ARTICLE Journal of Circulating Biomarkers Biodistribution, Uptake and Effects Caused by Cancer-derived Extracellular Vesicles Review Article Lilite Sadovska1,2, Cristina Bajo Santos1,2, Zane Kalniņa1 and Aija Linē1* 1 Latvian Biomedical Research and Study Centre, Riga, Latvia 2 Faculty of Biology, University of Latvia, Riga, Latvia Lilite Sadovska and Cristina Bajo Santos contributed equally to this work. *Corresponding author(s) E-mail: aija@biomed.lu.lv Received 09 January 2015; Accepted 12 March 2015 DOI: 10.5772/60522 © 2015 The Author(s). Licensee InTech. This is an open access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/3.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Abstract Extracellular vesicles (EVs) have recently emerged as important mediators of intercellular communication. They are released in the extracellular space by a variety of normal and cancerous cell types and have been found in all human body fluids. Cancer-derived EVs have been shown to carry lipids, proteins, mRNAs, non-coding and structural RNAs and even extra-chromosomal DNA, which can be taken up by recipient cells and trigger diverse physiological and pathological responses. An increasing body of evidence suggests that cancer-derived EVs mediate paracrine signalling between cancer cells. This leads to the increased invasiveness, proliferation rate and chemoresistance, as well as the acquisition of the cancer stem cell phenotype. This stimulates angiogenesis and the reprogramming of normal stromal cells into cancer-promoting cell types. Furthermore, cancer-derived EVs contribute to the forma‐ tion of the pre-metastatic niche and modulation of anti- tumour immune response. However, as most of these data are obtained by in vitro studies, it is not entirely clear which of these effects are recapitulated in vivo. In the current review, we summarize studies that assess the tissue distribution, trafficking, clearance and uptake of cancer- derived EVs in vivo and discuss the impact they have, both locally and systemically. Keywords Extracellular vesicles, biodistribution, traffick‐ ing, tumour microenvironment, immunosuppression, metastatic niche 1. Introduction Extracellular vesicles (EVs) are a heterogeneous population of nanosized membrane vesicles that are released in the extracellular space by almost all normal and cancer cell types. Currently, three broad categories of EVs have been defined. These are based on the mode of biogenesis: (i) exosomes, (ii) microvesicles and (iii) apoptotic bodies [1]. Exosomes are EVs of endocytic origin, which range from 50-150 nm in diameter. These are released into the extrac‐ ellular environment by a fusion of the multivesicular bodies (MVBs) with the plasma membrane. Microvesicles (sometimes also referred to as ectosomes or microparticles) are large EVs, ranging between 100-1000 nm in diameter, which are secreted by shedding or budding from the plasma membrane [2, 3]. Recently, some cancer cells have been found to secrete very large EVs (1-10 µm) called large oncosomes. These are due to the shedding of non-apoptotic plasma membrane blebs, which are characteristic of fast- migrating “aboeboid” tumour cells [4]. Currently, there is 1J Circ Biomark, 2015, 4:2 | doi: 10.5772/60522 no consensus about whether these EVs represent a subclass of microvesicles or an entirely new class of EVs. Apoptotic bodies are heterogeneous EVs, which contain cytoplasm with condensed organelles and/or nuclear fragments. They are released into the surrounding extracellular space by apoptotic/dying cells [1]. In vivo, apoptotic bodies are quickly cleared by macrophages and other phagocytes [5]. Although they have been found to carry miRNAs, which can be functionally active in the recipient cells [6], they are structurally and functionally very different from live cell- derived EVs and will not be discussed in this review. Although exosomes, microvesicles and oncosomes have distinct physical and biochemical properties, so far, no markers that can unambiguously distinguish these types of EVs have been identified. Furthermore, the current methods used for the fractionation of EVs cannot reliably separate various types of EVs [3]. Furthermore, recent studies have uncovered substantial differences in the EV biogenesis of various cell types. This suggests that discrim‐ inating these types of EVs could be more complex than initially thought [3, 7]. Therefore, in the current review, we will use the term EV to designate all types of live cell- secreted vesicles. Cancer-derived EVs have been shown to carry a variety of lipids, proteins, mRNAs, non-coding and structural RNAs and even extra-chromosomal DNA [8-10]. The molecular content of EVs partially reflects that of the parent cells. However, studies have shown that they are enriched in certain molecules, indicating the existence of specific mechanisms that sort cargo into EVs [11, 12]. Overall, these mechanisms are poorly understood and are likely to be related to the mode of EV biogenesis. The sorting of specific proteins into EVs can be mediated by the endosomal sorting complex for transport (ESCRT) machinery [13] or ESCRT-independent mechanisms such as tetraspanin [14] or ceramide-dependent pathways [15]. The sorting of RNAs into EVs can be mediated by the interaction of specific RNA-binding proteins, such as hnRNPA2B1, with cis-acting elements in the RNA sequence [12, 16]. EVs can be taken up by recipient cells and trigger diverse biological effects. Therefore, they have emerged as impor‐ tant mediators of intercellular communication, both in normal physiological processes and in the development of various diseases [8, 9, 17]. In cancer, EVs have been shown to mediate paracrine signalling between cancer cells, cross- talk between tumour and microenvironment, contribute to the formation of the pre-metastatic niche and interfere with the anti-tumour immune response. However, most of the data regarding their role in cancer come from in vitro studies and it is not entirely clear which of the uptake mechanisms are recapitulated in vivo. Most data are concerned with the fate of EVs when they are internalized by various cell types, as well as what mechanisms govern the trafficking of EVs in the body. In this review, we summarize studies that aim to assess the tissue distribu‐ tion, trafficking, clearance and uptake of cancer-derived EVs in vivo. We also discuss the impact that they have been shown to have, both locally and systemically. 2. Tissue Distribution of Cancer-derived EVs EVs have been found in various biological fluids, including blood, milk, urine, saliva, etc. [18]. Here, they represent a heterogeneous mixture of EVs derived from various cell types. Several lines of evidence suggest that cancer-derived EVs can be released into the circulation or other biofluids of cancer patients. At first, cancer patients have been found to have higher levels of circulating EVs, compared to healthy controls [19-21]. Secondly, EVs isolated from the biofluids of cancer patients or tumour-bearing animals were shown to contain cancer-associated markers, such as Melan-A [19], TYRP2 [22] and CA19-9 [23], and amplified or mutated oncogenes [9, 24]. However, until recently, very little was known about the half-life, clearance, trafficking and tissue distribution of cancer-derived EVs in the body. Data from in vivo studies that address these issues have only started to accumulate over the last few years. The main findings of these studies are summarized in Table 1. In these studies, two different approaches for studying the biodistribution of EVs have been exploited. One is based on administering exogenous EVs into the circula‐ tion of experimental animals, while the other is based on tumour models that produce labelled EVs endogenous‐ ly. Most of the studies that used exogenously produced EVs isolated them from a cell culture medium by differential ultracentrifugation and sucrose gradient, as described by Thery et al. 2006 [25]. After the intrave‐ nous administration has been conducted, the EVs are tracked in vivo. This is carried out either by labelling the EVs with fluorescent membrane dyes, such as DiI, PKH67 and PKH26 [22, 26-32], and loading them with superpar‐ amagnetic iron nanoparticles (SPION5), allowing magnet‐ ic resonance tracking [33]; or by using EVs engineered to display a membrane reporter [27, 30, 34]. EV reporter systems have been created by the genetic engineering of cell lines that produce EVs with membrane- anchored Gaussia luciferase (gLuc), biotin acceptor peptide (BAP) [27] or green fluorescent protein (GFP) [34]. Studies that are based on endogenously produced EVs have used either genetically engineered cancer cell lines, which produced GFP-tagged CD63 [34, 35], or human cancer xenografts in mice, where cancer-derived EVs were located by detecting human CD63 [32]. The EV detection methods vary depending on the method of EV labelling and the specific aim of each study. In vivo imaging system (IVIS) is the method of choice for the analysis of EV tissue