Next Article in Journal
Qualitative and Quantitative Assessment of Vascular Changes in Diabetic Macular Edema after Dexamethasone Implant Using Optical Coherence Tomography Angiography
Next Article in Special Issue
Manufacturing of Human Extracellular Vesicle-Based Therapeutics for Clinical Use
Previous Article in Journal
The Combination of Arginine Deprivation and 5-Fluorouracil Improves Therapeutic Efficacy in Argininosuccinate Synthetase Negative Hepatocellular Carcinoma
Previous Article in Special Issue
The Methods of Choice for Extracellular Vesicles (EVs) Characterization
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Extracellular Vesicles in Hematological Malignancies: From Biology to Therapy

1
Laboratory of Preclinical and Translational Research, IRCCS-Referral Cancer Center of Basilicata (CROB), 858028 Rionero in Vulture, Italy
2
Laboratory of Clinical Research and Advanced Diagnostics, IRCCS-Referral Cancer Center of Basilicata (CROB), 85028 Rionero in Vulture, Italy
3
CEINGE-Biotecnologie Avanzate scarl, Federico II University, 80138 Naples, Italy
4
Department of Molecular Medicine and Medical Biotechnologies, Federico II University, 80138 Naples, Italy
5
Scientific Direction, IRCCS-Referral Cancer Center of Basilicata (CROB), 85028 Rionero in Vulture, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2017, 18(6), 1183; https://doi.org/10.3390/ijms18061183
Submission received: 28 April 2017 / Revised: 29 May 2017 / Accepted: 30 May 2017 / Published: 2 June 2017

Abstract

:
Extracellular vesicles (EVs) are a heterogeneous group of particles, between 15 nanometers and 10 microns in diameter, released by almost all cell types in physiological and pathological conditions, including tumors. EVs have recently emerged as particularly interesting informative vehicles, so that they could be considered a true “cell biopsy”. Indeed, EV cargo, including proteins, lipids, and nucleic acids, generally reflects the nature and status of the origin cells. In some cases, EVs are enriched of peculiar molecular cargo, thus suggesting at least a degree of specific cellular packaging. EVs are identified as important and critical players in intercellular communications in short and long distance interplays. Here, we examine the physiological role of EVs and their activity in cross-talk between bone marrow microenvironment and neoplastic cells in hematological malignancies (HMs). In these diseases, HM EVs can modify tumor and bone marrow microenvironment, making the latter “stronger” in supporting malignancy, inducing drug resistance, and suppressing the immune system. Moreover, EVs are abundant in biologic fluids and protect their molecular cargo against degradation. For these and other “natural” characteristics, EVs could be potential biomarkers in a context of HM liquid biopsy and therapeutic tools. These aspects will be also analyzed in this review.

Graphical Abstract

1. Introduction

Extracellular vesicles (EVs), a heterogeneous group of lipid bilayer particles between 15 nanometers and 10 microns in size, are released by almost all (normal and neoplastic) cell types [1]. They are classified, in increasing order of size, into exosomes (Exo) [2], microvesicles (also referred as ectosomes or microparticles; MVs) [3], small and large oncosomes [4,5], according to their biophysical properties (i.e., size, shape) and mechanism of biogenesis [6].
EVs contain a set of cytoplasmic and surface proteins, lipids [7], nucleic acids (DNA, mRNA, and small and long noncoding RNA) [8,9,10,11,12] and even cellular organelles, such as mitochondria [13]. The components of EVs, currently known, are summarized in some databases such as EVpedia, Vesiclepedia, and ExoCarta [14,15,16].
The EV cargo generally reflects the nature and status of the origin cell [17,18]. In some cases, EV cytoplasmic and surface content suggest a degree of specific cellular packaging into EVs [6,19].
The release of EVs, both in physiological and pathological conditions, including tumors, is influenced by microenvironment [20,21], oxygen tension [22], glucose [23] and intracellular Ca2+ concentrations [3], and the status of donor cells [17,18].
Once released from a given cell, EVs can reach target cells where they can remain stably associated with plasma membrane or can be internalized through diverse endocytic pathways with subsequent transfer of EV cargo into cytoplasm and nucleus of recipient cells [24,25,26,27,28]. In particular, Rappa et al. described spathasomes as a mechanism to transfer EV biomaterials into the nucleus of recipient cells [28]. Due to the ability to transfer their cargo to target cells, EVs can alter the composition and function of recipient cells and can even induce epigenetic changes [29,30,31,32]. EVs exert effects in a pleiotropic manner, by directly activating cell surface receptors via protein and lipid ligands, or by integrating their membrane content into the plasma membrane of their cellular targets [33,34].
EVs can be isolated from diverse body fluids [35], including blood [36], saliva [37], semen [38], seminal plasma [18], mother’s milk [39], synovial fluid [40], nasal secretions [18], urine, feces [18,41], amnion [42] , ascites [43], and cerebrospinal fluids [44]. In particular, we reported that serum MVs are elevated in patients with different hematological malignancies (HMs) when compared to healthy subjects and, more importantly, they exposed surface specific HM associated markers [36].
EVs have emerged as particularly interesting tumor information vehicles, such that they could be considered for “cell biopsies”. Therefore, EVs could be used as minimally invasive biomarkers and even as a therapeutic tool. Their role was extensively studied in solid tumors [45,46,47,48] whereas in HMs it is in the process of being defined [49,50,51].
The aim of this review is to discuss the recent advances in the field of EVs as actors in HMs, underlining the EV role in the tumor-microenvironment cross-talk, in drug resistance, as well as their potential use as biomarkers and therapeutic tools.

2. Physiological Role of Extracellular Vesicles (EVs)

EVs play important roles in intercellular cross-talk in both short and long distances [1,52]; in fact, they are involved in numerous physiological processes, including stem cell renewal and differentiation [53], tissue repair [31], immune surveillance [33], and blood coagulation [33,54]. For these reasons, they have been highly preserved through evolution [9,18].
Ratajczak et al. reported that MVs isolated from mouse embryonic stem cells (SCs) efficiently enhanced survival and expansion of murine hematopoietic progenitor cells (HPCs) [55]. A recent study showed that MVs from megakaryocytes (Mks) induced differentiation of hematopoietic stem and progenitor cells (HSPCs) towards the Mk lineage [56].
EVs seem to also play important roles in tissue engineering and regenerative activities, including liver, nervous, vascular, reproductive, and renal systems [57,58]. Indeed, EVs released from injured tissues, acting on SCs, promoted the release of “regenerative” MVs for tissue repair [59]. Gatti et al. reported that intravenous administration of EVs derived from human mesenchymal stem/stromal cells (MSCs) stimulated tubular cell proliferation and protected against acute kidney injury [31].
All the immune system cells, including macrophages, B, T, NK, and dendritic cells (DCs), can release EVs. A lot of evidence suggests that EVs have an important role in the regulation of immunity, acting both as immune stimulators or suppressors [6,18,52,60]. EVs acted as immune suppressors by enhancing the function of regulatory T cells (T-regs), inhibiting NK and CD8+ cell activity [33,61], and affecting monocyte differentiation [62]. Recently, Jong et al. reported that activated NK cells produced EVs that were active against cancer cells, delivering cytotoxic proteins, such as perforin and granzymes A and B [63]. Moreover, EVs from embryonic SCs affect DC maturation and both T cell proliferation and differentiation, thus potentially contributing to innate immune suppression [64]. Interestingly, healthy MSC-derived EVs also induced immunosuppressive effects on purified T, B, and NK cells from healthy donors [65].
One of the best characterized physiological roles of EVs is their capacity to enhance coagulation [18]. An inverse correlation between the EV number and their capacity to form both thrombin and thrombin-antithrombin complexes has been clearly demonstrated [18,66]. Furthermore, EVs found in saliva and urine of healthy humans exposed coagulant tissue factor (TF) and initiated TF/factor VII-mediated coagulation [29].
EVs are also involved in fertilization, pregnancy, and fetus/mother communication [18,67]. In amniotic fluid and in human breast milk, EVs regulate the immune response of fetus to maximize its survival during pregnancy and growth [18].
Finally, EVs participate in myelin formation, as well as in neurite outgrowth and neuronal survival of nervous system [68,69].

3. Role of EVs in the Malignancy-Microenvironment Cross-Talk

The bone marrow (BM) hematopoietic niche consists of SCs, hematopoietic (both myeloid and lymphoid precursors), immune- and stromal cells (BMSCs); the latter includes vascular endothelial cells, pericytes, adipocytes, fibroblasts, osteoblasts/osteoclast, and MSCs, as well as extracellular matrix (ECM) [70,71]. All BM cell populations can significantly influence tumor microenvironment, via autocrine or paracrine mechanisms, through the secretion of a large variety of soluble factors, including EVs. In this context, EVs represent an important vehicle of growth factors, cytokines, enzymes, angiogenic molecules, and genetic materials that are able to reprogram and modify recipient cells [72]. The complex EV mediated bi-directional communications between tumor and BM microenvironment are summarized in Figure 1.

3.1. Autocrine Loop of Primary Tumor EVs

Several studies have suggested that, under different conditions, tumor cells produced EVs that modulated themselves, in an autocrine feedback loop [73,74,75], which is relevant for the concepts of tumor auto-sustaining and the increase of tumor aggressiveness.
It has been recently demonstrated that multiple myeloma (MM) EVs have a prominent role in promoting tumor plasma cell proliferation through both direct and indirect mechanisms. In particular, EVs derived from MM cells over-expressing CD147 (an extracellular matrix metalloproteinase inducer also known as basigin) enhanced neoplastic proliferation more than EVs released from CD147 downregulated cells, thus suggesting that CD147 is involved in EV induced MM proliferation [73].
Patel et al. reported that Exo derived from a pre-B acute lymphoblastic leukemia (ALL) cell line growing at high density induced a proliferative effect on the same non-growing low density cells, favoring their survival [76]. In addition, ALL-derived Exo also induced cell proliferation of leukemic and non-leukemic B cell lines in both an autocrine and paracrine manner [74].
Several leukemic cells, including erythromyeloblastoid, chronic myeloid leukemia (CML), and pre-B ALL cells, released their specific oncogenic fusion transcripts in MVs; interestingly, these EVs returned to leukemic cells with an autocrine loop [75]. In particular, CML-derived Exo promoted the proliferation and survival of CML cells, both in vitro and in vivo, by TGF-β1/TGF-β1 receptor engagement [77]. However, this EV autocrine loop did not work in a chronic lymphocytic leukemia (CLL) model [71]; a possible explanation could be that CLL cells did not express heparan sulphate (HS) receptors required for the EV uptake [78]. Further studies are needed to better define the autocrine processes involving EVs. Moreover, Paggetti et al. demonstrated that CLL-derived exosomes induce an inflammatory phenotype in endothelial and MSCs resembling the phenotype of cancer-associated fibroblasts. CLL-derived exosomes, in this way, create a favorable environment for promoting CLL progression [78]. Further studies are needed to better define the autocrine processes involving EVs.

3.2. EV Mediated Communication between Stem Cells and Malignancy

It is well established that the embryonic microenvironment is non-permissive for tumor development [79]. In this setting, Zhou et al. demonstrated in vitro and in vivo that Exo released by human embryonic SCs could reprogram cancer cells, inhibiting tumor growth and tumorigenicity. On the other hand, tumor cells can interact and modify the SCs through the EVs [79].
It has been demonstrated that leukemic SC derived MVs promote proliferation and migration and inhibit apoptosis of acute myeloid leukemia (AML) cells [80]. In addition, AML derived Exo suppressed residual HSPC functions by “decreasing” clonogenicity and by reprogramming stroma, respectively [81,82]. Recently, BM-AML-MVs promoted the survival of healthy hematopoietic stem cells (HSCs) without changing their immature phenotype and their ability to form colonies, but inducing leukemic-like functional characteristics, such as miR21 and miR29 over-expression [83]. Interestingly, researchers demonstrated an essential role of vacuolar protein sorting protein 33b (VPS33B) in Exo pathways of HSC and leukemia-initiating cells. In particular, the VPS33B deletion in an in vivo AML model led to impairment of both Exo maturation and secretion and delayed the onset of AML [84]. Finally, Koch et al. reported that diffuse large B-cell lymphomas possessed an infrastructure comprising hematopoietic stem cells (HSCs) and not HSCs. Exo mediated Wnt signaling modulated the transitions between clonogenic states [85].

3.3. Immuno-Cells and Tumor Cross-Talk

Depending on the type of cells from which they are released, EVs exert immune-modulatory activities, influencing both the activation and suppression of the immune response [86]. In particular, the release of EVs by neoplastic cells is one mechanism through which the antitumor immune response is suppressed or evaded [87].
For example, EVs derived from B and T cell lymphomas have been found to be enriched in major histocompatibility complex (MHC), T-cell receptor (TCR), Apo2 ligand (APO2L), Fas ligand (FasL), and natural-killer group-2 member-D (NKG2D), which had the ability to inhibit NK cell cytotoxicity, to promote T cell apoptosis, and to downregulate antigen (Ag) processing by Ag presenting cells [88,89,90].
In CLL, where NK cell dysfunction has been reported [91], the plasma soluble ligand BAG6 inactivated NK cell functions by binding their receptor NKp30. On the contrary, BAG6-positive Exo activated NK cells that could kill tumor cells [92]. Therefore, a possible explanation of CLL immune evasion could be a deregulated balance between BAG6-Exo and BAG6 soluble form.
Both MM cell and BMSC-Exo promoted the growth and enhanced the immunosuppressive activity of myeloid derived suppressor cells (MDSCs) in vitro and in MM xenograft [93,94]; moreover, MM-Exo reduced cytotoxic activity of NK cells against MM cells [95]. In addition, CD38-positive EVs released from MM cells could represent an MM strategy to escape immune system. In fact, ectoenzyme CD38 on MM EVs could generate an anergic immune system by converting nucleotides to adenosine, which is an immune-suppressor; in addition, CD38-positive EVs binding anti-CD38 mAb (Daratumumab) could be captured by FcR-positive cells such as NK, monocytes, and MDSCs [96,97]. The effects of EV internalization by immune cells are under investigation.
Exo from Burkitt’s lymphoma cell line (DG75), from latent membrane protein1 (LMP1)-transfected cell line (LMP-DG75) or from EBV (Epstein-Barr virus)-infected cell line (DG75-EBV) was used in order to mimic Exo produced during EBV infection or EBV-associated diseases. DG75 Exo were internalized by B cells in PBMCs, and led their proliferation and T cell independent class-switch recombination. Additionally, LMP1-DG75 Exo induced the differentiation of B cells into a plasmablast-like phenotype [98].
In Hodgkin lymphoma (HL), Hansen et al. reported that Hodgkin and Reed Stemberg (HRS) cells released CD30-positive EVs that mediate communication with supportive microenvironment. In particular, CD30+ EVs stimulated innate immune cells, such as healthy eosinophil-like EoL-1 cells and primary granulocytes to release Interleukin (IL)-8 [99].
TGFβ1 plays a significant role in leukemic Exo-mediated immune escape [100]. Exo from sera of AML patients containing membrane-associated TGFβ1 reduced the ability of NK cells to kill leukemic cells; furthermore, their presence in sera correlated to response to chemotherapy in AML [101]. TGFβ1 has also been found to be enriched in CML-Exo and treatment with TGF-β1 inhibitor significantly reduced Exo-stimulated cell proliferation and colony formation of CML cells [100].

