Multiple myeloma (MM) remains an incurable plasma cell malignancy that develops in the bone marrow (BM). This BM is partially responsible for protecting the MM cells against current standard-of-care therapies and for accommodating MM-related symptoms such as bone resorption and immune suppression. Increasing evidence has implicated extracellular vesicles (EV), including exosomes in the different processes within the BM. Exosomes are <150-nm-sized vesicles secreted by different cell types including MM cells. These vesicles contain protein and RNA cargo that they deliver to the recipient cell. In this way, they have been implicated in MM-related processes including osteolysis, angiogenesis, immune suppression, and drug resistance. Targeting exosome secretion could therefore potentially block these different processes. In this review, we will summarize the current findings of exosome-related processes in the BM and describe not only the current treatment strategies to counter them but also how exosomes can be harnessed to deliver toxic payloads. Finally, an overview of the different clinical studies that investigate EV cargo as potential MM biomarkers in liquid biopsies will be discussed.

In multiple myeloma (MM), malignant plasma cells (PCs) expand within the bone marrow (BM), leading to typical symptoms such as bone lesions, anemia, hypercalcemia, and recurrent infections. The worldwide incidence is currently 160 000.1 MM is often preceded by a precancerous asymptomatic stage called monoclonal gammopathy of undetermined significance (MGUS). Fifteen percent of patients with MGUS evolve to MM, with or without an intermediate state of smoldering MM (SMM). Currently, the percentage of BM PCs is used to stratify patients with MM, whereby PCs are defined as CD138+/CD38+. However, CD138 can be shed from the membrane, indicating a need for novel biomarkers.2 

Despite the development of novel treatment strategies, MM remains incurable; most MM patients relapse and become refractory to all therapies.3 The BM microenvironment is known to play a crucial role in both progression and relapse.4 Much effort has been invested in finding therapeutic targets by understanding niche-tumor crosstalk; however, this has mostly led to disappointing clinical impact. In recent years, a new communication mode has emerged, namely secreted extracellular vesicles (EVs). These EVs encompass a heterogeneous group of vesicles consisting of microvesicles (MVs), apoptotic bodies, exosomes, and exomeres, depending on their size and secretion pathway. Apoptotic bodies are the largest EVs (1-5 μm) that are released from the plasma membrane during late stages of apoptosis, whereas MVs are a heterogeneous group of vesicles with a size ranging from 100 nm to 1 μm, which bleb from the plasma membrane. Exomeres are small (<50 nm) nonmembranous vesicles.5 

In this review, we mainly focus on the role of exosomes in MM development and therapy and their possible value as biomarkers.

Exosome biogenesis and uptake6 

Exosomes are small vesicles (30-150 nm), defined by their endosomal origin. The process of exosome formation and uptake is described in detail in Figure 1. Briefly, early endosomes will mature into multivesicular bodies that will fuse with the plasma membrane to release their content as exosomes. Biogenesis of exosomes is either ESCRT (endosomal sorting complex required for transport) dependent, whereby ESCRT proteins form complexes with syndecans via syntenin,7 or it can be independently8 regulated by tetraspanins or the lipid ceramide.9 Exosome release is mediated by Rab GTPases and SNARE proteins.10,11 Their cargo includes lipids, proteins, small RNA, and possibly DNA.12 

Figure 1.

Exosome biogenesis and uptake. (A) [1] The process of exosome formation begins with the inward budding of the plasma membrane to form primary endocytic vesicles, which fuse together to create early endosomes. [2] Early endosomes mature into late endosomes at which point a second inward budding occurs to form intraluminal vesicles (ILVs). Late endosomes that contain several ILVs are called multivesicular bodies (MVBs). Biogenesis of exosomes can be either ESCRT (endosomal sorting complex required for transport) dependent or independent. ESCRT proteins (including TSG-101) can be divided in four multimeric complexes (ESCRT-0, -I, -II, -III) each with their defined role in vesicle formation. The accessory proteins Bro1/ALG-2-interacting protein X (ALIX) and vacuolar protein sorting (VPS4) ATPase are involved in stabilizing the complex. Syntenin is a multivalent protein that binds the cytosolic domain of syndecan but also directly interacts with ALIX. ESCRT independent exosome biogenesis involves lipids such as ceramide or tetraspanins. ESCRT dependent and independent mechanisms most likely also work together. [3] The last step in exosome biogenesis is their release into extracellular space. This step includes the transport of MVBs to the plasma membrane, followed by their docking and fusion. This process is regulated by proteins involved in cytoskeletal rearrangements and fusion machinery such as the Rab family of GTPases and the SNARE (soluble NSF attachment protein receptors) family proteins. Different Rab proteins have been implicated in vesicular trafficking of which Rab27a/b are best described. The SNARE protein family encompasses more than 60 members, which induce membrane fusion. The best described SNAREs involved in exosome release are VAMP3 and VAMP7. (B) Different uptake mechanisms have been described. [1] Specific uptake through receptors present on the membrane which is dependent on which ligands are expressed on the membrane of the exosomes. [2] Pinocytosis, during which actin-driven membrane ruffling is triggered in the recipient cell. Lamellipodia will form pinocytic cups in which exosomes, bound to the membrane, are “captured”. Once the pinocytic cups close, they are termed pinocytomes, which will shrink to the size of endosomes. [3] Clathrin-mediated endocytosis is a receptor mediated process whereby clathrin-coated vesicles will be formed, followed by invagination of the membrane and fusion to endosomes. Dynamin 2, clathrin and adaptor protein 2 (AP2) are the best characterized proteins involved in this process. Caveolin-dependent endocytosis is similar to clathrin but involves the presence of caveolae, small plasma membrane invaginations rich in caveolin, cholesterol and sphingolipids. [4] Plasma membrane fusion, during which the membrane of the exosome directly merges with the plasma membrane, releasing exosome content into the cytosol. Rab and SNARE family proteins contribute to this process. This figure was created with biorender.com.

Figure 1.

Exosome biogenesis and uptake. (A) [1] The process of exosome formation begins with the inward budding of the plasma membrane to form primary endocytic vesicles, which fuse together to create early endosomes. [2] Early endosomes mature into late endosomes at which point a second inward budding occurs to form intraluminal vesicles (ILVs). Late endosomes that contain several ILVs are called multivesicular bodies (MVBs). Biogenesis of exosomes can be either ESCRT (endosomal sorting complex required for transport) dependent or independent. ESCRT proteins (including TSG-101) can be divided in four multimeric complexes (ESCRT-0, -I, -II, -III) each with their defined role in vesicle formation. The accessory proteins Bro1/ALG-2-interacting protein X (ALIX) and vacuolar protein sorting (VPS4) ATPase are involved in stabilizing the complex. Syntenin is a multivalent protein that binds the cytosolic domain of syndecan but also directly interacts with ALIX. ESCRT independent exosome biogenesis involves lipids such as ceramide or tetraspanins. ESCRT dependent and independent mechanisms most likely also work together. [3] The last step in exosome biogenesis is their release into extracellular space. This step includes the transport of MVBs to the plasma membrane, followed by their docking and fusion. This process is regulated by proteins involved in cytoskeletal rearrangements and fusion machinery such as the Rab family of GTPases and the SNARE (soluble NSF attachment protein receptors) family proteins. Different Rab proteins have been implicated in vesicular trafficking of which Rab27a/b are best described. The SNARE protein family encompasses more than 60 members, which induce membrane fusion. The best described SNAREs involved in exosome release are VAMP3 and VAMP7. (B) Different uptake mechanisms have been described. [1] Specific uptake through receptors present on the membrane which is dependent on which ligands are expressed on the membrane of the exosomes. [2] Pinocytosis, during which actin-driven membrane ruffling is triggered in the recipient cell. Lamellipodia will form pinocytic cups in which exosomes, bound to the membrane, are “captured”. Once the pinocytic cups close, they are termed pinocytomes, which will shrink to the size of endosomes. [3] Clathrin-mediated endocytosis is a receptor mediated process whereby clathrin-coated vesicles will be formed, followed by invagination of the membrane and fusion to endosomes. Dynamin 2, clathrin and adaptor protein 2 (AP2) are the best characterized proteins involved in this process. Caveolin-dependent endocytosis is similar to clathrin but involves the presence of caveolae, small plasma membrane invaginations rich in caveolin, cholesterol and sphingolipids. [4] Plasma membrane fusion, during which the membrane of the exosome directly merges with the plasma membrane, releasing exosome content into the cytosol. Rab and SNARE family proteins contribute to this process. This figure was created with biorender.com.

Close modal

Exosomes can exert their function either by presenting molecules on their membrane or by releasing their cargo after internalization. The mechanisms behind EV uptake and cargo delivery are still poorly defined but include macro/micropinocytosis, clathrin-mediated endocytosis, caveolin-dependent endocytosis, plasma membrane fusion, and specific uptake through membrane receptors.13 

Exosome markers and isolation techniques

Minimal information for studies of extracellular vesicles guidelines14 dictate that exosomes should be characterized by a minimum of 3 protein markers: 1 transmembranic protein (including CD63, CD9, and CD82) associated with the plasma membrane and/or endosomes, 1 cytosolic protein (including elements of the ESCRT such as TSG101 and/or ALIX) or flotillins (Flot 1 and 2) that associate with membrane microdomains, and 1 negative marker for other intracellular proteins such as calreticulin, calnexin, prohibitin, or GM130. HSP70, ARF6, and tubulin are also valid as cytosolic markers, whereas syntenin has been put forward as the most abundant protein in exosomes.15 

A wide variety of isolation techniques14,16 is available to isolate exosomes, each with certain recovery/specificity. Precipitation kits such as Exoquick solutions or the ExoEasy kit are fast, accessible, and have a high yield but low specificity, making extra purification measures necessary. This technique should be approached with caution, especially for biomarker studies as it is nearly impossible to exclude coprecipitated cargo. Differential ultracentrifugation remains the most commonly used technique to isolate EVs17 and is considered to have intermediate specificity and yield but has as a drawback the need for heavy equipment that is not always available. Density gradient can further improve purity of exosome fractions by removing non-EV contaminants. Size exclusion chromatography is a valid alternative with similar characteristics. Combinations of different isolation techniques will increase specificity, which is necessary to detect specific exosomal cargo. One paper described that exosomes from serum from patients with MM, obtained by iodixanol and sucrose gradients, was pure, but those achieved with limited processing (serial centrifugation or 1-step precipitation kits) resulted in contamination by a residual matrix, embedding the exosomes.18 An overview of the different isolation techniques and markers for EV studies in MM are listed in Table 1.

Table 1.

Overview of the different isolation and marker strategies, used in MM studies

Vesicle typeIsolation methodProtocolSize MarkersReference
exo Isolation kits, ∗∗∗also used UC Exoquick ≤150 nm CD63, CD81,+Alix∗ or +Tsg101∗∗ 22,26,46,49,45∗,51,54∗∗,63∗∗∗ 
   CD63, CD81, Tsg101, syntenin 34  
   Hsp70, flotilin 1 60  
   Hsp70, CD63, CD9,+Alix∗ or +LAMP2∗∗ calreticulin negative 56,67∗,61∗∗ 
   HspP70/90, Alix, Tsg101, flotilin 1, calreticulin negative 23  
   Hsp70/90, flotilin 1, CD63,+Alix∗ calreticulin negative 42,52,65∗ 
   CD63, CD9, flotilin 1, calreticulin negative 77  
 ExoEasy kit ≤150 nm Tsg101 59  
  CD63, CD81 57,58  
 Total exosome isolation kit ≤150 nm CD63, Hsp70 50  
Differential centrifugation ≤100 nm LAMP1, Alix, galectin negative 78  
 ≤100 nm Alix, CD63, calnexin negative no explicit markers∗ 31,32,35∗,36∗ 
 100 nm Hsp70, CD63, Tsg101 25  
 ≤150 nm Rab5, LAMP2, CD63, GM130 negative 80  
 ≤150 nm Hsp70, Tsg101, CD81, CD63, calreticulin negative, no markers∗ only Hsp70∗∗ 44∗,47∗∗,48  
∗Also used exoquick for patient material   CD63, flotilin 1, clathrin only CD63∗ 62,72,73∗ 
 150 nm Tsg101, Alix, GM130, and α-tubulin negative 33  
∗Also used different kits for patient material ≤150 nm Tsg101, CD81 68∗ 
 ≤200 nm CD63, no markers∗ 27,66∗,69  
 50-350 nm Alix, CD9, LAMP1 40  
UC 100 000g, 1 h ≤100 nm CD63, Hsp70 37  
 120 000g, 2 h 150 nm CD63, Alix, Tsg101, Rab5 53  
MV Centrifugation 13 000g ≤200 nm No markers 24  
 16 000g No markers 28  
  10 99% 100 nm, 1%> 300 nm No markers 29  
  11 100 nm to 1μm Alix, Tsg101, CD63, CD9 39  
  12 ≤200 nm No markers 68  
ApoEV  13 150 nm CD63, CD9, flotilin 1, Alix, syntenin, annexin 79  
MV or exo? Concentration via centrifugal filters  150 nm Alix, CD81 76  
Vesicle typeIsolation methodProtocolSize MarkersReference
exo Isolation kits, ∗∗∗also used UC Exoquick ≤150 nm CD63, CD81,+Alix∗ or +Tsg101∗∗ 22,26,46,49,45∗,51,54∗∗,63∗∗∗ 
   CD63, CD81, Tsg101, syntenin 34  
   Hsp70, flotilin 1 60  
   Hsp70, CD63, CD9,+Alix∗ or +LAMP2∗∗ calreticulin negative 56,67∗,61∗∗ 
   HspP70/90, Alix, Tsg101, flotilin 1, calreticulin negative 23  
   Hsp70/90, flotilin 1, CD63,+Alix∗ calreticulin negative 42,52,65∗ 
   CD63, CD9, flotilin 1, calreticulin negative 77  
 ExoEasy kit ≤150 nm Tsg101 59  
  CD63, CD81 57,58  
 Total exosome isolation kit ≤150 nm CD63, Hsp70 50  
Differential centrifugation ≤100 nm LAMP1, Alix, galectin negative 78  
 ≤100 nm Alix, CD63, calnexin negative no explicit markers∗ 31,32,35∗,36∗ 
 100 nm Hsp70, CD63, Tsg101 25  
 ≤150 nm Rab5, LAMP2, CD63, GM130 negative 80  
 ≤150 nm Hsp70, Tsg101, CD81, CD63, calreticulin negative, no markers∗ only Hsp70∗∗ 44∗,47∗∗,48  
∗Also used exoquick for patient material   CD63, flotilin 1, clathrin only CD63∗ 62,72,73∗ 
 150 nm Tsg101, Alix, GM130, and α-tubulin negative 33  
∗Also used different kits for patient material ≤150 nm Tsg101, CD81 68∗ 
 ≤200 nm CD63, no markers∗ 27,66∗,69  
 50-350 nm Alix, CD9, LAMP1 40  
UC 100 000g, 1 h ≤100 nm CD63, Hsp70 37  
 120 000g, 2 h 150 nm CD63, Alix, Tsg101, Rab5 53  
MV Centrifugation 13 000g ≤200 nm No markers 24  
 16 000g No markers 28  
  10 99% 100 nm, 1%> 300 nm No markers 29  
  11 100 nm to 1μm Alix, Tsg101, CD63, CD9 39  
  12 ≤200 nm No markers 68  
ApoEV  13 150 nm CD63, CD9, flotilin 1, Alix, syntenin, annexin 79  
MV or exo? Concentration via centrifugal filters  150 nm Alix, CD81 76  

exo, exosomes; MV, microvesicles; UC, ultracentrifugation.

∗,∗∗,∗∗∗Asterisks refer to which marker or isolation method is exceptionally used (in addition to the generally listed markers) in the corresponding reference.

Size was measured via transmission electron microscope, and/or nanoparticle tracking analysis, dynamic light scattering, or flow cytometry. Different differential centrifugation protocols were used: (1) 300g (5 minutes), 1200g (20 minutes), 10 000g (30 minutes), 100 000g (60 minutes); (2) 300g (5 minutes), 3000g (15 minutes), 10 000g (30 minutes), 100 000g (90 minutes); (3) 800g (30 min), 16 000g (45 min), 100 000g (2 hours); (4) 300g (10 minutes), 4000g (20 minutes), 100 000g (4 hours + 1 hours); (5) 300g (10 minutes), 2000g (20 minutes), 100 000g (30/70 minutes); (6) 450g (5 minutes), 16 000g (15 minutes), 120 000g (20 minutes); (7) 500g (5 minutes), 2000g (10 minutes), 10 000g (30 minutes), 100 000g (60 minutes); (8) 2000g (30 minutes), 20 000g (70 minutes), 170 000g (90 minutes); (9) 300g (10 minutes), 12 000g (45 minutes), 100 000g (90 minutes). Different centrifugation protocols were used: (10) 1500g (15 minutes), 20 000g (60 minutes); (11) 200g, (5 minutes), 1500g (30 minutes), 16 000g (60 minutes); (12) 500g (5 minutes), 2000g (10 minutes), 10 000g (30 minutes); (13) 800g (10 minutes), 2000g (10 minutes), 16 000g (30 minutes).

Because most isolation techniques do not solely isolate exosomes, it has been proposed to use the term “small EVs” when considering exosomes. As characterization and functionality of exosomes/EVs are very method dependent, the International Society of Extracellular Vesicles has introduced EV-Track where authors can upload their manuscript to receive an EV-Metric score, based on components, which were argued to be indispensable for unambiguous interpretation and reproduction of EV experiments.19 

Increasing evidence has demonstrated that EVs, including exosomes, play a role in most of the BM-MM interactions and processes. These will be listed below and are depicted in Figure 2.

Figure 2.

Role of EVs in the MM microenvironment. MM cells reside in the hypoxic BM milieu, which will trigger MM EV release. These EVs will stimulate angiogenesis by delivering pro-angiogenic cargo including VEGF, miR-135b and piRNA-823, which will trigger proliferation and tube formation in ECs. Osteolysis will be activated by inducing osteoclast proliferation and activation through the delivery of EV cargo (AREG, IL32 and sXBP1) and by blocking osteoblast differentiation and activity through inhibitory cargo such as AREG, DKK1, lncRUNX2-AS1, and miR-129-5p. Moreover, osteoblasts are stimulated to secrete cMet and IL11 for the osteoclasts. Immune suppression is induced by the presence of CD73 and CD39, and activation of MDSC and TAMs via miR-1305 EV delivery, who in turn will inhibit NK- and T-cell proliferation. MM EVs have a direct impact on T cells by inhibiting CD8 activation and inducing T regulatory cell expansion. Direct effects on NK cells are more inconclusive and only occur after treating MM cells with cytotoxic compounds, releasing “chemoexosomes” (depicted with a ∗). IFNγ is released in response to chemoexosomes, but NK activation is uncertain. These chemoexosomes also contain cell cycle proteins and acid SMase, thereby inducing auto-resistance. Finally, MM cells trigger BMSC to transform into CAFs by delivering mir-146a, mir-21, WWC2, CD166, and CD44 as EV cargo. Transition toward CAFs is accompanied by miR-27b-3p and miR-214-3p upregulation. CAFs in turn will release growth and survival cargo to MM cells, containing IL6, FN, PSMA3, Circ_0007841, Bcl-XL, miR-10, miR-214-3p, miR-5100 and LINC00461. They will also stimulate TAMs via delivery of miR-let-7c. A downward arrow indicates less expression of specific cargo in tumoral EVs. This figure was created with biorender.com.

Figure 2.

Role of EVs in the MM microenvironment. MM cells reside in the hypoxic BM milieu, which will trigger MM EV release. These EVs will stimulate angiogenesis by delivering pro-angiogenic cargo including VEGF, miR-135b and piRNA-823, which will trigger proliferation and tube formation in ECs. Osteolysis will be activated by inducing osteoclast proliferation and activation through the delivery of EV cargo (AREG, IL32 and sXBP1) and by blocking osteoblast differentiation and activity through inhibitory cargo such as AREG, DKK1, lncRUNX2-AS1, and miR-129-5p. Moreover, osteoblasts are stimulated to secrete cMet and IL11 for the osteoclasts. Immune suppression is induced by the presence of CD73 and CD39, and activation of MDSC and TAMs via miR-1305 EV delivery, who in turn will inhibit NK- and T-cell proliferation. MM EVs have a direct impact on T cells by inhibiting CD8 activation and inducing T regulatory cell expansion. Direct effects on NK cells are more inconclusive and only occur after treating MM cells with cytotoxic compounds, releasing “chemoexosomes” (depicted with a ∗). IFNγ is released in response to chemoexosomes, but NK activation is uncertain. These chemoexosomes also contain cell cycle proteins and acid SMase, thereby inducing auto-resistance. Finally, MM cells trigger BMSC to transform into CAFs by delivering mir-146a, mir-21, WWC2, CD166, and CD44 as EV cargo. Transition toward CAFs is accompanied by miR-27b-3p and miR-214-3p upregulation. CAFs in turn will release growth and survival cargo to MM cells, containing IL6, FN, PSMA3, Circ_0007841, Bcl-XL, miR-10, miR-214-3p, miR-5100 and LINC00461. They will also stimulate TAMs via delivery of miR-let-7c. A downward arrow indicates less expression of specific cargo in tumoral EVs. This figure was created with biorender.com.

Close modal

Angiogenesis

The MM burden increases the natural hypoxia of the BM,20 thereby triggering angiogenesis.21 Exosomes derived from hypoxia-resistant MM cells were found to contain high levels of the microRNA miR-135b that suppressed its target “factor–inhibiting hypoxia inducible factor 1” when transferred into endothelial cells (ECs), leading to their proliferation in vitro and in vivo.22 Our group demonstrated that MM exosomes induced EC proliferation and tube formation via Stat3 phosphorylation. Moreover, we identified multiple angiogenic factors as cargo proteins, including angiogenin, basic fibroblast growth factor, and vascular endothelial growth factor (VEGF).23 More recently, MM MVs were found to contain the small silencing piwi-interacting RNA piRNA-823, which when transferred to ECs, promoted proliferation, invasion, and tube formation by enhancing expression of VEGF, interleukin 6 (IL-6), and ICAM-1. piRNA-823–transfected ECs enhanced tumor growth when co-inoculated with MM cells in a xenograft model.24 Exosomes from MM patient serum were found to induce more proliferation in ECs compared with MGUS exosomes. This was associated with an altered intracellular c-Src distribution and activation of the nuclear factor κB (NF-κB) pathway.25 By contrast, exosomes derived from bone marrow stromal cells (BMSCs) isolated from healthy young people strongly inhibited angiogenesis. This was linked to high expression of miR-340, which could inhibit the c-Met signaling pathway.26 Interestingly, treating MM cells with C6 ceramide blocked tube formation by delivering increased levels of exosomal miR-29b that blocked the PI3K-Akt pathway in ECs.27 Treating MM cells with bortezomib also altered the composition of MM MVs, with more proinflammatory cytokines and less proangiogenic cytokines such as VEGF and angiogenin. Furthermore, these MVs inhibited NF-κB signaling in human umbilical vein endothelial cells leading to less proliferation and migration/tube formation.28,29 

Osteolysis

MM cells trigger osteolysis by activating osteoclasts and inhibiting osteoblasts.30 MM exosomes were found to induce osteoclast formation and activity by activating the pAkt pathway.31 They contained activators of the unfolded protein response pathway, triggering IRE1α activation and downstream signaling.32 Also the presence of IL-32, which was upregulated in response to hypoxia, induced osteoclast activity both in vitro and in vivo.33 We further demonstrated that MM exosomes increased osteoclast formation and activity while simultaneously blocking osteoblast differentiation, inducing osteoblast apoptosis by downregulating Runx2, Osterix, and collagen 1A1 via the presence of DKK1 cargo. Injecting healthy mice with MM exosomes was sufficient to trigger bone disease.34 Similarly, amphiregulin was found to be present in MM exosomes, which could bind to epidermal growth factor receptor both in osteoclasts precursors and mesenchymal stem cells (MSCs), thereby activating the former and blocking the differentiation of the latter into osteoblasts.35 More recently it was discovered that miR-129-5p was upregulated in MM exosomes compared with SMM exosomes. This micro RNA (miRNA), once transferred to MSCs, inhibited the transcription factor Sp1 and its target alkaline phosphatase, thereby blocking osteoblast differentiation.36 MM exosomes could also block osteoblast differentiation from BMSC, by triggering IL-6 secretion from BMSCs via the Ape1/NF-κB pathway.37 In line with this, the presence of the long noncoding antisense RNA LncRUNX2-AS1 in human myeloma cell line (HMCL)-derived exosomes impaired osteogenic differentiation of MSCs by negatively regulating RUNX2 expression.38 Addition of MM MVs to MSC also induced elevated expression of miR-103a-3p, thereby inhibiting osteogenesis. Injecting MM MVs in a xenograft model further exacerbated MM bone disease.39 Finally, the presence of hepatocyte growth factor on exosomes of a subtype of patients with MM could induce c-Met signaling in osteoblast-like cells, thereby inducing the release of the osteoclast activating cytokine IL-11.40 

Immunosuppressive environment

The BM environment in MM is immunosuppressive because of the presence of cells such as myeloid derived suppressor cells (MDSCs) and tumor associated macrophages (TAM)s.41 Our group has demonstrated that MM- and MM BMSC-derived exosomes induced MDSC expansion and activation through pStat1 and 3 signaling, in vitro and in vivo. Treating healthy mice with MM exosomes increased the prevalence and activity of MDSCs, leading to inhibition of T-cell proliferation.23,42 Moreover, MVs isolated from the BM plasma of patients with MM, contained increased levels of the ectoenzymes CD39 and CD73, resulting in increased levels of adenosine, which can activate MDSC and inhibit T-cell function.43 Exosomes from HMCL, specifically with del13, could induce polarization of peripheral blood monocytes into M2 TAMs via the NF-κB pathway. These exosomes contained less miR-16, which was found to inhibit IKKa/B.44 Moreover, under hypoxia, MM cells were able to secrete increased amounts of exosomes containing, among others, miR-1305, which when transferred to THP1 monocytes, induced M2 polarization.45 Also, MM-MSC cells could stimulate polarization of PBMC-derived macrophages into M2 macrophages via the transfer of miR-let-7c.46 Available results are conflicting concerning the effect of MM exosomes on natural killer (NK) cells, which are activated by recognizing molecular patterns of ligands such as danger-associated molecular patterns or pathogen-associated molecular patterns. Two reports suggested that treatment of MM cells with low doses of genotoxic drugs such as melphalan induced a higher secretion of exosomes that could trigger NK proliferation, activation (CD69 expression), and interferon γ (IFNγ) release through the presentation of IL-15(RA)47 or via binding of HSP70 to TLR2.48 However, a more recent report indicated that MM exosomes could induce IFNγ release but also reduced NK cytotoxicity. Treating MM cells with either eicosapentaenoic or docosahexaenoic acid reduced the immunosuppressive effect while keeping the immune stimulatory response.49 Of note, in this report, the MM cells were not treated with a genotoxic drug, which could explain the different outcome. Finally, it was found that MM exosomes tended to induce apoptosis of CD4+ T cells derived from healthy donors (HDs) and patients with MM but increased viability in CD8+ T cells. However, the cytotoxic capacity of these CD8+ T cells was decreased. Furthermore, the viability of HD T regulatory cells increased, indicating that MM exosomes alter T-cell phenotype to a more suppressive profile.50 

Drug resistance

Several reports have evaluated whether exosomes are involved in the protection of MM cells by the BM environment. Patient BM-MSC–derived exosomes were found to induce proliferation and survival of MM cells, whereas those of healthy BM-MSCs reduced MM viability. MM BM-MSCs contained lower levels of the tumor suppressor miR-15a and higher levels of fibronectin and IL-6.51 Similarly, we found that BMSC-derived exosomes induced MM proliferation and survival via activation of different prosurvival pathways including pAkt, p53, and JNK. More importantly, BMSC exosomes could induce bortezomib resistance by increasing Bcl2 levels.52 BMSC exosomes also contained caspase3-cleaved Bcl-XL, which not only conveyed antiapoptotic signals to recipient MM cells but was necessary for exosomal uptake.53 Furthermore, BMSC could increase MM cell proliferation via miR-10a transfer. Intriguingly, blocking exosome secretion using the S1P receptor modulator FTY720 induced BMSC apoptosis by an intracellular overload of miR-10a, indicating that both cell types respond differently to the presence of miR-10. PTEN and CDK6 were affected in BMSC, whereas SMAD4 seemed to be a target in MM cells. Treatment of a xenograft with FTY720 led to a reduction in the number of MM-BMSC and abolished their prosurvival effect on the MM cells.54 Further investigation into the content of BMSC exosomes revealed that miR-573, miR-544, miR-545, miR-382, miR-16 (all down), and miR-10a (up) were able to distinguish MM patient BMSCs from MGUS and HD. Target gene analysis suggested EPHA8 as a target in MM for miR-10a, whereas miR-16 negatively impacted cyclin D1 and IGFR1.55 MM BMSC exosomes also contained miR-23b-3p, miR-27b-3p, miR-125b-5p, miR-214-3p, and miR-5100. However, MM cells only incorporated miR-214-3p and miR-5100, which induced MM cell proliferation and drug resistance via downregulation of PTEN.56 Moreover, LINC00461, present in BM MSC exosomes, was discovered to be a sponge for miR-15a/16 after transfer into MM cells, thereby releasing BCL-2, leading to enhanced MM survival.57 Exosomal transfer of the circular RNA Circ_0007841 from BM-MSCs to MM cells could also induce survival, proliferation, and migration of MM cells via targeting of the miR-338-3p/BRD4 axis, which activated the PI3K/AKT signaling pathway.58 BMSCs were also found to have high levels of HDAC3 in the presence of MM cells, which in turn supported MM proliferation. Targeting HDAC3 in BMSC, induced changes in their exosome output both on a quantitative and qualitative level, including downregulation of prosurvival miR-380, miR-382, miR-15b, miR-9986, and miR-5191, which led to MM cell growth arrest.59 To further underscore the role of exosomes in drug resistance, it was found that exosomes derived from MSCs of proteasome inhibitor-resistant patients induced resistance in sensitive HMCL. PSMA3 mRNA and the long noncoding RNA (lncRNA) PSMA3-AS1, which codes for 1 of the proteasome subunits, was strongly present and increased proteasome activity after transfer to MM cells. Targeting PSMA3-AS1, reduced tumor burden in a xenograft model and synergized with carfilzomib treatment.60 It was further demonstrated that bortezomib-resistant MM cells could transfer resistance to sensitive cells via exosomal HSP70 delivery. Intravenous immunoglobulin G treatment, which contained different immunoglobulins including antibodies against HSP70, caused apoptosis of MM cells because of an interruption of this loop, thereby enhancing sensitivity to bortezomib both in vitro and in vivo.61 

Not only the BM environment can induce drug resistance via exosomes, MM cells can also autologously induce therapy resistance. Treating MM cells with proteasome inhibitors led to increased secretion of exosomes, coined “chemoexosomes,” with an altered proteome profile, enriched in proteins that regulate cell cycle. Transfer of these exosomes to other MM cells enhanced proliferation and survival signals including pERK.62 Our group observed an increase in acid sphingomyelinase (ASM) expression in MM cell lines treated with melphalan or bortezomib, as well as in their exosomes. Exosomes high in ASM content were able to transfer the drug-resistant phenotype to chemosensitive cells, thereby suggesting a tumor-protective role for ASM.63 MM cells can also stimulate BMSCs to become cancer associated fibroblasts (CAFs) via exosomes, creating a mutual feedback loop. CAFs express fibroblast-specific protein 1 (FSP1) and, on activation by cancer cells, α-smooth muscle actin (αSMA) and fibroblast activation protein (FAP).64 Our group found that MM cells could transfer miR-146a via exosomes to MSCs, triggering through the NOTCH pathway enhanced cytokine secretion including IL-6 and CXCL10. This in turn stimulated MM growth and migration.65 MiR-146a transfer led to upregulation of αSMA and FAP in MSCs.66 MM exosomes further increased expression of miR-27b-3p and miR-214-3p in fibroblasts from patients with MGUS through transfer of exosomal WWC2, which regulates the Hippo pathway. These miRNAs modulated MCL1 via FBXW7 and PTEN and enhanced the expression of the CAF markers αSMA and FAP.67 Finally, when comparing the impact of HMCL derived exosomes to patient plasma-derived exosomes on healthy BMSCs, it was found that both were capable of inducing BMSC proliferation, whereas plasma exosomes also stimulated migration and adhesion to MM cells via the transfer of migration/adhesion related proteins (ie, MYH4, CD166, CD44, ANXA2, and FN1).68 

Strategies to successfully block exosome secretion are still limited. We targeted exosome secretion using the sphingomyelinase inhibitor GW4869, which blocks the conversion of sphingomyelin into ceramide. This inhibitor reduced the release of exosomes, countered myeloma bone disease, and significantly enhanced the antitumor effect of bortezomib34 in the 5T33MM murine model. Another report indicated that GW4869 decreased exosome secretion from MM cells but decreased at the same time the level of EV-related tumor suppressive miRNAs (miR-202, 15a, 16, and 29b). By contrast, C6-ceramide treatment increased secretion of exosomes with a higher load of tumor suppressive miRNAs, leading to reduced viability in neighboring MM cells.69 Heparanase, an endoglycosidase present in the BM that cleaves heparan sulfate, is a possible target to block exosome release and uptake.70 Endosomal heparanase allows clustering of syndecans, by trimming heparin sulfate from syndecans, thereby recruiting them via ALIX to ESCRT.71 Exogenous heparanase could enhance exosome secretion of MM cells with increased levels of syndecan-1, VEGF, hepatocyte growth factor,72 and heparanase62 as exosomal cargo. These exosomes stimulated adhesion, ECM degradation and MM invasion in surrounding BM cells. On the receiving side, heparan sulfate chains were able to function as receptors for exosomes via mutual binding to fibronectin, after which exosomes were internalized, thereby activating downstream signaling pathways such as p38 and ERK. More importantly, the heparanase inhibitor Roneparstat, capable of inhibiting exosome binding to the cells,73 has been shown to have potent anti-MM activity in vivo74 and is currently being evaluated in a phase 1 clinical study in patients with advanced refractory myeloma.75 Related to this, overexpression of miR-1252-5p in MM cells was discovered to reduce heparanase levels. Accordingly, mimics of this miRNA, when electroporated into HEK293T cell MVs, led to enhanced bortezomib sensitivity in MM cells.76 Heparin treatment also reduced uptake of BMSC exosomes in MM cells by 50%, whereas inhibitors of clathrin- and caveolin-dependent endocytosis were at least equally effective. The dynamin inhibitor dynasore could block the protumoral effects of BMSC exosomes and synergized with bortezomib in a xenograft model.77 

By contrast, exosomes can also be harnessed to present tumor antigens to the immune system. J558 myeloma cells were engineered to express the endogenous P1A tumor antigen and a transgenic form of membrane-bound HSP70, which served as an adjuvant danger signal. Exosomes from these cells could stimulate maturation of DCs in vitro and stimulate CD4+ T helper 1 and P1A-specific CD8+ cytotoxic T-lymphocyte responses in vivo.78 Apoptotic extracellular vesicles (apoEVs) derived from staurosporine-treated apoptotic MSCs could induce MM cell apoptosis by presenting FasL and triggering the trafficking of Fas to the surface of the cells. Treating 5TGM1 mice with apoEVs significantly prolonged their survival.79 Another study investigated the possibility of engineering tumor necrosis factor–related apoptosis-inducing ligand+ K562 exosomes. Although these exosomes were effective in inducing apoptosis in tumor necrosis factor–related apoptosis-inducing ligand death receptor (DR)5+ lymphoma cells, they were less potent in the DR5DR4+KMS11 MM cell line.80 Intriguingly, exosomes can be armed with a payload to induce selective killing of target cells. The group of Kalluri has genetically engineered MSC exosomes (iExosomes) to carry short interfering RNA specific to oncogenic KrasG12D, which suppressed the growth of pancreatic cancer in different animal models.81 This study has kick-started a phase 1 clinical trial to evaluate dose and side effects of the iExosomes.82 Although no such study has been performed yet in MM, extrapolating these data to relevant MM oncogenes is a promising approach. Different myeloma targeting molecules such as anti-CD138 or anti-BCMA monoclonal antibodies could be engineered into the exosomes to direct them specifically to MM cells.

BM biopsies are still the gold standard to diagnose MM but have as a disadvantage that they are invasive and do not reflect spatial tumor heterogeneity. Liquid biopsies, whereby bodily fluids are used to isolate tumor material, have emerged as possible alternatives to circumvent these limitations. EVs, including exosomes, could serve as tumor material for biomarker discovery, either to diagnose patients with MM or to monitor them during treatment.83 An overview of the different biomarker trials can be found in Table 2.

Table 2.

Results of the different biomarkers studies in MM using different EVs

TypeSamplesMethodology BiomarkerImpactReference
Protein      
MV N=2 MM Serum
UC
Immunoblotting 
Monoclonal Ig (FLC) Detectable in MM MVs 84  
exo N=10 5T33MM mice Blood
Exoquick
Immunocapture 
Monoclonal Ig Detectable in MM EVs earlier than M protein 85  
MV N=8 ND, 26PR, 18CR, 14PD, 18HD Plasma
UC
FC 
CD138 Higher in MM vs HD, correlates to MM progression 86  
MV N=115ND, 48CR, 20HD Blood/BM
UC
FC 
CD138 Higher in MM vs HD, correlates to MM progression 87  
MV N=14ND, 30PR, 12CR, 18PD, 24HD Plasma
UC
FC 
CD138/P-gp+/CD34 High CD138-P-pg+CD34+EV count associated with unresponsiveness to treatment 88  
MV N=61ND, 5PD, 29HD Plasma
UC
FC 
CD138 Higher in MM vs HD, correlation with bone disease 39  
MV N=12 ND, 28 HD Serum
UC
FC 
CD38 Higher in MM vs HD 89  
MV N=27MM, 11 MGUS, 14 SMM BM plasma
UC
FC 
CD38 Higher in MM vs MGUS and SMM 43  
MV N=10 MM, 20 ND Serum
Bench centrifugation
FC 
CD138/CD38 Single and double positive EVs higher in MM vs HD
Double positive correlate with tumor burden 
90  
exo N=8 ND,16 PD 8 MGUS, 16 HD Plasma
Exoquick
FC 
CD163/CD206 Higher in MM vs MGUS, HD and RR 93  
exo N=233 ND (of which 32 immunoblot), 13 HD Serum
UC
Elisa/immunoblot 
CD44 Higher in MM vs HD 91  
RNA      
exo N=156 ND, 5 HD Serum
Exoquick
Small RNAseq 
Let-7b, let-7e, miR-106a, miR-106b, miR-155, miR-16, miR-17, miR-18a, miR-20a Lower in MM vs HD, let-7b and miR-18a predictors for PFS and OS 96  
exo N=3 MM, 18 HD UC
qRT-PCR 
miR-155 Lower in MM vs HD 97  
exo N=20 MM, 20S MM, 16 HD Serum
Exoquick
qRT-PCR 
let-7c-5p, let-7d-5p, miR-185-5p, miR-103a-3p, miR20a-5p Lower in MM vs SMM and HD 98  
   miR4505 and miR4741 Higher in MM vs SMM and HD  
exo N=48 ND, 16 HD Serum
Exoquick
Microarray/qRT-PCR 
let-7c-5p, let-7d-5p, miR-140-3p, miR-185-5p, and miR-425-5p Lower in MM vs HD, negative correlation with disease progression and tumor burden 99  
exo N=115 MM (73 responder, 42 non-responder) Serum
UC
miRNA array 
MiR-17-5p, miR-20a-5p, miR- 15a-5p and miR-16-5p Lower in bortezomib resistant patients 101  
exo N=5 bz resistant, 5 bz sensitive MM Serum
UC
RNAseq 
482 lncRNA and 2099 mRNA FFAR1 and SP9 were increased, potential diagnostic marker HIST1H2BG and ITIH2 were decreased in resistance 102  
exo N=122 MM, 54 HD Serum
Exoquick
qRT-PCR 
circ-Myc Higher in MM vs HD, correlated to 17p deletion and translocation of t(4;14) 103  
exo N=25 MM, 5 HD Serum
Exoquick
RNA seq/qRT-PCR 
265 upregulated and 787 downregulated circRNAs (MM vs HD) chr2:2744228–2 744 407+correlated to peripheral neuropathy 104  
exo N=20 MM, 5 HD Serum
Exoquick
RNA seq/qRT-PCR 
circATP10A Prognostic potential, correlated to vascular endothelial growth factor B levels 106  
exo N=20 MM, 5 HD Serum
Exoquick
qRT-PCR 
circ-G042080 Predictor myocard damage 105  
exo N=56 MM, 49 MGUS, 36 HD Serum
miCURY exosome isolation kit
lncRNA array 
PRINS Negative correlation with PC in BM 100  
TypeSamplesMethodology BiomarkerImpactReference
Protein      
MV N=2 MM Serum
UC
Immunoblotting 
Monoclonal Ig (FLC) Detectable in MM MVs 84  
exo N=10 5T33MM mice Blood
Exoquick
Immunocapture 
Monoclonal Ig Detectable in MM EVs earlier than M protein 85  
MV N=8 ND, 26PR, 18CR, 14PD, 18HD Plasma
UC
FC 
CD138 Higher in MM vs HD, correlates to MM progression 86  
MV N=115ND, 48CR, 20HD Blood/BM
UC
FC 
CD138 Higher in MM vs HD, correlates to MM progression 87  
MV N=14ND, 30PR, 12CR, 18PD, 24HD Plasma
UC
FC 
CD138/P-gp+/CD34 High CD138-P-pg+CD34+EV count associated with unresponsiveness to treatment 88  
MV N=61ND, 5PD, 29HD Plasma
UC
FC 
CD138 Higher in MM vs HD, correlation with bone disease 39  
MV N=12 ND, 28 HD Serum
UC
FC 
CD38 Higher in MM vs HD 89  
MV N=27MM, 11 MGUS, 14 SMM BM plasma
UC
FC 
CD38 Higher in MM vs MGUS and SMM 43  
MV N=10 MM, 20 ND Serum
Bench centrifugation
FC 
CD138/CD38 Single and double positive EVs higher in MM vs HD
Double positive correlate with tumor burden 
90  
exo N=8 ND,16 PD 8 MGUS, 16 HD Plasma
Exoquick
FC 
CD163/CD206 Higher in MM vs MGUS, HD and RR 93  
exo N=233 ND (of which 32 immunoblot), 13 HD Serum
UC
Elisa/immunoblot 
CD44 Higher in MM vs HD 91  
RNA      
exo N=156 ND, 5 HD Serum
Exoquick
Small RNAseq 
Let-7b, let-7e, miR-106a, miR-106b, miR-155, miR-16, miR-17, miR-18a, miR-20a Lower in MM vs HD, let-7b and miR-18a predictors for PFS and OS 96  
exo N=3 MM, 18 HD UC
qRT-PCR 
miR-155 Lower in MM vs HD 97  
exo N=20 MM, 20S MM, 16 HD Serum
Exoquick
qRT-PCR 
let-7c-5p, let-7d-5p, miR-185-5p, miR-103a-3p, miR20a-5p Lower in MM vs SMM and HD 98  
   miR4505 and miR4741 Higher in MM vs SMM and HD  
exo N=48 ND, 16 HD Serum
Exoquick
Microarray/qRT-PCR 
let-7c-5p, let-7d-5p, miR-140-3p, miR-185-5p, and miR-425-5p Lower in MM vs HD, negative correlation with disease progression and tumor burden 99  
exo N=115 MM (73 responder, 42 non-responder) Serum
UC
miRNA array 
MiR-17-5p, miR-20a-5p, miR- 15a-5p and miR-16-5p Lower in bortezomib resistant patients 101  
exo N=5 bz resistant, 5 bz sensitive MM Serum
UC
RNAseq 
482 lncRNA and 2099 mRNA FFAR1 and SP9 were increased, potential diagnostic marker HIST1H2BG and ITIH2 were decreased in resistance 102  
exo N=122 MM, 54 HD Serum
Exoquick
qRT-PCR 
circ-Myc Higher in MM vs HD, correlated to 17p deletion and translocation of t(4;14) 103  
exo N=25 MM, 5 HD Serum
Exoquick
RNA seq/qRT-PCR 
265 upregulated and 787 downregulated circRNAs (MM vs HD) chr2:2744228–2 744 407+correlated to peripheral neuropathy 104  
exo N=20 MM, 5 HD Serum
Exoquick
RNA seq/qRT-PCR 
circATP10A Prognostic potential, correlated to vascular endothelial growth factor B levels 106  
exo N=20 MM, 5 HD Serum
Exoquick
qRT-PCR 
circ-G042080 Predictor myocard damage 105  
exo N=56 MM, 49 MGUS, 36 HD Serum
miCURY exosome isolation kit
lncRNA array 
PRINS Negative correlation with PC in BM 100  

bz, bortezomib; CR, complete response; exo, exosomes; FC, flow cytometry; HD, healthy donors; MV, microvesicles; ND, newly diagnosed MM; PC, plasma cells; PD, progressive disease; PR, partial response; RR, relapsed/refractory; UC, ultracentrifugation.

Methodology shown in order of source, isolation technique, and characterization technique.

Initial proteomic studies have focused on evaluating previously identified prognostic markers on MVs. Both the monoclonal immunoglobulin free light chain84 and the monoclonal immunoglobulin of the B-cell receptor have been demonstrated on MM-derived MVs.85 When examining PC markers, it was found that CD138 was more highly expressed in MM MVs compared with HD, which correlated to therapeutic response and disease stage.86,87 In a follow-up study, the multidrug resistance protein P-glycoprotein, together with the stem cell marker CD34 on CD138 MVs in patients with MM, correlated to unresponsiveness to treatment. This could be a consequence of CD138 shedding or an indication of the existence of a resistant immature phenotype.88 The levels of CD138+ MVs in the peripheral blood also correlated positively with the number of MM bone lesions.39 In addition, CD38+ MVs were more present in BM plasma and/or serum of patients with MM compared with patients with MGUS and SMM.43,89 Furthermore, using a simplified EV isolation technique that could be more easily implemented in routine clinical practice, the presence of CD38+, CD138+, and CD38+/CD138+ MVs was identified in patients with MM, whereby double-positive EVs correlated to BM-PC percentage.90 Other proteins such as MHC class I and CD44 were also found to be enriched in exosomes, derived either from MM cell lines or patient sera.91,92 Other cell types that are involved in MM progress could also release possible EV biomarkers. For example, CD163+ (hemoglobin-haptoglobin receptor) and CD206+ (mannose receptor) exosomes, derived from TAMs, were significantly elevated in patients with newly diagnosed MM compared with patients with HD, MGUS, and MM both in remission and relapse.93 As an alternative approach, patients could be stratified to either MM, MGUS, or asymptomatic MM, using either surface plasmon resonance spectroscopy or Raman spectroscopy to evaluate exosome levels.94,95 

Concerning small RNA, a first study identified 22 miRNAs that were significantly lower in MM patient exosomes, among which let-7b and miR-18a, which were significant predictors for progression-free survival (PFS) and shorter overall survival (OS).96 In this study, downregulation of miR-155 was only reported to correlate with PFS; however, it was also lower in another small cohort of patients with MM.97 Similarly, several downregulated miRNA (including let-7c-5p, let-7d-5p, and miR-185-5p) and 2 upregulated miRNAs were found in MM exosome samples compared with SMM and HD.98 The same downregulated miRNAs in exosomes of patients with newly diagnosed MM had a strong negative correlation with disease progression, β2-microglobulin, and plasma cell load.99 Also the lncRNA PRINS was deregulated in exosomes of patients with newly diagnosed MM compared with MGUS and HD, and its expression had a negative correlation with PC percentage.100 When comparing exosomal miRNAs between responders and nonresponders to bortezomib, miR-17-5p, miR-20a-5p, miR-15a-5p, and miR-16-5p were lower in the resistant group.101 Similarly, 482 lncRNAs and 2099 mRNAs were discovered to be deregulated in exosomes of bortezomib-resistant patients. Of these, FFAR1 and SP9 were increased, whereas HIST1H2BG and ITIH2 were decreased. Exosomal FFAR1 and SP9 could be potential independent prognostic indicators of survival in patients with MM.102 Also, circMyc levels were higher in MM patient exosomes and correlated to 17p deletion and translocation of t(4;14). Moreover, circMyc levels were higher in bortezomib-resistant patients vs responders and were an independent predictor of poor prognosis.103 

Concerning MM-induced pathologies, 265 upregulated circRNAs and 787 downregulated circular RNAs (circRNAs) in MM patient exosomes were screened for their impact on peripheral neuropathy. CircRNA chr2:27442282 744 407+ was found to be correlated with peripheral neuropathy characteristics and was predicted to downregulate miR-6829-3p and elevate GRIN2B.104 Similarly, exosomal circG042080 was identified as a possible predictor for MM-related myocardial damage.105 Moreover, a recent paper found that exosomal circATP10A in patients with MM had prognostic potential and correlated to vascular endothelial growth factor B protein levels, suggesting involvement in angiogenesis.106 

Other possible EV cargos such as DNA or lipids have not been evaluated yet in MM. Although circulating cell-free DNA could serve as a possible biomarker, the presence of double-stranded DNA inside small EVs remains controversial, which can be attributed to discrepancies in the preparation method and size of the isolated EVs.107 Lipids are important players in a variety of processes including membrane formation and intracellular signaling. It would seem logical that lipid biomarkers can be evaluated in exosomes because the distribution of lipids is expected to be very similar to the plasma membrane. However, the evaluation of lipids in exosomes is not straightforward because of several risks including co-isolation of lipoproteins, poor lipid extraction, exclusion of lipophages, and the complex nature of biological membranes.108 Nevertheless, with standardized isolation and processing methods, these hurdles could be avoided, making EVs a valuable pool for lipidomic analysis.

It is now clear that EVs, including exosomes, have an established role in MM pathobiology by exchanging various cargo between the MM cells and their environment. Therefore, they provide attractive therapeutic targets to tackle MM-induced BM changes and DR. However, progress on this front is still disappointing because exosomes remain elusive potential targets. Unresolved issues include a lack of selectivity and insufficient insight into exosomal secretion routes as outlined in Table 3. These issues need to be addressed before exosomal targeting can move forward. Moreover, more knowledge of the different secretion routes could pinpoint a target that might be highly specific to the release of certain unwanted cargo. Because the use of exosomes as drug delivery vehicles is closer to clinical implementation, knowledge of these secretion pathways is also necessary to better understand how certain payloads can be incorporated into exosomes. In regard to the biomarker studies, there has been a clear rise in the number of MM-related studies, especially evaluating small RNA. However, there is no consensus yet on which is the best EV biomarker. For now, EV analysis should not replace biopsies but could be used to improve risk stratification. Unresolved issues for biomarker implementation include clinically feasible isolation procedures and interference by unassociated EV release. Moreover, the development of more sophisticated “omics” approaches will most likely fast-forward this domain.

Table 3.

List of unresolved issues in EV studies and possible solutions

IssuePossible solution
Lack of selective targets in exosome secretion Differentiation by location. Unconventional exosome inhibitors such as proton pump inhibitors might work in the acidic BM109 
Chemotherapy triggering the release of chemoexosomes might upregulate potential targets 
Insufficient insight into the different secretion routes Cargo profiling on the different routes by KO studies 
Standardization of isolation techniques Large consortium studies to compare different techniques at different sites to determine if cargo results are valid 
Unsuitable isolation procedures for a clinical setting Research into more feasible strategies without compromising exosome purity 
Release of exosomes in the bloodstream, unassociated to MM, triggered by underlying pathologies such as chronic inflammation For flow cytometry: always add an MM marker (CD38/CD138)
For RNA analysis: an extra immunomagnetic isolation step, using an MM marker could be considered 
IssuePossible solution
Lack of selective targets in exosome secretion Differentiation by location. Unconventional exosome inhibitors such as proton pump inhibitors might work in the acidic BM109 
Chemotherapy triggering the release of chemoexosomes might upregulate potential targets 
Insufficient insight into the different secretion routes Cargo profiling on the different routes by KO studies 
Standardization of isolation techniques Large consortium studies to compare different techniques at different sites to determine if cargo results are valid 
Unsuitable isolation procedures for a clinical setting Research into more feasible strategies without compromising exosome purity 
Release of exosomes in the bloodstream, unassociated to MM, triggered by underlying pathologies such as chronic inflammation For flow cytometry: always add an MM marker (CD38/CD138)
For RNA analysis: an extra immunomagnetic isolation step, using an MM marker could be considered 

In conclusion, the different studies reported in this review highlight the potential use of exosomes in MM, both as a therapeutic target and as potential material for liquid biopsies.

Contribution: E.M. and K.V. wrote the manuscript and approved the final version.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Eline Menu, Department of Hematology and Immunology, Myeloma Center Brussels, Laarbeeklaan 103, B-1090 Brussels, Belgium; e-mail: eline.menu@vub.be.

1.
Ludwig
H
,
Novis Durie
S
,
Meckl
A
,
Hinke
A
,
Durie
B
.
Multiple myeloma incidence and mortality around the globe: interrelations between health access and quality, economic resources, and patient empowerment
.
Oncologist
.
2020
;
25
(
9
):
e1406
-
e1413
.
2.
Rajkumar
SV
,
Dimopoulos
MA
,
Palumbo
A
, et al
.
International Myeloma Working Group updated criteria for the diagnosis of multiple myeloma
.
Lancet Oncol
.
2014
;
15
(
12
):
e538
-
e548
.
3.
van de Donk
NWCJ
,
Pawlyn
C
,
Yong
KL
.
Multiple myeloma
.
Lancet
.
2021
;
397
(
10272
):
410
-
427
.
4.
Moser-Katz
T
,
Joseph
NS
,
Dhodapkar
MV
,
Lee
KP
,
Boise
LH
.
Game of bones: how myeloma manipulates its microenvironment
.
Front Oncol
.
2021
;
10
:
625199
.
5.
Anand
S
,
Samuel
M
,
Mathivanan
S
.
Exomeres: a new member of extracellular vesicles family
.
Subcell Biochem
.
2021
;
97
:
89
-
97
.
6.
Kalluri
R
,
LeBleu
VS
.
The biology, function, and biomedical applications of exosomes
.
Science
.
2020
;
367
(
6478
):
eaau6977
.
7.
Ghossoub
R
,
Lembo
F
,
Rubio
A
, et al
.
Syntenin-ALIX exosome biogenesis and budding into multivesicular bodies are controlled by ARF6 and PLD2
.
Nat Commun
.
2014
;
5
(
1
):
3477
.
8.
Kowal
J
,
Tkach
M
,
Théry
C
.
Biogenesis and secretion of exosomes
.
Curr Opin Cell Biol
.
2014
;
29
:
116
-
125
.
9.
Stuffers
S
,
Sem Wegner
C
,
Stenmark
H
,
Brech
A
.
Multivesicular endosome biogenesis in the absence of ESCRTs
.
Traffic
.
2009
;
10
(
7
):
925
-
937
.
10.
Blanc
L
,
Vidal
M
.
New insights into the function of Rab GTPases in the context of exosomal secretion
.
Small GTPases
.
2018
;
9
(
1-2
):
95
-
106
.
11.
Yoon
TY
,
Munson
M
.
SNARE complex assembly and disassembly
.
Curr Biol
.
2018
;
28
(
8
):
R397
-
R401
.
12.
Wei
H
,
Chen
Q
,
Lin
L
, et al
.
Regulation of exosome production and cargo sorting
.
Int J Biol Sci
.
2021
;
17
(
1
):
163
-
177
.
13.
Mulcahy
LA
,
Pink
RC
,
Carter
DR
.
Routes and mechanisms of extracellular vesicle uptake
.
J Extracell Vesicles
.
2014
;
3
(
1
):
3
.
14.
Théry
C
,
Witwer
KW
,
Aikawa
E
, 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
(
1
):
1535750
.
15.
Kugeratski
FG
,
Hodge
K
,
Lilla
S
, et al
.
Quantitative proteomics identifies the core proteome of exosomes with syntenin-1 as the highest abundant protein and a putative universal biomarker
.
Nat Cell Biol
.
2021
;
23
(
6
):
631
-
641
.
16.
Willms
E
,
Cabañas
C
,
Mäger
I
,
Wood
MJA
,
Vader
P
.
Extracellular vesicle heterogeneity: subpopulations, isolation techniques, and diverse functions in cancer progression
.
Front Immunol
.
2018
;
9
:
738
.
17.
Sidhom
K
,
Obi
PO
,
Saleem
A
.
A review of exosomal isolation methods: is size exclusion chromatography the best option?
.
Int J Mol Sci
.
2020
;
21
(
18
):
E6466
.
18.
Paolini
L
,
Zendrini
A
,
Di Noto
G
, et al
.
Residual matrix from different separation techniques impacts exosome biological activity
.
Sci Rep
.
2016
;
6
(
1
):
23550
.
19.
Van Deun
J
,
Hendrix
A
;
EV-TRACK consortium
.
Is your article EV-TRACKed?
.
J Extracell Vesicles
.
2017
;
6
(
1
):
1379835
.
20.
Spencer
JA
,
Ferraro
F
,
Roussakis
E
, et al
.
Direct measurement of local oxygen concentration in the bone marrow of live animals
.
Nature
.
2014
;
508
(
7495
):
269
-
273
.
21.
Vacca
A
,
Ribatti
D
,
Roncali
L
, et al
.
Bone marrow angiogenesis and progression in multiple myeloma
.
Br J Haematol
.
1994
;
87
(
3
):
503
-
508
.
22.
Umezu
T
,
Tadokoro
H
,
Azuma
K
,
Yoshizawa
S
,
Ohyashiki
K
,
Ohyashiki
JH
.
Exosomal miR-135b shed from hypoxic multiple myeloma cells enhances angiogenesis by targeting factor-inhibiting HIF-1
.
Blood
.
2014
;
124
(
25
):
3748
-
3757
.
23.
Wang
J
,
De Veirman
K
,
Faict
S
, et al
.
Multiple myeloma exosomes establish a favourable bone marrow microenvironment with enhanced angiogenesis and immunosuppression
.
J Pathol
.
2016
;
239
(
2
):
162
-
173
.
24.
Li
B
,
Hong
J
,
Hong
M
, et al
.
piRNA-823 delivered by multiple myeloma-derived extracellular vesicles promoted tumorigenesis through re-educating endothelial cells in the tumor environment
.
Oncogene
.
2019
;
38
(
26
):
5227
-
5238
.
25.
Di Noto
G
,
Chiarini
M
,
Paolini
L
, et al
.
Immunoglobulin free light chains and GAGs mediate multiple myeloma extracellular vesicles uptake and secondary NFκB nuclear translocation
.
Front Immunol
.
2014
;
5
:
517
.
26.
Umezu
T
,
Imanishi
S
,
Azuma
K
, et al
.
Replenishing exosomes from older bone marrow stromal cells with miR-340 inhibits myeloma-related angiogenesis
.
Blood Adv
.
2017
;
1
(
13
):
812
-
823
.
27.
Liu
L
,
Ye
Q
,
Liu
L
, et al
.
C6-ceramide treatment inhibits the proangiogenic activity of multiple myeloma exosomes via the miR-29b/Akt pathway
.
J Transl Med
.
2020
;
18
(
1
):
298
.
28.
Guo
HM
,
Sun
L
,
Yang
L
,
Liu
XJ
,
Nie
ZY
,
Luo
JM
.
Microvesicles shed from bortezomib-treated or lenalidomide-treated human myeloma cells inhibit angiogenesis in vitro
.
Oncol Rep
.
2018
;
39
(
6
):
2873
-
2880
.
29.
Zarfati
M
,
Avivi
I
,
Brenner
B
,
Katz
T
,
Aharon
A
.
Extracellular vesicles of multiple myeloma cells utilize the proteasome inhibitor mechanism to moderate endothelial angiogenesis
.
Angiogenesis
.
2019
;
22
(
1
):
185
-
196
.
30.
Ashcroft
AJ
,
Davies
FE
,
Morgan
GJ
.
Aetiology of bone disease and the role of bisphosphonates in multiple myeloma
.
Lancet Oncol
.
2003
;
4
(
5
):
284
-
292
.
31.
Raimondi
L
,
De Luca
A
,
Amodio
N
, et al
.
Involvement of multiple myeloma cell-derived exosomes in osteoclast differentiation
.
Oncotarget
.
2015
;
6
(
15
):
13772
-
13789
.
32.
Raimondi
L
,
De Luca
A
,
Fontana
S
, et al
.
Multiple myeloma-derived extracellular vesicles induce osteoclastogenesis through the activation of the XBP1/IRE1α axis
.
Cancers (Basel)
.
2020
;
12
(
8
):
E2167
.
33.
Zahoor
M
,
Westhrin
M
,
Aass
KR
, 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
(
27
):
2656
-
2666
.
34.
Faict
S
,
Muller
J
,
De Veirman
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
(
11
):
105
.
35.
Raimondo
S
,
Saieva
L
,
Vicario
E
, et al
.
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
(
1
):
2
.
36.
Raimondo
S
,
Urzì
O
,
Conigliaro
A
, et al
.
Extracellular vesicle microRNAs contribute to the osteogenic inhibition of mesenchymal stem cells in multiple myeloma
.
Cancers (Basel)
.
2020
;
12
(
2
):
E449
.
37.
Liu
Z
,
Liu
H
,
Li
Y
, et al
.
Multiple myeloma-derived exosomes inhibit osteoblastic differentiation and improve IL-6 secretion of BMSCs from multiple myeloma
.
J Investig Med
.
2020
;
68
(
1
):
45
-
51
.
38.
Li
B
,
Xu
H
,
Han
H
, et al
.
Exosome-mediated transfer of lncRUNX2-AS1 from multiple myeloma cells to MSCs contributes to osteogenesis
.
Oncogene
.
2018
;
37
(
41
):
5508
-
5519
.
39.
Zhang
L
,
Lei
Q
,
Wang
H
, et al
.
Tumor-derived extracellular vesicles inhibit osteogenesis and exacerbate myeloma bone disease
.
Theranostics
.
2019
;
9
(
1
):
196
-
209
.
40.
Strømme
O
,
Psonka-Antonczyk
KM
,
Stokke
BT
,
Sundan
A
,
Arum
CJ
,
Brede
G
.
Myeloma-derived extracellular vesicles mediate HGF/c-Met signaling in osteoblast-like cells
.
Exp Cell Res
.
2019
;
383
(
1
):
111490
.
41.
Rossi
M
,
Botta
C
,
Correale
P
,
Tassone
P
,
Tagliaferri
P
.
Immunologic microenviron ment and personalized treatment in multiple myeloma
.
Expert Opin Biol Ther
.
2013
;
13
(
suppl 1
):
S83
-
S93
.
42.
Wang
J
,
De Veirman
K
,
De Beule
N
, et al
.
The bone marrow microenvironment enhances multiple myeloma progression by exosome-mediated activation of myeloid-derived suppressor cells
.
Oncotarget
.
2015
;
6
(
41
):
43992
-
44004
.
43.
Morandi
F
,
Marimpietri
D
,
Horenstein
AL
, 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
(
8
):
e1458809
.
44.
Khalife
J
,
Ghose
J
,
Martella
M
, et al
.
MiR-16 regulates crosstalk in NF-κB tolerogenic inflammatory signaling between myeloma cells and bone marrow macrophages
.
JCI Insight
.
2019
;
4
(
21
):
129348
.
45.
Lee
JY
,
Ryu
D
,
Lim
SW
, et al
.
Exosomal miR-1305 in the oncogenic activity of hypoxic multiple myeloma cells: a biomarker for predicting prognosis
.
J Cancer
.
2021
;
12
(
10
):
2825
-
2834
.
46.
Tian
X
,
Sun
M
,
Wu
H
, et al
.
Exosome-derived miR-let-7c promotes angiogenesis in multiple myeloma by polarizing M2 macrophages in the bone marrow microenvironment
.
Leuk Res
.
2021
;
105
:
106566
.
47.
Borrelli
C
,
Ricci
B
,
Vulpis
E
, et al
.
Drug-induced senescent multiple myeloma cells elicit NK cell proliferation by direct or exosome-mediated IL15 Trans-presentation
.
Cancer Immunol Res
.
2018
;
6
(
7
):
860
-
869
.
48.
Vulpis
E
,
Cecere
F
,
Molfetta
R
, et al
.
Genotoxic stress modulates the release of exosomes from multiple myeloma cells capable of activating NK cell cytokine production: Role of HSP70/TLR2/NF-kB axis
.
OncoImmunology
.
2017
;
6
(
3
):
e1279372
.
49.
Moloudizargari
M
,
Redegeld
F
,
Asghari
MH
,
Mosaffa
N
,
Mortaz
E
.
Long-chain polyunsaturated omega-3 fatty acids reduce multiple myeloma exosome-mediated suppression of NK cell cytotoxicity
.
Daru
.
2020
;
28
(
2
):
647
-
659
.
50.
Shao
Q
,
Deng
L
,
Liu
H
, et al
.
Involvement of MM cell-derived exosomes in T lymphocytes immune responses
.
Oncol Lett
.
2020
;
20
(
4
):
31
.
51.
Roccaro
AM
,
Sacco
A
,
Maiso
P
, et al
.
BM mesenchymal stromal cell-derived exosomes facilitate multiple myeloma progression
.
J Clin Invest
.
2013
;
123
(
4
):
1542
-
1555
.
52.
Wang
J
,
Hendrix
A
,
Hernot
S
, et al
.
Bone marrow stromal cell-derived exosomes as communicators in drug resistance in multiple myeloma cells
.
Blood
.
2014
;
124
(
4
):
555
-
566
.
53.
Vardaki
I
,
Sanchez
C
,
Fonseca
P
, et al
.
Caspase-3-dependent cleavage of Bcl-xL in the stroma exosomes is required for their uptake by hematological malignant cells
.
Blood
.
2016
;
128
(
23
):
2655
-
2665
.
54.
Umezu
T
,
Imanishi
S
,
Yoshizawa
S
,
Kawana
C
,
Ohyashiki
JH
,
Ohyashiki
K
.
Induction of multiple myeloma bone marrow stromal cell apoptosis by inhibiting extracellular vesicle miR-10a secretion
.
Blood Adv
.
2019
;
3
(
21
):
3228
-
3240
.
55.
Peng
Y
,
Song
X
,
Lan
J
,
Wang
X
,
Wang
M
.
Bone marrow stromal cells derived exosomal miR-10a and miR-16 may be involved in progression of patients with multiple myeloma by regulating EPHA8 or IGF1R/CCND1
.
Medicine (Baltimore)
.
2021
;
100
(
4
):
e23447
.
56.
Saltarella
I
,
Lamanuzzi
A
,
Desantis
V
, et al
.
Myeloma cells regulate miRNA transfer from fibroblast-derived exosomes by expression of lncRNAs
.
J Pathol
.
2021
;
256
(
4
):
402
-
413
.
57.
Deng
M
,
Yuan
H
,
Liu
S
,
Hu
Z
,
Xiao
H
.
Exosome-transmitted LINC00461 promotes multiple myeloma cell proliferation and suppresses apoptosis by modulating microRNA/BCL-2 expression
.
Cytotherapy
.
2019
;
21
(
1
):
96
-
106
.
58.
Wang
Y
,
Lin
Q
,
Song
C
,
Ma
R
,
Li
X
.
Circ_0007841 promotes the progression of multiple myeloma through targeting miR-338-3p/BRD4 signaling cascade
.
Cancer Cell Int
.
2020
;
20
(
1
):
383
.
59.
Ho
M
,
Chen
T
,
Liu
J
, et al
.
Targeting histone deacetylase 3 (HDAC3) in the bone marrow microenvironment inhibits multiple myeloma proliferation by modulating exosomes and IL-6 trans-signaling
.
Leukemia
.
2020
;
34
(
1
):
196
-
209
.
60.
Xu
H
,
Han
H
,
Song
S
, et al
.
Exosome-transmitted PSMA3 and PSMA3-AS1 promote proteasome inhibitor resistance in multiple myeloma
.
Clin Cancer Res
.
2019
;
25
(
6
):
1923
-
1935
.
61.
Jones
RJ
,
Singh
RK
,
Shirazi
F
, et al
.
Intravenous immunoglobulin G suppresses heat shock protein (HSP)-70 expression and enhances the activity of HSP90 and proteasome inhibitors
.
Front Immunol
.
2020
;
11
:
1816
.
62.
Bandari
SK
,
Purushothaman
A
,
Ramani
VC
, et al
.
Chemotherapy induces secretion of exosomes loaded with heparanase that degrades extracellular matrix and impacts tumor and host cell behavior
.
Matrix Biol
.
2018
;
65
:
104
-
118
.
63.
Faict
S
,
Oudaert
I
,
D’Auria
L
, et al
.
The transfer of sphingomyelinase contributes to drug resistance in multiple myeloma
.
Cancers (Basel)
.
2019
;
11
(
12
):
E1823
.
64.
Frassanito
MA
,
Rao
L
,
Moschetta
M
, et al
.
Bone marrow fibroblasts parallel multiple myeloma progression in patients and mice: in vitro and in vivo studies
.
Leukemia
.
2014
;
28
(
4
):
904
-
916
.
65.
De Veirman
K
,
Wang
J
,
Xu
S
, et al
.
Induction of miR-146a by multiple myeloma cells in mesenchymal stromal cells stimulates their pro-tumoral activity
.
Cancer Lett
.
2016
;
377
(
1
):
17
-
24
.
66.
Cheng
Q
,
Li
X
,
Liu
J
, et al
.
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
.
67.
Frassanito
MA
,
Desantis
V
,
Di Marzo
L
, 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
(
2
):
241
-
253
.
68.
Reale
A
,
Carmichael
I
,
Xu
R
, et al
.
Human myeloma cell- and plasma-derived extracellular vesicles contribute to functional regulation of stromal cells
.
Proteomics
.
2021
;
21
(
13-14
):
e2000119
.
69.
Cheng
Q
,
Li
X
,
Wang
Y
,
Dong
M
,
Zhan
FH
,
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
(
4
):
561
-
568
.
70.
Purushothaman
A
,
Sanderson
RD
.
Heparanase: a dynamic promoter of myeloma progression
.
Adv Exp Med Biol
.
2020
;
1221
:
331
-
349
.
71.
Roucourt
B
,
Meeussen
S
,
Bao
J
,
Zimmermann
P
,
David
G
.
Heparanase activates the syndecan-syntenin-ALIX exosome pathway
.
Cell Res
.
2015
;
25
(
4
):
412
-
428
.
72.
Thompson
CA
,
Purushothaman
A
,
Ramani
VC
,
Vlodavsky
I
,
Sanderson
RD
.
Heparanase regulates secretion, composition, and function of tumor cell-derived exosomes
.
J Biol Chem
.
2013
;
288
(
14
):
10093
-
10099
.
73.
Purushothaman
A
,
Bandari
SK
,
Liu
J
,
Mobley
JA
,
Brown
EE
,
Sanderson
RD
.
Fibronectin on the surface of myeloma cell-derived exosomes mediates exosome-cell interactions
.
J Biol Chem
.
2016
;
291
(
4
):
1652
-
1663
.
74.
Ramani
VC
,
Zhan
F
,
He
J
, et al
.
Targeting heparanase overcomes chemoresistance and diminishes relapse in myeloma
.
Oncotarget
.
2016
;
7
(
2
):
1598
-
1607
.
75.
Galli
M
,
Chatterjee
M
,
Grasso
M
, et al
.
Phase I study of the heparanase inhibitor roneparstat: an innovative approach for ultiple myeloma therapy
.
Haematologica
.
2018
;
103
(
10
):
e469
-
e472
.
76.
Rodrigues-Junior
DM
,
Pelarin
MFA
,
Nader
HB
,
Vettore
AL
,
Pinhal
MAS
.
MicroRNA-1252-5p associated with extracellular vesicles enhances bortezomib sensitivity in multiple myeloma cells by targeting heparanase
.
OncoTargets Ther
.
2021
;
14
:
455
-
467
.
77.
Tu
C
,
Du
Z
,
Zhang
H
, et al
.
Endocytic pathway inhibition attenuates extracellular vesicle-induced reduction of chemosensitivity to bortezomib in multiple myeloma cells
.
Theranostics
.
2021
;
11
(
5
):
2364
-
2380
.
78.
Xie
Y
,
Bai
O
,
Zhang
H
, 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
(
11
):
2655
-
2666
.
79.
Wang
J
,
Cao
Z
,
Wang
P
, et al
.
Apoptotic extracellular vesicles ameliorate multiple myeloma by restoring Fas-mediated apoptosis
.
ACS Nano
.
2021
;
15
(
9
):
14360
-
14372
.
80.
Rivoltini
L
,
Chiodoni
C
,
Squarcina
P
, et al
.
TNF- (TRAIL)-armed exosomes deliver proapoptotic signals to tumor site
.
Clin Cancer Res
.
2016
;
22
(
14
):
3499
-
3512
.
81.
Kamerkar
S
,
LeBleu
VS
,
Sugimoto
H
, et al
.
Exosomes facilitate therapeutic targeting of oncogenic KRAS in pancreatic cancer
.
Nature
.
2017
;
546
(
7659
):
498
-
503
.
82.
Mendt
M
,
Kamerkar
S
,
Sugimoto
H
, et al
.
Generation and testing of clinical-grade exosomes for pancreatic cancer
.
JCI Insight
.
2018
;
3
(
8
):
99263
.
83.
Trino
S
,
Lamorte
D
,
Caivano
A
,
De Luca
L
,
Sgambato
A
,
Laurenzana
I
.
Clinical relevance of extracellular vesicles in hematological neoplasms: from liquid biopsy to cell biopsy
.
Leukemia
.
2021
;
35
(
3
):
661
-
678
.
84.
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
(
8
):
e70811
.
85.
Iaccino
E
,
Mimmi
S
,
Dattilo
V
, et al
.
Monitoring multiple myeloma by idiotype-specific peptide binders of tumor-derived exosomes
.
Mol Cancer
.
2017
;
16
(
1
):
159
.
86.
Krishnan
SR
,
Luk
F
,
Brown
RD
,
Suen
H
,
Kwan
Y
,
Bebawy
M
.
Isolation of human CD138(+) microparticles from the plasma of patients with multiple myeloma
.
Neoplasia
.
2016
;
18
(
1
):
25
-
32
.
87.
Liu
ZY
,
Tian
MY
,
Deng
L
, et al
.
The potential diagnostic power of CD138+ microparticles from the plasma analysis for multiple myeloma clinical monitoring
.
Hematol Oncol
.
2019
;
37
(
4
):
401
-
408
.
88.
Rajeev Krishnan
S
,
De Rubis
G
,
Suen
H
,
Joshua
D
,
Lam Kwan
Y
,
Bebawy
M
.
A liquid biopsy to detect multidrug resistance and disease burden in multiple myeloma
.
Blood Cancer J
.
2020
;
10
(
3
):
37
.
89.
Caivano
A
,
Laurenzana
I
,
De Luca
L
, 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
(
12
):
9739
-
9752
.
90.
Laurenzana
I
,
Trino
S
,
Lamorte
D
, et al
.
Analysis of amount, size, protein phenotype and molecular content of circulating extracellular vesicles identifies new biomarkers in multiple myeloma
.
Int J Nanomedicine
.
2021
;
16
:
3141
-
3160
.
91.
Harshman
SW
,
Canella
A
,
Ciarlariello
PD
, et al
.
Proteomic characterization of circulating extracellular vesicles identifies novel serum myeloma associated markers
.
J Proteomics
.
2016
;
136
:
89
-
98
.
92.
Harshman
SW
,
Canella
A
,
Ciarlariello
PD
, et al
.
Characterization of multiple myeloma vesicles by label-free relative quantitation
.
Proteomics
.
2013
;
13
(
20
):
3013
-
3029
.
93.
Kvorning
SL
,
Nielsen
MC
,
Andersen
NF
,
Hokland
M
,
Andersen
MN
,
Møller
HJ
.
Circulating extracellular vesicle-associated CD163 and CD206 in multiple myeloma
.
Eur J Haematol
.
2020
;
104
(
5
):
409
-
419
.
94.
Di Noto
G
,
Bugatti
A
,
Zendrini
A
, et al
.
Merging colloidal nanoplasmonics and surface plasmon resonance spectroscopy for enhanced profiling of multiple myeloma-derived exosomes
.
Biosens Bioelectron
.
2016
;
77
:
518
-
524
.
95.
Russo
M
,
Tirinato
L
,
Scionti
F
, et al
.
Raman spectroscopic stratification of multiple myeloma patients based on exosome profiling
.
ACS Omega
.
2020
;
5
(
47
):
30436
-
30443
.
96.
Manier
S
,
Liu
CJ
,
Avet-Loiseau
H
, et al
.
Prognostic role of circulating exosomal miRNAs in multiple myeloma
.
Blood
.
2017
;
129
(
17
):
2429
-
2436
.
97.
Caivano
A
,
La Rocca
F
,
Simeon
V
, et al
.
MicroRNA-155 in serum-derived extracellular vesicles as a potential biomarker for hematologic malignancies: a short report
.
Cell Oncol (Dordr)
.
2017
;
40
(
1
):
97
-
103
.
98.
Zhang
ZY
,
Li
YC
,
Geng
CY
,
Zhou
HX
,
Gao
W
,
Chen
WM
.
Serum exosomal microRNAs as novel biomarkers for multiple myeloma
.
Hematol Oncol
.
2019
;
37
(
4
):
409
-
417
.
99.
Zhang
ZY
,
Li
YC
,
Geng
CY
,
Wang
HJ
,
Chen
WM
.
Potential relationship between clinical significance and serum exosomal miRNAs in patients with multiple myeloma
.
BioMed Res Int
.
2019
;
2019
:
1575468
.
100.
Sedlarikova
L
,
Bollova
B
,
Radova
L
, et al
.
Circulating exosomal long noncoding RNA PRINS-First findings in monoclonal gammopathies
.
Hematol Oncol
.
2018
;
36
(
5
):
786
-
791
.
101.
Zhang
L
,
Pan
L
,
Xiang
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
(
21
):
30876
-
30891
.
102.
Tang
JX
,
Chen
Q
,
Li
Q
,
He
YH
,
Xiao
D
.
Exosomal mRNAs and lncRNAs involved in multiple myeloma resistance to bortezomib
.
Cell Biol Int
.
2021
;
45
(
5
):
965
-
975
.
103.
Luo
Y
,
Gui
R
.
Circulating exosomal CircMYC is associated with recurrence and bortezomib resistance in patients with multiple myeloma
.
Turk J Haematol
.
2020
;
37
(
4
):
248
-
262
.
104.
Zhang
Y
,
Pisano
M
,
Li
N
, et al
.
Exosomal circRNA as a novel potential therapeutic target for multiple myeloma-related peripheral neuropathy
.
Cell Signal
.
2021
;
78
:
109872
.
105.
Sun
R
,
Liu
W
,
Zhao
Y
, et al
.
Exosomal circRNA as a novel potential therapeutic target for multiple myeloma-related myocardial damage
.
Cancer Cell Int
.
2021
;
21
(
1
):
311
.
106.
Yu
M
,
Yu
J
,
Zhang
Y
, et al
.
A novel circRNA-miRNA-mRNA network revealed exosomal circ-ATP10A as a biomarker for multiple myeloma angiogenesis
.
Bioengineered
.
2021
.
107.
Hur
JY
,
Lee
KY
.
Characteristics and clinical application of extracellular vesicle-derived DNA
.
Cancers (Basel)
.
2021
;
13
(
15
):
3827
.
108.
Skotland
T
,
Sagini
K
,
Sandvig
K
,
Llorente
A
.
An emerging focus on lipids in extracellular vesicles
.
Adv Drug Deliv Rev
.
2020
;
159
:
308
-
321
.
109.
Guan
XW
,
Zhao
F
,
Wang
JY
, et al
.
Tumor microenvironment interruption: a novel anti-cancer mechanism of Proton-pump inhibitor in gastric cancer by suppressing the release of microRNA-carrying exosomes
.
Am J Cancer Res
.
2017
;
7
(
9
):
1913
-
1925
.
Sign in via your Institution