distribu‐ tion. However, studies that focus on the specific effects caused by cancer-derived EVs in specific organs also used flow cytometry, microscopy, immunohistochemistry, gLuc activity measurements, magnetic resonance, etc. 2 J Circ Biomark, 2015, 4:2 | doi: 10.5772/60522 Cell Line Animal Line EV Labelling Injection Site Detection Method Results Ref. Exogenously Administered EVs into Cancer-free Animals TS/A murine mammary tumour BALB/c PKH67 i.v. Flow cytometry EVs are taken up by bone marrow CD11b +Gr-1+ cells; suppress myeloid cell differentiation into DCs. [26] EL-4 mouse lymphoma C57BL/6 IRDye800 i.p. LI-COR imager EVs detected in the liver, lung, kidney and spleen, taken up by CD11b+Gr-1+ cells. [28] B16-BL6 mouse melanoma C57BL/6 gLuc-lactadherin i.v. via tail vein gLuc activity measurement Half-life of EVs in the blood is ~2 minutes At 10 to 60 minutes after injection, EVs are distributed mainly to the liver and lungs; at 4 hours – lungs and spleen. [30] BALB/c gLuc- lactadherin i.v. via tail vein LAS3000 IVIS C57BL/6 PKH26 i.v. via tail vein Fluorescent microscopy HEK293T human embryonic kidney Athymic nude mice gLuc i.v. via retro-orbital vein Bioluminescence imaging 30 minutes after injection, EVs are distributed to the spleen, liver, lungs and kidneys; actively taken up by liver and lung cells but not spleen cells. EVs are eliminated via hepatic and renal routes. [27] Biotin-Alexa680- streptavidin i.v. via tail vein FMT imaging B16-F10 mouse melanoma C57BL/6 SPION5 Footpad In vivo MRI EVs home to the subcapsular sinus of lymph nodes. [33] MDA-MB-231 human breast cancer Nude mice DiI i.v. via tail vein Flow cytometry, IF EVs are internalized by macrophages in the lungs and brain, resulting in the activation of NF-κB pathway. [32] K562 human chronic myelogenous leukaemia SD rats n.a. i.v. via tail vein n.a. EVs deliver hybrid BCR/ABL DNA to normal neutrophils; administration of EVs induce CML phenotype in mice and rats. [36] NOD/SCID mice i.v. via tail vein n.a. Exogenously Administered EVs into Cancer Bearing Animals BSp73ASML BDX rat pancreatic adenocarcinoma BDX rats n.a. Footpad – first EVs, then cells n.a. EVs from metastatic cells support the metastatic spread of non-metastatic cells to lymph nodes and lungs. [37] B16-F10 mouse melanoma C57BL/6 albino; i.v. injection of melanoma cells DiR Footpad IVIS EVs home to sentinel lymph nodes and enhance migration of melanoma cells to EV-rich sites in lymph nodes. [29] B16-F10 and B16-F1 mouse melanoma C57BL/6 mice with orthotopic B16-F10 tumours PKH67 i.v. Confocal microscopy EVs home to the lungs, bone marrow, liver and spleen. EVs enhance metastasis by bone marrow education via the transfer of MET to the bone marrow progenitors. [22] HEK293T human embryonic kidney RAG2-/- mice with breast cancer (HCC70) xenografts DiR i.v. via tail vein IVIS EGFR-targeted EVs home to tumour microenvironment and can deliver miRNAs to EGFR-expressing breast cancer cells. [31] HEK293T human embryonic kidney Athymic nude mice with glioma xenograft gLuc i.v. via tail vein Bioluminescence imaging Similar amounts of EVs are found in tumours, spleen and liver. [27] 4T1 mouse mammary tumour BALB/c with 4T1 orthotopic tumour DiR i.v., i.t. IVIS200 EVs are taken up in the liver and spleen, very little amounts travel to the tumour. Slower uptake and clearance in mice with impaired innate immunity. Intratumourally administered EVs stay associated with tumour. [38] Nude mice with 4T1 orthotopic tumour NOD.CB17- Prkdcscid/J with 4T1 orthotopic tumour Endogenously Produced EVs from Genetically Engineered Cancer Cell Lines MMT-060562 mouse breast cancer; MDA- MB-231 human breast cancer Nude mice with orthotopic MMT tumours or MDA- MB-231 xenografts CD63-GFP n.a. CLSM imaging Breast cancer cells secrete EVs in the primary and metastatic tumour microenvironment and blood circulation; EVs are taken up by cancer cells and CAFs. [34] 3Lilite Sadovska, Cristina Bajo Santos, Zane Kalniņa and Aija Linē: Biodistribution, Uptake and Effects Caused by Cancer-derived Extracellular Vesicles Cell Line Animal Line EV Labelling Injection Site Detection Method Results Ref. MDA-MB-231 human breast cancer Nude mice with MDA-MB-231 xenograft Human CD63 n.a. IHC EVs are taken up by macrophages in the lung, brain and lymph nodes; induce inflammatory processes in tumour microenvironment and axillary lymph nodes.* [32] H460 human lung cancer Nude mice with H460 xenograft hCD63-GFP n.a. Immunomagnetic separation, RT-PCR Human cancer-derived EVs carry mRNAs and are detectable in the blood and saliva. [35] gLuc – Gaussia luciferase; FMT – fluorescence mediated tomography; i.v. – intravenous; i.p. – intraperitoneal; EV – extracellular vesicle; DC – dendritic cell; BM – bone marrow; MDMC – monocyte derived myeloid cell; IVIS – In Vivo imaging system; SPION5 – super paramagnetic iron oxide nanoparticles; MRI – magnetic resonance imaging; PLN – Poplietal lymph node; IF – immunofluorescence; CLSM – confocal laser scanning microscopy; IHC – immunohistochemistry. Table 1. Studies investigating EV biodistribution and functions in vivo In cancer-free animals, exogenously administered cancer- derived EVs were distributed mainly to the liver, lungs, kidneys and spleen [28, 30] and were also detected in the lymph nodes [29, 33] and bone marrow [22, 26]. EVs derived from human embryonic kidney cells (HEK293T) were predominantly localized in the spleen, followed by the liver, lungs and kidneys. Additionally, lower amounts of EVs were also detected in the brain, heart and muscle [27]. However, when the EV-injected animals were trans‐ cardially perfused with PBS before collecting the organs, the highest amount of EVs was detected in the kidneys and not the spleen, followed by the liver and lungs. This suggests that EVs are actively taken up by the kidney, liver and lung cells but not the spleen cells. It also indicates that the accumulation of EVs in the nonperfused spleen may be due to the uptake of EVs by circulating lymphocytes and macrophages [27]. In the liver and lungs, they are likely to be taken up and degraded by phagocytic cells such as Kuppfer cells and alveolar macrophages [27]. However, a portion of the EVs may also be internalized by the kidney cells and released into the urine [27]. A recent study by Cai et al. (2014) demonstrated that, when injected in rats eliciting some characteristics of CML, EVs derived from the CML cell line K562 transferred BCR-ABL hybrid gene to normal neutrophils. This suggests that EV-mediated transfer of oncogenes may represent a novel mechanism of tumourigenesis [36]. Considering that cancer patients have substantially higher levels of EVs in the blood than healthy individuals [19-21] and that cell-free RNAs, part of which are likely to be packaged into EVs, are remarkably stable in the patients’ blood [39], it seemed plausible that EVs should be very stable in the biofluids. Unexpectedly, when injected in the blood circulation of immunocompetent mice, murine melanoma-derived EVs had a half-life of only approxi‐ mately two minutes. Furthermore, they were cleared from the circulation within four hours [30]. Similarly, when injected into athymic nude mice [27], HEK293T-derived EVs had a half-life of less than 30 minutes in vivo in most tissues. Moreover, lymphoma-derived EVs were taken up by CD11b+Gr-1+ cells within one hour after the injection into immunocompetent mice [28]. Such a short half-life of exogenously administrated EVs was very surprising and has to be taken into account when designing studies for the identification of EV-associated cancer biomarkers. In tumour-bearing animals, both the exogenously admin‐ istered EVs and endogenously produced EVs have also been found to accommodate the tumour microenviron‐ ment, lymph nodes and bone marrow. However, in such studies, the largest part of intravenously administered EVs was rapidly cleared from the circulation. Exogenously administered HEK293T-derived EVs accumulated in xenograft tumours at similar levels than the liver and spleen at 60 minutes post-injection [27]. Nevertheless, it remained unclear which cell types bind or internalize the EVs. Several other studies suggest that, in the tumour microenvironment, cancer-derived EVs can be internalized by other cancer cells, as well as by surrounding cells like cancer-associated fibroblasts (CAFs), endothelial cells and other stroma cells [31, 34, 40]. A number of studies have demonstrated that cancer-derived EVs are released into the blood circulation and are trafficked to the lymph nodes, bone marrow and lungs. Here, they promote metastatic niche formation and enhance the metastatic spread of cancer cells to these organs [22, 29, 34, 37]. Furthermore, when taken up by macrophages, cancer-derived EVs were shown to induce an inflammatory response [32]. Taken together, these studies provide a solid basis for the concept that cancer-derived EVs promote cancer development and progression in vivo by inducing various biological effects, both locally and systemically. 