Next Article in Journal
NR3C1 Glucocorticoid Receptor Gene Polymorphisms Are Associated with Membranous and IgA Nephropathies
Next Article in Special Issue
Molecular Mediators of RNA Loading into Extracellular Vesicles
Previous Article in Journal
Bench Research Informed by GWAS Results
Previous Article in Special Issue
Molecular Determinants for RNA Release into Extracellular Vesicles
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Role of Extracellular Vesicle-Based Cell-to-Cell Communication in Multiple Myeloma Progression

by
Ilaria Saltarella
1,†,
Aurelia Lamanuzzi
1,†,
Benedetta Apollonio
1,
Vanessa Desantis
1,2,
Giulia Bartoli
1,
Angelo Vacca
1 and
Maria Antonia Frassanito
3,*
1
Department of Biomedical Sciences and Human Oncology, Unit of Internal Medicine “Guido Baccelli”, University of Bari Medical School, Piazza Giulio Cesare 11, 70124 Bari, Italy
2
Department of Biomedical Sciences and Human Oncology, Pharmacology Section, University of Bari Medical School, Piazza Giulio Cesare 11, 70124 Bari, Italy
3
Department of Biomedical Sciences and Human Oncology, Unit of General Pathology, University of Bari “Aldo Moro”, 70124 Bari, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Cells 2021, 10(11), 3185; https://doi.org/10.3390/cells10113185
Submission received: 11 October 2021 / Revised: 4 November 2021 / Accepted: 12 November 2021 / Published: 16 November 2021
(This article belongs to the Special Issue Extracellular Vesicle-Associated Non-Coding RNAs)

Abstract

:
Multiple myeloma (MM) progression closely depends on the bidirectional crosstalk between tumor cells and the surrounding microenvironment, which leads to the creation of a tumor supportive niche. Extracellular vesicles (EVs) have emerged as key players in the pathological interplay between the malignant clone and near/distal bone marrow (BM) cells through their biologically active cargo. Here, we describe the role of EVs derived from MM and BM cells in reprogramming the tumor microenvironment and in fostering bone disease, angiogenesis, immunosuppression, drug resistance, and, ultimately, tumor progression. We also examine the emerging role of EVs as new therapeutic agents for the treatment of MM, and their potential use as clinical biomarkers for early diagnosis, disease classification, and therapy monitoring.

1. Introduction

Multiple myeloma (MM) is a hematologic neoplasm characterized by the expansion of monoclonal malignant plasma cells (MM cells) that accumulate in the bone marrow (BM). MM usually progresses from the preneoplastic and asymptomatic phases of monoclonal gammopathy of undetermined significance (MGUS) and smoldering myeloma (SMM) to symptomatic MM [1]. Despite the development of new drugs, MM is still an incurable malignancy. Several studies have shown that the crosstalk between cancer cells and BM microenvironment plays a crucial role in disease progression and drug resistance, with the tumor being able to hijack surrounding cells through various mechanisms of cell-to-cell communication, including autocrine/paracrine signaling and the release of extracellular vesicles (EVs) [2].
Based on their size, EVs were first classified into apoptotic/necrotic bodies (size 1000–5000 nm), microvesicles (200–1000 nm) and exosomes (30–150 nm) [3]. Regarding apoptotic/necrotic bodies originate from apoptotic and necrotic cells that break up into multiple vesicles, microvesicles are pinched off from the plasma membrane, while exosomes are formed by an active, energy-dependent and organized process [4]. Currently, the MISEV18 guidelines recommend the use of the term “EVs” for a heterogeneous family of vesicles (both small and medium/large vesicles) delimited by a lipid bilayer that are released from all eukaryotic cells in various body fluids (plasma, urine, saliva, and cerebrospinal fluid) [5].
Although exosomes were initially regarded as cellular waste with no biological activities [6], it is now widely accepted that EVs play an essential role in intercellular communication via their cargo, capable of reprogramming near and distal recipient cells [3,7].
EVs share multiple molecules with the parental cell, providing useful information about the genetic, molecular, and functional properties of the cell of origin [8,9]. Tumor cells produce and release a greater amount of EVs (tumor-derived EVs) than healthy proliferating cells [8]. Although some proteins involved in EVs biogenesis (i.e., p53 [10] and Rab proteins [11]) can be altered during tumor progression, the mechanisms leading to increased production and secretion of EVs by tumor cells are still unclear.
In recent years, several studies have documented the central role of EVs as key regulators of the bidirectional crosstalk between tumor cells and BM microenvironment during disease progression.
Here we provide an overview of the role of EVs derived from MM and BM cells in disease progression, drug resistance, bone resorption, immunosuppression and angiogenesis. We also discuss the emerging role of EVs as new therapeutic agents for the treatment of MM, and their potential use as clinical biomarkers for early diagnosis, disease classification, and therapy monitoring.

2. EVs Biogenesis

Two main pathways of biogenesis have been described for the generation of EVs: (i) direct budding of the plasma membrane with consequent release of EVs, classically referred to as microvesicles [4,12]; and (ii) endosomal biogenesis, an active and complex mechanism that ultimately leads to the production of EVs, classically referred to as exosomes (Figure 1A) [13].
Endosomal biogenesis begins with the inward budding of plasma membrane, giving rise to an early endosome, which in turn undergoes multiple invaginations leading to the formation of intraluminal vesicles (ILVs) incorporated into a mature multivesicular body (MVB) (Figure 1A). As a result, the ILVs express on their surface various proteins belonging to the plasma membrane of the cell of origin, and they enclose the parental cytosol.
EVs contain endosome-related membrane transport and fusion proteins that contribute to EVs biogenesis (annexins, flotillin, GTPases, and lipids, such as ceramides and sphingomyelin) [14,15], tetraspanins (CD9, CD63, CD81, and CD82) used as EVs markers [5,16], and proteins belonging to the accessory endosomal sorting complex required for transport (ESCRT) [17,18]. Activation of ESCRT pathway is responsible for EVs sorting, packaging, and transport [18]. Both the ESCRT machinery and its auxiliary proteins (Alix, Vps4, and VTA1) are localized on the cytoplasmic side of the endosomal membrane and are involved in the sorting and ubiquitination of proteins into ILVs. In particular, ESCRT-0 recognizes and binds to ubiquitinated proteins, while ESCRT-I interacts with ESCRT-II, forming another complex. The two complexes bind to ESCRT-III, which is involved in the budding process, and induces the detachment of ILVs from the MVB membrane by activating the ATPase protein Vps4 [19,20]. Alix associated with ESCRT proteins is involved in endosomal membrane budding and abscission, and cargo selection via syndecan binding [21,22].
In addition, two ESCRT-independent pathways have been identified: (i) tetraspanin-enriched domains (TEMs)-dependent, where tetraspanins act as sorting machinery [23], and (ii) sphingolipids-dependent, where ceramides trigger spontaneous negative curvature of the endosomal membrane [24].
At the end of the maturation process, the newly formed MVBs can fuse either with lysosomes for degradation or with plasma membranes for the release of EVs into the extracellular space [25] (Figure 1A). The release process is regulated by transmembrane proteins (e.g., Rab, N-ethylmaleimide-sensitive factor-NSF, and soluble NSF-attachment protein-SNAP) and membrane complexes (e.g., SNAP-attachment protein receptor-SNARE) [11,26,27]. SNARE proteins, localized on different intracellular membranes, are usually responsible for the fusion of different cellular compartments [28], and trigger the release of EVs through the formation of invadopodia [12,29].
When released into the extracellular space, EVs interact with local cells or enter the blood and lymphatic circulation, allowing interaction with distant cells [30].
Several mechanisms regulate the interaction of EVs with target cells, including fusion, internalization, micropinocytosis [31], and phagocytosis [32]. Interestingly, fusion of EVs is regulated by the same proteins involved in their release, i.e., SNAREs and Rab proteins, while internalization of EVs requires clathrin- and caveolin-mediated endocytosis [33,34], integrins [35], and membrane-associated lipids, including cholesterol, sphingolipids, and glycosylphosphatidylinositol (GPI)-anchored proteins [36]. Moreover, surface antigens of EVs (i.e., MHC-I/-II, tetraspanins, Fas ligand (FasL), and TNF related apoptosis inducing ligand (TRAIL)) can interact with their specific receptors on target cells and activate intracellular signaling [4,37,38] (Figure 1B).

3. EVs Cargo

The biological activity of EVs is closely dependent on their cargo, such as proteins, lipids, and nucleic acids, which are responsible for target cell reprogramming and provide important information about the parental cells by mirroring their cytoplasmic content (Figure 1B).
EVs proteins include endosomal, cytosolic, and nuclear proteins [39], involved in EVs biogenesis, transport, and fusion (e.g., HSP70, HSP90), integrins and adhesion molecules that play a role in target cells binding [40,41,42]. In addition, under pathological conditions, other molecules may also be included in the cargo of EVs. For example, tumor-derived EVs contain specific oncoproteins (HER family [43]) and immunosuppressive molecules (Fas-L, TRAIL, and immune checkpoints such as PD-L1 [44,45]) that promote neoplastic progression and immune evasion (Figure 1B).
Recent studies have shown that EVs can be ‘decorated’ with additional proteins besides the canonical exofacial molecules, collectively referred to as ‘corona’ [46,47,48,49,50,51,52]. Buzas et al. [46] showed that EVs associate with extracellular matrix proteins, complement, immunoglobulins, coagulation factors, lipoproteins, nucleic acids, and thiol-reactive antioxidants [46]. Furthermore, Toth et al. [51] confirmed the interaction between plasma proteins and EVs, and identified a number of proteins (ApoA1, ApoB, ApoC3, ApoE, complement factors 3 and 4B, fibrinogen α-chain, immunoglobulin heavy constant γ2 and γ4 chains) that form a ‘corona’ around EVs in blood plasma [51].
EVs also carry nucleic acids as DNAs (single-stranded, double-stranded, genomic, mitochondrial and reverse-transcribed complementary DNA) [53,54,55,56] and RNAs, including mRNAs and non-coding RNAs (microRNAs (miRs), long non-coding RNAs (lncRNAs), circular RNAs (circRNAs), tRNA-derived small RNA fragments, and YRNAs; for recent and detailed review see [57]) (Figure 1B). The lipid bilayer of EVs protects RNA from degradation and increases its stability. Several studies have highlighted the important role of the RNA content of EVs in modulating the transcriptome of target cells and in shaping the tumor microenvironment [58,59,60,61].
Since the molecular and genetic cargo of EVs partially reflects the composition of parental cells and EVs can be easily obtained from patients’ serum/plasma or other body fluids, several studies suggest that tumor-derived EVs may be useful for cancer diagnosis, prognosis and drug responses (see ‘Diagnostic potential’ section) [52,62,63,64].

