of protein concentration or nanoparticle tracking analysis. Quantitative RT-PCR, nucleic acid sequencing, Western blot, or ELISA are used for exosome RNA and protein identification. The International Society for Extracellular Vesicles (ISEV) has recently released minimal experimental requirements for definition of extracellular vesicles and their functions [21].

Roles of exosomes in cancer

Accumulating evidence indicates that exosomes play important roles in cancer. Exosomes transfer oncogenic proteins and nucleic acids to modulate the activity of recipient cells and play decisive roles in tumorigenesis, growth, progression, metastasis, and drug resistance (Fig. 2). Exosomes can act on various recipient cells. The uptake of exosomes may induce a persistent and efficient modulation of recipient cells. In this section, we will discuss about the roles of exosomes in cancer and the molecular mechanisms (Table 1).

Fig. 2
figure 2

Roles of exosomes in cancer. Exosomes are critically involved in tumor initiation, growth, progression, metastasis, and drug resistance by transferring oncogenic proteins and nucleic acids. Tumor-derived exosomes can activate endothelial cells to support tumor angiogenesis and thrombosis. Tumor-derived exosomes can convert fibroblasts and MSCs into myofibroblasts to facilitate tumor angiogenesis and metastasis. Tumor-derived exosomes contribute to create an immunosuppressive microenvironment by inducing apoptosis and impairing the function of effector T cells and NK cells, inhibiting DC differentiation, expanding MDSCs, as well as promoting Treg cell activity. Tumor-derived exosomes can mobilize neutrophils and skew M2 polarization of macrophages to promote tumor progression. Moreover, tumor-derived exosomes can help tumor cells develop drug resistance by transferring multidrug-resistant proteins and miRNAs, exporting tumoricidal drugs, and neutralizing antibody-based drugs. In turn, exosomes from activated T cells, macrophages, and stromal cells can promote tumor metastasis and drug resistance

Table 1 Overview on the function of exosomes in cancer

Tumorigenesis

Normal cells are transformed into cancer cells in the process of tumorigenesis. Exosomes from malignant cells have shown the potential to induce normal cell transformation. For instance, prostate cancer cell-derived exosomes could induce neoplastic transformation of adipose-derived stem cells (ASCs) [22], which is associated with trafficking of oncogenic proteins (Ras superfamily of GTPases), mRNA (K-ras and H-ras), as well as miRNAs (miR-125b, miR-130b, and miR-155) by exosomes. In addition, Melo et al. suggest that breast cancer cell-derived exosomes contain precursor microRNAs (pre-miRNAs) associated with RNA-induced silencing complex (RISC)-loading complex proteins, which could induce a rapid and efficient silencing of mRNAs in nontumorigenic epithelial cells, resulting in transcriptome reprogramming and oncogenic transformation [23]. They further demonstrate that the exosomes from serum specimen from breast cancer patients but not those from healthy donors induce tumor formation in mice when co-injected with the nontumorigenic epithelial cells, suggesting a potential mechanism for exosome in tumorigenesis. Cancer is composed of heterogeneous cell populations. Side population (SP) cells are a sub-population of cells that exhibit stem cell-like characteristics and can be isolated in cancer by adapting the Hoechst33342 staining method. Koch et al. demonstrate that in diffuse large B-cell lymphoma, side population cells could export Wnt3a via exosomes to neighboring cells, thus modulating SP-non-SP transitions and maintaining population equilibrium [24]. Altogether, these findings indicate that exosomes may contribute to tumor development and uncontrolled tumor progression by acting as a mediator in the transformation of normal cells to malignant cells and a modulator for the balance between cancer stem cells (CSCs) and non-CSCs.

Tumor growth

The promoting effects of exosomes from distinct sources on tumor cell proliferation have been widely reported. Cancer cells uptake exosomes that contain survivin, an anti-apoptotic protein, to protect them from genotoxic stress-induced cell death [25]. Exosomes from serum of glioblastoma patients contain EGFRvIII mRNA, which stimulate the proliferation of human glioma cells through a self-promoting way [26]. Colon cancer cell-derived exosomes are enriched in ΔNp73 mRNA. The proliferation potential of target cells is greatly enhanced by incubation with ΔNp73-containing exosomes [27]. The interaction between tumor stromal cells and tumor cells also efficiently promote tumor growth. Exosomes from chronic myelogenous leukemia (CML) cells stimulate bone marrow stromal cells to produce IL-8, which in turn promote the growth of leukemia cells [28]. Bone marrow mesenchymal stromal cells (BM-MSCs) from multiple myeloma (MM) patients release exosomes that express increased levels of oncogenic proteins, cytokines, and adhesion molecules to facilitate the growth of MM cells [29]. Thus, exosomes from tumor cells and microenvironment could act coordinately to promote tumor growth.

Tumor angiogenesis

The formation of new blood vessels is required for tumor growth and progression. Proteomic analysis has revealed that abundant angiogenic factors are present in malignant mesothelioma-derived exosomes [30]. Exosome uptake induces upregulation of angiogenesis-related genes and results in enhanced endothelial cell proliferation, migration, and sprouting [31]. Exosomes derived from hypoxic glioblastoma cells are more potent to induce angiogenesis [32]. Exosomes from metastatic breast cancer cells contain miR-105. Exosome-mediated transfer of miR-105 degrades ZO-1 protein, disturbs tight junctions, and induces vascular permeability in distant organs [33]. Exosomal miR-92a from K562 leukemia cells targets integrin α5 to enhance endothelial cell migration and tube formation [34]. MiR-210 is significantly enriched in exosomes from hypoxic K562 cells, which promotes the angiogenic activity of endothelial cells [35]. Multiple myeloma cells grown under hypoxic condition produce more exosomes containing miR-135b, which directly suppresses FIH-1, an inhibitor of HIF-1, to enhance endothelial tube formation in endothelial cells [36]. Exosomes are critically involved in tumor angiogenesis by directly delivering angiogenic proteins into endothelial cells or modulating the angiogenic function of endothelial cells by exosomal miRNAs.

Tumor metastasis

Exosomes contribute to tumor metastasis by enhancing tumor cell migration and invasion, establishing pre-metastatic niche, and remodeling the extracellular matrix. EBV-positive nasopharyngeal carcinoma (NPC) cell-derived exosomes contain HIF-1α, which increases migration and invasiveness of EBV-negative NPC cells [37]. Metastatic cancer cells secrete increased level of miRNA with tumor-suppressor function, which may suggest another mechanism for the role of exosomes in metastasis [38]. The formation of pre-metastatic niche is a prerequisite for tumor metastasis. Exosomes from highly metastatic melanoma enhance the metastatic ability of primary tumors by converting bone marrow progenitor cells to a pro-vasculogenic and pre-metastatic phenotype via the MET receptor [39]. Gastrointestinal stromal tumor cells release exosomes containing protein tyrosine kinase to convert progenitor smooth muscle cells to a pre-metastatic phenotype [40]. Suetsugu et al. show that highly metastatic breast cancer cells can transfer their own exosomes to other cancer cells and normal lung tissue cells in vitro and in vivo by using fluorescent protein imaging method [41], which provides direct evidence for the involvement of exosomes from highly metastatic cancer cells in educating stromal cells. Luga and colleagues have shown that exosomes produced by stromal cells are taken up by breast cancer cells and are then loaded with Wnt11, which is associated with stimulation of the invasiveness and metastasis of the breast cancer cells [42]. Exosomes from activated CD8+ T cells promote cancer cell invasion and lung metastasis via the Fas/FasL pathway [43], which adds another layer of mechanism for the role of tumor-infiltrating lymphocytes in cancer metastasis. Exosome-mediated transfer of oncogenic microRNAs into cancer cells is associated with enhanced metastatic potential. IL-4-activated macrophage-derived exosomes transfer miR-223 to co-cultivated breast cancer cells, leading to increase of cell invasion [44]. Exosome-mediated delivery of miR-221/222 from MSCs to gastric cancer cells greatly enhances gastric cancer cell migration [45]. Fabbri et al. suggest that miRNAs in tumor-secreted exosomes can directly bind toll-like receptor (TLR) in immune cells to promote tumor metastasis [46]. Recently, Costa-Silva and colleagues demonstrate that MIF-containing exosomes from pancreatic ductal adenocarcinoma (PDAC) cells induce TGF-β production in liver Kupffer cells, which in turn upregulates fibronectin (FN) expression by hepatic stellate cells and enhances recruitment of bone marrow-derived cells, finally leading to the formation of liver pre-metastatic niche [47], suggesting a complicated network that involves cancer cells, stromal cells, and immune cells in exosome-initiated pre-metastatic niche formation. Intriguingly, Zomer et al. use the Cre-LoxP system to visualize extracellular vesicle (EV) exchange between tumor cells in living mice [48]. They show that the less malignant tumor cells that take up EVs released by malignant tumor cells display enhanced migratory behavior and metastatic capacity, indicating that the metastatic behavior can be phenocopied through extracellular vesicle exchange. Taken together, these findings reveal that the intercellular communication mediated by exosomes may be an important mechanism for tumor metastasis.