3. Uptake of EVs in Tumour Microenvironment It has become clear that, within the tumour microenviron‐ ment, the cellular composition is complex. Furthermore, relationships between different cell types are no less sophisticated than those in any healthy organs [41]. Therefore, the role of intercellular communication in the acquisition of various cancer phenotypes, invasive growth and metastasis and drug resistance is increasing‐ ly recognized. It can be mediated by soluble signalling molecules, cell-cell or cell-matrix adhesion, gap junc‐ 4 J Circ Biomark, 2015, 4:2 | doi: 10.5772/60522 tions and EVs [42]. In the tumour microenvironment, cancer-derived EVs have been shown to be taken up by other cancer cells and stromal cells, such as fibroblasts, as well as endothelial cells and tumour-infiltrated immune cells. EVs can exert their effects in the recipient cells either by binding to the cell surface receptors or delivering their content inside the recipient cell. Contra‐ ry to the single-molecule signals, EVs have the potential to affect multiple signalling pathways inside the recipi‐ ent cell. Hence, they provide more efficient means for phenotypic reprogramming or synchronizing the physiological state of the surrounding cells [43, 44]. Internalization can occur either through a fusion with the cell membrane [45], endocytosis and micropinocytosis [46, 47] or phagocytosis [48]. However, it is not yet entirely clear which of these mechanisms lead to the degradation of EV components and which, eventually, result in the release of EV content into the cytoplasm. Here, it can alter the physiological functions of the recipient cell. The recipient cell specificity is likely to be determined by the composition of adhesion molecules and lipid content on the surface of EVs and the respec‐ tive ligands on the cellular surface [49-53]. For example, the internalization of glioblastoma-derived EVs was found to depend on the expression of heparan sulfate proteoglycans on the recipient cells [50]. Meanwhile, the uptake of rat tumour-derived EVs by specific cell types depended on the expression of tetraspanin Tspan8 and integrin α4 on the EVs [54]. Interestingly, low extracellu‐ lar pH (pH 6.0) has also been shown to increase the release and uptake of melanoma-derived EVs [45]. This finding is particularly significant for understanding the EV function in the tumour microenvironment, as hypoxia and extracellular acidosis are common features of the vast majority of solid cancers. Extracellular acidosis arises by switching metabolism to glycolysis, resulting in the increased production and excretion of acidic metabo‐ lites, such as lactic and carbonic acids, and can be as low as 5.9 [55]. Recently, low pH-dependent EV-mediated elimination of cisplatin was also shown to serve as a mechanism of chemoresistance to cisplatin [56]. 4. Paracrine Effects Caused by Cancer-derived EVs in the Tumour Microenvironment 4.1 Cancer Cell Cross-talk The uptake of cancer-derived EVs by other cancer cells can lead to increased invasiveness and metastatic potential [57-60], anchorage-independent growth [24, 61], prolifera‐ tion and chemoresistance [60, 62, 63], as well as inducing the epithelial-mesenchymal transition (EMT) [59, 64] (Figure 1). Moreover, cancer-derived EVs are shown to drive the oncogenic conversion of non-tumourigenic cells [65, 66]. These effects can be mediated by the transfer of functionally active proteins, such as HIF1α [59], EGFRvIII [67], miRNAs [61, 63] and possibly, other non-coding RNAs, mRNAs and fragments of genomic DNA carrying various cancer genes and transposable elements [9, 68, 69]. In 1980, Poste and Nicolson provided the first experimental evidence that EVs, shed from highly metastatic melanoma cells, could increase the metastatic potential of poorly metastatic melanoma cells [57]. Later on, several independ‐ ent studies confirmed these findings in vitro and in vivo. A study by Hao et al. (2006) demonstrated that, when i.v. was injected into C57BL/6 mice, the EVs released from highly metastatic melanoma cell line were taken up by poorly metastatic cells, which acquired the capacity to form metastatic colonies in the lungs. Similarly, EVs derived from highly invasive (but not from non-invasive) triple- negative breast cancer cells significantly increased the proliferation, migration and invasion capacity of other breast cancer cell lines [60]. In line with this, EVs from prostate cancer (PC) patients’ sera have been found to enhance the proliferation and invasion of PC cell lines [62]. A study by Aga et al. (2014) demonstrated that EVs derived from EBV-infected nasopharyngeal carcinoma contained the functionally active transcription factor, HIF1α. Further‐ more, the uptake of HIF1α-containing EVs (but not EVs containing mutant HIF1α) resulted in the downregulation of E-cadherin and upregulation of N-cadherin in recipient cells [59]. Changes in E- and N-cadherin expression are markers for EMT that confer mesenchymal properties to epithelial cells. This is associated with invasion and metastasis, as well as the acquisition of cancer stem cell phenotype [70]. Furthermore, EVs from drug-resistant cell line variants have been shown to confer resistance to non- resistant PC or breast cancer cells [62, 63]. Al-Nedawi et al. (2008) demonstrated that glioma cells with a mutated form of epidermal growth factor receptor (EGFRvIII) release it via EVs, which were taken up by indolent glioma cells [24]. This resulted in the activation of MAPK and Akt signalling pathways. It also led to changes in the expression of EGFRvIII-regulated genes, resulting in the morphological transformation and increase in anchor‐ age-independent growth capacity [24]. Furthermore, breast and colorectal cancer (CRC) cells have been shown to release Amphiregulin (AREG), an EGFR ligand, via EVs [71]. When taken up by breast cancer cells, EV-packaged AREG displayed greater membrane stability and increased invasiveness. Interestingly, the AREG level in EVs corre‐ lated with the mutant KRAS status of the donor cells [71]. Later on, the same group demonstrated that mutant KRAS status affects the composition of the EV proteome. When taken up by CRC cells with wt KRAS, EVs derived from CRC cells with mutated KRAS contained many tumour- promoting proteins, including KRAS and EGFR, and enhanced colony formation [72]. Another study demon‐ strated that EVs derived from a hepatocellular carcinoma (HCC) were enriched in specific miRNAs that could be taken up by other HCC cells. This resulted in the modula‐ tion of TAK1 signalling pathway and enhanced anchorage- independent growth [61]. 5Lilite Sadovska, Cristina Bajo Santos, Zane Kalniņa and Aija Linē: Biodistribution, Uptake and Effects Caused by Cancer-derived Extracellular Vesicles A recent study by Melo et al. (2014) highlights a novel mechanism that could potentially impact our understand‐ ing of the physiological role of cancer-derived EVs. This study demonstrated that, in contrast to those produced by normal cells, breast cancer EVs bear a RISC-loading complex. This is associated with pre-miRNAs that are capable of cell-independent miRNA biogenesis. These EVs were able to mediate the rapid and efficient silencing of target mRNAs of recipient cells and, importantly, were shown to have a protumourigenic effect on non-tumouri‐ genic epithelial cells. It was demonstrated that the impact was dependent on the presence of Dicer within cancer EVs [65]. The authors proposed that cancer EVs are capable of inducing an oncogenic “field effect” by subjugating neighbouring normal cells to cooperate in cancer progres‐ sion. In addition, another excellent study by Abd Elmageed et al. (2014) showed, for the first time, that EVs produced by prostate cancer cells are capable of inducing a neoplastic transformation of tumour-trophic mesenchymal stem cells. The EV-primed adipose-derived tissue stem cells under‐ went a mesenchymal-to-epithelial transition, adopted genetic instability and oncogenic transformation, and were able to form tumours in vivo. This was achieved through the oncogenic factor transfer by prostate cancer cell- derived EVs, which included oncogenic miRNAs, K-ras and H-ras transcripts and oncoproteins [66]. Figure 1. Local and systemic effects that are triggered by cancer-derived EVs. Locally cancer-derived EVs have been reported to promote proliferation, invasiveness and chemoresistance, and to induce EMT in cancer cells in a paracrine manner, and to stimulate angiogenesis and reprogramming of stromal cells into CAFs. Systemically, cancer-derived EVs have been shown to contribute to the generation of metastatic microenvironment by reprogramming BMDCs, regulating gene expression in the lungs and lymph nodes and modulating anti-tumour immune response. BMDCs, bone marrow-derived cells; ECM, extracellular matrix; EMT, epithelial-mesenchymal transition; CAF, cancer-associated fibroblast; DC, dendritic cells; MDSC, myeloid-derived suppressor cell; NK, natural killer cell; TCR, T cell receptor. However, several recent