3.4. EV Mediated Bi-Directional Communication between BM Stroma and Tumor

EVs participate in the cross-talk between malignant cells and BMSCs [93,102,103,104], exerting either an anti- or a pro-tumor growth effect, depending on cancer type and stage of disease [62,105].
Ghosh et al. found that CLL-MVs play an important role in the activation of CLL microenvironment in favor of disease progression [106]. In particular, CLL-MVs can activate the AKT signaling pathway in CLL-BMSCs by inducing the production of vascular endothelial growth factor (VEGF), a survival factor for CLL cells [107]. Moreover, CLL-Exo facilitate a transition of stromal cells towards cancer-associated fibroblast phenotype and promote cell migration and angiogenesis in vitro and in vivo [78,108].
Fei and colleagues demonstrated the protective effects of stroma versus ALL cells following the release of EVs contained Galectin-3 (GAL3) [109]. GAL3was internalized by ALL cells, where it stimulated transcription of endogenous GAL3 mRNA and was a protection toward drug treatment [109]. Also, ALL Exo could be captured from stromal cells, inducing a metabolic switch from oxidative phosphorylation to aerobic glycolysis [110].
The interaction between BMSCs and MM cells plays a key role in MM pathogenesis [93]. BMSC-derived Exo induced survival, proliferation, and migration of MM cells in an in vivo MM mouse model, while normal BM-MSC-Exo significantly reduced MM cell proliferation [102]. Interestingly, Exo from cells of MM patients, under hypoxia conditions, accelerate in vitro angiogenesis; this effect is due to miR135b, which targets the HIF1-dependent signaling pathway [111]. Notably, it has been reported that MM-MVs carrying multiple angiogenesis-related proteins enhanced angiogenesis by modulating the STAT3 pathway and that endothelial cells stimulated with MM-MVs secrete IL-6 and VEGF, two important factors for MM cell growth [112]. Moreover, Exo from MM cells and MM patient sera also interacted with osteoclasts (OCs), supporting their growth and migration and inducing a differentiation toward multinuclear OCs which eroded dentine discs, thus suggesting a possible role in the development of bone disease of MM patients [113]. Vardaki and colleagues firstly provided evidence that Bcl-xL, a caspase-3 substrate, was presented in the BM-derived fibroblast Exo and that the consequent cleaved Bcl-xL (clBcl-xL) was exposed on Exo surface. The clBcl-xL was necessary for EV uptake by both MM and aggressive lymphoma cells, driving their proliferation [114].
Exo derived from adult T-cell leukemia/lymphoma (ATL) cells transferred epigenetic regulators, such as miR21 and miR155, induced changes in cellular morphology, and promoted proliferation of human MSCs to support the creation of a favorable microenvironment for leukemia [115].
Horiguchi et al. demonstrated that EVs derived from leukemic cells were efficiently transferred into MSCs [116]. In particular, they found that EV miR-7977 derived from AML/myelodysplastic syndrome (MDS) CD34+ cells, was transferred into BM MSCs and reduced their ability to support CD34+ cells. Huan et al. studied the role of Exo in developing BM AML niche [117]. They reported that AML-Exo altered the proliferative and migratory responses of both BMSCs and hematopoietic progenitor cell lines, resulting in the re-programming of the microenvironment. As recently suggested by Muntion et al., MVs derived from MSCs of MDS patients strongly modify CD34+ cell properties, promoting their cell viability and clonogenic capacity and altering micro-RNA and gene expression profiling [118].
EVs released by MSCs from patients with myeloproliferative neoplasms (MPN) were found to be selectively enriched in miR155, and they induced an increase in granulocyte colony forming unit number from neoplastic CD34+ [119]. Moreover, Exo released by CML cells stimulate BMSCs to produce IL-8 which, in turn, promotes both in vitro and in vivo leukemic cell survival [104]. MVs containing “leukemic” transcripts from CML cells transferred these mRNA in healthy MSCs, increasing their proliferation [75]. In addition, CML-Exo were able to enhance angiogenesis in vitro; this effect was amplified when the Exo were released in hypoxic conditions [120]. BCR-ABL1-positive MVs from CML also induced malignant transformation of normal mononuclear cells through genomic instability via different mechanisms, which led to DNA breakage and recombination [121]. Interestingly, Gutkin et al. demonstrated that enzyme telomerase (hTERT) mRNA may be transferred, via Exo, from cancer cells, such as T-cell leukemia and CML cells, to hTERT negative fibroblasts. In these cells, hTERT mRNA was translated into an active form that transforms them into telomerase-positive cells [122]. Of interest, hTERT expression in hTERT negative cells affected their biological properties towards a tumor like-phenotype, characterized by extended or unlimited life span, prevention and reversal of senescence, resistance to DNA damage, and prevention of apoptosis [19,123].

3.5. EVs and Extracellular Matrix (ECM)

Purushothaman and colleagues revealed a new mechanism for Exo-mediated cross-talk within MM cells [124]. In particular, these authors demonstrated that surface heparan sulfate (sHS)-positive (sHS+) Exo were able to bind fibronectin (fibro), then act as ligand for sHS+ target cells. Specifically, the interaction between MM cells and fibro-sHS+ Exo induced the activation of p38 and pERK and the production of DKK1 and MMP9, two crucial factors for MM invasion. Moreover, anti-fibro antibodies (Abs) inhibited the invasion of endothelial cells mediated by MM Exo [124].
Redzic et al. demonstrated that EVs from multiple cancer cells stimulate the secretion of EMMPRIN, MMP9, and IL-6, important mediators of migration and inflammation, in human monocytic leukemia cells, thus suggesting a further manner in which EVs drive tumor progression [125].
Finally, in 3D Matrigel culture and in HL tissues, CD30+ EVs from HL cells predominantly stuck to long actin- and tubulin-based protrusions; this could be a further network to guide CD30+ EVs into the microenvironment towards distant cells, thereby easing their communication [99].

3.6. Malignancy-Microenvironment Cross-Talk and Drug Resistance

Drug resistance in cancer is a multi-factorial process due to intracellular mechanisms (e.g., gene alterations or epigenetic changes) and to interactions of cancer cells with the microenvironment, resulting in decreased drug accumulation, increased efflux, drug compartmentalization, and alterations of cellular pathways [126]. Emerging evidence has highlighted the role of EVs to confer drug resistance in cancer cells, for example, by sequestering drugs and/or by reducing their free concentration available to cells [127]. In this setting, EVs shed from donor drug-resistant to recipient drug-sensitive cells can transfer drug resistance through different modulators, such as drug-efflux pumps, miRNAs, and long noncoding RNAs [128].
Recently, Koch at al. demonstrated that lymphoma cell lines treated with anthracyclines, drugs currently used for the therapy of aggressive lymphoma, secreted anthracycline-containing Exo [129]. Moreover, the silencing of ABCA3 transporter using the cyclooxygenase inhibitor indomethacin suppressed the biogenesis of Exo, with a consequent increase in drug susceptibility of lymphoma cells in vitro and in vivo. Therefore, the authors suggested that the targeting of Exo biogenesis could provide a promising approach to overcome drug resistance and to enhance antitumor efficacy of anthracyclines [129].
EVs could exert a negative effect on the therapy efficacy in HMs. For example, BMSC-Exo inhibited bortezomib mediated cell apoptosis in MM cells [103]; likewise, GAL3 Exo from stromal cells have been reported to activate the NFkB pathway in ALL cells, inducing an anti-apoptosis effect and drug resistance [109].
CD20-positive Exo in CLL can bind anti-CD20 monoclonal Ab, Rituximab, reducing its free action on CLL cells [78,130]. Similarly, in B cell lymphoma mouse models, tumor cells evaded complement-mediated killing of immunotherapy through the action of lymphoma Exo, which binds complement [131].
In an AML setting, Viola et al. found that stromal Exo trafficking could be a candidate mechanism for extrinsic chemo-resistance which increases tyrosine kinase inhibitor resistance [132]. An AML cell line carrying FLT3 internal tandem duplication (ITD) mutation was exposed to Exo derived from normal or AML BMSCs. Both Exo types protected AML cells from cytarabine effects, while only AML-BMSC-Exo protected AML cells from FLT3 inhibitor treatment [132]. Recently, it has also been shown that AML cells resistant to apoptosis could modulate, via EVs, the expression profiling of apoptosis-related proteins of blasts sensitive to chemotherapy [133]. Finally, a multi-resistant AML cell line transferred its chemo resistance to sensitive promyelocytic leukemia cells through EVs [134]. These results indicate a first proof of concept that circulating AML-EVs could become potential biomarkers for therapy resistance. However, further studies are needed to define their clinical implications.
Interestingly, in another setting, EVs can enhance drug efficacy to kill both tumor cells and their supportive microenvironment. In HL, for example, the anti-CD30 Ab drug conjugate, Brentuximab Vedotin (SGN-35), bound CD30+ EVs released by HRS cells, forming SGN-35/CD30+ EV complex [135]. The authors reported that SGN-35 directly killed tumor cells (CD30+ cells). In addition, the tumor supportive microenvironment, including mast cells and eosinophils (CD30 negative cells), was damaged by SGN-35/CD30+ EV internalization [135].

4. Clinical Potential of EVs as Biomarkers and Therapeutics

EVs are abundant in biofluids, protect their molecular cargo against degradation, and may deliver genetic/proteic/lipidic signatures associated with specific phenotypes. They could be, therefore, considered a full-fledged form of “cell biopsy”, with several advantages in respect to circulating tumor cells, that are less present in the circulation, and in respect to cell-free circulating biomarkers, including proteins, microRNA, and others, which are susceptible to degradation and have short half-lives [136]. The aforementioned characteristics, together with the possibility “to engineer” EVs, make them attractive as new possible biomarkers and therapeutic tools in HMs.