Tumor drug resistance

Exosomes contribute to the development of therapy resistance in tumor cells through a variety of mechanisms. Tumor-derived exosomes can transfer multi-drug resistance (MDR)-associated proteins and miRNAs to target cells [49, 50]. In addition, exosomes participate in the process of tumor resistance by mediating drug efflux. The drugs and their metabolites can be encapsulated and exported by exosomes [51, 52]. Melanosomal sequestration of cytotoxic drugs contributes to the intractability of malignant melanomas [53]. Moreover, exosomes may counteract the effect of antibody drugs by modulating their binding to tumor cells. Lymphoma exosomes carry CD20, which bind therapeutic anti-CD20 antibodies and protect target cells from antibody attack [54]. Exosomes from HER2-overexpressing breast cancer cells express active HER2 and can bind to the HER2 antibody trastuzumab to inhibit its activity [55]. Exosomes secreted by stromal cells also contribute to tumor drug resistance. BM-MSC-derived exosomes induce multiple myeloma cells resistant to bortezomib through the activation of several survival relevant pathways [56]. Therefore, exosomes released by cancer cells and stromal cells may have a potential to modulate sensitivity of cancer cells to distinct therapies.

Tumor immune escape

Initially reported as tumor-associated antigens and tumor immune response stimulators, the recent studies have shown that tumor-derived exosomes might rather perform immunosuppressive functions. Tumor exosomes block the differentiation of murine myeloid precursor cells into dendritic cells (DC) [57]. Tumor exosome-carried TGF-β1 skews IL-2 responsiveness in favor of regulatory T cells and away from cytotoxic cells [58]. Human nasopharyngeal carcinoma-derived exosomes recruit, expand, and regulate the function of regulatory T cells through CCL20 [59]. NPC cell-derived exosomes impair T cell function, which is associated with upregulated miRNAs in the exosomes [60]. Tumor cell-derived exosomes switch the differentiation of myeloid cells to myeloid-derived suppressor cells (MDSCs) and induce accelerated lung metastasis in a MyD88-dependent manner [61, 62]. Hsp72 on tumor-derived exosomes promotes the immunosuppressive activity of MDSCs via autocrine activation of IL-6/STAT3 pathway [63]. Breast cancer cell-derived exosomes simulate the activation of NF-κB and enhance the secretion of pro-inflammatory cytokines in macrophages [64]. Exosomes from human prostate cancer cells express ligands for NKG2D on their surface and downregulate NKG2D expression on natural killer (NK) and CD8+ T cells, leading to the impairment of their cytotoxic function [65]. Collectively, these data suggest that tumor-derived exosomes interfere on multiple levels with the immune system to drive tumor immune evasion.

Tumor-stroma interaction

Tumor stroma is believed to be critically involved in tumor development and progression. Webber et al. suggest that prostate cancer cells could trigger differentiation of fibroblasts into myofibroblasts through exosomal TGF-β [66]. In addition, prostate cancer exosomes triggered TGFβ1-dependent fibroblast differentiation resemble stromal cells isolated from cancerous prostate tissue [67], which accelerates tumor growth by supporting angiogenesis. MSCs function as precursors for tumor myofibroblast. The research from our lab suggests that tumor cell-derived exosomes could induce differentiation of human MSCs to carcinoma-associated fibroblasts (CAFs) [68]. Adipose tissue-derived MSCs treated with breast cancer-derived exosomes also display the characteristics of myofibroblasts [69]. Moreover, stromal communication with cancer cells modulates therapy response. Boelens et al. suggest that exosomes transferred from stromal cells to breast cancer cells constitute a juxtacrine NOTCH3 pathway to expand therapy-resistant tumor-initiating cells [70]. Luga et al. demonstrate that fibroblast-secreted exosomes mobilize autocrine Wnt-planar cell polarity (PCP) signaling to drive breast cancer cell invasion and metastasis [42]. Therefore, exosomes may mediate a reciprocal interplay between tumor cells and stromal cells to synergistically promote tumor progression.

Tumor thrombosis

Tissue factor (TF) overexpression is closely associated with tumor progression. TF can get incorporated into tumor-derived exosomes. The hypercoagulable state in cancer patients may be partially influenced by the release of TF-bearing exosomes from tumor cells. Garnier et al. demonstrate that exosomes link the procoagulant status with metastatic phenotype in cancer. Induction of EMT changes in epithelial cancer cells results in the release of exosomes containing elevated level of tissue factor. Importantly, TF-rich exosomes can be transferred to endothelial cells and cause their exaggerated procoagulant conversion [71], suggesting that EMT influences tumor-vascular interaction through altered TF-containing exosomes. However, the exact roles of exosomes in tumor thrombosis and consequent impact on tumor growth, progression, and metastasis remain to be further explored.

Exosomes as cancer biomarkers and targets

The findings that exosomes play critical roles in almost all aspects of cancer provide opportunities for the development of exosomes as ideal diagnostic biomarkers and therapeutic targets. Exosome-shuttled proteins and nucleic acids have been suggested as novel diagnostic and prognostic indicators for a variety of cancers. Moreover, utilizing tumor-derived exosomes as vaccines and exosomes from distinct sources as carriers for drugs and small molecules have been proved to be effective in pre-clinical studies and clinical trials.

Exosomes as cancer diagnostic biomarkers

Exosomes are readily accessible in nearly all body fluids including blood, urine, saliva, and ascites. Exosomes contain bioactive molecules that reflect the pathological state of the originated cells, thus providing an enriched source of biomarkers (Table 2). The level of exosomes is elevated in the plasma of some cancer patients as compared to healthy controls. There is a positive correlation between the abundance of tumor exosomes and tumor stage in ovarian cancer patients [72]. Tumor is characterized by a specific miRNA profile. The majority of circulating microRNAs is concentrated in exosomes [73]. Exosomal miRNAs have been suggested as diagnostic and prognostic indicators for lung cancer, esophageal squamous cell carcinoma, prostate cancer, breast cancer, glioblastoma, ovarian cancer, and other cancer types [7480]. Exosomal miRNAs are positively correlated with the stage and degree of cancer progression. In addition to miRNAs, long non-coding RNAs (LncRNAs) are also detected in exosomes [81, 82]. LncRNA from serum of gastric cancer patients is defined as a novel exosomal biomarker [83, 84].