findings have challenged the paradigm of the pro-tumourigenic role of cancer-derived EVs, suggesting that cancer-derived EVs can also have opposite or, so far, unknown roles. For example, a recent 6 J Circ Biomark, 2015, 4:2 | doi: 10.5772/60522 study by Gabriel et al. (2013) demonstrated that EVs derived from various cancer cell lines and PC patients’ plasma contained a functionally active tumour-suppressor protein PTEN that suppressed the proliferation of PTEN- deficient recipient cells [107]. As the authors did not detect it in the EVs derived from normal cells and plasma from healthy individuals, they suggested that this might represent a mechanism used by cancer cells to downregu‐ late PTEN level [107]. Meanwhile, another study found a functional PTEN protein in EVs, produced by mouse embryonic fibroblasts and human embryonic kidney cells [108]. This suggests that this could be a more wide spread phenomenon, provoking questions regarding its biological significance in normal physiological processes and cancer. Moreover, it remains to be determined which cell types actively take up PTEN-containing EVs in vivo and whether they exert the same physiological effects in cancerous and normal cells. Taken together, several lines of evidence strongly support the concept that cancer-derived EVs act paracrinally to synchronize the physiological state in subpopulations of cells. They do this by delivering signalling molecules that are not endogenously expressed in the recipient cells and thus, driving the cancer progression. Furthermore, they may even induce the acquisition of cancer cell phenotype in non-malignant cells. However, various EV subpopula‐ tions differ in their molecular content and may cause opposite effects in the recipient cells. Thus, further studies that dissect the heterogeneity of EVs and the recipient cell selectivity are urgently needed. 4.2 Promotion of Angiogenesis Cancer-derived EVs have also been shown to promote angiogenesis. A number of independent studies have demonstrated that cancer-derived EVs can be taken up by the endothelial cells in vitro and in vivo. This results in morphological changes, migration and proliferation of endothelial cells, tube formation and neovascularization [40, 67, 73-78]. Apparently, these effects can be mediated by the transfer of angiogenic proteins [73] and oncogenic proteins [67], as well as various mRNAs and miRNAs [40, 78, 79]. While the angiogenic proteins, such as angiogenin, IL-6, IL-8, TIMP-1 and VEGF, are likely to impact in a paracrine manner, the delivery of oncogenic EGFR to the endothelial cells was shown to trigger the endogenous expression of VEGF, followed by the autocrine activation of VEGF receptor-2 signalling [67]. Several studies empha‐ size the role of EV-shuttled miRNAs in the endothelial cell migration and neovascularization. A study by Umezu et al. (2012) demonstrated that the uptake of leukaemia cell- derived EVs carrying miR-92a enhanced endothelial cell migration and tube formation [79]. Meanwhile, Zhuang et al. (2012) showed that tumour-secreted miR-9 triggered the activation of JAK-STAT pathway in the endothelial cells, resulting in enhanced migration and tumour angiogenesis [78]. Furthermore, the administration of anti-miR-9 or JAK inhibitors suppressed these effects in vitro and in vivo [78]. Likewise, the treatment of mice carrying human carcinoma xenografts with Diannexin, a drug that binds phosphati‐ dilserine and blocks EV exchange, resulted in the reduction of tumour growth rate and microvascular density [67]. Collectively, these studies demonstrated, both in vitro and in vivo, that cancer-derived EVs have a pro-angiogenic capacity and therefore, might represent an attractive target for therapeutic intervention. However, to date, it is not entirely clear at what stages of maturation endothe‐ lial cells are targeted by cancer-derived EVs in vivo. 4.3 Acquisition of CAF Phenotype Among the different components of the tumour stroma, cancer-associated fibroblasts (CAFs) are one of the main elements. CAFs are defined as all the fibroblastic, non- neoplastic, non-vascular, non-epithelial and non-inflam‐ matory cells with a stable karyotype found in a tumour [80]. CAFs promote tumour progression by secreting soluble growth factors, cytokines and chemokines that stimulate proliferation and migration of cancer cells, induce angio‐ genesis, modify tumour metabolism, stimulate acquisition of cancer stem cell phenotype and modulate the immune response [80, 81]. CAFs can originate from resident tissue fibroblasts, mesenchymal stem cells, myofibroblasts and even epithelial and endothelial cells via EMT or EndMT, respectively [80]. This process is accompanied by persistent changes in their gene methylation pattern [82, 83]. Once the CAF phenotype is acquired, two autocrine signalling loops, mediated by TGF-β and SDF-1 cytokines, maintain them in this differentiation state in an autocrine manner [84]. A growing body of evidence suggests that cancer-derived EVs play a crucial role in the reprogramming of these cells into CAFs via the transfer of TGFβ. Thus, for instance, TGFβ1 containing PC-derived EVs was found to be taken up by primary lung fibroblasts, resulting in their differen‐ tiation into tumour-promoting CAFs [85, 86]. These cells supported angiogenesis in vitro and tumour growth in vivo. Interestingly, this effect could not be achieved by using soluble TGFβ1 and appeared to depend on heparan sulphate chains on the EV surface. Moreover, EV-deficient (Rab27a knock-down) cancer cells failed to achieve activa‐ tion of the tumour stroma [86]. Likewise, gastric cancer- derived EVs were shown to trigger the differentiation of umbilical cord-derived mesenchymal stem cells into CAFs by the transfer of TGFβ and the subsequent activation of TGFβ/Smad pathway [87]. Furthermore, breast cancer- derived EVs were found to convert adipose tissue-derived mesenchymal stem cells into CAFs expressing various tumour-promoting factors [88]. Hence, cancer-derived EVs seem to play a crucial role in the induction of CAF phenotype via the transfer of TGFβ. 7Lilite Sadovska, Cristina Bajo Santos, Zane Kalniņa and Aija Linē: Biodistribution, Uptake and Effects Caused by Cancer-derived Extracellular Vesicles 5. Systemic Effects Caused by Cancer-derived EVs 5.1 Formation of Pre-metastatic Niche The first evidence that cancer-derived EVs can contribute to the generation of metastatic microenvironment was provided by Jung et al. (2009) [37]. In this study, rats received injections of a conditioned medium containing EVs and a soluble matrix obtained from highly metastatic pancreatic cancer cells, followed by the injection of the respective cancer cells. This showed that the conditioned medium promoted the settlement of a non-invasive variant of these cells in the lymph nodes and lungs. This suggested that highly metastatic cells deliver messages that elicit alterations in pre-metastatic organs. This allows the homing, settling and growing of poorly metastatic cells [37]. Another study demonstrated that, when injected in the mouse footpad, mouse melanoma-derived EVs, home to sentinel lymph nodes, enhance the migration of melanoma cells to the EV-rich sites in the lymph nodes. In the lymph nodes, EVs were found to regulate the expression of a variety of genes involved in migration, extracellular matrix deposition and vascular proliferation [29]. Moreover, EVs derived from putative renal cancer stem cells were found to increase the number of lung metastases, when intrave‐ nously injected in SCID mice [74]. An elegant study by Peinado et al. (2012) demonstrated that, when injected in mice, EVs derived from highly malignant mouse melano‐ ma, home to the lungs and bone marrow, enhance endo‐ thelial permeability at pre-metastatic sites and promote the development of distant metastasis. These EVs were found to “educate” bone marrow-derived cells (BMDCs) by transferring the MET oncoprotein, resulting in the activa‐ tion of the MET pathway in BMDCs. They become condi‐ tioned to support tumour vasculogenesis, invasion and metastasis. Furthermore, higher amounts of MET were found in circulating EVs isolated from patients with stage three and four melanoma than in healthy controls. This shows that this finding may also have relevance in a clinical setting [22]. A number of preclinical studies suggest that cancer- derived EVs have a systemic effect on the conditioning of a pre-metastatic niche and hence, set the basis for a new therapeutic strategy. However, it remains to be determined whether or not this type of signalling represents a common mechanism of metastasis in various human cancers. 