Next Article in Journal
Established and Emerging Mechanisms in the Pathogenesis of Arrhythmogenic Cardiomyopathy: A Multifaceted Disease
Next Article in Special Issue
Binding of RNA Aptamers to Membrane Lipid Rafts: Implications for Exosomal miRNAs Transfer from Cancer to Immune Cells
Previous Article in Journal
Exploiting Manipulated Small Extracellular Vesicles to Subvert Immunosuppression at the Tumor Microenvironment through Mannose Receptor/CD206 Targeting
Previous Article in Special Issue
Exosomes in Angiogenesis and Anti-angiogenic Therapy in Cancers
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Myeloid Cell Modulation by Tumor-Derived Extracellular Vesicles

1
Skin Cancer Unit, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
2
Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, 68167 Mannheim, Germany
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2020, 21(17), 6319; https://doi.org/10.3390/ijms21176319
Submission received: 29 July 2020 / Revised: 28 August 2020 / Accepted: 29 August 2020 / Published: 31 August 2020
(This article belongs to the Special Issue EVs in Cross-Talk between Cancer and Immune Cells)

Abstract

:
Extracellular vesicles (EV) can carry proteins, RNA and DNA, thus serving as communication tools between cells. Tumor cells secrete EV, which can be taken up by surrounding cells in the tumor microenvironment as well as by cells in distant organs. Tumor-derived EV (TEV) contain factors induced by tumor-associated hypoxia such as heat shock proteins or a variety of microRNA (miRNA). The interaction of TEV with tumor and host cells can promote cancer angiogenesis, invasion and metastasis. Myeloid cells are widely presented in tissues, comprise the majority of immune cells and play an essential role in immune reactions and tissue remodeling. However, in cancer, the differentiation of myeloid cells and their functions are impaired, resulting in tumor promotion. Such alterations are due to chronic inflammatory conditions associated with cancer and are mediated by the tumor secretome, including TEV. A high capacity of myeloid cells to clear EV from circulation put them in the central position in EV-mediated formation of pre-metastatic niches. The exposure of myeloid cells to TEV could trigger numerous signaling pathways. Progenitors of myeloid cells alter their differentiation upon the contact with TEV, resulting in the generation of myeloid-derived suppressor cells (MDSC), inhibiting anti-tumor function of T and natural killer (NK) cells and promoting thereby tumor progression. Furthermore, TEV can augment MDSC immunosuppressive capacity. Different subsets of mature myeloid cells such as monocytes, macrophages, dendritic cells (DC) and granulocytes take up TEV and acquire a protumorigenic phenotype. However, the delivery of tumor antigens to DC by TEV was shown to enhance their immunostimulatory capacity. The present review will discuss a diverse and complex EV-mediated crosstalk between tumor and myeloid cells in the context of the tumor type, TEV-associated cargo molecules and type of recipient cells.

Graphical Abstract

1. Introduction

1.1. Extracellular Vesicles

Initially thought to eliminate unneeded cell compounds, extracellular vesicles (EV) are now recognized as means of intracellular communication [1]. Current understanding of EV biogenesis enables their classification into exosomes and microvesicles, providing specific markers for EV characterization [1,2]. Exosomes are considered as smaller (50–150 nm) EV that originate from the endosomal system; thus, tetraspanins such as cluster of differentiation (CD)9, CD63, CD81 are used as exosome markers [1,3]. Microvesicles are larger (50–1000 nm) EV that shed directly from the cellular membrane, and, therefore, annexin A1 was suggested as a marker for microvesicles [1,2,3]. In addition to the differences in generation, EV subtypes show differential distribution of cargo molecules such as proteins, RNA and DNA that has been comprehensively reviewed elsewhere [1,2]. According to the guidelines proposed by the International Society for Extracellular Vesicles, the current review will use the definition “extracellular vesicles” since the final consensus on specific EV markers has not yet been reached [4].
In view of the function of EV as mediators of intercellular communication, an explicable interest is rising regarding the EV-mediated triggering of phenotypical changes in target cells, which can be induced by surface signaling and/or the uptake of EV [5]. Membrane-associated proteins, lipids or sugars on EV can interact with surface molecules of target cells, triggering intracellular signaling cascades and mediate the internalization of EV, in which lectin family receptors, adhesion molecules and numerous other receptor-ligand interactions are involved [5,6,7]. Moreover, EV are also considered to carry so called “surface cargo” of adsorbed plasma-derived molecules: immunoglobulins, complement proteins, cytokines, coagulation factors, enzymes, thiols, lipoproteins, DNA modulating the surface EV-cell interactions [8,9]. Although it is technically challenging to prove, which molecule is incorporated into EV membrane and which is absorbed, the diversity of EV surface components highlights the importance of surface interactions between EV and recipient cells.
The involvement of membrane lipid rafts in the function of EV is less well studied. The rafts can activate signaling pathways or perform sorting functions by regulating proteins associated with them [10]. It has been reported that EV express increased levels of certain lipids and thus differ from the cellular membrane of cells, from which they are derived [11]. Moreover, the regulation of the PI3K/Akt pathway by lipid rafts through the participation of insulin receptors or endothelial growth factor receptors, which are transported by EV, was already reported [10]. However, the biological functions of lipids found in EV have hardly been investigated.
Various studies have shown that microRNA (miRNA) essentially contribute to EV function and that their function can be lost when miRNA is depleted by knocking down crucial enzymes of miRNA biogenesis [12,13,14]. Since miRNA and other non-coding RNA play a pivotal role in EV-mediated cancer-host cell interaction [15], we provide in this review an insight into miRNA biogenesis and sorting into EV. EV contain large numbers of RNA that is intact and functional in recipient cells [16]. During the generation of EV, cytosolic RNA (miRNA, other small non-coding RNA, long non-coding RNA, mRNA, tRNA, and rRNA) are taken up into the lumen [16,17]; this could be confirmed by the observation that intact RNA can still be isolated from EV treated with RNAse. The RNA profile of EV often does not reflect that of the original cells, suggesting that the RNA recruitment into EV is a selective process [16]. Since EV secreted by different cell types share an overlapping set of miRNA, the existence of a common mechanism for selective miRNA export is conceivable [18]. However, the exact process by which miRNA is sorted into EV is unknown. Several studies suggest an involvement of RNA-binding proteins in the packaging of miRNA into EV by recognizing specific short miRNA motifs [19,20,21,22,23,24]. Post-transcriptional modifications of RNA seem to play a crucial role in this process [23,25].

1.2. Myeloid Cells

Myeloid cells differentiate from hematopoietic stem cells (HSC), represent the majority among immune cells and include monocytes, macrophages, DC and granulocytes [26]. HSC differentiate into common myeloid progenitors followed by granulocyte-macrophage progenitors (GMP) (Figure 1) [27]. Granulocytes (neutrophils, basophils and eosinophils) are developed from GMP under the influence of granulocyte colony-stimulating factor (G-CSF) through the stages of myeloblasts, promyelocytes, myelocytes, metamyelocytes and band cells. Monocytes and DC develop from GMP via monocyte/macrophage and DC progenitors induced by the macrophage colony-stimulating factor (M-CSF) [27].
Terminally differentiated macrophages, granulocytes and DC play a major role in the immune defense against pathogens and in the tissue remodeling [26,27]. However, myeloid cells can be reprogrammed to promote tumor progression and angiogenesis [28,29]. Under the conditions of chronic inflammation or cancer, GM-CSF, G-CSF, M-CSF, tumor necrosis factor (TNF)-α, interleukin (IL)-1β, IL-6, transforming growth factor (TGF)-β and IL-10 are constantly secreted, leading to the alteration of myelopoiesis and generation of myeloid-derived suppressor cells (MDSC) [30,31,32]. CCL2-dependent recruitment of inflammatory monocytes (Ly6ChiCD11b+CD11cMHCIIVCAM1CCR2+) to the cancer site and their exposure to M-CSF and other tumor-derived factors leads to the generation of tumor-associated macrophages (TAM) that demonstrate self-renewal capacity [29,33] Both MDSC and TAM are key players in cancer progression and will be discussed in this review.
In addition to these soluble factors, abundant EV production by tumor cells contributes to the modulation of host immune cells during cancer progression [34]. Macrophages are known to perform clearance of circulating EV [35,36,37]. Furthermore, fully differentiated cells like macrophages and mature DC were demonstrated to uptake EV more efficiently than monocytes or immature DC [38]. Taking into account the importance of EV for tumor development and metastasis and a high capacity of myeloid cells for EV internalization, the current review will discuss various aspects of myeloid cell modulation by TEV, leading to immune escape.

2. Tumor-Derived Extracellular Vesicles (TEV)

Tumors secrete EV, which can be locally taken up by neighboring cancer cells and cells from the tumor microenvironment promoting cancer growth [34]. Tumors can also release EV into the circulation and promote metastasis formation at distant organs [34]. These systemic TEV effects were shown to be organ-specific and dependent on the integrin expression on TEV [39].
In comparison to EV secreted by normal cells, TEV present a similar morphology but a distinct molecular composition, which reflects the stressed state of tumor cells [40]. Additionally, tumor cells secrete EV to a higher extent than normal cells [40]. Being one of the hallmarks of cancer, hypoxia largely accounts for these properties of TEV. Hypoxia and acidic pH lead to cellular stress in tumors and can induce increased release of EV with altered miRNA, protein and lipid cargo, promoting cancer angiogenesis, invasion and metastasis [41,42,43,44]. Moreover, TEV composition and secretion can be modulated by anti-cancer treatment [45].
Since EV can carry biomarkers that are relevant for tumor diagnostics, they are widely studied as a liquid biopsy source [46]. The identification of TEV among the whole EV population isolated from the peripheral blood or other liquids from cancer patients requires the recognition of specific tumor markers in the respective EV fractions. To meet these needs, several approaches to EV isolation and characterization are suggested. Recent advances have been made for the isolation of TEV in melanoma, when antibodies against melanoma antigen chondroitin sulfate proteoglycan (CSPG)4 were used for on-beads immunocapturing of melanoma-derived EV [47]. Size fractionation might aid identification of tumor-specific EV by reducing their heterogeneity [48]. For instance, human epidermal growth factor receptor 2, a marker of breast cancer, has been demonstrated to be stronger expressed in EV with the size of 20–100 nm, while prostate-specific membrane antigen (PSMA), a prostate cancer marker, was higher in the EV fraction of 200–1000 nm [48].
An analytical microfluidic platform has been developed for the isolation of tumor-specific EV-RNA, achieving 94% specificity and 10-fold yield increase as compared to other methods [49]. Simultaneous analysis of exosome markers (CD9, CD63 and CD81) with PSMA in nanoscale flow cytometry has been shown to be a useful method for characterizing the origin of TEV in prostate cancer [50]. Surface-enhanced Raman spectroscopy (SERS) nanotags enabled EV phenotyping by using simultaneously anti-CD63-conjugated magnetic beads and specific detection antibody-coated SERS nanotags [51]. Since heat-shock protein (HSP)70 is highly expressed in cancer cells and is associated with a worse prognosis, prospective clinical trial was initiated to study the capability of HSP70 as TEV biomarker [52]. This study has demonstrated that the HSP70 level in TEV is higher in metastatic than non-metastatic patients and that is inversely correlated with the therapy response.
Delivery of TEV to the immune cells can lead to various dysfunctions, causing tumor immune escape [53]. TEV can directly inhibit effector immune cells [54] or elicit their protumorigenic effects by modulating myeloid cells [55]. In contrast, TEV as carriers of tumor antigens might trigger an anti-tumor immune response via the transfer of tumor antigens to DC [56]. Therefore, understanding the complexity of EV-mediated crosstalk between tumor and myeloid cells is crucial for restoring and maintaining an efficient anti-tumor immune response.

3. EV in Crosstalk between Tumor Cells and Myeloid Progenitors

The model for the compartment of myeloid cells has been recently revised by Bassler et al. [26] and suggested to be rather a “motorway junction” on a hematopoietic “autobahn” than a “trunk of a tree”. The new model highlighted the ability of myeloid progenitors to dynamically respond to extracellular signals during stress, inflammation etc. Moreover, single-cell RNA-sequencing analysis indicates that HSC and progenitor cells exhibit an early precommitment for developing into distinct cell types [26,57]. Such precommitment becomes more pronounced during aging resulting in a shift from the lymphatic towards the myeloid compartment [58] that is in agreement with immunosuppression accompanying cancer development in elderly patients.
Immature myeloid progenitor cells are continuously generated in the bone marrow under physiological conditions [32]. They show no suppressive potential against natural killer (NK) and T cells and normally differentiate to mature myeloid cells; however, in cancer, their normal differentiation is affected. In that case, immature myeloid progenitor cells may obtain immunosuppressive capacity typical for MDSC [59]. EV can contribute here in several ways. Thus, an uptake of acute myeloid leukemia-derived EV, expressing c-Myc, by myeloid progenitor cells resulted in the selective proliferation of MDSC [60]. Moreover, it has been demonstrated that EV derived from Lewis lung carcinoma or breast cancer cells block the differentiation of myeloid progenitors into DC in vitro [61,62]. Melanoma-derived EV have been shown to transfer MET kinase to circulating vasculogenic and hematopoietic myeloid progenitor cells in vivo, enhancing bone marrow mobilization [63]. This transfer led to an increased formation of lung and bone metastases. Patients with metastatic melanoma showed increased total MET and phospho-MET levels in circulating EV. Moreover, these patients had elevated MET expression in circulating CD45- C-KITlow/+ tyrosine kinase with immunoglobulin-like and EGF-like domains (TIE)2+ bone marrow progenitor cells [63]. Furthermore, a direct immunosuppressive capacity towards effector T cell was obtained by murine immature myeloid cells through melanoma EV-mediated programmed death ligand (PD-L)1 upregulation via Toll-like receptor (TLR) signaling [64]. The upregulation of PD-L1 by myeloid and tumor cells induces T cell anergy through the interaction with PD-1 on T cells and represents a critically important immune escape mechanism shared by different cancers [65]. The blocking of PD-1/PD-L1 axis by anti-PD-1 antibodies significantly increased the efficiency of cancer immunotherapy [65].

4. EV in Crosstalk between Tumor Cells and Monocytes

Monocytes represent a subset of myeloid cells circulating in the peripheral blood and migrating into tissues during inflammation [66]. They play a key role in phagocytosis, tissue homeostasis and resolving overactivated immune responses, preventing thereby a tissue damage [66]. Monocytes have a high plasticity to differentiate into macrophages or DC, but this plasticity is either altered or reduced in cancer [67]. Being constantly produced by tumor cells, released into the peripheral blood and taken up by monocytes, TEV contribute to these changes. In fact, treatment of CD14+ monocytes with melanoma-derived or colorectal carcinoma-derived EV impaired their ability to differentiate into DC [68]. Furthermore, upon the exposure to melanoma TEV, monocytes change their cytokine secretion profile, upregulate immunosuppressive markers and gain an ability to inhibit the function of effector immune cells [69].
TEV derived from different cancer types share some similarity and induce the production of IL-6 [64,68,70,71], IL-10 [64,72,73,74], TNF-α [64,68,71], TGF-β, IL-1β, chemokine (C-C motif) ligand (CCL)2 and CCL4 [64,68,70,71] by monocytes. It has been demonstrated that IL-6 production triggered by TEV via TLR2 and TLR4 is needed for the activation of signal transducer and activator of transcription (STAT)3 [71]. TGF-β produced by monocytes upon treatment with TEV derived from melanoma or colorectal carcinoma cells was shown to mediate an inhibition of T cell activity [68].
Several groups have reported an upregulation of PD-L1 and downregulation of human leukocyte antigen (HLA)-DR in monocytes treated with TEV. In one study, melanoma-derived EV have been demonstrated to skew monocyte differentiation towards suppressive CD14+HLA-DR-/low cells, which also lose the expression of costimulatory molecules CD80 and CD86 [68]. Huber et al. [69] found that a special set of miRNA (miRNA-146a, -155, -125b, -100, -125a, -146b, -99b and let-7e) is responsible for HLA-DR downregulation, secretion of IL-6 and CCL2 by monocytes upon the exposure to melanoma EV, leading to the inhibition of T cell effector functions. Furthermore, melanoma-derived EV were found to convert monocytes into immunosuppressive MDSC via PD-L1 upregulation triggered by the signaling via the HSP86/TLR4/Nuclear factor (NF)-κB axis [64]. In chronic lymphocytic leukemia (CLL), the transfer of noncoding Y RNA hY4 to monocytes resulted in the upregulation of PD-L1 in a TLR7-dependent manner [70]. In addition, this study demonstrated that targeting TLR7 in vivo led to the reduction of tumor load. Moreover, glioblastoma stem cell-derived EV have been shown to upregulate PD-L1 on monocytes and skew them towards M2-like cells associated with the phosphorylation of STAT3, p70S6 kinase and extracellular signal-regulated kinase (Erk)1/2 [75]. In contrast, Lorgulescu et al. [76] stated that glioma-derived EV were not capable of inducing PD-L1 expression in peripheral monocytes. However, they caused a significant downregulation of HLA-DR expression. Pancreatic cancer-derived EV could downregulate HLA-DR, induce arginase-1 (Arg-1) expression, increase production of reactive oxygen species (ROS) and the levels of phosphorylated STAT1 and STAT3 in monocytes [77].
It has been demonstrated an acquisition of a tumor-associated macrophages (TAM)-like phenotype by monocytes exposed to TEV. The interaction between EV secreted by head and neck cancer cells (PCI-1 cells) and monocytes has been shown to activate the NF-κB pathway resulting in a TAM-like phenotype [78]. Hypopharyngeal carcinoma cells (FaDu cells) overexpressing the transcription factor Snail, a key inducer of epithelial to mesenchymal transition (EMT), have been shown to secrete miRNA-21-enriched EV, which increased the expression of M2 markers (CD163, CD206) and the pro-angiogenic ability of CD14+ monocytes [73]. Wang et al. [79] observed that monocytes exposed to gastric cancer-derived EV develop into PD-1+ macrophages, producing M2-like cytokines and inducing the suppression of CD8 T cell activity. Interestingly, a study with EV from poorly metastatic melanoma cells noted that these EV could stimulate an innate immune response and block metastasis into the lungs [80]. When mice were preconditioned with such EV, they developed significantly less melanoma metastases. The study showed further that this effect was mediated by the pigment epithelium-derived factor expressed in EV delivered to non-classical Ly6Clow monocytes. Therefore, existing evidence underlines the acquisition of immunosuppressive potential by monocytes upon the exposure to TEV, which was dependent on the metastatic potential of the EV-producing cancer cells.

5. EV in Crosstalk between Tumor Cells and Macrophages

Macrophages are terminally differentiated tissue-resident cells derived from yolk-sac embryonic precursors and replenished by circulating monocytes during inflammation [26]. Macrophages show high plasticity and constantly adapt to environmental changes by modifying their functional state. The main functions of macrophages include elimination of infectious agents, promotion of wound healing and regulation of adaptive immunity [81]. Two types of macrophages are described. M1 macrophages express high levels of IL-1β, IL-6, IL-12, IL-23, and TNF-α (pro-inflammatory cytokines), ROS, nitric oxide (NO), and low levels of IL-10 and TGF-β (anti-inflammatory cytokines). They are activated by interferon (IFN)-γ or bacterial lipopolysaccharides (LPS) and play a role in the elimination of tumor cells. In contrast, M2 macrophages express high levels of IL-10 and low levels of IL-12, are activated by IL-4, IL-10, IL-13 and glucocorticoid hormones and promote tumor progression [82,83]. TAM have characteristics similar to M2 macrophages. They produce CCL22, prostaglandin E2 and TGF β, leading to immunosuppression [59]. Similar to MDSC, TAM express PD-L1 and Arg-1, causing T cell anergy and apoptosis [84,85]. Using these mechanisms, TAM promote tumor growth, metastasis and angiogenesis, suppress anti-tumor immunity and protect tumor cells from chemotherapy-induced apoptosis [86,87]. Furthermore, their accumulation has been shown to be associated with a poor clinical outcome [88]. Since macrophages clear EV from systemic circulation as mentioned above, these cells could be an important target of TEV.
EV are known to facilitate pre-metastatic niche formation by polarizing macrophages towards M2 phenotype in ovarian cancer [89]. Initiation of pre-metastatic niche formation by TEV has also been shown in pancreatic cancer by the stimulation of TGF-β production by Kupffer cells and of fibronectin production by hepatic stellate cells, leading to the recruitment of bone marrow-derived macrophages [90].
Growing evidences suggested a role of TEV in regulating the polarization of macrophages [91]. Thus, EV produced by colorectal cancer and melanoma cells were reported to induce mixed M1 and M2 polarization and cytokine production [74,92,93]. Breast cancer-derived EV have been demonstrated to induce an IL-6-mediated pathway via gp130/STAT3 signaling, leading to TAM polarization [94]. Hypoxia-induced melanoma TEV have been demonstrated to promote M2 polarization of macrophages, to transfer let-7a miRNA and increase tumor growth [95]. Cheng et al. [96] stated that TEV derived from liver cancer cells can upregulate PD-L1 expression as well as cytokine secretion by macrophages through STAT3 signaling, and that the treatment with melatonin modulated the function of TEV, leading to the attenuation of immunosuppressive capacity of macrophages.
Extensive data show the involvement of miRNA in the TEV-mediated modulation of macrophage phenotype. In hypoxic glioma-derived EV, miRNA-1246 was found to be the most enriched miRNA. The transduction of macrophages with miRNA 1246 resulted in the activation of STAT3 and the inhibition of NF-κB signaling, leading to the skewing of macrophages towards a tumorigenic phenotype in vitro and in vivo [97]. Similarly, human macrophages transduced with miRNA-222-3p, which is significantly increased in EV isolated from serum of patients with epithelial ovarian cancer, upregulated the expression of genes linked to M2 polarization and showed increased IL-10 secretion in vitro [98]. In addition, miRNA-222-3p caused the downregulation of suppressor of cytokine signaling (SOCS)3 in these macrophages, induced STAT3 signaling and a TAM-like phenotypic polarization. As a result, miRNA-222-3p transfected macrophages promoted the growth of ovarian cancer cells in vitro and in vivo [98].
A study with hepatocellular carcinoma (HCC)-derived EV indicated that miRNA-146a 5p from EV contributed to M2-like macrophage polarization, and that the suppression of Sal-like protein (SALL)4, a transcription factor for miRNA-146-5p, reversed T cell exhaustion induced by these macrophages in vivo [99]. Furthermore, miRNA-150 found to be present in TEV accumulated in the plasma of HCC patients, promoted tumor development through increasing vascular endothelial growth factor (VEGF) secretion by TAM [100]. The inhibition of miRNA-150 in tumor-bearing mice suppressed VEGF secretion and tumor growth in this study. Additionally, the delivery of miRNA-21 to tumor-associated macrophages led to increased xenograft tumor progression in mice [73].
In another study, the delivery of miRNA-23a by EV derived from liver cancer cells to macrophages led to the upregulation of PD-L1 expression via a Phosphatidylinositol 3-kinase (PI3K)/Protein kinase Akt pathway resulting in the attenuation of CD8 T cell functions [101]. In contrast, the treatment of murine 4T1 breast cancer cells with epigallocatechin gallate induced the secretion of miRNA-16 containing EV, which inhibited TAM infiltration and M2-like polarization by the suppression of NF-κB activation [102]. Taken together, macrophages are highly affected by TEV, leading to protumor effects in most cases, although the exact outcome of this interaction should be related to the content of TEV and the type of cancer cells producing these EV.

6. EV in Crosstalk between Tumor Cells and Dendritic Cells

DC are terminally differentiated myeloid cells that present antigens to T cells and play a pivotal role in crosslinking innate and adaptive immunity [103]. During normal hematopoiesis, DC originate from HSC, whereas under inflammatory conditions, they arise from monocytes [26,103]. DC need a contact with bacteria, viruses or damaged tissues to be activated and can promote the differentiation of naïve T cells, inducing an immune response [104]. Mature DC infiltrate tumors and recruit anti-tumor effector immune cells [105]. However, in cancer, VEGF, M-CSF, IL-6, adenosine and hypoxia accumulated in the tumor microenvironment affect DC functions, leading to the enrichment of immature, functionally incompetent DC [106]. This results in a weak stimulation of the anti-tumor immune response and accumulation of immunosuppressive MDSC [107]. In contrast to monocytes, which are more likely exposed to TEV in the circulation, DC contact these vesicles in the tumor microenvironment and draining lymph nodes [108,109,110].
It has been observed that TEV derived from murine thymoma and melanoma cell lines are internalized by DC in the skin and trafficked to the draining lymph node, resulting in the inhibition of DC maturation and TGF-β production [111]. Besides, melanoma TEV have been shown to induce IL-6 production in DC, leading to STAT3-dependent matrix metalloproteinase 9 production by tumor cells, promoting thereby their invasiveness [112]. Such effect on DC was demonstrated to be TLR2/4-dependent and mediated by HSP72 and HSP105 expressed on the TEV surface. In vitro DC treatment with EV from lung cancer cells caused downregulation of CD80, CD86 and major histocompatibility complex (MHC)-II, whereas immunosuppressive markers including PD-L1 were upregulated [61]. In this study, the authors also found that Lewis lung carcinoma-derived EV inhibited DC migration to draining lymph nodes, downregulating chemokine receptors and reducing antigen-specific CD4 T cell proliferation in a PD-L1-dependent manner. The delivery of miRNA-212-3p by pancreatic cancer-derived EV led to the inhibition of the expression of RFXAP, a transcription factor for MHC-II, resulting in the downregulation of MHCII in vitro [113]. The authors concluded that TEV, containing miRNA-212-3p, induced tolerogenic DC. In another study, miRNA-203 delivered by pancreatic cancer-derived EV mediated the downregulation of TLR4 expression on DC [114].
In contrast to the above described inhibition of DC immunostimulatory activity, TEV were also reported to convey tumor antigens to DC resulting in the activation CD8 T cell anti-tumor response [56]. Two studies demonstrated that TEV loaded with miRNA-155, miRNA 142 or let 7i by electroporation could promote the maturation of DC, causing an increase in T cell proliferation and cytotoxicity against tumor target cells in vitro [115,116]. Moreover, HCC-TEV improved DC therapeutic efficiency resulting in the elevation of CD8 T cell numbers, an increase in IFN-γ production and a decrease in the production of IL-10 and TGF β [117]. Furthermore, TEV-pulsed DC were shown to reduce numbers of regulatory T cells (Treg) and augment the therapeutic effect of antibodies against PD-1 and sorafenib in an HCC mouse model [118]. Therapeutic administration of TEV-loaded DC also showed a beneficial effect alone and in combination with MDSC depletion in pancreatic cancer [119].

7. EV in Crosstalk between Tumor Cells and Granulocytes

Granulocytes are myeloid cells, containing granules with defensive factors and enzymes, which play an important role in the innate immune response [120]. Granulocytes can be divided into three subsets: neutrophils, basophils and eosinophils. Neutrophils play a critical role in cancer [121]. On one hand, they show a strong cytotoxic activity and have the ability to recruit other immune cells due to chemokine release [122]. On the other hand, similar to monocytes and macrophages, neutrophils show a great plasticity and develop protumorigenic features under the exposure to TGF-β, G-CSF and IFN-β, promoting tumor angiogenesis and metastasis [121,123]. Furthermore, a high neutrophil-to-lymphocyte ratio was found to be associated with decreased overall survival (OS) of patients in different types of cancer [124]. The infiltration of the tumor with mature neutrophils was proposed to be a prognostic factor for tumor recurrence [125,126].
Eosinophils are also known to play a role in cancer development. An accumulation of eosinophils has been observed in the tumor tissue and peripheral blood of cancer patients [127]. Moreover, this was reported to be associated with a beneficial OS and progression-free survival (PFS) in melanoma patients [128,129].
During infection, neutrophils form neutrophil extracellular traps (NET) to eliminate pathogens. However, in cancer, NET formation could contribute to cancer-associated thrombosis, leading to thromboembolic complications [130,131]. Interestingly, breast cancer-derived EV were demonstrated to induce NET formation in neutrophils and enhance thrombosis when administrated in vivo [132].
TEV could also influence the phenotype of neutrophils. Thus, colorectal cancer stem cell (CRCSC)-derived EV increased the survival of neutrophils and endowed neutrophils with a protumor phenotype that was mediated by IL-1β [133]. In contrast, different populations of myeloid progenitors were not affected by CRCSC-derived EV in vivo, suggesting that this effect on neutrophils was not dependent on the proliferation of myeloid progenitors [133]. Furthermore, Cheng et al. [134] reported a correlation between the abundance of the major miRNA in CRCSC-derived EV, miRNA-146a, and increased numbers of tumor-infiltrating neutrophils.
Neutrophils could be polarized towards a protumorigenic phenotype upon the exposure to gastric cancer-derived EV [135]. High mobility group box (HMGB)1 was found to be a key factor transported by gastric cancer-derived EV that triggers TLR4 signaling followed by the activation of NF-κB and protumor activation of neutrophils [135].
Data on TEV impact on eosinophils and basophils are relatively scarce. It was found that EV derived from epithelial lung cancer cells contain γ-glutamyl transpeptidase 1 and contributed to the conversion of leukotriene C4 produced by eosinophils into leukotriene D4, which is known to mediate a number of protumor effects [136].

8. EV in Crosstalk between Tumor Cells and MDSC

MDSC represent a heterogeneous population of myeloid cells generated under chronic inflammatory conditions or cancer and mediate a profound immunosuppression in the tumor microenvironment [32]. In mice, two MDSC subsets have been described: CD11b+Ly6G-Ly6Chigh monocytic (M-MDSC) and CD11b+Ly6G+Ly6Clow polymorphonuclear (PMN-MDSC). Human MDSC could be divided into three subpopulations: CD11b+CD14+HLA-DRlow/-CD15- M-MDSC, CD11b+CD14-HLA-DRlow/-CD15+ PMN-MDSC and CD11b+CD14-HLA-DRlow/-CD15- early (e)MDSC [137]. MDSC are known to strongly inhibit the anti-tumor activity of T and NK cells through different mechanisms. They express PD-L1 and Fas antigen ligand, inducing T cell anergy and apoptosis, increase the production of immunosuppressive adenosine, ROS, NO and expression of Arg-1, leading to the down-regulation of T cell receptor ζ-chain expression as well as stimulate Treg generation [138,139,140]. In melanoma patients, the high frequency of MDSC has been shown to correlate with an advanced stage, disease progression, decreased OS and PFS and with low response to immunotherapy [141,142].
Several studies found that HSP triggered TLR signaling, leading to the increase of MDSC immunosuppressive potential. Diao et al. [143] have demonstrated that renal cell carcinoma cells produce TEV containing HSP70 that stimulate the TLR2/Myeloid differentiation primary response protein MyD88 (MyD88)/STAT3 signaling in MDSC, leading to tumor progression. HSP70 has been shown to be expressed on TEV isolated from patients with breast, pulmonary or ovary cancer to a higher extent than in EV derived from healthy donors [144]. In line with these data, renal cancer TEV showed abundant expression of HSP70. Therefore, blocking EV-HSP70 with A8 peptide prevented its interaction with TLR2 and blocked EV-mediated activation of MDSC. Moreover, A8 treatment potentiated 5-FU therapy since the latter enhanced the HSP70 expression on EV [144]. Triggering of the TLR2/MyD88 signaling pathway by TEV derived from thymoma, mammary carcinoma or colon carcinoma cells and containing HSP72, has been shown to induce IL-6 production by MDSC and autocrine activation of STAT3 [145]. Another study reported that TEV induced IL-6 release from MDSC and postulated that the role of TLR2 signaling could be dependent on the type of TEV-producing cancer cells [146]. Comparing the effects of EV derived from melanoma cells in vitro and in vivo, they found that only EV isolated from in vitro cultured tumor cells exerted a TLR2-dependent effect on MDSC in their system. Furthermore, it has been demonstrated that the inactivation of prostaglandin E2 and TGF-β blocks MDSC induction via TEV derived from mammary tumor cells in vivo [147]. The authors suggested that in vivo MDSC generation was dependent on the delivery of prostaglandin E2 and TGF-β by TEV. It was earlier shown that the expression of receptors for prostaglandin E2 and the activation of MDSC was supported by such interactions [148].
TEV have been shown to transfer functional RNA from cancer cells to MDSC [149]. In oral squamous cell carcinoma (OSCC), hypoxic TEV enhanced the suppressive effect of MDSC on γδ T cells through a miRNA-21/Phosphatase and tensin homolog (PTEN)/PD-L1 axis [150]. This study further showed that targeting miRNA-21 in combination with anti-PD-1 therapy had an anti-tumor effect on OSCC-bearing mice. Hypoxia-induced glioma-derived EV have been demonstrated to induce MDSC via miRNA-10a and miRNA-21 since MDSC from TEV-treated mice showed an enhanced suppressive effect on CD8 T cells that was attenuated by inhibiting miRNA-10a or miRNA-21 [151]. A similar effect has been shown to be mediated by the transfer of miRNA-29a and miRNA-92a in hypoxia-induced glioma TEV [152]. Interestingly, these miRNA promoted proliferation of MDSC, while miRNA-92a also enhanced their immunosuppressive function. Bruns et al. [153] stated that a transfer of miRNA-155 by CLL-derived EV contributed to MDSC induction, which was prevented by vitamin D, repressing the expression of miRNA-155 in CLL cells. Gastric cancer-derived EV were demonstrated to deliver miRNA-107 to MDSC, leading to their expansion and an upregulation of Arg-1 expression [154].
Not only cancer cells themselves but also tumor-educated mesenchymal stem cells (MSC) could modulate MDSC promoting breast cancer progression [155]. MSC-derived EV, containing TGF-β, complement component C1q and semaphorins could enhance immunosuppressive capacity of M-MDSC via PD-L1 upregulation and drive their differentiation towards M2-macrophages, showing an increased Arg-1 activity and IL-10 secretion [155]. Therefore, EV derived from cancer and stroma cells could support tumor development through the activation of MDSC.

9. Conclusions

TEV are transferred to myeloid cells and modulate their differentiation and phenotype to induce the formation of pre-metastatic niches at distant sites further supporting tumor progression (Figure 2). Such TEV capacity is attributed to their cargo molecules (oncogenes, various HSP, numerous miRNA etc.), which could trigger surface signaling or exert intracellular effects after being internalized (Table 1). Sorting of these cargo molecules into TEV is regulated by tumor-associated hypoxia [41,42,43,44,95,97,150,151,152] and cytotoxic chemotherapy [45]. Exposure of monocytes to TEV induced their conversion into immunosuppressive MDSC [102], whereas DC could acquire immunostimulatory capacity under the influence of TEV containing tumor-associated antigens [56,115,116,117,118,119], leading to anti-tumor effects. Therefore, the state of cells producing TEV, cargo of TEV and state of recipient cells are the factors, determining the outcome of TEV-mediated crosstalk between tumors and myeloid cells.
Numerous studies implicate EV as a delivery system for anti-cancer drugs or vaccine vectors to improve an anti-cancer immunity [156,157]. Other therapeutic strategies aim at disrupting EV-mediated protumorigenic effects by interfering with the secretion, uptake and signaling of EV [40]. Among them are anti-CD9 and anti-CD63 antibodies, which induced macrophage-dependent depletion of circulating EV [158]. This approach showed a therapeutic benefit in mice with breast tumors based on the prevention of metastasis without affecting primary tumor growth [158]. In addition, inhibiting TEV-associated miRNA with anti-miRNA oligonucleotides was demonstrated to reduce the growth of human oral squamous cell carcinoma cells in nude mice [159]. Therefore, a combination of blocking immunoinhibitory effects of TEV and EV application as an immunostimulatory vector could become an effective anti-cancer therapy.

Funding

The study was funded by the Deutsche Forschungsgemeinschaft (DFG; German Research Foundation; project number 25933240/RTG 2099 to J. Utikal and V. Umansky), the Cooperation Program in Cancer Research of the Deutsches Krebsforschungszentrum (DKFZ) and the Israeli Ministry of Science and Technology (MOST, CA181 to V. Umansky) and the German Federal Ministry of Education and Research (SERPENTINE project in the ERA PerMed network to V. Umansky).

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

Arg-1Arginase-1
CCLChemokine (C-C motif) ligand
CDCluster of differentiation
CLLChronic lymphocytic leukemia
CSPGChondroitin sulfate proteoglycan
CRCSCColorectal cancer stem cell
DCDendritic cells
EMTEpithelial to mesenchymal transition
Erk Extracellular signal-regulated kinase
EVExtracellular vesicles
G-CSFGranulocyte colony-stimulating factor
GMPGranulocyte-macrophage progenitors
HCCHepatocellular carcinoma
HLAHuman leukocyte antigen
HMGB1High mobility group box 1
HSCHematopoietic stem cells
HSPHeat-shock protein
IFNInterferon
ILInterleukin
LPSLipopolysaccharides
M-CSFMacrophage colony-stimulating factor
MDSCMyeloid-derived suppressor cells
MHCMajor histocompatibility complex
miRNAMicroRNA
M-MDSCMonocytic MDSC
MSCMesenchymal stem cells
MyD88Myeloid differentiation primary response protein MyD88
NETNeutrophil extracellular traps
NFNuclear factor
NKNatural Killer
NONitric oxide
OSOverall survival
OSCCOral squamous cell carcinoma
PDProgrammed cell death protein
PD-LProgrammed death ligand
PFSProgression-free survival
PI3KPhosphatidylinositol 3-kinase
PMN-MDSCPolymorphonuclear MDSC
PSMAProstate-specific membrane antigen
PTENPhosphatase and tensin homolog
ROSReactive oxygen species
SALL Sal-like protein
SERSSurface-enhanced Raman spectroscopy
SOCS Suppressor of cytokine signaling
STATSignal transducer and activator of transcription
TAMTumor-associated macrophages
TIE Tyrosine kinase with immunoglobulin-like and EGF-like domains
TEVTumor-derived extracellular vesicles
TGFTransforming growth factor
TLRToll-like receptor
TNFTumor necrosis factor
TregRegulatory T cells
VEGFVascular endothelial growth factor

References

  1. 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]
  2. Jeppesen, D.K.; Fenix, A.M.; Franklin, J.L.; Higginbotham, J.N.; Zhang, Q.; Zimmerman, L.J.; Liebler, D.C.; Ping, J.; Liu, Q.; Evans, R.; et al. Reassessment of Exosome Composition. Cell 2019, 177, 428–445.e18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Hessvik, N.P.; Llorente, A. Current knowledge on exosome biogenesis and release. Cell. Mol. Life Sci. 2018, 75, 193–208. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Théry, C.; Witwer, K.W.; Aikawa, E.; Alcaraz, M.J.; Anderson, J.D.; Andriantsitohaina, R.; Antoniou, A.; Arab, T.; Archer, F.; Atkin-Smith, G.K.; et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): A position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J. Extracell. Vesicles 2018, 7, 1535750. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Mathieu, M.; Martin-Jaular, L.; Lavieu, G.; Théry, C. Specificities of secretion and uptake of exosomes and other extracellular vesicles for cell-to-cell communication. Nat. Cell Biol. 2019, 21, 9–17. [Google Scholar] [CrossRef] [PubMed]
  6. Gonda, A.; Kabagwira, J.; Senthil, G.N.; Wall, N.R. Internalization of Exosomes through Receptor-Mediated Endocytosis. Mol. Cancer Res. 2019, 17, 337–347. [Google Scholar] [CrossRef] [Green Version]
  7. Skotland, T.; Hessvik, N.P.; Sandvig, K.; Llorente, A. Exosomal lipid composition and the role of ether lipids and phosphoinositides in exosome biology. J. Lipid Res. 2019, 60, 9–18. [Google Scholar] [CrossRef] [Green Version]
  8. Buzás, E.I.; Tóth, E.Á.; Sódar, B.W.; Szabó-Taylor, K.É. Molecular interactions at the surface of extracellular vesicles. Semin. Immunopathol. 2018, 40, 453–464. [Google Scholar] [CrossRef] [Green Version]
  9. Hida, K.; Zheng, L.; Wauben, M.; Ikuo, M.; Shen, L.; Gho, Y.S.; Takakura, Y.; Thery, C. ISEV2019 Abstract Book. J. Extracell. Vesicles 2019, 8, 1593587. [Google Scholar]
  10. Elsherbini, A.; Bieberich, E. Ceramide and Exosomes: A Novel Target in Cancer Biology and Therapy. In Advances in Cancer Research; Academic Press Inc.: Cambridge, MA, USA, 2018; Volume 140, pp. 121–154. [Google Scholar]
  11. Record, M.; Carayon, K.; Poirot, M.; Silvente-Poirot, S. Exosomes as new vesicular lipid transporters involved in cell–cell communication and various pathophysiologies. Biochim. Biophys. Acta Mol. Cell Biol. Lipids 2014, 1841, 108–120. [Google Scholar] [CrossRef]
  12. Fan, M.; Krutilina, R.; Sun, J.; Sethuraman, A.; Yang, C.H.; Wu, Z.H.; Yue, J.; Pfeffer, L.M. Comprehensive analysis of MicroRNA (miRNA) targets in breast cancer cells. J. Biol. Chem. 2013, 288, 27480–27493. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Collino, F.; Bruno, S.; Incarnato, D.; Dettori, D.; Neri, F.; Provero, P.; Pomatto, M.; Oliviero, S.; Tetta, C.; Quesenberry, P.J.; et al. AKI recovery induced by mesenchymal stromal cell-derived extracellular vesicles carrying micrornas. J. Am. Soc. Nephrol. 2015, 26, 2349–2360. [Google Scholar] [CrossRef] [PubMed]
  14. Ying, W.; Riopel, M.; Bandyopadhyay, G.; Dong, Y.; Birmingham, A.; Seo, J.B.; Ofrecio, J.M.; Wollam, J.; Hernandez-Carretero, A.; Fu, W.; et al. Adipose Tissue Macrophage-Derived Exosomal miRNAs Can Modulate In Vivo and In Vitro Insulin Sensitivity. Cell 2017, 171, 372–384. [Google Scholar] [CrossRef] [PubMed]
  15. Schwarzenbach, H.; Gahan, P. MicroRNA Shuttle from Cell-To-Cell by Exosomes and Its Impact in Cancer. Non Coding RNA 2019, 5, 28. [Google Scholar] [CrossRef] [Green Version]
  16. Valadi, H.; Ekström, K.; Bossios, A.; Sjöstrand, M.; Lee, J.J.; Lötvall, J.O. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat. Cell Biol. 2007, 9, 654–659. [Google Scholar] [CrossRef] [Green Version]
  17. Huang, X.; Yuan, T.; Tschannen, M.; Sun, Z.; Jacob, H.; Du, M.; Liang, M.; Dittmar, R.L.; Liu, Y.; Liang, M.; et al. Characterization of human plasma-derived exosomal RNAs by deep sequencing. BMC Genom. 2013, 14, 319. [Google Scholar] [CrossRef] [Green Version]
  18. Guduric-Fuchs, J.; O’Connor, A.; Camp, B.; O’Neill, C.L.; Medina, R.J.; Simpson, D.A. Selective extracellular vesicle-mediated export of an overlapping set of microRNAs from multiple cell types. BMC Genom. 2012, 13, 357. [Google Scholar] [CrossRef] [Green Version]
  19. Villarroya-Beltri, C.; Gutiérrez-Vázquez, C.; Sánchez-Cabo, F.; Pérez-Hernández, D.; Vázquez, J.; Martin-Cofreces, N.; Martinez-Herrera, D.J.; Pascual-Montano, A.; Mittelbrunn, M.; Sánchez-Madrid, F. Sumoylated hnRNPA2B1 controls the sorting of miRNAs into exosomes through binding to specific motifs. Nat. Commun. 2013, 4, 2980. [Google Scholar] [CrossRef] [Green Version]
  20. Temoche-Diaz, M.M.; Shurtleff, M.J.; Nottingham, R.M.; Yao, J.; Fadadu, R.P.; Lambowitz, A.M.; Schekman, R. Distinct mechanisms of microRNA sorting into cancer cell-derived extracellular vesicle subtypes. Elife 2019, 8, e47544. [Google Scholar] [CrossRef]
  21. Shurtleff, M.J.; Temoche-Diaz, M.M.; Karfilis, K.V.; Ri, S.; Schekman, R. Y-box protein 1 is required to sort microRNAs into exosomes in cells and in a cell-free reaction. Elife 2016, 5, e19276. [Google Scholar] [CrossRef]
  22. Kossinova, O.A.; Gopanenko, A.V.; Tamkovich, S.N.; Krasheninina, O.A.; Tupikin, A.E.; Kiseleva, E.; Yanshina, D.D.; Malygin, A.A.; Veńyaminova, A.G.; Kabilov, M.R.; et al. Cytosolic YB-1 and NSUN2 are the only proteins recognizing specific motifs present in mRNAs enriched in exosomes. Biochim. Biophys. Acta Proteins Proteom. 2017, 1865, 664–673. [Google Scholar] [CrossRef] [PubMed]
  23. Statello, L.; Maugeri, M.; Garre, E.; Nawaz, M.; Wahlgren, J.; Papadimitriou, A.; Lundqvist, C.; Lindfors, L.; Collén, A.; Sunnerhagen, P.; et al. Identification of RNA-binding proteins in exosomes capable of interacting with different types of RNA: RBP-facilitated transport of RNAs into exosomes. PLoS ONE 2018, 13, e0195969. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Janas, T.; Janas, M.M.; Sapoń, K.; Janas, T. Mechanisms of RNA loading into exosomes. FEBS Lett. 2015, 589, 1391–1398. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Koppers-Lalic, D.; Hackenberg, M.; Bijnsdorp, I.V.; van Eijndhoven, M.A.J.; Sadek, P.; Sie, D.; Zini, N.; Middeldorp, J.M.; Ylstra, B.; de Menezes, R.X.; et al. Nontemplated Nucleotide Additions Distinguish the Small RNA Composition in Cells from Exosomes. Cell Rep. 2014, 8, 1649–1658. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Bassler, K.; Schulte-Schrepping, J.; Warnat-Herresthal, S.; Aschenbrenner, A.C.; Schultze, J.L. The Myeloid Cell Compartment—Cell by Cell. Annu. Rev. Immunol. 2019, 37, 269–293. [Google Scholar] [CrossRef] [PubMed]
  27. Weiskopf, K.; Schnorr, P.J.; Pang, W.W.; Chao, M.P.; Chhabra, A.; Seita, J.; Feng, M.; Weissman, I.L. Myeloid Cell Origins, Differentiation, and Clinical Implications. Microbiol. Spectr. 2016, 4, 857–875. [Google Scholar]
  28. Coussens, L.M.; Pollard, J.W. Leukocytes in Mammary Development and Cancer. Cold Spring Harb. Perspect. Biol. 2011, 3, a003285. [Google Scholar] [CrossRef] [Green Version]
  29. Ugel, S.; De Sanctis, F.; Mandruzzato, S.; Bronte, V. Tumor-induced myeloid deviation: When myeloid-derived suppressor cells meet tumor-associated macrophages. J. Clin. Investig. 2015, 125, 3365–3376. [Google Scholar] [CrossRef] [Green Version]
  30. Marvel, D.; Gabrilovich, D.I. Myeloid-derived suppressor cells in the tumor microenvironment: Expect the unexpected. J. Clin. Investig. 2015, 125, 3356–3364. [Google Scholar] [CrossRef]
  31. Landskron, G.; De la Fuente, M.; Thuwajit, P.; Thuwajit, C.; Hermoso, M.A. Chronic Inflammation and Cytokines in the Tumor Microenvironment. J. Immunol. Res. 2014, 2014, 1–19. [Google Scholar] [CrossRef] [Green Version]
  32. Veglia, F.; Perego, M.; Gabrilovich, D. Myeloid-derived suppressor cells coming of age. Nat. Immunol. 2018, 19, 108–119. [Google Scholar] [CrossRef] [PubMed]
  33. Franklin, R.A.; Liao, W.; Sarkar, A.; Kim, M.V.; Bivona, M.R.; Liu, K.; Pamer, E.G.; Li, M.O. The cellular and molecular origin of tumor-associated macrophages. Science 2014, 344, 921–925. [Google Scholar] [CrossRef] [Green Version]
  34. Latifkar, A.; Hur, Y.H.; Sanchez, J.C.; Cerione, R.A.; Antonyak, M.A. New insights into extracellular vesicle biogenesis and function. J. Cell Sci. 2019, 132, jcs222406. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Hyenne, V.; Ghoroghi, S.; Collot, M.; Bons, J.; Follain, G.; Harlepp, S.; Mary, B.; Bauer, J.; Mercier, L.; Busnelli, I.; et al. Studying the Fate of Tumor Extracellular Vesicles at High Spatiotemporal Resolution Using the Zebrafish Embryo. Dev. Cell 2019, 48, 554–572.e7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Verweij, F.J.; Revenu, C.; Arras, G.; Dingli, F.; Loew, D.; Pegtel, D.M.; Follain, G.; Allio, G.; Goetz, J.G.; Zimmermann, P.; et al. Live Tracking of Inter-organ Communication by Endogenous Exosomes In Vivo. Dev. Cell 2019, 48, 573–589.e4. [Google Scholar] [CrossRef] [Green Version]
  37. Imai, T.; Takahashi, Y.; Nishikawa, M.; Kato, K.; Morishita, M.; Yamashita, T.; Matsumoto, A.; Charoenviriyakul, C.; Takakura, Y. Macrophage-dependent clearance of systemically administered B16BL6-derived exosomes from the blood circulation in mice. J. Extracell. Vesicles 2015, 4, 26238. [Google Scholar] [CrossRef]
  38. Czernek, L.; Chworos, A.; Duechler, M. The Uptake of Extracellular Vesicles Is Affected by the Differentiation Status of Myeloid Cells. Scand. J. Immunol. 2015, 82, 506–514. [Google Scholar] [CrossRef] [Green Version]
  39. Hoshino, A.; Costa-Silva, B.; Shen, T.-L.; Rodrigues, G.; Hashimoto, A.; Tesic Mark, M.; Molina, H.; Kohsaka, S.; Di Giannatale, A.; Ceder, S.; et al. Tumour exosome integrins determine organotropic metastasis. Nature 2015, 527, 329–335. [Google Scholar] [CrossRef] [Green Version]
  40. Whiteside, T.L. Tumor-Derived Exosomes and Their Role in Cancer Progression. In Advances in Clinical Chemistry; Academic Press Inc.: Cambridge, MA, USA, 2016; Volume 74, pp. 103–141. [Google Scholar]
  41. Shao, C.; Yang, F.; Miao, S.; Liu, W.; Wang, C.; Shu, Y.; Shen, H. Role of hypoxia-induced exosomes in tumor biology. Mol. Cancer 2018, 17, 120. [Google Scholar] [CrossRef]
  42. Kumar, A.; Deep, G. Hypoxia in tumor microenvironment regulates exosome biogenesis: Molecular mechanisms and translational opportunities. Cancer Lett. 2020, 479, 23–30. [Google Scholar] [CrossRef]
  43. Ramteke, A.; Ting, H.; Agarwal, C.; Mateen, S.; Somasagara, R.; Hussain, A.; Graner, M.; Frederick, B.; Agarwal, R.; Deep, G. Exosomes secreted under hypoxia enhance invasiveness and stemness of prostate cancer cells by targeting adherens junction molecules. Mol. Carcinog. 2015, 54, 554–565. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Boussadia, Z.; Lamberti, J.; Mattei, F.; Pizzi, E.; Puglisi, R.; Zanetti, C.; Pasquini, L.; Fratini, F.; Fantozzi, L.; Felicetti, F.; et al. Acidic microenvironment plays a key role in human melanoma progression through a sustained exosome mediated transfer of clinically relevant metastatic molecules. J. Exp. Clin. Cancer Res. 2018, 37, 245. [Google Scholar] [CrossRef] [PubMed]
  45. Keklikoglou, I.; Cianciaruso, C.; Güç, E.; Squadrito, M.L.; Spring, L.M.; Tazzyman, S.; Lambein, L.; Poissonnier, A.; Ferraro, G.B.; Baer, C.; et al. Chemotherapy elicits pro-metastatic extracellular vesicles in breast cancer models. Nat. Cell Biol. 2019, 21, 190–202. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Nazarenko, I. Extracellular Vesicles: Recent Developments in Technology and Perspectives for Cancer Liquid Biopsy. In Recent Results in Cancer Research; Springer: New York, NY, USA, 2020; Volume 215, pp. 319–344. [Google Scholar]
  47. Sharma, P.; Ludwig, S.; Muller, L.; Hong, C.S.; Kirkwood, J.M.; Ferrone, S.; Whiteside, T.L. Immunoaffinity-based isolation of melanoma cell-derived exosomes from plasma of patients with melanoma. J. Extracell. Vesicles 2018, 7, 1435138. [Google Scholar] [CrossRef]
  48. Kim, D.; Woo, H.-K.; Lee, C.; Min, Y.; Kumar, S.; Sunkara, V.; Jo, H.-G.; Lee, Y.J.; Kim, J.; Ha, H.K.; et al. EV-Ident: Identifying Tumor-Specific Extracellular Vesicles by Size Fractionation and Single-Vesicle Analysis. Anal. Chem. 2020, 92, 6010–6018. [Google Scholar] [CrossRef]
  49. Reátegui, E.; van der Vos, K.E.; Lai, C.P.; Zeinali, M.; Atai, N.A.; Aldikacti, B.; Floyd, F.P.; Khankhel, A.H.; Thapar, V.; Hochberg, F.H.; et al. Engineered nanointerfaces for microfluidic isolation and molecular profiling of tumor-specific extracellular vesicles. Nat. Commun. 2018, 9, 175. [Google Scholar] [CrossRef]
  50. Padda, R.S.; Deng, F.K.; Brett, S.I.; Biggs, C.N.; Durfee, P.N.; Brinker, C.J.; Williams, K.C.; Leong, H.S. Nanoscale flow cytometry to distinguish subpopulations of prostate extracellular vesicles in patient plasma. Prostate 2019, 79, 592–603. [Google Scholar] [CrossRef]
  51. Zhang, W.; Jiang, L.; Diefenbach, R.J.; Campbell, D.H.; Walsh, B.J.; Packer, N.H.; Wang, Y. Enabling Sensitive Phenotypic Profiling of Cancer-Derived Small Extracellular Vesicles Using Surface-Enhanced Raman Spectroscopy Nanotags. ACS Sens. 2020, 5, 764–771. [Google Scholar] [CrossRef]
  52. Chanteloup, G.; Cordonnier, M.; Isambert, N.; Bertaut, A.; Marcion, G.; Garrido, C.; Gobbo, J. Membrane-bound exosomal HSP70 as a biomarker for detection and monitoring of malignant solid tumours: A pilot study. Pilot Feasibility Stud. 2020, 6, 35. [Google Scholar] [CrossRef] [Green Version]
  53. Whiteside, T.L. Exosomes carrying immunoinhibitory proteins and their role in cancer. Clin. Exp. Immunol. 2017, 189, 259–267. [Google Scholar] [CrossRef] [Green Version]
  54. Sharma, P.; Diergaarde, B.; Ferrone, S.; Kirkwood, J.M.; Whiteside, T.L. Melanoma cell-derived exosomes in plasma of melanoma patients suppress functions of immune effector cells. Sci. Rep. 2020, 10, 92. [Google Scholar] [CrossRef] [PubMed]
  55. Altevogt, P.; Bretz, N.P.; Ridinger, J.; Utikal, J.; Umansky, V. Novel insights into exosome-induced, tumor-associated inflammation and immunomodulation. Semin. Cancer Biol. 2014, 28, 51–57. [Google Scholar] [CrossRef] [PubMed]
  56. Wolfers, J.; Lozier, A.; Raposo, G.; Regnault, A.; Théry, C.; Masurier, C.; Flament, C.; Pouzieux, S.; Faure, F.; Tursz, T.; et al. Tumor-derived exosomes are a source of shared tumor rejection antigens for CTL cross-priming. Nat. Med. 2001, 7, 297–303. [Google Scholar] [CrossRef] [PubMed]
  57. Laurenti, E.; Göttgens, B. From haematopoietic stem cells to complex differentiation landscapes. Nature 2018, 553, 418–426. [Google Scholar] [CrossRef] [PubMed]
  58. Young, K.; Borikar, S.; Bell, R.; Kuffler, L.; Philip, V.; Trowbridge, J.J. Progressive alterations in multipotent hematopoietic progenitors underlie lymphoid cell loss in aging. J. Exp. Med. 2016, 213, 2259–2267. [Google Scholar] [CrossRef]
  59. Gabrilovich, D.I.; Ostrand-Rosenberg, S.; Bronte, V. Coordinated regulation of myeloid cells by tumours. Nat. Rev. Immunol. 2012, 12, 253–268. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Pyzer, A.R.; Stroopinsky, D.; Rajabi, H.; Washington, A.; Tagde, A.; Coll, M.; Fung, J.; Bryant, M.P.; Cole, L.; Palmer, K.; et al. MUC1-mediated induction of myeloid-derived suppressor cells in patients with acute myeloid leukemia. Blood 2017, 129, 1791–1801. [Google Scholar] [CrossRef] [Green Version]
  61. Ning, Y.; Shen, K.; Wu, Q.; Sun, X.; Bai, Y.; Xie, Y.; Pan, J.; Qi, C. Tumor exosomes block dendritic cells maturation to decrease the T cell immune response. Immunol. Lett. 2018, 199, 36–43. [Google Scholar] [CrossRef]
  62. Yu, S.; Liu, C.; Su, K.; Wang, J.; Liu, Y.; Zhang, L.; Li, C.; Cong, Y.; Kimberly, R.; Grizzle, W.E.; et al. Tumor Exosomes Inhibit Differentiation of Bone Marrow Dendritic Cells. J. Immunol. 2007, 178, 6867–6875. [Google Scholar] [CrossRef] [Green Version]
  63. Peinado, H.; Alečković, M.; Lavotshkin, S.; Matei, I.; Costa-Silva, B.; Moreno-Bueno, G.; Hergueta-Redondo, M.; Williams, C.; García-Santos, G.; Ghajar, C.M.; 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] [Green Version]
  64. Fleming, V.; Hu, X.; Weller, C.; Weber, R.; Groth, C.; Riester, Z.; Hüser, L.; Sun, Q.; Nagibin, V.; Kirschning, C.; et al. Melanoma Extracellular Vesicles Generate Immunosuppressive Myeloid Cells by Upregulating PD-L1 via TLR4 Signaling. Cancer Res. 2019, 79, 4715–4728. [Google Scholar] [CrossRef] [PubMed]
  65. Topalian, S.L.; Drake, C.G.; Pardoll, D.M. Immune checkpoint blockade: A common denominator approach to cancer therapy. Cancer Cell 2015, 27, 450–461. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Shi, C.; Pamer, E.G. Monocyte recruitment during infection and inflammation. Nat. Rev. Immunol. 2011, 11, 762–774. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Engblom, C.; Pfirschke, C.; Pittet, M.J. The role of myeloid cells in cancer therapies. Nat. Rev. Cancer 2016, 16, 447–462. [Google Scholar] [CrossRef]
  68. Valenti, R.; Huber, V.; Filipazzi, P.; Pilla, L.; Sovena, G.; Villa, A.; Corbelli, A.; Fais, S.; Parmiani, G.; Rivoltini, L. Human Tumor-Released Microvesicles Promote the Differentiation of Myeloid Cells with Transforming Growth Factor-β–Mediated Suppressive Activity on T Lymphocytes. Cancer Res. 2006, 66, 9290–9298. [Google Scholar] [CrossRef] [Green Version]
  69. Huber, V.; Vallacchi, V.; Fleming, V.; Hu, X.; Cova, A.; Dugo, M.; Shahaj, E.; Sulsenti, R.; Vergani, E.; Filipazzi, P.; et al. Tumor-derived microRNAs induce myeloid suppressor cells and predict immunotherapy resistance in melanoma. J. Clin. Investig. 2018, 128, 5505–5516. [Google Scholar] [CrossRef] [Green Version]
  70. Haderk, F.; Schulz, R.; Iskar, M.; Cid, L.L.; Worst, T.; Willmund, K.V.; Schulz, A.; Warnken, U.; Seiler, J.; Benner, A.; et al. Tumor-derived exosomes modulate PD-L1 expression in monocytes. Sci. Immunol. 2017, 2, eaah5509. [Google Scholar] [CrossRef] [Green Version]
  71. Bretz, N.P.; Ridinger, J.; Rupp, A.-K.; Rimbach, K.; Keller, S.; Rupp, C.; Marmé, F.; Umansky, L.; Umansky, V.; Eigenbrod, T.; et al. Body Fluid Exosomes Promote Secretion of Inflammatory Cytokines in Monocytic Cells via Toll-like Receptor Signaling. J. Biol. Chem. 2013, 288, 36691–36702. [Google Scholar] [CrossRef] [Green Version]
  72. Lenart, M.; Rutkowska-Zapala, M.; Baj-Krzyworzeka, M.; Szatanek, R.; Węglarczyk, K.; Smallie, T.; Ziegler-Heitbrock, L.; Zembala, M.; Siedlar, M. Hyaluronan carried by tumor-derived microvesicles induces IL-10 production in classical (CD14++ CD16−) monocytes via PI3K/Akt/mTOR-dependent signalling pathway. Immunobiology 2017, 222, 1–10. [Google Scholar] [CrossRef]
  73. Hsieh, C.-H.; Tai, S.-K.; Yang, M.-H. Snail-overexpressing Cancer Cells Promote M2-Like Polarization of Tumor-Associated Macrophages by Delivering MiR-21-Abundant Exosomes. Neoplasia 2018, 20, 775–788. [Google Scholar] [CrossRef]
  74. Baj-Krzyworzeka, M.; Mytar, B.; Szatanek, R.; Surmiak, M.; Węglarczyk, K.; Baran, J.; Siedlar, M. Colorectal cancer-derived microvesicles modulate differentiation of human monocytes to macrophages. J. Transl. Med. 2016, 14, 36. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Gabrusiewicz, K.; Li, X.; Wei, J.; Hashimoto, Y.; Marisetty, A.L.; Ott, M.; Wang, F.; Hawke, D.; Yu, J.; Healy, L.M.; et al. Glioblastoma stem cell-derived exosomes induce M2 macrophages and PD-L1 expression on human monocytes. Oncoimmunology 2018, 7, e1412909. [Google Scholar] [CrossRef] [PubMed]
  76. Iorgulescu, J.B.; Ivan, M.E.; Safaee, M.; Parsa, A.T. The limited capacity of malignant glioma-derived exosomes to suppress peripheral immune effectors. J. Neuroimmunol. 2016, 290, 103–108. [Google Scholar] [CrossRef] [PubMed]
  77. Javeed, N.; Gustafson, M.P.; Dutta, S.K.; Lin, Y.; Bamlet, W.R.; Oberg, A.L.; Petersen, G.M.; Chari, S.T.; Dietz, A.B.; Mukhopadhyay, D. Immunosuppressive CD14 + HLA-DR lo/neg monocytes are elevated in pancreatic cancer and “primed” by tumor-derived exosomes. Oncoimmunology 2017, 6, e1252013. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Gärtner, K.; Battke, C.; Dünzkofer, J.; Hüls, C.; von Neubeck, B.; Kellner, M.; Fiestas, E.; Fackler, S.; Lang, S.; Zeidler, R. Tumor-derived extracellular vesicles activate primary monocytes. Cancer Med. 2018, 7, 2013–2020. [Google Scholar] [CrossRef]
  79. Wang, F.; Li, B.; Wei, Y.; Zhao, Y.; Wang, L.; Zhang, P.; Yang, J.; He, W.; Chen, H.; Jiao, Z.; et al. Tumor-derived exosomes induce PD1+ macrophage population in human gastric cancer that promotes disease progression. Oncogenesis 2018, 7, 41. [Google Scholar] [CrossRef] [Green Version]
  80. Plebanek, M.P.; Angeloni, N.L.; Vinokour, E.; Li, J.; Henkin, A.; Martinez-Marin, D.; Filleur, S.; Bhowmick, R.; Henkin, J.; Miller, S.D.; et al. Pre-metastatic cancer exosomes induce immune surveillance by patrolling monocytes at the metastatic niche. Nat. Commun. 2017, 8, 1319. [Google Scholar] [CrossRef]
  81. Mosser, D.M.; Edwards, J.P. Exploring the full spectrum of macrophage activation. Nat. Rev. Immunol. 2008, 8, 958–969. [Google Scholar] [CrossRef]
  82. Mantovani, A.; Sozzani, S.; Locati, M.; Allavena, P.; Sica, A. Macrophage polarization: Tumor-associated macrophages as a paradigm for polarized M2 mononuclear phagocytes. Trends Immunol. 2002, 23, 549–555. [Google Scholar] [CrossRef]
  83. Shapouri-Moghaddam, A.; Mohammadian, S.; Vazini, H.; Taghadosi, M.; Esmaeili, S.-A.; Mardani, F.; Seifi, B.; Mohammadi, A.; Afshari, J.T.; Sahebkar, A. Macrophage plasticity, polarization, and function in health and disease. J. Cell. Physiol. 2018, 233, 6425–6440. [Google Scholar] [CrossRef]
  84. Kuang, D.-M.; Zhao, Q.; Peng, C.; Xu, J.; Zhang, J.-P.; Wu, C.; Zheng, L. Activated monocytes in peritumoral stroma of hepatocellular carcinoma foster immune privilege and disease progression through PD-L1. J. Exp. Med. 2009, 206, 1327–1337. [Google Scholar] [CrossRef]
  85. Rodriguez, P.C.; Quiceno, D.G.; Zabaleta, J.; Ortiz, B.; Zea, A.H.; Piazuelo, M.B.; Delgado, A.; Correa, P.; Brayer, J.; Sotomayor, E.M.; et al. Arginase I Production in the Tumor Microenvironment by Mature Myeloid Cells Inhibits T-Cell Receptor Expression and Antigen-Specific T-Cell Responses. Cancer Res. 2004, 64, 5839–5849. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Mantovani, A.; Sica, A. Macrophages, innate immunity and cancer: Balance, tolerance, and diversity. Curr. Opin. Immunol. 2010, 22, 231–237. [Google Scholar] [CrossRef] [PubMed]
  87. Zheng, Y.; Cai, Z.; Wang, S.; Zhang, X.; Qian, J.; Hong, S.; Li, H.; Wang, M.; Yang, J.; Yi, Q. Macrophages are an abundant component of myeloma microenvironment and protect myeloma cells from chemotherapy drug–induced apoptosis. Blood 2009, 114, 3625–3628. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Qian, B.-Z.; Pollard, J.W. Macrophage Diversity Enhances Tumor Progression and Metastasis. Cell 2010, 141, 39–51. [Google Scholar] [CrossRef] [Green Version]
  89. Feng, W.; Dean, D.C.; Hornicek, F.J.; Shi, H.; Duan, Z. Exosomes promote pre-metastatic niche formation in ovarian cancer. Mol. Cancer 2019, 18, 124. [Google Scholar] [CrossRef] [Green Version]
  90. Costa-Silva, B.; Aiello, N.M.; Ocean, A.J.; Singh, S.; Zhang, H.; Thakur, B.K.; Becker, A.; Hoshino, A.; Mark, M.T.; Molina, H.; et al. Pancreatic cancer exosomes initiate pre-metastatic niche formation in the liver. Nat. Cell Biol. 2015, 17, 816–826. [Google Scholar] [CrossRef]
  91. Baig, M.S.; Roy, A.; Rajpoot, S.; Liu, D.; Savai, R.; Banerjee, S.; Kawada, M.; Faisal, S.M.; Saluja, R.; Saqib, U.; et al. Tumor-derived exosomes in the regulation of macrophage polarization. Inflamm. Res. 2020, 69, 435–451. [Google Scholar] [CrossRef]
  92. Bardi, G.T.; Smith, M.A.; Hood, J.L. Melanoma exosomes promote mixed M1 and M2 macrophage polarization. Cytokine 2018, 105, 63–72. [Google Scholar] [CrossRef]
  93. Popēna, I.; Ābols, A.; Saulīte, L.; Pleiko, K.; Zandberga, E.; Jēkabsons, K.; Endzeliņš, E.; Llorente, A.; Linē, A.; Riekstiņa, U. Effect of colorectal cancer-derived extracellular vesicles on the immunophenotype and cytokine secretion profile of monocytes and macrophages. Cell Commun. Signal. 2018, 16, 17. [Google Scholar] [CrossRef] [Green Version]
  94. Ham, S.; Lima, L.G.; Chai, E.P.Z.; Muller, A.; Lobb, R.J.; Krumeich, S.; Wen, S.W.; Wiegmans, A.P.; Möller, A. Breast Cancer-Derived Exosomes Alter Macrophage Polarization via gp130/STAT3 Signaling. Front. Immunol. 2018, 9, 871. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Park, J.E.; Dutta, B.; Tse, S.W.; Gupta, N.; Tan, C.F.; Low, J.K.; Yeoh, K.W.; Kon, O.L.; Tam, J.P.; Sze, S.K. Hypoxia-induced tumor exosomes promote M2-like macrophage polarization of infiltrating myeloid cells and microRNA-mediated metabolic shift. Oncogene 2019, 38, 5158–5173. [Google Scholar] [CrossRef]
  96. Cheng, L.; Liu, J.; Liu, Q.; Liu, Y.; Fan, L.; Wang, F.; Yu, H.; Li, Y.; Bu, L.; Li, X.; et al. Exosomes from Melatonin Treated Hepatocellularcarcinoma Cells Alter the Immunosupression Status through STAT3 Pathway in Macrophages. Int. J. Biol. Sci. 2017, 13, 723–734. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Qian, M.; Wang, S.; Guo, X.; Wang, J.; Zhang, Z.; Qiu, W.; Gao, X.; Chen, Z.; Xu, J.; Zhao, R.; et al. Hypoxic glioma-derived exosomes deliver microRNA-1246 to induce M2 macrophage polarization by targeting TERF2IP via the STAT3 and NF-κB pathways. Oncogene 2020, 39, 428–442. [Google Scholar] [CrossRef] [PubMed]
  98. Ying, X.; Wu, Q.; Wu, X.; Zhu, Q.; Wang, X.; Jiang, L.; Chen, X.; Wang, X. Epithelial ovarian cancer-secreted exosomal miR-222-3p induces polarization of tumor-associated macrophages. Oncotarget 2016, 7, 43076–43087. [Google Scholar] [CrossRef] [Green Version]
  99. Yin, C.; Han, Q.; Xu, D.; Zheng, B.; Zhao, X.; Zhang, J. SALL4-mediated upregulation of exosomal miR-146a-5p drives T-cell exhaustion by M2 tumor-associated macrophages in HCC. Oncoimmunology 2019, 8, e1601479. [Google Scholar] [CrossRef]
  100. Liu, Y.; Zhao, L.; Li, D.; Yin, Y.; Zhang, C.-Y.; Li, J.; Zhang, Y. Microvesicle-delivery miR-150 promotes tumorigenesis by up-regulating VEGF, and the neutralization of miR-150 attenuate tumor development. Protein Cell 2013, 4, 932–941. [Google Scholar] [CrossRef] [Green Version]
  101. Liu, J.; Fan, L.; Yu, H.; Zhang, J.; He, Y.; Feng, D.; Wang, F.; Li, X.; Liu, Q.; Li, Y.; et al. Endoplasmic Reticulum Stress Causes Liver Cancer Cells to Release Exosomal miR-23a-3p and Up-regulate Programmed Death Ligand 1 Expression in Macrophages. Hepatology 2019, 70, 241–258. [Google Scholar] [CrossRef]
  102. Jang, J.-Y.; Lee, J.-K.; Jeon, Y.-K.; Kim, C.-W. Exosome derived from epigallocatechin gallate treated breast cancer cells suppresses tumor growth by inhibiting tumor-associated macrophage infiltration and M2 polarization. BMC Cancer 2013, 13, 421. [Google Scholar] [CrossRef] [Green Version]
  103. Collin, M.; Bigley, V. Human dendritic cell subsets: An update. Immunology 2018, 154, 3–20. [Google Scholar] [CrossRef]
  104. Shortman, K.; Heath, W.R. The CD8 + dendritic cell subset. Immunol. Rev. 2010, 234, 18–31. [Google Scholar] [CrossRef]
  105. Tran Janco, J.M.; Lamichhane, P.; Karyampudi, L.; Knutson, K.L. Tumor-Infiltrating Dendritic Cells in Cancer Pathogenesis. J. Immunol. 2015, 194, 2985–2991. [Google Scholar] [CrossRef] [Green Version]
  106. Pinzon-Charry, A.; Maxwell, T.; López, J.A. Dendritic cell dysfunction in cancer: A mechanism for immunosuppression. Immunol. Cell Biol. 2005, 83, 451–461. [Google Scholar] [CrossRef] [PubMed]
  107. Gabrilovich, D. Mechanisms and functional significance of tumour-induced dendritic-cell defects. Nat. Rev. Immunol. 2004, 4, 941–952. [Google Scholar] [CrossRef] [PubMed]
  108. Hood, J.L. The association of exosomes with lymph nodes. Semin. Cell Dev. Biol. 2017, 67, 29–38. [Google Scholar] [CrossRef] [PubMed]
  109. Maus, R.L.G.; Jakub, J.W.; Nevala, W.K.; Christensen, T.A.; Noble-Orcutt, K.; Sachs, Z.; Hieken, T.J.; Markovic, S.N. Human Melanoma-Derived Extracellular Vesicles Regulate Dendritic Cell Maturation. Front. Immunol. 2017, 8, 358. [Google Scholar] [CrossRef] [Green Version]
  110. Maus, R.L.G.; Jakub, J.W.; Hieken, T.J.; Nevala, W.K.; Christensen, T.A.; Sutor, S.L.; Flotte, T.J.; Markovic, S.N. Identification of novel, immune-mediating extracellular vesicles in human lymphatic effluent draining primary cutaneous melanoma. Oncoimmunology 2019, 8, e1667742. [Google Scholar] [CrossRef] [Green Version]
  111. Yang, C.; Kim, S.-H.; Bianco, N.R.; Robbins, P.D. Tumor-Derived Exosomes Confer Antigen-Specific Immunosuppression in a Murine Delayed-Type Hypersensitivity Model. PLoS ONE 2011, 6, e22517. [Google Scholar] [CrossRef] [Green Version]
  112. Shen, Y.; Guo, D.; Weng, L.; Wang, S.; Ma, Z.; Yang, Y.; Wang, P.; Wang, J.; Cai, Z. Tumor-derived exosomes educate dendritic cells to promote tumor metastasis via HSP72/HSP105-TLR2/TLR4 pathway. Oncoimmunology 2017, 6, e1362527. [Google Scholar] [CrossRef]
  113. Ding, G.; Zhou, L.; Qian, Y.; Fu, M.; Chen, J.; Chen, J.; Xiang, J.; Wu, Z.; Jiang, G.; Cao, L. Pancreatic cancer-derived exosomes transfer miRNAs to dendritic cells and inhibit RFXAP expression via miR-212-3p. Oncotarget 2015, 6, 29877–29888. [Google Scholar] [CrossRef] [Green Version]
  114. Zhou, M.; Chen, J.; Zhou, L.; Chen, W.; Ding, G.; Cao, L. Pancreatic cancer derived exosomes regulate the expression of TLR4 in dendritic cells via miR-203. Cell. Immunol. 2014, 292, 65–69. [Google Scholar] [CrossRef]
  115. Taghikhani, A.; Hassan, Z.M.; Ebrahimi, M.; Moazzeni, S. microRNA modified tumor-derived exosomes as novel tools for maturation of dendritic cells. J. Cell. Physiol. 2019, 234, 9417–9427. [Google Scholar] [CrossRef]
  116. Asadirad, A.; Hashemi, S.M.; Baghaei, K.; Ghanbarian, H.; Mortaz, E.; Zali, M.R.; Amani, D. Phenotypical and functional evaluation of dendritic cells after exosomal delivery of miRNA-155. Life Sci. 2019, 219, 152–162. [Google Scholar] [CrossRef] [PubMed]
  117. Rao, Q.; Zuo, B.; Lu, Z.; Gao, X.; You, A.; Wu, C.; Du, Z.; Yin, H. Tumor-derived exosomes elicit tumor suppression in murine hepatocellular carcinoma models and humans in vitro. Hepatology 2016, 64, 456–472. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Shi, S.; Rao, Q.; Zhang, C.; Zhang, X.; Qin, Y.; Niu, Z. Dendritic Cells Pulsed with Exosomes in Combination with PD-1 Antibody Increase the Efficacy of Sorafenib in Hepatocellular Carcinoma Model. Transl. Oncol. 2018, 11, 250–258. [Google Scholar] [CrossRef] [PubMed]
  119. Xiao, L.; Erb, U.; Zhao, K.; Hackert, T.; Zöller, M. Efficacy of vaccination with tumor-exosome loaded dendritic cells combined with cytotoxic drug treatment in pancreatic cancer. Oncoimmunology 2017, 6, e1319044. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. Mayadas, T.N.; Cullere, X.; Lowell, C.A. The Multifaceted Functions of Neutrophils. Annu. Rev. Pathol. Mech. Dis. 2014, 9, 181–218. [Google Scholar] [CrossRef] [Green Version]
  121. Coffelt, S.B.; Wellenstein, M.D.; de Visser, K.E. Neutrophils in cancer: Neutral no more. Nat. Rev. Cancer 2016, 16, 431–446. [Google Scholar] [CrossRef] [Green Version]
  122. Mantovani, A.; Cassatella, M.A.; Costantini, C.; Jaillon, S. Neutrophils in the activation and regulation of innate and adaptive immunity. Nat. Rev. Immunol. 2011, 11, 519–531. [Google Scholar] [CrossRef]
  123. Moses, K.; Brandau, S. Human neutrophils: Their role in cancer and relation to myeloid-derived suppressor cells. Semin. Immunol. 2016, 28, 187–196. [Google Scholar] [CrossRef]
  124. Templeton, A.J.; McNamara, M.G.; Šeruga, B.; Vera-Badillo, F.E.; Aneja, P.; Ocaña, A.; Leibowitz-Amit, R.; Sonpavde, G.; Knox, J.J.; Tran, B.; et al. Prognostic Role of Neutrophil-to-Lymphocyte Ratio in Solid Tumors: A Systematic Review and Meta-Analysis. JNCI J. Natl. Cancer Inst. 2014, 106, dju124. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Jensen, H.K.; Donskov, F.; Marcussen, N.; Nordsmark, M.; Lundbeck, F.; von der Maase, H. Presence of Intratumoral Neutrophils Is an Independent Prognostic Factor in Localized Renal Cell Carcinoma. J. Clin. Oncol. 2009, 27, 4709–4717. [Google Scholar] [CrossRef] [PubMed]
  126. Li, Y.-W.; Qiu, S.-J.; Fan, J.; Zhou, J.; Gao, Q.; Xiao, Y.-S.; Xu, Y.-F. Intratumoral neutrophils: A poor prognostic factor for hepatocellular carcinoma following resection. J. Hepatol. 2011, 54, 497–505. [Google Scholar] [CrossRef] [PubMed]
  127. Simon, S.C.S.; Utikal, J.; Umansky, V. Opposing roles of eosinophils in cancer. Cancer Immunol. Immunother. 2019, 68, 823–833. [Google Scholar] [CrossRef]
  128. Moreira, A.; Leisgang, W.; Schuler, G.; Heinzerling, L. Eosinophilic count as a biomarker for prognosis of melanoma patients and its importance in the response to immunotherapy. Immunotherapy 2017, 9, 115–121. [Google Scholar] [CrossRef]
  129. Simon, S.C.S.; Hu, X.; Panten, J.; Grees, M.; Renders, S.; Thomas, D.; Weber, R.; Schulze, T.J.; Utikal, J.; Umansky, V. Eosinophil accumulation predicts response to melanoma treatment with immune checkpoint inhibitors. Oncoimmunology 2020, 9, 1727116. [Google Scholar] [CrossRef] [Green Version]
  130. Demers, M.; Wagner, D. NETosis: A New Factor in Tumor Progression and Cancer-Associated Thrombosis. Semin. Thromb. Hemost. 2014, 40, 277–283. [Google Scholar] [CrossRef] [Green Version]
  131. Yipp, B.G.; Kubes, P. NETosis: How vital is it? Blood 2013, 122, 2784–2794. [Google Scholar] [CrossRef]
  132. 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]
  133. Hwang, W.-L.; Lan, H.-Y.; Cheng, W.-C.; Huang, S.-C.; Yang, M.-H. Tumor stem-like cell-derived exosomal RNAs prime neutrophils for facilitating tumorigenesis of colon cancer. J. Hematol. Oncol. 2019, 12, 10. [Google Scholar] [CrossRef] [Green Version]
  134. Cheng, W.; Liao, T.; Lin, C.; Yuan, L.E.; Lan, H.; Lin, H.; Teng, H.; Chang, H.; Lin, C.; Yang, C.; et al. RAB27B-activated secretion of stem-like tumor exosomes delivers the biomarker microRNA-146a-5p, which promotes tumorigenesis and associates with an immunosuppressive tumor microenvironment in colorectal cancer. Int. J. Cancer 2019, 145, 2209–2224. [Google Scholar] [CrossRef]
  135. 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] [PubMed]
  136. Lukic, A.; Ji, J.; Idborg, H.; Samuelsson, B.; Palmberg, L.; Gabrielsson, S.; Rådmark, O. Pulmonary epithelial cancer cells and their exosomes metabolize myeloid cell-derived leukotriene C 4 to leukotriene D 4. J. Lipid Res. 2016, 57, 1659–1669. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Bronte, V.; Brandau, S.; Chen, S.-H.; Colombo, M.P.; Frey, A.B.; Greten, T.F.; Mandruzzato, S.; Murray, P.J.; Ochoa, A.; Ostrand-Rosenberg, S.; et al. Recommendations for myeloid-derived suppressor cell nomenclature and characterization standards. Nat. Commun. 2016, 7, 12150. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Groth, C.; Hu, X.; Weber, R.; Fleming, V.; Altevogt, P.; Utikal, J.; Umansky, V. Immunosuppression mediated by myeloid-derived suppressor cells (MDSCs) during tumour progression. Br. J. Cancer 2019, 120, 16–25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. Gabrilovich, D.I.; Nagaraj, S. Myeloid-derived suppressor cells as regulators of the immune system. Nat. Rev. Immunol. 2009, 9, 162–174. [Google Scholar] [CrossRef]
  140. Petrova, V.; Arkhypov, I.; Weber, R.; Groth, C.; Altevogt, P.; Utikal, J.; Umansky, V. Modern Aspects of Immunotherapy with Checkpoint Inhibitors in Melanoma. Int. J. Mol. Sci. 2020, 21, 2367. [Google Scholar] [CrossRef] [Green Version]
  141. Weber, J.; Gibney, G.; Kudchadkar, R.; Yu, B.; Cheng, P.; Martinez, A.J.; Kroeger, J.; Richards, A.; McCormick, L.; Moberg, V.; et al. Phase I/II Study of Metastatic Melanoma Patients Treated with Nivolumab Who Had Progressed after Ipilimumab. Cancer Immunol. Res. 2016, 4, 345–353. [Google Scholar] [CrossRef] [Green Version]
  142. Jiang, H.; Gebhardt, C.; Umansky, L.; Beckhove, P.; Schulze, T.J.; Utikal, J.; Umansky, V. Elevated chronic inflammatory factors and myeloid-derived suppressor cells indicate poor prognosis in advanced melanoma patients. Int. J. Cancer 2015, 136, 2352–2360. [Google Scholar] [CrossRef]
  143. 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, 35. [Google Scholar] [CrossRef]
  144. 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] [PubMed]
  145. 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]
  146. Xiang, X.; Liu, Y.; Zhuang, X.; Zhang, S.; Michalek, S.; Taylor, D.D.; Grizzle, W.; Zhang, H.-G. TLR2-Mediated Expansion of MDSCs Is Dependent on the Source of Tumor Exosomes. Am. J. Pathol. 2010, 177, 1606–1610. [Google Scholar] [CrossRef] [PubMed]
  147. Xiang, X.; Poliakov, A.; Liu, C.; Liu, Y.; Deng, Z.B.; Wang, J.; Cheng, Z.; Shah, S.V.; Wang, G.J.; Zhang, L.; et al. Induction of myeloid-derived suppressor cells by tumor exosomes. Int. J. Cancer 2009, 124, 2621–2633. [Google Scholar] [CrossRef] [Green Version]
  148. Sinha, P.; Clements, V.K.; Fulton, A.M.; Ostrand-Rosenberg, S. Prostaglandin E2 promotes tumor progression by inducing myeloid-derived suppressor cells. Cancer Res. 2007, 67, 4507–4513. [Google Scholar] [CrossRef] [Green Version]
  149. Ridder, K.; Sevko, A.; Heide, J.; Dams, M.; Rupp, A.-K.; Macas, J.; Starmann, J.; Tjwa, M.; Plate, K.H.; Sültmann, H.; et al. Extracellular vesicle-mediated transfer of functional RNA in the tumor microenvironment. Oncoimmunology 2015, 4, e1008371. [Google Scholar] [CrossRef] [Green Version]
  150. Li, L.; Cao, B.; Liang, X.; Lu, S.; Luo, H.; Wang, Z.; Wang, S.; Jiang, J.; Lang, J.; Zhu, G. Microenvironmental oxygen pressure orchestrates an anti- and pro-tumoral γδ T cell equilibrium via tumor-derived exosomes. Oncogene 2019, 38, 2830–2843. [Google Scholar] [CrossRef]
  151. Guo, X.; Qiu, W.; Liu, Q.; Qian, M.; Wang, S.; Zhang, Z.; Gao, X.; Chen, Z.; Xue, H.; Li, G. Immunosuppressive effects of hypoxia-induced glioma exosomes through myeloid-derived suppressor cells via the miR-10a/Rora and miR-21/Pten Pathways. Oncogene 2018, 37, 4239–4259. [Google Scholar] [CrossRef]
  152. Guo, X.; Qiu, W.; Wang, J.; Liu, Q.; Qian, M.; Wang, S.; Zhang, Z.; Gao, X.; Chen, Z.; Guo, Q.; et al. Glioma exosomes mediate the expansion and function of myeloid-derived suppressor cells through microRNA-29a/ Hbp1 and microRNA-92a/Prkar1a pathways. Int. J. Cancer 2019, 144, 3111–3126. [Google Scholar] [CrossRef]
  153. Bruns, H.; Böttcher, M.; Qorraj, M.; Fabri, M.; Jitschin, S.; Dindorf, J.; Busch, L.; Jitschin, R.; Mackensen, A.; Mougiakakos, D. CLL-cell-mediated MDSC induction by exosomal miR-155 transfer is disrupted by vitamin D. Leukemia 2017, 31, 985–988. [Google Scholar] [CrossRef]
  154. Ren, W.H.; Zhang, X.R.; Li, W.B.; Feng, Q.; Feng, H.J.; Tong, Y.; Rong, H.; Wang, W.; Zhang, D.; Zhang, Z.Q.; et al. Exosomal miRNA-107 induces myeloid-derived suppressor cell expansion in gastric cancer. Cancer Manag. Res. 2019, 11, 4023–4040. [Google Scholar] [CrossRef] [PubMed]
  155. Biswas, S.; Mandal, G.; Roy Chowdhury, S.; Purohit, S.; Payne, K.K.; Anadon, C.; Gupta, A.; Swanson, P.; Yu, X.; Conejo-Garcia, J.R.; et al. Exosomes Produced by Mesenchymal Stem Cells Drive Differentiation of Myeloid Cells into Immunosuppressive M2-Polarized Macrophages in Breast Cancer. J. Immunol. 2019, 203, 3447–3460. [Google Scholar] [CrossRef] [PubMed]
  156. Gilligan, K.E.; Dwyer, R.M. Engineering Exosomes for Cancer Therapy. Int. J. Mol. Sci. 2017, 18, 1122. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Ohno, S.; Drummen, G.; Kuroda, M. Focus on Extracellular Vesicles: Development of Extracellular Vesicle-Based Therapeutic Systems. Int. J. Mol. Sci. 2016, 17, 172. [Google Scholar] [CrossRef] [Green Version]
  158. Nishida-Aoki, N.; Tominaga, N.; Takeshita, F.; Sonoda, H.; Yoshioka, Y.; Ochiya, T. Disruption of Circulating Extracellular Vesicles as a Novel Therapeutic Strategy against Cancer Metastasis. Mol. Ther. 2017, 25, 181–191. [Google Scholar] [CrossRef] [Green Version]
  159. Yamayoshi, A.; Oyama, S.; Kishimoto, Y.; Konishi, R.; Yamamoto, T.; Kobori, A.; Harada, H.; Ashihara, E.; Sugiyama, H.; Murakami, A. Development of Antibody–Oligonucleotide Complexes for Targeting Exosomal MicroRNA. Pharmaceutics 2020, 12, 545. [Google Scholar] [CrossRef]
Figure 1. Differentiation of myeloid cells. Hematopoietic stem cells (HSC) differentiate into the common myeloid progenitors (CMP) and further into the granulocyte-macrophage progenitors (GMP). The granulocyte colony-stimulating factor (G-CSF) generates the differentiation of GMP via myeloblasts, promyeloblasts, myelocytes, and band forms into granulocytes (shown by the arrows), whereas macrophage colony-stimulating factor (M-CSF) induces GMP development towards monocytes, macrophages or DC via monocyte/macrophage and dendritic cell progenitors (MDP) (shown by the arrows).
Figure 1. Differentiation of myeloid cells. Hematopoietic stem cells (HSC) differentiate into the common myeloid progenitors (CMP) and further into the granulocyte-macrophage progenitors (GMP). The granulocyte colony-stimulating factor (G-CSF) generates the differentiation of GMP via myeloblasts, promyeloblasts, myelocytes, and band forms into granulocytes (shown by the arrows), whereas macrophage colony-stimulating factor (M-CSF) induces GMP development towards monocytes, macrophages or DC via monocyte/macrophage and dendritic cell progenitors (MDP) (shown by the arrows).
Ijms 21 06319 g001
Figure 2. Tumor-derived extracellular vesicles (TEV) affect myeloid cell differentiation and function. Tumor cells secrete TEV containing tumor-derived factors (proteins, mRNA, miRNA etc.), which interact with myeloid progenitors (partially blocking their differentiation to mature myeloid cells that is shown with “x”), monocytes, neutrophils and myeloid-derived suppressor cells (MDSC), resulting in tumor-promoting immunosuppression (shown with arrows). Exposure of dendritic cells (DC) to TEV leads to either acquisition of immunosuppressive function or induction of immunostimulatory capacity (shown with arrows).
Figure 2. Tumor-derived extracellular vesicles (TEV) affect myeloid cell differentiation and function. Tumor cells secrete TEV containing tumor-derived factors (proteins, mRNA, miRNA etc.), which interact with myeloid progenitors (partially blocking their differentiation to mature myeloid cells that is shown with “x”), monocytes, neutrophils and myeloid-derived suppressor cells (MDSC), resulting in tumor-promoting immunosuppression (shown with arrows). Exposure of dendritic cells (DC) to TEV leads to either acquisition of immunosuppressive function or induction of immunostimulatory capacity (shown with arrows).
Ijms 21 06319 g002
Table 1. Effects of TEV on myeloid cells.
Table 1. Effects of TEV on myeloid cells.
Cell TypesTEV-Associated MoleculesEffectsSources of TEVReferences
myeloid progenitorsc-Mycaccumulation of MDSChuman cell line of myeloid leukemia; patient-derived cells[60]
not shownblock of differentiation, accumulation of MDSCmurine cell lines of lung cancer and breast cancer[61,62]
MET kinaseactivation of the Erk pathway, pro-metastatic behaviormurine cell line of skin cancer[63]
HSP90αTLR4-dependent MDSC inductionmurine and human cell lines of skin cancer[64]
monocytesnot shownimmunosuppressive phenotypehuman cell lines of colorectal cancer, skin cancer, pancreatic cancer, gastric cancer and malignant brain tumor[68,75,76,77,79]
Y RNA hY4activation of TLR7-dependent signaling, protumorigenic phenotypehuman cell line of lymphocytic leukemia[70]
unknown proteinsactivation of TLR-dependent signalingamniotic fluid and malignant ascites from patients with ovarian cancer[71]
hyaluronanactivation of PI3K/Akt signaling, production of anti-inflammatory cytokineshuman cell line of pancreatic cancer[72]
miRNA-21immunosuppressive phenotypehuman cell line of head and neck cancer[73]
not showncontact time-dependent effect (early contact: immunosuppressive; late contact: proinflammatory)human cell line of colon cancer[74]
set of miRNAconversion of monocytes into MDSCmurine and human cell lines of skin cancer[69]
not shownsecretion of pro-inflammatory cytokineshuman cell line of head and neck cancer[78]
pigment epithelium-derived factorcancer cell clearance at the pre-metastatic nichemurine and human cell lines of skin cancer[80]
macrophagesmacrophage migration inhibitory factorTGF-β-dependent formation of pre-metastatic nichesmurine and human cell lines of pancreatic cancer[90]
not shownaltered secretion of cytokinesmurine cell lines of skin cancer; human cell lines of colorectal cancer and liver cancer[92,93,96]
gp130cytokine secretion, induction of STAT3 signalingmurine cell line of breast cancer[94]
let-7a, chemoattractantsmacrophage recruitment, immunosuppressive phenotypemurine cell line of skin cancer under hypoxic conditions[95]
miRNA-1246immunosuppressive phenotype, induction of STAT3 signaling, increased motility of glioma cellshuman cell line of glioma under hypoxic conditions[97]
miRNA-222immunosuppressive phenotype, induction of STAT3 signalinghuman cell line of ovarian cancer[98]
miRNA-146aimmunosuppressive phenotype, induction of STAT3 signalingmurine and human cell lines of liver cancer[99]
miRNA-150upregulation of VEGF, immunosuppressive phenotypeHEK cells, overexpressing miR-150[100]
miRNA-23aimmunosuppressive phenotype, activation of PI3K/Akt signalinghuman cell line of liver cancer[101]
DCnot showninhibition of DC maturation, production of anti-inflammatory cytokinesmurine cell lines of thymic cancer and skin cancer[111]
HSP72, HSP105TLR signaling-dependent matrix metallopeptidase 9 productionmurine cell lines of skin cancer and breast cancer; tumor tissue from patients with breast cancer[112]
not shownimmunosuppressive phenotype, decreased migration of DC to the draining lymph nodesmurine cell lines of lung cancer and breast cancer[61]
miRNA-212-3pdownregulation of MHCII expressionhuman cell line of pancreatic cancer[113]
miRNA-203downregulation of TLR4 expressionhuman cell line of pancreatic cancer[114]
miRNA-155, miRNA-142 or let-7ienhanced maturation of DC and increased stimulation of T cellsmurine cell lines of breast cancer and colon cancer[115,116]
tumor antigensdecreased Treg, increased survival of mice treated with sorafenib and anti-PD-1 antibodiesmurine cell line of liver cancer[118]
not shownincreased survival of micemurine cell line of pancreatic cancer[119]
Granulocytesnot showninduction of NET formation by neutrophilsmurine cell line of breast cancer; plasma from breast tumor-bearing mice[132]
not shownincreased neutrophil survival, protumorigenic phenotypemurine cell line of colorectal cancer[133]
miRNA-146aincreased neutrophil infiltration of tumorshuman cell line of colorectal cancer; serum from patients with colorectal cancer[134]
HMGB1TLR4-dependent NF-κB activation, increased autophagic responsehuman cell line of gastric cancer[135]
γ-glutamyl transpeptidase 1conversion of eosinophil-derived leukotriene C4 into leukotriene D4human cell line of lung cancer[136]
MDSCHSP70TLR2 signaling-dependent cytokine release and induction of STAT3, activation of MDSC, tumor progressionmurine cell lines of kidney cancer, skin cancer and colon cancer; human cell lines of colon cancer, prostate cancer, cervix cancer[143,144]
HSP72TLR2 signaling-dependent induction of STAT3, activation of MDSCmurine cell lines of colon cancer, lymphatic cancer; tissue from tumor-bearing mice (breast cancer, skin cancer, lymphoma); human cell line of lung cancer[145,146]
miRNA-21PTEN-dependent PD-L1 upregulation, enhanced suppression of γδ T cell functionshuman cell line of oral cancer under hypoxic conditions[150]
miRNA-10a, miRNA-21, miRNA-29a, miRNA-92aexpansion and activation of MDSCmurine cell line of glioma under hypoxic conditions[151,152]
miRNA-155activation of MDSChuman cell line of lymphocytic leukemia[153]
miRNA-107expansion of MDSC with increased Arg-1 expressionhuman cell line of gastric cancer, serum from patients with gastric cancer[154]
TGF-β, C1q, semaphorinsupregulated PD-L1 expression, differentiation towards M2 macrophages, tumor progressionhuman and murine MSC isolated from breast tumors[155]
prostaglandin E2 and TGF-β

Share and Cite

MDPI and ACS Style

Arkhypov, I.; Lasser, S.; Petrova, V.; Weber, R.; Groth, C.; Utikal, J.; Altevogt, P.; Umansky, V. Myeloid Cell Modulation by Tumor-Derived Extracellular Vesicles. Int. J. Mol. Sci. 2020, 21, 6319. https://doi.org/10.3390/ijms21176319

AMA Style

Arkhypov I, Lasser S, Petrova V, Weber R, Groth C, Utikal J, Altevogt P, Umansky V. Myeloid Cell Modulation by Tumor-Derived Extracellular Vesicles. International Journal of Molecular Sciences. 2020; 21(17):6319. https://doi.org/10.3390/ijms21176319

Chicago/Turabian Style

Arkhypov, Ihor, Samantha Lasser, Vera Petrova, Rebekka Weber, Christopher Groth, Jochen Utikal, Peter Altevogt, and Viktor Umansky. 2020. "Myeloid Cell Modulation by Tumor-Derived Extracellular Vesicles" International Journal of Molecular Sciences 21, no. 17: 6319. https://doi.org/10.3390/ijms21176319

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