Table 2 Exosomes from distinct biofluids of cancer patients as biomarkers

Exosomes as cancer therapy targets

Exosome-based immunotherapy

Dendritic cell-derived exosomes (dexosomes) have been developed as immunotherapeutic anticancer agents [85]. Tumor peptide-pulsed DC-derived exosomes suppress growth of established murine tumors in a T cell-dependent manner [86]. Exosomes secreted by living tumor cells contain and transfer tumor antigens to dendritic cells and induce potent CD8+ T cell-dependent antitumor effects on mouse tumors [87]. Dexosomes have entered clinical trials for colorectal cancer, metastatic melanoma, and non-small cell lung cancer and have achieved modest therapeutic effects [88].

Exosome removal for cancer therapy

The removal of exosomes from advanced cancer patients is a novel strategy to treat cancer [89]. Exosome depletion by dimethyl amiloride (DMA) in mice restores the anti-tumor efficacy of cyclophosphamide (CTX) through the inhibition of MDSC functions. Amiloride, a drug used to treat high blood pressure, inhibits exosome formation and blunts MDSC suppressor functions in colorectal cancer patients [63]. The biotechnology company Aethlon Medical has developed an adjunct therapeutic method HER2osome, which is able to reduce tumor-secreted HER2 positive exosomes in the circulation and thus inhibit HER2-positive breast cancer progression. However, further work is needed to evaluate the clinical safety of such a treatment strategy based on exosome removal.

Exosomes as anti-cancer drug delivery vehicles

The use of exosomes as nucleic acid or drug delivery vehicles has gained considerable interest due to their excellent biodistribution and biocompatibility [90]. Exosome-mediated delivery of therapeutic short interfering RNA (siRNA) to the target cells has been tested. The exosome-delivered siRNA is effective at causing post-transcriptional gene silencing and inducing cell death in recipient cancer cells [9193]. To improve drug delivery efficacy to tumors, the researchers have modified exosomes with targeting ligands such as iRGD-Lamp2b. The modified exosomes show highly efficient targeting to αV integrin-positive breast cancer cells, and intravenous injection of these exosomes obviously inhibits tumor growth [94]. In addition, exosomes have been utilized as effective vehicle for drug delivery [95]. Exosomes from MSCs have been tested as the vehicle to package and deliver active drugs such as paclitaxel [96].

Conclusion

The rapid expansion of the number of published studies on exosomes clearly shows that research on exosomes and their functions is now a very exciting field. Exosomes are small particles with big roles and are emerging as major players in intercellular communication. Exosomes have been suggested as active transporters for proteins, DNA, mRNA, and non-coding RNAs. The roles of exosomes in cancer have been gradually realized. Although some reports have suggested anti-tumor roles of exosomes due to their potential to elicit immune response, most of the reports have revealed the various pro-tumor effects of exosomes, which is further supported by the observations that the level of circulating exosomes is increased in cancer patients and correlated with tumor progression. In this review, we discussed several aspects of exosome biology in cancer. Cancer cells communicate with the surrounding and distant cells via exosomes, which constitutes a bi-directional interaction network to synergistically promote cancer development, progression, metastasis, and drug resistance. However, the exact mechanisms mediating the complex roles of exosomes in cancer have not yet fully elucidated. Exosomes would be ideal biomarkers for cancer diagnosis and targeted therapy because they closely represent the state of their parental cells and are relatively stable in the circulation and could be easily collected from body fluids. The potential of exosomal contents for diagnostic and prognostic biomarkers have been investigated in various cancers. It is required to develop faster and more convenient methods for validating the proposed exosomal cargos as biomarkers in specimens from human cancer patients. The use of nanotechnology to load exosomes with small molecules or drugs for cancer therapy has also been exploited. Improvements in developing new strategies to obtain a large amount of exosomes from appropriate donor cells, efficiently introducing the therapeutic agents into exosomes, and optimizing the targeted delivery of exosomes to particular tissues will facilitate the use of exosomes as natural carrier in clinical therapy. Future studies of exosomes will not only shed lights on their roles in the pathogenesis of cancer but will open new avenues for cancer diagnosis and therapeutics.

Abbreviations

APC:

antigen-presenting cell

ASCs:

adipose-derived stem cells

BM-MSCs:

bone marrow mesenchymal stromal cells

CAFs:

carcinoma-associated fibroblasts

CTX:

cyclophosphamide

Dexosomes:

dendritic cell-derived exosomes

DMA:

dimethyl amiloride

EMT:

epithelial-mesenchymal transition

ESCRT:

endosomal sorting complex required for transport

esRNA:

exosomal shuttle RNA

FIH-1:

factor-inhibiting hypoxia-inducible factor 1

GTPases:

guanosine triphosphatases

HIF-1:

hypoxia-inducible factor 1

HSPGs:

heparan sulfate proteoglycans

ICAM:

intercellular adhesion molecule

ILVs:

Intraluminal vesicles

LncRNA:

long non-coding RNA

MACS:

magnetic activated cell sorting

MDR:

multi-drug resistance

MDSCs:

myeloid-derived suppressor cells

MVBs:

multivesicular bodies

NPC:

nasopharyngeal carcinoma

PCP:

planar cell polarity

RISC:

RNA-induced silencing complex

siRNA:

short interfering RNA

SP:

side population

TEM:

transmission electron microscopy

TF:

tissue factor

TIM:

T cell immunoglobulin and mucin domain molecule

TLR:

toll-like receptor

TSAP6:

tumor suppressor-activated pathway 6

References

  1. Thery C, Zitvogel L, Amigorena S. Exosomes: composition, biogenesis and function. Nat Rev Immunol. 2002;2(8):569–79.

    CAS  PubMed  Google Scholar 

  2. Trajkovic K, Hsu C, Chiantia S, Rajendran L, Wenzel D, Wieland F, et al. Ceramide triggers budding of exosome vesicles into multivesicular endosomes. Science. 2008;319(5867):1244–7.

    CAS  PubMed  Google Scholar 

  3. Baietti MF, Zhang Z, Mortier E, Melchior A, Degeest G, Geeraerts A, et al. Syndecan-syntenin-ALIX regulates the biogenesis of exosomes. Nat Cell Biol. 2012;14(7):677–85.

    CAS  PubMed  Google Scholar 

  4. Ostrowski M, Carmo NB, Krumeich S, Fanget I, Raposo G, Savina A, et al. Rab27a and Rab27b control different steps of the exosome secretion pathway. Nat Cell Biol. 2010;12(1):1–13.

    Google Scholar 

  5. Hsu C, Morohashi Y, Yoshimura S, Manrique-Hoyos N, Jung S, Lauterbach MA, et al. Regulation of exosome secretion by Rab35 and its GTPase-activating proteins TBC1D10A-C. J Cell Biol. 2010;189(2):223–32.

    CAS  PubMed Central  PubMed  Google Scholar 

  6. Savina A, Furlan M, Vidal M, Colombo MI. Exosome release is regulated by a calcium-dependent mechanism in K562 cells. J Biol Chem. 2003;278(22):20083–90.

    CAS  PubMed  Google Scholar 

  7. Parolini I, Federici C, Raggi C, Lugini L, Palleschi S, De Milito A, et al. Microenvironmental pH is a key factor for exosome traffic in tumor cells. J Biol Chem. 2009;284(49):34211–22.

    CAS  PubMed Central  PubMed  Google Scholar 

  8. Yu JL, May L, Lhotak V, Shahrzad S, Shirasawa S, Weitz JI, et al. Oncogenic events regulate tissue factor expression in colorectal cancer cells: implications for tumor progression and angiogenesis. Blood. 2005;105(4):1734–41.

    CAS  PubMed  Google Scholar 

  9. Yu X, Harris SL, Levine AJ. The regulation of exosome secretion: a novel function of the p53 protein. Cancer Res. 2006;66(9):4795–801.

    CAS  PubMed  Google Scholar 

  10. Lespagnol A, Duflaut D, Beekman C, Blanc L, Fiucci G, Marine JC, et al. Exosome secretion, including the DNA damage-induced p53-dependent secretory pathway, is severely compromised in TSAP6/Steap3-null mice. Cell Death Differ. 2008;15(11):1723–33.