4. EVs in MM Progression and Drug Resistance

MM depends on the BM milieu that co-evolves with the tumor and promotes cancer cell proliferation, drug resistance, and disease progression by inducing bone resorption, immunosuppression, and angiogenesis [65] (Figure 2). Among BM stromal cells (BMSCs), fibroblasts (FBs) contribute to the formation of the tumor supportive niche and promote disease progression and resistance to bortezomib-induced apoptosis [66,67]. Frassanito et al. [68] have shown that EVs promote the bidirectional crosstalk between MM cells and FBs in the tumor microenvironment [68].
Specifically, MM-derived EVs (MM-EVs) contain the WWC2 protein, which activates the Hippo pathway, which in turn triggers a de novo synthesis of miR-27b-3p and miR-214-3p into recipient FBs [68]. Moreover, MM-EVs activate FBs by increasing the expression of fibroblast activation protein (FAP) and alpha-smooth muscle actin (αSMA) promoting their transformation into cancer associated fibroblasts (CAFs). Taken together, these data support evidence that MM-EVs can directly reprogram surrounding cells in the tumor microenvironment during disease progression. Similarly, Cheng et al. [69] demonstrated that EVs derived from the OPM2 MM cell line contain high levels of miR-21 and miR-146, which trigger cell proliferation, IL-6 release, and conversion of mesenchymal stromal cells (MSCs) into CAFs, as documented by overexpression of FAP, αSMA, and SDF-1 [69]. Knockdown of both miRs in MSCs prevents their transformation and IL-6 release. Moreover, MM-EVs induce secretion of IL-6 by BMSCs via activation of APE1/NF-kB pathway [70]. Finally, EVs from both plasma and MM cells of patients promote cell proliferation, migration, and adhesion of recipient BMSCs by transferring multiple oncogenic factors highlighting the role of MM-EVs in modifying the BM niche [71].
On the other hand, MSCs-EVs from MM patients and healthy donors (HD) differentially affect the phenotype and the intracellular signaling of MM cells: MM MSCs-EVs enhance viability, proliferation, and migration of MM cells, while HD MSCs-EVs do not [72].
EVs from BMSCs support myeloma progression by delivering miRs, pro-survival cytokines and chemokines [73]. Roccaro et al. [74] demonstrated that MSCs-EVs contain oncogenic proteins, cytokines and adhesion molecules, including IL-6, MCP-1, junction plakoglobin, fibronectin, and express low level of the tumor-suppressive miR-15a compared to EVs derived from healthy MSCs, which promote tumor growth and dissemination of MM cells in vivo [74]. Recently, proteomic analysis of MSCs-EV cargo revealed that MM MSCs-EVs heterogeneously express higher levels of ribosomal proteins than their HD counterpart. EVs enriched with ribosomal proteins promote the viability and proliferation of MM cells [72,75]. Moreover, EVs derived from the BM of 5T33 mice contain pro-tumor proteins, including MCP-1, MIP-1α, and SDF-1, which promote proliferation, survival, and resistance to bortezomib-induced apoptosis of RPMI8226 cells via activation of various intracellular pathways, including Jun N-terminal kinase, p38, p53, and Akt [76]. Similarly, our group has demonstrated the involvement of MM FBs-EVs in drug resistance. Specifically, MM cells co-cultured with MM FBs-EVs selectively overexpress miR-214-3p and miR-5100, which trigger MM cells proliferation and resistance to bortezomib-induced apoptosis via activation of intracellular pathways involved in cell apoptosis and proliferation, i.e., MAPK, AKT/mTOR, p53 (unpublished data).

5. EVs in MM Bone Resorption

Bone disease is a common feature of MM and it is associated with severe pain, pathologic fracture, spinal cord compression, vertebral collapse, and hypercalcemia [77]. It is characterized by the disruption of the physiological balance between osteoblasts (OBs) and osteoclasts (OCs), leading to a decrease in bone formation and an increase in bone resorption resulting in lytic lesions [77].
MM cells are important regulators of bone disease via cell-to-cell contact with BMSCs, which activate intracellular pathways involved in the regulation of OCs and OBs activities [78]. Recent studies demonstrated the involvement of MM-EVs in bone disease via the transfer of miRNAs, lncRNAs, and cytokines.
Raimondi et al. [79] demonstrated the ability of MM-EVs to trigger differentiation of OCs and promote bone resorption in both mice and humans. Specifically, MM-EVs induce the migration of OCs precursors (pOCs) by increasing the expression of CXCR4, and promoting their differentiation into multinuclear OCs capable to excavate bone lacunae. The uptake of EVs in pOCs induces the expression of markers of bone resorption activity, including tartrate-resistant acid phosphatase (TRAP), proteolytic enzyme cathepsin K (CTSK) and matrix metalloproteinases 9 (MMP-9). In addition, MM-EVs promote the survival of pOCs and inhibit their apoptosis [79].
Other studies documented the osteoclastogenic effect of EVs via direct and indirect mechanisms [80,81]. Proteomic analysis of MM1.s-EVs revealed that they directly contribute to bone resorption by containing several proteins involved in the unfolded protein response (UPR) pathway via the IRE1α/XBP1 axis. Selective inhibition of IRE1α partially counteracts EVs-induced OCs differentiation and bone resorption [80]. In addition, MM1.s-EVs contain the EGFR ligand amphiregulin (AREG), which directly activates the EGFR pathway in recipient pOCs cells, triggering OC differentiation. AREG-enriched EVs from MM cells activate OCs differentiation indirectly via MSCs. Indeed, MSCs uptake MM-EVs and release the pro-osteoclastogenic cytokine IL-8, which inhibits OBs differentiation by reducing the expression of OBs markers, i.e., alkaline phosphatase (ALP), osteocalcin (OCN), collagen type I alpha 1 (COL1A1) [81]. A recent study has shown that MM-EVs can contribute to bone resorption and lytic lesions by delivering the pro-inflammatory cytokine IL-32. IL-32 represents a poor prognostic factor that correlates with osteoclast activity and lytic lesions in MM patients and negatively with progression free survival [82].
RUNX2-AS1 is a natural antisense transcript derived from intron 7 of the RUNX2 gene, the major transcription factor associated with OB differentiation [83,84]. RUNX2-AS1 binds to RUNX2 pre-mRNA and affects the splicing of RUNX2, reducing its expression [83,84]. MM-EVs contain the lncRNA RUNX2-AS1, which decreases the expression of RUNX2 in MSCs as well as their osteogenic potential by regulating the expression of osteopontin (OPN) [85]. The osteolytic effect of MM-EVs was confirmed in vivo using the 5TGM1 mouse model: EVs significantly increase differentiation of OCs and improve their resorptive activity by reducing trabecular bone volume. Interestingly, analysis of EVs from the RPMI8226 MM cells revealed the presence of OPN (personal data not shown), which may be involved in bone resorption and support disease progression via angiogenesis [83].
Moreover, 5TGM1-derived EVs express the Wnt ligand DKK-1. DKK-1 downregulates Wnt signaling and negatively regulates OBs differentiation by decreasing the expression of master regulator genes for OBs differentiation, including RUNX2, Osterix, Col1A1 and ALP [78]. Liu et al. [70] demonstrated that MM-EVs inhibit the differentiation of BMSCs in OB by decreasing the mRNA levels of Ocn, Osterix, and Runx2. Co-culture of BMSCs with MM-EVs significantly reduced the number of OBs as well as their activity [70]. Moreover, EVs support bone disease by reprogramming the expression profile of OBs and OCs through miRs transfer. Raimondo et al. [86] demonstrated the overexpression of miR-129-5p in EVs from BM plasma of MM patients compared to SMM patients. Interestingly, co-cultures of MSCs with MM-EVs determine an increase in miR-129-5p, which inhibits the transcription factor Sp1, a positive modulator OB differentiation, and of its target gene Alp [86].
Collectively, these data suggest that the miRs cargo of MM-EVs may support bone disease during MM progression (Figure 2).

6. EVs in Immunosuppression

Immune dysregulation is a hallmark of MM and has been associated to disease progression from MGUS to symptomatic MM [87]. Immune dysfunction in MM patients includes quantitative, phenotypic, and functional abnormalities in dendritic cells (DCs), T cells, natural killer cells (NK), T regulatory cells (Treg), and myeloid-derived suppressor cells (MDSCs) [87,88,89,90,91]. MDSCs represent an immature myeloid cell population capable of repressing both innate and adaptive immunity [92]. MM patients exhibit increased levels of peripheral and BM MDSCs, suggesting that they have a role in immune escape of MM cells [93]. Uptake of MM- and BMSCs-EVs supports the expansion of murine MDSCs [94,95]. Both BMSCs- and MM-EVs activate Stat1 and Stat3 in target MDSCs and increase the levels of anti-apoptotic proteins Mcl-1 and Bcl-XL, demonstrating that EVs directly modulate the survival and apoptotic pathways of MDSCs [94,95]. Accordingly, inhibition of Stat1/3 pathways reduced MDSCs survival. In vivo studies confirmed the ability of BMSCs-EVs to maintain BM immunosuppression by inducing the expansion of MDSCs, which in turn suppress T cell proliferation and function through the release of nitric oxide (NO). Analysis of BM immune cells from naïve mice inoculated with 5T33MMvt EVs showed an increase of MDSCs, immature myeloid cells and eosinophils resembling the BM immune features of 5T33MM tumor bearing mice, implying that MM cells trigger BM immunosuppression via the release of EVs [94,95].
In addition, EVs contribute to BM immunosuppression by producing the immunosuppressive molecule adenosine (ADO). ADO is produced by adenosinergic ectoenzymes, i.e., CD39/CD73 or CD38/CD203a (PC-1)/CD73 from ATP or NAD+, respectively [96]. Morandi et al. [96] have shown that EVs from MM patients express higher surface levels of adenosinergic ectoenzymes, including CD38, CD39, CD73, and CD203a and, thus, resemble the phenotype of parental cells. In vitro analysis of EVs from MM, MGUS and SMM patients showed that MM-EVs efficiently metabolize ADO precursors, resulting in increased ADO production compared to MGUS and SMM. Interestingly, increased ADO levels as well as expression of CD38 on MM-EVs support resistance to the anti-CD38 monoclonal antibody treatment [97,98].
Thus, EVs from MM patients exert their immunomodulatory effect by maintaining the expansion of MDSCs and the production of the immunosuppressive molecule ADO (Figure 2).

7. EVs in Angiogenesis

The pleiotropic effect of EVs in MM also affects angiogenesis. Several studies documented the ability of BM-EVs to promote BM angiogenesis by transferring cytokines and miRs [94,99,100] (Figure 2).
MM-EVs from human RPMI8226 and murine 5T33MMvt cells contain pro-angiogenic proteins, i.e., angiogenin, HGF, MMP-9, VEGF, PDGF and SDF1-1α ([95] and personal data). They modulate cell signaling pathways, including Stat3, Jnk, and p53 in recipient BMSCs and STR10 endothelial cells (ECs), resulting in enhanced cell viability. Murine MM-EVs trigger angiogenesis in the chorioallantoic membrane (CAM) assay by promoting capillary and vessel formation [95].
The BM milieu of MM patients is characterized by an aberrant expression of HIF-1α and by the presence of hypoxic regions that further stimulate angiogenesis [101,102]. Umezu et al. [99] demonstrated the strong ability of hypoxia-resistant MM cells to induce angiogenesis by using an in vitro model that reproduced the hypoxic conditions of MM BM stroma. Hypoxia resistant MM cells release higher levels of EVs than normal MM cells and promote in vitro and in vivo angiogenesis through the delivery of miR-135b. In turn, miR-135b promotes the activation of hypoxia-induced angiogenic response by downregulating the factor inhibiting HIF-1 (FIH-1), a protein that binds to and inhibits the function of HIF-1α. Inhibition of FIH-1 leads to an increase in HIF-1α levels in target ECs and promotion of angiogenesis under hypoxic conditions [99]. Moreover, miR-let-7c embedded into MSCs-EVs promotes angiogenesis via M2 polarization of BM-resident and peripheral blood macrophages by inducing overexpression of CD206 and of miR-let-7c. M2 macrophages in turn trigger angiogenesis in vitro in a miR-let-7c–dependent manner [103].
Furthermore, Li et al. [100] have shown that EVs derived from MM cells, as well as circulating EVs, contain high levels of the small PIWI-interacting RNA 823 (piRNA-823), which correlates with disease progression, poor prognosis, and angiogenesis. Transfer of piRNA-823 into recipient EA.hy926 ECs prevents their apoptosis and increases ECs proliferation, invasion, angiogenesis, and tumor growth in vivo. Based on these results, it can be speculated that piRNA-823 may represent a therapeutic target to counteract angiogenesis and tumor growth.

8. Diagnostic Potential of EVs

Several studies have aimed to identify non-invasive biomarkers that are useful for disease diagnosis, prediction of therapeutic intervention, prognosis, and monitoring of disease progression.
The different EVs content among MM, MGUS, and HD patients, as well as their systemic circulation in all body fluids, including plasma, serum, and urine, make EVs suitable for non-invasive liquid biopsy.
Manier et al. [62] first suggested the prognostic significance of circulating EVs miR-let7-b and miR-18a in patients with MM [62]. Moreover, comparison of serum-derived miRs from MM, SMM and HD patients identified miR-140-3p, miR-20a-5p, miR-185-5p, and miR-4741 as oncogenic miRs highly correlated with malignant transformation, suggesting that they may serve as circulating biomarker of disease progression [63]. Similarly, Sedlarikova et al. [104] identified lncRNA PRINS as differentially expressed in MGUS, MM patients and HD, indicating the diagnostic potential of lncPRINS [104].
Circulating EVs might also provide information on the efficacy of therapeutic strategies and could be used as biomarkers for primary and acquired drug resistance. Zhang et al. [105] found a correlation between downregulation of EVs miRs (miR-16-5p, miR-15a-5p and miR-20a-5p, miR-17-5p) and bortezomib resistance, indicating the potential use of EVs as circulating biomarkers for therapy management of MM patients [105]. Moreover, MSCs-EVs contain high levels of PSMA3 mRNA and lncRNA PSMA3-AS1, which mediate resistance to proteasome inhibitors and correlate to poor prognosis [106].
Due to the great diagnostic potential of EVs, highly sensitive methods, such as flow cytometry, digital PCR, microscopic imaging, and Raman spectroscopy, have been used to identify of EVs and define their cargo [107].
Marchisio et al. [108] described a method based on polychromatic flow cytometry to identify circulating EVs derived from different cells, i.e., ECs, leukocytes and platelets [108]. Applying this approach to MM could help to define the origin of circulating EVs based on the expression of specific surface antigens, i.e., CD38 for MM-EVs [98], CD31 for ECs-EVs [108]. Therefore, an increase in CD38+ and CD31+ EVs in the peripheral blood could predict the progression of MGUS to MM as well as the therapeutic response; thus, avoiding the BM biopsies of patients.
In addition, Raman spectroscopy and surface-enhanced Raman scattering were used to distinguish EVs from MGUS, asymptomatic and symptomatic MM based on their spectra. Multivariate analysis of these spectra should improve stratification of MM patients and their follow-up and risk of progression [109]. Recently, Laurenzana et al. [110] presented a new method for isolating EVs from peripheral blood in a single centrifugation step. They applied this method to characterize EVs from HD and MM patients by analyzing the size, concentration, and genetic content of EVs. The authors demonstrated increased levels of CD38+CD138+ EVs in the sera of MM patients. Interestingly, the number of CD38+CD138+ EVs correlates with plasmacytosis and disease stage [110].
Overall, these studies highlight the promising role of EVs as novel biomarkers for distinguishing clinical disease phase, monitoring MM progression and patient outcome, and predicting the efficacy of therapeutic strategies.

9. Therapeutic Perspective

Since EVs are known to play an important role in MM progression, several studies have focused on inhibiting EVs-mediated crosstalk by blocking the release and/or uptake of EVs to prevent their tumor-supportive activity [111] (Figure 3A).
Thompson et al. [112] demonstrated that heparanase induces release of EVs by tumor cells and affects their protein cargo by increasing the levels of syndecan-1, VEGF, and HGF [112]. Inhibition of heparanase activity through SST0001 suppresses MM cell growth and angiogenesis [113] (Figure 3A). The sphingolipid C6 ceramide affects MM cell proliferation, apoptosis, and EV release, and increases the levels of tumor-suppressive miRs, including miR-202, miR-16, miR-29b, and miR-15a embedded in MM-EVs [114].
GW4869, a neutral sphingomyelinase that prevents EVs budding from the plasma membrane [115], is cytotoxic for several MM cell lines and primary MM cells by binding phosphatidylserine expressed on their surface. Moreover, GW4869 is able to retard the growth of MM cells expressing phosphatidylserine in a mouse xenograft model [115]. Treatment of 5TGM1 mice with GW4869 reduces osteolysis by increasing OB activity and synergizes with bortezomib, leading to a reduction in tumor growth and microvessel density [116] (Figure 3A).
Inhibition of MM-EVs endocytosis by BMSCs by various endocytosis inhibitors (i.e., heparin, dynasore and omeprazole), prevents activation of intracellular pathways, i.e., STAT1, STAT3, and ERK1/2, which are associated with cell survival and proliferation [117]. Moreover, Tu et al. [118] have shown that EVs confer resistance to bortezomib. Blockade of endocytosis by chemical inhibitors reduces the internalization of BMSCs-EVs and sensitizes MM cells to bortezomib treatment in vitro and in vivo [118]. Other studies have explored the possibility of enhancing the EVs immune activity [119,120]. EVs derived from membrane-bound HSP70- or TNFα-engineered MM cells stimulate the anti-MM immune response in wild-type BALB/c mice by inducing DCs maturation [119,120]. Recently, Wang et al. [121] demonstrated that apoptotic EVs (apoEVs) derived from MSCs induce MM cells apoptosis and inhibit tumor growth by Fas pathway activation, suggesting a potential use of apoEVs as an anti-MM therapy (Figure 3B) [121].
Finally, since EVs transfer proteins and nucleic acids to neighboring and distant cells, they can be used as new therapeutic tools to deliver drugs, miRs, or lncRNAs to tumor cells [122]. To date, there are no anti-MM therapies based on EVs as a therapeutic agent. However, ongoing studies are investigating the possibility of encapsulating chemotherapeutic agents, e.g., doxorubicin, to ensure more specific drug delivery to tumor cells without side effects [123]. This strategy could be used for various anti-MM drugs, including monoclonal antibodies (e.g., anti-CD38, anti-SLAMF7 mAbs) to improve their efficacy [124]. Synthetic nanoparticles formulation has been developed for miRs delivery. The miR-34 based-therapy MRX34 was the first miR therapy in its class to be approved for the treatment of solid and hematological cancers, including MM. MRX34 delivers a miR-34 mimic encapsulated in a liposomal nanoparticle that causes miR-34 overexpression. Although the clinical trial was discontinued due to immune-mediated adverse effects, it provides evidence for the concept of miR delivery by EVs for cancer therapy [125].

10. Concluding Remarks

EVs are important mediators of intercellular communication by facilitating bidirectional crosstalk between cancer cells and tumor microenvironments in several diseases, including MM. The aberrant cargo packaged in the EVs isolated from the MM BM milieu is able to directly reprogram recipient cells and promote bone disease, angiogenesis, immunosuppression, and drug resistance, ultimately leading to tumor progression (Table 1).
The different EV cargo observed in pre-malignant and malignant stages, as well as the presence of EVs in almost all body fluids, makes them ideal candidates for non-invasive liquid biopsy to differentiate MGUS from overt MM, monitor MM progression, predict patient outcome, and predict the efficacy of therapeutic strategies.
Although the pathogenic role of EVs in MM is now widely documented, their application as a novel drug delivery tool is still in its infancy, and further investigation is needed to identify therapeutic strategies based on EVs.

Author Contributions

Conceptualization, I.S., A.L., A.V. and M.A.F.; writing of original draft, I.S., A.L., B.A. and M.A.F.; supervision of the manuscript, V.D., G.B. and A.V. All authors have read and agreed to the published version of the manuscript.

Funding

INNOLABS–POR Puglia FESR-FSE 2014-2020 (Telemielolab) to AV.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

This work was supported by INNOLABS–POR Puglia FESR-FSE 2014-2020 (Telemielolab) to A.V.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Korde, N.; Kristinsson, S.Y.; Landgren, O. Monoclonal gammopathy of undetermined significance (MGUS) and smoldering multiple myeloma (SMM): Novel biological insights and development of early treatment strategies. Blood 2011, 117, 5573–5581. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Di Marzo, L.; Desantis, V.; Solimando, A.G.; Ruggieri, S.; Annese, T.; Nico, B.; Fumarulo, R.; Vacca, A.; Frassanito, M.A. Microenvironment drug resistance in multiple myeloma: Emerging new players. Oncotarget 2016, 7, 60698–60711. [Google Scholar] [CrossRef] [Green Version]
  3. Brinton, L.T.; Sloane, H.S.; Kester, M.; Kelly, K.A. Formation and role of exosomes in cancer. Cell. Mol. Life Sci. 2015, 72, 659–671. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Thery, C.; Ostrowski, M.; Segura, E. Membrane vesicles as conveyors of immune responses. Nat. Rev. Immunol. 2009, 9, 581–593. [Google Scholar] [CrossRef]
  5. Thery, C.; Witwer, K.W.; Aikawa, E.; Alcaraz, M.J.; Anderson, J.D.; Andriantsitohaina, R.; Antoniou, A.; Arab, T.; Archer, F.; Atkin-Smith, G.K.; et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): A position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J. Extracell. Vesicles 2018, 7, 1535750. [Google Scholar] [CrossRef] [Green Version]
  6. Johnstone, R.M.; Adam, M.; Hammond, J.R.; Orr, L.; Turbide, C. Vesicle formation during reticulocyte maturation. Association of plasma membrane activities with released vesicles (exosomes). J. Biol. Chem. 1987, 262, 9412–9420. [Google Scholar] [CrossRef]
  7. Peinado, H.; Aleckovic, M.; Lavotshkin, S.; Matei, I.; Costa-Silva, B.; Moreno-Bueno, G.; Hergueta-Redondo, M.; Williams, C.; Garcia-Santos, G.; Ghajar, C.; et al. Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat. Med. 2012, 18, 883–891. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Whiteside, T.L. Tumor-Derived Exosomes and Their Role in Cancer Progression. Adv. Clin. Chem. 2016, 74, 103–141. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Larssen, P.; Wik, L.; Czarnewski, P.; Eldh, M.; Lof, L.; Ronquist, K.G.; Dubois, L.; Freyhult, E.; Gallant, C.J.; Oelrich, J.; et al. Tracing Cellular Origin of Human Exosomes Using Multiplex Proximity Extension Assays. Mol. Cell. Proteom. 2017, 16, 502–511. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Yu, X.; Harris, S.L.; Levine, A.J. The regulation of exosome secretion: A novel function of the p53 protein. Cancer Res. 2006, 66, 4795–4801. [Google Scholar] [CrossRef] [Green Version]
  11. Ostrowski, M.; Carmo, N.B.; Krumeich, S.; Fanget, I.; Raposo, G.; Savina, A.; Moita, C.F.; Schauer, K.; Hume, A.N.; Freitas, R.P.; et al. Rab27a and Rab27b control different steps of the exosome secretion pathway. Nat. Cell. Biol. 2010, 12, 19–30. [Google Scholar] [CrossRef] [Green Version]
  12. Bebelman, M.P.; Smit, M.J.; Pegtel, D.M.; Baglio, S.R. Biogenesis and function of extracellular vesicles in cancer. Pharmacol. Ther. 2018, 188, 1–11. [Google Scholar] [CrossRef]
  13. Kowal, J.; Tkach, M.; Thery, C. Biogenesis and secretion of exosomes. Curr. Opin. Cell Biol. 2014, 29, 116–125. [Google Scholar] [CrossRef] [Green Version]
  14. White, I.J.; Bailey, L.M.; Aghakhani, M.R.; Moss, S.E.; Futter, C.E. EGF stimulates annexin 1-dependent inward vesiculation in a multivesicular endosome subpopulation. EMBO J. 2006, 25, 1–12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Lachenal, G.; Pernet-Gallay, K.; Chivet, M.; Hemming, F.J.; Belly, A.; Bodon, G.; Blot, B.; Haase, G.; Goldberg, Y.; Sadoul, R. Release of exosomes from differentiated neurons and its regulation by synaptic glutamatergic activity. Mol. Cell Neurosci. 2011, 46, 409–418. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Andreu, Z.; Yanez-Mo, M. Tetraspanins in extracellular vesicle formation and function. Front. Immunol. 2014, 5, 442. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Zhang, Y.; Liu, Y.; Liu, H.; Tang, W.H. Exosomes: Biogenesis, biologic function and clinical potential. Cell Biosci. 2019, 9, 19. [Google Scholar] [CrossRef] [PubMed]
  18. Colombo, M.; Moita, C.; van Niel, G.; Kowal, J.; Vigneron, J.; Benaroch, P.; Manel, N.; Moita, L.F.; Thery, C.; Raposo, G. Analysis of ESCRT functions in exosome biogenesis, composition and secretion highlights the heterogeneity of extracellular vesicles. J. Cell Sci. 2013, 126, 5553–5565. [Google Scholar] [CrossRef] [Green Version]
  19. Henne, W.M.; Buchkovich, N.J.; Emr, S.D. The ESCRT pathway. Dev. Cell 2011, 21, 77–91. [Google Scholar] [CrossRef] [Green Version]
  20. Vietri, M.; Radulovic, M.; Stenmark, H. The many functions of ESCRTs. Nat. Rev. Mol. Cell. Biol. 2020, 21, 25–42. [Google Scholar] [CrossRef]
  21. Villarroya-Beltri, C.; Baixauli, F.; Gutierrez-Vazquez, C.; Sanchez-Madrid, F.; Mittelbrunn, M. Sorting it out: Regulation of exosome loading. Semin. Cancer Biol. 2014, 28, 3–13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Ghossoub, R.; Lembo, F.; Rubio, A.; Gaillard, C.B.; Bouchet, J.; Vitale, N.; Slavik, J.; Machala, M.; Zimmermann, P. Syntenin-ALIX exosome biogenesis and budding into multivesicular bodies are controlled by ARF6 and PLD2. Nat. Commun. 2014, 5, 3477. [Google Scholar] [CrossRef] [Green Version]
  23. Perez-Hernandez, D.; Gutierrez-Vazquez, C.; Jorge, I.; Lopez-Martin, S.; Ursa, A.; Sanchez-Madrid, F.; Vazquez, J.; Yanez-Mo, M. The intracellular interactome of tetraspanin-enriched microdomains reveals their function as sorting machineries toward exosomes. J. Biol. Chem. 2013, 288, 11649–11661. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Trajkovic, K.; Hsu, C.; Chiantia, S.; Rajendran, L.; Wenzel, D.; Wieland, F.; Schwille, P.; Brugger, B.; Simons, M. Ceramide triggers budding of exosome vesicles into multivesicular endosomes. Science 2008, 319, 1244–1247. [Google Scholar] [CrossRef] [PubMed]
  25. Riches, A.; Campbell, E.; Borger, E.; Powis, S. Regulation of exosome release from mammary epithelial and breast cancer cells—A new regulatory pathway. Eur. J. Cancer 2014, 50, 1025–1034. [Google Scholar] [CrossRef]
  26. Stenmark, H. Rab GTPases as coordinators of vesicle traffic. Nat. Rev. Mol. Cell Biol. 2009, 10, 513–525. [Google Scholar] [CrossRef] [PubMed]
  27. Wei, Y.; Wang, D.; Jin, F.; Bian, Z.; Li, L.; Liang, H.; Li, M.; Shi, L.; Pan, C.; Zhu, D.; et al. Pyruvate kinase type M2 promotes tumour cell exosome release via phosphorylating synaptosome-associated protein 23. Nat. Commun. 2017, 8, 14041. [Google Scholar] [CrossRef] [Green Version]
  28. Han, J.; Pluhackova, K.; Bockmann, R.A. The Multifaceted Role of SNARE Proteins in Membrane Fusion. Front. Physiol. 2017, 8, 5. [Google Scholar] [CrossRef] [Green Version]
  29. Hoshino, D.; Kirkbride, K.C.; Costello, K.; Clark, E.S.; Sinha, S.; Grega-Larson, N.; Tyska, M.J.; Weaver, A.M. Exosome secretion is enhanced by invadopodia and drives invasive behavior. Cell Rep. 2013, 5, 1159–1168. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Isaac, R.; Reis, F.C.G.; Ying, W.; Olefsky, J.M. Exosomes as mediators of intercellular crosstalk in metabolism. Cell Metab. 2021, 33, 1744–1762. [Google Scholar] [CrossRef] [PubMed]
  31. Fitzner, D.; Schnaars, M.; van Rossum, D.; Krishnamoorthy, G.; Dibaj, P.; Bakhti, M.; Regen, T.; Hanisch, U.K.; Simons, M. Selective transfer of exosomes from oligodendrocytes to microglia by macropinocytosis. J. Cell Sci. 2011, 124, 447–458. [Google Scholar] [CrossRef] [Green Version]
  32. Feng, D.; Zhao, W.L.; Ye, Y.Y.; Bai, X.C.; Liu, R.Q.; Chang, L.F.; Zhou, Q.; Sui, S.F. Cellular internalization of exosomes occurs through phagocytosis. Traffic 2010, 11, 675–687. [Google Scholar] [CrossRef]
  33. Tian, T.; Zhu, Y.L.; Zhou, Y.Y.; Liang, G.F.; Wang, Y.Y.; Hu, F.H.; Xiao, Z.D. Exosome uptake through clathrin-mediated endocytosis and macropinocytosis and mediating miR-21 delivery. J. Biol. Chem. 2014, 289, 22258–22267. [Google Scholar] [CrossRef] [Green Version]
  34. Svensson, K.J.; Christianson, H.C.; Wittrup, A.; Bourseau-Guilmain, E.; Lindqvist, E.; Svensson, L.M.; Morgelin, M.; Belting, M. Exosome uptake depends on ERK1/2-heat shock protein 27 signaling and lipid Raft-mediated endocytosis negatively regulated by caveolin-1. J. Biol. Chem. 2013, 288, 17713–17724. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. 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] [Green Version]
  36. Toda, Y.; Takata, K.; Nakagawa, Y.; Kawakami, H.; Fujioka, S.; Kobayashi, K.; Hattori, Y.; Kitamura, Y.; Akaji, K.; Ashihara, E. Effective internalization of U251-MG-secreted exosomes into cancer cells and characterization of their lipid components. Biochem. Biophys. Res. Commun. 2015, 456, 768–773. [Google Scholar] [CrossRef] [Green Version]
  37. Munich, S.; Sobo-Vujanovic, A.; Buchser, W.J.; Beer-Stolz, D.; Vujanovic, N.L. Dendritic cell exosomes directly kill tumor cells and activate natural killer cells via TNF superfamily ligands. Oncoimmunology 2012, 1, 1074–1083. [Google Scholar] [CrossRef] [Green Version]
  38. Tkach, M.; Kowal, J.; Zucchetti, A.E.; Enserink, L.; Jouve, M.; Lankar, D.; Saitakis, M.; Martin-Jaular, L.; Thery, C. Qualitative differences in T-cell activation by dendritic cell-derived extracellular vesicle subtypes. EMBO J. 2017, 36, 3012–3028. [Google Scholar] [CrossRef]
  39. Choi, D.S.; Kim, D.K.; Kim, Y.K.; Gho, Y.S. Proteomics of extracellular vesicles: Exosomes and ectosomes. Mass Spectrom. Rev. 2015, 34, 474–490. [Google Scholar] [CrossRef]
  40. Shimaoka, M.; Kawamoto, E.; Gaowa, A.; Okamoto, T.; Park, E.J. Connexins and Integrins in Exosomes. Cancers 2019, 11, 106. [Google Scholar] [CrossRef] [Green Version]
  41. Runz, S.; Keller, S.; Rupp, C.; Stoeck, A.; Issa, Y.; Koensgen, D.; Mustea, A.; Sehouli, J.; Kristiansen, G.; Altevogt, P. Malignant ascites-derived exosomes of ovarian carcinoma patients contain CD24 and EpCAM. Gynecol. Oncol. 2007, 107, 563–571. [Google Scholar] [CrossRef]
  42. Wyciszkiewicz, A.; Kalinowska-Lyszczarz, A.; Nowakowski, B.; Kazmierczak, K.; Osztynowicz, K.; Michalak, S. Expression of small heat shock proteins in exosomes from patients with gynecologic cancers. Sci. Rep. 2019, 9, 9817. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Ciravolo, V.; Huber, V.; Ghedini, G.C.; Venturelli, E.; Bianchi, F.; Campiglio, M.; Morelli, D.; Villa, A.; Della Mina, P.; Menard, S.; et al. Potential role of HER2-overexpressing exosomes in countering trastuzumab-based therapy. J. Cell Physiol. 2012, 227, 658–667. [Google Scholar] [CrossRef]
  44. Baxevanis, C.N.; Anastasopoulou, E.A.; Voutsas, I.F.; Papamichail, M.; Perez, S.A. Immune biomarkers: How well do they serve prognosis in human cancers? Expert. Rev. Mol. Diagn. 2015, 15, 49–59. [Google Scholar] [CrossRef]
  45. Wieckowski, E.U.; Visus, C.; Szajnik, M.; Szczepanski, M.J.; Storkus, W.J.; Whiteside, T.L. Tumor-derived microvesicles promote regulatory T cell expansion and induce apoptosis in tumor-reactive activated CD8+ T lymphocytes. J. Immunol. 2009, 183, 3720–3730. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Buzas, E.I.; Toth, E.A.; Sodar, B.W.; Szabo-Taylor, K.E. Molecular interactions at the surface of extracellular vesicles. Semin. Immunopathol. 2018, 40, 453–464. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Baranyai, T.; Herczeg, K.; Onodi, Z.; Voszka, I.; Modos, K.; Marton, N.; Nagy, G.; Mager, I.; Wood, M.J.; El Andaloussi, S.; et al. Isolation of Exosomes from Blood Plasma: Qualitative and Quantitative Comparison of Ultracentrifugation and Size Exclusion Chromatography Methods. PLoS ONE 2015, 10, e0145686. [Google Scholar] [CrossRef] [Green Version]
  48. Gyorgy, B.; Szabo, T.G.; Turiak, L.; Wright, M.; Herczeg, P.; Ledeczi, Z.; Kittel, A.; Polgar, A.; Toth, K.; Derfalvi, B.; et al. Improved flow cytometric assessment reveals distinct microvesicle (cell-derived microparticle) signatures in joint diseases. PLoS ONE 2012, 7, e49726. [Google Scholar] [CrossRef]
  49. Karimi, N.; Cvjetkovic, A.; Jang, S.C.; Crescitelli, R.; Hosseinpour Feizi, M.A.; Nieuwland, R.; Lotvall, J.; Lasser, C. Detailed analysis of the plasma extracellular vesicle proteome after separation from lipoproteins. Cell. Mol. Life Sci. 2018, 75, 2873–2886. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Onodi, Z.; Pelyhe, C.; Terezia Nagy, C.; Brenner, G.B.; Almasi, L.; Kittel, A.; Mancek-Keber, M.; Ferdinandy, P.; Buzas, E.I.; Giricz, Z. Isolation of High-Purity Extracellular Vesicles by the Combination of Iodixanol Density Gradient Ultracentrifugation and Bind-Elute Chromatography From Blood Plasma. Front. Physiol. 2018, 9, 1479. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Toth, E.A.; Turiak, L.; Visnovitz, T.; Cserep, C.; Mazlo, A.; Sodar, B.W.; Forsonits, A.I.; Petovari, G.; Sebestyen, A.; Komlosi, Z.; et al. Formation of a protein corona on the surface of extracellular vesicles in blood plasma. J. Extracell. Vesicles 2021, 10, e12140. [Google Scholar] [CrossRef]
  52. Palviainen, M.; Saraswat, M.; Varga, Z.; Kitka, D.; Neuvonen, M.; Puhka, M.; Joenvaara, S.; Renkonen, R.; Nieuwland, R.; Takatalo, M.; et al. Extracellular vesicles from human plasma and serum are carriers of extravesicular cargo-Implications for biomarker discovery. PLoS ONE 2020, 15, e0236439. [Google Scholar] [CrossRef] [PubMed]
  53. Gusachenko, O.N.; Zenkova, M.A.; Vlassov, V.V. Nucleic acids in exosomes: Disease markers and intercellular communication molecules. Biochemistry 2013, 78, 1–7. [Google Scholar] [CrossRef] [PubMed]
  54. Balaj, L.; Lessard, R.; Dai, L.; Cho, Y.J.; Pomeroy, S.L.; Breakefield, X.O.; Skog, J. Tumour microvesicles contain retrotransposon elements and amplified oncogene sequences. Nat. Commun. 2011, 2, 180. [Google Scholar] [CrossRef] [PubMed]
  55. Lee, T.H.; Chennakrishnaiah, S.; Audemard, E.; Montermini, L.; Meehan, B.; Rak, J. Oncogenic ras-driven cancer cell vesiculation leads to emission of double-stranded DNA capable of interacting with target cells. Biochem. Biophys. Res. Commun. 2014, 451, 295–301. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Kahlert, C.; Melo, S.A.; Protopopov, A.; Tang, J.; Seth, S.; Koch, M.; Zhang, J.; Weitz, J.; Chin, L.; Futreal, A.; et al. Identification of double-stranded genomic DNA spanning all chromosomes with mutated KRAS and p53 DNA in the serum exosomes of patients with pancreatic cancer. J. Biol. Chem. 2014, 289, 3869–3875. [Google Scholar] [CrossRef] [Green Version]
  57. Veziroglu, E.M.; Mias, G.I. Characterizing Extracellular Vesicles and Their Diverse RNA Contents. Front. Genet. 2020, 11, 700. [Google Scholar] [CrossRef]
  58. Al-Nedawi, K.; Meehan, B.; Micallef, J.; Lhotak, V.; May, L.; Guha, A.; Rak, J. Intercellular transfer of the oncogenic receptor EGFRvIII by microvesicles derived from tumour cells. Nat. Cell Biol. 2008, 10, 619–624. [Google Scholar] [CrossRef] [PubMed]
  59. Tanaka, Y.; Kamohara, H.; Kinoshita, K.; Kurashige, J.; Ishimoto, T.; Iwatsuki, M.; Watanabe, M.; Baba, H. Clinical impact of serum exosomal microRNA-21 as a clinical biomarker in human esophageal squamous cell carcinoma. Cancer 2013, 119, 1159–1167. [Google Scholar] [CrossRef]
  60. Conigliaro, A.; Costa, V.; Lo Dico, A.; Saieva, L.; Buccheri, S.; Dieli, F.; Manno, M.; Raccosta, S.; Mancone, C.; Tripodi, M.; et al. CD90+ liver cancer cells modulate endothelial cell phenotype through the release of exosomes containing H19 lncRNA. Mol. Cancer 2015, 14, 155. [Google Scholar] [CrossRef]
  61. Li, J.; Li, Z.; Jiang, P.; Peng, M.; Zhang, X.; Chen, K.; Liu, H.; Bi, H.; Liu, X.; Li, X. Circular RNA IARS (circ-IARS) secreted by pancreatic cancer cells and located within exosomes regulates endothelial monolayer permeability to promote tumor metastasis. J. Exp. Clin. Cancer Res. 2018, 37, 177. [Google Scholar] [CrossRef] [Green Version]
  62. 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]
  63. Zhang, Z.Y.; Li, Y.C.; Geng, C.Y.; Zhou, H.X.; Gao, W.; Chen, W.M. Serum exosomal microRNAs as novel biomarkers for multiple myeloma. Hematol. Oncol. 2019, 37, 409–417. [Google Scholar] [CrossRef] [PubMed]
  64. Taylor, D.D.; Gercel-Taylor, C. MicroRNA signatures of tumor-derived exosomes as diagnostic biomarkers of ovarian cancer. Gynecol. Oncol. 2008, 110, 13–21. [Google Scholar] [CrossRef]
  65. Manier, S.; Sacco, A.; Leleu, X.; Ghobrial, I.M.; Roccaro, A.M. Bone marrow microenvironment in multiple myeloma progression. J. Biomed. Biotechnol. 2012, 2012, 157496. [Google Scholar] [CrossRef] [PubMed]
  66. Frassanito, M.A.; Rao, L.; Moschetta, M.; Ria, R.; Di Marzo, L.; De Luisi, A.; Racanelli, V.; Catacchio, I.; Berardi, S.; Basile, A.; et al. Bone marrow fibroblasts parallel multiple myeloma progression in patients and mice: In vitro and in vivo studies. Leukemia 2014, 28, 904–916. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Frassanito, M.A.; De Veirman, K.; Desantis, V.; Di Marzo, L.; Vergara, D.; Ruggieri, S.; Annese, T.; Nico, B.; Menu, E.; Catacchio, I.; et al. Halting pro-survival autophagy by TGFbeta inhibition in bone marrow fibroblasts overcomes bortezomib resistance in multiple myeloma patients. Leukemia 2016, 30, 640–648. [Google Scholar] [CrossRef]
  68. Frassanito, M.A.; Desantis, V.; Di Marzo, L.; Craparotta, I.; Beltrame, L.; Marchini, S.; Annese, T.; Visino, F.; Arciuli, M.; Saltarella, I.; et al. Bone marrow fibroblasts overexpress miR-27b and miR-214 in step with multiple myeloma progression, dependent on tumour cell-derived exosomes. J. Pathol. 2019, 247, 241–253. [Google Scholar] [CrossRef] [PubMed]
  69. Cheng, Q.; Li, X.; Liu, J.; Ye, Q.; Chen, Y.; Tan, S.; Liu, J. Multiple Myeloma-Derived Exosomes Regulate the Functions of Mesenchymal Stem Cells Partially via Modulating miR-21 and miR-146a. Stem Cells Int. 2017, 2017, 9012152. [Google Scholar] [CrossRef] [PubMed]
  70. Liu, Z.; Liu, H.; Li, Y.; Shao, Q.; Chen, J.; Song, J.; Fu, R. Multiple myeloma-derived exosomes inhibit osteoblastic differentiation and improve IL-6 secretion of BMSCs from multiple myeloma. J. Investig. Med. 2020, 68, 45–51. [Google Scholar] [CrossRef]
  71. Reale, A.; Carmichael, I.; Xu, R.; Mithraprabhu, S.; Khong, T.; Chen, M.; Fang, H.; Savvidou, I.; Ramachandran, M.; Bingham, N.; et al. Human myeloma cell- and plasma-derived extracellular vesicles contribute to functional regulation of stromal cells. Proteomics 2021, 21, e2000119. [Google Scholar] [CrossRef] [PubMed]
  72. Dabbah, M.; Attar-Schneider, O.; Tartakover Matalon, S.; Shefler, I.; Jarchwsky Dolberg, O.; Lishner, M.; Drucker, L. Microvesicles derived from normal and multiple myeloma bone marrow mesenchymal stem cells differentially modulate myeloma cells’ phenotype and translation initiation. Carcinogenesis 2017, 38, 708–716. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Watanabe, T. Realization of Osteolysis, Angiogenesis, Immunosuppression, and Drug Resistance by Extracellular Vesicles: Roles of RNAs and Proteins in Their Cargoes and of Ectonucleotidases of the Immunosuppressive Adenosinergic Noncanonical Pathway in the Bone Marrow Niche of Multiple Myeloma. Cancers 2021, 13, 2969. [Google Scholar] [CrossRef]
  74. 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]
  75. Dabbah, M.; Lishner, M.; Jarchowsky-Dolberg, O.; Tartakover-Matalon, S.; Brin, Y.S.; Pasmanik-Chor, M.; Neumann, A.; Drucker, L. Ribosomal proteins as distinct “passengers” of microvesicles: New semantics in myeloma and mesenchymal stem cells’ communication. Transl. Res. 2021, 236, 117–132. [Google Scholar] [CrossRef] [PubMed]
  76. 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] [Green Version]
  77. Terpos, E.; Ntanasis-Stathopoulos, I.; Gavriatopoulou, M.; Dimopoulos, M.A. Pathogenesis of bone disease in multiple myeloma: From bench to bedside. Blood Cancer J. 2018, 8, 7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Faict, S.; Muller, J.; De Veirman, K.; De Bruyne, E.; Maes, K.; Vrancken, L.; Heusschen, R.; De Raeve, H.; Schots, R.; Vanderkerken, K.; et al. Exosomes play a role in multiple myeloma bone disease and tumor development by targeting osteoclasts and osteoblasts. Blood Cancer J. 2018, 8, 105. [Google Scholar] [CrossRef] [Green Version]
  79. 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] [Green Version]
  80. Raimondi, L.; De Luca, A.; Fontana, S.; Amodio, N.; Costa, V.; Carina, V.; Bellavia, D.; Raimondo, S.; Siragusa, S.; Monteleone, F.; et al. Multiple Myeloma-Derived Extracellular Vesicles Induce Osteoclastogenesis through the Activation of the XBP1/IRE1alpha Axis. Cancers 2020, 12, 2167. [Google Scholar] [CrossRef] [PubMed]
  81. Raimondo, S.; Saieva, L.; Vicario, E.; Pucci, M.; Toscani, D.; Manno, M.; Raccosta, S.; Giuliani, N.; Alessandro, R. Multiple myeloma-derived exosomes are enriched of amphiregulin (AREG) and activate the epidermal growth factor pathway in the bone microenvironment leading to osteoclastogenesis. J. Hematol. Oncol. 2019, 12, 2. [Google Scholar] [CrossRef] [PubMed]
  82. Zahoor, M.; Westhrin, M.; Aass, K.R.; Moen, S.H.; Misund, K.; Psonka-Antonczyk, K.M.; Giliberto, M.; Buene, G.; Sundan, A.; Waage, A.; et al. Hypoxia promotes IL-32 expression in myeloma cells, and high expression is associated with poor survival and bone loss. Blood Adv. 2017, 1, 2656–2666. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Colla, S.; Morandi, F.; Lazzaretti, M.; Rizzato, R.; Lunghi, P.; Bonomini, S.; Mancini, C.; Pedrazzoni, M.; Crugnola, M.; Rizzoli, V.; et al. Human myeloma cells express the bone regulating gene Runx2/Cbfa1 and produce osteopontin that is involved in angiogenesis in multiple myeloma patients. Leukemia 2005, 19, 2166–2176. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Saeki, Y.; Mima, T.; Ishii, T.; Ogata, A.; Kobayashi, H.; Ohshima, S.; Ishida, T.; Tabunoki, Y.; Kitayama, H.; Mizuki, M.; et al. Enhanced production of osteopontin in multiple myeloma: Clinical and pathogenic implications. Br. J. Haematol. 2003, 123, 263–270. [Google Scholar] [CrossRef] [PubMed]
  85. Li, B.; Xu, H.; Han, H.; Song, S.; Zhang, X.; Ouyang, L.; Qian, C.; Hong, Y.; Qiu, Y.; Zhou, W.; et al. Exosome-mediated transfer of lncRUNX2-AS1 from multiple myeloma cells to MSCs contributes to osteogenesis. Oncogene 2018, 37, 5508–5519. [Google Scholar] [CrossRef] [PubMed]
  86. Raimondo, S.; Urzi, O.; Conigliaro, A.; Bosco, G.L.; Parisi, S.; Carlisi, M.; Siragusa, S.; Raimondi, L.; Luca, A.; Giavaresi, G.; et al. Extracellular Vesicle microRNAs Contribute to the Osteogenic Inhibition of Mesenchymal Stem Cells in Multiple Myeloma. Cancers 2020, 12, 449. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Lopes, R.; Caetano, J.; Ferreira, B.; Barahona, F.; Carneiro, E.A.; Joao, C. The Immune Microenvironment in Multiple Myeloma: Friend or Foe? Cancers 2021, 13, 625. [Google Scholar] [CrossRef]
  88. Zelle-Rieser, C.; Thangavadivel, S.; Biedermann, R.; Brunner, A.; Stoitzner, P.; Willenbacher, E.; Greil, R.; Johrer, K. T cells in multiple myeloma display features of exhaustion and senescence at the tumor site. J. Hematol. Oncol. 2016, 9, 116. [Google Scholar] [CrossRef] [Green Version]
  89. Frassanito, M.A.; Ruggieri, S.; Desantis, V.; Di Marzo, L.; Leone, P.; Racanelli, V.; Fumarulo, R.; Dammacco, F.; Vacca, A. Myeloma cells act as tolerogenic antigen-presenting cells and induce regulatory T cells in vitro. Eur. J. Haematol. 2015, 95, 65–74. [Google Scholar] [CrossRef]
  90. Prabhala, R.H.; Pelluru, D.; Fulciniti, M.; Prabhala, H.K.; Nanjappa, P.; Song, W.; Pai, C.; Amin, S.; Tai, Y.T.; Richardson, P.G.; et al. Elevated IL-17 produced by TH17 cells promotes myeloma cell growth and inhibits immune function in multiple myeloma. Blood 2010, 115, 5385–5392. [Google Scholar] [CrossRef] [PubMed]
  91. Leone, P.; Solimando, A.G.; Malerba, E.; Fasano, R.; Buonavoglia, A.; Pappagallo, F.; De Re, V.; Argentiero, A.; Silvestris, N.; Vacca, A.; et al. Actors on the Scene: Immune Cells in the Myeloma Niche. Front. Oncol. 2020, 10, 599098. [Google Scholar] [CrossRef] [PubMed]
  92. Veglia, F.; Sanseviero, E.; Gabrilovich, D.I. Myeloid-derived suppressor cells in the era of increasing myeloid cell diversity. Nat. Rev. Immunol. 2021, 21, 485–498. [Google Scholar] [CrossRef] [PubMed]
  93. Favaloro, J.; Liyadipitiya, T.; Brown, R.; Yang, S.; Suen, H.; Woodland, N.; Nassif, N.; Hart, D.; Fromm, P.; Weatherburn, C.; et al. Myeloid derived suppressor cells are numerically, functionally and phenotypically different in patients with multiple myeloma. Leuk. Lymphoma 2014, 55, 2893–2900. [Google Scholar] [CrossRef] [PubMed]
  94. Wang, J.; De Veirman, K.; De Beule, N.; Maes, K.; De Bruyne, E.; Van Valckenborgh, E.; Vanderkerken, K.; Menu, E. The bone marrow microenvironment enhances multiple myeloma progression by exosome-mediated activation of myeloid-derived suppressor cells. Oncotarget 2015, 6, 43992–44004. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. 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]
  96. Morandi, F.; Marimpietri, D.; Horenstein, A.L.; Bolzoni, M.; Toscani, D.; Costa, F.; Castella, B.; Faini, A.C.; Massaia, M.; Pistoia, V.; et al. Microvesicles released from multiple myeloma cells are equipped with ectoenzymes belonging to canonical and non-canonical adenosinergic pathways and produce adenosine from ATP and NAD(+). Oncoimmunology 2018, 7, e1458809. [Google Scholar] [CrossRef] [Green Version]
  97. Saltarella, I.; Desantis, V.; Melaccio, A.; Solimando, A.G.; Lamanuzzi, A.; Ria, R.; Storlazzi, C.T.; Mariggio, M.A.; Vacca, A.; Frassanito, M.A. Mechanisms of Resistance to Anti-CD38 Daratumumab in Multiple Myeloma. Cells 2020, 9, 167. [Google Scholar] [CrossRef] [Green Version]
  98. 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] [Green Version]
  99. 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]
  100. Li, B.; Hong, J.; Hong, M.; Wang, Y.; Yu, T.; Zang, S.; Wu, Q. piRNA-823 delivered by multiple myeloma-derived extracellular vesicles promoted tumorigenesis through re-educating endothelial cells in the tumor environment. Oncogene 2019, 38, 5227–5238. [Google Scholar] [CrossRef] [PubMed]
  101. Ria, R.; Catacchio, I.; Berardi, S.; De Luisi, A.; Caivano, A.; Piccoli, C.; Ruggieri, V.; Frassanito, M.A.; Ribatti, D.; Nico, B.; et al. HIF-1alpha of bone marrow endothelial cells implies relapse and drug resistance in patients with multiple myeloma and may act as a therapeutic target. Clin. Cancer Res. 2014, 20, 847–858. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Bhaskar, A.; Tiwary, B.N. Hypoxia inducible factor-1 alpha and multiple myeloma. Int. J. Adv. Res. 2016, 4, 706–715. [Google Scholar]
  103. Tian, X.; Sun, M.; Wu, H.; Chen, C.; Li, H.; Qiu, S.; Wang, T.; Han, J.; Xiao, Q.; Chen, K. Exosome-derived miR-let-7c promotes angiogenesis in multiple myeloma by polarizing M2 macrophages in the bone marrow microenvironment. Leuk. Res. 2021, 105, 106566. [Google Scholar] [CrossRef] [PubMed]
  104. Sedlarikova, L.; Bollova, B.; Radova, L.; Brozova, L.; Jarkovsky, J.; Almasi, M.; Penka, M.; Kuglik, P.; Sandecka, V.; Stork, M.; et al. Circulating exosomal long noncoding RNA PRINS-First findings in monoclonal gammopathies. Hematol. Oncol. 2018, 36, 786–791. [Google Scholar] [CrossRef] [PubMed]
  105. Zhang, L.; Pan, L.; Xiang, B.; Zhu, H.; Wu, Y.; Chen, M.; Guan, P.; Zou, X.; Valencia, C.A.; Dong, B.; et al. Potential role of exosome-associated microRNA panels and in vivo environment to predict drug resistance for patients with multiple myeloma. Oncotarget 2016, 7, 30876–30891. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Xu, H.; Han, H.; Song, S.; Yi, N.; Qian, C.; Qiu, Y.; Zhou, W.; Hong, Y.; Zhuang, W.; Li, Z.; et al. Exosome-Transmitted PSMA3 and PSMA3-AS1 Promote Proteasome Inhibitor Resistance in Multiple Myeloma. Clin. Cancer Res. 2019, 25, 1923–1935. [Google Scholar] [CrossRef] [PubMed]
  107. Wang, S.; Khan, A.; Huang, R.; Ye, S.; Di, K.; Xiong, T.; Li, Z. Recent advances in single extracellular vesicle detection methods. Biosens. Bioelectron. 2020, 154, 112056. [Google Scholar] [CrossRef]
  108. Marchisio, M.; Simeone, P.; Bologna, G.; Ercolino, E.; Pierdomenico, L.; Pieragostino, D.; Ventrella, A.; Antonini, F.; Del Zotto, G.; Vergara, D.; et al. Flow Cytometry Analysis of Circulating Extracellular Vesicle Subtypes from Fresh Peripheral Blood Samples. Int. J. Mol. Sci. 2020, 22, 48. [Google Scholar] [CrossRef] [PubMed]
  109. Russo, M.; Tirinato, L.; Scionti, F.; Coluccio, M.L.; Perozziello, G.; Riillo, C.; Mollace, V.; Gratteri, S.; Malara, N.; Di Martino, M.T.; et al. Raman Spectroscopic Stratification of Multiple Myeloma Patients Based on Exosome Profiling. ACS Omega 2020, 5, 30436–30443. [Google Scholar] [CrossRef]
  110. Laurenzana, I.; Trino, S.; Lamorte, D.; Girasole, M.; Dinarelli, S.; De Stradis, A.; Grieco, V.; Maietti, M.; Traficante, A.; Statuto, T.; et al. Analysis of Amount, Size, Protein Phenotype and Molecular Content of Circulating Extracellular Vesicles Identifies New Biomarkers in Multiple Myeloma. Int. J. Nanomed. 2021, 16, 3141–3160. [Google Scholar] [CrossRef] [PubMed]
  111. Raimondo, S.; Giavaresi, G.; Lorico, A.; Alessandro, R. Extracellular Vesicles as Biological Shuttles for Targeted Therapies. Int. J. Mol. Sci. 2019, 20, 1848. [Google Scholar] [CrossRef] [Green Version]
  112. Thompson, C.A.; Purushothaman, A.; Ramani, V.C.; Vlodavsky, I.; Sanderson, R.D. Heparanase regulates secretion, composition, and function of tumor cell-derived exosomes. J. Biol. Chem. 2013, 288, 10093–10099. [Google Scholar] [CrossRef] [Green Version]
  113. Ritchie, J.P.; Ramani, V.C.; Ren, Y.; Naggi, A.; Torri, G.; Casu, B.; Penco, S.; Pisano, C.; Carminati, P.; Tortoreto, M.; et al. SST0001, a chemically modified heparin, inhibits myeloma growth and angiogenesis via disruption of the heparanase/syndecan-1 axis. Clin. Cancer Res. 2011, 17, 1382–1393. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Cheng, Q.; Li, X.; Wang, Y.; Dong, M.; Zhan, F.H.; Liu, J. The ceramide pathway is involved in the survival, apoptosis and exosome functions of human multiple myeloma cells in vitro. Acta Pharmacol. Sin. 2018, 39, 561–568. [Google Scholar] [CrossRef] [PubMed]
  115. Menck, K.; Sonmezer, C.; Worst, T.S.; Schulz, M.; Dihazi, G.H.; Streit, F.; Erdmann, G.; Kling, S.; Boutros, M.; Binder, C.; et al. Neutral sphingomyelinases control extracellular vesicles budding from the plasma membrane. J. Extracell. Vesicles 2017, 6, 1378056. [Google Scholar] [CrossRef] [PubMed]
  116. Vuckovic, S.; Vandyke, K.; Rickards, D.A.; McCauley Winter, P.; Brown, S.H.J.; Mitchell, T.W.; Liu, J.; Lu, J.; Askenase, P.W.; Yuriev, E.; et al. The cationic small molecule GW4869 is cytotoxic to high phosphatidylserine-expressing myeloma cells. Br. J. Haematol. 2017, 177, 423–440. [Google Scholar] [CrossRef]
  117. Zheng, Y.; Tu, C.; Zhang, J.; Wang, J. Inhibition of multiple myelomaderived exosomes uptake suppresses the functional response in bone marrow stromal cell. Int. J. Oncol. 2019, 54, 1061–1070. [Google Scholar] [CrossRef]
  118. Tu, C.; Du, Z.; Zhang, H.; Feng, Y.; Qi, Y.; Zheng, Y.; Liu, J.; Wang, J. Endocytic pathway inhibition attenuates extracellular vesicle-induced reduction of chemosensitivity to bortezomib in multiple myeloma cells. Theranostics 2021, 11, 2364–2380. [Google Scholar] [CrossRef] [PubMed]
  119. 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] [Green Version]
  120. Xie, Y.; Bai, O.; Zhang, H.; Li, W.; Xiang, J. Tumor necrosis factor gene-engineered J558 tumor cell-released exosomes stimulate tumor antigen P1A-specific CD8+ CTL responses and antitumor immunity. Cancer Biother. Radiopharm. 2010, 25, 21–28. [Google Scholar] [CrossRef] [PubMed]
  121. Wang, J.; Cao, Z.; Wang, P.; Zhang, X.; Tang, J.; He, Y.; Huang, Z.; Mao, X.; Shi, S.; Kou, X. Apoptotic Extracellular Vesicles Ameliorate Multiple Myeloma by Restoring Fas-Mediated Apoptosis. ACS Nano 2021, 15, 14360–14372. [Google Scholar] [CrossRef] [PubMed]
  122. Desantis, V.; Saltarella, I.; Lamanuzzi, A.; Melaccio, A.; Solimando, A.G.; Mariggio, M.A.; Racanelli, V.; Paradiso, A.; Vacca, A.; Frassanito, M.A. MicroRNAs-Based Nano-Strategies as New Therapeutic Approach in Multiple Myeloma to Overcome Disease Progression and Drug Resistance. Int. J. Mol. Sci. 2020, 21, 3084. [Google Scholar] [CrossRef]
  123. 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]
  124. Wang, X.; He, L.; Huang, X.; Zhang, S.; Cao, W.; Che, F.; Zhu, Y.; Dai, J. Recent Progress of Exosomes in Multiple Myeloma: Pathogenesis, Diagnosis, Prognosis and Therapeutic Strategies. Cancers 2021, 13, 1635. [Google Scholar] [CrossRef] [PubMed]
  125. Hong, D.S.; Kang, Y.K.; Borad, M.; Sachdev, J.; Ejadi, S.; Lim, H.Y.; Brenner, A.J.; Park, K.; Lee, J.L.; Kim, T.Y.; et al. Phase 1 study of MRX34, a liposomal miR-34a mimic, in patients with advanced solid tumours. Br. J. Cancer 2020, 122, 1630–1637. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic representation of EV biogenesis (A) and cargo (B). For more details, see the main text.
Figure 1. Schematic representation of EV biogenesis (A) and cargo (B). For more details, see the main text.
Cells 10 03185 g001
Figure 2. Schematic representation EVs-mediated interaction in BM microenvironment. MM cells release EVs that contain oncogenic proteins, cytokines, miRs, and other ncRNAs that support: (i) disease progression and drug resistance supporting fibroblast (FBs) activation, conversion of mesenchymal stromal cells (MSCs) into cancer-associated fibroblasts (CAFs) and IL-6 release by bone marrow stromal cells (BMSCs); (ii) bone resorption by increasing proliferation and function of osteoclasts (OCs) and preventing their apoptosis, and by inhibiting osteoblast differentiation; (iii) angiogenesis through the release of pro-angiogenic cytokines and chemokines, which promote the viability of endothelial cells (ECs) and BMSCs and the release of pro-angiogenic factors; and (iv) immunosuppression, which promotes the expansion of myeloid derived suppressor cells (MDSCs) and the production of the immunosuppressive adenosine (ADO). Finally, EVs from BMSCs, in turn, promote the proliferation and viability of MM cells, thus supporting disease progression and drug resistance.
Figure 2. Schematic representation EVs-mediated interaction in BM microenvironment. MM cells release EVs that contain oncogenic proteins, cytokines, miRs, and other ncRNAs that support: (i) disease progression and drug resistance supporting fibroblast (FBs) activation, conversion of mesenchymal stromal cells (MSCs) into cancer-associated fibroblasts (CAFs) and IL-6 release by bone marrow stromal cells (BMSCs); (ii) bone resorption by increasing proliferation and function of osteoclasts (OCs) and preventing their apoptosis, and by inhibiting osteoblast differentiation; (iii) angiogenesis through the release of pro-angiogenic cytokines and chemokines, which promote the viability of endothelial cells (ECs) and BMSCs and the release of pro-angiogenic factors; and (iv) immunosuppression, which promotes the expansion of myeloid derived suppressor cells (MDSCs) and the production of the immunosuppressive adenosine (ADO). Finally, EVs from BMSCs, in turn, promote the proliferation and viability of MM cells, thus supporting disease progression and drug resistance.
Cells 10 03185 g002
Figure 3. Schematic representation of EV therapeutic perspectives: (A) inhibition of EVs release and uptake, (B) EVs as therapeutic tools. For more details see the main text.
Figure 3. Schematic representation of EV therapeutic perspectives: (A) inhibition of EVs release and uptake, (B) EVs as therapeutic tools. For more details see the main text.
Cells 10 03185 g003
Table 1. Biological and clinical relevance of role of EVs in MM.
Table 1. Biological and clinical relevance of role of EVs in MM.
Cargo MoleculeEVs SourceTarget CellsBiological Effect in Target CellsStudies ModelClinical RelevanceClinical ApplicationRef
WWC2MM cellsFBsHippo pathway activation;
miR-27b-3p and miR214-3p
In vitroTransition MGUS to MMPrognostic *[68]
IL-6, MCP-1, fibronectin, miR-15aMSCsMM cellsTumor growthIn vitro and in vivoTumor progressionPrognostic *[74]
MCP-1, MIP-1α, SDF-1BMSCsMM cellsActivation of JNK, p38, p53, AKT pathwaysIn vitroTumor progressionPrognostic *[76]
RUNX2-AS1MM cellsMSCsDown-regulation of RUNX2 expression and of osteogenic potentialIn vitro and in vivoTumor-associated bone lossPrognostic *[85]
Not specifiedBMSCs,
MM cells
MSDCsIncreased STATs-mediated viabilityIn vitro and in vivoImmunosuppressionPrognostic *[94,95]
miR-129-5pMM cells, BM plasmaMSCsDown-regulation of Sp1 and reduced osteogenic differentiationIn vitro and ex vivoBone lesions, transition SMM to MMPrognostic[86]
Angiogenin, HGF, MMP-9, VEGFMM cellsECsEnhanced viabilityIn vitro and in vivoIncreased angiogenesisPrognostic[95]
IL32MM cellspreOCsIncreased osteoclast activityIn vitro and ex vivoOsteolytic bone disease, reduced PFSPrognostic[82]
miR-135bHypoxic MM cell linesECsDownregulation FIH-1In vitro and in vivoIncreased angiogenesisPrognostic[99]
let-7b and miR-18aPlasmaNot specifiedNot specifiedEx vivoNegative correlation with PFS and OSPrognostic[62]
Adenosinergic ectoenzymesBM plasmaNot specifiedIncreased ADOEx vivoImmunosuppression, HD to MM transitionDiagnostic[96]
miR-20a-5p, miR-103a-3p, miR-4505SerumNot specifiedNot specifiedEx vivoHD and MM transition transitionDiagnostic[63]
Let-7c-5p, miR-185-5p, miR-4741SerumNot specifiedNot specifiedEx vivoSMM to MM transitionDiagnostic[63]
lncRNA PRINSSerumNot specifiedNot specifiedEx vivoCorrelation with biochemical parameters in MGUS and MM patientsDiagnostic[104]
PSMA3 mRNA, PSMA3-AS1MSCsNot specifiedIncreased PSMA3 protein levels and increased proteasome activityIn vitro and ex vivoResistance to proteasome inhibitors, high levels correlating to OS and PFFPrognostic, Response to therapy[106]
miR-16-5p, miR-15a-5p, miR-20a-5p, miR-17-5pSerumNot specifiedNot specifiedEx vivoResistance to bortezomibPrognostic,
Response to therapy
[105]
UPR proteinsMM cellsMacrophagesOCs terminal differentiationIn vitroBone resorptionPrognostic, Therapeutic target[80]
piRNA-823MM cells, serumECsIncreased survival, proliferation, and angiogenesisIn vitro and ex vivoIncreased angiogenesis, poor prognosisPrognostic,
Therapeutic target
[100]
Summary of the cited studies on the role of EVs in MM. The specific EVs cargo composition, source, EVs molecular and biological effects, and their clinical implications are listed. * Prognostic value of the specific cargo molecule wasn’t directly addressed in the indicated study. SMM: Smouldering Multiple Myeloma; MM: Multiple Myeloma; MGUS: Monoclonal gammopathy of undetermined significance; OCs: Osteoclasts; PFS: Patient Free Survival; MSCs: Mesenchymal Stromal Cells, BMSCs: Bone Marrow Stromal Cells; MDSCs: Myeloid-Derived Suppressor Cells; ADO: Adenosine; ECs: Endothelial Cells; OS: Overall Survival.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Saltarella, I.; Lamanuzzi, A.; Apollonio, B.; Desantis, V.; Bartoli, G.; Vacca, A.; Frassanito, M.A. Role of Extracellular Vesicle-Based Cell-to-Cell Communication in Multiple Myeloma Progression. Cells 2021, 10, 3185. https://doi.org/10.3390/cells10113185

AMA Style

Saltarella I, Lamanuzzi A, Apollonio B, Desantis V, Bartoli G, Vacca A, Frassanito MA. Role of Extracellular Vesicle-Based Cell-to-Cell Communication in Multiple Myeloma Progression. Cells. 2021; 10(11):3185. https://doi.org/10.3390/cells10113185

Chicago/Turabian Style

Saltarella, Ilaria, Aurelia Lamanuzzi, Benedetta Apollonio, Vanessa Desantis, Giulia Bartoli, Angelo Vacca, and Maria Antonia Frassanito. 2021. "Role of Extracellular Vesicle-Based Cell-to-Cell Communication in Multiple Myeloma Progression" Cells 10, no. 11: 3185. https://doi.org/10.3390/cells10113185

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