4.1. EVs as Biomarkers

We have previously reported that serum MV count in CLL, non-Hodgkin’s lymphoma (NHL), Waldenstrom’s macroglobulinemia (WM), HL, MM, AML, MPN, and MDS patients are high compared to healthy subjects [36].
In CLL, our data are in agreement with those provided by Gosh et al. about the amount of CLL-EVs [106]. Moreover, we reported that total number of MVs in CLL patients positively correlates with advanced clinical stages, it is predictive for overall survival (OS), and, in patients with initial stages, it also correlates with time to treatment. Furthermore, serum CLL-MVs were preferentially positive for CD19 and CD37 [137]. In a different setting, Boysen et al. showed that plasma CD52+ MVs were preferentially released from CLL cells and that they can be a predictive biomarker of progression [107]. Recently, an antibody microarray (DotScan) of plasma CLL-EVs gave a profile of surface proteins. These EVs expressed moderate or high levels of CD5, CD19, CD31, CD44, CD55, CD62L, CD82, HLA-A, B, C, and HLA-DR and low levels of CD21, CD49c, and CD63 [138].
Regarding the EV genetic cargo, our and other data suggest that serum EV miR155 level is a promising prognostic/predictive biomarker in CLL, independent of clinical stage [139,140].
In HL, we demonstrated that HL MVs are positive for CD30, the phenotypic marker of HRS cells, and that patients with higher HL stage showed fewer CD30+ MV than low stage HL subjects [36]. Further studies will be performed to clarify this data. Van Eijndhoven et al. confirmed our results in terms of high levels of EVs in patients with respect to controls [141]. Moreover, we and others reported that plasma EV miRNA levels, including miR155, miR127, miR21, and let7, reflect the presence of the tumor and could be a diagnostic tool for therapy response and relapse monitoring in HL [139,141].
In WM, we showed, for the first time, higher levels of serum MVs and EV miR155 with respect to controls [36,139]. The miR155 amount also had a positive correlation trend with the International Prognostic Scoring System. Interestingly, Exo miRNA content may also differ at different phases of disease [142] and this could potentially be used as a marker of progression in patients with WM.
MM circulating EVs are enriched in CD38, CD138, CD44, superficial HSP70, and CD147 [36,73,143,144]. In these studies, the number of serum MVs CD38+ positively correlated with clinical international stage system, plasma CD138+ MVs were associated with disease phase and therapeutic response, and increased plasma CD147+ MVs were found at disease progression. Finally, Harshman et al. identified, by EV proteomic profiling, CD44 as a novel associated marker in MM [145].
In regard to circulating MM EV cargo, different authors have reported that MM EVs contained c-SRC, ZNF224, CCL2, IL-6, fibro and various miRNAs, including miR15 and 16 and miR135b [145,146]. In particular, Exo miR135b was found to enhance angiogenesis in MM bone marrow; on the contrary, its expression level in plasma Exo was significantly lower when compared to controls, thus suggesting a possible role in local BM rather than in circulation. Recently, Manier et al. indicated serum Exo derived microRNAs, let7b and miR18a as significant predictors of progression free survival (PFS) and OS in MM patients [147], supporting the possibility that circulating miRNA from Exo could be predictive of MM prognosis.
Very little information is currently available about the role of circulating EVs in MPNs, though we demonstrated that CD13+ MVs were higher in the serum of CML and primary myelofibrosis patients with respect to healthy subjects [36]. Regarding CML, a differential miRNA signature between K562 cell derived-Exo and K562 cells has also been found [148]; preliminary data indicated that plasma Exo miR215 could have a possible role in selecting CML patients who can discontinue imatinib therapy [149].
We also reported increased levels of circulating serum MVs (in particular of those positive for CD13, a “mature” myeloid surface marker) in MDS patients with respect to healthy controls. Interestingly, higher risk MDS showed the highest amount of CD13+ MVs, comparable with those detected in AML [36].
Circulating AML-EVs are enriched in CD34 [150], CD 117 [151], and CD13 [36]. Plasma AML Exo have a considerably greater amount of proteins with respect to controls [132]; in particular, protein content of Exo from AML cells resistant to chemotherapy could give relevant information about drug resistance [132]. Therefore, AML-Exo TGFβ1 levels could be a novel biomarker of response to therapy and could reflect the presence/absence of residual disease after therapy [101].
Exo from AML cells contain mRNA that are important for AML prognosis (FLT3-ITD, NPM1), for treatment, (FLT3-ITD, IGF-IR, CXCR4), and for behavior of leukemic niche (IGF-IR, CXCR4, MMP4) [117]. In regard to miRNA cargo in AML-Exo, we reported that EV miR155 was over-expressed in AML serum when compared to healthy subjects [139]. Its amount was directly correlated with white blood count cells and complex karyotypes and could be further investigated as a biomarker in AML patients. Interestingly, in an AML xenograft mouse model, Exo miR155 and other miRNAs were indicative of AML presence, thus suggesting their potential use for minimal residual disease detection [81,152].
Finally, in the allogeneic stem cell transplant (alloSCT) setting, EVs have been reported as potential biomarkers of acute graft versus host disease (GvHD). In particular, in MM patients after allograft, CD46 and CD25-positive EVs were associated with an increased risk of developing a GvHD, while CD31 and CD106-positive EVs were correlated with a reduced incidence of this complication [153]. Furthermore, plasma EVs from HMs patients undergoing alloSCT contains myeloid, erythroid, and megakaryocyte lineage specific transcripts. Interestingly, the presence of these EV mRNAs seems to precede and predict the recovery of white blood cells, reticulocyte, and platelet count in blood, thus providing a novel potential biomarker of allogeneic stem cells engraftment [154].
All the information about EVs as biomarkers in HM is summarized in Table 1.

4.2. EVs for Therapy

EVs are currently under investigation as attractive and possible tools to integrate within novel therapeutic strategies in immunotherapy, as therapeutic targets, and as mechanisms of drug delivery [155].

4.2.1. Immunotherapy

EVs may act not only as Ag carriers, but also as modulators of direct and indirect Ag presentation [64]. For instance, specific Ags carried by tumor EVs can be taken up and processed by DCs; at this point, DCs can prime tumor-specific cytotoxic T-lymphocytes to initiate the immune response [155,156,157]. Indeed, some studies have indicated the possibility of using tumor-derived Exo as vaccines in lymphoma because they have been shown to be immunogenic and effective for DC vaccination. For example, Yao et al. showed that CML Exo-pulsed DC induced strong cytotoxic antileukemic immune responses as well as protective immunity against leukemia cells in vitro and in vivo [158,159]. In experiments reported by Shen et al., HSP70+ Exo were derived from a human acute promyelocytic leukemia cell line. These HSP70+ Exo were pulsed on DC and showed to be much more effective in inducing leukemia-specific cytotoxic T lymphocytes than non-pulsed DC [160]. Exo carrying proteins important for antigen presentation were shown to elicit T cell cytotoxic activity in vitro and in vivo and to induce Th1-type antibody responses via activation of B cells [161]. Thus, combining several immune-stimulatory signals in EVs could generate EV-based vaccines that might induce potent innate and adaptive immune responses [162]. To date, the possibility of using EVs as a tool in cell free-vaccines is under consideration.
Another potential immunotherapeutic application of EVs is their use as agents to restore NK cell activity in some HMs. In particular, in CLL, where NK cell dysfunction has been reported [91], BAG6-positive Exo represents a trigger for NK cytotoxicity [92].
In the scenario of EVs as an immune-therapeutic tool, it is important to underline the role of MSC-EVs. By recapitulating both immune-modulator and cytoprotective features of parental cells, they have therapeutic/protective effects in both tissue repair and regeneration [163,164]. Moreover, MSC-EVs could be used to improve stem cell engraftment in alloSCT. In this setting, we recently demonstrated that BM-MSC-EVs are able to modify the phenotype of CD34-positive hematopoietic stem cells and increase their migration from peripheral blood to BM in an in vivo model [165]. Finally, after alloSCT, MSC-EVs have been also employed to treat GvHD refractory in therapy [166].

4.2.2. EVs as Therapeutic Target

The importance of EVs in intercellular cross-talk of tumors renders them as potential therapeutic targets. Different approaches can be applied to stop EV-mediated communications: (i) reducing the amount of EVs by interfering with their biogenesis and release; (ii) blocking EV uptake by target cells; (iii) interfering with their path in target cells; and (iv) removing EVs from the circulation.
Exo biogenesis could be attenuated by small-molecule inhibitors of enzymes or proteins such as ALIX involved in Exo formation [167]. Exo secretion can also be regulated by increasing intracellular Ca2+ levels (as observed in K562 CML cells) [168] or by the use of dimethyl amiloride (DMA), an inhibitor of Na+/Ca2+ exchange [169]. In particular, DMA was shown to reduce Exo secretion in mice bearing EL4 lymphoma. It induced inhibition of tumor growth accompanied by suppression of MDSC activity [169]. Targeting Exo biogenesis could also provide a promising approach to overcome drug resistance and to enhance anti-lymphoma efficacy of anthracyclines and anthracenediones [129].
Uptake of EVs could be targeted and/or attenuated by blocking surface adhesion molecules, such as phosphatidylserine, ICAM1, sHS, proteoglycans, or other receptors important for EV internalization [170,171]. The targeting of intracellular signaling pathways activated by Exo or oncogenes delivered to recipient cells using inhibitory RNAs is also being explored [172].
There are no data regarding the depletion of circulating EVs in HM. On the contrary, depletion of cancer-derived Exo was applied to suppress cancer metastasis. Recently, Nishida et al. demonstrated that the administration of human-specific anti-CD9 and anti-CD63 Abs, binding these Ags on Exo surface, suppressed metastasis to the lungs, lymph nodes, and thoracic cavity in a human breast cancer xenograft mouse model [173].

4.2.3. Therapeutic Drug Delivery

EVs can be exploited as drug delivery tools thanks to a series of interesting “natural” features:
(1)
An architecture that protects their cargo from nucleases and proteases;
(2)
Size (nanometer) and specific composition (lipid bilayer) that minimize recognition by the mononuclear phagocyte system [174];
(3)
Low immunogenicity (patient self-derived EVs reduced the adaptive immune system activation);
(4)
Specific lipids and proteins, such as CD55 and CD59, which stabilize EVs in bodily fluids [52,175], stimulate membrane fusion between cells;
(5)
Tropism to target specific cells and tissues [176].
EVs have been evaluated as a drug delivery vehicle for diverse therapeutic cargos, including both small molecules (e.g., doxorubicin, curcumin, etc.) and macromolecules (i.e., RNA, DNA, and proteins). There are different EV loading strategies, such as ex vivo extracellular EV loading and in vitro intracellular loading during EV biogenesis [177].
Several drugs, like doxorubicin, withferin A, and celastrol, delivered by Exo and compared to free drugs administration, improved anti-tumor effects in in vivo models, such as colon adenocarcinoma and breast cancer [178]. Furthermore, mixing curcumin with Exo improved the bioavailability and anti-inflammatory efficacy of this drug in a model of LPS(Lipopolysaccharide)-induced septic shock [179].
Regarding to macromolecule delivery, different miRNA and silencing RNA were loaded in EVs from different cells [3,180]. For example, Lunavat et al. demonstrated that Exo delivered cMyc silencing RNA reduced cMyc transcript and induced apoptosis in mouse lymphoma cells [180].

4.2.4. Storage Conditions and Bio-Distribution of EVs

Nowadays, there are no protocols for EV manipulation for therapeutically purposes. EV purification and storage processes, in fact, need to be standardized in order to make them more reproducible and safe and to preserve EV functional properties. It has been demonstrated that purification methods of EVs can dramatically affect their integrity, influencing their subsequent bio-distribution in vivo [181,182]. Furthermore, the natural matrix of EVs might interfere with their integrity. EVs need to be stored as a “pure” pool for therapeutic applications [182]. A large number of solvents and buffers such as water, Phosphate Buffered Saline (PBS), Dimethyl sulfoxide (DMSO), and glycerol have been tested for EV storage; among them, the last two induced a full or partial lyses of EVs [183] while PBS was widely used but it interfered with EV quantification based on single particle detection [184]. Recently, Lener et al. suggested storing EVs in isotonic buffers to prevent pH shifts also during the freezing and thawing procedures [182]. In addition, concerning the storage temperature, it seems that EVs integrity is more preserved at −80 and −20 °C than at −190 or at 4 °C [183,185]. However, in consideration of the wide spectrum of potential EV-based applications, ad hoc purification and storage protocols need to be developed.

5. Conclusions

In HM, EVs are important intercellular “communicators”. They are involved in the tumor auto-sustaining, in making the stroma “stronger” in supporting malignancy, in inducing tumor drug resistance, and in immune system suppression. In addition, EVs could be potential biomarkers in the context of HM liquid biopsy and are interesting therapeutic tools thanks to their abundance in biologic fluids, to their natural capacity to protect molecular cargo, and, finally, to the possibility of engineering them.
Overall, several questions such as the lack of EV population specific identity and cell origin, the insufficient information about their biology and activity in disease and health, the lack of standardized isolation methods, and the need an implementation of EVs as drug delivery platform, still remain open. Recently, an EV-TRACK platform was created to recruit all EV biological and technical information [186] in order to render more transparent the field of EVs.

Acknowledgments

The study was supported by Current Research funds of Italian Ministry of Health.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

ABCA3ATP-binding cassette member A3
AgAntigen
GvHDGraft versus Host Disease
ALIXALG-2-interacting protein X
ALLAcute Lymphoblastic Leukemia
AMLAcute Myeloid Leukemia
APO2LApo2 Ligand
BLAdult T-Cell Leukemia/Lymphoma Burkitt’s Lymphoma
BMBone Marrow
BMSCsBone Marrow Stromal Cells
c-SRCProto-oncogene tyrosine-protein kinase Src
CCL2C-C Motif Chemokine Ligand 2
clBcl-xLCleaved Bcl-xL
CLLChronic Lymphocytic
CMLChronic Myeloid Leukemia
CXCR4Chemokine receptor type 4
DCsDendritic Cells
DKK1Dickkopf-1
DMADimethyl Amiloride
ECMExtracellular Matrix
EMMPRINExtracellular Matrix Metalloproteinase Inducer
EVsExtracellular Vesicles
ExoExosomes
FasLFas Ligand
fibroFibronectin
FLT3fms-Like Tyrosine Kinase 3
GAL3Galectin-3
HIF-1Hypoxia-Inducible Factor 1
HLHodgkin Lymphoma
HMHematological Malignancy
HPCsHematopoietic Progenitor Cells
HPS-70Heat Shock Protein 70
HRSHodgkin and Reed Stemberg
HSHeparan Sulphate
HSCsHematopoietic Stem Cells
HSCTHematopoietic Stem Cell Transplantation
HSPCsHematopoietic Stem and Progenitor Cells
hTERTEnzyme Telomerase
ICAM1Intercellular Adhesion Molecule-1
IGF-IRInsulin-like Growth Factor-I Receptor
ILInterleukin
ITDInternal Tandem Duplication
MDSMyelodysplastic Syndrome
MDSCsMyeloid Derived Suppressor Cells
MHCMajor Histocompatibility Complex
MksMegakaryocytes
MMMultiple Myeloma
MMP4Matrix metallopeptidase 4
MMP9Matrix metallopeptidase 9
MPNMyeloproliferative Neoplasm
MSCsMesenchymal Stem/Stromal Cells
MVsMicrovesicles
NFkBNuclear Factor kappaB
NHLNon-Hodgkin’s Lymphoma
NKG2DNatural-Killer Group-2 Member-D
NPM1Nucleophosmin 1
OCsOsteoclasts
OVOverall Survival
PFSProgression Free Survival
SCsStem Cells
sHSSurface Heparan Sulphate
TCRT-cell Receptor
TFTissue Factor
TGFTumor Growth Factor
Th1T helper 1
VEGFVascular Endothelial Growth Factor
WMWaldenstrom’s Macroglobulinemia
ZNF224Zinc Finger Protein 224