    CAS  PubMed  Google Scholar 

  11. Thompson CA, Purushothaman A, Ramani VC, Vlodavsky I, Sanderson RD. Heparanase regulates secretion, composition, and function of tumor cell-derived exosomes. J Biol Chem. 2013;288(14):10093–9.

    CAS  PubMed Central  PubMed  Google Scholar 

  12. Riches A, Campbell E, Borger E, Powis S. Regulation of exosome release from mammary epithelial and breast cancer cells - a new regulatory pathway. Eur J Cancer. 2014;50(5):1025–34.

    CAS  PubMed  Google Scholar 

  13. Miyanishi M, Tada K, Koike M, Uchiyama Y, Kitamura T, Nagata S. Identification of Tim4 as a phosphatidylserine receptor. Nature. 2007;450(7168):435–9.

    CAS  PubMed  Google Scholar 

  14. Segura E, Nicco C, Lombard B, Veron P, Raposo G, Batteux F, et al. ICAM-1 on exosomes from mature dendritic cells is critical for efficient naive T-cell priming. Blood. 2005;106(1):216–23.

    CAS  PubMed  Google Scholar 

  15. Tian T, Wang Y, Wang H, Zhu Z, Xiao Z. Visualizing of the cellular uptake and intracellular trafficking of exosomes by live-cell microscopy. J Cell Biochem. 2010;111(2):488–96.

    CAS  PubMed  Google Scholar 

  16. Feng D, Zhao WL, Ye YY, Bai XC, Liu RQ, Chang LF, et al. Cellular internalization of exosomes occurs through phagocytosis. Traffic. 2010;11(5):675–87.

    CAS  PubMed  Google Scholar 

  17. Escrevente C, Keller S, Altevogt P, Costa J. Interaction and uptake of exosomes by ovarian cancer cells. BMC Cancer. 2011;11:108.

    CAS  PubMed Central  PubMed  Google Scholar 

  18. Christianson HC, Svensson KJ, van Kuppevelt TH, Li JP, Belting M. Cancer cell exosomes depend on cell-surface heparan sulfate proteoglycans for their internalization and functional activity. Proc Natl Acad Sci U S A. 2013;110(43):17380–5.

    CAS  PubMed Central  PubMed  Google Scholar 

  19. Thakur BK, Zhang H, Becker A, Matei I, Huang Y, Costa-Silva B, et al. Double-stranded DNA in exosomes: a novel biomarker in cancer detection. Cell Res. 2014;24(6):766–9.

    CAS  PubMed Central  PubMed  Google Scholar 

  20. Valadi H, Ekstrom K, Bossios A, Sjostrand M, Lee JJ, Lotvall JO. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol. 2007;9(6):654–9.

    CAS  PubMed  Google Scholar 

  21. Lotvall J, Hill AF, Hochberg F, Buzas EI, Di Vizio D, Gardiner C, et al. Minimal experimental requirements for definition of extracellular vesicles and their functions: a position statement from the International Society for Extracellular Vesicles. J Extracell Vesicles. 2014;3:26913.

    PubMed  Google Scholar 

  22. Abd Elmageed ZY, Yang Y, Thomas R, Ranjan M, Mondal D, Moroz K, et al. Neoplastic reprogramming of patient-derived adipose stem cells by prostate cancer cell-associated exosomes. Stem Cells. 2014;32(4):983–97.

    CAS  PubMed Central  PubMed  Google Scholar 

  23. Melo SA, Sugimoto H, O'Connell JT, Kato N, Villanueva A, Vidal A, et al. Cancer exosomes perform cell-independent microRNA biogenesis and promote tumorigenesis. Cancer Cell. 2014;26(5):707–21.

    CAS  PubMed  Google Scholar 

  24. Koch R, Demant M, Aung T, Diering N, Cicholas A, Chapuy B, et al. Populational equilibrium through exosome-mediated Wnt signaling in tumor progression of diffuse large B-cell lymphoma. Blood. 2014;123(14):2189–98.

    CAS  PubMed  Google Scholar 

  25. Khan S, Aspe JR, Asumen MG, Almaguel F, Odumosu O, Acevedo-Martinez S, et al. Extracellular, cell-permeable survivin inhibits apoptosis while promoting proliferative and metastatic potential. Br J Cancer. 2009;100(7):1073–86.

    CAS  PubMed Central  PubMed  Google Scholar 

  26. Al-Nedawi K, Meehan B, Micallef J, Lhotak V, May L, Guha A, et al. Intercellular transfer of the oncogenic receptor EGFRvIII by microvesicles derived from tumour cells. Nat Cell Biol. 2008;10(5):619–24.

    CAS  PubMed  Google Scholar 

  27. Soldevilla B, Rodriguez M, San Millan C, Garcia V, Fernandez-Perianez R, Gil-Calderon B, et al. Tumor-derived exosomes are enriched in DeltaNp73, which promotes oncogenic potential in acceptor cells and correlates with patient survival. Hum Mol Genet. 2014;23(2):467–78.

    CAS  PubMed  Google Scholar 

  28. Corrado C, Raimondo S, Saieva L, Flugy AM, De Leo G, Alessandro R. Exosome-mediated crosstalk between chronic myelogenous leukemia cells and human bone marrow stromal cells triggers an interleukin 8-dependent survival of leukemia cells. Cancer Lett. 2014;348(1–2):71–6.

    CAS  PubMed  Google Scholar 

  29. Roccaro AM, Sacco A, Maiso P, Azab AK, Tai YT, Reagan M, et al. BM mesenchymal stromal cell-derived exosomes facilitate multiple myeloma progression. J Clin Invest. 2013;123(4):1542–55.

    CAS  PubMed Central  PubMed  Google Scholar 

  30. Park JE, Tan HS, Datta A, Lai RC, Zhang H, Meng W, et al. Hypoxic tumor cell modulates its microenvironment to enhance angiogenic and metastatic potential by secretion of proteins and exosomes. Mol Cell Proteomics. 2010;9(6):1085–99.

    CAS  PubMed Central  PubMed  Google Scholar 

  31. Nazarenko I, Rana S, Baumann A, McAlear J, Hellwig A, Trendelenburg M, et al. Cell surface tetraspanin Tspan8 contributes to molecular pathways of exosome-induced endothelial cell activation. Cancer Res. 2010;70(4):1668–78.

    CAS  PubMed  Google Scholar 

  32. Kucharzewska P, Christianson HC, Welch JE, Svensson KJ, Fredlund E, Ringner M, et al. Exosomes reflect the hypoxic status of glioma cells and mediate hypoxia-dependent activation of vascular cells during tumor development. Proc Natl Acad Sci U S A. 2013;110(18):7312–7.

    CAS  PubMed Central  PubMed  Google Scholar 

  33. Zhou W, Fong MY, Min Y, Somlo G, Liu L, Palomares MR, et al. Cancer-secreted miR-105 destroys vascular endothelial barriers to promote metastasis. Cancer Cell. 2014;25(4):501–15.

    CAS  PubMed Central  PubMed  Google Scholar 

  34. Umezu T, Ohyashiki K, Kuroda M, Ohyashiki JH. Leukemia cell to endothelial cell communication via exosomal miRNAs. Oncogene. 2013;32(22):2747–55.

    CAS  PubMed  Google Scholar 

  35. Tadokoro H, Umezu T, Ohyashiki K, Hirano T, Ohyashiki JH. Exosomes derived from hypoxic leukemia cells enhance tube formation in endothelial cells. J Biol Chem. 2013;288(48):34343–51.

