Next Article in Journal
RPS27a and RPL40, Which Are Produced as Ubiquitin Fusion Proteins, Are Not Essential for p53 Signalling
Next Article in Special Issue
Good Cop, Bad Cop: Profiling the Immune Landscape in Multiple Myeloma
Previous Article in Journal
Serum N-Glycosylation RPLC-FD-MS Assay to Assess Colorectal Cancer Surgical Interventions
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of Extracellular Vesicles in the Pathogenesis of Hematological Malignancies: Interaction with Tumor Microenvironment; a Potential Biomarker and Targeted Therapy

1
Department of Cellular and Molecular Biology, The University of Texas at Tyler Health Science Center, Tyler, TX 75708, USA
2
Department of Pulmonary Immunology, The University of Texas at Tyler Health Science Center, Tyler, TX 75708, USA
*
Author to whom correspondence should be addressed.
Biomolecules 2023, 13(6), 897; https://doi.org/10.3390/biom13060897
Submission received: 17 April 2023 / Revised: 21 May 2023 / Accepted: 25 May 2023 / Published: 27 May 2023

Abstract

:
The tumor microenvironment (TME) plays an important role in the development and progression of hematological malignancies. In recent years, studies have focused on understanding how tumor cells communicate within the TME. In addition to several factors, such as growth factors, cytokines, extracellular matrix (ECM) molecules, etc., a growing body of evidence has indicated that extracellular vesicles (EVs) play a crucial role in the communication of tumor cells within the TME, thereby contributing to the pathogenesis of hematological malignancies. The present review focuses on how EVs derived from tumor cells interact with the cells in the TME, such as immune cells, stromal cells, endothelial cells, and ECM components, and vice versa, in the context of various hematological malignancies. EVs recovered from the body fluids of cancer patients often carry the bioactive molecules of the originating cells and hence can be considered new predictive biomarkers for specific types of cancer, thereby also acting as potential therapeutic targets. Here, we discuss how EVs influence hematological tumor progression via tumor–host crosstalk and their use as biomarkers for hematological malignancies, thereby benefiting the development of potential therapeutic targets.

1. Introduction

Intercellular communication, an essential biological process of multicellular organisms, is mediated by three different mechanisms: (1) cytoplasmic bridges; (2) direct interactions between adjacent cells via membrane proteins; and (3) cellular secretary molecules [1,2]. Recently, a fourth mechanism has been discovered, which includes the intercellular transfer of extracellular vesicles (EVs) [3,4,5,6,7,8,9,10]. EVs are membrane-bound entities released by almost all types of cells into the extracellular environment [11,12]. EVs are known to transport bioactive molecules, such as proteins, lipids, and nucleic acids in the form of DNA, RNA, miRNA (miR), etc., between cells [13,14]. Detectable levels of EVs are found in nearly all biological fluids, such as blood, urine, synovial fluid, and saliva, and they are even found in the interstitial spaces between cells [12,15,16,17]. Since they are protected from degradation by extracellular proteases and RNases, EVs can be stably stored for long-term use [18]. Depending on their biogenesis, size, release mechanism, content, and function, EVs can be broadly classified into microvesicles, exosomes, and apoptotic bodies (Figure 1) [11,12,19,20].
Microvesicles (MVs). MVs are a type of EV generated by direct outward budding of plasma membrane from cells [13,21]. The formation and release of MVs from cells typically require the interplay of cytoskeletal components, such as actin and microtubules; molecular motors, such as kinesin and myosin; and fusion machinery, such as SNAREs and tethering factors [22,23]. MVs typically ranging from 100 nm to 1 µm in diameter [11,12,15,19,21]. Because MVs are generated from plasma membrane by outward budding, they carry cytosolic and plasma membrane-associated proteins, e.g., proteins clustered at the plasma membrane, such as tetraspanins, which could serve as markers for MVs regardless of the originating cells [24,25]. Other cytoskeletal proteins, such as heat shock proteins, integrins, and proteins containing post-translational modifications, such as glycosylation and phosphorylation, have also been shown to be present in MVs [26,27,28].
Exosomes. Exosomes are the other subtype of the EV of endocytic origin [15,29] with a typical diameter of 30 to 150 nm [30,31]. Specifically, exosomes are formed by inward budding of early endosomal membrane, which matures into multivesicular bodies (MVBs) [12,15,32]. MVBs eventually fuse with the plasma membrane, releasing their content of exosomes into the extracellular space [32,33,34,35,36]. The regulation of MVBs and the formation and subsequent release of exosomes are mediated by the endosomal sorting complexes required for transport (ESCRT) pathway [37,38,39]. Since exosome generation is mediated by the ESCRT pathway, ESCRT and its accessory proteins (Alix, TSG101, HSC70, and HSP90β) are believed to be present in all exosomes regardless of the type of originating cells and hence serve as exosome markers [40,41,42,43,44]. Other than the ESCRT pathway, exosome generation is also thought to be dependent on sphingomyelinase enzymes since, in some instances, cells with ESCRT deficiency also produce significant numbers of CD63+ exosomes [45,46,47,48]. Both exosomes and MVs have been shown to participate actively in cell–cell communication, maintenance of cells, and tumor progression by transporting their cargo between cells [49,50]. EVs are readily taken up by the recipient cells, either by direct fusion with the plasma membrane or by fusion with the endosomal membrane after endocytosis [51,52,53,54].
Apoptotic bodies. Apoptotic bodies, which are in general not considered to be a true form of EV, are larger, ranging from 50 nm to 5 µm in diameter [55,56,57], and are released from cells undergoing programmed cell death [19,56]. These bodies are generated by the separation of plasma membrane from the cytoskeleton due to enhanced hydrostatic pressure during contraction of cells [58,59]. In contrast with MVs and exosomes, apoptotic bodies contain cellular organelles, chromatin, and a few glycosylated proteins [19,42,60,61]. Hence, higher levels of nuclear proteins (such as histones), mitochondrial proteins (such as HSP60), Golgi, and endoplasmic reticulum-associated proteins (such as GRP78) are expected to be observed in apoptotic bodies.

2. Tumor-Derived EVs

Tumor-derived EVs are distinguished from normal cell-secreted EVs due to the presence of unique tumor-specific ‘labels’ [62,63,64,65]. Tumor-derived EVs have been shown to carry oncogenic proteins or nucleic acids (such as DNA, RNA, miRNAs, etc.) which facilitate tumor progression. Oncogenic bioactive molecules are enriched in tumor-derived EVs compared to normal cell-derived EVs [66,67,68]. For example, chromosome segregation 1 like protein (CSE1L), a transmembrane protein, is enriched in tumor-derived EVs, not only triggering Ras-dependent EVs biogenesis but also promoting metastasis of B16F10 and melanoma cells [69]. Adriamycin-resistant breast cancer cell-derived EVs were shown to carry transient receptor potential cation channel subfamily C member 5 (TrpC5) and to transfer of EV. TrpC5 confers endothelial cell resistance against chemotherapeutic regimens [70]. On the other hand, the transfer of oncogenic nucleic acids such as miRNAs, specifically miR-221 from highly aggressive breast tumor cells to nonaggressive cancer cells, via EVs, contributing to the promotion of epithelial-to-mesenchymal transition (EMT) [71] and leading to the induction of proliferation and metastasis while preventing drug-induced apoptosis of EVs’ fused recipient cells [5]. Therefore, tumor cells more often induce oncogenic transformations into normal healthy cells via the transfer of oncogenic bioactive molecules through EVs [72,73].

3. Hematological Malignancies

Hematological malignancies are defined as tumors that commence in blood-forming tissues, such as bone marrow or cells of the immune system, resulting in leukemias, lymphomas, and myelomas [74,75]. Hematological malignancies are considered to be among the leading causes of cancer-related deaths worldwide [76,77,78]. In the United States itself, an estimated 184,710 new cases of hematological neoplasms were reported with 57,380 deaths in 2023, and the incidence increases with age [79]. GLOBOCAN 2020 reported non-Hodgkin lymphoma to be the predominant hematological cancer worldwide with 544,352 new cases and 259,793 deaths, followed by leukemia, with 474,519 new cases and 311,594 deaths worldwide [80]. The outbreak of COVID-19 further increased the death rate of patients suffering from various hematological malignancies [81,82,83,84,85].
EVs are known to play an important role in cell–cell communication via the transfer of bioactive cargo molecules. However, the role of EVs in the crosstalk of tumor cells with cells in the tumor microenvironment (TME) and other distant cells remains to be completely determined in the context of the pathogenesis of hematological malignancies. The present review highlights how hematological cancer cells communicate with the cells in the TME and vice versa via the release of EVs. The first part of the review focuses on the modern classification of hematological malignancies, followed by understanding the EVs’ role in cell–cell and cell–extracellular matrix communication in the TME. The final part of the review reveals how EVs could be used as biomarkers for hematological malignancies and their contributions to drug resistance.

4. Hematological Malignancies: The Modern Classification

Over the years, different classification schemes have been established to define various hematological malignancies; however, the World Health Organization (WHO) classification is the most prevalent [86]. Based on features such as morphology, lineage, clinical attributes, cytogenetics, and molecular pattern, the WHO 5th edition (WHO5) broadly categorizes hematological neoplasms into myeloid neoplasms, lymphoid neoplasms, myeloid/lymphoid neoplasms with eosinophilia and tyrosine kinase gene fusions, mastocytosis, histiocytic/dendritic neoplasms, and mixed myeloid and lymphoid neoplasms [87,88]. In the present section, we discuss a detailed classification of myeloid- and lymphoid neoplasms, followed by a brief description of other hematological neoplasms (Table 1).
Myeloid neoplasms. This type of neoplasm is obtained from bone marrow progenitor cells, which differentiate into megakaryocytes, granulocytes, erythrocytes, and monocytes. Exceptions exist, such as chronic myeloid leukemia (CML), in which the originating cells are pluripotent hematopoietic stem cells that differentiate into lymphoid cells. Myeloid neoplasms are further classified as follows.
(1). Myeloproliferative neoplasms (MPNs): MPNs, a type of clonal stem cell disorder, are caused by genetic mutations, resulting in abnormal activation of pro-growth signaling, ultimately leading to uncontrolled proliferation of myeloid progenitors, such as megakaryocytes, granulocytes, or erythrocytes. MPNs are sub-classified into: (A) chronic myeloid leukemia (CML)—caused by excessive proliferation of the granulocyte lineage, leading to increased myeloid maturation, which is associated with the BCR:ABL1 fusion gene; (B) classic BCR:ABL1-negative MPNs—these neoplasms lack the BCR:ABL1 fusion gene, which includes polycythemia vera (PV), characterized by increased red blood cells; essential thrombocytopenia (ET), a clonal disorder associated with thrombocytosis; and primary myelofibrosis (PF), caused by a polyclonal increase in fibroblasts leading to bone marrow fibrosis; (C) chronic neutrophilic leukemia (CNL)—caused by overproduction of matured granulocytes; (D) chronic eosinophilic leukemia (CEL)—caused by clonal proliferation of morphologically abnormal eosinophils and their precursors, resulting in hypereosinophilia; and (E) myeloproliferative neoplasm, unclassifiable—these neoplasms are classes of MPNs not satisfying the criteria to categorize into specific MPN types or having overlapping MPN subtype characteristics.
(2). Myelodysplastic neoplasms/syndromes (MDS): These neoplasms are caused by ineffective blood cell production, leading to dysplasia or cytopenia and often resulting in bone marrow failure. In accordance with WHO5, MDS are classified with respect to genetic abnormalities or morphology. According to genetic abnormalities, MDS are subtyped into: (A) MDS with low blasts and isolated del(5q)—5q deletion, bone marrow blasts (BMB) < 5% in bone marrow and peripheral blasts (PB) < 2%; and SF3B1 or TP53 mutation; (B) MDS with low blasts and SF3B1 mutation—absence of 5q deletion, BMB < 5% and PB < 2%, SF3B1 mutation, and ≥15% ring sideroblasts; and (C) MDS with biallelic TP53 inactivation—equal to or more than two TP53 mutations or one mutation with TP53 copy number loss or copy neutrality, resulting in loss of heterozygosity, complex cytogenetics, BMB, and PB < 20%. Morphologically defined MDS can be classified into: (A) MDS with low blasts—BMB < 5% and PB < 2%; (B) MDS hypoblastic—BM cellularity < 20%; and (C) MDS with increased blasts (MDS-IB)—these neoplasms include MDS-IB1, BMB blasts 5–9%, and PB blasts 2–4%; MDS-IB2, BMB blasts 10–19%, and PB blasts 5–19%; MDS with increased blasts and fibrosis; BM fibrosis with BMB blasts 5–19%; and PB blasts 2–19%.
(3). Acute myeloid leukemia (AML): These neoplasms are characterized by highly aggressive and genetically heterogenous myeloid malignancies. They are further classified as: (A) AML with recurrent genetic abnormalities—caused by genetic abnormalities and subcategorized into characteristic chromosomal changes due to inversions or reciprocal chromosomal rearrangements and no associated karyotypic abnormalities; (B) AML with myelodysplasia-related features—observed in patients with a history of MDS or MDS/myeloproliferative neoplasms (MPNs) and lacking the genetic features of AML; (C). therapy-related AML—complications arise after treatment with chemo- or radiation therapy; (D) AML not otherwise specified (NOS)—having recognizable clinical, morphologic, and immunophenotypic features but lacking karyotypic and diagnostic features of AML; (E) myeloid sarcoma—these neoplasms are masses of tumor blasts outside the bone marrow but effacing into the tissues; and (F) myeloid proliferations related to Down syndrome (DS)—occurring in Down syndrome, which is associated with an increased risk of AML.
(4). Myeloid neoplasms with mutated TP53: These neoplasms are new types of myeloid neoplasms, which are further classified as MDS, MDS/AML, and AML depending on TP53 mutations and blast counts. They are categorized into—MDS with mutated TP53, <10% blasts; MDS/AML with mutated TP53, 10–19% blasts; and AML with mutated TP53, >20% blasts.
Lymphoid neoplasms. Lymphoid neoplasms are typical types of cancer, affecting cells that normally give rise to B-lymphocytes, such as lymphocytes; plasma cells; and T-lymphocytes, such as Treg, TH, and TC. Depending on the B-cell and T-cell/NK-cell lineage, lymphoid neoplasms can be classified into precursor lymphoid neoplasms, mature B cell neoplasms, Hodgkin lymphoma, and mature T cell or NK cell lineage neoplasms.
(1). Precursor lymphoid neoplasms: These neoplasms are highly aggressive and are referred to as acute lymphoblastic leukemia/lymphoblastic lymphoma (ALL/LBL) because they evolved from either leukemic or lymphatic cells. They are further classified as—B-cell ALL/LBL—arising from precursor B-cells and representing most of ALL/LBLs; and T cell ALL/LBL—derived from T-cell precursors comprising 15% all ALL/LBL populations.
(2). Mature B-cell neoplasms: According to WHO5, lymphoid neoplasms of matured B-cells are further classified as: (A) chronic lymphocytic leukemia (CLL)/small lymphocytic lymphoma (SLL)—small, matured-appearing lymphocytes; (B) lymphoplasmacytic lymphoma (LPL)—derived from post-germinal centers of B-cells; (C) ,monoclonal gammopathies—a new subtype of lymphoid neoplasms for primary cold agglutinin disease, which are further classified as primary cold agglutinin disease, monoclonal gammopathy of undetermined significance (MGUS) of non-IgM type, or amyloidosis; (D) plasma cell neoplasms—related to terminally differentiated germinal centers of B-cells secreting monoclonal IgM; (E) hairy cell leukemia—tumors related to the post-germinal centers of B-cells with the appearance of a hairy morphology; (F) marginal zone lymphoma (MZL)—neoplasms associated with a marginal zone of matured B-cells; (G) follicular lymphoma (FL)—the most common lymphoma, which is associated with the neoplastic germinal center of B-cells and further classified as—classic FL, proliferation of small (centrocytes) and large (centroblasts) neoplastic follicular cells; FL grade 3B, uncontrolled proliferation of centroblasts; and FL with unusual features, comprising FL with blastoid features and FL with predominantly diffuse growth pattern; (H) mantle cell lymphoma (MCL)—related to neoplasms of naïve B-cells and antigen-stimulated B-cells; (I) diffuse large B-cell lymphoma (DLBCL)—associated with matured B-cells; and (J) high-grade B-cell lymphomas—comprising Burkitt lymphoma (BL), a highly aggressive B-cell neoplasm; and other high-grade B-cell lymphomas associated with MYC and BCL-2 rearrangements.
(3). Hodgkin lymphoma (HL): These neoplasms are derived from germinal center and post-germinal center B-cells, comprising the minority population of neoplastic lymphomas and subdivided into: (A) classic HL (cHL)—further classified into nodular sclerosis cHL, mixed cellularity cHL, lymphocyte-rich cHL, or lymphocyte-depleted cHL; and (B) nodular lymphocyte-predominant HL (NLPHL)—these neoplasms are uncommon, retaining immunophenotypic features of germinal neoplastic B-cells.
(4). Mature T-cell or NK-cell lineage lymphomas: These neoplasms are further categorized into: (A) peripheral T cell lymphoma (PTCL)—comprising peripheral T cell lymphoma that does not fit the criteria of T-cell lymphoma, NOS; anaplastic large cell lymphoma (ALCL)—according to the expression of anaplastic lymphoma kinase (ALK), these neoplasms are subcategorized into “ALCL, ALK-positive” or “ALCL, ALK-negative”; and “breast implant-associated ALCL”; and follicular helper T-cell lymphoma—including “angioimmunoblastic T-cell lymphoma (AITL)” and related follicular TH cells; extranodal NK-/T-cell lymphoma, nasal type—these neoplasms are Epstein–Barr virus (EBV)-associated neoplasm arises in the lymph nodes; subcutaneous panniculitis-like T cell lymphoma; hepatosplenic T-cell lymphoma—highly aggressive and related to immunosuppression; and primary intestinal T-cell lymphomas—aggressive T-cell lymphomas in the intestinal tract; (B) primary cutaneous peripheral T-cell lymphomas—related to cutaneous peripheral T-cells; (C) adult T-cell leukemia-lymphoma (ATL)—associated with peripheral T-cells originating from T-cell leukemia virus (HTLV) type 1-infected CD4+ T-cells; (D) T-cell large granular lymphocyte (TLGL) leukemia—developed from clonally expanded large granular T-cells; (E) T-cell prolymphocytic leukemia—highly aggressive and comprising medium-sized matured T-cells; (F) NK-cell large granular lymphocyte (NKLGL) leukemia—highly aggressive, associated with malignant NK-cells; and (G) aggressive NK-cell leukemia—considered a nasal type since more often shown to be associated with EBV infection.
Myeloid/lymphoid neoplasms with eosinophilia and tyrosine kinase (TK) gene fusions. Eosinophilia is defined as an increase in eosinophil count in the peripheral blood and tissues. More often, myeloid/lymphoid neoplasms are shown to be associated with hyper-eosinophilia (HE), which is defined as an absolute eosinophil count (AEC) >1.5 × 109/L in the peripheral blood and hypereosinophilic syndrome (HES) in a situation of HE in which eosinophil-mediated organ damage or dysfunction is observed. According to the pathogenic mechanisms leading to the expansion of eosinophils, HES are subclassified as primary, secondary, and idiopathic. According to the gene fusion and rearrangements of PDGFRA, PDGFRB, JAK2, FGFR1, ETV6:ABL1, and FLT3, WHO5 defined these types of neoplasms as a separate class. A brief classification of these hematological neoplasms is shown in Table 1.
Mastocytosis. These neoplasms are rare, heterogenous forms of neoplasms, resulting from neoplastic mast cells in tissues and various organs. Constitutive activation of KIT receptor is shown to contribute to this form of hematological neoplasm. WHO5 classified this type of neoplasm into cutaneous mastocytosis (CM), in which the disease is limited to skin only; and systemic mastocytosis (SM), involving extracutaneous organ infiltration. A brief discussion is provided in Table 1.
Histiocytic/dendritic neoplasms. This type of neoplasm is related to cells that have developed into antigen-presenting dendritic cells and tissue macrophages, further classified in Table 1. The MAPK pathway has been shown to play an important role in histiocytic/dendritic neoplasms.
Mixed myeloid and lymphoid neoplasms. Sometimes, neoplasms express the markers of myeloid and lymphoid cells or none, with examples being “acute undifferentiated leukemia” and “mixed phenotype acute leukemia (MPAL).” A detailed classification is shown in Table 1.

5. EVs in Cell-Cell and Cell-Extracellular Matrix Communication in the TME

The TME is the environment surrounding the tumor cells in the body [89,90]. It consists of immune cells, stromal cells, fibroblasts, extracellular matrix (ECM), and cells of the blood and lymphatic vessels [89,90]. Tumor cells and their TME are in constant interaction, thereby regulating each other either positively or negatively [91]. Dynamic interaction between cancer cells and TME components not only supports tumor growth and development [92,93] but also promotes local invasion and metastatic dissemination of cancer [94,95]. In hypoxic and acidic conditions, the TME often promotes angiogenesis, a process of restoring nutrient and oxygen supply, as well as removing metabolic waste [96,97,98]. Additionally, the infiltration of various immune cells into the TME performs various pro- and anti-tumorigenic functions [99,100,101,102]. EVs play an important role in intercellular communication via the transfer of bioactive cargo molecules between cells [103]. Tumor-derived EVs also act as a communicating vehicle between cancer cells and cells in the TME and in some instances also with distant cells. On the other hand, cells in the TME often release EVs that interact with tumor cells, influencing tumor development and progression.
Effects of hematological malignancy-derived EVs on immune cells. Immune cells play a major role in the elimination of tumors through diverse mechanisms, and the evasion of immune surveillance serves as an important step for developing tumor niches and successful establishment of tumors. Immune evasion, a strategy facilitated by tumor-derived EVs is utilized in different ways to target various immune cells (Figure 2). Tumor-derived EVs, through their receptor-mediated uptake, can introduce several suppressive factors, e.g., miRNA, DNA, pro-apoptotic factors, metabolites, and various enzymes, into immune cells (Table 2). They can also alter the activation of immune cells through inhibitory cell surface receptors [104,105,106,107].
In activated T-cells, tumor-derived EVs induce the down-regulation of CD3ζ and JAK3 expression via transcriptional regulation, thereby facilitating Fas/FasL-mediated apoptosis of CD8(+) T-cells [108]. Chronic lymphocytic leukemia (CLL)-derived EVs were shown to down-regulate CD69 expression in T-cells via miR-363 transfer, thereby affecting effector T-cell migration [109,110,111]. Tumor cells are often shown to evade the host immune system via the activation of the PD-L1/PD-1 pathway. PD-L1 is expressed on the surfaces of various tumor cells, whereas its receptor, PD-1, is present on T-cells. PD-L1 binding to PD-1 results in the apoptosis of T-cells, thereby evading host immune responses. The upregulation of PD-1 has been observed in various T-cell populations after exposure to diffuse large B cell lymphoma (DLBCL)-derived EVs [112]. EVs released from B-cell lymphoma (BCL) under chemotherapy are enriched with CD39 and CD73, and they hydrolyze ATP, which is generated from chemotherapy-treated tumor cells and transformed into adenosine [113], which in turn affects the immune system by inhibiting T-cell activity and proliferation [114].
Table 2. The effect of EVs, derived from hematological malignancy on immune cell function.
Table 2. The effect of EVs, derived from hematological malignancy on immune cell function.
Originating CellsEffector CellsMalignanciesFunctionsReferences
Leukemic cellsCD8(+)T-cellsSolid tumors or AML Down-regulates CD3ζ and JAK3 expression and promotes Fas/FasL-mediated T-cell apoptosis[108]
Leukemic cellsCD4(+)T-cellsCLLDown-regulates CD69 expression via miR-363 transfer and affects effector T-cell migration[109,110,111]
Lymphoma cells T-cellsDLBCLInduces PD-1 expression in T-cells and enhances T-cell apoptosis[112]
Lymphoma cells T-cellsBCLCarries CD39 and CD73 and hydrolyzes ATP to generate adenosine to inhibit T-cell activity and proliferation [114]
Lymphoma cellsNK-cells, APCsBCL, TCLCarries MHC, APO2L, FASL, TCR, and NKG2D and inhibits NK-cells cytotoxicity and antigen processing of APCs[115,116,117]
Non-leukemic cellsNK-cellsCLLCarries BAG6 and activates NK-cells, but activated NK-cells are eliminated by lymphocytes[118]
Myeloma cellsMDSCs,MMInduces growth and immunosuppressive activity[119,120]
NK-cells, Reduces NK-cells’ cytotoxicity[121]
Immune cells Carries ectoenzyme, CD38, which converts nucleotides into adenosine to suppress immune system[122]
Leukemic cellsNK-cellsAMLEVs’-bound TGFβ1 reduces NK-cells’ cytotoxicity[123]
Lymphoma cellsMonocytes -Releases TNF-α, IL-1β, and IL-6 and prevents monocyte differentiation into dendritic cells [124]
Lymphoma cells MacrophagesDLBCLTransfers MyD88 and stimulates pro-inflammatory NF-κB signaling pathway [125]
Leukemic cells MacrophagesCMLPolarizes macrophages to M2-phenotype to induce TNF-α and IL-10 expression and down-regulates NO and ROS generation[126]
Tumor cellsNeutrophilsCATPromotes NET formation, reduced generation of suppressor cells [127,128,129]
Lymphoma cellsMDSCsLymphomaCarries HSP72 and promotes suppressive functions [124]
Abbreviations of malignancies: AML, acute myeloid leukemia; CLL, chronic lymphocytic leukemia; DLBCL, diffuse large B cell lymphoma; BCL, B-cell lymphoma; TCL, T-cell lymphoma; MM, multiple myeloma; CAT, cancer associated thrombosis; CML, chronic myelogenous leukemia.
EVs are known to carry surface receptors of the originating cells. For example, EVs from B- and T-cell lymphomas are capable of carrying cell surface molecules, such as major histocompatibility complex (MHC), Apo2 ligand (APO2L), Fas ligand (FASL), T-cell receptor (TCR), and natural-killer group-2 member-D (NKG2D), which not only inhibit the cytotoxicity of NK-cells, thereby promoting T-cell apoptosis, but also down-regulate the processing of antigens by antigen presenting cells (APCs) [115,116,117]. The plasma soluble ligand, BAG6, in CLL patients binds to the receptor NKp30 on NK-cells, causing NK-cell inactivation [118]. In contrast, BAG6, carried by the EVs, activates NK cells, which have the ability to kill tumor cells [118]. Hence, the dysregulated balance between the soluble form of BAG6 and its EVs’ bound form determines the immune evasion of CLL. EVs from multiple myeloma (MM) cells suppress the immune system through different mechanisms. First, MM-derived EVs enhance the growth and immunosuppressive activity of myeloid-derived suppressor cells (MDSCs) in both in vitro and in vivo MM xenograft murine models [119,120]. Second, EVs from MM cells reduce NK-cells’ cytotoxic activity [121]. Third, the ectoenzyme, CD38, on MM-derived EVs converts nucleotides into adenosine, which is a well-known suppressor of the immune system [122].Finally, CD38-positive EVs were shown to be internalized by FcR-positive cells, such as monocytes, MDSCs, and NK cells, after binding to an anti-CD38 mAb (daratumumab), although the effects are still under investigation [123]. TGFβ1, on the other hand, was shown to play an important role in immune-evasive mechanisms of leukemic EVs [130]. EVs from the sera of acute myeloid leukemia (AML) patients are enriched with membrane-bound TGFβ1, which significantly reduces the killing properties of NK-cells [131]. Moreover, TGFβ1 on chronic myelogenous leukemia (CML) EVs stimulates the proliferation and colony formation of CML cells [130].
Monocytes are the highly dynamic cells differentiated into macrophages and dendritic cells to effectively protect the body from tumor assault. The fusion of lymphoma-derived EVs with monocytes results in the release of TNF-α, IL-1β, and IL-6, which in turn impair monocytic differentiation into dendritic cells [123]. DLBCL-derived EVs readily transfer MyD88 to the macrophages, thereby stimulating the pro-inflammatory signaling pathway, NF-κB, independent of TLR and IL-1R activation [125]. Similarly, CML-derived EVs have been demonstrated to alter the macrophage polarization to a more M2 phenotype within the tumor microenvironment, hence demonstrating up-regulation of IL-10 and TNF-α expression while down-regulating macrophage NO and ROS generation [126]. More often, in a tumor environment, M2 macrophages are converted into tumor-associated macrophages (TAMs) with the potential of releasing pro-tumorigenic growth factors, cytokines, and chemokines to enhance tumor progression [132,133,134,135].
Like monocytes, granulocytes, such as neutrophils, are highly plastic cells that can be readily influenced by tumor-derived EVs. Tumor-derived EV-treated neutrophils have been shown to promote NET formation and reduce the generation of suppressor cells, which are beneficial for tumor progression [127,128,129].
MDSCs, the primary cells associated with reducing the tumorigenicity of T-cells and NK-cells, have been shown to be modulated by heat shock proteins (HSPs) inside the EVs, such as HSP70 and HSP72 [124,136,137].
Crosstalk between tumor cells and stromal cells through EVs. Stromal cells refer to a highly heterogenous population of cells that provide structural and physiological support for hematopoietic cells. In cancer, stromal cells often contribute to disease progression by supporting growth, development, and metastasis of tumors (Figure 3). Table 3 also summarizes interactions between tumor cells with cells in the TME via the release of EVs.
In CLL patients, activation of the tumor microenvironment, thereby promoting disease progression, is favored by tumor cell-secreted EVs [138]. CLL-derived EVs activate the AKT survival pathway in stromal cells, in turn releasing vascular endothelial growth factor (VEGF), thereby contributing to tumor survival [139]. Again, CLL-derived EVs facilitate the conversion of stromal cells into cancer-associated fibroblasts (CAFs), promoting tumor metastasis and angiogenesis [140,141].
In acute lymphoblastic leukemia (ALL), EV-mediated transfer of galectin 3 (GAL3) from stromal cells to ALL cells stimulates endogenous GAL3 expression, which confers protection against drug treatment [142]. On the other hand, EVs derived from ALL cells induce a metabolic shift from oxidative phosphorylation to aerobic glycolysis in stromal cells [143].
Table 3. Tumor cells communicate with the stromal cells and vice versa via EVs transfer.
Table 3. Tumor cells communicate with the stromal cells and vice versa via EVs transfer.
Originating Cells Effector CellsMalignanciesFunctionsReferences
Tumor cellsStromal cellsCLLTumor-derived EVs induce stromal cells to release VEGF to promote tumor survival[139]
Tumor cellsStromal cellsCLLTumor-derived EVs convert stromal cells into CAFs, thereby promoting metastasis and angiogenesis[140,141]
Stromal cells Cancer cells ALLStromal cell-derived EVs induce GAL3 expression in cancer cells, hence inducing drug resistance[142]
Tumor cells Stromal cellsALL Switch oxidative phosphorylation to aerobic glycolysis in favor of cancer progression [143]
Stromal cellsTumor cellsMMStromal cells from MM induce proliferation, migration, and survival of tumor cells [144]
Tumor cells Endothelial cellsMMTumor cell-derived EVs in hypoxic conditions affect miR-135b, targeting HIF-1 pathway to promote angiogenesis[145]
Tumor cellsEndothelial cellsMM Tumor cell-secreted EVs activate endothelial STAT3 pathway, which promotes angiogenesis and tumor growth[146]
Tumor cellsOsteoclastsMM MM-derived EVs support osteoclast growth and migration[147]
Fibroblasts Tumor cells MM EVs carry clBcl-xL, which helps in EV uptake by the tumor cells to promote tumor proliferation[148]
Tumor cells MSCsATL Tumor cell-derived EVs transfer miR-155 and miR-21 to the MSCs, thereby inducing MSC proliferation[149]
Leukemic CD34+ cells MSCs AML AML CD34+ cell-derived EVs reduce further development of CD34+ cells from MSCs via miR-7977 [150]
MSCs CD34+ cellsMPNEVs carry miR-155 from MSCs, increasing granulocyte CFU numbers in neoplastic CD34+ cells[151]
Leukemic cells MSCsCMLCML-derived EVs induce IL-8 release from MSCs, thereby promoting CML survival[152]
BCR-ABL + tumor cells MononuclearCMLInduce genome instability, leading to malignant transformation of cells [153]
Tumor cells FibroblastsTCL, CML Transfer of hTERT mRNA via the EVs results in induced hTERT expression in the fibroblasts, leading to genome instability[154,155]
Abbreviations of malignancies: CLL, chronic lymphocytic leukemia; ALL, acute lymphocytic leukemia; MM, multiple myeloma; ATL, adult T-cell leukemia/lymphoma; AML, acute myeloid leukemia; MPN, myeloproliferative neoplasm; CML, chronic myelogenous leukemia, TCL, T-cell leukemia.
In the case of MM, stromal cell-derived EVs from MM induce proliferation, migration, and survival of tumor cells, whereas normal stromal cell-secreted EVs prevent tumor proliferation [144]. Under hypoxic conditions, MM-derived EVs were shown to affect miR-135b, which targets the HIF-1 pathway to induce tumor angiogenesis [145]. In another study, MM-derived EVs were reported to modulate the STAT3 pathway in endothelial cells, not only promoting angiogenesis but also inducing tumor growth via the release of VEGF and IL-6, respectively [146]. Moreover, EVs from MM are also capable of inducing growth and migration of osteoclasts (OCs), thus promoting the development of bone diseases in MM patients [147]. Stromal fibroblast-derived EVs actively carry Bcl-xL and its cleaved counterpart, clBcl-xL, facilitating the uptake of EVs by MM cells, hence promoting tumor proliferation [148].
Tumor cells from adult T-cell leukemia/lymphoma (ATL) often release miR-155 and miR-21 through EVs, triggering mesenchymal stem cell (MSC) proliferation and aiding the development of a friendly environment for leukemic progression [149]. Another study indicated that AML CD34+ cell-derived EVs efficiently transfer miR-7977 to MSCs to reduce proliferation of CD34+ cells [150].
miR-155 is found to be selectively packaged in the EVs of MSCs from myeloproliferative neoplasms (MPNs), increasing granulocyte CFU numbers in neoplastic CD34+ cells [151]. Moreover, leukemic cell survival in CML is enhanced by IL-8 production from MSCs upon incorporation of CML cell-derived EVs [152]. Another study showed that EVs from BCR-ABL+ CML tumor cells induced genomic instability in normal mononuclear cells, leading to malignant transformations [153]. Additionally, hTERT mRNA is known to be transported from TCL and CML cells to fibroblasts via EVs, leading to ectopic hTERT expression in fibroblasts [154] and a resultant switch to a tumor-like phenotype [155].
Interaction of EVs with the endothelium in the context of hematological malignancies. Hematological neoplasm-derived EVs are believed to communicate with the cells of the endothelium and vice versa. Hematological tumors, endowed with angiogenic-promoting ability, are dependent on the vascular endothelium for growth, migration, and invasion [156]. More often, tumor-EVs have been shown to activate the endothelial cells, contributing to tumor angiogenesis, whereas endothelial EVs in the TME induce tumor cells to remodel ECM components, thereby providing supplements for the growth of the tumor [157]. For example, EV-mediated transfer of VEGF and VEGF receptor from AML to endothelial cells promotes endothelial glycolysis, leading to vascular remodeling and chemoresistance [158,159]. Multiple signaling pathways appear to be regulated in MM, leading to increased viability of a bone marrow endothelial cell line (STR10), enhanced, angiogenesis and immunosuppression, further facilitating the progression of MM [120,159]. Piwi-interacting RNA-823 (piRNA-823), essentially carried by MM-derived EVs, transforms the endothelial cells, in turn promoting MM growth [159,160]. However, treatment of MM with a protease inhibitor, bortezomib, bestowed anti-angiogenic properties on MM-derived EVs [161]. CML-derived EVs induce the expression of VCAM-1 and IL-8 in endothelial cells, thereby promoting angiogenesis [162]. However, pretreatment of CML cells with curcumin decreases endothelial proliferation and migration via the release of miR-21-enriched EVs [163]. Moreover, CML-EVs, packaged with miR-126, have been found to negatively regulate the expression of VCAM-1 and CXCL12 in endothelial cells, thereby restricting CML motility and adhesion [159,164]. In CLL, tumor-derived EVs are shown to up-regulate the expression of ICAM-1, CXCL1, and IL-34 in endothelial cells, leading to angiogenesis and CLL proliferation [140]. In PML, EVs carry a significant amount of retinoic acid receptor-α (RAR-α), and the transfer of PML-EVs’ RAR-α to the endothelial cells results in the acquisition of tissue factor, hence imparting pro-coagulant properties to the endothelial cells [159,165].
Interaction of EVs with extracellular matrix (ECM). ECM, the physical scaffold, plays a pivotal role not only in the communication of cells with nearby cells but also in the growth, function, and movement of cells [166,167,168]. Tumor cells often communicate with the ECM via the release of EVs [169,170]. For example, surface heparan sulfate (sHS)-positive EVs from MM cells bind to one ECM component, fibronectin, thereby acting as a ligand for sHS-positive target cells. The binding of EVs’ fibronectin with sHS-positive cells triggers the activation of MAPKs (p38 and ERK1/2), resulting in the production of MMP-9 and DKK1, which essentially regulate the invasion of MM cells [171]. Cancer cell-derived EVs often induce the secretion of EMMPRIN, MMP-9, and IL-6 from human monocytic cell lines, thereby modulating the ECM to promote migration and inflammation, ultimately leading to tumor progression [172]. CD30+ EVs have often been shown to stick to long actin or tubulin protrusions of HL cells grown in 3D-matrigel or tissues, which could be a guiding mechanism of EVs to reach distant cells, thereby enabling cell–cell communication [173]. Figure 4 illustrates the contribution of EVs to the progression of hematological tumors by interacting with the ECM.
EVs in lymphatic malignancies. Lymphoma is a hematological malignancy associated with the lymphatic system [174,175]. A growing body of evidence has indicated that EVs actively participate in the pathogenesis of various lymphomas [176,177]. The incorporation of EVs, generated from EBV-associated lymphomas into monocytes or macrophages, transforms the immune regulatory mechanisms, leading to tumor evasion [178]. Lymphoma-derived EVs have often been shown to promote the angiogenic process by delivering angiogenic mRNA, miRNA, and proteins such as VEGF [179]. EBV lymphoma-induced macrophages have been shown to release secreted phospholipase A2 of group X (sPLA2-X), which hydrolyzes lymphoma-derived EVs’ phospholipids, thereby allowing for better uptake of EVs and associated lipid mediator signaling in TAMs, contributing to lymphoma growth [180]. HL-derived EVs were demonstrated to be internalized by TME fibroblasts and promote the release of pro-inflammatory cytokines, growth factors, and angiogenic factors, which together contribute to growth of lymphomas [181]. Moreover, HL-derived large EVs were shown to promote the release of IL-1β from monocytes depending on CD44 transfer, whereas both large and small HL-EVs confer immunomodulatory effects through eATP [182]. A recent study indicated that plasma EVs of pediatric HL can be used as a potential biomarker for relapse occurrence of HL [183]. DLBCL-derived EVs were found to be taken up by the tonsillar cells and stromal cells, which contribute to the progression of DLBCL [184]. Moreover, the PD-L1+ EV population was shown to be elevated in the plasma of DLBCL patients and could serve as a biomarker for DLBCL [185]. A recent study also indicated that DLBCL-tumor-derived EVs carry miR-125b-5p, which is readily taken up by DLBCL cells and reduces DLBCL sensitivity to rituximab via miR-125b-5p-mediated targeting of TNFAIP3, reflecting the ability of DLBCL-EVs to influence other cells in the TME [186]. Burkitt lymphoma-derived EVs not only inhibit autophagy and apoptosis but also promote lymphoma growth via miR-106a-mediated targeting of Beclin1 [187].

6. EVs: The Biomarker of Hematological Malignancies: Contribution in Drug Resistance

EVs as biomarkers. Biomarker identification, expressed differentially in different cell populations, as well as patients, is of prime importance for the diagnostic, prognostic, and therapeutic relevance of cancer. EVs carrying bioactive molecules such as proteins, DNA, RNA, miRNAs, etc., of the originating cells serve as excellent biological markers for the detection of cancer [188]. The release of EVs has been shown to be increased significantly during the development of cancer; hence, EVs in the body fluids of cancer patients serve as prognostic biomarkers for cancer [189,190,191]. The half-life of an EV-based biomarker is relatively longer since the content of the EVs is protected from extracellular proteases and nucleases, contributing to biomarker stability [192]. More often, at the disease stage, response to therapy, tumor burden, and survival are determined by the total protein content of the EVs from the plasma of melanomas and other solid tumors [131,193,194]. A detailed discussion about how EVs can be considered biomarkers in the context of different types of hematological malignancies appears below.
AML. EVs’ protein content in AML reflects the extent of the disease and correlates with the post-therapy likelihood of relapse. Low protein-containing EVs predict the long-term disease survival of AML. Moreover, a high level of TGF-β is observed in the EVs of AML patients upon diagnosis, which is dramatically reduced following chemotherapy, suggesting that EVs’ TGF-β levels mark the effect of chemotherapy in AML [189]. Moreover, EVs in the circulation of AML patients have been shown to be enriched with CD13, CD117, and CD34 [195,196]. EVs from AML cells also carry signature mRNA and miRNA molecules, thereby plausibly serving as future biomarkers [197].Signature mRNA molecules in the circulating EVs of AML patients often aid in the diagnosis and treatment of AML patients. For example, EVs’ FLT3-ITD and NPM1 mRNAs serve for determining AML prognosis; FLT3-ITD, IGF-IR, and CXCR4 mRNAs help in AML treatment, whereas IGF-IR, CXCR4, and MMP4 mRNAs aid in understanding the behavior of the tumor niche [197]. Moreover, miRNAs, such as miR-155, have been shown to be over-expressed in the serum EVs of AML patients compared to healthy individuals [198]. Therefore, AML-derived EVs could serve as diagnostic and prognostic biomarkers [131].
CML. CD13+ EVs were found to be up-regulated in the sera of CML patients compared to healthy individuals [195]. miR-215 is up-regulated in the plasma EVs of CML patients, and after successful treatment with imatinib, EVs’ miR-215 level is significantly reduced, serving as a predictor of successful treatment and discontinuing the drug [199].
MM. In the case of MM, plasma EVs, enriched with CD38, CD138, and superficially CD147, are considered to be biomarkers at different stages of the diseased condition [66,195,200]. For example, CD38+ EVs are associated with the clinical stages, and CD138+ EVs are correlated with disease stages and therapeutic response, whereas CD147+ EVs are related to the progression of MM. Moreover, CD44 enriched plasma EVs of MM patients are considered to be novel biomarkers for MM [201]. On the other hand, decreases in let-7b and miR-18a in the plasma EVs are associated with poor survival of MM patients [202].
CLL. Total EV populations in the plasma of CLL patients not only positively correlate with the advanced stages and overall patient survival but also with the duration of treatment in the initial stages of CLL [203]; these EVs are positive for CD19 and CD37. Another group reported that the level of plasma CD52+ EVs is increased in CLL patients and can be used as a predictive biomarker for CLL [139]. Furthermore, a moderate to high level of CD19, CD5, CD44, CD31, CD55, CD82, CD62L, HLA-A, B, C, and HLA-DR expression and a low level of CD49c, CD21, and CD63 expression was observed in CLL-EVs, which could be used as biomarkers for CLL [204]. A separate report indicated that EVs’ miR-155 could be considered a biomarker for patients suffering from B-cell CLL [205]. Chemotherapy-resistant individuals were found to high higher miR-155-laden EVs in the plasma, whereas their population was dramatically reduced in patients experiencing complete response [205].
HL. Patients with higher HL stages were shown to have a reduced level of CD30+ [a marker for Hodgkin and Reed–Sternberg (HRS) cells] EVs in the plasma [195]. The presence of miR-155, miR-127, miR21, and let-7 in the plasma EVs of HL patients often serves as a diagnostic tool for the detection and therapy of tumors in HL patients [198,206].
WL. In Waldenstrom macroglobulinemia (WM), a higher level of EVs and associated miR-155 was reported in the plasma compared to healthy controls, which could be used as a potential biomarker for WL [195,198]. However, EVs’ miRNA composition may change depending on the disease stages [207]. Table 4 summarizes how EVs act as a biomarker in different hematological malignancies.
EVs in drug resistance. Cancer cells often acquire resistance by decreased accumulation of drugs inside the cells, increased drug efflux outside the cells, compartmentalization of drugs, and alteration of cellular pathways targeted by specific drugs [208]. EVs have been shown to play an important role in modulating the drug resistance properties of various hematological neoplastic cells by various novel mechanisms. For example, BMSC-derived EVs confer resistance on MM cells against the chemotherapeutic drug bortezomib via the activation of diverse survival pathways, such as AKT, NOTCH1, NF-κB, and STAT3 [209]. Similarly, GAL3+ EVs from stromal cells confer anti-apoptosis and drug resistance to ALL cells by activating the NF-ĸB survival pathway [142]. In another study, BCL-derived EVs were found to express surface CD20, which helps BCLs to escape the humoral immune system by intercepting the chemotherapeutic drug rituximab [210]. The authors also indicated that sequestration of rituximab by EVs may contribute to the reduction in efficacy of pharmacological treatment [210]. Cancer cells often exert drug resistance properties by expelling the drugs into the extracellular space with the help of ATP binding cassette (ABC) transporter. ABCA3 transporters, highly expressed in leukemic cells, are localized on the exosome membrane within the MVBs, where drugs are effectively sequestered [210,211,212,213]. The release of exosomes is accompanied by the expulsion of drugs from the cancer cells, thereby contributing to drug resistance [210,214,215,216]. Furthermore, lymphoma cells, treated with chemotherapeutic drugs called anthracyclines, were shown to efflux the drug outside the cells through the release of EVs [217]. However, treatment of the cells with ABCA3 inhibitor increased the susceptibility of the drug via the suppression of EV biogenesis [217]. EV-mediated evasion of humoral immunotherapy was also reported to be dependent on ABCA3 in BCLs [210]. In the case of AML, exosomes from normal or AML bone marrow stromal cells (BMSCs) protect the leukemic cells owing to FLT3-ITD mutations against cytarabine; however, only AML-BMSC-exosomes impart protection against FLT3 inhibitor treatment [218]. Moreover, AML cells transfer their chemo-resistant properties to sensitive PML cells via the release of EVs [219].
In contrast to the above findings, EVs also effect potentiated chemotherapy-mediated elimination of cancer and other TME cells. For example, treatment with the anti-CD30 Ab drug conjugate Brentuximab Vedotin (SGN-35) in HL patients resulted in the binding of SGN-35 with HRS-derived CD30+ EVs and SGN-35/CD30+ EVs, not only killing the CD30+ tumor cells directly but also targeting CD30- cells in the TME, such as eosinophils and mast cells, resulting in severe damage to EV internalization [220]. Figure 5 describes how EVs influence chemotherapeutic treatment in the context of various hematological malignancies.
In summary, these studies indicate that EVs play a pivotal role in the regulation of hematological malignancies by modulating therapeutic resistance and tumor progression. Advancement of our understanding of cancer-associated EVs’ biology and novel techniques for isolating and characterizing the EVs will certainly help in predicting biomarkers for different hematological malignancies and in developing suitable targets.

7. Conclusions

A growing body of evidence indicates that EVs play a master role in cell–cell communication by delivering important messages among different types of cells. In recent years, the pace of EV research has been tremendous, and the ease at which EVs can be isolated with minimally invasive procedures has opened up possibilities for liquid biopsies and associated EVs to be used for real time monitoring of disease progression and treatment responses [221,222,223]. A recent multi-omics approach to longitudinally characterize EVs has been successful in elucidating hallmarks of COVID-19 throughout its progression [224]. Multi-omics studies have opened up avenues for quick, detailed characterization of EVs using new high-throughput techniques, as well as strict standardization, which could serve as an important benchmark for proper identification and could resolve potential problems arising due to the heterogeneity of EVs across patient population [225]. Some of the challenges could be overcome by utilizing EVs as an additional source of information when multiplexed with presently used biomarkers for better delineating disease mechanisms.
EVs have also shown promising results in drug delivery because of their inherent physiological stability and better immunological tolerance. They have been utilized to deliver different payloads ranging from onco-therapies to biologics and RNA therapeutics. Moreover, the growing body of evidence indicates the use of EVs as directed therapeutics or indirectly by stimulating immune cells [226,227]. Despite their therapeutic potential, several challenges remain to be overcome before the mass adoption of EV-based drugs. Roadblocks include several aspects of manufacturing, including heterogeneous production, which suffers from low yield and increasing complexity for scaling up production. On the bright side, there has been significant progress toward addressing these issues using microfluidic devices, along with bioreactors, to expedite production, as well as utilizing EV mimetics to address the problem of heterogeneity [228].
The findings reviewed here have raised new questions with plausible interpretations for understanding how cancer cells communicate with other hematologic cells in the TME and vice versa via the transfer of EVs. Moreover, EVs can be considered biomarkers for hematological malignancies, thereby helping not only in the diagnosis and prognosis but also in the development of therapeutic strategies against such malignancies.
Therefore, it is only a matter of time before the widespread adoption of EVs for multiple strategies. The increasing wealth of biological knowledge would only increase their potential applications, but future standardization practices across different fields must not be avoided to achieve a uniform approach to transforming EVs from bench to bedside applications.

Author Contributions

K.D. contributed to the conceptualization, literature search, experimental data collection, study design, critical review, and preparation of the final version of the manuscript. T.M. contributed to the literature search and experimental data collection and wrote the initial draft of the manuscript. P.S. contributed to the literature search and experimental data collection. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors thank Sara Rose for editing the manuscript for the English language.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

EVextracellular vesicle
MVmicrovesicle
SNAREsoluble N-ethylmaleimide-sensitive factor activating protein receptor
MVBmultivesicular body
ESCRTendosomal sorting complexes required for transport
TSG101tumor susceptibility gene 101
HSC70heat shock cognate 70-kDa protein
HSP90βheat shock protein 90 beta
CDcluster of differentiation
GRPGolgi and endoplasmic reticulum associated protein
CSE1Lchromosome segregation 1 like
TrpC5transient receptor potential cation channel subfamily C member 5
EMTepithelial to mesenchymal transition
JAKJanus kinase
PD-L1programmed death ligand 1
PD1programmed cell death protein 1
CLLchronic lymphocytic leukemia
DLBCLdiffuse large B cell lymphoma
BCL B-cell lymphoma
ATPadenosine triphosphate
MHC major histocompatibility complex
APO2LApo2 ligand
TCR T-cell receptor
NKG2Dnatural-killer group-2 member-D
APCantigen presenting cell
NKp30 natural killer protein 30
BAG6 Bcl2-associated athanogene cochaperone 6
MMmultiple myeloma
MDSC myeloid-derived suppressor cell
TGFβ1 transforming growth factor beta 1
AMLacute myeloid leukemia
NK natural killer
TNF-α tumor necrosis factor α
IL interleukin
MyD88myeloid differentiation primary response 88
NF-κBnuclear factor kappa-light-chain-enhancer of activated B cells
TLR Toll-like receptor
IL-1Rinterleukin-1 receptor
CMLchronic myelogenous leukemia
NO nitric oxide
ROS reactive oxygen species
TAMtumor-associated macrophage
NETneutrophil extracellular trap
VEGFvascular endothelial growth factor
CAFcancer-associated fibroblast
ALL acute lymphoblastic leukemia
GAL3 galectin 3
HIF hypoxia inducible factor
STATsignal transducer and activator of transcription
OC osteoclast
clBcl-xLcleaved Bcl-xL
MSCmesenchymal stem cell
ATL adult T-cell leukemia/lymphoma
MPNmyeloproliferative neoplasm
CFU colony forming unit
BCR B-cell receptor
hTERThuman telomerase reverse transcriptase
TCL T-cell lymphoma
sHSsurface heparan sulfate
MAPKmitogen-activated protein kinase
ERK extracellular signal-regulated kinase
MMP matrix metalloproteinase
EMMPRINextracellular matrix metalloproteinase inducer
NOTCHneurogenic locus notch homolog protein
ABCATP binding cassette
GLOBOCANGlobal Cancer Observatory
WHO World Health Organization
ABL1 Abelson murine leukemia viral oncogene homolog 1
PV polycythemia vera
ET essential thrombocytopenia
PFprimary myelofibrosis
CNL chronic neutrophilic leukemia
CELchronic eosinophilic leukemia
MDSmyelodysplastic neoplasms/syndrome
BMBbone marrow blasts
PB peripheral blasts
SF3B1splicing factor 3B subunit 1
TP53 tumor protein 53
IB increased blasts
NOSnot otherwise specified
DSDown syndrome
SLL small lymphocytic lymphoma
LPLlymphoplasmacytic lymphoma
MGUS monoclonal gammopathy of undetermined significance
IgM immunoglobulin M
MZLmarginal zone lymphoma
FL follicular lymphoma
MCL mantle cell lymphoma
BL Burkitt lymphoma
HL Hodgkin lymphoma
cHL classic HL
NLPHLnodular lymphocyte-predominant HL
PTCLperipheral T-cell lymphoma
ALCL anaplastic large cell lymphoma
ALK anaplastic lymphoma kinase
AITLangioimmunoblastic T-cell lymphoma
EBV Epstein–Barr virus
HTLV human T-lymphotropic virus
TLGLT-cell large granular lymphocyte leukemia
NKLGLNK-cell large granular lymphocyte leukemia
TK tyrosine kinase
HE hyper eosinophilia
AEC absolute eosinophil count
HEShypereosinophilic syndrome
PDGFR platelet-derived growth factor receptor
FGFR1fibroblast growth factor receptor 1
ETV6ETS (erythroblast transformation specific) variant transcription factor 6
FLT3 FMS-like tyrosine kinase 3
CM cutaneous mastocytosis
SM systemic mastocytosis
MPAL mixed phenotype acute leukemia
piRNA Piwi-interacting RNA
VCAM-1 vascular cell adhesion molecule 1
CXCL chemokine (C-X-C motif) ligand
PML progressive multifocal leukoencephalopathy
RAR-α retinoic acid receptor α
sPLA2-X soluble phospholipase A2 of group X
TNFAIP3 tumor necrosis factor alpha-induced protein 3
ITDinternal tandem duplication
IGF-IR insulin-like growth factor 1 (IGF-1) receptor
NPM1nucleophosmin gene 1
HLA human leukocyte antigen
HRS Hodgkin and Reed–Sternberg
WM Waldenstrom macroglobulinemia
BMSCbone-marrow stromal cell
HM hematological malignant

References

  1. Combarnous, Y.; Nguyen, T.M.D. Cell Communications among Microorganisms, Plants, and Animals: Origin, Evolution, and Interplays. Int. J. Mol. Sci. 2020, 21, 8052. [Google Scholar] [CrossRef] [PubMed]
  2. Huang, D.; Wang, R. Exploring the mechanisms of cell reprogramming and transdifferentiation via intercellular communication. Phys. Rev. E 2020, 102, 012406. [Google Scholar] [CrossRef] [PubMed]
  3. Das, K.; Prasad, R.; Roy, S.; Mukherjee, A.; Sen, P. The Protease Activated Receptor2 Promotes Rab5a Mediated Generation of Pro-metastatic Microvesicles. Sci. Rep. 2018, 8, 7357. [Google Scholar] [CrossRef] [PubMed]
  4. Das, K.; Prasad, R.; Singh, A.; Bhattacharya, A.; Roy, A.; Mallik, S.; Mukherjee, A.; Sen, P. Protease-activated receptor 2 promotes actomyosin dependent transforming microvesicles generation from human breast cancer. Mol. Carcinog. 2018, 57, 1707–1722. [Google Scholar] [CrossRef]
  5. Das, K.; Paul, S.; Singh, A.; Ghosh, A.; Roy, A.; Ansari, S.A.; Prasad, R.; Mukherjee, A.; Sen, P. Triple-negative breast cancer-derived microvesicles transfer microRNA221 to the recipient cells and thereby promote epithelial-to-mesenchymal transition. J. Biol. Chem. 2019, 294, 13681–13696. [Google Scholar] [CrossRef]
  6. Das, K.; Keshava, S.; Ansari, S.A.; Kondreddy, V.; Esmon, C.T.; Griffin, J.H.; Pendurthi, U.R.; Rao, L.V.M. Factor VIIa induces extracellular vesicles from the endothelium: A potential mechanism for its hemostatic effect. Blood 2021, 137, 3428–3442. [Google Scholar] [CrossRef]
  7. Das, K.; Keshava, S.; Pendurthi, U.R.; Rao, L.V.M. Factor VIIa suppresses inflammation and barrier disruption through the release of EEVs and transfer of microRNA 10a. Blood 2022, 139, 118–133. [Google Scholar] [CrossRef]
  8. Das, K.; Pendurthi, U.R.; Manco-Johnson, M.; Martin, E.J.; Brophy, D.F.; Rao, L.V.M. Factor VIIa treatment increases circulating extracellular vesicles in hemophilia patients: Implications for the therapeutic hemostatic effect of FVIIa. J. Thromb. Haemost. 2022, 20, 1928–1933. [Google Scholar] [CrossRef]
  9. Das, K.; Rao, L.V.M. The Role of microRNAs in Inflammation. Int. J. Mol. Sci. 2022, 23, 15479. [Google Scholar] [CrossRef]
  10. Gonzalez Fernandez, J.; Moncayo Arlandi, J.; Ochando, A.; Simon, C.; Vilella, F. The role of extracellular vesicles in intercellular communication in human reproduction. Clin. Sci. 2023, 137, 281–301. [Google Scholar] [CrossRef]
  11. Zaborowski, M.P.; Balaj, L.; Breakefield, X.O.; Lai, C.P. Extracellular Vesicles: Composition, Biological Relevance, and Methods of Study. Bioscience 2015, 65, 783–797. [Google Scholar] [CrossRef]
  12. Yanez-Mo, M.; Siljander, P.R.; Andreu, Z.; Zavec, A.B.; Borras, F.E.; Buzas, E.I.; Buzas, K.; Casal, E.; Cappello, F.; Carvalho, J.; et al. Biological properties of extracellular vesicles and their physiological functions. J. Extracell. Vesicles 2015, 4, 27066. [Google Scholar] [CrossRef]
  13. van Niel, G.; D’Angelo, G.; Raposo, G. Shedding light on the cell biology of extracellular vesicles. Nat. Rev. Mol. Cell Biol. 2018, 19, 213–228. [Google Scholar] [CrossRef]
  14. Chen, Y.; Zhao, Y.; Yin, Y.; Jia, X.; Mao, L. Mechanism of cargo sorting into small extracellular vesicles. Bioengineered 2021, 12, 8186–8201. [Google Scholar] [CrossRef]
  15. Raposo, G.; Stoorvogel, W. Extracellular vesicles: Exosomes, microvesicles, and friends. J. Cell Biol. 2013, 200, 373–383. [Google Scholar] [CrossRef]
  16. Neven, K.Y.; Nawrot, T.S.; Bollati, V. Extracellular Vesicles: How the External and Internal Environment Can Shape Cell-To-Cell Communication. Curr. Environ. Health Rep. 2017, 4, 30–37. [Google Scholar] [CrossRef]
  17. Muraoka, S.; Hirano, M.; Isoyama, J.; Ishida, M.; Tomonaga, T.; Adachi, J. Automated Proteomics Sample Preparation of Phosphatidylserine-Positive Extracellular Vesicles from Human Body Fluids. ACS Omega 2022, 7, 41472–41479. [Google Scholar] [CrossRef]
  18. Pulliero, A.; Pergoli, L.; La Maestra, S.; Micale, R.T.; Camoirano, A.; Bollati, V.; Izzotti, A.; De Flora, S. Extracellular vesicles in biological fluids. A biomarker of exposure to cigarette smoke and treatment with chemopreventive drugs. J. Prev. Med. Hyg. 2019, 60, E327–E336. [Google Scholar] [CrossRef]
  19. Borges, F.T.; Reis, L.A.; Schor, N. Extracellular vesicles: Structure, function, and potential clinical uses in renal diseases. Braz. J. Med. Biol. Res. 2013, 46, 824–830. [Google Scholar] [CrossRef]
  20. Liu, Y.J.; Wang, C. A review of the regulatory mechanisms of extracellular vesicles-mediated intercellular communication. Cell Commun. Signal. 2023, 21, 77. [Google Scholar] [CrossRef]
  21. Clancy, J.W.; Schmidtmann, M.; D’Souza-Schorey, C. The ins and outs of microvesicles. FASEB Bioadv. 2021, 3, 399–406. [Google Scholar] [CrossRef] [PubMed]
  22. Cai, H.; Reinisch, K.; Ferro-Novick, S. Coats, tethers, Rabs, and SNAREs work together to mediate the intracellular destination of a transport vesicle. Dev. Cell 2007, 12, 671–682. [Google Scholar] [CrossRef] [PubMed]
  23. Plawinski, L.; Cras, A.; Hernandez Lopez, J.R.; de la Pena, A.; Van der Heyden, A.; Belle, C.; Toti, F.; Angles-Cano, E. Distinguishing Plasmin-Generating Microvesicles: Tiny Messengers Involved in Fibrinolysis and Proteolysis. Int. J. Mol. Sci. 2023, 24, 1571. [Google Scholar] [CrossRef] [PubMed]
  24. Escola, J.M.; Kleijmeer, M.J.; Stoorvogel, W.; Griffith, J.M.; Yoshie, O.; Geuze, H.J. Selective enrichment of tetraspan proteins on the internal vesicles of multivesicular endosomes and on exosomes secreted by human B-lymphocytes. J. Biol. Chem. 1998, 273, 20121–20127. [Google Scholar] [CrossRef] [PubMed]
  25. Zoller, M. Tetraspanins: Push and pull in suppressing and promoting metastasis. Nat. Rev. Cancer 2009, 9, 40–55. [Google Scholar] [CrossRef]
  26. Di Vizio, D.; Morello, M.; Dudley, A.C.; Schow, P.W.; Adam, R.M.; Morley, S.; Mulholland, D.; Rotinen, M.; Hager, M.H.; Insabato, L.; et al. Large oncosomes in human prostate cancer tissues and in the circulation of mice with metastatic disease. Am. J. Pathol. 2012, 181, 1573–1584. [Google Scholar] [CrossRef]
  27. Heijnen, H.F.; Schiel, A.E.; Fijnheer, R.; Geuze, H.J.; Sixma, J.J. Activated platelets release two types of membrane vesicles: Microvesicles by surface shedding and exosomes derived from exocytosis of multivesicular bodies and alpha-granules. Blood 1999, 94, 3791–3799. [Google Scholar] [CrossRef]
  28. Morello, M.; Minciacchi, V.R.; de Candia, P.; Yang, J.; Posadas, E.; Kim, H.; Griffiths, D.; Bhowmick, N.; Chung, L.W.; Gandellini, P.; et al. Large oncosomes mediate intercellular transfer of functional microRNA. Cell Cycle 2013, 12, 3526–3536. [Google Scholar] [CrossRef]
  29. Rech, J.; Getinger-Panek, A.; Gałka, S.; Bednarek, I. Origin and Composition of Exosomes as Crucial Factors in Designing Drug Delivery Systems. Appl. Sci. 2022, 12, 12259. [Google Scholar] [CrossRef]
  30. Kowal, J.; Tkach, M.; Thery, C. Biogenesis and secretion of exosomes. Curr. Opin. Cell Biol. 2014, 29, 116–125. [Google Scholar] [CrossRef]
  31. Kim, Y.B.; Lee, G.B.; Moon, M.H. Size Separation of Exosomes and Microvesicles Using Flow Field-Flow Fractionation/Multiangle Light Scattering and Lipidomic Comparison. Anal. Chem. 2022, 94, 8958–8965. [Google Scholar] [CrossRef]
  32. 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]
  33. Simons, M.; Raposo, G. Exosomes—Vesicular carriers for intercellular communication. Curr. Opin. Cell Biol. 2009, 21, 575–581. [Google Scholar] [CrossRef]
  34. Thery, C.; Ostrowski, M.; Segura, E. Membrane vesicles as conveyors of immune responses. Nat. Rev. Immunol. 2009, 9, 581–593. [Google Scholar] [CrossRef]
  35. Mathivanan, S.; Simpson, R.J. ExoCarta: A compendium of exosomal proteins and RNA. Proteomics 2009, 9, 4997–5000. [Google Scholar] [CrossRef]
  36. Gurung, S.; Perocheau, D.; Touramanidou, L.; Baruteau, J. The exosome journey: From biogenesis to uptake and intracellular signalling. Cell Commun. Signal. 2021, 19, 47. [Google Scholar] [CrossRef]
  37. Babst, M.; Katzmann, D.J.; Estepa-Sabal, E.J.; Meerloo, T.; Emr, S.D. Escrt-III: An endosome-associated heterooligomeric protein complex required for mvb sorting. Dev. Cell 2002, 3, 271–282. [Google Scholar] [CrossRef]
  38. Wollert, T.; Hurley, J.H. Molecular mechanism of multivesicular body biogenesis by ESCRT complexes. Nature 2010, 464, 864–869. [Google Scholar] [CrossRef]
  39. Lee, Y.J.; Shin, K.J.; Jang, H.J.; Ryu, J.S.; Lee, C.Y.; Yoon, J.H.; Seo, J.K.; Park, S.; Lee, S.; Je, A.R.; et al. GPR143 controls ESCRT-dependent exosome biogenesis and promotes cancer metastasis. Dev. Cell 2023, 58, 320–334.e328. [Google Scholar] [CrossRef]
  40. Tauro, B.J.; Greening, D.W.; Mathias, R.A.; Ji, H.; Mathivanan, S.; Scott, A.M.; Simpson, R.J. Comparison of ultracentrifugation, density gradient separation, and immunoaffinity capture methods for isolating human colon cancer cell line LIM1863-derived exosomes. Methods 2012, 56, 293–304. [Google Scholar] [CrossRef]
  41. Morita, E.; Sandrin, V.; Chung, H.Y.; Morham, S.G.; Gygi, S.P.; Rodesch, C.K.; Sundquist, W.I. Human ESCRT and ALIX proteins interact with proteins of the midbody and function in cytokinesis. EMBO J. 2007, 26, 4215–4227. [Google Scholar] [CrossRef] [PubMed]
  42. Thery, C.; Boussac, M.; Veron, P.; Ricciardi-Castagnoli, P.; Raposo, G.; Garin, J.; Amigorena, S. Proteomic analysis of dendritic cell-derived exosomes: A secreted subcellular compartment distinct from apoptotic vesicles. J. Immunol. 2001, 166, 7309–7318. [Google Scholar] [CrossRef] [PubMed]
  43. van Niel, G.; Porto-Carreiro, I.; Simoes, S.; Raposo, G. Exosomes: A common pathway for a specialized function. J. Biochem. 2006, 140, 13–21. [Google Scholar] [CrossRef] [PubMed]
  44. Geminard, C.; De Gassart, A.; Blanc, L.; Vidal, M. Degradation of AP2 during reticulocyte maturation enhances binding of hsc70 and Alix to a common site on TFR for sorting into exosomes. Traffic 2004, 5, 181–193. [Google Scholar] [CrossRef] [PubMed]
  45. Stuffers, S.; Sem Wegner, C.; Stenmark, H.; Brech, A. Multivesicular endosome biogenesis in the absence of ESCRTs. Traffic 2009, 10, 925–937. [Google Scholar] [CrossRef]
  46. Buschow, S.I.; van Balkom, B.W.; Aalberts, M.; Heck, A.J.; Wauben, M.; Stoorvogel, W. MHC class II-associated proteins in B-cell exosomes and potential functional implications for exosome biogenesis. Immunol. Cell Biol. 2010, 88, 851–856. [Google Scholar] [CrossRef]
  47. Theos, A.C.; Truschel, S.T.; Tenza, D.; Hurbain, I.; Harper, D.C.; Berson, J.F.; Thomas, P.C.; Raposo, G.; Marks, M.S. A lumenal domain-dependent pathway for sorting to intralumenal vesicles of multivesicular endosomes involved in organelle morphogenesis. Dev. Cell 2006, 10, 343–354. [Google Scholar] [CrossRef]
  48. van Niel, G.; Charrin, S.; Simoes, S.; Romao, M.; Rochin, L.; Saftig, P.; Marks, M.S.; Rubinstein, E.; Raposo, G. The tetraspanin CD63 regulates ESCRT-independent and -dependent endosomal sorting during melanogenesis. Dev. Cell 2011, 21, 708–721. [Google Scholar] [CrossRef]
  49. Doyle, L.M.; Wang, M.Z. Overview of Extracellular Vesicles, Their Origin, Composition, Purpose, and Methods for Exosome Isolation and Analysis. Cells 2019, 8, 727. [Google Scholar] [CrossRef]
  50. Kalluri, R.; McAndrews, K.M. The role of extracellular vesicles in cancer. Cell 2023, 186, 1610–1626. [Google Scholar] [CrossRef]
  51. Mulcahy, L.A.; Pink, R.C.; Carter, D.R. Routes and mechanisms of extracellular vesicle uptake. J. Extracell. Vesicles 2014, 3, 24641. [Google Scholar] [CrossRef]
  52. O’Brien, K.; Ughetto, S.; Mahjoum, S.; Nair, A.V.; Breakefield, X.O. Uptake, functionality, and re-release of extracellular vesicle-encapsulated cargo. Cell Rep. 2022, 39, 110651. [Google Scholar] [CrossRef]
  53. Montecalvo, A.; Larregina, A.T.; Shufesky, W.J.; Stolz, D.B.; Sullivan, M.L.; Karlsson, J.M.; Baty, C.J.; Gibson, G.A.; Erdos, G.; Wang, Z.; et al. Mechanism of transfer of functional microRNAs between mouse dendritic cells via exosomes. Blood 2012, 119, 756–766. [Google Scholar] [CrossRef]
  54. Parolini, I.; Federici, C.; Raggi, C.; Lugini, L.; Palleschi, S.; De Milito, A.; Coscia, C.; Iessi, E.; Logozzi, M.; Molinari, A.; et al. Microenvironmental pH is a key factor for exosome traffic in tumor cells. J. Biol. Chem 2009, 284, 34211–34222. [Google Scholar] [CrossRef]
  55. Wang, Y.; Khan, H.M.; Zhou, C.; Liao, X.; Tang, P.; Song, P.; Gui, X.; Li, H.; Chen, Z.; Liu, S.; et al. Apoptotic cell-derived micro/nanosized extracellular vesicles in tissue regeneration. Nanotechnol. Rev. 2022, 11, 957–972. [Google Scholar] [CrossRef]
  56. Jeppesen, D.K.; Zhang, Q.; Franklin, J.L.; Coffey, R.J. Extracellular vesicles and nanoparticles: Emerging complexities. Trends Cell Biol. 2023. [Google Scholar] [CrossRef]
  57. Bano, R.; Ahmad, F.; Mohsin, M. A perspective on the isolation and characterization of extracellular vesicles from different biofluids. RSC Adv. 2021, 11, 19598–19615. [Google Scholar] [CrossRef]
  58. Wickman, G.; Julian, L.; Olson, M.F. How apoptotic cells aid in the removal of their own cold dead bodies. Cell Death Differ. 2012, 19, 735–742. [Google Scholar] [CrossRef]
  59. Zhang, M.; Lin, Y.; Chen, R.; Yu, H.; Li, Y.; Chen, M.; Dou, C.; Yin, P.; Zhang, L.; Tang, P. Ghost messages: Cell death signals spread. Cell Commun. Signal. 2023, 21, 6. [Google Scholar] [CrossRef]
  60. Escrevente, C.; Keller, S.; Altevogt, P.; Costa, J. Interaction and uptake of exosomes by ovarian cancer cells. BMC Cancer 2011, 11, 108. [Google Scholar] [CrossRef]
  61. Kerr, J.F.; Wyllie, A.H.; Currie, A.R. Apoptosis: A basic biological phenomenon with wide-ranging implications in tissue kinetics. Br. J. Cancer 1972, 26, 239–257. [Google Scholar] [CrossRef] [PubMed]
  62. Maisano, D.; Mimmi, S.; Dattilo, V.; Marino, F.; Gentile, M.; Vecchio, E.; Fiume, G.; Nistico, N.; Aloisio, A.; de Santo, M.P.; et al. A novel phage display based platform for exosome diversity characterization. Nanoscale 2022, 14, 2998–3003. [Google Scholar] [CrossRef] [PubMed]
  63. Luo, H.; Zhang, H.; Mao, J.; Cao, H.; Tao, Y.; Zhao, G.; Zhang, Z.; Zhang, N.; Liu, Z.; Zhang, J.; et al. Exosome-based nanoimmunotherapy targeting TAMs, a promising strategy for glioma. Cell Death Dis. 2023, 14, 235. [Google Scholar] [CrossRef] [PubMed]
  64. Raghavan, K.S.; Francescone, R.; Franco-Barraza, J.; Gardiner, J.C.; Vendramini-Costa, D.B.; Luong, T.; Pourmandi, N.; Andren, A.; Kurimchak, A.; Ogier, C.; et al. NetrinG1(+) cancer-associated fibroblasts generate unique extracellular vesicles that support the survival of pancreatic cancer cells under nutritional stress. Cancer Res. Commun. 2022, 2, 1017–1036. [Google Scholar] [CrossRef] [PubMed]
  65. Fathi, M.; Martinez-Paniagua, M.; Rezvan, A.; Montalvo, M.J.; Mohanty, V.; Chen, K.; Mani, S.A.; Varadarajan, N. Identifying signatures of EV secretion in metastatic breast cancer through functional single-cell profiling. iScience 2023, 26, 106482. [Google Scholar] [CrossRef]
  66. Arendt, B.K.; Walters, D.K.; Wu, X.; Tschumper, R.C.; Jelinek, D.F. Multiple myeloma dell-derived microvesicles are enriched in CD147 expression and enhance tumor cell proliferation. Oncotarget 2014, 5, 5686–5699. [Google Scholar] [CrossRef]
  67. McMasters, A.; McMasters, K.M.; Hao, H. Exosome to Promote Cancer Progression via Its Bioactive Cargoes. Arch. Cancer Biol. Ther. 2021, 2, 29–34. [Google Scholar]
  68. Barlin, M.; Erdmann-Gilmore, P.; Mudd, J.L.; Zhang, Q.; Seymour, R.W.; Guo, Z.; Miessner, J.R.; Goedegebuure, S.P.; Bi, Y.; Osorio, O.A.; et al. Proteins in Tumor-Derived Plasma Extracellular Vesicles Indicate Tumor Origin. Mol. Cell. Proteom. 2023, 22, 100476. [Google Scholar] [CrossRef]
  69. Liao, C.F.; Lin, S.H.; Chen, H.C.; Tai, C.J.; Chang, C.C.; Li, L.T.; Yeh, C.M.; Yeh, K.T.; Chen, Y.C.; Hsu, T.H.; et al. CSE1L, a novel microvesicle membrane protein, mediates Ras-triggered microvesicle generation and metastasis of tumor cells. Mol. Med. 2012, 18, 1269–1280. [Google Scholar] [CrossRef]
  70. Dong, Y.; Pan, Q.; Jiang, L.; Chen, Z.; Zhang, F.; Liu, Y.; Xing, H.; Shi, M.; Li, J.; Li, X.; et al. Tumor endothelial expression of P-glycoprotein upon microvesicular transfer of TrpC5 derived from adriamycin-resistant breast cancer cells. Biochem. Biophys. Res. Commun. 2014, 446, 85–90. [Google Scholar] [CrossRef]
  71. Jiang, J.; Li, J.; Zhou, X.; Zhao, X.; Huang, B.; Qin, Y. Exosomes Regulate the Epithelial-Mesenchymal Transition in Cancer. Front. Oncol. 2022, 12, 864980. [Google Scholar] [CrossRef]
  72. Chulpanova, D.S.; Pukhalskaia, T.V.; Rizvanov, A.A.; Solovyeva, V.V. Contribution of Tumor-Derived Extracellular Vesicles to Malignant Transformation of Normal Cells. Bioengineering 2022, 9, 245. [Google Scholar] [CrossRef]
  73. Robado de Lope, L.; Sanchez-Herrero, E.; Serna-Blasco, R.; Provencio, M.; Romero, A. Cancer as an infective disease: The role of EVs in tumorigenesis. Mol. Oncol. 2023, 17, 390–406. [Google Scholar] [CrossRef]
  74. Salem, A.E.; Shah, H.R.; Covington, M.F.; Koppula, B.R.; Fine, G.C.; Wiggins, R.H.; Hoffman, J.M.; Morton, K.A. PET-CT in Clinical Adult Oncology: I. Hematologic Malignancies. Cancers 2022, 14, 5941. [Google Scholar] [CrossRef]
  75. Sochacka-Cwikla, A.; Maczynski, M.; Regiec, A. FDA-Approved Drugs for Hematological Malignancies-The Last Decade Review. Cancers 2021, 14, 87. [Google Scholar] [CrossRef]
  76. Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer Statistics, 2021. CA Cancer J. Clin. 2021, 71, 7–33. [Google Scholar] [CrossRef]
  77. Chen, L.; Zheng, Y.; Yu, K.; Chen, S.; Wang, W.; Gale, R.P.; Liu, Z.X.; Liang, Y. Changing causes of death in persons with haematological cancers 1975–2016. Leukemia 2022, 36, 1850–1860. [Google Scholar] [CrossRef]
  78. Wedding, U. Palliative care of patients with haematological malignancies: Strategies to overcome difficulties via integrated care. Lancet Healthy Longev. 2021, 2, e746–e753. [Google Scholar] [CrossRef]
  79. Siegel, R.L.; Miller, K.D.; Wagle, N.S.; Jemal, A. Cancer statistics, 2023. CA Cancer J. Clin. 2023, 73, 17–48. [Google Scholar] [CrossRef]
  80. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef]
  81. Vijenthira, A.; Gong, I.Y.; Fox, T.A.; Booth, S.; Cook, G.; Fattizzo, B.; Martin-Moro, F.; Razanamahery, J.; Riches, J.C.; Zwicker, J.; et al. Outcomes of patients with hematologic malignancies and COVID-19: A systematic review and meta-analysis of 3377 patients. Blood 2020, 136, 2881–2892. [Google Scholar] [CrossRef] [PubMed]
  82. Langerbeins, P.; Hallek, M. COVID-19 in patients with hematologic malignancy. Blood 2022, 140, 236–252. [Google Scholar] [CrossRef] [PubMed]
  83. Pinana, J.L.; Martino, R.; Garcia-Garcia, I.; Parody, R.; Morales, M.D.; Benzo, G.; Gomez-Catalan, I.; Coll, R.; De La Fuente, I.; Luna, A.; et al. Risk factors and outcome of COVID-19 in patients with hematological malignancies. Exp. Hematol. Oncol. 2020, 9, 21. [Google Scholar] [CrossRef] [PubMed]
  84. Pagano, L.; Salmanton-Garcia, J.; Marchesi, F.; Busca, A.; Corradini, P.; Hoenigl, M.; Klimko, N.; Koehler, P.; Pagliuca, A.; Passamonti, F.; et al. COVID-19 infection in adult patients with hematological malignancies: A European Hematology Association Survey (EPICOVIDEHA). J. Hematol. Oncol. 2021, 14, 168. [Google Scholar] [CrossRef] [PubMed]
  85. Hus, I.; Szymczyk, A.; Manko, J.; Drozd-Sokolowska, J. COVID-19 in Adult Patients with Hematological Malignancies-Lessons Learned after Three Years of Pandemic. Biology 2023, 12, 545. [Google Scholar] [CrossRef]
  86. Lekkala, M.R.; Liesveld, J. Hematologic Malignancies, Classification, and Update on Modern Interventions. In Orofacial Supportive Care in Cancer: A Contemporary Oral Oncology Perspective; Nair, R., Ed.; Springer International Publishing: Cham, Switzerland, 2022; pp. 11–22. [Google Scholar]
  87. Khoury, J.D.; Solary, E.; Abla, O.; Akkari, Y.; Alaggio, R.; Apperley, J.F.; Bejar, R.; Berti, E.; Busque, L.; Chan, J.K.C.; et al. The 5th edition of the World Health Organization Classification of Haematolymphoid Tumours: Myeloid and Histiocytic/Dendritic Neoplasms. Leukemia 2022, 36, 1703–1719. [Google Scholar] [CrossRef]
  88. Alaggio, R.; Amador, C.; Anagnostopoulos, I.; Attygalle, A.D.; Araujo, I.B.O.; Berti, E.; Bhagat, G.; Borges, A.M.; Boyer, D.; Calaminici, M.; et al. The 5th edition of the World Health Organization Classification of Haematolymphoid Tumours: Lymphoid Neoplasms. Leukemia 2022, 36, 1720–1748. [Google Scholar] [CrossRef]
  89. Bejarano, L.; Jordao, M.J.C.; Joyce, J.A. Therapeutic Targeting of the Tumor Microenvironment. Cancer Discov. 2021, 11, 933–959. [Google Scholar] [CrossRef]
  90. Farc, O.; Cristea, V. An overview of the tumor microenvironment, from cells to complex networks (Review). Exp. Ther. Med. 2021, 21, 96. [Google Scholar] [CrossRef]
  91. Anderson, N.M.; Simon, M.C. The tumor microenvironment. Curr. Biol. 2020, 30, R921–R925. [Google Scholar] [CrossRef]
  92. Wang, Q.; Shao, X.; Zhang, Y.; Zhu, M.; Wang, F.X.C.; Mu, J.; Li, J.; Yao, H.; Chen, K. Role of tumor microenvironment in cancer progression and therapeutic strategy. Cancer Med. 2023. [Google Scholar] [CrossRef]
  93. Frisbie, L.; Buckanovich, R.J.; Coffman, L. Carcinoma-Associated Mesenchymal Stem/Stromal Cells: Architects of the Pro-tumorigenic Tumor Microenvironment. Stem Cells 2022, 40, 705–715. [Google Scholar] [CrossRef]
  94. Das, K.; Prasad, R.; Ansari, S.A.; Roy, A.; Mukherjee, A.; Sen, P. Matrix metalloproteinase-2: A key regulator in coagulation proteases mediated human breast cancer progression through autocrine signaling. Biomed. Pharmacother. 2018, 105, 395–406. [Google Scholar] [CrossRef]
  95. Baumann, Z.; Auf der Maur, P.; Bentires-Alj, M. Feed-forward loops between metastatic cancer cells and their microenvironment-the stage of escalation. EMBO Mol. Med. 2022, 14, e14283. [Google Scholar] [CrossRef]
  96. Kim, H.J.; Ji, Y.R.; Lee, Y.M. Cross-talk between angiogenesis and immune regulation in the tumor microenvironment. Arch. Pharm. Res. 2022, 45, 401–416. [Google Scholar] [CrossRef]
  97. Chakraborty, S.; Njah, K.; Hong, W. Agrin Mediates Angiogenesis in the Tumor Microenvironment. Trends Cancer 2020, 6, 81–85. [Google Scholar] [CrossRef]
  98. Sun, R.; Kong, X.; Qiu, X.; Huang, C.; Wong, P.P. The Emerging Roles of Pericytes in Modulating Tumor Microenvironment. Front. Cell Dev. Biol. 2021, 9, 676342. [Google Scholar] [CrossRef]
  99. Arner, E.N.; Rathmell, J.C. Metabolic programming and immune suppression in the tumor microenvironment. Cancer Cell 2023, 41, 421–433. [Google Scholar] [CrossRef]
  100. Noe, J.T.; Ding, C.; Geller, A.E.; Rendon, B.E.; Yan, J.; Mitchell, R.A. A Tumor-admixture Model to Interrogate Immune Cell-dependent Tumorigenesis. Bio-Protoc. 2023, 13, e4630. [Google Scholar] [CrossRef]
  101. Kubelkova, K.; Bostik, V.; Joshi, L.; Macela, A. Innate Immune Recognition, Integrated Stress Response, Infection, and Tumorigenesis. Biology 2023, 12, 499. [Google Scholar] [CrossRef]
  102. Goff, S.L.; Danforth, D.N. The Role of Immune Cells in Breast Tissue and Immunotherapy for the Treatment of Breast Cancer. Clin. Breast Cancer 2021, 21, e63–e73. [Google Scholar] [CrossRef] [PubMed]
  103. Pitt, J.M.; Kroemer, G.; Zitvogel, L. Extracellular vesicles: Masters of intercellular communication and potential clinical interventions. J. Clin. Investig. 2016, 126, 1139–1143. [Google Scholar] [CrossRef] [PubMed]
  104. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [PubMed]
  105. Czernek, L.; Duchler, M. Functions of Cancer-Derived Extracellular Vesicles in Immunosuppression. Arch. Immunol. Ther. Exp. 2017, 65, 311–323. [Google Scholar] [CrossRef]
  106. Zhang, X.; Yuan, X.; Shi, H.; Wu, L.; Qian, H.; Xu, W. Exosomes in cancer: Small particle, big player. J. Hematol. Oncol. 2015, 8, 83. [Google Scholar] [CrossRef]
  107. LeBien, T.W.; Tedder, T.F. B lymphocytes: How they develop and function. Blood 2008, 112, 1570–1580. [Google Scholar] [CrossRef]
  108. Whiteside, T.L. Immune modulation of T-cell and NK (natural killer) cell activities by TEXs (tumour-derived exosomes). Biochem. Soc. Trans. 2013, 41, 245–251. [Google Scholar] [CrossRef]
  109. Radulovic, K.; Rossini, V.; Manta, C.; Holzmann, K.; Kestler, H.A.; Niess, J.H. The early activation marker CD69 regulates the expression of chemokines and CD4 T cell accumulation in intestine. PLoS ONE 2013, 8, e65413. [Google Scholar] [CrossRef]
  110. Shiow, L.R.; Rosen, D.B.; Brdickova, N.; Xu, Y.; An, J.; Lanier, L.L.; Cyster, J.G.; Matloubian, M. CD69 acts downstream of interferon-alpha/beta to inhibit S1P1 and lymphocyte egress from lymphoid organs. Nature 2006, 440, 540–544. [Google Scholar] [CrossRef]
  111. Smallwood, D.T.; Apollonio, B.; Willimott, S.; Lezina, L.; Alharthi, A.; Ambrose, A.R.; De Rossi, G.; Ramsay, A.G.; Wagner, S.D. Extracellular vesicles released by CD40/IL-4-stimulated CLL cells confer altered functional properties to CD4+ T cells. Blood 2016, 128, 542–552. [Google Scholar] [CrossRef]
  112. Chen, Z.; You, L.; Wang, L.; Huang, X.; Liu, H.; Wei, J.Y.; Zhu, L.; Qian, W. Dual effect of DLBCL-derived EXOs in lymphoma to improve DC vaccine efficacy in vitro while favor tumorgenesis in vivo. J. Exp. Clin. Cancer Res. 2018, 37, 190. [Google Scholar] [CrossRef]
  113. Zhang, F.; Li, R.; Yang, Y.; Shi, C.; Shen, Y.; Lu, C.; Chen, Y.; Zhou, W.; Lin, A.; Yu, L.; et al. Specific Decrease in B-Cell-Derived Extracellular Vesicles Enhances Post-Chemotherapeutic CD8(+) T Cell Responses. Immunity 2019, 50, 738–750.e7. [Google Scholar] [CrossRef]
  114. Ghiringhelli, F.; Bruchard, M.; Chalmin, F.; Rebe, C. Production of adenosine by ectonucleotidases: A key factor in tumor immunoescape. J. Biomed. Biotechnol. 2012, 2012, 473712. [Google Scholar] [CrossRef]
  115. Busch, A.; Quast, T.; Keller, S.; Kolanus, W.; Knolle, P.; Altevogt, P.; Limmer, A. Transfer of T cell surface molecules to dendritic cells upon CD4+ T cell priming involves two distinct mechanisms. J. Immunol. 2008, 181, 3965–3973. [Google Scholar] [CrossRef]
  116. Hedlund, M.; Nagaeva, O.; Kargl, D.; Baranov, V.; Mincheva-Nilsson, L. Thermal- and oxidative stress causes enhanced release of NKG2D ligand-bearing immunosuppressive exosomes in leukemia/lymphoma T and B cells. PLoS ONE 2011, 6, e16899. [Google Scholar] [CrossRef]
  117. Xie, Y.; Zhang, H.; Li, W.; Deng, Y.; Munegowda, M.A.; Chibbar, R.; Qureshi, M.; Xiang, J. Dendritic cells recruit T cell exosomes via exosomal LFA-1 leading to inhibition of CD8+ CTL responses through downregulation of peptide/MHC class I and Fas ligand-mediated cytotoxicity. J. Immunol. 2010, 185, 5268–5278. [Google Scholar] [CrossRef]
  118. Reiners, K.S.; Topolar, D.; Henke, A.; Simhadri, V.R.; Kessler, J.; Sauer, M.; Bessler, M.; Hansen, H.P.; Tawadros, S.; Herling, M.; et al. Soluble ligands for NK cell receptors promote evasion of chronic lymphocytic leukemia cells from NK cell anti-tumor activity. Blood 2013, 121, 3658–3665. [Google Scholar] [CrossRef]
  119. Wang, J.; Faict, S.; Maes, K.; De Bruyne, E.; Van Valckenborgh, E.; Schots, R.; Vanderkerken, K.; Menu, E. Extracellular vesicle cross-talk in the bone marrow microenvironment: Implications in multiple myeloma. Oncotarget 2016, 7, 38927–38945. [Google Scholar] [CrossRef]
  120. 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]
  121. Garg, T.K.; Gann, J.I.; Malaviarachchi, P.A.; Stone, K.; Macleod, V.; Greenway, A.D.; Akel, N.S.; Edmondson, R.D.; Davies, F.E.; Epstein, J.; et al. Myeloma-Derived Exosomes and Soluble Factors Suppress Natural Killer Cell Function. Blood 2016, 128, 2066. [Google Scholar] [CrossRef]
  122. 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]
  123. Malavasi, F.; Chillemi, A.; Castella, B.; Schiavoni, I.; Incarnato, D.; Oliva, S.; Horenstein, A.L. CD38 and Antibody Therapy: What Can Basic Science Add? Blood 2016, 128, SCI-36. [Google Scholar] [CrossRef]
  124. Chalmin, F.; Ladoire, S.; Mignot, G.; Vincent, J.; Bruchard, M.; Remy-Martin, J.P.; Boireau, W.; Rouleau, A.; Simon, B.; Lanneau, D.; et al. Membrane-associated Hsp72 from tumor-derived exosomes mediates STAT3-dependent immunosuppressive function of mouse and human myeloid-derived suppressor cells. J. Clin. Investig. 2010, 120, 457–471. [Google Scholar] [CrossRef] [PubMed]
  125. Mancek-Keber, M.; Lainscek, D.; Bencina, M.; Chen, J.G.; Romih, R.; Hunter, Z.R.; Treon, S.P.; Jerala, R. Extracellular vesicle-mediated transfer of constitutively active MyD88(L265P) engages MyD88(wt) and activates signaling. Blood 2018, 131, 1720–1729. [Google Scholar] [CrossRef] [PubMed]
  126. Jafarzadeh, N.; Safari, Z.; Pornour, M.; Amirizadeh, N.; Forouzandeh Moghadam, M.; Sadeghizadeh, M. Alteration of cellular and immune-related properties of bone marrow mesenchymal stem cells and macrophages by K562 chronic myeloid leukemia cell derived exosomes. J. Cell. Physiol. 2019, 234, 3697–3710. [Google Scholar] [CrossRef]
  127. Demers, M.; Wagner, D.D. NETosis: A new factor in tumor progression and cancer-associated thrombosis. Semin. Thromb. Hemost. 2014, 40, 277–283. [Google Scholar] [CrossRef]
  128. Leal, A.C.; Mizurini, D.M.; Gomes, T.; Rochael, N.C.; Saraiva, E.M.; Dias, M.S.; Werneck, C.C.; Sielski, M.S.; Vicente, C.P.; Monteiro, R.Q. Tumor-Derived Exosomes Induce the Formation of Neutrophil Extracellular Traps: Implications For The Establishment of Cancer-Associated Thrombosis. Sci. Rep. 2017, 7, 6438. [Google Scholar] [CrossRef]
  129. Zhang, X.; Shi, H.; Yuan, X.; Jiang, P.; Qian, H.; Xu, W. Tumor-derived exosomes induce N2 polarization of neutrophils to promote gastric cancer cell migration. Mol. Cancer 2018, 17, 146. [Google Scholar] [CrossRef]
  130. Zhou, J.; Wang, S.; Sun, K.; Chng, W.J. The emerging roles of exosomes in leukemogeneis. Oncotarget 2016, 7, 50698–50707. [Google Scholar] [CrossRef]
  131. Hong, C.S.; Muller, L.; Whiteside, T.L.; Boyiadzis, M. Plasma exosomes as markers of therapeutic response in patients with acute myeloid leukemia. Front. Immunol. 2014, 5, 160. [Google Scholar] [CrossRef]
  132. Mantovani, A.; Sica, A.; Sozzani, S.; Allavena, P.; Vecchi, A.; Locati, M. The chemokine system in diverse forms of macrophage activation and polarization. Trends Immunol. 2004, 25, 677–686. [Google Scholar] [CrossRef]
  133. Rogers, T.L.; Holen, I. Tumour macrophages as potential targets of bisphosphonates. J. Transl. Med. 2011, 9, 177. [Google Scholar] [CrossRef]
  134. Quatromoni, J.G.; Eruslanov, E. Tumor-associated macrophages: Function, phenotype, and link to prognosis in human lung cancer. Am. J. Transl. Res. 2012, 4, 376–389. [Google Scholar]
  135. Schiavoni, G.; Gabriele, L.; Mattei, F. The tumor microenvironment: A pitch for multiple players. Front. Oncol. 2013, 3, 90. [Google Scholar] [CrossRef]
  136. Diao, J.; Yang, X.; Song, X.; Chen, S.; He, Y.; Wang, Q.; Chen, G.; Luo, C.; Wu, X.; Zhang, Y. Exosomal Hsp70 mediates immunosuppressive activity of the myeloid-derived suppressor cells via phosphorylation of Stat3. Med. Oncol. 2015, 32, 453. [Google Scholar] [CrossRef]
  137. Gobbo, J.; Marcion, G.; Cordonnier, M.; Dias, A.M.M.; Pernet, N.; Hammann, A.; Richaud, S.; Mjahed, H.; Isambert, N.; Clausse, V.; et al. Restoring Anticancer Immune Response by Targeting Tumor-Derived Exosomes With a HSP70 Peptide Aptamer. J. Natl. Cancer Inst. 2016, 108, djv330. [Google Scholar] [CrossRef]
  138. Ghosh, A.K.; Secreto, C.R.; Knox, T.R.; Ding, W.; Mukhopadhyay, D.; Kay, N.E. Circulating microvesicles in B-cell chronic lymphocytic leukemia can stimulate marrow stromal cells: Implications for disease progression. Blood 2010, 115, 1755–1764. [Google Scholar] [CrossRef]
  139. Boysen, J.; Nelson, M.; Magzoub, G.; Maiti, G.P.; Sinha, S.; Goswami, M.; Vesely, S.K.; Shanafelt, T.D.; Kay, N.E.; Ghosh, A.K. Dynamics of microvesicle generation in B-cell chronic lymphocytic leukemia: Implication in disease progression. Leukemia 2017, 31, 350–360. [Google Scholar] [CrossRef]
  140. Paggetti, J.; Haderk, F.; Seiffert, M.; Janji, B.; Distler, U.; Ammerlaan, W.; Kim, Y.J.; Adam, J.; Lichter, P.; Solary, E.; et al. Exosomes released by chronic lymphocytic leukemia cells induce the transition of stromal cells into cancer-associated fibroblasts. Blood 2015, 126, 1106–1117. [Google Scholar] [CrossRef]
  141. Farahani, M.; Rubbi, C.; Liu, L.; Slupsky, J.R.; Kalakonda, N. CLL Exosomes Modulate the Transcriptome and Behaviour of Recipient Stromal Cells and Are Selectively Enriched in miR-202-3p. PLoS ONE 2015, 10, e0141429. [Google Scholar] [CrossRef]
  142. Fei, F.; Joo, E.J.; Tarighat, S.S.; Schiffer, I.; Paz, H.; Fabbri, M.; Abdel-Azim, H.; Groffen, J.; Heisterkamp, N. B-cell precursor acute lymphoblastic leukemia and stromal cells communicate through Galectin-3. Oncotarget 2015, 6, 11378–11394. [Google Scholar] [CrossRef] [PubMed]
  143. Johnson, S.M.; Dempsey, C.; Chadwick, A.; Harrison, S.; Liu, J.; Di, Y.; McGinn, O.J.; Fiorillo, M.; Sotgia, F.; Lisanti, M.P.; et al. Metabolic reprogramming of bone marrow stromal cells by leukemic extracellular vesicles in acute lymphoblastic leukemia. Blood 2016, 128, 453–456. [Google Scholar] [CrossRef] [PubMed]
  144. 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]
  145. 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]
  146. Liu, Y.; Zhu, X.J.; Zeng, C.; Wu, P.H.; Wang, H.X.; Chen, Z.C.; Li, Q.B. Microvesicles secreted from human multiple myeloma cells promote angiogenesis. Acta Pharmacol. Sin. 2014, 35, 230–238. [Google Scholar] [CrossRef]
  147. 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]
  148. Vardaki, I.; Sanchez, C.; Fonseca, P.; Olsson, M.; Chioureas, D.; Rassidakis, G.; Ullen, A.; Zhivotovsky, B.; Bjorkholm, M.; Panaretakis, T. Caspase-3-dependent cleavage of Bcl-xL in the stroma exosomes is required for their uptake by hematological malignant cells. Blood 2016, 128, 2655–2665. [Google Scholar] [CrossRef]
  149. El-Saghir, J.; Nassar, F.; Tawil, N.; El-Sabban, M. ATL-derived exosomes modulate mesenchymal stem cells: Potential role in leukemia progression. Retrovirology 2016, 13, 73. [Google Scholar] [CrossRef]
  150. Horiguchi, H.; Kobune, M.; Kikuchi, S.; Yoshida, M.; Murata, M.; Murase, K.; Iyama, S.; Takada, K.; Sato, T.; Ono, K.; et al. Extracellular vesicle miR-7977 is involved in hematopoietic dysfunction of mesenchymal stromal cells via poly(rC) binding protein 1 reduction in myeloid neoplasms. Haematologica 2016, 101, 437–447. [Google Scholar] [CrossRef]
  151. Ramos, T.L.; Sánchez-Abarca, L.I.; Rosón, B.; Redondo, A.; Rodríguez, C.; Rodríguez, H.; Ortega, R.; Rico, A.; Preciado, S.; López Villar, O.; et al. Extracellular Vesicles Play an Important Role in Intercellular Communication Between Bone Marrow Stroma and Hematopoietic Progenitor Cells in Myeloproliferative Neoplasms. Blood 2016, 128, 1957. [Google Scholar] [CrossRef]
  152. Corrado, C.; Raimondo, S.; Saieva, L.; Flugy, A.M.; De Leo, G.; Alessandro, R. Exosome-mediated crosstalk between chronic myelogenous leukemia cells and human bone marrow stromal cells triggers an interleukin 8-dependent survival of leukemia cells. Cancer Lett. 2014, 348, 71–76. [Google Scholar] [CrossRef]
  153. Zhu, X.; You, Y.; Li, Q.; Zeng, C.; Fu, F.; Guo, A.; Zhang, H.; Zou, P.; Zhong, Z.; Wang, H.; et al. BCR-ABL1-positive microvesicles transform normal hematopoietic transplants through genomic instability: Implications for donor cell leukemia. Leukemia 2014, 28, 1666–1675. [Google Scholar] [CrossRef]
  154. Gutkin, A.; Uziel, O.; Beery, E.; Nordenberg, J.; Pinchasi, M.; Goldvaser, H.; Henick, S.; Goldberg, M.; Lahav, M. Tumor cells derived exosomes contain hTERT mRNA and transform nonmalignant fibroblasts into telomerase positive cells. Oncotarget 2016, 7, 59173–59188. [Google Scholar] [CrossRef]
  155. Jaiswal, R.K.; Kumar, P.; Yadava, P.K. Telomerase and its extracurricular activities. Cell. Mol. Biol. Lett. 2013, 18, 538–554. [Google Scholar] [CrossRef]
  156. Mancini, S.J.C.; Balabanian, K.; Corre, I.; Gavard, J.; Lazennec, G.; Le Bousse-Kerdiles, M.C.; Louache, F.; Maguer-Satta, V.; Mazure, N.M.; Mechta-Grigoriou, F.; et al. Deciphering Tumor Niches: Lessons From Solid and Hematological Malignancies. Front. Immunol. 2021, 12, 766275. [Google Scholar] [CrossRef]
  157. Gao, J.; Zhang, X.; Jiang, L.; Li, Y.; Zheng, Q. Tumor endothelial cell-derived extracellular vesicles contribute to tumor microenvironment remodeling. Cell Commun. Signal. 2022, 20, 97. [Google Scholar] [CrossRef]
  158. Wang, B.; Wang, X.; Hou, D.; Huang, Q.; Zhan, W.; Chen, C.; Liu, J.; You, R.; Xie, J.; Chen, P.; et al. Exosomes derived from acute myeloid leukemia cells promote chemoresistance by enhancing glycolysis-mediated vascular remodeling. J. Cell. Physiol. 2019, 234, 10602–10614. [Google Scholar] [CrossRef]
  159. Sun, G.; Gu, Q.; Zheng, J.; Cheng, H.; Cheng, T. Emerging roles of extracellular vesicles in normal and malignant hematopoiesis. J. Clin. Investig. 2022, 132. [Google Scholar] [CrossRef]
  160. 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]
  161. 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, 185–196. [Google Scholar] [CrossRef]
  162. Taverna, S.; Flugy, A.; Saieva, L.; Kohn, E.C.; Santoro, A.; Meraviglia, S.; De Leo, G.; Alessandro, R. Role of exosomes released by chronic myelogenous leukemia cells in angiogenesis. Int. J. Cancer 2012, 130, 2033–2043. [Google Scholar] [CrossRef] [PubMed]
  163. Taverna, S.; Fontana, S.; Monteleone, F.; Pucci, M.; Saieva, L.; De Caro, V.; Cardinale, V.G.; Giallombardo, M.; Vicario, E.; Rolfo, C.; et al. Curcumin modulates chronic myelogenous leukemia exosomes composition and affects angiogenic phenotype via exosomal miR-21. Oncotarget 2016, 7, 30420–30439. [Google Scholar] [CrossRef] [PubMed]
  164. Taverna, S.; Amodeo, V.; Saieva, L.; Russo, A.; Giallombardo, M.; De Leo, G.; Alessandro, R. Exosomal shuttling of miR-126 in endothelial cells modulates adhesive and migratory abilities of chronic myelogenous leukemia cells. Mol. Cancer 2014, 13, 169. [Google Scholar] [CrossRef] [PubMed]
  165. Fang, Y.; Garnier, D.; Lee, T.H.; D’Asti, E.; Montermini, L.; Meehan, B.; Rak, J. PML-RARa modulates the vascular signature of extracellular vesicles released by acute promyelocytic leukemia cells. Angiogenesis 2016, 19, 25–38. [Google Scholar] [CrossRef]
  166. Frantz, C.; Stewart, K.M.; Weaver, V.M. The extracellular matrix at a glance. J. Cell Sci. 2010, 123, 4195–4200. [Google Scholar] [CrossRef]
  167. Karamanos, N.K.; Theocharis, A.D.; Piperigkou, Z.; Manou, D.; Passi, A.; Skandalis, S.S.; Vynios, D.H.; Orian-Rousseau, V.; Ricard-Blum, S.; Schmelzer, C.E.H.; et al. A guide to the composition and functions of the extracellular matrix. FEBS J. 2021, 288, 6850–6912. [Google Scholar] [CrossRef]
  168. Dolega, M.E.; Monnier, S.; Brunel, B.; Joanny, J.F.; Recho, P.; Cappello, G. Extracellular matrix in multicellular aggregates acts as a pressure sensor controlling cell proliferation and motility. Elife 2021, 10, e63258. [Google Scholar] [CrossRef]
  169. Patel, N.J.; Ashraf, A.; Chung, E.J. Extracellular Vesicles as Regulators of the Extracellular Matrix. Bioengineering 2023, 10, 136. [Google Scholar] [CrossRef]
  170. Sheta, M.; Taha, E.A.; Lu, Y.; Eguchi, T. Extracellular Vesicles: New Classification and Tumor Immunosuppression. Biology 2023, 12, 110. [Google Scholar] [CrossRef]
  171. Purushothaman, A.; Bandari, S.K.; Liu, J.; Mobley, J.A.; Brown, E.E.; Sanderson, R.D. Fibronectin on the Surface of Myeloma Cell-derived Exosomes Mediates Exosome-Cell Interactions. J. Biol. Chem. 2016, 291, 1652–1663. [Google Scholar] [CrossRef]
  172. Redzic, J.S.; Kendrick, A.A.; Bahmed, K.; Dahl, K.D.; Pearson, C.G.; Robinson, W.A.; Robinson, S.E.; Graner, M.W.; Eisenmesser, E.Z. Extracellular vesicles secreted from cancer cell lines stimulate secretion of MMP-9, IL-6, TGF-beta1 and EMMPRIN. PLoS ONE 2013, 8, e71225. [Google Scholar] [CrossRef]
  173. Hansen, H.P.; Engels, H.M.; Dams, M.; Paes Leme, A.F.; Pauletti, B.A.; Simhadri, V.L.; Durkop, H.; Reiners, K.S.; Barnert, S.; Engert, A.; et al. Protrusion-guided extracellular vesicles mediate CD30 trans-signalling in the microenvironment of Hodgkin’s lymphoma. J. Pathol. 2014, 232, 405–414. [Google Scholar] [CrossRef]
  174. Mugnaini, E.N.; Ghosh, N. Lymphoma. Prim. Care Clin. Off. Pract. 2016, 43, 661–675. [Google Scholar] [CrossRef]
  175. Cordone, I.; Masi, S.; Giannarelli, D.; Pasquale, A.; Conti, L.; Telera, S.; Pace, A.; Papa, E.; Marino, M.; de Fabritiis, P.; et al. Major Differences in Lymphocyte Subpopulations Between Cerebrospinal Fluid and Peripheral Blood in Non-Hodgkin Lymphoma Without Leptomeningeal Involvement: Flow Cytometry Evidence of a Cerebral Lymphatic System. Front. Oncol. 2021, 11, 685786. [Google Scholar] [CrossRef]
  176. Kulka, M.; Brennan, K.; Mc Gee, M. Investigation of canine extracellular vesicles in diffuse large B-cell lymphomas. PLoS ONE 2022, 17, e0274261. [Google Scholar] [CrossRef]
  177. Matthiesen, R.; Gameiro, P.; Henriques, A.; Bodo, C.; Moraes, M.C.S.; Costa-Silva, B.; Cabecadas, J.; Gomes da Silva, M.; Beck, H.C.; Carvalho, A.S. Extracellular Vesicles in Diffuse Large B Cell Lymphoma: Characterization and Diagnostic Potential. Int. J. Mol. Sci. 2022, 23, 3327. [Google Scholar] [CrossRef]
  178. Higuchi, H.; Yamakawa, N.; Imadome, K.I.; Yahata, T.; Kotaki, R.; Ogata, J.; Kakizaki, M.; Fujita, K.; Lu, J.; Yokoyama, K.; et al. Role of exosomes as a proinflammatory mediator in the development of EBV-associated lymphoma. Blood 2018, 131, 2552–2567. [Google Scholar] [CrossRef]
  179. Yang, J.; Li, W.; He, X.; Zhang, G.; Yue, L.; Chai, Y. VEGF overexpression is a valuable prognostic factor for non-Hodgkin’s lymphoma evidence from a systemic meta-analysis. Dis. Markers 2015, 2015, 786790. [Google Scholar] [CrossRef]
  180. Kudo, K.; Miki, Y.; Carreras, J.; Nakayama, S.; Nakamoto, Y.; Ito, M.; Nagashima, E.; Yamamoto, K.; Higuchi, H.; Morita, S.Y.; et al. Secreted phospholipase A(2) modifies extracellular vesicles and accelerates B cell lymphoma. Cell Metab. 2022, 34, 615–633.e618. [Google Scholar] [CrossRef]
  181. Dorsam, B.; Bosl, T.; Reiners, K.S.; Barnert, S.; Schubert, R.; Shatnyeva, O.; Zigrino, P.; Engert, A.; Hansen, H.P.; von Strandmann, E.P. Hodgkin Lymphoma-Derived Extracellular Vesicles Change the Secretome of Fibroblasts Toward a CAF Phenotype. Front. Immunol. 2018, 9, 1358. [Google Scholar] [CrossRef]
  182. Maltoni, F.; Ciardiello, C.; Nanni, L.; Barone, M.; Narimanfar, G.; Casadei, B.; Argnani, L.; Broccoli, A.; Forte, D.; Budillon, A.; et al. Critical Role of Hodgkin Lymphoma Cells-Derived Large and Small Extracellular Vesicles in the Regulation of the Immune-Inflammatory Microenvironment. Blood 2022, 140, 11509–11511. [Google Scholar] [CrossRef]
  183. Repetto, O.; Lovisa, F.; Elia, C.; Enderle, D.; Romanato, F.; Buffardi, S.; Sala, A.; Pillon, M.; Steffan, A.; Burnelli, R.; et al. Proteomic Exploration of Plasma Exosomes and Other Small Extracellular Vesicles in Pediatric Hodgkin Lymphoma: A Potential Source of Biomarkers for Relapse Occurrence. Diagnostics 2021, 11, 917. [Google Scholar] [CrossRef] [PubMed]
  184. Rutherford, S.C.; Fachel, A.A.; Li, S.; Sawh, S.; Muley, A.; Ishii, J.; Saxena, A.; Dominguez, P.M.; Caldas Lopes, E.; Agirre, X.; et al. Extracellular vesicles in DLBCL provide abundant clues to aberrant transcriptional programming and genomic alterations. Blood 2018, 132, e13–e23. [Google Scholar] [CrossRef] [PubMed]
  185. Li, J.W.; Shi, D.; Wan, X.C.; Hu, J.; Su, Y.F.; Zeng, Y.P.; Hu, Z.J.; Yu, B.H.; Zhang, Q.L.; Wei, P.; et al. Universal extracellular vesicles and PD-L1+ extracellular vesicles detected by single molecule array technology as circulating biomarkers for diffuse large B cell lymphoma. Oncoimmunology 2021, 10, 1995166. [Google Scholar] [CrossRef]
  186. Zhang, L.; Zhou, S.; Zhou, T.; Li, X.; Tang, J. Potential of the tumor-derived extracellular vesicles carrying the miR-125b-5p target TNFAIP3 in reducing the sensitivity of diffuse large B cell lymphoma to rituximab. Int. J. Oncol. 2021, 58, 31. [Google Scholar] [CrossRef]
  187. Tang, J.; Hu, P.; Zhou, S.; Zhou, T.; Li, X.; Zhang, L. Lymphoma cell-derived extracellular vesicles inhibit autophagy and apoptosis to promote lymphoma cell growth via the microRNA-106a/Beclin1 axis. Cell Cycle 2022, 21, 1280–1293. [Google Scholar] [CrossRef]
  188. Tickner, J.A.; Urquhart, A.J.; Stephenson, S.A.; Richard, D.J.; O’Byrne, K.J. Functions and therapeutic roles of exosomes in cancer. Front. Oncol. 2014, 4, 127. [Google Scholar] [CrossRef]
  189. Szczepanski, M.J.; Szajnik, M.; Welsh, A.; Whiteside, T.L.; Boyiadzis, M. Blast-derived microvesicles in sera from patients with acute myeloid leukemia suppress natural killer cell function via membrane-associated transforming growth factor-beta1. Haematologica 2011, 96, 1302–1309. [Google Scholar] [CrossRef]
  190. van Doormaal, F.F.; Kleinjan, A.; Di Nisio, M.; Buller, H.R.; Nieuwland, R. Cell-derived microvesicles and cancer. Neth. J. Med. 2009, 67, 266–273. [Google Scholar]
  191. Zocco, D.; Ferruzzi, P.; Cappello, F.; Kuo, W.P.; Fais, S. Extracellular vesicles as shuttles of tumor biomarkers and anti-tumor drugs. Front. Oncol. 2014, 4, 267. [Google Scholar] [CrossRef]
  192. 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]
  193. 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]
  194. Filipazzi, P.; Burdek, M.; Villa, A.; Rivoltini, L.; Huber, V. Recent advances on the role of tumor exosomes in immunosuppression and disease progression. Semin. Cancer Biol. 2012, 22, 342–349. [Google Scholar] [CrossRef]
  195. Caivano, A.; Laurenzana, I.; De Luca, L.; La Rocca, F.; Simeon, V.; Trino, S.; D’Auria, F.; Traficante, A.; Maietti, M.; Izzo, T.; et al. High serum levels of extracellular vesicles expressing malignancy-related markers are released in patients with various types of hematological neoplastic disorders. Tumour Biol. 2015, 36, 9739–9752. [Google Scholar] [CrossRef]
  196. Hong, C.S.; Muller, L.; Boyiadzis, M.; Whiteside, T.L. Isolation and characterization of CD34+ blast-derived exosomes in acute myeloid leukemia. PLoS ONE 2014, 9, e103310. [Google Scholar] [CrossRef]
  197. Huan, J.; Hornick, N.I.; Shurtleff, M.J.; Skinner, A.M.; Goloviznina, N.A.; Roberts, C.T., Jr.; Kurre, P. RNA trafficking by acute myelogenous leukemia exosomes. Cancer Res. 2013, 73, 918–929. [Google Scholar] [CrossRef]
  198. Caivano, A.; La Rocca, F.; Simeon, V.; Girasole, M.; Dinarelli, S.; Laurenzana, I.; De Stradis, A.; De Luca, L.; Trino, S.; Traficante, A.; et al. MicroRNA-155 in serum-derived extracellular vesicles as a potential biomarker for hematologic malignancies—A short report. Cell. Oncol. 2017, 40, 97–103. [Google Scholar] [CrossRef]
  199. Ohyashiki, K.; Umezu, T.; Katagiri, S.; Kobayashi, C.; Azuma, K.; Tauchi, T.; Okabe, S.; Fukuoka, Y.; Ohyashiki, J.H. Downregulation of Plasma miR-215 in Chronic Myeloid Leukemia Patients with Successful Discontinuation of Imatinib. Int. J. Mol. Sci. 2016, 17, 570. [Google Scholar] [CrossRef]
  200. Krishnan, S.R.; Luk, F.; Brown, R.D.; Suen, H.; Kwan, Y.; Bebawy, M. Isolation of Human CD138(+) Microparticles from the Plasma of Patients with Multiple Myeloma. Neoplasia 2016, 18, 25–32. [Google Scholar] [CrossRef]
  201. Harshman, S.W.; Canella, A.; Ciarlariello, P.D.; Agarwal, K.; Branson, O.E.; Rocci, A.; Cordero, H.; Phelps, M.A.; Hade, E.M.; Dubovsky, J.A.; et al. Proteomic characterization of circulating extracellular vesicles identifies novel serum myeloma associated markers. J. Proteom. 2016, 136, 89–98. [Google Scholar] [CrossRef]
  202. 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]
  203. De Luca, L.; D’Arena, G.; Simeon, V.; Trino, S.; Laurenzana, I.; Caivano, A.; La Rocca, F.; Villani, O.; Mansueto, G.; Deaglio, S.; et al. Characterization and prognostic relevance of circulating microvesicles in chronic lymphocytic leukemia. Leuk. Lymphoma 2017, 58, 1424–1432. [Google Scholar] [CrossRef] [PubMed]
  204. Belov, L.; Matic, K.J.; Hallal, S.; Best, O.G.; Mulligan, S.P.; Christopherson, R.I. Extensive surface protein profiles of extracellular vesicles from cancer cells may provide diagnostic signatures from blood samples. J. Extracell. Vesicles 2016, 5, 25355. [Google Scholar] [CrossRef] [PubMed]
  205. Ferrajoli, A.; Shanafelt, T.D.; Ivan, C.; Shimizu, M.; Rabe, K.G.; Nouraee, N.; Ikuo, M.; Ghosh, A.K.; Lerner, S.; Rassenti, L.Z.; et al. Prognostic value of miR-155 in individuals with monoclonal B-cell lymphocytosis and patients with B chronic lymphocytic leukemia. Blood 2013, 122, 1891–1899. [Google Scholar] [CrossRef]
  206. van Eijndhoven, M.A.; Zijlstra, J.M.; Groenewegen, N.J.; Drees, E.E.; van Niele, S.; Baglio, S.R.; Koppers-Lalic, D.; van der Voorn, H.; Libregts, S.F.; Wauben, M.H.; et al. Plasma vesicle miRNAs for therapy response monitoring in Hodgkin lymphoma patients. JCI Insight 2016, 1, e89631. [Google Scholar] [CrossRef]
  207. Bouyssou, J.M.C.; Aljawai, Y.; Yosef, A.; Manier, S.; Rawal, R.; Kokubun, K.; Perilla Glen, A.; Sacco, A.; Castillo, J.J.; Treon, S.P.; et al. Profiling of Circulating Exosomes in Patients with Waldenström Macroglobulinemia. Blood 2016, 128, 2940. [Google Scholar] [CrossRef]
  208. Cesi, G.; Walbrecq, G.; Margue, C.; Kreis, S. Transferring intercellular signals and traits between cancer cells: Extracellular vesicles as “homing pigeons”. Cell Commun. Signal. 2016, 14, 13. [Google Scholar] [CrossRef]
  209. 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]
  210. Aung, T.; Chapuy, B.; Vogel, D.; Wenzel, D.; Oppermann, M.; Lahmann, M.; Weinhage, T.; Menck, K.; Hupfeld, T.; Koch, R.; et al. Exosomal evasion of humoral immunotherapy in aggressive B-cell lymphoma modulated by ATP-binding cassette transporter A3. Proc. Natl. Acad. Sci. USA 2011, 108, 15336–15341. [Google Scholar] [CrossRef]
  211. Chapuy, B.; Koch, R.; Radunski, U.; Corsham, S.; Cheong, N.; Inagaki, N.; Ban, N.; Wenzel, D.; Reinhardt, D.; Zapf, A.; et al. Intracellular ABC transporter A3 confers multidrug resistance in leukemia cells by lysosomal drug sequestration. Leukemia 2008, 22, 1576–1586. [Google Scholar] [CrossRef]
  212. Chapuy, B.; Panse, M.; Radunski, U.; Koch, R.; Wenzel, D.; Inagaki, N.; Haase, D.; Truemper, L.; Wulf, G.G. ABC transporter A3 facilitates lysosomal sequestration of imatinib and modulates susceptibility of chronic myeloid leukemia cell lines to this drug. Haematologica 2009, 94, 1528–1536. [Google Scholar] [CrossRef]
  213. Efferth, T.; Gillet, J.P.; Sauerbrey, A.; Zintl, F.; Bertholet, V.; de Longueville, F.; Remacle, J.; Steinbach, D. Expression profiling of ATP-binding cassette transporters in childhood T-cell acute lymphoblastic leukemia. Mol. Cancer Ther. 2006, 5, 1986–1994. [Google Scholar] [CrossRef]
  214. Azmi, A.S.; Bao, B.; Sarkar, F.H. Exosomes in cancer development, metastasis, and drug resistance: A comprehensive review. Cancer Metastasis Rev. 2013, 32, 623–642. [Google Scholar] [CrossRef]
  215. Jones, P.M.; George, A.M. The ABC transporter structure and mechanism: Perspectives on recent research. Cell. Mol. Life Sci. 2004, 61, 682–699. [Google Scholar] [CrossRef]
  216. Lee, C.H. Reversing agents for ATP-binding cassette drug transporters. Methods Mol. Biol. 2010, 596, 325–340. [Google Scholar] [CrossRef]
  217. Koch, R.; Aung, T.; Vogel, D.; Chapuy, B.; Wenzel, D.; Becker, S.; Sinzig, U.; Venkataramani, V.; von Mach, T.; Jacob, R.; et al. Nuclear Trapping through Inhibition of Exosomal Export by Indomethacin Increases Cytostatic Efficacy of Doxorubicin and Pixantrone. Clin. Cancer Res. 2016, 22, 395–404. [Google Scholar] [CrossRef]
  218. Wojtuszkiewicz, A.; Schuurhuis, G.J.; Kessler, F.L.; Piersma, S.R.; Knol, J.C.; Pham, T.V.; Jansen, G.; Musters, R.J.; van Meerloo, J.; Assaraf, Y.G.; et al. Exosomes Secreted by Apoptosis-Resistant Acute Myeloid Leukemia (AML) Blasts Harbor Regulatory Network Proteins Potentially Involved in Antagonism of Apoptosis. Mol. Cell. Proteom. 2016, 15, 1281–1298. [Google Scholar] [CrossRef]
  219. Bouvy, C.; Wannez, A.; Laloy, J.; Chatelain, C.; Dogne, J.M. Transfer of multidrug resistance among acute myeloid leukemia cells via extracellular vesicles and their microRNA cargo. Leuk. Res. 2017, 62, 70–76. [Google Scholar] [CrossRef]
  220. Hansen, H.P.; Trad, A.; Dams, M.; Zigrino, P.; Moss, M.; Tator, M.; Schon, G.; Grenzi, P.C.; Bachurski, D.; Aquino, B.; et al. CD30 on extracellular vesicles from malignant Hodgkin cells supports damaging of CD30 ligand-expressing bystander cells with Brentuximab-Vedotin, in vitro. Oncotarget 2016, 7, 30523–30535. [Google Scholar] [CrossRef]
  221. McKiernan, J.; Donovan, M.J.; Margolis, E.; Partin, A.; Carter, B.; Brown, G.; Torkler, P.; Noerholm, M.; Skog, J.; Shore, N.; et al. A Prospective Adaptive Utility Trial to Validate Performance of a Novel Urine Exosome Gene Expression Assay to Predict High-grade Prostate Cancer in Patients with Prostate-specific Antigen 2–10ng/ml at Initial Biopsy. Eur. Urol. 2018, 74, 731–738. [Google Scholar] [CrossRef]
  222. Wang, W.W.; Sorokin, I.; Aleksic, I.; Fisher, H.; Kaufman, R.P., Jr.; Winer, A.; McNeill, B.; Gupta, R.; Tilki, D.; Fleshner, N.; et al. Expression of Small Noncoding RNAs in Urinary Exosomes Classifies Prostate Cancer into Indolent and Aggressive Disease. J. Urol. 2020, 204, 466–475. [Google Scholar] [CrossRef] [PubMed]
  223. Ramirez-Garrastacho, M.; Bajo-Santos, C.; Line, A.; Martens-Uzunova, E.S.; de la Fuente, J.M.; Moros, M.; Soekmadji, C.; Tasken, K.A.; Llorente, A. Extracellular vesicles as a source of prostate cancer biomarkers in liquid biopsies: A decade of research. Br. J. Cancer 2022, 126, 331–350. [Google Scholar] [CrossRef] [PubMed]
  224. Lam, S.M.; Zhang, C.; Wang, Z.; Ni, Z.; Zhang, S.; Yang, S.; Huang, X.; Mo, L.; Li, J.; Lee, B.; et al. A multi-omics investigation of the composition and function of extracellular vesicles along the temporal trajectory of COVID-19. Nat. Metab. 2021, 3, 909–922. [Google Scholar] [CrossRef] [PubMed]
  225. Shaba, E.; Vantaggiato, L.; Governini, L.; Haxhiu, A.; Sebastiani, G.; Fignani, D.; Grieco, G.E.; Bergantini, L.; Bini, L.; Landi, C. Multi-Omics Integrative Approach of Extracellular Vesicles: A Future Challenging Milestone. Proteomes 2022, 10, 12. [Google Scholar] [CrossRef]
  226. Herrmann, I.K.; Wood, M.J.A.; Fuhrmann, G. Extracellular vesicles as a next-generation drug delivery platform. Nat. Nanotechnol. 2021, 16, 748–759. [Google Scholar] [CrossRef]
  227. Urabe, F.; Kosaka, N.; Ito, K.; Kimura, T.; Egawa, S.; Ochiya, T. Extracellular vesicles as biomarkers and therapeutic targets for cancer. Am. J. Physiol. Cell Physiol. 2020, 318, C29–C39. [Google Scholar] [CrossRef]
  228. Wang, Z.; Mo, H.; He, Z.; Chen, A.; Cheng, P. Extracellular vesicles as an emerging drug delivery system for cancer treatment: Current strategies and recent advances. Biomed. Pharmacother. 2022, 153, 113480. [Google Scholar] [CrossRef]
Figure 1. EV biogenesis and uptake by target recipient cells. EVs mainly consist of MVs and exosomes. MVs are generated by plasma membrane outward budding, whereas exosomes are of endocytic origin. Both carry mRNAs, miRNAs, proteins, and other bioactive molecules. EVs are taken up by recipient cells, either by direct fusion with the plasma membrane or by the endocytic pathway. After EV uptake, the EVs’ cargo is released into the recipient cells, hence acting as intercellular communicators between cells.
Figure 1. EV biogenesis and uptake by target recipient cells. EVs mainly consist of MVs and exosomes. MVs are generated by plasma membrane outward budding, whereas exosomes are of endocytic origin. Both carry mRNAs, miRNAs, proteins, and other bioactive molecules. EVs are taken up by recipient cells, either by direct fusion with the plasma membrane or by the endocytic pathway. After EV uptake, the EVs’ cargo is released into the recipient cells, hence acting as intercellular communicators between cells.
Biomolecules 13 00897 g001
Figure 2. Effect of EVs generated during hematological malignancies on different immune cells. EVs, derived from tumor cells not only (1) induce apoptosis of T-cells, but also reduce T-cell (2) migration and (3) proliferation. Moreover, tumor-derived EVs (4) decrease the cytotoxicity of NK-cells and (5) restrain the processing of antigen by APCs. On the other hand, tumor-derived EVs (6) induce immunosuppressive functions of MDSCs, as well as (7) promote MDSCs growth. Again, tumor-derived EVs (8) not only prevent the generation of ROS in macrophages but also (9) promote macrophages’ pro-inflammatory response. (10) The differentiation of monocytes into dendritic cells is often perturbed by the incorporation of tumor-derived EVs into monocytes. All of these processes contribute to the development and progression of the tumor.
Figure 2. Effect of EVs generated during hematological malignancies on different immune cells. EVs, derived from tumor cells not only (1) induce apoptosis of T-cells, but also reduce T-cell (2) migration and (3) proliferation. Moreover, tumor-derived EVs (4) decrease the cytotoxicity of NK-cells and (5) restrain the processing of antigen by APCs. On the other hand, tumor-derived EVs (6) induce immunosuppressive functions of MDSCs, as well as (7) promote MDSCs growth. Again, tumor-derived EVs (8) not only prevent the generation of ROS in macrophages but also (9) promote macrophages’ pro-inflammatory response. (10) The differentiation of monocytes into dendritic cells is often perturbed by the incorporation of tumor-derived EVs into monocytes. All of these processes contribute to the development and progression of the tumor.
Biomolecules 13 00897 g002
Figure 3. Crosstalk between tumor cells and cells in the TME through EVs. (1) EVs derived from tumor cells (tEVs) target stromal cells to release VEGF, which in turn induces growth of the tumor. tEVs induce the conversion of stromal cells into CAFs, which promote (2) tumor metastasis and angiogenesis. Moreover, tEV-fused stromal cells (3) induce the rate of aerobic glycolysis in tumor cells. On the other hand, (4) stromal cell-derived EVs (sEVs) promote proliferation and migration and confer drug resistance to tumor cells. (5) tEVs, upon fusion with endothelial cells, often promote angiogenesis, whereas (6) tEV-fused endothelial cells in turn contribute to tumor growth. (7) tEVs also promote the growth and migration of osteoclasts. More often, tEVs are shown to induce genomic instability in (8) several mononuclear cells and (9) fibroblasts, resulting in their transformation into pro-cancerous cells. (10) On the other hand, fibroblast-derived EVs (fEVs) induce the proliferation of the tumor. (11) Moreover, tEVs are also shown to induce the proliferation of MSCs, which in turn contribute to the progression of the tumor.
Figure 3. Crosstalk between tumor cells and cells in the TME through EVs. (1) EVs derived from tumor cells (tEVs) target stromal cells to release VEGF, which in turn induces growth of the tumor. tEVs induce the conversion of stromal cells into CAFs, which promote (2) tumor metastasis and angiogenesis. Moreover, tEV-fused stromal cells (3) induce the rate of aerobic glycolysis in tumor cells. On the other hand, (4) stromal cell-derived EVs (sEVs) promote proliferation and migration and confer drug resistance to tumor cells. (5) tEVs, upon fusion with endothelial cells, often promote angiogenesis, whereas (6) tEV-fused endothelial cells in turn contribute to tumor growth. (7) tEVs also promote the growth and migration of osteoclasts. More often, tEVs are shown to induce genomic instability in (8) several mononuclear cells and (9) fibroblasts, resulting in their transformation into pro-cancerous cells. (10) On the other hand, fibroblast-derived EVs (fEVs) induce the proliferation of the tumor. (11) Moreover, tEVs are also shown to induce the proliferation of MSCs, which in turn contribute to the progression of the tumor.
Biomolecules 13 00897 g003
Figure 4. The interaction of HM-derived EVs with ECM leads to tumor progression. 1. HL-derived CD30+ EVs invade through ECM to reach distant innate immune cells (here shown is a dendritic cell, for example) that are positive for CD30 ligand (CD30L+), leading to tumor cell–immune cell interaction. 2. HM cells, such as MM-released sHS+ EVs, interact with ECM fibronectin, which further interacts with other ECM cells, thereby releasing MMP-9 and DKK1, which contribute to MM invasion through ECM. 3. HC-derived EVs induce monocytes to secrete EMMPRIN, MMP-9, and IL-6, which not only promote tumor invasion but also influence tumor inflammation, ultimately leading to tumor progression through ECM.
Figure 4. The interaction of HM-derived EVs with ECM leads to tumor progression. 1. HL-derived CD30+ EVs invade through ECM to reach distant innate immune cells (here shown is a dendritic cell, for example) that are positive for CD30 ligand (CD30L+), leading to tumor cell–immune cell interaction. 2. HM cells, such as MM-released sHS+ EVs, interact with ECM fibronectin, which further interacts with other ECM cells, thereby releasing MMP-9 and DKK1, which contribute to MM invasion through ECM. 3. HC-derived EVs induce monocytes to secrete EMMPRIN, MMP-9, and IL-6, which not only promote tumor invasion but also influence tumor inflammation, ultimately leading to tumor progression through ECM.
Biomolecules 13 00897 g004
Figure 5. EVs influence chemotherapeutic drug treatment in various hematological malignancies. 1. Stromal cells contributing to the survival of hematological malignant (HM) cells. Bone-marrow stromal cell (BMSCs)-derived EVs activate the survival pathways in MM cells; stromal cell-derived EVs also activate the survival pathway in ALL cells. 2. EV-mediated humoral immune escape. BCL-derived EVs help BCL cells to escape humoral immune responses of the host. 3. EV-mediated expulsion of chemotherapeutic drugs. Lymphoma cells expel the administered chemotherapeutic drugs, anthracyclines, via the release of EVs. 4. EV-dependent chemoresistance transfer. Chemo-resistant AML cells transfer their chemo-resistant properties to chemo-sensitive PML cells via EV secretion. 5. and 6. HM-EV-mediated mast cell and eosinophil damage in the TME and killing of tumor cells. Treatment with an anti-CD30 bound drug conjugate, Brentuximab Vedotin (SGN-35), in HL patients results in the binding of SGN-35 with CD30+ HL-EVs, which in turn either is internalized by the CD30- eosinophils and mast cell in the TME, resulting in cellular damage, or directly kills CD30+ tumor cells. Green dotted arrow indicates the survival of the tumor, whereas red dotted arrow represents death or damage of the tumor cells.
Figure 5. EVs influence chemotherapeutic drug treatment in various hematological malignancies. 1. Stromal cells contributing to the survival of hematological malignant (HM) cells. Bone-marrow stromal cell (BMSCs)-derived EVs activate the survival pathways in MM cells; stromal cell-derived EVs also activate the survival pathway in ALL cells. 2. EV-mediated humoral immune escape. BCL-derived EVs help BCL cells to escape humoral immune responses of the host. 3. EV-mediated expulsion of chemotherapeutic drugs. Lymphoma cells expel the administered chemotherapeutic drugs, anthracyclines, via the release of EVs. 4. EV-dependent chemoresistance transfer. Chemo-resistant AML cells transfer their chemo-resistant properties to chemo-sensitive PML cells via EV secretion. 5. and 6. HM-EV-mediated mast cell and eosinophil damage in the TME and killing of tumor cells. Treatment with an anti-CD30 bound drug conjugate, Brentuximab Vedotin (SGN-35), in HL patients results in the binding of SGN-35 with CD30+ HL-EVs, which in turn either is internalized by the CD30- eosinophils and mast cell in the TME, resulting in cellular damage, or directly kills CD30+ tumor cells. Green dotted arrow indicates the survival of the tumor, whereas red dotted arrow represents death or damage of the tumor cells.
Biomolecules 13 00897 g005
Table 1. Classification of hematological neoplasms according to World Health Organization, 5th Edition [87,88].
Table 1. Classification of hematological neoplasms according to World Health Organization, 5th Edition [87,88].
TypeSub-ClassSub-Sub-ClassCharacteristics
Myeloid neoplasmMPNCMLExcessive granulocyte proliferation leading to increased myeloid maturation
Classic BCR:ABL1-negative MPN: (1) PVIncreased red blood cells
(2) ETClonal disorder related to thrombocytosis
(3) PFPolyclonal increase in fibroblasts leading to BM fibrosis
CNLOverproduction of matured granulocytes
CELClonal proliferation of abnormal eosinophils resulting in hypereosinophilia
Myeloproliferative neoplasm, unclassifiableDoes not have specific MPN characteristics or possess overlapping MPN features
MDSAccording to genetic abnormalities: (1) Low blasts and isolated del(5q)5q deletion, BMB < 5% in BM, and PB < 2%, SF3B1/TP53 mutation
(2) Low blasts and SF3B1 mutationBMB < 5% and PB < 2%, SF3B1 mutation, ≥15% ring sideroblasts
(3) Biallelic TP53 inactivation BMB and PB < 20%; copy number loss/neutral results in loss of heterozygosity, complex cytogenetics
According to morphology: (1) Low blastsBMB < 5% and PB < 2%
(2) HypoblasticBM cellularity < 20%
(3) MDS-IBMDS-IB1: BMB 5–9% and PB 2–4%; MDS-IB2; BMB 10–19% and PB 5–19%; MDS with increased blasts and fibrosis: BM fibrosis with BMB 5–19% and PB 2–19%
AMLAML with recurring genetic abnormalitiesCategorized into characteristic chromosomal rearrangements and no karyotypic abnormalities
AML with myelodysplasia-related featuresLacks genetic features of AML but with history of MDS or MDS/MPN
Therapy-related AMLArises after chemo- or radiation therapy
AML-NOSWith clinical, morphologic, and immunophenotypic features but lacking AML diagnostic karyotype
Myeloid sarcomaTumor mass outside BM effaces into tissues
Myeloid proliferation related to DSDS, associated with increased AML
Myeloid neoplasm with mutated TP53MDS-mutated TP53<10% blasts
MDS/AML-mutated TP5310–19% blasts
AML-mutated TP53>20% blasts
Lymphoid neoplasmPrecursor lymphoid neoplasmB-cell ALL/LBLArises from B-cell precursor, representing most ALL/LBL
T-cell ALL/LBLArises from T-cell precursor, representing 15% ALL/LBL
Mature B-cell neoplasmCLL/SLLSmall, mature-appearing lymphocytes
LPLDerived from post-germinal center of B-cells
Monoclonal gammopathyA type of lymphoid neoplasm for primary cold agglutinin disease
Plasma cell neoplasmAssociated with terminally differentiated germinal centers of B-cells
Hairy cell leukemiaAssociated with post-germinal centers of B-cells
MZLAssociated with marginal zone of matured B-cells
FL: (1) Classic FLProliferation of centrocytes and centroblasts
(2) FL grade 3BUncontrolled proliferation of centroblasts
(3) FL-unusual featureComprising FL with blastoid features and FL with diffused growth pattern
MCLNeoplasm of naïve and antigen-presenting B-cells
DLBCLNeoplasm of matured B-cells
High-grade B-cell lymphoma Comprising aggressive BL and other high-grade B-cell lymphoma
HLcHLGerminal and post-germinal centers of B-cells; again classified as nodular sclerosis cHL, mixed cellularity cHL, and lymphocyte rich- or lymphocyte-depleted cHL
NLPHLAssociated with germinal neoplastic B-cells
Matured T-cell/NK-cell lineage lymphomaPTCL: (1) PTCL-NOSNot fitting the criteria of T-cell lymphomas
(2) ALCLAccording to the expression of ALK, sub-categorized into “ALCL, ALK-positive” or “ALCL, ALK-negative” and “breast implant-associated ALCL.”
(3) Follicular THComprising AITL and related follicular TH cell lymphomas
(4) Extranodal EBV-associated neoplasm arises in lymph nodes
NK/T-cell lymphoma nasal type
(5) Hepatosplenic T-cell lymphomaAggressive and related to immunosuppression
(6) Primary intestinal T-cell lymphomaAggressive T-cell lymphoma in intestinal tract
Primary cutaneous T-cell lymphomaLymphoma of cutaneous peripheral T-cells
ATLAssociated with peripheral T-cells from HTLV-1 infected CD4+ T-cells
TLGL leukemiaDerived from clonally expanded granular T-cells
T-cell polymorphocytic leukemia Highly aggressive, medium-sized matured T-cells
NKLGL leukemiaAggressive, associated with malignant NK-cells
AggressiveAggressive, associated with malignant NK-cells
AggressiveNasal type, associated with EBV infection
NK-cell leukemia
Myeloid and lymphoid neoplasm with eosinophilia and TK gene fusionHESPrimary (or neoplastic)Clonal eosinophilic expansion reaching underlying stem cells and myeloid and eosinophilic neoplasm
Secondary (or reactive)Polyclonal eosinophilic expansion mediated by overproduction of eosinophilic cytokines during parasitic infections; certain solid tumor T-cell lymphomas
IdiopathicThe underlying cause of HE remains unknown
Specific syndrome associated with HE-Disease complications and clinical presentation are not fully defined, e.g., EGPA and some immunodeficiencies
HEUS -Persistent unexplained HE; difficult to predict whether the patients will develop clinical manifestations leading to HES
MastocytosisSMIndolent SM70% of SM is found to be indolent SM, which may or may not cause skin lesions of maculopapular cutaneous mastocytosis (MPCM)
Smoldering SMRare, shows same phenotypic effects as indolent SM
Aggressive SMHighly aggressive, manifesting round, rather than spindle-shaped, mast cells with median survival from month to years
MCLSame phenotypic responses as aggressive SM
SM-AHNSimilar to SM phenotype but requiring urgent treatment depending on the disease stage in BM and extracutaneous sites
Histiocytic or dendritic neoplasmHistiocytic sarcoma-Malignant proliferation of cells with morphologic and immunophenotypic features of mature tissue histiocytes
Tumors of Langerhans cellsLangerhans cell histiocytosisMalignancies of cells expressing CD1a, langerin, and S100
Langerhans cell sarcomaHigh-grade malignancy with same features as Langerhans cell histiocytosis
Indeterminate dendritic cell tumor-Rare, involving proliferation of cells, spindle-shaped to ovoid, similar to IDC
Interdigitating dendritic cell sarcoma Very rare, involving spindle to ovoid cells resembling interdigitating dendritic cells
Follicular dendritic cell sarcoma Neoplastic proliferation of spindle to ovoid cells; morphologically and immunophenotypically similar to follicular dendritic cells
Inflammatory pseudotumor-like follicular/fibroblastic dendritic cell sarcoma Neoplastic spindle cells residing in lymphoplasmacytic infiltrate involving liver and spleen
Fibroblastic reticular cell tumor Very rare, spindle cells with cytokeratin involving skin, spleen, and lymph nodes
Disseminated juvenile xanthogranuloma Proliferation and dissemination of small oval histiocytes resembling dermal juvenile xanthogranuloma of skin and soft tissues
Erdheim–Chester disease Involving foamy histiocytes of bones leading to clonal proliferation and associated with heart, CNS, and retroperitoneum
Mixed myeloid and lymphoid neoplasmMPALPh+ MPALWith Philadelphia chromosome (Ph)
Ph categories: (1) MPAL with t(v;11q23.3); KMT2A-rearrangedMPAL associated with detectable t(v;11q23.3) and KMT2A rearrangement
(2) MPAL B/myeloid NOSMPAL with B-lymphoblast immunophenotype lacking Ph chromosome or t(v;11q23.3)
(3) MPAL T/myeloid, NOSMPAL with T-lymphoblast immunophenotype lacking Ph chromosome or t(v;11q23.3)
Abbreviations not provided in the text nor in the table: BCR, B-cell receptor; ABL1, Abelson murine leukemia viral oncogene homolog 1; SF3B1, splicing factor 3b subunit 1; TP53, tumor protein p53; EGPA, eosinophilic granulomatosis with polyangiitis; HEUS, hypereosinophilia of undetermined origin; SM-AHN, systemic mastocytosis with an associated hematological neoplasm; IDC, indeterminate cells; CNS, central nervous system; KMT2A, lysine methyltransferase 2A.
Table 4. EVs as biomarkers of different types of hematological malignancies.
Table 4. EVs as biomarkers of different types of hematological malignancies.
DiseaseEVs’ BiomarkerUp-/Down-Regulation References
AMLTGF-βUp[189]
CD13, CD117, CD34 Up[195,196]
FLT-ITD, NPM1, IGF-IR, CXCR4, MMP4 mRNA-[197]
miR-155Up[198]
CMLCD13Up[195]
miR-215Up[199]
MMCD38, CD138, CD147Up[66,195,200]
CD44Up [201]
let-7b, miR-18aDown[202]
CLLCD19, CD37Up[203]
CD52Up[139]
CD19, CD5, CD44, CD31, CD55, CD82, CD62L, HLA-A, B, C, HLA-DR Up[204]
CD49c, CD21, CD63Down[204]
miR-155Up[205]
HLCD30Down[195]
miR-155, miR-127, miR-21, let-7Up[195,206]
WLmiR-155Up[195,207]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Das, K.; Mukherjee, T.; Shankar, P. The Role of Extracellular Vesicles in the Pathogenesis of Hematological Malignancies: Interaction with Tumor Microenvironment; a Potential Biomarker and Targeted Therapy. Biomolecules 2023, 13, 897. https://doi.org/10.3390/biom13060897

AMA Style

Das K, Mukherjee T, Shankar P. The Role of Extracellular Vesicles in the Pathogenesis of Hematological Malignancies: Interaction with Tumor Microenvironment; a Potential Biomarker and Targeted Therapy. Biomolecules. 2023; 13(6):897. https://doi.org/10.3390/biom13060897

Chicago/Turabian Style

Das, Kaushik, Tanmoy Mukherjee, and Prem Shankar. 2023. "The Role of Extracellular Vesicles in the Pathogenesis of Hematological Malignancies: Interaction with Tumor Microenvironment; a Potential Biomarker and Targeted Therapy" Biomolecules 13, no. 6: 897. https://doi.org/10.3390/biom13060897

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