References

  1. Maas, S.L.; Breakefield, X.O.; Weaver, A.M. Extracellular vesicles: Unique intercellular delivery vehicles. Trends Cell Biol. 2017, 27, 172–188. [Google Scholar] [CrossRef] [PubMed]
  2. Cocucci, E.; Meldolesi, J. Ectosomes and exosomes: Shedding the confusion between extracellular vesicles. Trends Cell Biol. 2015, 25, 364–372. [Google Scholar] [CrossRef] [PubMed]
  3. Vader, P.; Breakefield, X.O.; Wood, M.J. Extracellular vesicles: Emerging targets for cancer therapy. Trends Mol. Med. 2014, 20, 385–393. [Google Scholar] [CrossRef] [PubMed]
  4. Minciacchi, V.R.; Freeman, M.R.; di Vizio, D. Extracellular vesicles in cancer: Exosomes, microvesicles and the emerging role of large oncosomes. Semin. Cell Dev. Biol. 2015, 40, 41–51. [Google Scholar] [CrossRef] [PubMed]
  5. Meehan, B.; Rak, J.; di Vizio, D. Oncosomes-large and small: What are they, where they came from? J. Extracell. Vesicles 2016, 5, 33109. [Google Scholar] [CrossRef] [PubMed]
  6. Zaborowski, M.P.; Balaj, L.; Breakefield, X.O.; Lai, C.P. Extracellular vesicles: Composition, biological relevance, and methods of study. Bioscience 2015, 65, 783–797. [Google Scholar] [CrossRef] [PubMed]
  7. Skotland, T.; Sandvig, K.; Llorente, A. Lipids in exosomes: Current knowledge and the way forward. Prog. Lipid Res. 2017, 66, 30–41. [Google Scholar] [CrossRef] [PubMed]
  8. Kim, K.M.; Abdelmohsen, K.; Mustapic, M.; Kapogiannis, D.; Gorospe, M. RNA in extracellular vesicles. Wiley Interdiscip. Rev. RNA 2017. [Google Scholar] [CrossRef] [PubMed]
  9. Raposo, G.; Stoorvogel, W. Extracellular vesicles: Exosomes, microvesicles, and friends. J. Cell Biol. 2013, 200, 373–383. [Google Scholar] [CrossRef] [PubMed]
  10. Bellingham, S.A.; Coleman, B.M.; Hill, A.F. Small RNA deep sequencing reveals a distinct miRNA signature released in exosomes from prion-infected neuronal cells. Nucleic Acids Res. 2012, 40, 10937–10949. [Google Scholar] [CrossRef] [PubMed]
  11. Nolte-’t Hoen, E.N.; Buermans, H.P.; Waasdorp, M.; Stoorvogel, W.; Wauben, M.H.; ’t Hoen, P.A. Deep sequencing of RNA from immune cell-derived vesicles uncovers the selective incorporation of small non-coding RNA biotypes with potential regulatory functions. Nucleic Acids Res. 2012, 40, 9272–9285. [Google Scholar] [CrossRef] [PubMed]
  12. Mateescu, B.; Kowal, E.J.; van Balkom, B.W.; Bartel, S.; Bhattacharyya, S.N.; Buzas, E.I.; Buck, A.H.; de Candia, P.; Chow, F.W.; Das, S.; et al. Obstacles and opportunities in the functional analysis of extracellular vesicle RNA—An ISEV position paper. J. Extracell. Vesicles 2017, 6, 1286095. [Google Scholar] [CrossRef] [PubMed]
  13. Falchi, A.M.; Sogos, V.; Saba, F.; Piras, M.; Congiu, T.; Piludu, M. Astrocytes shed large membrane vesicles that contain mitochondria, lipid droplets and ATP. Histochem. Cell Biol. 2013, 139, 221–231. [Google Scholar] [CrossRef] [PubMed]
  14. Kim, D.K.; Lee, J.; Kim, S.R.; Choi, D.S.; Yoon, Y.J.; Kim, J.H.; Go, G.; Nhung, D.; Hong, K.; Jang, S.C.; et al. EVpedia: A community web portal for extracellular vesicles research. Bioinformatics 2015, 31, 933–939. [Google Scholar] [CrossRef] [PubMed]
  15. Kalra, H.; Simpson, R.J.; Ji, H.; Aikawa, E.; Altevogt, P.; Askenase, P.; Bond, V.C.; Borras, F.E.; Breakefield, X.; Budnik, V.; et al. Vesiclepedia: A compendium for extracellular vesicles with continuous community annotation. PLoS Biol. 2012, 10, e1001450. [Google Scholar] [CrossRef] [PubMed]
  16. Keerthikumar, S.; Chisanga, D.; Ariyaratne, D.; Al Saffar, H.; Anand, S.; Zhao, K.; Samuel, M.; Pathan, M.; Jois, M.; Chilamkurti, N.; et al. Exocarta: A web-based compendium of exosomal cargo. J. Mol. Biol. 2016, 428, 688–692. [Google Scholar] [CrossRef] [PubMed]
  17. Dozio, V.; Sanchez, J.C. Characterisation of extracellular vesicle-subsets derived from brain endothelial cells and analysis of their protein cargo modulation after TNF exposure. J. Extracell. Vesicles 2017, 6, 1302705. [Google Scholar] [CrossRef] [PubMed]
  18. Yanez-Mo, M.; Siljander, P.R.; Andreu, Z.; Zavec, A.B.; Borras, F.E.; Buzas, E.I.; Buzas, K.; Casal, E.; Cappello, F.; Carvalho, J.; et al. Biological properties of extracellular vesicles and their physiological functions. J. Extracell. Vesicles 2015, 4, 27066. [Google Scholar] [CrossRef] [PubMed]
  19. Witas, R.; Chaput, D.; Khan, H.; Stevens, S.M., Jr.; Kang, D. Isolation and proteomic analysis of microvesicles and exosomes from HT22 cells and primary neurons. Methods Mol. Biol. 2017, 1598, 255–267. [Google Scholar] [PubMed]
  20. Dostert, G.; Mesure, B.; Menu, P.; Velot, E. How do mesenchymal stem cells influence or are influenced by microenvironment through extracellular vesicles communication? Front. Cell Dev. Biol. 2017, 5, 6. [Google Scholar] [CrossRef] [PubMed]
  21. Naito, Y.; Yoshioka, Y.; Yamamoto, Y.; Ochiya, T. How cancer cells dictate their microenvironment: Present roles of extracellular vesicles. Cell. Mol. Life Sci. 2017, 74, 697–713. [Google Scholar] [CrossRef] [PubMed]
  22. Kucharzewska, P.; Christianson, H.C.; Welch, J.E.; Svensson, K.J.; Fredlund, E.; Ringner, M.; Morgelin, M.; Bourseau-Guilmain, E.; Bengzon, J.; Belting, M. Exosomes reflect the hypoxic status of glioma cells and mediate hypoxia-dependent activation of vascular cells during tumor development. Proc. Natl. Acad. Sci. USA 2013, 110, 7312–7317. [Google Scholar] [CrossRef] [PubMed]
  23. Rice, G.E.; Scholz-Romero, K.; Sweeney, E.; Peiris, H.; Kobayashi, M.; Duncombe, G.; Mitchell, M.D.; Salomon, C. The effect of glucose on the release and bioactivity of exosomes from first trimester trophoblast cells. J. Clin. Endocrinol. Metab. 2015, 100, E1280–E1288. [Google Scholar] [CrossRef] [PubMed]
  24. Turturici, G.; Tinnirello, R.; Sconzo, G.; Geraci, F. Extracellular membrane vesicles as a mechanism of cell-to-cell communication: Advantages and disadvantages. Am. J. Phys. Cell Phys. 2014, 306, C621–C633. [Google Scholar] [CrossRef] [PubMed]
  25. Aubertin, K.; Silva, A.K.; Luciani, N.; Espinosa, A.; Djemat, A.; Charue, D.; Gallet, F.; Blanc-Brude, O.; Wilhelm, C. Massive release of extracellular vesicles from cancer cells after photodynamic treatment or chemotherapy. Sci. Rep. 2016, 6, 35376. [Google Scholar] [CrossRef] [PubMed]
  26. Prada, I.; Meldolesi, J. Binding and fusion of extracellular vesicles to the plasma membrane of their cell targets. Int. J. Mol. Sci. 2016, 17, 1296. [Google Scholar] [CrossRef] [PubMed]
  27. Mulcahy, L.A.; Pink, R.C.; Carter, D.R. Routes and mechanisms of extracellular vesicle uptake. J. Extracell. Vesicles 2014, 3, 24641. [Google Scholar] [CrossRef] [PubMed]
  28. Rappa, G.; Santos, M.F.; Green, T.M.; Karbanova, J.; Hassler, J.; Bai, Y.; Barsky, S.H.; Corbeil, D.; Lorico, A. Nuclear transport of cancer extracellular vesicle-derived biomaterials through nuclear envelope invagination-associated late endosomes. Oncotarget 2017, 8, 14443–14461. [Google Scholar] [CrossRef] [PubMed]
  29. Yuana, Y.; Sturk, A.; Nieuwland, R. Extracellular vesicles in physiological and pathological conditions. Blood Rev. 2013, 27, 31–39. [Google Scholar] [CrossRef] [PubMed]
  30. Valadi, H.; Ekstrom, K.; Bossios, A.; Sjostrand, M.; Lee, J.J.; Lotvall, J.O. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat. Cell Biol. 2007, 9, 654–659. [Google Scholar] [CrossRef] [PubMed]
  31. Gatti, S.; Bruno, S.; Deregibus, M.C.; Sordi, A.; Cantaluppi, V.; Tetta, C.; Camussi, G. Microvesicles derived from human adult mesenchymal stem cells protect against ischaemia-reperfusion-induced acute and chronic kidney injury. Nephrol. Dial. Transplant. 2011, 26, 1474–1483. [Google Scholar] [CrossRef] [PubMed]
  32. Qian, Z.; Shen, Q.; Yang, X.; Qiu, Y.; Zhang, W. The role of extracellular vesicles: An epigenetic view of the cancer microenvironment. BioMed Res. Int. 2015, 2015, 649161. [Google Scholar] [CrossRef] [PubMed]
  33. Andaloussi, S.E.; Mager, I.; Breakefield, X.O.; Wood, M.J. Extracellular vesicles: Biology and emerging therapeutic opportunities. Nat. Rev. Drug Discov. 2013, 12, 347–357. [Google Scholar] [CrossRef] [PubMed]
  34. French, K.C.; Antonyak, M.A.; Cerione, R.A. Extracellular vesicle docking at the cellular port: Extracellular vesicle binding and uptake. Semin. Cell Dev. Biol. 2017. [Google Scholar] [CrossRef] [PubMed]
  35. Cappello, F.; Logozzi, M.; Campanella, C.; Bavisotto, C.C.; Marcilla, A.; Properzi, F.; Fais, S. Exosome levels in human body fluids: A tumor marker by themselves? Eur. J. Pharm. Sci. 2017, 96, 93–98. [Google Scholar] [CrossRef] [PubMed]
  36. Caivano, A.; Laurenzana, I.; de Luca, L.; La Rocca, F.; Simeon, V.; Trino, S.; D’Auria, F.; Traficante, A.; Maietti, M.; Izzo, T.; et al. High serum levels of extracellular vesicles expressing malignancy-related markers are released in patients with various types of hematological neoplastic disorders. Tumour Biol. 2015, 36, 9739–9752. [Google Scholar] [CrossRef] [PubMed]
  37. Ogawa, Y.; Miura, Y.; Harazono, A.; Kanai-Azuma, M.; Akimoto, Y.; Kawakami, H.; Yamaguchi, T.; Toda, T.; Endo, T.; Tsubuki, M.; et al. Proteomic analysis of two types of exosomes in human whole saliva. Biol. Pharm. Bull. 2011, 34, 13–23. [Google Scholar] [CrossRef] [PubMed]
  38. Aalberts, M.; van Dissel-Emiliani, F.M.; van Adrichem, N.P.; van Wijnen, M.; Wauben, M.H.; Stout, T.A.; Stoorvogel, W. Identification of distinct populations of prostasomes that differentially express prostate stem cell antigen, Annexin A1, and GLIPR2 in humans. Biol. Reprod. 2012, 86, 82. [Google Scholar] [CrossRef] [PubMed]
  39. Melnik, B.C.; John, S.M.; Schmitz, G. Milk: An exosomal microRNA transmitter promoting thymic regulatory T cell maturation preventing the development of atopy? J. Transl. Med. 2014, 12, 43. [Google Scholar] [CrossRef] [PubMed]
  40. Kolhe, R.; Hunter, M.; Liu, S.; Jadeja, R.N.; Pundkar, C.; Mondal, A.K.; Mendhe, B.; Drewry, M.; Rojiani, M.V.; Liu, Y.; et al. Gender-specific differential expression of exosomal miRNA in synovial fluid of patients with osteoarthritis. Sci. Rep. 2017, 7, 2029. [Google Scholar] [CrossRef] [PubMed]
  41. Van Balkom, B.W.; Pisitkun, T.; Verhaar, M.C.; Knepper, M.A. Exosomes and the kidney: Prospects for diagnosis and therapy of renal diseases. Kidney Int. 2011, 80, 1138–1145. [Google Scholar] [CrossRef] [PubMed]
  42. Balbi, C.; Piccoli, M.; Barile, L.; Papait, A.; Armirotti, A.; Principi, E.; Reverberi, D.; Pascucci, L.; Becherini, P.; Varesio, L.; et al. First characterization of human amniotic fluid stem cell extracellular vesicles as a powerful paracrine tool endowed with regenerative potential. Stem Cells Transl. Med. 2017, 6, 1340–1355. [Google Scholar] [CrossRef] [PubMed]