    CAS  PubMed Central  PubMed  Google Scholar 

  36. Umezu T, Tadokoro H, Azuma K, Yoshizawa S, Ohyashiki K, Ohyashiki JH. Exosomal miR-135b shed from hypoxic multiple myeloma cells enhances angiogenesis by targeting factor-inhibiting HIF-1. Blood. 2014;124(25):3748–57.

    CAS  PubMed Central  PubMed  Google Scholar 

  37. Aga M, Bentz GL, Raffa S, Torrisi MR, Kondo S, Wakisaka N, et al. Exosomal HIF1alpha supports invasive potential of nasopharyngeal carcinoma-associated LMP1-positive exosomes. Oncogene. 2014;33(37):4613–22.

    CAS  PubMed Central  PubMed  Google Scholar 

  38. Ostenfeld MS, Jeppesen DK, Laurberg JR, Boysen AT, Bramsen JB, Primdal-Bengtson B, et al. Cellular disposal of miR23b by RAB27-dependent exosome release is linked to acquisition of metastatic properties. Cancer Res. 2014;74(20):5758–71.

    CAS  PubMed  Google Scholar 

  39. Peinado H, Aleckovic M, Lavotshkin S, Matei I, Costa-Silva B, Moreno-Bueno G, et al. Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat Med. 2012;18(6):883–91.

    CAS  PubMed Central  PubMed  Google Scholar 

  40. Atay S, Banskota S, Crow J, Sethi G, Rink L, Godwin AK. Oncogenic KIT-containing exosomes increase gastrointestinal stromal tumor cell invasion. Proc Natl Acad Sci U S A. 2014;111(2):711–6.

    CAS  PubMed Central  PubMed  Google Scholar 

  41. Suetsugu A, Honma K, Saji S, Moriwaki H, Ochiya T, Hoffman RM. Imaging exosome transfer from breast cancer cells to stroma at metastatic sites in orthotopic nude-mouse models. Adv Drug Deliv Rev. 2013;65(3):383–90.

    CAS  PubMed  Google Scholar 

  42. Luga V, Zhang L, Viloria-Petit AM, Ogunjimi AA, Inanlou MR, Chiu E, et al. Exosomes mediate stromal mobilization of autocrine Wnt-PCP signaling in breast cancer cell migration. Cell. 2012;151(7):1542–56.

    CAS  PubMed  Google Scholar 

  43. Cai Z, Yang F, Yu L, Yu Z, Jiang L, Wang Q, et al. Activated T cell exosomes promote tumor invasion via Fas signaling pathway. J Immunol. 2012;188(12):5954–61.

    CAS  PubMed  Google Scholar 

  44. Yang M, Chen J, Su F, Yu B, Lin L, Liu Y, et al. Microvesicles secreted by macrophages shuttle invasion-potentiating microRNAs into breast cancer cells. Mol Cancer. 2011;10:117.

    CAS  PubMed Central  PubMed  Google Scholar 

  45. Wang M, Zhao C, Shi H, Zhang B, Zhang L, Zhang X, et al. Deregulated microRNAs in gastric cancer tissue-derived mesenchymal stem cells: novel biomarkers and a mechanism for gastric cancer. Br J Cancer. 2014;110(5):1199–210.

    CAS  PubMed Central  PubMed  Google Scholar 

  46. Fabbri M, Paone A, Calore F, Galli R, Gaudio E, Santhanam R, et al. MicroRNAs bind to toll-like receptors to induce prometastatic inflammatory response. Proc Natl Acad Sci U S A. 2012;109(31):E2110–2116.

    CAS  PubMed Central  PubMed  Google Scholar 

  47. Costa-Silva B, Aiello NM, Ocean AJ, Singh S, Zhang H, Thakur BK, et al. Pancreatic cancer exosomes initiate pre-metastatic niche formation in the liver. Nat Cell Biol. 2015;17(6):816–26.

    PubMed  Google Scholar 

  48. Zomer A, Maynard C, Verweij FJ, Kamermans A, Schafer R, Beerling E, et al. In Vivo imaging reveals extracellular vesicle-mediated phenocopying of metastatic behavior. Cell. 2015;161(5):1046–57.

    CAS  PubMed Central  PubMed  Google Scholar 

  49. Corcoran C, Rani S, O'Brien K, O'Neill A, Prencipe M, Sheikh R, et al. Docetaxel-resistance in prostate cancer: evaluating associated phenotypic changes and potential for resistance transfer via exosomes. PLoS One. 2012;7(12):e50999.

    CAS  PubMed Central  PubMed  Google Scholar 

  50. Wei Y, Lai X, Yu S, Chen S, Ma Y, Zhang Y, et al. Exosomal miR-221/222 enhances tamoxifen resistance in recipient ER-positive breast cancer cells. Breast Cancer Res Treat. 2014;147(2):423–31.

    CAS  PubMed  Google Scholar 

  51. Shedden K, Xie XT, Chandaroy P, Chang YT, Rosania GR. Expulsion of small molecules in vesicles shed by cancer cells: association with gene expression and chemosensitivity profiles. Cancer Res. 2003;63(15):4331–7.

    CAS  PubMed  Google Scholar 

  52. Safaei R, Larson BJ, Cheng TC, Gibson MA, Otani S, Naerdemann W, et al. Abnormal lysosomal trafficking and enhanced exosomal export of cisplatin in drug-resistant human ovarian carcinoma cells. Mol Cancer Ther. 2005;4(10):1595–604.

    CAS  PubMed  Google Scholar 

  53. Chen KG, Valencia JC, Lai B, Zhang G, Paterson JK, Rouzaud F, et al. Melanosomal sequestration of cytotoxic drugs contributes to the intractability of malignant melanomas. Proc Natl Acad Sci U S A. 2006;103(26):9903–7.

    CAS  PubMed Central  PubMed  Google Scholar 

  54. Aung T, Chapuy B, Vogel D, Wenzel D, Oppermann M, Lahmann M, et al. Exosomal evasion of humoral immunotherapy in aggressive B-cell lymphoma modulated by ATP-binding cassette transporter A3. Proc Natl Acad Sci U S A. 2011;108(37):15336–41.

    CAS  PubMed Central  PubMed  Google Scholar 

  55. Ciravolo V, Huber V, Ghedini GC, Venturelli E, Bianchi F, Campiglio M, et al. Potential role of HER2-overexpressing exosomes in countering trastuzumab-based therapy. J Cell Physiol. 2012;227(2):658–67.

    CAS  PubMed  Google Scholar 

  56. Battke C, Ruiss R, Welsch U, Wimberger P, Lang S, Jochum S, et al. Tumour exosomes inhibit binding of tumour-reactive antibodies to tumour cells and reduce ADCC. Cancer Immunol Immunother. 2011;60(5):639–48.

    CAS  PubMed  Google Scholar 

  57. Yu S, Liu C, Su K, Wang J, Liu Y, Zhang L, et al. Tumor exosomes inhibit differentiation of bone marrow dendritic cells. J Immunol. 2007;178(11):6867–75.

    CAS  PubMed  Google Scholar 

  58. Clayton A, Mitchell JP, Court J, Mason MD, Tabi Z. Human tumor-derived exosomes selectively impair lymphocyte responses to interleukin-2. Cancer Res. 2007;67(15):7458–66.

    CAS  PubMed  Google Scholar 

  59. Mrizak D, Martin N, Barjon C, Jimenez-Pailhes AS, Mustapha R, Niki T, et al. Effect of nasopharyngeal carcinoma-derived exosomes on human regulatory T cells. J Natl Cancer Inst. 2014;107(1):363.

    PubMed  Google Scholar 

  60. Ye SB, Li ZL, Luo DH, Huang BJ, Chen YS, Zhang XS, et al. Tumor-derived exosomes promote tumor progression and T-cell dysfunction through the regulation of enriched exosomal microRNAs in human nasopharyngeal carcinoma. Oncotarget. 2014;5(14):5439–52.

    PubMed Central  PubMed  Google Scholar 

  61. Xiang X, Poliakov A, Liu C, Liu Y, Deng ZB, Wang J, et al. Induction of myeloid-derived suppressor cells by tumor exosomes. Int J Cancer. 2009;124(11):2621–33.

    CAS  PubMed Central  PubMed  Google Scholar 

  62. Liu Y, Xiang X, Zhuang X, Zhang S, Liu C, Cheng Z, et al. Contribution of MyD88 to the tumor exosome-mediated induction of myeloid derived suppressor cells. Am J Pathol. 2010;176(5):2490–9.

    CAS  PubMed Central  PubMed  Google Scholar 

  63. Chalmin F, Ladoire S, Mignot G, Vincent J, Bruchard M, Remy-Martin JP, et al. Membrane-associated Hsp72 from tumor-derived exosomes mediates STAT3-dependent immunosuppressive function of mouse and human myeloid-derived suppressor cells. J Clin Invest. 2010;120(2):457–71.

    CAS  PubMed Central  PubMed  Google Scholar 

  64. Chow A, Zhou W, Liu L, Fong MY, Champer J, Van Haute D, et al. Macrophage immunomodulation by breast cancer-derived exosomes requires toll-like receptor 2-mediated activation of NF-kappaB. Sci Rep. 2014;4:5750.

    CAS  PubMed Central  PubMed  Google Scholar 

  65. Lundholm M, Schroder M, Nagaeva O, Baranov V, Widmark A, Mincheva-Nilsson L, et al. Prostate tumor-derived exosomes down-regulate NKG2D expression on natural killer cells and CD8+ T cells: mechanism of immune evasion. PLoS One. 2014;9(9):e108925.

    PubMed Central  PubMed  Google Scholar 

  66. Webber J, Steadman R, Mason MD, Tabi Z, Clayton A. Cancer exosomes trigger fibroblast to myofibroblast differentiation. Cancer Res. 2010;70(23):9621–30.

    CAS  PubMed  Google Scholar 

  67. Webber JP, Spary LK, Sanders AJ, Chowdhury R, Jiang WG, Steadman R, et al. Differentiation of tumour-promoting stromal myofibroblasts by cancer exosomes. Oncogene. 2015;34(3):290–302.

    CAS  PubMed  Google Scholar 

  68. Gu J, Qian H, Shen L, Zhang X, Zhu W, Huang L, et al. Gastric cancer exosomes trigger differentiation of umbilical cord derived mesenchymal stem cells to carcinoma-associated fibroblasts through TGF-beta/Smad pathway. PLoS One. 2012;7(12):e52465.

    CAS  PubMed Central  PubMed  Google Scholar 

  69. Cho JA, Park H, Lim EH, Lee KW. Exosomes from breast cancer cells can convert adipose tissue-derived mesenchymal stem cells into myofibroblast-like cells. Int J Oncol. 2012;40(1):130–8.

    CAS  PubMed  Google Scholar 

  70. Boelens MC, Wu TJ, Nabet BY, Xu B, Qiu Y, Yoon T, et al. Exosome transfer from stromal to breast cancer cells regulates therapy resistance pathways. Cell. 2014;159(3):499–513.

    CAS  PubMed  Google Scholar 

  71. Garnier D, Magnus N, Lee TH, Bentley V, Meehan B, Milsom C, et al. Cancer cells induced to express mesenchymal phenotype release exosome-like extracellular vesicles carrying tissue factor. J Biol Chem. 2012;287(52):43565–72.

    CAS  PubMed Central  PubMed  Google Scholar 

  72. Taylor DD, Gercel-Taylor C. MicroRNA signatures of tumor-derived exosomes as diagnostic biomarkers of ovarian cancer. Gynecol Oncol. 2008;110(1):13–21.

    CAS  PubMed  Google Scholar 

  73. Gallo A, Tandon M, Alevizos I, Illei GG. The majority of microRNAs detectable in serum and saliva is concentrated in exosomes. PLoS One. 2012;7(3):e30679.

    CAS  PubMed Central  PubMed  Google Scholar 

  74. Wang W, Chen Y. Circulating miRNAs in cancer: from detection to therapy. J Hematol Oncol. 2014;7(1):86.

    CAS  PubMed Central  PubMed  Google Scholar 

  75. Skog J, Wurdinger T, van Rijn S, Meijer DH, Gainche L, Sena-Esteves M, et al. Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers. Nat Cell Biol. 2008;10(12):1470–6.

    CAS  PubMed Central  PubMed  Google Scholar 

  76. Rabinowits G, Gercel-Taylor C, Day JM, Taylor DD, Kloecker GH. Exosomal microRNA: a diagnostic marker for lung cancer. Clin Lung Cancer. 2009;10(1):42–6.

    CAS  PubMed  Google Scholar 

  77. Lau C, Kim Y, Chia D, Spielmann N, Eibl G, Elashoff D, et al. Role of pancreatic cancer-derived exosomes in salivary biomarker development. J Biol Chem. 2013;288(37):26888–97.

    CAS  PubMed Central  PubMed  Google Scholar 

  78. Nilsson J, Skog J, Nordstrand A, Baranov V, Mincheva-Nilsson L, Breakefield XO, et al. Prostate cancer-derived urine exosomes: a novel approach to biomarkers for prostate cancer. Br J Cancer. 2009;100(10):1603–7.

    CAS  PubMed Central  PubMed  Google Scholar 

  79. Corcoran C, Friel AM, Duffy MJ, Crown J, O'Driscoll L. Intracellular and extracellular microRNAs in breast cancer. Clin Chem. 2011;57(1):18–32.

    CAS  PubMed  Google Scholar 

  80. Tanaka Y, Kamohara H, Kinoshita K, Kurashige J, Ishimoto T, Iwatsuki M, et al. Clinical impact of serum exosomal microRNA-21 as a clinical biomarker in human esophageal squamous cell carcinoma. Cancer. 2013;119(6):1159–67.

    CAS  PubMed  Google Scholar 

  81. Kogure T, Yan IK, Lin WL, Patel T. Extracellular vesicle-mediated transfer of a novel long noncoding RNA TUC339: a mechanism of intercellular signaling in human hepatocellular cancer. Genes Cancer. 2013;4(7–8):261–72.

    CAS  PubMed Central  PubMed  Google Scholar 

  82. Takahashi K, Yan IK, Kogure T, Haga H, Patel T. Extracellular vesicle-mediated transfer of long non-coding RNA ROR modulates chemosensitivity in human hepatocellular cancer. FEBS Open Bio. 2014;4:458–67.

    CAS  PubMed Central  PubMed  Google Scholar 

  83. Li Q, Shao Y, Zhang X, Zheng T, Miao M, Qin L, et al. Plasma long noncoding RNA protected by exosomes as a potential stable biomarker for gastric cancer. Tumour Biol. 2015;36(3):2007–12.

    CAS  PubMed  Google Scholar 

  84. Wang J, Zhou Y, Lu J, Sun Y, Xiao H, Liu M, et al. Combined detection of serum exosomal miR-21 and HOTAIR as diagnostic and prognostic biomarkers for laryngeal squamous cell carcinoma. Med Oncol. 2014;31(9):148.

    PubMed  Google Scholar 

  85. Pitt JM, Charrier M, Viaud S, Andre F, Besse B, Chaput N, et al. Dendritic cell-derived exosomes as immunotherapies in the fight against cancer. J Immunol. 2014;193(3):1006–11.

    CAS  PubMed  Google Scholar 

  86. Zitvogel L, Regnault A, Lozier A, Wolfers J, Flament C, Tenza D, et al. Eradication of established murine tumors using a novel cell-free vaccine: dendritic cell-derived exosomes. Nat Med. 1998;4(5):594–600.