5.2 Modulation of Anti-tumour Immune Response Cancer-derived EVs have been reported to both stimulate and suppress anti-tumour immune responses [11]. They are shown to contain tumour-associated antigens, such as CEA and MART1, and are efficiently taken up by dendritic cells. In turn, these cross-present the antigens to CD8+ T cells, resulting in potent anti-tumour effects [89, 90]. Moreover, as they bear MHC class I molecules, it has been suggested that they could directly stimulate CD8+ T cells [11]. In fact, several pre-clinical and clinical studies based on the immunization with cancer-derived EVs, in combi‐ nation with various cytokines, have shown the induction of beneficial tumour-specific CD8+ T cell responses. Such studies suggest that they may represent an attractive approach for cancer immunotherapy [91-94]. On the other hand, increasing evidence suggests that cancer-derived EVs can suppress the anti-tumour immune response in a variety of ways. For instance, melanoma- derived EVs have been shown to be enriched for FasL and induced Fas-mediated apoptosis in T cells [95]. Subsequent studies have described similar immune evasion mecha‐ nisms, mediated by FasL or TRAIL expression, on EVs in prostate and colorectal cancer and glioma [96-99]. More‐ over, FasL expression can also lead to the TCR impairment due to the downregulation of CD3-ζ chain, which has been reported in ovarian [100, 101] and head and neck squamous carcinoma patients [98]. Contrary to the effector T cells, CD4+CD25highTregs are resistant to FasL-induced apoptosis and cancer-derived EVs have been reported to stimulate the expansion and suppressive functions of Tregs [98, 102, 103]. Ovarian cancer-derived EVs have been shown to promote the proliferation of CD4+CD25+FOXP3+T cells, convert CD4+CD25neg T cells into CD4+CD25+Tregs and upregulate Treg suppressor functions (e.g., production of perforin, Granzyme B, IL-10, etc.), when added to the culture of peripheral blood T cells obtained from healthy donors [102]. At least partially, this effect seems to be mediated by EV-transferred TGFβ1. This is because the pre- treatment of malignant-effusion derived EVs, with neutral‐ izing antibodies against TGFβ1, reduced the expansion and suppressive functions of Tregs [104]. However, another recent study demonstrated that CD4+CD25highTreg expan‐ sion and IL-10 secretion is promoted by cancer cell-secreted miR-214 that targets PTEN in CD4+ T cells [105]. Other EV-mediated immunosuppressive mechanisms include T cell inhibition via the production of extracellular adenosine by EV-expressed CD39 and CD73 [106], and the downregulation of the activating receptor NKG2D on NK and CD8+ T cells by EV-transferred TGFβ [103, 107]. Cancer-derived EVs have also been shown to suppress the cytotoxic activity of NK cells by expressing MICA, which triggers the downregulation of NKG2D from the cell surface and reduces NK cytotoxicity [108]. In addition, cancer-derived EVs have multiple effects on myeloid precursors, dendritic cells and macrophages. Cancer-derived EVs have been shown to block the differ‐ entiation of myeloid precursors into dendritic cells and promote the generation of myeloid-derived suppressor cells via Stat3 activation [26, 109, 110]. In turn, this results in the suppression of effector T cell proliferation, activation and cytolytic functions and the induction of Treg cells [109, 111]. In macrophages, melanoma and breast cancer- derived EVs (but not those from non-cancerous cells) have been reported to activate NF-κB signalling and to alter the cytokine and chemokine profile, favouring the production 8 J Circ Biomark, 2015, 4:2 | doi: 10.5772/60522 of pro-inflammatory cytokines. However these changes were complex and not consistent with M1 or M2 polariza‐ tion [32, 112]. Hence, the role of cancer-derived EVs in the macrophage-mediated tumour-promoting or anti-tumour effects is not entirely clear and, presumably, may vary depending on their content and physiological state of their cell-of-origin. 6. Concluding Remarks and Future Directions Collectively, these studies strongly support the para‐ digm of the cancer-promoting role of cancer-derived EVs. They suggest that inhibition of the formation or uptake of cancer-derived EVs or their components could be a novel therapeutic avenue. In fact, several studies have demonstrated that blocking the production or uptake of EVs or specific miRNAs carried by EVs reduced tu‐ mour growth and angiogenesis. This clearly shows a therapeutic benefit [67, 78, 113]. In addition, natural or genetically engineered EVs can be exploited as tools for delivery of virus-like particles or other gene therapy products, allowing to evade pre-existing neutralizing antibodies against the viral vectors and to increase transduction efficiency [114, 115]. Moreover, cancer-derived EVs seem to have very diverse effects on immune cells, which may lead to the stimulation or suppression of anti-tumour immune responses. Hence, a deeper understanding of mechanisms and how they impact the functions of various immune cell subsets could help to develop novel strategies for shifting the balance towards immunostimulatory tumour microenvironment. However, it remains unclear whether it is possible to entirely stop cancer progression by inhibiting the forma‐ tion or uptake of cancer-derived EVs. It seems likely that cancers differ in their ability to produce EVs and in the degree to which they depend on the EV-mediated signal‐ ling. However, to the best of our knowledge, the levels of EVs and their effects have not been systematically studied during the course of disease progression and compared among different cancer types. Another layer of complexity is added by the heterogeneity of EV biogenesis and the composition of their molecular cargo. Currently, there is great controversy regarding what types of EVs each cell type produces and which of them carry molecular cargo that are capable of eliciting biologically significant effects. For instance, many studies have reported that exosomes are enriched in miRNAs, suggesting that they function as vehicles for the intercellular transfer of miRNAs [8, 116-120]. Nonetheless, a recent study by Chevillet et al. (2014) has challenged this view by demonstrating that, on average, most exosomes harbour less than one molecule of a given miRNA. However, it remains unclear whether rare exosomes in the population carry many copies of a given miRNA or whether a larger fraction of exosomes carries a low concentration of miRNAs [121]. In this regard, a recent study by Thakur et al. (2014) demonstrated that exosomes carry dsDNA representing the whole genomic DNA. However, only a subset (~10%) of exosomes contained DNA [122]. Hence, the characterization of EV subpopula‐ tions carrying cancer-derived molecular cargo seems to be of paramount importance for designing studies aimed at the discovery of EV-associated biomarkers and therapeutic targeting of EVs. 7. Conflict of Interest The authors declare no conflicts of interest. 8. Acknowledgements This study was supported by the Latvian Council of Science (grant no. 625/2014). 9. References [1] Kalra H, Simpson RJ, Ji H, Aikawa E, Altevogt P, Askenase P, Bond VC, Borras FE, Breakefield X, Budnik V, Buzas E, Camussi G, Clayton A, Cocucci E, Falcon-Perez JM, Gabrielsson S, Gho YS, Gupta D, Harsha HC, Hendrix A, Hill AF, Inal JM, Jenster G, Kramer-Albers EM, Lim SK, Llorente A, Lotvall J, Marcilla A, Mincheva-Nilsson L, Nazarenko I, Nieuwland R, Nolte-'t Hoen EN, Pandey A, Patel T, Piper MG, Pluchino S, Prasad TS, Rajendran L, Raposo G, Record M, Reid GE, Sanchez-Madrid F, Schiffelers RM, Siljander P, Stensballe A, Stoorvogel W, Taylor D, Thery C, Valadi H, van Balkom BW, Vazquez J, Vidal M, Wauben MH, Yanez-Mo M, Zoeller M, Mathivanan S (2012) Vesiclepedia: A Compendium for Extracellular Vesicles with Continuous Community Annotation. PLoS.Biol. 10(12):e1001450. [2] Thery C, Ostrowski M, Segura E (2009) Membrane Vesicles as Conveyors of Immune Responses. Nat.Rev.Immunol. 9(8):581-593. [3] Colombo M, Raposo G, Thery C (2014) Biogenesis, Secretion, and Intercellular Interactions of Exo‐ somes and Other Extracellular Vesicles. Annu Rev Cell Dev Biol. 30:255-289. [4] Morello M, Minciacchi VR, de Candia P, Yang J, Posadas E, Kim H, Griffiths D, Bhowmick N, Chung LW, Gandellini P, Freeman MR, Demichelis F, Di Vizio D (2013) Large Oncosomes Mediate Intercel‐ lular Transfer of Functional microRNA. Cell Cycle. 12(22):3526-3536. [5] Weigert A, Jennewein C, Brune B (2009) The Liaison Between Apoptotic Cells and Macrophages-- The End Programs the Beginning. Biol Chem. 390(5-6): 379-390. [6] Zernecke A, Bidzhekov K, Noels H, Shagdarsuren E, Gan L, Denecke B, Hristov M, Koppel T, Jahan‐ tigh MN, Lutgens E, Wang S, Olson EN, Schober A, Weber C (2009) Delivery of MicroRNA-126 by 9Lilite Sadovska, Cristina Bajo Santos, Zane Kalniņa and Aija Linē: Biodistribution, Uptake and Effects Caused by Cancer-derived Extracellular Vesicles Apoptotic Bodies Induces CXCL12-dependent Vascular Protection. Sci.Signal. 2(100):ra81. [7] Colombo M, Moita C, van NG, Kowal J, Vigneron J, Benaroch P, Manel N, Moita LF, Thery C, Raposo G (2013) Analysis of ESCRT Functions in Exosome Biogenesis, Composition and Secretion Highlights the Heterogeneity of Extracellular Vesicles. J. Cell Sci. 126(Pt 24):5553-5565. [8] Valadi H, Ekstrom K, Bossios A, Sjostrand M, Lee JJ, Lotvall JO (2007) Exosome-mediated Transfer of mRNAs and microRNAs is a Novel Mechanism of Genetic Exchange Between Cells. Nat. Cell Biol. 9(6):654-659. [9] Balaj L, Lessard R, Dai L, Cho YJ, Pomeroy SL, Breakefield XO, Skog J (2011) Tumour Microvesi‐ cles Contain Retrotransposon Elements and Ampli‐ fied Oncogene Sequences. Nat.Commun. 