  43. Menay, F.; Herschlik, L.; de Toro, J.; Cocozza, F.; Tsacalian, R.; Gravisaco, M.J.; di Sciullo, M.P.; Vendrell, A.; Waldner, C.I.; Mongini, C. Exosomes isolated from ascites of T-cell lymphoma-bearing mice expressing surface CD24 and HSP-90 induce a tumor-specific immune response. Front. Immunol. 2017, 8, 286. [Google Scholar] [CrossRef] [PubMed]
  44. Lee, J.; McKinney, K.Q.; Pavlopoulos, A.J.; Han, M.H.; Kim, S.H.; Kim, H.J.; Hwang, S. Exosomal proteome analysis of cerebrospinal fluid detects biosignatures of neuromyelitis optica and multiple sclerosis. Clin. Chim. Acta 2016, 462, 118–126. [Google Scholar] [CrossRef] [PubMed]
  45. Fais, S.; O’Driscoll, L.; Borras, F.E.; Buzas, E.; Camussi, G.; Cappello, F.; Carvalho, J.; Cordeiro da Silva, A.; del Portillo, H.; El Andaloussi, S.; et al. Evidence-based clinical use of nanoscale extracellular vesicles in nanomedicine. ACS Nano 2016, 10, 3886–3899. [Google Scholar] [CrossRef] [PubMed]
  46. Surman, M.; Stepien, E.; Hoja-Lukowicz, D.; Przybylo, M. Deciphering the role of ectosomes in cancer development and progression: Focus on the proteome. Clin. Exp. Metastasis 2017. [Google Scholar] [CrossRef] [PubMed]
  47. Peinado, H.; Zhang, H.; Matei, I.R.; Costa-Silva, B.; Hoshino, A.; Rodrigues, G.; Psaila, B.; Kaplan, R.N.; Bromberg, J.F.; Kang, Y.; et al. Pre-metastatic niches: Organ-specific homes for metastases. Nat. Rev. Cancer 2017, 17, 302–317. [Google Scholar] [CrossRef] [PubMed]
  48. Whiteside, T.L. Exosomes carrying immunoinhibitory proteins and their role in cancer. Clin. Exp. Immunol. 2017. [Google Scholar] [CrossRef] [PubMed]
  49. Raimondo, S.; Corrado, C.; Raimondi, L.; de Leo, G.; Alessandro, R. Role of extracellular vesicles in hematological malignancies. BioMed Res. Int. 2015, 2015, 821613. [Google Scholar] [CrossRef] [PubMed]
  50. Boyiadzis, M.; Whiteside, T.L. Information transfer by exosomes: A new frontier in hematologic malignancies. Blood Rev. 2015, 29, 281–290. [Google Scholar] [CrossRef] [PubMed]
  51. Zhang, L.; Valencia, C.A.; Dong, B.; Chen, M.; Guan, P.J.; Pan, L. Transfer of micrornas by extracellular membrane microvesicles: A nascent crosstalk model in tumor pathogenesis, especially tumor cell-microenvironment interactions. J. Hematol. Oncol. 2015, 8, 14. [Google Scholar] [CrossRef] [PubMed]
  52. Robbins, P.D.; Morelli, A.E. Regulation of immune responses by extracellular vesicles. Nat. Rev. Immunol. 2014, 14, 195–208. [Google Scholar] [CrossRef] [PubMed]
  53. Nawaz, M.; Fatima, F.; Vallabhaneni, K.C.; Penfornis, P.; Valadi, H.; Ekstrom, K.; Kholia, S.; Whitt, J.D.; Fernandes, J.D.; Pochampally, R.; et al. Extracellular vesicles: Evolving factors in stem cell biology. Stem Cells Int. 2016, 2016, 1073140. [Google Scholar] [CrossRef] [PubMed]
  54. Nomura, S.; Niki, M.; Nisizawa, T.; Tamaki, T.; Shimizu, M. Microparticles as biomarkers of blood coagulation in cancer. Biomark. Cancer 2015, 7, 51–56. [Google Scholar] [CrossRef] [PubMed]
  55. Ratajczak, J.; Wysoczynski, M.; Hayek, F.; Janowska-Wieczorek, A.; Ratajczak, M.Z. Membrane-derived microvesicles: Important and underappreciated mediators of cell-to-cell communication. Leukemia 2006, 20, 1487–1495. [Google Scholar] [CrossRef] [PubMed]
  56. Jiang, J.; Woulfe, D.S.; Papoutsakis, E.T. Shear enhances thrombopoiesis and formation of microparticles that induce megakaryocytic differentiation of stem cells. Blood 2014, 124, 2094–2103. [Google Scholar] [CrossRef] [PubMed]
  57. Lamichhane, T.N.; Sokic, S.; Schardt, J.S.; Raiker, R.S.; Lin, J.W.; Jay, S.M. Emerging roles for extracellular vesicles in tissue engineering and regenerative medicine. Tissue Eng. Part B Rev. 2015, 21, 45–54. [Google Scholar] [CrossRef] [PubMed]
  58. Xin, H.; Li, Y.; Cui, Y.; Yang, J.J.; Zhang, Z.G.; Chopp, M. Systemic administration of exosomes released from mesenchymal stromal cells promote functional recovery and neurovascular plasticity after stroke in rats. J. Cereb. Blood Flow Metab. 2013, 33, 1711–1715. [Google Scholar] [CrossRef] [PubMed]
  59. Lee, Y.; El Andaloussi, S.; Wood, M.J. Exosomes and microvesicles: Extracellular vesicles for genetic information transfer and gene therapy. Hum. Mol. Genet. 2012, 21, R125–R134. [Google Scholar] [CrossRef] [PubMed]
  60. Tannetta, D.; Dragovic, R.; Alyahyaei, Z.; Southcombe, J. Extracellular vesicles and reproduction-promotion of successful pregnancy. Cell. Mol. Immunol. 2014, 11, 548–563. [Google Scholar] [CrossRef] [PubMed]
  61. Clayton, A.; Mitchell, J.P.; Court, J.; Linnane, S.; Mason, M.D.; Tabi, Z. Human tumor-derived exosomes down-modulate NKG2D expression. J. Immunol. 2008, 180, 7249–7258. [Google Scholar] [CrossRef] [PubMed]
  62. Yu, B.; Zhang, X.; Li, X. Exosomes derived from mesenchymal stem cells. Int. J. Mol. Sci. 2014, 15, 4142–4157. [Google Scholar] [CrossRef] [PubMed]
  63. Jong, A.Y.; Wu, C.H.; Li, J.; Sun, J.; Fabbri, M.; Wayne, A.S.; Seeger, R.C. Large-scale isolation and cytotoxicity of extracellular vesicles derived from activated human natural killer cells. J. Extracell. Vesicles 2017, 6, 1294368. [Google Scholar] [PubMed]
  64. Burrello, J.; Monticone, S.; Gai, C.; Gomez, Y.; Kholia, S.; Camussi, G. Stem cell-derived extracellular vesicles and immune-modulation. Front. Cell Dev. Biol. 2016, 4, 83. [Google Scholar] [CrossRef] [PubMed]
  65. Di Trapani, M.; Bassi, G.; Midolo, M.; Gatti, A.; Kamga, P.T.; Cassaro, A.; Carusone, R.; Adamo, A.; Krampera, M. Differential and transferable modulatory effects of mesenchymal stromal cell-derived extracellular vesicles on T, B and NK cell functions. Sci. Rep. 2016, 6, 24120. [Google Scholar] [CrossRef] [PubMed]
  66. Berckmans, R.J.; Sturk, A.; van Tienen, L.M.; Schaap, M.C.; Nieuwland, R. Cell-derived vesicles exposing coagulant tissue factor in saliva. Blood 2011, 117, 3172–3180. [Google Scholar] [CrossRef] [PubMed]
  67. Ng, Y.H.; Rome, S.; Jalabert, A.; Forterre, A.; Singh, H.; Hincks, C.L.; Salamonsen, L.A. Endometrial exosomes/microvesicles in the uterine microenvironment: A new paradigm for embryo-endometrial cross talk at implantation. PLoS ONE 2013, 8, e58502. [Google Scholar] [CrossRef] [PubMed]
  68. Bakhti, M.; Winter, C.; Simons, M. Inhibition of myelin membrane sheath formation by oligodendrocyte-derived exosome-like vesicles. J. Biol. Chem. 2011, 286, 787–796. [Google Scholar] [CrossRef] [PubMed]
  69. Wang, S.; Cesca, F.; Loers, G.; Schweizer, M.; Buck, F.; Benfenati, F.; Schachner, M.; Kleene, R. Synapsin I is an oligomannose-carrying glycoprotein, acts as an oligomannose-binding lectin, and promotes neurite outgrowth and neuronal survival when released via Glia-derived exosomes. J. Neurosci. 2011, 31, 7275–7290. [Google Scholar] [CrossRef] [PubMed]
  70. Mendelson, A.; Frenette, P.S. Hematopoietic stem cell niche maintenance during homeostasis and regeneration. Nat. Med. 2014, 20, 833–846. [Google Scholar] [CrossRef] [PubMed]
  71. Anthony, B.A.; Link, D.C. Regulation of hematopoietic stem cells by bone marrow stromal cells. Trends Immunol. 2014, 35, 32–37. [Google Scholar] [CrossRef] [PubMed]
  72. Javeed, N.; Mukhopadhyay, D. Exosomes and their role in the micro-/macro-environment: A comprehensive review. J. Biomed. Res. 2016, 30, 1–9. [Google Scholar]
  73. Arendt, B.K.; Walters, D.K.; Wu, X.; Tschumper, R.C.; Jelinek, D.F. Multiple myeloma dell-derived microvesicles are enriched in CD147 expression and enhance tumor cell proliferation. Oncotarget 2014, 5, 5686–5699. [Google Scholar] [CrossRef] [PubMed]
  74. Wu, X.-H. Leukemia-derived exosomes induce paracrine and autocrine cell proliferation in pediatric ALL. In Proceedings of the 58th ASH Annual Meeting and Exposition, San Diego, CA, USA, 3–6 December 2016; p. 4080. [Google Scholar]
  75. Milani, G.; Lana, T.; Bresolin, S.; Aveic, S.; Pasto, A.; Frasson, C.; Te Kronnie, G. Expression profiling of circulating microvesicles reveals intercellular transmission of oncogenic pathways. Mol. Cancer Res. 2017. [Google Scholar] [CrossRef] [PubMed]
  76. Patel, S.J.; Darie, C.C.; Clarkson, B.D. Exosome mediated growth effect on the non-growing pre-B acute lymphoblastic leukemia cells at low starting cell density. Am. J. Transl. Res. 2016, 8, 3614–3629. [Google Scholar] [PubMed]
  77. Raimondo, S.; Saieva, L.; Corrado, C.; Fontana, S.; Flugy, A.; Rizzo, A.; de Leo, G.; Alessandro, R. Chronic myeloid leukemia-derived exosomes promote tumor growth through an autocrine mechanism. Cell Commun. Signal. 2015, 13, 8. [Google Scholar] [CrossRef] [PubMed]
  78. Paggetti, J.; Haderk, F.; Seiffert, M.; Janji, B.; Distler, U.; Ammerlaan, W.; Kim, Y.J.; Adam, J.; Lichter, P.; Solary, E.; et al. Exosomes released by chronic lymphocytic leukemia cells induce the transition of stromal cells into cancer-associated fibroblasts. Blood 2015, 126, 1106–1117. [Google Scholar] [CrossRef] [PubMed]
  79. Zhou, S.; Abdouh, M.; Arena, V.; Arena, M.; Arena, G.O. Reprogramming malignant cancer cells toward a benign phenotype following exposure to human embryonic stem cell microenvironment. PLoS ONE 2017, 12, e0169899. [Google Scholar] [CrossRef] [PubMed]
  80. Wang, Y.; Cheng, Q.; Liu, J.; Dong, M. Leukemia stem cell-released microvesicles promote the survival and migration of myeloid leukemia cells and these effects can be inhibited by microRNA34a overexpression. Stem Cells Int. 2016, 2016, 9313425. [Google Scholar] [CrossRef] [PubMed]
  81. Hornick, N.I.; Doron, B.; Abdelhamed, S.; Huan, J.; Harrington, C.A.; Shen, R.; Cambronne, X.A.; Chakkaramakkil Verghese, S.; Kurre, P. AML suppresses hematopoiesis by releasing exosomes that contain microRNAs targeting c-MYB. Sci. Signal. 2016, 9, ra88. [Google Scholar] [CrossRef] [PubMed]
  82. Huan, J.; Hornick, N.I.; Goloviznina, N.A.; Kamimae-Lanning, A.N.; David, L.L.; Wilmarth, P.A.; Mori, T.; Chevillet, J.R.; Narla, A.; Roberts, C.T., Jr.; et al. Coordinate regulation of residual bone marrow function by paracrine trafficking of AML exosomes. Leukemia 2015, 29, 2285–2295. [Google Scholar] [CrossRef] [PubMed]
  83. Razmkhah, F.; Soleimani, M.; Mehrabani, D.; Karimi, M.H.; Amini Kafi-Abad, S.; Ramzi, M.; Iravani Saadi, M.; Kakoui, J. Leukemia microvesicles affect healthy hematopoietic stem cells. Tumour Biol. 2017, 39. [Google Scholar] [CrossRef] [PubMed]
  84. Gu, H.; Chen, C.; Hao, X.; Wang, C.; Zhang, X.; Li, Z.; Shao, H.; Zeng, H.; Yu, Z.; Xie, L.; et al. Sorting protein VPS33B regulates exosomal autocrine signaling to mediate hematopoiesis and leukemogenesis. J. Clin. Investig. 2016, 126, 4537–4553. [Google Scholar] [CrossRef] [PubMed]
  85. Koch, R.; Demant, M.; Aung, T.; Diering, N.; Cicholas, A.; Chapuy, B.; Wenzel, D.; Lahmann, M.; Guntsch, A.; Kiecke, C.; et al. Populational equilibrium through exosome-mediated Wnt signaling in tumor progression of diffuse large B-cell lymphoma. Blood 2014, 123, 2189–2198. [Google Scholar] [CrossRef] [PubMed]