    CAS  PubMed  Google Scholar 

  87. Wolfers J, Lozier A, Raposo G, Regnault A, Thery C, Masurier C, et al. Tumor-derived exosomes are a source of shared tumor rejection antigens for CTL cross-priming. Nat Med. 2001;7(3):297–303.

    CAS  PubMed  Google Scholar 

  88. Tan A, De La Pena H, Seifalian AM. The application of exosomes as a nanoscale cancer vaccine. Int J Nanomedicine. 2010;5:889–900.

    CAS  PubMed Central  PubMed  Google Scholar 

  89. Marleau AM, Chen CS, Joyce JA, Tullis RH. Exosome removal as a therapeutic adjuvant in cancer. J Transl Med. 2012;10:134.

    CAS  PubMed Central  PubMed  Google Scholar 

  90. van den Boorn JG, Dassler J, Coch C, Schlee M, Hartmann G. Exosomes as nucleic acid nanocarriers. Adv Drug Deliv Rev. 2013;65(3):331–5.

    PubMed  Google Scholar 

  91. Shtam TA, Kovalev RA, Varfolomeeva EY, Makarov EM, Kil YV, Filatov MV. Exosomes are natural carriers of exogenous siRNA to human cells in vitro. Cell Commun Signal. 2013;11:88.

    CAS  PubMed Central  PubMed  Google Scholar 

  92. Zhang Y, Li L, Yu J, Zhu D, Li X, Gu H, et al. Microvesicle-mediated delivery of transforming growth factor beta1 siRNA for the suppression of tumor growth in mice. Biomaterials. 2014;35(14):4390–400.

    CAS  PubMed  Google Scholar 

  93. Alvarez-Erviti L, Seow Y, Yin H, Betts C, Lakhal S, Wood MJ. Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes. Nat Biotechnol. 2011;29(4):341–5.

    CAS  PubMed  Google Scholar 

  94. Tian Y, Li S, Song J, Ji T, Zhu M, Anderson GJ, et al. A doxorubicin delivery platform using engineered natural membrane vesicle exosomes for targeted tumor therapy. Biomaterials. 2014;35(7):2383–90.

    CAS  PubMed  Google Scholar 

  95. Smyth T, Kullberg M, Malik N, Smith-Jones P, Graner MW, Anchordoquy TJ. Biodistribution and delivery efficiency of unmodified tumor-derived exosomes. J Control Release. 2015;199:145–55.

    CAS  PubMed  Google Scholar 

  96. Pascucci L, Cocce V, Bonomi A, Ami D, Ceccarelli P, Ciusani E, 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. 2014;192:262–70.

    CAS  PubMed  Google Scholar 

  97. Demory Beckler M, Higginbotham JN, Franklin JL, Ham AJ, Halvey PJ, Imasuen IE, et al. Proteomic analysis of exosomes from mutant KRAS colon cancer cells identifies intercellular transfer of mutant KRAS. Mol Cell Proteomics. 2013;12(2):343–55.

    PubMed Central  PubMed  Google Scholar 

  98. Fedele C, Singh A, Zerlanko BJ, Iozzo RV, Languino LR. The alphavbeta6 integrin is transferred intercellularly via exosomes. J Biol Chem. 2015;290(8):4545–51.

    CAS  PubMed  Google Scholar 

  99. Khan S, Jutzy JM, Aspe JR, McGregor DW, Neidigh JW, Wall NR. Survivin is released from cancer cells via exosomes. Apoptosis. 2011;16(1):1–12.

    CAS  PubMed Central  PubMed  Google Scholar 

  100. Menck K, Klemm F, Gross JC, Pukrop T, Wenzel D, Binder C. Induction and transport of Wnt5a during macrophage-induced malignant invasion is mediated by two types of extracellular vesicles. Oncotarget. 2013;4(11):2057–66.

    PubMed Central  PubMed  Google Scholar 

  101. Clayton A, Al-Taei S, Webber J, Mason MD, Tabi Z. Cancer exosomes express CD39 and CD73, which suppress T cells through adenosine production. J Immunol. 2011;187(2):676–83.

    CAS  PubMed  Google Scholar 

  102. Chowdhury R, Webber JP, Gurney M, Mason MD, Tabi Z, Clayton A. Cancer exosomes trigger mesenchymal stem cell differentiation into pro-angiogenic and pro-invasive myofibroblasts. Oncotarget. 2015;6(2):715–31.

    PubMed Central  PubMed  Google Scholar 

  103. Clayton A, Mitchell JP, Court J, Linnane S, Mason MD, Tabi Z. Human tumor-derived exosomes down-modulate NKG2D expression. J Immunol. 2008;180(11):7249–58.

    CAS  PubMed  Google Scholar 

  104. Ashiru O, Boutet P, Fernandez-Messina L, Aguera-Gonzalez S, Skepper JN, Vales-Gomez M, et al. Natural killer cell cytotoxicity is suppressed by exposure to the human NKG2D ligand MICA*008 that is shed by tumor cells in exosomes. Cancer Res. 2010;70(2):481–9.

    CAS  PubMed Central  PubMed  Google Scholar 

  105. Xiao H, Lasser C, Shelke GV, Wang J, Radinger M, Lunavat TR, et al. Mast cell exosomes promote lung adenocarcinoma cell proliferation—role of KIT-stem cell factor signaling. Cell Commun Signal. 2014;12:64.

    PubMed Central  PubMed  Google Scholar 

  106. Diao J, Yang X, Song X, Chen S, He Y, Wang Q, et al. Exosomal Hsp70 mediates immunosuppressive activity of the myeloid-derived suppressor cells via phosphorylation of Stat3. Med Oncol. 2015;32(2):453.

    PubMed  Google Scholar 

  107. Sagar G, Sah RP, Javeed N, Dutta SK, Smyrk TC, Lau JS, et al. Pathogenesis of pancreatic cancer exosome-induced lipolysis in adipose tissue. Gut 2015, doi:gutjnl-2014-308350

  108. Deng Z, Mu J, Tseng M, Wattenberg B, Zhuang X, Egilmez NK, et al. Enterobacteria-secreted particles induce production of exosome-like S1P-containing particles by intestinal epithelium to drive Th17-mediated tumorigenesis. Nat Commun. 2012;6:6956.

    Google Scholar 

  109. Zhuang G, Wu X, Jiang Z, Kasman I, Yao J, Guan Y, et al. Tumour-secreted miR-9 promotes endothelial cell migration and angiogenesis by activating the JAK-STAT pathway. EMBO J. 2012;31(17):3513–23.

    CAS  PubMed Central  PubMed  Google Scholar 

  110. Singh R, Pochampally R, Watabe K, Lu Z, Mo YY. Exosome-mediated transfer of miR-10b promotes cell invasion in breast cancer. Mol Cancer. 2014;13:256.

    CAS  PubMed Central  PubMed  Google Scholar 

  111. Chen WX, Liu XM, Lv MM, Chen L, Zhao JH, Zhong SL, et al. Exosomes from drug-resistant breast cancer cells transmit chemoresistance by a horizontal transfer of microRNAs. PLoS One. 2014;9(4):e95240.

    PubMed Central  PubMed  Google Scholar 

  112. Kogure T, Lin WL, Yan IK, Braconi C, Patel T. Intercellular nanovesicle-mediated microRNA transfer: a mechanism of environmental modulation of hepatocellular cancer cell growth. Hepatology. 2011;54(4):1237–48.

    CAS  PubMed Central  PubMed  Google Scholar 

  113. Fong MY, Zhou W, Liu L, Alontaga AY, Chandra M, Ashby J, et al. Breast-cancer-secreted miR-122 reprograms glucose metabolism in premetastatic niche to promote metastasis. Nat Cell Biol. 2015;17(2):183–94.