2:180. [10] Ohshima K, Kanto K, Hatakeyama K, Ide T, Wakabayashi-Nakao K, Watanabe Y, Sakura N, Terashima M, Yamaguchi K, Mochizuki T (2014) Exosome-mediated Extracellular Release of Polya‐ denylate-binding Protein 1 in Human Metastatic Duodenal Cancer Cells. Proteomics. 14(20): 2297-2306. [11] Robbins PD, Morelli AE (2014) Regulation of Immune Responses by Extracellular Vesicles. Nat Rev Immunol. 14(3):195-208. [12] Villarroya-Beltri C, Baixauli F, Gutierrez-Vazquez C, Sanchez-Madrid F, Mittelbrunn M (2014) Sorting it Out: Regulation of Exosome Loading. Semin Cancer Biol. 28:3-13. [13] Hurley JH, Hanson PI (2010) Membrane Budding and Scission by the ESCRT Machinery: It's All in the Neck. Nat Rev Mol Cell Biol. 11(8):556-566. [14] Yanez-Mo M, Barreiro O, Gordon-Alonso M, Sala- Valdes M, Sanchez-Madrid F (2009) Tetraspanin- enriched Microdomains: A Functional Unit in Cell Pplasma Membranes. Trends Cell Biol. 19(9): 434-446. [15] Trajkovic K, Hsu C, Chiantia S, Rajendran L, Wenzel D, Wieland F, Schwille P, Brugger B, Simons M (2008) Ceramide Triggers Budding of Exosome Vesicles into Multivesicular Endosomes. Science. 319(5867):1244-1247. [16] Villarroya-Beltri C, Gutierrez-Vazquez C, Sanchez- Cabo F, Perez-Hernandez D, Vazquez J, Martin- Cofreces N, Martinez-Herrera DJ, Pascual-Montano A, Mittelbrunn M, Sanchez-Madrid F (2013) Sumoylated hnRNPA2B1 Controls the Sorting of Mirnas into Exosomes Through Binding to Specific Motifs. Nat Commun. 4:2980. [17] Raposo G, Stoorvogel W (2013) Extracellular Vesicles: Exosomes, Microvesicles, and Friends. J.Cell Biol. 200(4):373-383. [18] Vlassov AV, Magdaleno S, Setterquist R, Conrad R (2012) Exosomes: Current Knowledge of Their Composition, Biological Functions, and Diagnostic and Therapeutic Potentials. Biochim Biophys Acta. 1820(7):940-948. [19] Eldh M, Bagge RO, Lasser C, Svanvik J, Sjostrand M, Mattsson J, Lindner P, Choi DS, Gho YS, Lotvall J (2014) MicroRNA in Exosomes Isolated Directly from the Liver Circulation in Patients with Meta‐ static Uveal Melanoma. BMC Cancer. 14(1):962. [20] Rabinowits G, Gercel-Taylor C, Day JM, Taylor DD, Kloecker GH (2009) Exosomal microRNA: A Diagnostic Marker for Lung Cancer. Clin. Lung Cancer. 10(1):42-46. [21] Logozzi M, de MA, Lugini L, Borghi M, Calabro L, Spada M, Perdicchio M, Marino ML, Federici C, Iessi E, Brambilla D, Venturi G, Lozupone F, Santinami M, Huber V, Maio M, Rivoltini L, Fais S (2009) High Levels of Exosomes Expressing CD63 and Caveolin-1 in Plasma of Melanoma Patients. PLoS.One. 4(4):e5219. [22] Peinado H, Aleckovic M, Lavotshkin S, Matei I, Costa-Silva B, Moreno-Bueno G, Hergueta-Redon‐ do M, Williams C, Garcia-Santos G, Ghajar C, Nitadori-Hoshino A, Hoffman C, Badal K, Garcia BA, Callahan MK, Yuan J, Martins VR, Skog J, Kaplan RN, Brady MS, Wolchok JD, Chapman PB, Kang Y, Bromberg J, Lyden D (2012) Melanoma Exosomes Educate Bone Marrow Progenitor Cells Toward a Pro-metastatic Phenotype Through MET. Nat Med. 18(6):883-891. [23] Javeed N, Sagar G, Dutta SK, Smyrk T, Lau JS, Bhattacharya S, Truty MJ, Petersen GM, Kaufman RJ, Chari ST, Mukhopadhyay D (2014) Pancreatic Cancer-derived Exosomes Causes Paraneoplastic Beta-cell Dysfunction. Clin Cancer Res. [24] Al-Nedawi K, Meehan B, Micallef J, Lhotak V, May L, Guha A, Rak J (2008) Intercellular Transfer of the Oncogenic Receptor Egfrviii by Microvesicles Derived from Tumour Cells. Nat Cell Biol. 10(5): 619-624. [25] Thery C, Clayton A, Amigorena S, Raposo G (2006) Isolation and Characterisation of Exosomes from Cell Culture Supernatants and Biological Fluids. Current Protocols in Cell Biology. USA: John Wiley & Sons, Inc.; p. 3.22.21 - 23.22.29. [26] Yu S, Liu C, Su K, Wang J, Liu Y, Zhang L, Li C, Cong Y, Kimberly R, Grizzle WE, Falkson C, Zhang HG (2007) Tumor Exosomes Inhibit Differentiation of Bone Marrow Dendritic Cells. The Journal of Immunology. 178(11):6867-6875. [27] Lai CP, Mardini O, Ericsson M, Prabhakar S, Maguire CA, Chen JW, Tannous BA, Breakefield XO (2014) Dinamic Biodistribution of Extracellular Vesicles in Vivo Using a Ultimodal Imaging Report‐ er. American Chemical Society, Nano. 8(1):483-494. [28] Sun D, Zhuang X, Xiang X, Liu Y, Zhang S, Liu C, Barnes S, Grizzle W, Miller D, Zhang HG (2010) A 10 J Circ Biomark, 2015, 4:2 | doi: 10.5772/60522 Novel Nanoparticle Drug Delivery System: The Anti-Inflammatory Activity of Curcumin is En‐ hanced When Encapsulated in Exosomes. Mol Ther. 18(9):1606-1614. [29] Hood JL, San RS, Wickline SA (2011) Exosomes Released by Melanoma Cells Prepare Sentinel Lymph Nodes for Tumor Metastasis. Cancer Res. 71(11):3792-3801. [30] Takahashi Y, Nishikawa M, Shinotsuka H, Matsui Y, Ohara S, Imai T, Takakura Y (2013) Visualization and in Vivo Tracking of the Exosomes of Murine Melanoma B16-BL6 Cells in Mice After Intravenous Injection. J Biotechnol. 165(2):77-84. [31] Ohno S, Takanashi M, Sudo K, Ueda S, Ishikawa A, Matsuyama N, Fujita K, Mizutani T, Ohgi T, Ochiya T, Gotoh N, Kuroda M (2013) Systemically Injected Exosomes Targeted to EGFR Deliver Antitumor Microrna to Breast Cancer Cells. Mol Ther. 21(1): 185-191. [32] Chow A, Zhou W, Liu L, Fong MY, Champer J, Van Haute D, Chin AR, Ren X, Gugiu BG, Meng Z, Huang W, Ngo V, Kortylewski M, Wang SE (2014) Macrophage immunomodulation by Breast Cancer- derived Exosomes Requires Toll-like Receptor 2- mediated Activation of NF-kappaB. Sci Rep. 4:5750. [33] Hu L, Wickline SA, Hood JL (2014) Magnetic Resonance Imaging of Melanoma Exosomes in Lymph Nodes. Magn Reson Med. [34] Suetsugu A, Honma K, Saji S, Moriwaki H, Ochiya T, Hoffman RM (2013) Imaging Exosome Transfer from Breast Cancer Cells to Stroma at Metastatic Sites in Orthotopic Nude-Mouse Models. Adv Drug Deliv Rev. 65(3):383-390. [35] Yang J, Wei F, Schafer C, Wong DT (2014) Detection of Tumor Cell-Specific Mrna and Protein in Exo‐ some-like Microvesicles from Blood and Saliva. PLoS One. 9(11):e110641. [36] Cai J, Wu G, Tan X, Han Y, Chen C, Li C, Wang N, Zou X, Chen X, Zhou F, He D, Zhou L, Jose PA, Zeng C (2014) Transferred BCR/ABL DNA from K562 Extracellular Vesicles Causes Chronic Myeloid Leukemia in Immunodeficient Mice. PLoS One. 9(8):e105200. [37] Jung T, Castellana D, Klingbeil P, Cuesta Hernan‐ dez I, Vitacolonna M, Orlicky DJ, Roffler SR, Brodt P, Zoller M (2009) CD44v6 Dependence of Preme‐ tastatic Niche Preparation by Exosomes. Neoplasia. 11(10):1093-1105. [38] Smyth T, Kullberg M, Malik N, Smith-Jones P, Graner MW, Anchordoquy TJ (2015) Biodistribu‐ tion and Delivery Efficiency of Unmodified Tumor- Derived Exosomes. J Control Release. 199:145-155. [39] Zandberga E, Kozirovskis V, Abols A, Andrejeva D, Purkalne G, Line A (2013) Cell-free microRNAs as Diagnostic, Prognostic, and Predictive Biomarkers for Lung Cancer. Genes Chromosomes Cancer. 52(4):356-369. [40] Hong BS, Cho JH, Kim H, Choi EJ, Rho S, Kim J, Kim JH, Choi DS, Kim YK, Hwang D, Gho YS (2009) Colorectal Cancer Cell-derived Microvesicles are Enriched in Cell Cycle-related Mrnas that Promote Proliferation of Endothelial Cells. BMC Genomics. 10:556. [41] Hanahan D* WR, * (2011) Hallmarks of Cancer: The Next Generation. Cell. 144:29. [42] Brandner JM, Haass NK (2013) Melanoma's Con‐ nections to the Tumour Microenvironment. Pathol‐ ogy. 45(5):443-452. [43] Lotvall J, Valadi H (2007) Cell to Cell Signalling via Exosomes Through esRNA. Cell Adh Migr. 1(3): 156-158. [44] Rak J (2013) Extracellular Vesicles - Biomarkers and Effectors of the Cellular Interactome in Cancer. Front Pharmacol. 4:21. [45] Parolini I, Federici C, Raggi C, Lugini L, Palleschi S, De Milito A, Coscia C, Iessi E, Logozzi M, Molinari A, Colone M, Tatti M, Sargiacomo M, Fais S (2009) Microenvironmental pH is a Key Factor for Exo‐ some Traffic in Tumor Cells. J Biol Chem. 284(49): 34211-34222. [46] Tian T, Wang Y, Wang H, Zhu Z, Xiao Z (2010) Visualizing of the Cellular Uptake and Intracellular Trafficking of Exosomes by Live-cell Microscopy. J Cell Biochem. 111(2):488-496. [47] Tian T, Zhu YL, Zhou YY, Liang GF, Wang YY, Hu FH, Xiao ZD (2014) Exosome Uptake Through Clathrin-mediated Endocytosis and Macropinocy‐ tosis and Mediating Mir-21 Delivery. J Biol Chem. 289(32):22258-22267. [48] Feng D, Zhao WL, Ye YY, Bai XC, Liu RQ, Chang LF, Zhou Q, Sui SF (2010) Cellular Internalization of Exosomes Occurs Through Phagocytosis. Traffic. 11(5):675-687. [49] Smyth TJ, Redzic JS, Graner MW, Anchordoquy TJ (2014) Examination of the Specificity of Tumor Cell Derived Exosomes with Tumor Cells in Vitro. Biochim Biophys Acta. 1838(11):2954-2965. [50] Christianson HC, Svensson KJ, van Kuppevelt TH, Li JP, Belting M (2013) Cancer Cell Exosomes Depend on Cell-surface Heparan Sulfate Proteogly‐ cans for Their Internalization and Functional Activity. Proc Natl Acad Sci U S A. 110(43): 17380-17385. [51] Svensson KJ, Christianson HC, Wittrup A, Bour‐ seau-Guilmain E, Lindqvist E, Svensson LM, Morgelin M, Belting M (2013) Exosome Uptake Depends on ERK1/2-heat Shock Protein 27 Signal‐ ing and Lipid Raft-mediated Endocytosis Negative‐ ly Regulated by Caveolin-1. J Biol Chem. 288(24): 17713-17724. 11Lilite Sadovska, Cristina Bajo Santos, Zane Kalniņa and Aija Linē: Biodistribution, Uptake and Effects Caused by Cancer-derived Extracellular Vesicles [52] Rana S, Claas C, Kretz CC, Nazarenko I, Zoeller M (2011) Activation-induced Internalization Differs for the Tetraspanins CD9 and Tspan8: Impact on Tumor Cell Motility. Int J Biochem Cell Biol. 43(1): 106-119. [53] Morelli AE, Larregina AT, Shufesky WJ, Sullivan ML, Stolz DB, Papworth GD, Zahorchak AF, Logar AJ, Wang Z, Watkins SC, Falo LD, Jr., Thomson AW (2004) Endocytosis, Intracellular Sorting, and Processing of Exosomes by Dendritic Cells. Blood. 104(10):3257-3266. [54] Rana S, Yue S, Stadel D, Zoller M (2012) Toward Tailored Exosomes: The Exosomal Tetraspanin Web Contributes to Target Cell Selection. Int J Biochem Cell Biol. 44(9):1574-1584. [55] Ivanova L, Zandberga E, Silina K, Kalnina Z, Abols A, Endzelins E, Vendina I, Romanchikova N, Hegmane A, Trapencieris P, Eglitis J, Line A (2014) Prognostic Relevance of Carbonic Anhydrase IX Expression is Distinct in Various Subtypes of Breast Cancer and its Silencing Suppresses Self-Renewal Capacity of Breast Cancer Cells. Cancer Chemother Pharmacol. [56] Federici C, Petrucci F, Caimi S, Cesolini A, Logozzi M, Borghi M, D'Ilio S, Lugini L, Violante N, Azzarito T, Majorani C, Brambilla D, Fais S (2014) Exosome Release and Low Ph Belong to a Framework of Resistance of Human Melanoma Cells to Cisplatin. PLoS One. 9(2):e88193. [57] Poste G, Nicolson GL (1980) Arrest and Metastasis of Blood-Borne Tumor Cells Are Modified by Fusion of Plasma Membrane Vesicles from Highly Metastatic Cells. Proc Natl Acad Sci U S A. 77(1): 399-403. [58] Hao S, Ye Z, Li F, Meng Q, Qureshi M, Yang J, Xiang J (2006) Epigenetic Transfer of Metastatic Activity by Uptake of Highly Metastatic B16 Melanoma Cell- released Exosomes. Exp Oncol. 28(2):126-131. [59] Aga M, Bentz GL, Raffa S, Torrisi MR, Kondo S, Wakisaka N, Yoshizaki T, Pagano JS, Shackelford J (2014) Exosomal HIF1alpha Supports Invasive Potential of Nasopharyngeal Carcinoma-associated LMP1-positive Exosomes. Oncogene. 33(37): 4613-4622. [60] O'Brien K, Rani S, Corcoran C, Wallace R, Hughes L, Friel AM, McDonnell S, Crown J, Radomski MW, O'Driscoll L (2013) Exosomes from triple-negative breast cancer cells can transfer phenotypic traits representing their cells of origin to secondary cells. Eur J Cancer. 49(8):1845-1859. [61] Kogure T LW, Yan IK,Braconi C,Patel T (2011) Intercellular Nanovesicle-Mediated microRNA Transfer: A Mechanism of Environmental Modula‐ tion of Hepatocellular Cancer Cell Growth. HEPA‐ TOLOGY. 54(4):1237-1248. [62] Corcoran C RS, O'Brien K, O'Neill A, Prencipe M, Sheikh R, Webb G, MsDermott R, Watson W, Crown J, O'Driscoll L (2012) Docetaxel-Resistance in Prostate Cancer: Evaluating Associated Phenotypic Changes and Potential for Resistance Transfer Via Exosomes. PLoS One. 7(12): e50999. [63] Chen WX, Liu XM, Lv MM, Chen L, Zhao JH, Zhong SL, Ji MH, Hu Q, Luo Z, Wu JZ, Tang JH (2014) Exosomes from Drug-resistant Breast Cancer Cells Transmit Chemoresistance by a Horizontal Transfer of microRNAs. PLoS One. 9(4):e95240. [64] Galindo-Hernandez O, Serna-Marquez N, Castillo- Sanchez R, Salazar EP (2014) Extracellular vesicles from MDA-MB-231 Breast Cancer Cells Stimulated with Linoleic Acid Promote an EMT-like Process in MCF10A Cells. Prostaglandins Leukot Essent Fatty Acids. 91(6):299-310. [65] Melo SA, Sugimoto H, O'Connell JT, Kato N, Villanueva A, Vidal A, Qiu L, Vitkin E, Perelman LT, Melo CA, Lucci A, Ivan C, Calin GA, Kalluri R (2014) Cancer Exosomes Perform Cell-independent Microrna Biogenesis and Promote Tumorigenesis. Cancer Cell. 26(5):707-721. [66] Abd Elmageed ZY, Yang Y, Thomas R, Ranjan M, Mondal D, Moroz K, Fang Z, Rezk BM, Moparty K, Sikka SC, Sartor O, Abdel-Mageed AB (2014) Neoplastic Reprogramming of Patient-Derived Adipose Stem Cells by Prostate Cancer Cell- associated Exosomes. Stem Cells. 32(4):983-997. [67] Al-Nedawi K, Meehan B, Kerbel RS, Allison AC, Rak J (2009) Endothelial Expression of Autocrine VEGF Upon the Uptake of Tumor-Derived Micro‐ vesicles Containing Oncogenic EGFR. Proc Natl Acad Sci U S A. 106(10):3794-3799. [68] Lazaro-Ibanez E, Sanz-Garcia A, Visakorpi T, Escobedo-Lucea C, Siljander P, Ayuso-Sacido A, Yliperttula M (2014) Different gDNA Content in the Subpopulations of Prostate Cancer Extracellular Vesicles: Apoptotic Bodies, Microvesicles, and Exosomes. Prostate. 74(14):1379-1390. [69] Lee TH, Chennakrishnaiah S, Audemard E, Mon‐ termini L, Meehan B, Rak J (2014) Oncogenic Ras- driven Cancer Cell Vesiculation Leads to Emission of Double-Stranded DNA Capable of Interacting with Target Cells. Biochem Biophys Res Commun. 451(2):295-301. [70] Scheel C, Weinberg RA (2012) Cancer Stem Cells and Epithelial-Mesenchymal Transition: Concepts and Molecular Links. Semin Cancer Biol. 22(5-6): 396-403. [71] Higginbotham JN, Demory Beckler M, Gephart JD, Franklin JL, Bogatcheva G, Kremers GJ, Piston DW, Ayers GD, McConnell RE, Tyska MJ, Coffey RJ (2011) Amphiregulin Exosomes Increase Cancer Cell Invasion. Curr Biol. 21(9):779-786. 12 J Circ Biomark, 2015, 4:2 | doi: 10.5772/60522 [72] Demory Beckler M, Higginbotham JN, Franklin JL, Ham AJ, Halvey PJ, Imasuen IE, Whitwell C, Li M, Liebler DC, Coffey RJ (2013) Proteomic Analysis of Exosomes from Mutant KRAS Colon Cancer Cells Identifies Intercellular Transfer of Mutant KRAS. Mol Cell Proteomics. 12(2):343-355. [73] Skog J, Wurdinger T, van Rijn S, Meijer DH, Gainche L, Sena-Esteves M, Curry WT, Jr., Carter BS, Krichevsky AM, Breakefield XO (2008) Glioblasto‐ ma Microvesicles Transport RNA and Proteins that Promote Tumour Growth and Provide Diagnostic Biomarkers. Nat Cell Biol. 10(12):1470-1476. [74] Grange C, Tapparo M, Collino F, Vitillo L, Damasco C, Deregibus MC, Tetta C, Bussolati B, Camussi G (2011) Microvesicles Released from Human Renal Cancer Stem Cells Stimulate Angiogenesis and Formation of Lung Premetastatic Niche. Cancer Res. 71(15):5346-5356. [75] Taverna S FA, * Saieva L,* Kohn EC, Santoro A, Meraviglia S, De Leo G, Alessandro R (2012) Role of Exosomes Released by Chronic Myelogenous Leukemia Cells in Angiogenesis. International Journal of Cancer. 130(9):2033–2043. [76] Liu Y, Zhu XJ, Zeng C, Wu PH, Wang HX, Chen ZC, Li QB (2014) Microvesicles Secreted from Human Multiple Myeloma Cells Promote Angiogenesis. Acta Pharmacol Sin. 35(2):230-238. [77] Hood JL, Pan H, Lanza GM, Wickline SA, Consor‐ tium for Translational Research in Advanced I, Nanomedicine (2009) Paracrine Induction of Endothelium by Tumor Exosomes. Lab Invest. 89(11):1317-1328. [78] Zhuang G, Wu X, Jiang Z, Kasman I, Yao J, Guan Y, Oeh J, Modrusan Z, Bais C, Sampath D, Ferrara N (2012) Tumour-secreted miR-9 Promotes Endothe‐ lial Cell Migration and Angiogenesis by Activating the JAK-STAT Pathway. EMBO J. [79] Umezu T, Ohyashiki K, Kuroda M, Ohyashiki JH (2012) Leukemia Cell to Endothelial Cell Commu‐ nication Via Exosomal miRNAs. Oncogene. [80] Öhlund D., Elyada E., D. T (2014) Fibroblast Heterogeneity in the Cancer Wound. The Journal of Experimental Medicine; p. 1503-1523. [81] Orimo A, Gupta PB, Sgroi DC, Arenzana-Seisdedos F, Delaunay T, Naeem R, Carey VJ, Richardson AL, Weinberg RA (2005) Stromal Fibroblasts Present in Invasive Human Breast Carcinomas Promote Tumor Growth and Angiogenesis Through Elevat‐ ed SDF-1/CXCL12 Secretion. Cell. 121(3):335-348. [82] Mrazek AA, Carmical JR, Wood TG, Hellmich MR, Eltorky M, Bohanon FJ, Chao C (2014) Colorectal Cancer-Associated Fibroblasts are Genotypically Distinct. Curr Cancer Ther Rev. 10(2):97-218. [83] Jiang L, Gonda TA, Gamble MV, Salas M, Seshan V, Tu S, Twaddell WS, Hegyi P, Lazar G, Steele I, Varro A, Wang TC, Tycko B (2008) Global Hypomethyla‐ tion of Genomic DNA in Cancer-Associated Myofibroblasts. Cancer Res. 68(23):9900-9908. [84] Kojima Y, Acar A, Eaton EN, Mellody KT, Scheel C, Ben-Porath I, Onder TT, Wang ZC, Richardson AL, Weinberg RA, Orimo A (2010) Autocrine TGF-beta and Stromal Cell-derived Factor-1 (SDF-1) Signal‐ ing Drives the Evolution of Tumor-Promoting Mammary Stromal Myofibroblasts. Proc Natl Acad Sci U S A. 107(46):20009-20014. [85] Webber J, Steadman R, Mason MD, Tabi Z, Clayton A (2010) Cancer Exosomes Trigger Fibroblast to Myofibroblast Differentiation. Cancer Res. 70(23): 9621-9630. [86] Webber JP, Spary LK, Sanders AJ, Chowdhury R, Jiang WG, Steadman R, Wymant J, Jones AT, Kynaston H, Mason MD, Tabi Z, Clayton A (2014) Differentiation of Tumour-Promoting Stromal Myofibroblasts by Cancer Exosomes. Oncogene. [87] Gu J. QH, Shen L., Zhang X, Zhu W, Huang L, Yan Y, Mao F, Zhao C, Shi Y, Xu W (2012) Gastric Cancer Exosomes Trigger Differentiation of Umbilical Cord Derived Mesenchymal Stem Cells to Carcinoma- Associated Fibroblasts through TGF-b/Smad Pathway. PLoS One. 7(12). [88] Cho JA, Park H, Lim EH, Lee KW (2012) Exosomes from Breast Cancer Cells Can Convert Adipose Tissue-Derived Mesenchymal Stem Cells into Myofibroblast-Like Cells. Int J Oncol. 