  86. Bruno, S.; Deregibus, M.C.; Camussi, G. The secretome of mesenchymal stromal cells: Role of extracellular vesicles in immunomodulation. Immunol. Lett. 2015, 168, 154–158. [Google Scholar] [CrossRef] [PubMed]
  87. Robbins, P.D.; Dorronsoro, A.; Booker, C.N. Regulation of chronic inflammatory and immune processes by extracellular vesicles. J. Clin. Investig. 2016, 126, 1173–1180. [Google Scholar] [CrossRef] [PubMed]
  88. Busch, A.; Quast, T.; Keller, S.; Kolanus, W.; Knolle, P.; Altevogt, P.; Limmer, A. Transfer of T cell surface molecules to dendritic cells upon CD4+ T cell priming involves two distinct mechanisms. J. Immunol. 2008, 181, 3965–3973. [Google Scholar] [CrossRef] [PubMed]
  89. Hedlund, M.; Nagaeva, O.; Kargl, D.; Baranov, V.; Mincheva-Nilsson, L. Thermal- and oxidative stress causes enhanced release of NKG2D ligand-bearing immunosuppressive exosomes in leukemia/lymphoma T and B cells. PLoS ONE 2011, 6, e16899. [Google Scholar] [CrossRef]
  90. Xie, Y.; Zhang, H.; Li, W.; Deng, Y.; Munegowda, M.A.; Chibbar, R.; Qureshi, M.; Xiang, J. Dendritic cells recruit T cell exosomes via exosomal LFA-1 leading to inhibition of CD8+ CTL responses through downregulation of peptide/MHC class I and Fas ligand-mediated cytotoxicity. J. Immunol. 2010, 185, 5268–5278. [Google Scholar] [CrossRef] [PubMed]
  91. Baier, C.; Fino, A.; Sanchez, C.; Farnault, L.; Rihet, P.; Kahn-Perles, B.; Costello, R.T. Natural killer cells modulation in hematological malignancies. Front. Immunol. 2013, 4, 459. [Google Scholar] [CrossRef] [PubMed]
  92. Reiners, K.S.; Topolar, D.; Henke, A.; Simhadri, V.R.; Kessler, J.; Sauer, M.; Bessler, M.; Hansen, H.P.; Tawadros, S.; Herling, M.; et al. Soluble ligands for NK cell receptors promote evasion of chronic lymphocytic leukemia cells from NK cell anti-tumor activity. Blood 2013, 121, 3658–3665. [Google Scholar] [CrossRef] [PubMed]
  93. Wang, J.; Faict, S.; Maes, K.; de Bruyne, E.; van Valckenborgh, E.; Schots, R.; Vanderkerken, K.; Menu, E. Extracellular vesicle cross-talk in the bone marrow microenvironment: Implications in multiple myeloma. Oncotarget 2016, 7, 38927–38945. [Google Scholar] [PubMed]
  94. Wang, J.; de Veirman, K.; Faict, S.; Frassanito, M.A.; Ribatti, D.; Vacca, A.; Menu, E. Multiple myeloma exosomes establish a favourable bone marrow microenvironment with enhanced angiogenesis and immunosuppression. J. Pathol. 2016, 239, 162–173. [Google Scholar] [CrossRef] [PubMed]
  95. Garg, T.K.; Gann, J.I.; Malaviarachchi, P.A.; Stone, K.; Macleod, V.; Greenway, A.D.; Morgan, G.J. Myeloma-derived exosomes and soluble factors suppress natural killer cell function. Proceedings of 58th ASH Annual Meeting and Exposition, San Diego, CA, USA, 3–6 December 2016; p. 2066. [Google Scholar]
  96. Chillemi, A.; Quarona, V.; Antonioli, L.; Ferrari, D.; Horenstein, A.L.; Malavasi, F. Roles and modalities of ectonucleotidases in remodeling the multiple myeloma niche. Front. Immunol. 2017, 8, 305. [Google Scholar] [CrossRef] [PubMed]
  97. Malavasi, F. CD38 and antibody therapy: What can basic science add? Proceedings of 58th ASH Annual Meeting and Exposition, San Diego, CA, USA, 3–6 December 2016; p. SCI-36. [Google Scholar]
  98. Gutzeit, C.; Nagy, N.; Gentile, M.; Lyberg, K.; Gumz, J.; Vallhov, H.; Puga, I.; Klein, E.; Gabrielsson, S.; Cerutti, A.; et al. Exosomes derived from Burkitt’s lymphoma cell lines induce proliferation, differentiation, and class-switch recombination in B cells. J. Immunol. 2014, 192, 5852–5862. [Google Scholar] [CrossRef] [PubMed]
  99. Hansen, H.P.; Engels, H.M.; Dams, M.; Paes Leme, A.F.; Pauletti, B.A.; Simhadri, V.L.; Durkop, H.; Reiners, K.S.; Barnert, S.; Engert, A.; et al. Protrusion-guided extracellular vesicles mediate CD30 trans-signalling in the microenvironment of Hodgkin’s lymphoma. J. Pathol. 2014, 232, 405–414. [Google Scholar] [CrossRef] [PubMed]
  100. Zhou, J.; Wang, S.; Sun, K.; Chng, W.J. The emerging roles of exosomes in leukemogeneis. Oncotarget 2016, 7, 50698–50707. [Google Scholar] [CrossRef] [PubMed]
  101. Hong, C.S.; Muller, L.; Whiteside, T.L.; Boyiadzis, M. Plasma exosomes as markers of therapeutic response in patients with acute myeloid leukemia. Front. Immunol. 2014, 5, 160. [Google Scholar] [CrossRef] [PubMed]
  102. Roccaro, A.M.; Sacco, A.; Maiso, P.; Azab, A.K.; Tai, Y.T.; Reagan, M.; Azab, F.; Flores, L.M.; Campigotto, F.; Weller, E.; et al. BM mesenchymal stromal cell-derived exosomes facilitate multiple myeloma progression. J. Clin. Investig. 2013, 123, 1542–1555. [Google Scholar] [CrossRef] [PubMed]
  103. Wang, J.; Hendrix, A.; Hernot, S.; Lemaire, M.; de Bruyne, E.; van Valckenborgh, E.; Lahoutte, T.; de Wever, O.; Vanderkerken, K.; Menu, E. Bone marrow stromal cell-derived exosomes as communicators in drug resistance in multiple myeloma cells. Blood 2014, 124, 555–566. [Google Scholar] [CrossRef] [PubMed]
  104. Corrado, C.; Raimondo, S.; Saieva, L.; Flugy, A.M.; 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, 71–76. [Google Scholar] [CrossRef] [PubMed]
  105. Bruno, S.; Collino, F.; Iavello, A.; Camussi, G. Effects of mesenchymal stromal cell-derived extracellular vesicles on tumor growth. Front. Immunol. 2014, 5, 382. [Google Scholar] [CrossRef] [PubMed]
  106. Ghosh, A.K.; Secreto, C.R.; Knox, T.R.; Ding, W.; Mukhopadhyay, D.; Kay, N.E. Circulating microvesicles in B-cell chronic lymphocytic leukemia can stimulate marrow stromal cells: Implications for disease progression. Blood 2010, 115, 1755–1764. [Google Scholar] [CrossRef] [PubMed]
  107. Boysen, J.; Nelson, M.; Magzoub, G.; Maiti, G.P.; Sinha, S.; Goswami, M.; Vesely, S.K.; Shanafelt, T.D.; Kay, N.E.; Ghosh, A.K. Dynamics of microvesicle generation in B-cell chronic lymphocytic leukemia: Implication in disease progression. Leukemia 2017, 31, 350–360. [Google Scholar] [CrossRef] [PubMed]
  108. Farahani, M.; Rubbi, C.; Liu, L.; Slupsky, J.R.; Kalakonda, N. Cll exosomes modulate the transcriptome and behaviour of recipient stromal cells and are selectively enriched in miR-202-3p. PLoS ONE 2015, 10, e0141429. [Google Scholar] [CrossRef] [PubMed]
  109. Fei, F.; Joo, E.J.; Tarighat, S.S.; Schiffer, I.; Paz, H.; Fabbri, M.; Abdel-Azim, H.; Groffen, J.; Heisterkamp, N. B-cell precursor acute lymphoblastic leukemia and stromal cells communicate through galectin-3. Oncotarget 2015, 6, 11378–11394. [Google Scholar] [CrossRef] [PubMed]
  110. Johnson, S.M.; Dempsey, C.; Chadwick, A.; Harrison, S.; Liu, J.; Di, Y.; McGinn, O.J.; Fiorillo, M.; Sotgia, F.; Lisanti, M.P.; et al. Metabolic reprogramming of bone marrow stromal cells by leukemic extracellular vesicles in acute lymphoblastic leukemia. Blood 2016, 128, 453–456. [Google Scholar] [CrossRef] [PubMed]
  111. Umezu, T.; Tadokoro, H.; Azuma, K.; Yoshizawa, S.; Ohyashiki, K.; Ohyashiki, J.H. Exosomal miR-135b shed from hypoxic multiple myeloma cells enhances angiogenesis by targeting factor-inhibiting HIF-1. Blood 2014, 124, 3748–3757. [Google Scholar] [CrossRef] [PubMed]
  112. Liu, Y.; Zhu, X.J.; Zeng, C.; Wu, P.H.; Wang, H.X.; Chen, Z.C.; Li, Q.B. Microvesicles secreted from human multiple myeloma cells promote angiogenesis. Acta Pharm. Sin. 2014, 35, 230–238. [Google Scholar] [CrossRef] [PubMed]
  113. Raimondi, L.; De Luca, A.; Amodio, N.; Manno, M.; Raccosta, S.; Taverna, S.; Bellavia, D.; Naselli, F.; Fontana, S.; Schillaci, O.; et al. Involvement of multiple myeloma cell-derived exosomes in osteoclast differentiation. Oncotarget 2015, 6, 13772–13789. [Google Scholar] [CrossRef] [PubMed]
  114. Vardaki, I.; Sanchez, C.; Fonseca, P.; Olsson, M.; Chioureas, D.; Rassidakis, G.; Ullen, A.; Zhivotovsky, B.; Bjorkholm, M.; Panaretakis, T. Caspase-3-dependent cleavage of Bcl-xL in the stroma exosomes is required for their uptake by hematological malignant cells. Blood 2016, 128, 2655–2665. [Google Scholar] [CrossRef] [PubMed]
  115. El-Saghir, J.; Nassar, F.; Tawil, N.; El-Sabban, M. ATL-derived exosomes modulate mesenchymal stem cells: Potential role in leukemia progression. Retrovirology 2016, 13, 73. [Google Scholar] [CrossRef] [PubMed]
  116. Horiguchi, H.; Kobune, M.; Kikuchi, S.; Yoshida, M.; Murata, M.; Murase, K.; Iyama, S.; Takada, K.; Sato, T.; Ono, K.; et al. Extracellular vesicle miR-7977 is involved in hematopoietic dysfunction of mesenchymal stromal cells via poly(rC) binding protein 1 reduction in myeloid neoplasms. Haematologica 2016, 101, 437–447. [Google Scholar] [CrossRef] [PubMed]
  117. Huan, J.; Hornick, N.I.; Shurtleff, M.J.; Skinner, A.M.; Goloviznina, N.A.; Roberts, C.T., Jr.; Kurre, P. RNA trafficking by acute myelogenous leukemia exosomes. Cancer Res. 2013, 73, 918–929. [Google Scholar] [CrossRef] [PubMed]
  118. Muntion, S.; Ramos, T.L.; Diez-Campelo, M.; Roson, B.; Sanchez-Abarca, L.I.; Misiewicz-Krzeminska, I.; Preciado, S.; Sarasquete, M.E.; de Las Rivas, J.; Gonzalez, M.; et al. Microvesicles from mesenchymal stromal cells are involved in HPC-microenvironment crosstalk in myelodysplastic patients. PLoS ONE 2016, 11, e0146722. [Google Scholar] [CrossRef] [PubMed]
  119. Ramos, T.L.; Sánchez-Abarca, L.I.; Rosón, B. Extracellular vesicles play an important role in intercellular communication between bone marrow stroma and hematopoietic progenitor cells in myeloproliferative neoplasms. In Proceedings of the 58th ASH Annual Meeting and Exposition, San Diego, CA, USA, 3–6 December 2016; p. 1957. [Google Scholar]
  120. Umezu, T.; Ohyashiki, K.; Kuroda, M.; Ohyashiki, J.H. Leukemia cell to endothelial cell communication via exosomal miRNAs. Oncogene 2013, 32, 2747–2755. [Google Scholar] [CrossRef] [PubMed]
  121. Zhu, X.; You, Y.; Li, Q.; Zeng, C.; Fu, F.; Guo, A.; Zhang, H.; Zou, P.; Zhong, Z.; Wang, H.; et al. BCR-ABL1-positive microvesicles transform normal hematopoietic transplants through genomic instability: Implications for donor cell leukemia. Leukemia 2014, 28, 1666–1675. [Google Scholar] [CrossRef] [PubMed]
  122. Gutkin, A.; Uziel, O.; Beery, E.; Nordenberg, J.; Pinchasi, M.; Goldvaser, H.; Henick, S.; Goldberg, M.; Lahav, M. Tumor cells derived exosomes contain hTERT mRNA and transform nonmalignant fibroblasts into telomerase positive cells. Oncotarget 2016, 7, 59173–59188. [Google Scholar] [CrossRef] [PubMed]
  123. Jaiswal, R.K.; Kumar, P.; Yadava, P.K. Telomerase and its extracurricular activities. Cell. Mol. Biol. Lett. 2013, 18, 538–554. [Google Scholar] [CrossRef] [PubMed]
  124. Purushothaman, A.; Bandari, S.K.; Liu, J.; Mobley, J.A.; Brown, E.E.; Sanderson, R.D. Fibronectin on the surface of myeloma cell-derived exosomes mediates exosome-cell interactions. J. Biol. Chem. 2016, 291, 1652–1663. [Google Scholar] [CrossRef] [PubMed]