    CAS  PubMed  Google Scholar 

  114. Bovy N, Blomme B, Freres P, Dederen S, Nivelles O, Lion M, et al. Endothelial exosomes contribute to the antitumor response during breast cancer neoadjuvant chemotherapy via microRNA transfer. Oncotarget. 2015;6(12):10253–66.

    PubMed  Google Scholar 

  115. Gernapudi R, Yao Y, Zhang Y, Wolfson B, Roy S, Duru N, et al. Targeting exosomes from preadipocytes inhibits preadipocyte to cancer stem cell signaling in early-stage breast cancer. Breast Cancer Res Treat. 2015;150(3):685–95.

    PubMed  Google Scholar 

  116. Lim PK, Bliss SA, Patel SA, Taborga M, Dave MA, Gregory LA, et al. Gap junction-mediated import of microRNA from bone marrow stromal cells can elicit cell cycle quiescence in breast cancer cells. Cancer Res. 2011;71(5):1550–60.

    CAS  PubMed  Google Scholar 

  117. Hong CS, Muller L, Boyiadzis M, Whiteside TL. Isolation and characterization of CD34+ blast-derived exosomes in acute myeloid leukemia. PLoS One. 2014;9(8):e103310.

    PubMed Central  PubMed  Google Scholar 

  118. Beckham CJ, Olsen J, Yin PN, Wu CH, Ting HJ, Hagen FK, et al. Bladder cancer exosomes contain EDIL-3/Del1 and facilitate cancer progression. J Urol. 2014;192(2):583–92.

    CAS  PubMed  Google Scholar 

  119. Eichelser C, Stuckrath I, Muller V, Milde-Langosch K, Wikman H, Pantel K, et al. Increased serum levels of circulating exosomal microRNA-373 in receptor-negative breast cancer patients. Oncotarget. 2014;5(20):9650–63.

    PubMed Central  PubMed  Google Scholar 

  120. Liu J, Sun H, Wang X, Yu Q, Li S, Yu X, et al. Increased exosomal microRNA-21 and microRNA-146a levels in the cervicovaginal lavage specimens of patients with cervical cancer. Int J Mol Sci. 2014;15(1):758–73.

    PubMed Central  PubMed  Google Scholar 

  121. Ogata-Kawata H, Izumiya M, Kurioka D, Honma Y, Yamada Y, Furuta K, et al. Circulating exosomal microRNAs as biomarkers of colon cancer. PLoS One. 2014;9(4):e92921.

    PubMed Central  PubMed  Google Scholar 

  122. Yoshioka Y, Kosaka N, Konishi Y, Ohta H, Okamoto H, Sonoda H, et al. Ultra-sensitive liquid biopsy of circulating extracellular vesicles using ExoScreen. Nat Commun. 2014;5:3591.

    PubMed Central  PubMed  Google Scholar 

  123. Matsumura T, Sugimachi K, Iinuma H, Takahashi Y, Kurashige J, Sawada G, et al. Exosomal microRNA in serum is a novel biomarker of recurrence in human colorectal cancer. Br J Cancer 2015, doi:bjc2015201

  124. Sugimachi K, Matsumura T, Hirata H, Uchi R, Ueda M, Ueo H, et al. Identification of a bona fide microRNA biomarker in serum exosomes that predicts hepatocellular carcinoma recurrence after liver transplantation. Br J Cancer. 2015;112(3):532–8.

    CAS  PubMed  Google Scholar 

  125. Wang H, Hou L, Li A, Duan Y, Gao H, Song X. Expression of serum exosomal microRNA-21 in human hepatocellular carcinoma. Biomed Res Int. 2014;2014:864894.

    PubMed Central  PubMed  Google Scholar 

  126. Li Y, Zhang Y, Qiu F, Qiu Z. Proteomic identification of exosomal LRG1: a potential urinary biomarker for detecting NSCLC. Electrophoresis. 2011;32(15):1976–83.

    CAS  PubMed  Google Scholar 

  127. Logozzi M, De Milito A, Lugini L, Borghi M, Calabro L, Spada M, et al. High levels of exosomes expressing CD63 and caveolin-1 in plasma of melanoma patients. PLoS One. 2009;4(4):e5219.

    PubMed Central  PubMed  Google Scholar 

  128. Klibi J, Niki T, Riedel A, Pioche-Durieu C, Souquere S, Rubinstein E, et al. Blood diffusion and Th1-suppressive effects of galectin-9-containing exosomes released by Epstein-Barr virus-infected nasopharyngeal carcinoma cells. Blood. 2009;113(9):1957–66.

    CAS  PubMed  Google Scholar 

  129. Li J, Sherman-Baust CA, Tsai-Turton M, Bristow RE, Roden RB, Morin PJ. Claudin-containing exosomes in the peripheral circulation of women with ovarian cancer. BMC Cancer. 2009;9:244.

    PubMed Central  PubMed  Google Scholar 

  130. Madhavan B, Yue S, Galli U, Rana S, Gross W, Muller M, et al. Combined evaluation of a panel of protein and miRNA serum-exosome biomarkers for pancreatic cancer diagnosis increases sensitivity and specificity. Int J Cancer. 2015;136(11):2616–27.

    CAS  PubMed  Google Scholar 

  131. Gabriel K, Ingram A, Austin R, Kapoor A, Tang D, Majeed F, et al. Regulation of the tumor suppressor PTEN through exosomes: a diagnostic potential for prostate cancer. PLoS One. 2013;8(7):e70047.

    CAS  PubMed Central  PubMed  Google Scholar 

  132. Khan S, Jutzy JM, Valenzuela MM, Turay D, Aspe JR, Ashok A, et al. Plasma-derived exosomal survivin, a plausible biomarker for early detection of prostate cancer. PLoS One. 2012;7(10):e46737.

    CAS  PubMed Central  PubMed  Google Scholar 

  133. Mitchell PJ, Welton J, Staffurth J, Court J, Mason MD, Tabi Z, et al. Can urinary exosomes act as treatment response markers in prostate cancer? J Transl Med. 2009;7:4.

    PubMed Central  PubMed  Google Scholar 

  134. Huang X, Yuan T, Liang M, Du M, Xia S, Dittmar R, et al. Exosomal miR-1290 and miR-375 as prognostic markers in castration-resistant prostate cancer. Eur Urol. 2015;67(1):33–41.

    CAS  PubMed  Google Scholar 

  135. Isin M, Uysaler E, Ozgur E, Koseoglu H, Sanli O, Yucel OB, et al. Exosomal lncRNA-p21 levels may help to distinguish prostate cancer from benign disease. Front Genet. 2015;6:168.

    PubMed Central  PubMed  Google Scholar 

Download references

Acknowledgements

We thank members of the Xu Lab for their excellent work and helpful discussions. This work was supported by the following: Major Research Plan of the National Natural Science Foundation of China (grant no. 91129718), the National Natural Science Foundation of China (grant no. 81201660), the Natural Science Foundation of the Jiangsu Province (grant no. BK20141303), Jiangsu Province’s Project of Scientific and Technological Innovation and Achievements Transformation (grant no. BL2012055), Jiangsu Province’s Outstanding Medical Academic Leader and Sci-tech Innovation Team Program (grant no. LJ201117), Jiangsu Province’s Qing Lan project, Foundation for Young Academic Leader of Jiangsu University, Starting Foundation for Senior Talents of Jiangsu University (grant no. 13JDG086).

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Xu Zhang or Wenrong Xu.

Additional information

Competing interests

The authors declare that they have no competing interest

Authors’ contributions

XZ and WRX were responsible for the conception and design of the manuscript. All authors participated in the drafting of the manuscript and approved its final version. All authors read and approved the final manuscript.

Rights and permissions

This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly credited. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhang, X., Yuan, X., Shi, H. et al. Exosomes in cancer: small particle, big player. J Hematol Oncol 8, 83 (2015). https://doi.org/10.1186/s13045-015-0181-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13045-015-0181-x

Keyword