40(1):130-138. [89] Andre F, Andersen M, Wolfers J, Lozier A, Raposo G, Serra V, Ruegg C, Flament C, Angevin E, Amigorena S, Zitvogel L (2001) Exosomes in Cancer Immunotherapy: Preclinical Data. Adv Exp Med Biol. 495:349-354. [90] Wolfers J, Lozier A, Raposo G, Regnault A, Thery C, Masurier C, Flament C, Pouzieux S, Faure F, Tursz T, Angevin E, Amigorena S, Zitvogel L (2001) Tumor-derived Exosomes are a Source of Shared Tumor Rejection Antigens for CTL Cross-priming. Nat Med. 7(3):297-303. [91] Dai S, Wan T, Wang B, Zhou X, Xiu F, Chen T, Wu Y, Cao X (2005) More Efficient Induction of HLA- A*0201-Restricted and Carcinoembryonic Antigen (CEA)-Specific CTL Response by Immunization with Exosomes Prepared from Heat-Stressed CEA- Positive Tumor Cells. Clin Cancer Res. 11(20): 7554-7563. [92] Dai S, Wei D, Wu Z, Zhou X, Wei X, Huang H, Li G (2008) Phase I Clinical Trial of Autologous Ascites- derived Exosomes Combined with GM-CSF for Colorectal Cancer. Mol Ther. 16(4):782-790. [93] Dai S, Zhou X, Wang B, Wang Q, Fu Y, Chen T, Wan T, Yu Y, Cao X (2006) Enhanced Induction of Dendritic Cell Maturation and HLA-A*0201- Restricted CEA-Specific CD8(+) CTL Response by Exosomes Derived from IL-18 Gene-Modified CEA- 13Lilite Sadovska, Cristina Bajo Santos, Zane Kalniņa and Aija Linē: Biodistribution, Uptake and Effects Caused by Cancer-derived Extracellular Vesicles positive Tumor Cells. J Mol Med (Berl). 84(12): 1067-1076. [94] Xie Y, Bai O, Zhang H, Li W, Xiang J (2010) Tumor Necrosis Factor Gene-Engineered J558 Tumor Cell- released Exosomes Stimulate Tumor Antigen P1A- Specific CD8+ CTL Responses and Antitumor Immunity. Cancer Biother Radiopharm. 25(1): 21-28. [95] Andreola G, Rivoltini L, Castelli C, Huber V, Perego P, Deho P, Squarcina P, Accornero P, Lozupone F, Lugini L, Stringaro A, Molinari A, Arancia G, Gentile M, Parmiani G, Fais S (2002) Induction of Lymphocyte Apoptosis by Tumor Cell Secretion of FasL-bearing Microvesicles. Journal of Experimen‐ tal Medicine. 195(10):1303-1316. [96] Huber V, Fais S, Iero M, Lugini L, Canese P, Squarcina P, Zaccheddu A, Colone M, Arancia G, Gentile M, Seregni E, Valenti R, Ballabio G, Belli F, Leo E, Parmiani G, Rivoltini L (2005) Human Colorectal Cancer Cells Induce T-Cell Death Through Release of Proapoptotic Microvesicles: Role in Immune Escape. Gastroenterology. 128(7): 1796-1804. [97] Abusamra AJ, Zhong Z, Zheng X, Li M, Ichim TE, Chin JL, Min WP (2005) Tumor Exosomes Express‐ ing Fas ligand Mediate CD8+ T-cell Apoptosis. Blood Cells Mol Dis. 35(2):169-173. [98] Wieckowski EU, Visus C, Szajnik M, Szczepanski MJ, Storkus WJ, Whiteside TL (2009) Tumor- derived Microvesicles Promote Regulatory T Cell Expansion and Induce Apoptosis in Tumor- Reactive Activated CD8+ T Lymphocytes. J Immu‐ nol. 183(6):3720-3730. [99] Liu Z-M, Wang Y-B, Yuan X-H (2013) Exosomes from Murine-derived GL26 Cells Promote Glioblas‐ toma Tumor Growth by Reducing Number and Function of CD8+T Cells. Asian Pacific Journal of Cancer Prevention. 14(1):309-314. [100] Taylor DD G-TC, Lyons KS, Stanson J, Whiteside TL (2003) T-Cell Apoptosis and Suppression of T-Cell Receptor/CD3 by Fas Ligand-containing Membrane Vesicles Shed fromOvarian Tumors. Clinical Cancer Research. 9:5113-5119. [101] Taylor DD, Gercel-Taylor C (2005) Tumour-derived Exosomes and Their Role in Cancer-associated T- cell Signalling Defects. Br J Cancer. 92(2):305-311. [102] Szajnik M, Czystowska M, Szczepanski MJ, Man‐ dapathil M, Whiteside TL (2010) Tumor-derived Microvesicles Induce, Expand and Up-regulate Biological Activities of Human Regulatory T Cells (Treg). PLoS One. 5(7):e11469. [103] Clayton A, Mitchell JP, Court J, Mason MD, Tabi Z (2007) Human Tumor-derived Exosomes Selective‐ ly Impair Lymphocyte Responses to Interleukin-2. Cancer Res. 67(15):7458-7466. [104] Wada J, Onishi H, Suzuki H, Yamasaki A, Nagai S, Morisaki T, Katano M (2010) Surface-bound TGF- beta1 on Effusion-derived Exosomes Participates in Maintenance of Number and Suppressive Function of Regulatory T-Cells in Malignant Effusions. Anticancer Res. 30(9):3747-3757. [105] Yin Y, Cai X, Chen X, Liang H, Zhang Y, Li J, Wang Z, Zhang W, Yokoyama S, Wang C, Li L, Hou D, Dong L, Xu T, Hiroi T, Yang F, Ji H, Zhang J, Zen K, Zhang CY (2014) Tumor-secreted Mir-214 Induces Regulatory T Cells: A Major Link Between Immune Evasion and Tumor Growth. Cell Res. 24(10): 1164-1180. [106] Clayton A, Al-Taei S, Webber J, Mason MD, Tabi Z (2011) Cancer Exosomes Express CD39 and CD73, Which Suppress T Cells Through Adenosine Production. J Immunol. 187(2):676-683. [107] Clayton A, Mitchell JP, Court J, Linnane S, Mason MD, Tabi Z (2008) Human Tumor-derived Exo‐ somes Down-modulate NKG2D Expression. The Journal of Immunology. 180(11):7249-7258. [108] Ashiru O, Boutet P, Fernandez-Messina L, Aguera- Gonzalez S, Skepper JN, Vales-Gomez M, Reyburn HT (2010) 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. 70(2):481-489. [109] Valenti R, Huber V, Filipazzi P, Pilla L, Sovena G, Villa A, Corbelli A, Fais S, Parmiani G, Rivoltini L (2006) Human Tumor-released Microvesicles Promote the Differentiation of Myeloid Cells with Transforming Growth Factor-beta-mediated Suppressive Activity on T Lymphocytes. Cancer Res. 66(18):9290-9298. [110] Chalmin F LS, Mignot G, Vincent J, Bruchard M, Remy-Martin JP, Boireau W, Rouleau A, Simon B, Lanneau D,De Thonel A, Multhoff G, Hamman A, Martin F, Chauffert B, Solary E, Zitvogel L, Garrido C, Ryffel B,Borg C, Apetoh L, Rébé C,Ghiringhelli F (2010) Membrane-associated Hsp72 from Tumor- Derived Exosomes Mediates STAT3-dependent Immunosuppressive Function of Mouse and Human Myeloid-derived Suppressor Cells. The Journal of Clinical Investigation. 120(2):15. [111] Gabrilovich DI, Nagaraj S (2009) Myeloid-derived Suppressor Cells as Regulators of the Immune system. Nat Rev Immunol. 9(3):162-174. [112] Marton A, Vizler C, Kusz E, Temesfoi V, Szathmary Z, Nagy K, Szegletes Z, Varo G, Siklos L, Katona RL, Tubak V, Howard OM, Duda E, Minarovits J, Buzas K (2012) Melanoma Cell-derived Exosomes Alter Macrophage and Dendritic Cell Functions in Vitro. Immunol Lett. 148(1):34-38. [113] Bobrie A, Krumeich S, Reyal F, Recchi C, Moita LF, Seabra MC, Ostrowski M, Thery C (2012) Rab27a Supports Exosome-dependent and -Independent 14 J Circ Biomark, 2015, 4:2 | doi: 10.5772/60522 Mechanisms That Modify the Tumor Microenvir‐ onment and Can Promote Tumor Progression. Cancer Res. 72(19):4920-4930. [114] Gyorgy B, Fitzpatrick Z, Crommentuijn MH, Mu D, Maguire CA (2014) Naturally Enveloped AAV Vectors for Shielding Neutralizing Antibodies and Robust Gene Delivery in Vivo. Biomaterials. 35(26): 7598-7609. [115] Maguire CA, Balaj L, Sivaraman S, Crommentuijn MH, Ericsson M, Mincheva-Nilsson L, Baranov V, Gianni D, Tannous BA, Sena-Esteves M, Breakefield XO, Skog J (2012) Microvesicle-associated AAV Vector as a Novel Gene Delivery System. Mol Ther. 20(5):960-971. [116] Ekstrom K, Valadi H, Sjostrand M, Malmhall C, Bossios A, Eldh M, Lotvall J (2012) Characterization of Mrna and Microrna in Human Mast Cell-derived Exosomes and Their Transfer to Other Mast Cells and Blood CD34 Progenitor Cells. J Extracell Vesicles. 1. [117] Eldh M, Ekstrom K, Valadi H, Sjostrand M, Olsson B, Jernas M, Lotvall J (2010) Exosomes Communi‐ cate Protective Messages During Oxidative Stress; Possible Role of Exosomal Shuttle RNA. PLoS One. 5(12):e15353. [118] Lasser C, O'Neil SE, Ekerljung L, Ekstrom K, Sjostrand M, Lotvall J (2011) RNA-containing Exosomes in Human Nasal Secretions. Am J Rhinol Allergy. 25(2):89-93. [119] Cheng L, Sharples RA, Scicluna BJ, Hill AF (2014) Exosomes Provide a Protective and Enriched Source of Mirna for Biomarker Profiling Compared to Intracellular and Cell-free Blood. J Extracell Vesi‐ cles. 3. [120] Li M, Zeringer E, Barta T, Schageman J, Cheng A, Vlassov AV (2014) Analysis of the RNA Content of the Exosomes Derived from Blood Serum and Urine and its Potential as Biomarkers. Philos Trans R Soc Lond B Biol Sci. 369(1652). [121] Chevillet JR, Kang Q, Ruf IK, Briggs HA, Vojtech LN, Hughes SM, Cheng HH, Arroyo JD, Meredith EK, Gallichotte EN, Pogosova-Agadjanyan EL, Morrissey C, Stirewalt DL, Hladik F, Yu EY, Higano CS, Tewari M (2014) Quantitative and Stoichiomet‐ ric Analysis of the Microrna Content of Exosomes. Proc Natl Acad Sci U S A. 111(41):14888-14893. [122] Thakur BK, Zhang H, Becker A, Matei I, Huang Y, Costa-Silva B, Zheng Y, Hoshino A, Brazier H, Xiang J, Williams C, Rodriguez-Barrueco R, Silva JM, Zhang W, Hearn S, Elemento O, Paknejad N, Manova-Todorova K, Welte K, Bromberg J, Peinado H, Lyden D (2014) Double-stranded DNA in Exosomes: a Novel Biomarker in Cancer Detection. Cell Res. 24(6):766-769. 15Lilite Sadovska, Cristina Bajo Santos, Zane Kalniņa and Aija Linē: Biodistribution, Uptake and Effects Caused by Cancer-derived Extracellular Vesicles