  125. Redzic, J.S.; Kendrick, A.A.; Bahmed, K.; Dahl, K.D.; Pearson, C.G.; Robinson, W.A.; Robinson, S.E.; Graner, M.W.; Eisenmesser, E.Z. Extracellular vesicles secreted from cancer cell lines stimulate secretion of MMP-9, IL-6, TGF-β1 and EMMPRIN. PLoS ONE 2013, 8, e71225. [Google Scholar] [CrossRef] [PubMed]
  126. Cesi, G.; Walbrecq, G.; Margue, C.; Kreis, S. Transferring intercellular signals and traits between cancer cells: Extracellular vesicles as “homing pigeons”. Cell Commun. Signal. 2016, 14, 13. [Google Scholar] [CrossRef] [PubMed]
  127. Gong, J.; Luk, F.; Jaiswal, R.; George, A.M.; Grau, G.E.; Bebawy, M. Microparticle drug sequestration provides a parallel pathway in the acquisition of cancer drug resistance. Eur. J. Pharm. 2013, 721, 116–125. [Google Scholar] [CrossRef] [PubMed]
  128. Sousa, D.; Lima, R.T.; Vasconcelos, M.H. Intercellular transfer of cancer drug resistance traits by extracellular vesicles. Trends Mol. Med. 2015, 21, 595–608. [Google Scholar] [CrossRef] [PubMed]
  129. Koch, R.; Aung, T.; Vogel, D.; Chapuy, B.; Wenzel, D.; Becker, S.; Sinzig, U.; Venkataramani, V.; von Mach, T.; Jacob, R.; et al. Nuclear trapping through inhibition of exosomal export by indomethacin increases cytostatic efficacy of doxorubicin and pixantrone. Clin. Cancer Res. 2016, 22, 395–404. [Google Scholar] [CrossRef] [PubMed]
  130. D’Arena, G.; Ruggieri, V.; D’Auria, F.; La Rocca, F.; Simeon, V.; Statuto, T.; Caivano, A.; Telesca, D.; del Vecchio, L.; Musto, P. Lenalidomide differently modulates CD20 antigen surface expression on chronic lymphocytic leukemia B-cells. Leuk. Lymphoma 2015, 56, 2458–2459. [Google Scholar] [CrossRef] [PubMed]
  131. Aung, T.; Chapuy, B.; Vogel, D.; Wenzel, D.; Oppermann, M.; Lahmann, M.; Weinhage, T.; Menck, K.; Hupfeld, T.; Koch, R.; et al. Exosomal evasion of humoral immunotherapy in aggressive B-cell lymphoma modulated by ATP-binding cassette transporter A3. Proc. Natl. Acad. Sci. USA 2011, 108, 15336–15341. [Google Scholar] [CrossRef] [PubMed]
  132. Viola, S.; Traer, E.; Huan, J.; Hornick, N.I.; Tyner, J.W.; Agarwal, A.; Loriaux, M.; Johnstone, B.; Kurre, P. Alterations in acute myeloid leukaemia bone marrow stromal cell exosome content coincide with gains in tyrosine kinase inhibitor resistance. Br. J. Haematol. 2016, 172, 983–986. [Google Scholar] [CrossRef] [PubMed]
  133. Wojtuszkiewicz, A.; Schuurhuis, G.J.; Kessler, F.L.; Piersma, S.R.; Knol, J.C.; Pham, T.V.; Jansen, G.; Musters, R.J.; van Meerloo, J.; Assaraf, Y.G.; et al. Exosomes secreted by apoptosis-resistant acute myeloid leukemia (AML) blasts harbor regulatory network proteins potentially involved in antagonism of apoptosis. Mol. Cell. Proteom. 2016, 15, 1281–1298. [Google Scholar] [CrossRef] [PubMed]
  134. Bouvy, C.; Wannez, A.; Chatelain, C.J.; Dogné, J.-M. Transfer of multidrug resistance among acute myeloid leukemia cells via extracellular vesicles and their micrornas cargo. In Proceedings of the 58th ASH Annual Meeting and Exposition, San Diego, CA, USA, 3–6 December 2016; p. 2864. [Google Scholar]
  135. Hansen, H.P.; Trad, A.; Dams, M.; Zigrino, P.; Moss, M.; Tator, M.; Schon, G.; Grenzi, P.C.; Bachurski, D.; Aquino, B.; et al. CD30 on extracellular vesicles from malignant Hodgkin cells supports damaging of CD30 ligand-expressing bystander cells with brentuximab-vedotin, in vitro. Oncotarget 2016, 7, 30523–30535. [Google Scholar] [CrossRef] [PubMed]
  136. Siravegna, G.; Marsoni, S.; Siena, S.; Bardelli, A. Integrating liquid biopsies into the management of cancer. Nat. Rev. Clin. Oncol. 2017. [Google Scholar] [CrossRef] [PubMed]
  137. De Luca, L.; D’Arena, G.; Simeon, V.; Trino, S.; Laurenzana, I.; Caivano, A.; La Rocca, F.; Villani, O.; Mansueto, G.; Deaglio, S.; et al. Characterization and prognostic relevance of circulating microvesicles in chronic lymphocytic leukemia. Leuk. Lymphoma 2017, 58, 1424–1432. [Google Scholar] [CrossRef] [PubMed]
  138. Belov, L.; Matic, K.J.; Hallal, S.; Best, O.G.; Mulligan, S.P.; Christopherson, R.I. Extensive surface protein profiles of extracellular vesicles from cancer cells may provide diagnostic signatures from blood samples. J. Extracell. Vesicles 2016, 5, 25355. [Google Scholar] [CrossRef] [PubMed]
  139. Caivano, A.; La Rocca, F.; Simeon, V.; Girasole, M.; Dinarelli, S.; Laurenzana, I.; de Stradis, A.; de Luca, L.; Trino, S.; Traficante, A.; et al. MicroRNA-155 in serum-derived extracellular vesicles as a potential biomarker for hematologic malignancies—A short report. Cell. Oncol. 2017, 40, 97–103. [Google Scholar] [CrossRef] [PubMed]
  140. Yeh, Y.Y.; Ozer, H.G.; Lehman, A.M.; Maddocks, K.; Yu, L.; Johnson, A.J.; Byrd, J.C. Characterization of CLL exosomes reveals a distinct microRNA signature and enhanced secretion by activation of BCR signaling. Blood 2015, 125, 3297–3305. [Google Scholar] [CrossRef] [PubMed]
  141. Van Eijndhoven, M.A.; Zijlstra, J.M.; Groenewegen, N.J.; Drees, E.E.; van Niele, S.; Baglio, S.R.; Koppers-Lalic, D.; van der Voorn, H.; Libregts, S.F.; Wauben, M.H.; et al. Plasma vesicle miRNAs for therapy response monitoring in Hodgkin lymphoma patients. JCI Insight 2016, 1, e89631. [Google Scholar] [CrossRef] [PubMed]
  142. Bouyssou, J.M.C.; Aljawai, Y.; Yosef, A.; Manier, S.; Rawal, R.; Kokubun, K.; Roccaro, A.M. Profiling of Circulating Exosomes in Patients with Waldenström Macroglobulinemia. In Proceedings of the 58th ASH Annual Meeting and Exposition, San Diego, CA, USA, 3–6 December 2016; Volume 128, p. 2940. [Google Scholar]
  143. Krishnan, S.R.; Luk, F.; Brown, R.D.; Suen, H.; Kwan, Y.; Bebawy, M. Isolation of human CD138+ microparticles from the plasma of patients with multiple myeloma. Neoplasia 2016, 18, 25–32. [Google Scholar] [CrossRef] [PubMed]
  144. Xie, Y.; Bai, O.; Zhang, H.; Yuan, J.; Zong, S.; Chibbar, R.; Slattery, K.; Qureshi, M.; Wei, Y.; Deng, Y.; et al. Membrane-bound HSP70-engineered myeloma cell-derived exosomes stimulate more efficient CD8+ CTL- and NK-mediated antitumour immunity than exosomes released from heat-shocked tumour cells expressing cytoplasmic HSP70. J. Cell. Mol. Med. 2010, 14, 2655–2666. [Google Scholar] [CrossRef] [PubMed]
  145. Harshman, S.W.; Canella, A.; Ciarlariello, P.D.; Agarwal, K.; Branson, O.E.; Rocci, A.; Cordero, H.; Phelps, M.A.; Hade, E.M.; Dubovsky, J.A.; et al. Proteomic characterization of circulating extracellular vesicles identifies novel serum myeloma associated markers. J. Proteom. 2016, 136, 89–98. [Google Scholar] [CrossRef] [PubMed]
  146. Di Noto, G.; Paolini, L.; Zendrini, A.; Radeghieri, A.; Caimi, L.; Ricotta, D. C-src enriched serum microvesicles are generated in malignant plasma cell dyscrasia. PLoS ONE 2013, 8, e70811. [Google Scholar] [CrossRef] [PubMed]
  147. Manier, S.; Liu, C.J.; Avet-Loiseau, H.; Park, J.; Shi, J.; Campigotto, F.; Salem, K.Z.; Huynh, D.; Glavey, S.V.; Rivotto, B.; et al. Prognostic role of circulating exosomal mirnas in multiple myeloma. Blood 2017, 129, 2429–2436. [Google Scholar] [CrossRef] [PubMed]
  148. Feng, D.Q.; Huang, B.; Li, J.; Liu, J.; Chen, X.M.; Xu, Y.M.; Chen, X.; Zhang, H.B.; Hu, L.H.; Wang, X.Z. Selective miRNA expression profile in chronic myeloid leukemia K562 cell-derived exosomes. Asian Pac. J. Cancer Prev. 2013, 14, 7501–7508. [Google Scholar] [CrossRef] [PubMed]
  149. Ohyashiki, K.; Umezu, T.; Katagiri, S.; Kobayashi, C.; Azuma, K.; Tauchi, T.; Okabe, S.; Fukuoka, Y.; Ohyashiki, J.H. Downregulation of plasma miR-215 in chronic myeloid leukemia patients with successful discontinuation of imatinib. Int. J. Mol. Sci. 2016, 17, 570. [Google Scholar] [CrossRef] [PubMed]
  150. Hong, C.S.; Muller, L.; Boyiadzis, M.; Whiteside, T.L. Isolation and characterization of CD34+ blast-derived exosomes in acute myeloid leukemia. PLoS ONE 2014, 9, e103310. [Google Scholar] [CrossRef] [PubMed]
  151. Szczepanski, M.J.; Szajnik, M.; Welsh, A.; Whiteside, T.L.; Boyiadzis, M. Blast-derived microvesicles in sera from patients with acute myeloid leukemia suppress natural killer cell function via membrane-associated transforming growth factor-β1. Haematologica 2011, 96, 1302–1309. [Google Scholar] [CrossRef] [PubMed]
  152. Boyiadzis, M.; Whiteside, T.L. Plasma-derived exosomes in acute myeloid leukemia for detection of minimal residual disease: Are we ready? Expert Rev. Mol. Diagn. 2016, 16, 623–629. [Google Scholar] [CrossRef] [PubMed]
  153. Lia, G.; Brunello, L.; Omedè, P.; Astolfi, M.; Drandi, D.; Giaccone, L.; Evangelista, A. Extracellular Vesicles as Potential Biomarker for Acute Graft-Versus-Host-Disease. In Proceedings of the 58th ASH Annual Meeting and Exposition, San Diego, CA, USA, 3–6 December 2016; p. 2239. [Google Scholar]
  154. Aoki, J.; Ohashi, K.; Mitsuhashi, M.; Murakami, T.; Oakes, M.; Kobayashi, T.; Doki, N.; Kakihana, K.; Sakamaki, H. Posttransplantation bone marrow assessment by quantifying hematopoietic cell-derived mRNAs in plasma exosomes/microvesicles. Clin. Chem. 2014, 60, 675–682. [Google Scholar] [CrossRef] [PubMed]
  155. H Rashed, M.; Bayraktar, E.; K Helal, G.; Abd-Ellah, M.F.; Amero, P.; Chavez-Reyes, A.; Rodriguez-Aguayo, C. Exosomes: From garbage bins to promising therapeutic targets. Int. J. Mol. Sci. 2017, 18, 538. [Google Scholar] [CrossRef] [PubMed]
  156. Zhang, B.; Yin, Y.; Lai, R.C.; Lim, S.K. Immunotherapeutic potential of extracellular vesicles. Front. Immunol. 2014, 5, 518. [Google Scholar] [CrossRef] [PubMed]
  157. Syn, N.L.; Wang, L.; Chow, E.K.; Lim, C.T.; Goh, B.C. Exosomes in cancer nanomedicine and immunotherapy: Prospects and challenges. Trends Biotechnol. 2017. [Google Scholar] [CrossRef] [PubMed]
  158. Yao, Y.; Wang, C.; Wei, W.; Shen, C.; Deng, X.; Chen, L.; Ma, L.; Hao, S. Dendritic cells pulsed with leukemia cell-derived exosomes more efficiently induce antileukemic immunities. PLoS ONE 2014, 9, e91463. [Google Scholar] [CrossRef] [PubMed]
  159. Yao, Y.; Wei, W.; Sun, J.; Chen, L.; Deng, X.; Ma, L.; Hao, S. Proteomic analysis of exosomes derived from human lymphoma cells. Eur. J. Med. Res. 2015, 20, 8. [Google Scholar] [CrossRef] [PubMed]
  160. Shen, C.; Hao, S.G.; Zhao, C.X.; Zhu, J.; Wang, C. Antileukaemia immunity: Effect of exosomes against NB4 acute promyelocytic leukaemia cells. J. Int. Med. Res. 2011, 39, 740–747. [Google Scholar] [CrossRef] [PubMed]
  161. Qazi, K.R.; Gehrmann, U.; Domange Jordo, E.; Karlsson, M.C.; Gabrielsson, S. Antigen-loaded exosomes alone induce Th1-type memory through a B-cell-dependent mechanism. Blood 2009, 113, 2673–2683. [Google Scholar] [CrossRef] [PubMed]
  162. Gehrmann, U.; Naslund, T.I.; Hiltbrunner, S.; Larssen, P.; Gabrielsson, S. Harnessing the exosome-induced immune response for cancer immunotherapy. Semin. Cancer Biol. 2014, 28, 58–67. [Google Scholar] [CrossRef] [PubMed]
  163. Rani, S.; Ryan, A.E.; Griffin, M.D.; Ritter, T. Mesenchymal stem cell-derived extracellular vesicles: Toward cell-free therapeutic applications. Mol. Ther. 2015, 23, 812–823. [Google Scholar] [CrossRef] [PubMed]
  164. De Luca, L.; Trino, S.; Laurenzana, I.; Lamorte, D.; Caivano, A.; del Vecchio, L.; Musto, P. Mesenchymal stem cell derived extracellular vesicles: A role in hematopoietic transplantation? Int. J. Mol. Sci. 2017, 18, 1022. [Google Scholar] [CrossRef] [PubMed]
  165. De Luca, L.; Trino, S.; Laurenzana, I.; Simeon, V.; Calice, G.; Raimondo, S.; Podesta, M.; Santodirocco, M.; di Mauro, L.; La Rocca, F.; et al. miRNAs and piRNAs from bone marrow mesenchymal stem cell extracellular vesicles induce cell survival and inhibit cell differentiation of cord blood hematopoietic stem cells: A new insight in transplantation. Oncotarget 2016, 7, 6676–6692. [Google Scholar] [PubMed]
  166. Kordelas, L.; Rebmann, V.; Ludwig, A.K.; Radtke, S.; Ruesing, J.; Doeppner, T.R.; Epple, M.; Horn, P.A.; Beelen, D.W.; Giebel, B. MSC-derived exosomes: A novel tool to treat therapy-refractory graft-versus-host disease. Leukemia 2014, 28, 970–973. [Google Scholar] [CrossRef] [PubMed]
  167. Baietti, M.F.; Zhang, Z.; Mortier, E.; Melchior, A.; Degeest, G.; Geeraerts, A.; Ivarsson, Y.; Depoortere, F.; Coomans, C.; Vermeiren, E.; et al. Syndecan-syntenin-alix regulates the biogenesis of exosomes. Nat. Cell Biol. 2012, 14, 677–685. [Google Scholar] [CrossRef] [PubMed]
  168. Marchaland, J.; Cali, C.; Voglmaier, S.M.; Li, H.; Regazzi, R.; Edwards, R.H.; Bezzi, P. Fast subplasma membrane Ca2+ transients control exo-endocytosis of synaptic-like microvesicles in astrocytes. J. Neurosci. 2008, 28, 9122–9132. [Google Scholar] [CrossRef] [PubMed]
  169. Chalmin, F.; Ladoire, S.; Mignot, G.; Vincent, J.; Bruchard, M.; Remy-Martin, J.P.; Boireau, W.; Rouleau, A.; Simon, B.; Lanneau, D.; et al. Membrane-associated HSP72 from tumor-derived exosomes mediates STAT3-dependent immunosuppressive function of mouse and human myeloid-derived suppressor cells. J. Clin. Investig. 2010, 120, 457–471. [Google Scholar] [CrossRef] [PubMed]
  170. Christianson, H.C.; Svensson, K.J.; van Kuppevelt, T.H.; Li, J.P.; Belting, M. Cancer cell exosomes depend on cell-surface heparan sulfate proteoglycans for their internalization and functional activity. Proc. Natl. Acad. Sci. USA 2013, 110, 17380–17385. [Google Scholar] [CrossRef] [PubMed]
  171. Lima, L.G.; Chammas, R.; Monteiro, R.Q.; Moreira, M.E.; Barcinski, M.A. Tumor-derived microvesicles modulate the establishment of metastatic melanoma in a phosphatidylserine-dependent manner. Cancer Lett. 2009, 283, 168–175. [Google Scholar] [CrossRef] [PubMed]
  172. Shtam, T.A.; Kovalev, R.A.; Varfolomeeva, E.Y.; Makarov, E.M.; Kil, Y.V.; Filatov, M.V. Exosomes are natural carriers of exogenous siRNA to human cells in vitro. Cell Commun. Signal. 2013, 11, 88. [Google Scholar] [CrossRef] [PubMed]
  173. Nishida-Aoki, N.; Tominaga, N.; Takeshita, F.; Sonoda, H.; Yoshioka, Y.; Ochiya, T. Disruption of circulating extracellular vesicles as a novel therapeutic strategy against cancer metastasis. Mol. Ther. 2017, 25, 181–191. [Google Scholar] [CrossRef] [PubMed]
  174. Gustafson, H.H.; Holt-Casper, D.; Grainger, D.W.; Ghandehari, H. Nanoparticle uptake: The phagocyte problem. Nano Today 2015, 10, 487–510. [Google Scholar] [CrossRef] [PubMed]
  175. Teissier, E.; Pecheur, E.I. Lipids as modulators of membrane fusion mediated by viral fusion proteins. Eur. Biophys. J. 2007, 36, 887–899. [Google Scholar] [CrossRef] [PubMed]
  176. Hoshino, A.; Costa-Silva, B.; Shen, T.L.; Rodrigues, G.; Hashimoto, A.; Tesic Mark, M.; Molina, H.; Kohsaka, S.; di Giannatale, A.; Ceder, S.; et al. Tumour exosome integrins determine organotropic metastasis. Nature 2015, 527, 329–335. [Google Scholar] [CrossRef] [PubMed]
  177. Barile, L.; Vassalli, G. Exosomes: Therapy delivery tools and biomarkers of diseases. Pharm. Ther. 2017, 174, 63–78. [Google Scholar] [CrossRef] [PubMed]
  178. Tian, Y.; Li, S.; Song, J.; Ji, T.; Zhu, M.; Anderson, G.J.; Wei, J.; Nie, G. A doxorubicin delivery platform using engineered natural membrane vesicle exosomes for targeted tumor therapy. Biomaterials 2014, 35, 2383–2390. [Google Scholar] [CrossRef] [PubMed]
  179. Sun, D.; Zhuang, X.; Xiang, X.; Liu, Y.; Zhang, S.; Liu, C.; Barnes, S.; Grizzle, W.; Miller, D.; Zhang, H.G. A novel nanoparticle drug delivery system: The anti-inflammatory activity of curcumin is enhanced when encapsulated in exosomes. Mol. Ther. 2010, 18, 1606–1614. [Google Scholar] [CrossRef] [PubMed]
  180. Lunavat, T.R.; Jang, S.C.; Nilsson, L.; Park, H.T.; Repiska, G.; Lasser, C.; Nilsson, J.A.; Gho, Y.S.; Lotvall, J. RNAi delivery by exosome-mimetic nanovesicles-implications for targeting c-Myc in cancer. Biomaterials 2016, 102, 231–238. [Google Scholar] [CrossRef] [PubMed]
  181. Wiklander, O.P.; Nordin, J.Z.; O’Loughlin, A.; Gustafsson, Y.; Corso, G.; Mager, I.; Vader, P.; Lee, Y.; Sork, H.; Seow, Y.; et al. Extracellular vesicle in vivo biodistribution is determined by cell source, route of administration and targeting. J. Extracell. Vesicles 2015, 4, 26316. [Google Scholar] [CrossRef] [PubMed]
  182. Lener, T.; Gimona, M.; Aigner, L.; Borger, V.; Buzas, E.; Camussi, G.; Chaput, N.; Chatterjee, D.; Court, F.A.; Del Portillo, H.A.; et al. Applying extracellular vesicles based therapeutics in clinical trials—An ISEV position paper. J. Extracell. Vesicles 2015, 4, 30087. [Google Scholar] [CrossRef] [PubMed]
  183. Lorincz, A.M.; Timar, C.I.; Marosvari, K.A.; Veres, D.S.; Otrokocsi, L.; Kittel, A.; Ligeti, E. Effect of storage on physical and functional properties of extracellular vesicles derived from neutrophilic granulocytes. J. Extracell. Vesicles 2014, 3, 25465. [Google Scholar] [CrossRef] [PubMed]
  184. Sokolova, V.; Ludwig, A.K.; Hornung, S.; Rotan, O.; Horn, P.A.; Epple, M.; Giebel, B. Characterisation of exosomes derived from human cells by nanoparticle tracking analysis and scanning electron microscopy. Colloids Surf. B Biointerfaces 2011, 87, 146–150. [Google Scholar] [CrossRef] [PubMed]
  185. Kalra, H.; Adda, C.G.; Liem, M.; Ang, C.S.; Mechler, A.; Simpson, R.J.; Hulett, M.D.; Mathivanan, S. Comparative proteomics evaluation of plasma exosome isolation techniques and assessment of the stability of exosomes in normal human blood plasma. Proteomics 2013, 13, 3354–3364. [Google Scholar] [CrossRef] [PubMed]
  186. Consortium, E.-T.; van Deun, J.; Mestdagh, P.; Agostinis, P.; Akay, O.; Anand, S.; Anckaert, J.; Martinez, Z.A.; Baetens, T.; Beghein, E.; et al. EV-TRACK: Transparent reporting and centralizing knowledge in extracellular vesicle research. Nat. Methods 2017, 14, 228–232. [Google Scholar]
Figure 1. A summary drawing of the role of Extracellular vesicles (EVs) in bone marrow (BM) microenvironment of different hematological malignancies (HMs). BM cells release EVs which can act, through both autocrine and paracrine (gray arrows), on recipient cells. Tumor EVs can render themselves more aggressive and “stronger” stroma in supporting the tumors. In addition, neoplasm EVs interact with extracellular matrix to promote tumor invasion. Moreover, they enhance angiogenesis, inhibit the immune system, and induce a suppression of hematopoietic stem and progenitor cell (HSPC) functions and a stem cell (SC) malignant transformation. MMP9: Matrix metallopeptidase 9; IL6: interleukine 6; MDSC: Myeloid Derived Suppressor Cells; MSCs: mesenchymal stem/stromal cells; NK: natural killer cells.
Figure 1. A summary drawing of the role of Extracellular vesicles (EVs) in bone marrow (BM) microenvironment of different hematological malignancies (HMs). BM cells release EVs which can act, through both autocrine and paracrine (gray arrows), on recipient cells. Tumor EVs can render themselves more aggressive and “stronger” stroma in supporting the tumors. In addition, neoplasm EVs interact with extracellular matrix to promote tumor invasion. Moreover, they enhance angiogenesis, inhibit the immune system, and induce a suppression of hematopoietic stem and progenitor cell (HSPC) functions and a stem cell (SC) malignant transformation. MMP9: Matrix metallopeptidase 9; IL6: interleukine 6; MDSC: Myeloid Derived Suppressor Cells; MSCs: mesenchymal stem/stromal cells; NK: natural killer cells.
Ijms 18 01183 g001
Table 1. EVS as biomarkers in HMs.
Table 1. EVS as biomarkers in HMs.
DiseaseBiofluidsEv TypeTumor BiomarkersReferences
CLLSerum, plasmaMVsCD19, CD37, CD52[36,107,137]
SerumMVs and ExomiR155[139,140]
HLSerumMVsCD30[36]
Serum, plasmaMVs and ExomiR155, miR127, miR21, miR24[139,141]
WMSerumMVs and ExomiR155[139,140]
MMSerum, plasma, cell mediumMVs and ExoCD38, CD138, CD147, CD44, cSRC, ZNF224, CCL2, IL6[36,73,101,143,144]
PlasmaMVs and ExomiR15, miR16, miR135, let-7b, miR181[145,147]
MM patients after allograftSerumEVsCD46, CD31, CD106, CD25[153]
CMLSerumMVsCD13[36]
PlasmaExomiR 215[149]
PMFSerumMVsCD13[36]
MDSSerumMVsCD13[36]
AMLSerum, plasma, cell mediumMVs and ExoCD34, CD117, CD33, CD13, TGFβ1[36,132,150,151]
Serum, plasmaEVsmiR155[139,152]
Cell mediumExoFLT3-ITD, NPM1, IGF-IR, CXCR4 mRNAs[117]
HM patients after allograftPlasmaEVsDEFA3, HBB, ITGA2B, ITGB3 mRNAs[154]
CLL = chronic lymphocytic leukemia; HL = Hodgkin lymphoma; WM = Waldenstrom’s macroglobulinemia; MM = multiple myeloma; CML = chronic myeloid leukemia; PMF = Primary myelofibrosis; MDS = myelodysplastic syndrome; AML = acute myeloid leukemia; FLT3-ITD = FMS-like tyrosine Kinase 3 with internal tandem duplication; NPM1 = Nucleophosmin 1; IGF-IR = Insulin-like growth factor 1receptor; CXCR4 = Chemokine receptor-4; DEFA3 = Defensin,a 3, neutrophil-specific; HBB = Hemoglobin b; ITGA2B = Integrin a 2; ITGB3 = Integrin b 3, platelet glycoprotein IIIA.

Share and Cite

MDPI and ACS Style

Caivano, A.; La Rocca, F.; Laurenzana, I.; Trino, S.; De Luca, L.; Lamorte, D.; Del Vecchio, L.; Musto, P. Extracellular Vesicles in Hematological Malignancies: From Biology to Therapy. Int. J. Mol. Sci. 2017, 18, 1183. https://doi.org/10.3390/ijms18061183

AMA Style

Caivano A, La Rocca F, Laurenzana I, Trino S, De Luca L, Lamorte D, Del Vecchio L, Musto P. Extracellular Vesicles in Hematological Malignancies: From Biology to Therapy. International Journal of Molecular Sciences. 2017; 18(6):1183. https://doi.org/10.3390/ijms18061183

Chicago/Turabian Style

Caivano, Antonella, Francesco La Rocca, Ilaria Laurenzana, Stefania Trino, Luciana De Luca, Daniela Lamorte, Luigi Del Vecchio, and Pellegrino Musto. 2017. "Extracellular Vesicles in Hematological Malignancies: From Biology to Therapy" International Journal of Molecular Sciences 18, no. 6: 1183. https://doi.org/10.3390/ijms18061183

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop