Abstract
Gliomas are the most common primary malignant tumors in the central nervous system. However, conventional treatments, such as surgical resection and postoperative combined chemo‐ and radio‐therapy, are ineffective in improving patients' long‐term survival. The tumor microenvironment (TME) consists of stromal cells, tumor components, and innate and acquired immune cells, and these cells, along with the extracellular matrix, regulate and communicate intercellularly to promote TME formation. The immune microenvironment plays a vital role in the development of glioma. Exosomes, which are extracellular vesicles (EVs), facilitate intercellular communication and regulation within the TME. Tumor cells can release exosomes to transmit messages, induce macrophage polarization, and inhibit immune cell activity, ultimately promoting metastasis and immune evasion. Moreover, immune cells can regulate tumorigenesis and progression through exosomes. This review summarized the biological properties of exosomes and their effects on the tumor microenvironment and provides an overview of the interactions between glioma cells and immune cells.
Keywords: exosomes, glioma, immune cells, immunotherapy, tumor microenvironment
Abstract
The tumor immune microenvironment contains parenchyma tumor cells and immune cells. By releasing exosomes, tumor cells can transmit messages, induce macrophage polarization, and inhibit immune cell activity, thus promoting metastasis and immune escape. Immune cells can also regulate glioma cells genesis and progression through exosomes.
1. INTRODUCTION
Gliomas are malignant tumors originating from the central nervous system and are characterized clinically by high morbidity, mortality, high infiltrating, and poor prognosis. 1 , 2 The main treatments for glioma are direct surgical resection, radiotherapy, and temozolomide chemotherapy. 3 , 4 , 5 , 6 , 7 Gliomas are classified as astrocytic, oligodendroglial, and ventricular canal tumors based on their malignancy and phenotype. 8 , 9 , 10 Diffuse gliomas can be further typed as astrocytic, oligodendroglial, or rare mixed oligodendroglial‐astrocytic of World Health Organization (WHO) grade II (low grade), III (anaplastic), or IV (glioblastoma). According to the WHO histological classification, gliomas are classified as WHO grade I–IV, with glioblastoma, a WHO grade IV glioma, accounting for approximately 45% of malignant brain tumors. 11 , 12 The pathogenesis of glioma remains unclear, with tumorigenesis resulting from a combination of environmental and genetic factors.
The tumor microenvironment (TME) consists of tumor cells and surrounding components. 13 , 14 It includes innate and adaptive immune cells (T lymphocytes and B lymphocytes), mesenchymal fibroblasts, and vascular and lymphatic vessel networks. Various chemokines secreted through autocrine or paracrine make up the TME. 15 , 16 , 17 Alterations in the microenvironment affect tumorigenesis and progression. The immune cell is fundamental in determining the fate of cancer and its invasiveness and metastatic capacity. 18 , 19 The clinical outcome of cancer patients is interrelated to the composition of immune cells that infiltrate tumors. 20 , 21
Exosomes are extracellular vesicles of 30–150 nm in diameter released in the form of exocytosis. The intake of exosomes relies on the endocytic system after fusion with the cell membrane. 22 , 23 , 24 Exosome contains various bioactive components, 25 , 26 , 27 , 28 and most cells (e.g., mast cells, tumor cells, dendritic cells, neuron‐shaped glial cells) can generate exosomes. 29 , 30 , 31 The outer membrane of exosomes has the property of a cytosolic phospholipid bilayer, effectively maintaining the activity and preserving stability in various body fluids. 32 , 33 The exosomes are abundant in TME and function as the most critical information exchange tool between tumor cells and TME. 34 , 35 , 36 , 37 , 38
Exosomes derived from tumor cells promote immune escape by directly inhibiting immune cells or regulating the expression of related cytokines. 39 , 40 Moreover, immune cells in the TME influence tumor cell growth, metastasis, and drug resistance by secreting exosomes. 41 , 42 , 43 This article reviewed the regulation of exosomes in the glioma TME and their role in the malignant progression of glioma.
2. OVERVIEW OF EXOSOMES
2.1. Mechanism of exosome formation
Exosomes are generated in normal physiological conditions or response to external environmental stimuli. They originate from intracellular invaginations, where the cell membrane forms multiple vesicles. These vesicles fuse to form early intracellular vesicles, which further mature and bud inward to form luminal vesicles or intraluminal vesicles (ILVs). 44 , 45 , 46 ILVs‐rich intracellular bodies are called multi‐vesicular bodies (MVBs). Upon fusion with lysosomes intracellularly, MVBs are degraded, while the other part is secreted outward to form exosomes under the regulation of Rab enzymes of the GTPase family. 47 , 48 , 49 Exosomes are extracellular vesicles with a diameter of 30–150 nm generated by almost all cell types. 50 , 51 Exosomes consist of a lipid bilayer containing transmembrane proteins and wrap around cytoplasmic proteins, lipids, or nucleic acids. 52 , 53 Exosomes can regulate cellular communication by transporting specific exosomal contents. 54 , 55 Exosomes are closely associated with normal physiological homeostasis and various diseases, including cancer. Exosomes are essential mediators of tumorigenesis, proliferation, angiogenesis, and distant metastasis. 56 , 57 Currently, the extraction of exosomes has become commercially available. The advantages and disadvantages of different methods are presented in Table 1.
TABLE 1.
The separation techniques for exosomes.
Detection methods | Advantages | Disadvantages |
---|---|---|
Ultracentrifugation | The obtained exosomes are not contaminated by the separation reagent, and the number of separations is large, and the processed sample is small | The instrument is expensive, the sample volume is large, the time consumption is long, and protein contamination still exists when exosomes are observed by electron microscopy |
Sucrose Density Gradient Centrifugation | High purity of exosomes | The preliminary preparation is complicated, time‐consuming, and cannot completely separate exosomes from proteins |
Polyethylene Glycol (PEG) | Simple operation, no special equipment required, more economical, and high yield of exosomes | Precipitation of some non‐exosome hydrophobic substances resulting in insufficient exosome purity |
Kit method | Simple, less time‐consuming, and higher yield of exosomes | The obtained exosome precipitate contains many impurities, and samples from different sources need to be extracted using different kits, and the kits are more expensive |
SEC | High purity of exosomes enables the isolation of structurally intact and functionally active vesicles | Special equipment required |
Ultrafiltration | Rapid separation of exosomes of different sizes with high capture efficiency | Filters can easily become clogged with vesicles and other macromolecular substances, causing the membrane to become overstressed and broken |
Separation method is based on surface component affinity | Higher recovery and less impurity protein | Based on special protein adsorption |
ACE separation method | The ability to simplify the exosome extraction and recovery process, significantly reducing processing steps and time‐consuming | Requires an AC electric field to be applied |
Microfluidic chip method | Simple operation and high capture rate | Requires nanofilters |
2.2. Exosomes functions
Exosomes are widely distributed in the body fluid of tumor patients after being released. 58 , 59 They can enter and deliver biologically active substances to target cells, affecting gene expression, protein synthesis, and other processes that ultimately regulate the target cells' function. 60 , 61 Exosomal surface proteins bind directly to target cell receptors and stimulate signaling pathways, and they can also fuse with target cell membranes and deliver functional proteins, miRNA, and other biomolecules. 22 , 62 The mRNA of exosomes can translate corresponding proteins, and miRNA and siRNA regulate target cells by affecting the expression of related genes. 63 , 64 Phagocytose exosomes can either be re‐released in target cells or degraded by the lysosomal pathway. 65 , 66
The exosomal function is closely related to their cellular origin and the protein and RNA they contain. Exosomes from different sources have different purposes at different physiological and pathological stages. 67 , 68 Exosomes can regulate physiological activities and maintain intracellular homeostasis, support the body's immune tolerance, and participate in normal physiological processes, and pathological processes. 69 , 70 Tumor cell‐derived exosomes contain antigens, genetics, and other biologically active substances that are vital in tumorigenesis, proliferation, invasion, and metastasis. 71 Proteins, miRNAs, and even DNA in tumor‐derived exosomes are potential markers for non‐invasive diagnosis. 72 , 73 As natural carriers, the phospholipid bilayer structure of exosomes protects the stability of proteins, miRNA, and other biologically active substances. Exosomes have similar biological activities as parental cells, are widely distributed, exist for a long time in the body, and have the advantages of a long half‐life and natural nontoxicity. Due to their nanoscale structure, exosomes can evade phagocytosis while freely shuttling between cells and matrix, with solid penetration ability and low immunogenicity. 74 Exosomes can also serve as a means of delivering drugs and miRNAs for tumor therapy. Tumor cell‐derived exosomes are also potential for tumor vaccine development. 75
2.3. Immune system cells‐derived exosomes (IEXs)
IEXs have a comprehensive spectrum of functions in the immune system. IEXs regulate multiple immune signaling pathways, including modulation, antigen presentation, antitumor immunity, and immune system suppression. Dendritic cell‐derived exosomes (DEX) can enhance antitumor immunity and activate specific T cells in the direct pathway by expressing the MHCII‐peptide complexes, costimulatory molecules, and binding to T‐cell receptors. In the indirect pathway, DEXs deliver the MHC II‐peptide complex to other DCs, a process named MHC‐dressing. Exosomes can transfer antigenic peptides from activated to inactivated DCs, which increases the number of MHCII‐peptide complexes on the surface of DCs to activate the T cells. Macrophage‐derived exosomes contain over 5100 proteins, and exosomal miR‐21‐5p and miR‐155‐5p secreted from nonclassical macrophages (M2) regulate tumor cells' migration, proliferation, invasion, and angiogenesis. Neutrophils‐derived exosomes have been shown to have unique protein profiles that change in activated and non‐activated situations. Immune cells generate exosomes with their properties that create an optimal microenvironment for function by activating other immune cells, inhibiting immune responses, and participating in the licensing phenomenon of APCs. In the following, we will discuss the characteristics of the exosomes of each cell involved in the glioma interaction with the immune system.
3. TUMOR MICROENVIRONMENT
Tumor cells typically invade normal tissues and establish a tumor microenvironment (TME) consisting of immune cells, stromal cells, vascular endothelial cells, and extracellular matrix (ECM). 76 , 77 , 78 , 79 Tumor cells can recruit and activate these components to form a tumor‐inhibiting inflammatory microenvironment, which can prevent tumor progression during the early stages of colonization or proliferation 80 , 81 (Figure 1). However, persistent tumor antigen stimulation and immune activation can lead to a depleted or remodeled state of the effector cells in the microenvironment, resulting in an immunosuppressive microenvironment that promotes malignant tumor phenotypes 82 , 83 , 84 (Figure 1). Immunotherapeutic strategies targeted at the TME can stimulate or restore the innate tumor‐suppressive capacity of the immune system, create a favorable immune microenvironment, and produce a comprehensive response. 85 , 86 , 87
FIGURE 1.
The primary cells involved in the tumor immune microenvironment. In the tumor immune microenvironment, some cells (NKT1, M1, N1, DC, ILC1, NK1, and Th1) directly kill tumor cells, while others (MDSC, M2, N2, ILC2, NK2, Th2, and Treg) cause immunosuppression. N1, N1‐polarized neutrophils; N2, N2‐polarized neutrophils.
Tumor metastasis is the primary cause of mortality. The pre‐metastatic microenvironment of tumors refers explicitly to the microenvironment in which the primary tumor focus is prepared for distant dissemination and colonization of tumor cells (Figure 2). The pre‐metastatic microenvironment includes tumor‐derived secretory factors, extracellular vesicles, macrophages, neutrophils, and T cells. With the manipulation of the primary tumor, multiple changes occur at distant sites, including the formation of PMN, inflammation, immunosuppression, angiogenesis/vascular permeability, organophilic, reprogramming, and lymphangiogenesis.
FIGURE 2.
The primary process of tumor metastasis. Tumor metastasis mainly includes invasion, intravasation, circulation, extravasation, and colonization. It also provides for the involvement of macrophages, neutrophils, and T cells.
The glioma microenvironment refers to the internal and external environment closely associated with glioma development, proliferation, and metastasis, including the structure, function, and metabolism of the tumor‐host tissue and the intrinsic environment of glioma cells. 88 , 89 , 90 Glioma cells require large amounts of nutrients to meet their metabolic needs. The plasticity of glioma metabolism allows them to adapt to a depleted or changing nutritional environment, reshaping the tumor immune microenvironment. 91 , 92 , 93 The tumor‐promoting effect of glioma‐associated microglia (GAMs), macrophages, regulatory T cells (Treg), and the inactivation of natural killer (NK) cells in TME can reduce the antitumor effect and are closely related to the formation of glioma immunosuppressive microenvironment. 88 , 94 In addition to immunosuppressive factors, the TME accumulates metabolites from tumor proliferation, such as adenosine and lactate. 95 , 96 Hypoxic TME increases the release of ATP and AMP. The nucleotidases CD39 and CD73 catalyze the conversion of ATP to AMP and AMP to adenosine extracellularly, resulting in a significant increase in adenosine levels in immune regulation. 97 , 98
4. EXOSOME‐MEDIATED INTERACTION IN GLIOMA
4.1. Interaction between glioma and immune cells in the TME
Glioma is a primary malignant tumor in the central nervous system (CNS), characterized by aggressive growth and difficulty in clinical cure. These features may be related to the low immunogenicity of glioma cells and the tumor immunosuppressive microenvironment. 99 Immune cells in the TME compete with glioma cells for nutrients, and the metabolites produced during this competition can affect immune cell differentiation and function. Glioma cells can influence the process of immune cells by secreting exosomes, and promote their malignant progression (Table 2). Similarly, immune cells can influence glioma progression by delivering substances to glioma cells through exosomes (Table 3).
TABLE 2.
Overview of glioma cell‐derived exosome cargos and their biological effects in TME.
Donor cell | Exosomal cargos | Receiving cell | Biological effects | References |
---|---|---|---|---|
Glioma cells | / | Monocytes | Suppress T‐cell immune responses | 107 |
Glioma cells | miR‐10a/miR‐21/RORA/PTEN | MDSCs | Induce MDSCs expansion and activation | 108 |
Glioma cells | miR‐29a/miR‐92a /HBP1/PRKRA1A | MDSCs | Activate the proliferation of MDSCs | 150 |
Glioma cells | SDF‐1α/CXCR4 | MDSCs | Induces MDSC recruitment | 109 |
Glioma cells | miR‐1246/DUSP/ERK | MDSCs | Drive differentiation and activation of MDSCs | 151 |
Glioma cells | miR‐1298‐5p/SETD7 /MSH2 | MDSCs | Promote immunosuppression of MDSCs | 110 |
Glioma cells | miR‐1983/TLR7/MyD88‐IRF5/IRF7/IFN‐β | NK cells | Stimulate NK cell responses | 114 |
GL26 cells | IFN‐γ and Granzyme B | CD8+ cells | Inhibit cell growth | 121 |
GSCs | / | CD4+ T | Promote cell growth | 107 |
Glioma cells | CD73 | T cells | Promote clonal proliferation of T cells | 122 |
GBM cells | miR‐451/miR‐21/c‐Myc | Microglia | Inhibit cell growth | 136 |
GBM cells | miR‐214‐5p/CXCR5 | Microglia | Regulate the inflammatory response of microglia | 137 |
Glioma cells | miR‐1246/TERF2IP/STAT3/NF‐κB | Macrophage | Promote M2 macrophage polarization | 138 |
GBM cells | Arginase‐1 | Macrophage | Promote M2 macrophage polarization | 139 |
Glioma cells | IL‐6‐pSTAT3‐miR‐155‐3p‐autophagy‐pSTAT3 | Macrophage | Promote M2 macrophage polarization | 141 |
Glioma cells | Circ‐NEIL3/HECTD4/IGF2BP3 | TAMs | Confer immunosuppressive properties on TAMs | 143 |
TABLE 3.
Overview of immune cell‐derived exosome cargos and their biological effects in TME.
Donor cell | Exosomal cargos | Receiving cell | Biological effects | References |
---|---|---|---|---|
DCs | c‐Cbl/PI3K/Akt/ERK | CD4+ and CD8+ T cells | Promote cell proliferation and inhibit tumor growth | 102 |
DCs | LGALS9 | T cells | Promotes DCs tumor antigen‐presenting activity and durable antitumor immunity | 103 |
NK cells | IL‐15 | Glioma cells | Inhibits the growth of glioblastoma | 152 |
Neutrophils | DOX | Glioma cells | Inhibits the growth of glioma | 128 |
TAMs | miR‐27a‐3p/miR‐22‐3p/miR‐221‐3p | GSCs | Induction of GSC proneuromesenchymal transition | 153 |
M2 microglia | miR‐7239‐3p/BMAL1 | Glioma cells | Promote glioma proliferation and migration | 140 |
M2 macrophages | miR‐15a/miR‐92/CCND1/RAP1B | Glioma cells | Inhibit tumor cell migration and invasion | 154 |
TAMs | Lnc‐TAL/ENO1/p38/MAPK | M2 microglia | Lead to chemotherapy resistance | 155 |
4.2. Dendritic cells (DCs)
Studies have shown that dendritic cell‐derived exosomes (DEX) can enhance antitumor immunity and activate specific T cells to combat tumor cells. Using methods to upregulate and downregulate exosome production by immune cells is a novel way to regulate immunity against tumors and infected cells, as well as immune reactions in some autoimmune and allergic diseases. DCs are the cells in the T cell‐mediated immune response to cancer within the organism. DCs and their precursors in tumors can be recruited and respond to several molecular signals, including cell death, inactivation, and successful maturation in the TME. 100 Immature DCs cannot initiate T‐cell responses to tumors and may induce immune tolerance, while mature DCs can migrate to tumor‐draining lymph nodes to create T‐cell responses, recruit T cells into the TME, and produce immunostimulatory cytokines to regulate the TME. 101
Research suggests that chaperone‐rich cell lysates (CRCLs) may play an essential role in developing antitumor vaccines. DCs from CRCLs loaded with GL261‐derived glioma cells can significantly prolong the survival of tumor‐bearing mice and inhibit tumor growth in vivo by secreting exosomes. The CRCL‐GL261‐DCs promote cell proliferation and cytotoxic T lymphocyte (CTL) activity of CD4+ and CD8+ T cells in vitro. Mechanism findings suggest that CRCL‐GL261‐DCs can negatively regulate Casitas B cell line lymphoma (Cbl)‐b and c‐Cbl signaling to lead to activation of phosphatidylinositol 3‐kinase (PI3K)/Akt and extracellular signaling‐regulatory kinase (ERK) signaling in T cells. 102 Cerebrospinal fluid (CSF) exosomes from GBM patients contain the unique protein‐LGALS9 ligand, which binds to the TIM3 receptor of DCs in CSF and further interferes DCs recognition, inhibits the presentation of antigen, resulting in the deactivation of cytotoxic T cell‐mediated antitumor immune responses. Blockade of the tumor secretory exosome LGALS9 in a mouse GBM tumor model significantly promoted DCs antigen presentation activity and durable antitumor immunity. 103 Targeting LGALS9 may be an effective therapeutic tool for GBM.
4.3. Myeloid‐derived suppressor cells (MDSCs)
Myeloid‐derived suppressor cells protect tumor cells from immune attack by negatively regulating the immune response, depleting essential amino acids like arginine and cysteine, necessary for T cell activation, and the production of reactive oxygen and nitrogen species that repress T cell function. 104 MDSCs also induce immune tolerance by recruiting regulatory T cells and Th17 cells through cytokine secretion such as TGF‐β, IL‐10, IFN‐γ, and upregulating ligands like CD86. 100 Additionally, MDSCs suppress innate immunity by inducing macrophage polarization towards the M2 phenotype and inhibiting NK cell‐mediated cytotoxic effects. 105 Apart from immunosuppressive functions, MDSCs upregulate vascular endothelial growth factors promoting angiogenesis and tumor growth acceleration. 106 Stimulated PBMCs from healthy donors with anti‐CD3, anti‐CD28, and IL‐2 with glioma stem cell‐derived exosomes significantly inhibited T cell activation, proliferation, and Th1 cytokine production but enhanced purified CD4+ T cell proliferation without affecting cell viability. 107 Glioma‐derived exosomes repress T‐cell immune responses by acting on monocyte maturation rather than interacting directly with T cells. Hypoxia‐induced glioma‐derived exosomes can more effectively induce MDSCs than normoxia‐induced exosomes by promoting the expression of miR‐10a and miR‐21, targeting RORA, and PTEN. 108 Additionally, hypoxia‐induced glioma cells can stimulate functional MDSC differentiation by transferring exosomal miR‐92a, activating the proliferative ability of MDSCs by targeting HBP1 and PRKAR1A. Glioma cell line‐derived exosomes, BATF2‐Exo, can inhibit MDSC chemotaxis by inhibiting intracellular SDF‐1α, while exosomal miR‐1246 activates MDSC differentiation and activation via the DUSP3/ERK pathway. 109 Furthermore, the upregulation of POU5F1 and hnRNPA1 enhances miR‐1246 transcription and packaging. Suppression of hypoxia‐driven exosomal miR‐1246 expression in glioma cells and PD‐L1 expression in MDSCs is a promising new approach for treating glioma. Finally, enrichment of miR‐1298‐5p in CSF exosomes significantly inhibits glioma progression by promoting the immunosuppressive effects of MDSCs and glioma through targeting SETD7 and MSH2. 110
4.4. Natural killer (NK) cells
NK cells represent a significant subset of lymphocytes in innate immunity. They enhance cytotoxicity in the immune response by releasing various cytokines, including perforin, interferon‐γ (IFN‐γ), tumor necrosis factor (TNF), granulocyte‐macrophage colony‐stimulating factor (GM‐CSF), and macrophage inflammatory protein‐1 (MIP‐1). 111 , 112 Interleukin‐15 (IL‐15) significantly increases the potential of NK cell‐derived exosomes (NK‐Exo) for immunotherapy. NK‐Exo containing IL‐15 (NK‐Exo‐IL‐15) demonstrated greater cytolytic activity against glioblastoma and significantly increased NK cell cytotoxicity. Neither NK‐Exo nor NK‐Exo‐IL‐15 was toxic to normal cells or mice. NK‐Exo‐IL‐15 inhibited the proliferation of mouse glioblastoma xenografts compared to NK‐Exo. 113 Glioma‐released exosome‐derived miR‐1983 plays a vital role in the innate antiglioma NK‐mediated circuit regulated by galectin‐1 (Gal‐1). As an endogenous TLR7 ligand, miR‐1983 activates TLR7 in pDC and cDC via the 5′‐UGUUU‐3′ motif at its 3′ end and further activates downstream signaling, stimulating IFN‐β secretion via MyD88‐IRF5/IRF7, and ultimately eradicating gliomas through stimulation of NK cells. 114
4.5. T cells and B cells
Lymphocytes have a complex role in tumor immune escape and inhibition of tumor growth. 115 , 116 CD4+ T cells and CD8+ T cells can hinder tumor growth and metastasis through specific immune responses. 117 , 118 However, regulatory B cells (Bregs) utilize immune resistance through Tregs and can promote tumor immune escape. 119 , 120 GL26 cell‐derived exosomes significantly suppressed the percentage of CD8+ T cells in splenocytes by inhibiting the release of IFN‐γ and granzyme. 121 Glioma stem cell (GSC)‐derived exosomes function as mediators of intercellular communication and promote tumor immune escape by inhibiting T cell activation, proliferation, and Th1 cytokine production while enhancing the proliferation of purified CD4+ T cells. Furthermore, glioma‐derived exosomes in PBMC directly promote the production of IL‐10 and arginase 1 by unstimulated CD14+ monocytes and down‐regulate HLA‐DR, producing a phenotype similar to monocyte myeloid‐derived suppressor cells (Mo MDSCs). 107 The concentration of exosome‐derived CD73+ in tumor‐derived extracellular vesicles (TDEVs) was significantly higher in the body fluids of GBM patients. In vitro, results showed that T cells could take up CD73+ TDEVs released from GBM cells. In vivo, defects in exosome synthesis and CD73 expression significantly inhibited tumor growth in GBM‐bearing mice and restored clonal proliferation of T cells in central and peripheral regions. 122
4.6. Macrophage
The macrophage‐derived exosomes contain over 5100 proteins, with their composition changing upon activation. Activated macrophage exosomes promote inflammation by triggering the NLRP3 receptor‐dependent inflammasome, TOLL‐like receptors (TLR), and TNF‐related signaling pathways. Additionally, macrophage‐derived exosomes inhibit the expression of β1 integrins, thus inhibiting endothelial and tumor migration. Exosomes generated from nonclassical macrophages (M2) containing high levels of miR‐21‐5p and miR‐155‐5p molecules regulate tumor migration, proliferation, invasion, and angiogenesis. The miR‐21‐5p enhances the proliferation and drug resistance of tumor cells by targeting PTEN, P21, PCD, and apoptotic protease activating factor 1 (APAF1). On the other hand, miR‐130b‐3p expression level increases in MB patients' plasma exosomes, inhibiting the proliferative ability of tumor cells via inhibiting the SIK1 and p53 signaling pathways. The expression levels of miR‐101‐3p and miR‐423‐5p are significantly higher in MB patients' plasma exosomes than in healthy controls. 123 Moreover, exosomes carrying miR‐101‐3p and miR‐423‐5p suppress tumor cell proliferation, migration, and invasion and enhance cell apoptosis, which could be tumor‐suppressive by targeting the FoxP4 gene. Furthermore, miR‐101‐3p can target EZH2 to increase its tumor‐suppressive effect. 124
4.7. Neutrophils
Neutrophils are the most abundant type of leukocytes and play a crucial role in the inflammatory response's progression. 125 Their exosomes are essential for innate immunity, as neutrophils are among the first cells present at the site of inflammation. However, whether neutrophils exhibit pro‐ or antitumor properties in a microenvironmentally relevant manner depends on multiple cytokine receptors on their surface, enabling them to respond to various signals. 126 , 127 Activated neutrophil‐derived exosomes can alter airway smooth muscle cells' proliferation and biological properties, suggesting their role in airway structural changes in asthma. A bio‐inspired neutrophil‐exosome (NEs‐Exos) system has been identified for delivering doxorubicin drugs for glioma treatment. In vivo experiments have shown that drug‐loaded NEs‐Exos can penetrate the BBB and migrate to the brain. Moreover, NEs‐Exos can chemically respond to inflammatory stimuli and target infiltrating tumor cells in inflamed brain tumors. Intravenous injection of NEs‐Exos/DOX effectively inhibits tumor growth and prolongs survival in a glioma mouse model. 128
4.8. Tumor‐associated macrophages (TAMs)
Tumor‐associated macrophages play a crucial role in tumor growth, invasion, and metastasis. These inflammatory immune cells have two forms of activation, the M1 type and the M2 type. 129 M1 TAMs have antitumor properties and effectively identify and destroy cancer cells through phagocytosis and cytotoxicity. 130 , 131 They perform key functions, such as secreting toxic intermediates and various inflammatory factors, activating Th1 cells, participating in the Th1 immune response, and having proinflammatory and antitumor effects. 132 , 133 In contrast, M2 TAMs can promote tumor progression by secreting cytokines that regulate tumor growth and angiogenesis and reduce patient survival. 134 , 135 The expression of miR‐451/miR‐21 is significantly increased in GBM‐derived exosomes, leading to the malignant progression of GBM by targeting c‐Myc mRNA expression. 136 Additionally, exosomal miR‐214‐5p derived from GBM can regulate the inflammatory response of microglia via targeting CXCR5. 137 Studies have shown that HGDEs, unlike GDEs, significantly promote glioma proliferation, migration, and invasion by inducing M2 macrophage polarization. MiR‐1246 enriched in the CSF of GBM patients can inhibit M2 macrophage polarization by targeting TERF2IP. 138
TAMs reprogrammed by GBex produce immunosuppressive and tumor growth‐promoting proteins, including Arginase‐1 that promotes glioblastoma growth. However, the pro‐growth effect of Arginase‐1 carried by TAM‐derived exosomes can be reversed by the selective Arginase‐1 inhibitor nor‐NOHA. 139 MiR‐7239‐3p in M2 microglia exosomes promotes glioma proliferation and migration, while miR‐92 inhibits tumor cell migration and invasion by targeting CCND1 or RAP1B and regulating the PI3K/AKT/mTOR signaling pathway. 140 H‐GDEs promote autophagy and M2‐like macrophage polarization, thereby promoting glioma progression through the IL‐6‐pSTAT3‐miR‐155‐3p‐autophagy‐pSTAT3 positive feedback loop. 141 Additionally, the miRNA‐124 delivered by HEK293T‐derived EV exerted synergistic antitumor effects by inhibiting the growth of human GBM cells and suppressing M2 microglia polarization. 142 Finally, circ‐NEIL3, upregulated in glioma tissue and stabilized by hnRNPA2B1, can promote tumorigenesis and oncogenic progression by blocking HECTD4‐mediated ubiquitination and by driving macrophage infiltration into the TME. 143
5. PROSPECT AND CONCLUSION
The significance of TME in tumor progression is increasingly recognized, as it actively participates in the process of tumor advancement. Tumor cells in TME account for about 30% of the population. Immune cells within the tumor have been found to play either a tumor‐suppressing or promoting role, while some have a bidirectional regulatory role. 144 , 145 The functions of both tumor cells and immune cells in the TME are reciprocal, as the behavior of tumor cells is closely linked to tumorigenesis and progression, which then influences the biological behavior of stromal cells in the TME. Immunotherapy strategies have shown positive effects and potential cures for advanced high‐grade tumor patients; however, TME evolves dynamically through compensatory feedback mechanisms, blocking immunotherapeutic effects, generating drug resistance, and even tumor progression. 77 , 146 , 147
Under normal physiological conditions, PD‐1/PD‐L1 signaling pathway activation induces peripheral immune tolerance, which maintains T‐cell immune homeostasis, and prevents immune overactivation‐mediated tissue damage. At the same time, tumor cells use the immunosuppressive function of PD‐1/PD‐L1 to evade host immune surveillance and produce tumor growth‐promoting effects (Figure 3). Given the complex nature of the tumor immune microenvironment, improvements are still needed in molecules or signaling pathways targeting TME for immunotherapy to be effective. The main barriers to the effectiveness of immunotherapy currently include inadequate responses of the host immune system to tumor antigens, low infiltration of immune cells in solid tumors, and formation of immunosuppressive TME. 86 , 148
FIGURE 3.
PD‐1 inhibitors activate T cells. The binding of PD‐1 on the surface of T cells to PD‐L1 on the surface of tumor cells inhibits the activation of T cells, while the application of PD‐1 inhibitors promotes the attack of T cells on tumor cells.
Various immunotherapeutic agents targeting TME have been developed in this stage, however, the combination of diverse immunosuppressive signals makes it challenging for single‐targeted therapeutic regimens to have long‐lasting effects. In the tumor microenvironment, the metabolism between glioma cells and other immune cells promotes the immune escape of glioma cells. Therefore, it is critical to explore the mechanisms by which immune cells obtain sufficient nutrients to maintain their antitumor activity. Although the metabolites of glioma cells can influence the differentiation and function of immune cells, further studies are required to determine whether the intrinsic mechanisms by which metabolites affect immune cells differ in different gliomas. The combination of tumor immunotherapy with metabolic enzyme target inhibitors may maintain the metabolic fitness of TIL cells. 149 However, glioma cells and immune cells often use the same metabolic pathways for proliferation, so attention needs to be paid to their potential therapeutic toxicity. Selective targeting of tumor cell‐specific metabolic markers may avoid damage to immune cells, significantly promote antitumor immune responses, and reduce the adverse effects of immunotherapy. Additionally, the differential expression of amino acid and nutrient transport proteins between glioma cells and lymphocytes, especially the carriers involved in the interaction, should be considered. These differences make it possible to selectively inhibit tumor cell metabolic pathways.
AUTHOR CONTRIBUTIONS
Original draft preparation, allocation, and revision: Xu Guo and Rui Sui. Supplement and edition: Haozhe Piao. All authors have read and agreed to the published version of the manuscript.
FUNDING INFORMATION
This Work is supported by the Fundamental Research Funds for the Central University(2021‐YGJC‐17).
CONFLICT OF INTEREST STATEMENT
The authors declare no competing interests.
ACKNOWLEDGMENTS
We thank the generous support of Liaoning Cancer Hospital & Institute (Shenyang) and Dalian University of Technology(Dalian).
Guo X, Sui R, Piao H. Exosomes‐mediated crosstalk between glioma and immune cells in the tumor microenvironment. CNS Neurosci Ther. 2023;29:2074‐2085. doi: 10.1111/cns.14239
DATA AVAILABILITY STATEMENT
The data in the current study are available from the corresponding authors upon reasonable request.
REFERENCES
- 1. Parmigiani E, Taylor V, Giachino C. Oncogenic and tumor‐suppressive functions of NOTCH signaling in glioma. Cell. 2020;9:2304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Diamandis P, Aldape KD. Insights from molecular profiling of adult glioma. J Clin Oncol. 2017;35:2386‐2393. [DOI] [PubMed] [Google Scholar]
- 3. Elmore KB, Schaff LR. DNA repair mechanisms and therapeutic targets in glioma. Curr Oncol Rep. 2021;23:87. [DOI] [PubMed] [Google Scholar]
- 4. Qi Y, Liu B, Sun Q, Xiong X, Chen Q. Immune checkpoint targeted therapy in glioma: status and hopes. Front Immunol. 2020;11:578877. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Kamran N, Alghamri MS, Nunez FJ, et al. Current state and future prospects of immunotherapy for glioma. Immunotherapy. 2018;10:317‐339. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Chen YH, Zeng WJ, Wen ZP, Cheng Q, Chen XP. Under explored epigenetic modulators: role in glioma chemotherapy. Eur J Pharmacol. 2018;833:201‐209. [DOI] [PubMed] [Google Scholar]
- 7. Grek CL, Sheng Z, Naus CC, Sin WC, Gourdie RG, Ghatnekar GG. Novel approach to temozolomide resistance in malignant glioma: connexin43‐directed therapeutics. Curr Opin Pharmacol. 2018;41:79‐88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Li X, Liu M, Zhao J, et al. Research progress about glioma stem cells in the immune microenvironment of glioma. Front Pharmacol. 2021;12:750857. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Cheng F, Guo D. MET in glioma: signaling pathways and targeted therapies. J Exp Clin Cancer Res. 2019;38:270. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Pan T, Wu F, Li L, et al. The role m(6)a RNA methylation is CNS development and glioma pathogenesis. Mol Brain. 2021;14:119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Coleman C, Stoller S, Grotzer M, Stucklin AG, Nazarian J, Mueller S. Pediatric hemispheric high‐grade glioma: targeting the future. Cancer Metastasis Rev. 2020;39:245‐260. [DOI] [PubMed] [Google Scholar]
- 12. Harris M, Svensson F, Kopanitsa L, Ladds G, Bailey D. Emerging patents in the therapeutic areas of glioma and glioblastoma. Expert Opin Ther Pat. 2018;28:573‐590. [DOI] [PubMed] [Google Scholar]
- 13. Zhao Y, Guo S, Deng J, et al. VEGF/VEGFR‐targeted therapy and immunotherapy in non‐small cell lung cancer: targeting the tumor microenvironment. Int J Biol Sci. 2022;18:3845‐3858. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Vahala D, Choi YS. Modelling the tumor microenvironment: recapitulating Nano‐ and Micro‐scale properties that regulate tumor progression. Front Cell Dev Biol. 2022;10:908799. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Radin DP, Tsirka SE. Interactions between tumor cells, neurons, and microglia in the glioma microenvironment. Int J Mol Sci. 2020;21:8476. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Pandey N, Anastasiadis P, Carney CP, et al. Nanotherapeutic treatment of the invasive glioblastoma tumor microenvironment. Adv Drug Deliv Rev. 2022;188:114415. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Kwak SB, Kim SJ, Kim J, et al. Tumor regionalization after surgery: roles of the tumor microenvironment and neutrophil extracellular traps. Exp Mol Med. 2022;54:720‐729. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Zhou J, Wang L, Peng C, Peng F. Co‐targeting tumor angiogenesis and immunosuppressive tumor microenvironment: a perspective in Ethnopharmacology. Front Pharmacol. 2022;13:886198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Huang Y, Kanada M, Ye J, et al. Exosome‐mediated remodeling of the tumor microenvironment: from local to distant intercellular communication. Cancer Lett. 2022;543:215796. [DOI] [PubMed] [Google Scholar]
- 20. Liu J, Chen T, Li S, Liu W, Wang P, Shang G. Targeting matrix metalloproteinases by E3 ubiquitin ligases as a way to regulate the tumor microenvironment for cancer therapy. Semin Cancer Biol. 2022;86:259‐268. [DOI] [PubMed] [Google Scholar]
- 21. Kim SJ, Khadka D, Seo JH. Interplay between solid tumors and tumor microenvironment. Front Immunol. 2022;13:882718. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Paskeh MDA, Entezari M, Mirzaei S, et al. Emerging role of exosomes in cancer progression and tumor microenvironment remodeling. J Hematol Oncol. 2022;15:83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Fridman ES, Ginini L, Gil Z. The role of extracellular vesicles in metabolic reprogramming of the tumor microenvironment. Cell. 2022;11:1433. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Xie S, Zhang Q, Jiang L. Current knowledge on exosome biogenesis, cargo‐sorting mechanism and therapeutic implications. Membranes. 2022;12:498. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Lee J, Lee JH, Chakraborty K, Hwang J, Lee YK. Exosome‐based drug delivery systems and their therapeutic applications. RSC Adv. 2022;12:18475‐18492. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Zhang Y, Liu Q, Zhang X, et al. Recent advances in exosome‐mediated nucleic acid delivery for cancer therapy. J Nanobiotechnol. 2022;20:279. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Cheng J, Wang X, Yuan X, Liu G, Chu Q. Emerging roles of exosome‐derived biomarkers in cancer theranostics: messages from novel protein targets. Am J Cancer Res. 2022;12:2226‐2248. [PMC free article] [PubMed] [Google Scholar]
- 28. Yang K, Zhou Q, Qiao B, et al. Exosome‐derived noncoding RNAs: function, mechanism, and application in tumor angiogenesis. Mol Ther Nucleic Acids. 2022;27:983‐997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Nafar S, Nouri N, Alipour M, Fallahi J, Zare F, Tabei SMB. Exosome as a target for cancer treatment. J Invest Med. 2022;70:1212‐1218. [DOI] [PubMed] [Google Scholar]
- 30. Moon B, Chang S. Exosome as a delivery vehicle for cancer therapy. Cell. 2022;11:316. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Waqas MY, Javid MA, Nazir MM, et al. Extracellular vesicles and exosome: insight from physiological regulatory perspectives. J Physiol Biochem. 2022;78:573‐580. [DOI] [PubMed] [Google Scholar]
- 32. Pathania AS, Prathipati P, Challagundla KB. New insights into exosome mediated tumor‐immune escape: clinical perspectives and therapeutic strategies. Biochim Biophys Acta Rev Cancer. 2021;1876:188624. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Noonin C, Thongboonkerd V. Exosome‐inflammasome crosstalk and their roles in inflammatory responses. Theranostics. 2021;11:4436‐4451. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Cheng J, Meng J, Zhu L, Peng Y. Exosomal noncoding RNAs in glioma: biological functions and potential clinical applications. Mol Cancer. 2020;19:66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Aili Y, Maimaitiming N, Mahemuti Y, Qin H, Wang Y, Wang Z. The role of exosomal miRNAs in glioma: biological function and clinical application. Front Oncol. 2021;11:686369. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Quezada C, Torres Á, Niechi I, et al. Role of extracellular vesicles in glioma progression. Mol Aspects Med. 2018;60:38‐51. [DOI] [PubMed] [Google Scholar]
- 37. Wu X, Wang X, Wang J, et al. The roles of exosomes as future therapeutic agents and diagnostic tools for glioma. Front Oncol. 2021;11:733529. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Kelly WJ, Giles AJ, Gilbert M. T lymphocyte‐targeted immune checkpoint modulation in glioma. J Immunother Cancer. 2020;8:e000379. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Ma Q, Long W, Xing C, et al. Cancer stem cells and immunosuppressive microenvironment in glioma. Front Immunol. 2018;9:2924. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Boyd NH, Tran AN, Bernstock JD, et al. Glioma stem cells and their roles within the hypoxic tumor microenvironment. Theranostics. 2021;11:665‐683. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Han C, Zhang C, Wang H, Zhao L. Exosome‐mediated communication between tumor cells and tumor‐associated macrophages: implications for tumor microenvironment. Onco Targets Ther. 2021;10:1887552. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. He C, Li L, Wang L, Meng W, Hao Y, Zhu G. Exosome‐mediated cellular crosstalk within the tumor microenvironment upon irradiation. Cancer Biol Med. 2021;18:21‐33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Xu Z, Zeng S, Gong Z, Yan Y. Exosome‐based immunotherapy: a promising approach for cancer treatment. Mol Cancer. 2020;19:160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Yang E, Wang X, Gong Z, Yu M, Wu H, Zhang D. Exosome‐mediated metabolic reprogramming: the emerging role in tumor microenvironment remodeling and its influence on cancer progression. Signal Transduct Target Ther. 2020;5:242. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Popowski K, Lutz H, Hu S, George A, Dinh PU, Cheng K. Exosome therapeutics for lung regenerative medicine. J Extracell Vesicles. 2020;9:1785161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Kim YK, Choi Y, Nam GH, Kim IS. Functionalized exosome harboring bioactive molecules for cancer therapy. Cancer Lett. 2020;489:155‐162. [DOI] [PubMed] [Google Scholar]
- 47. Zhao X, Wu D, Ma X, Wang J, Hou W, Zhang W. Exosomes as drug carriers for cancer therapy and challenges regarding exosome uptake. Biomed Pharmacother. 2020;128:110237. [DOI] [PubMed] [Google Scholar]
- 48. Li C, Hou X, Zhang P, et al. Exosome‐based tumor therapy: opportunities and challenges. Curr Drug Metab. 2020;21:339‐351. [DOI] [PubMed] [Google Scholar]
- 49. Pandian SRK, Vijayakumar KK, Kunjiappan S, Babkiewicz E, Maszczyk P. Emerging role of exosomes in hematological malignancies. Clin Exp Med. 2022. doi: 10.1007/s10238-022-00850-z. Online ahead of print. [DOI] [PubMed] [Google Scholar]
- 50. Yue B, Yang H, Wang J, et al. Exosome biogenesis, secretion and function of exosomal miRNAs in skeletal muscle myogenesis. Cell Prolif. 2020;53:e12857. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Wu Q, Zhou L, Lv D, Zhu X, Tang H. Exosome‐mediated communication in the tumor microenvironment contributes to hepatocellular carcinoma development and progression. J Hematol Oncol. 2019;12:53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Yang D, Zhang W, Zhang H, et al. Progress, opportunity, and perspective on exosome isolation – efforts for efficient exosome‐based theranostics. Theranostics. 2020;10:3684‐3707. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Song Q, Yu H, Han J, Lv J, Lv Q, Yang H. Exosomes in urological diseases – biological functions and clinical applications. Cancer Lett. 2022;544:215809. [DOI] [PubMed] [Google Scholar]
- 54. Schmid M, Jensen TH. The nuclear RNA exosome and its cofactors. Adv Exp Med Biol. 2019;1203:113‐132. [DOI] [PubMed] [Google Scholar]
- 55. Hou R, Li Y, Sui Z, et al. Advances in exosome isolation methods and their applications in proteomic analysis of biological samples. Anal Bioanal Chem. 2019;411:5351‐5361. [DOI] [PubMed] [Google Scholar]
- 56. Tang Q, Yang S, He G, et al. Tumor‐derived exosomes in the cancer immune microenvironment and cancer immunotherapy. Cancer Lett. 2022;548:215823. [DOI] [PubMed] [Google Scholar]
- 57. Guo W, Qiao T, Dong B, Li T, Liu Q, Xu X. The effect of hypoxia‐induced exosomes on anti‐tumor immunity and its implication for immunotherapy. Front Immunol. 2022;13:915985. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Kumar A, Deep G. Hypoxia in tumor microenvironment regulates exosome biogenesis: molecular mechanisms and translational opportunities. Cancer Lett. 2020;479:23‐30. [DOI] [PubMed] [Google Scholar]
- 59. Hou BR, Jiang C, Wang ZN, Ren HJ. Exosome‐mediated crosstalk between microglia and neural stem cells in the repair of brain injury. Neural Regen Res. 2020;15:1023‐1024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Wan Z, Gao X, Dong Y, et al. Exosome‐mediated cell‐cell communication in tumor progression. Am J Cancer Res. 2018;8:1661‐1673. [PMC free article] [PubMed] [Google Scholar]
- 61. Hu C, Chen M, Jiang R, Guo Y, Wu M, Zhang X. Exosome‐related tumor microenvironment. J Cancer. 2018;9:3084‐3092. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Seo N, Akiyoshi K, Shiku H. Exosome‐mediated regulation of tumor immunology. Cancer Sci. 2018;109:2998‐3004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Kimiz‐Gebologlu I, Oncel SS. Exosomes: large‐scale production, isolation, drug loading efficiency, and biodistribution and uptake. J Control Release. 2022;347:533‐543. [DOI] [PubMed] [Google Scholar]
- 64. Zhao Y, Liu L, Sun R, et al. Exosomes in cancer immunoediting and immunotherapy. Asian J Pharm Sci. 2022;17:193‐205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Li X, Li X, Zhang B, He B. The role of cancer stem cell‐derived exosomes in cancer progression. Stem Cells Int. 2022;2022:9133658. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Panigrahi AR, Srinivas L, Panda J. Exosomes: insights and therapeutic applications in cancer. Transl Oncol. 2022;21:101439. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Zargar MJ, Kaviani S, Vasei M, Soufi Zomorrod M, Heidari Keshel S, Soleimani M. Therapeutic role of mesenchymal stem cell‐derived exosomes in respiratory disease. Stem Cell Res Ther. 2022;13:194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Meng F, Xue X, Yin Z, Gao F, Wang X, Geng Z. Research progress of exosomes in bone diseases: mechanism, diagnosis and therapy. Front Bioeng Biotechnol. 2022;10:866627. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Han L, Zhao Z, Yang K, et al. Application of exosomes in the diagnosis and treatment of pancreatic diseases. Stem Cell Res Ther. 2022;13:153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Mukherjee A, Bisht B, Dutta S, Paul MK. Current advances in the use of exosomes, liposomes, and bioengineered hybrid nanovesicles in cancer detection and therapy. Acta Pharmacol Sin. 2022;43:2759‐2776. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Tan Y, Tang F, Li J, et al. Tumor‐derived exosomes: the emerging orchestrators in melanoma. Biomed Pharmacother. 2022;149:112832. [DOI] [PubMed] [Google Scholar]
- 72. Yu D, Li Y, Wang M, et al. Exosomes as a new frontier of cancer liquid biopsy. Mol Cancer. 2022;21:56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Li S, Chen L. Exosomes in pathogenesis, diagnosis, and treatment of hepatocellular carcinoma, front. Oncologia. 2022;12:793432. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Hao Q, Wu Y, Wu Y, Wang P, Vadgama JV. Tumor‐derived exosomes in tumor‐induced immune suppression. Int J Mol Sci. 2022;23:1461. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Chen L, Wang L, Zhu L, et al. Exosomes as drug carriers in anti‐cancer therapy. Front Cell Dev Biol. 2022;10:728616. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Raju GSR, Pavitra E, Varaprasad GL, et al. Nanoparticles mediated tumor microenvironment modulation: current advances and applications. J Nanobiotechnol. 2022;20:274. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Wang J, Mi S, Ding M, Li X, Yuan S. Metabolism and polarization regulation of macrophages in the tumor microenvironment. Cancer Lett. 2022;543:215766. [DOI] [PubMed] [Google Scholar]
- 78. Jia Z, Jia J, Yao L, Li Z. Crosstalk of exosomal non‐coding RNAs in the tumor microenvironment: novel frontiers. Front Immunol. 2022;13:900155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Liang L, Xu X, Li J, Yang C. Interaction between microRNAs and myeloid‐derived suppressor cells in tumor microenvironment. Front Immunol. 2022;13:883683. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Li Y, Zhao W, Wang Y, Wang H, Liu S. Extracellular vesicle‐mediated crosstalk between pancreatic cancer and stromal cells in the tumor microenvironment. J Nanobiotechnol. 2022;20:208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Zhou H, Wang M, Zhang Y, et al. Functions and clinical significance of mechanical tumor microenvironment: cancer cell sensing, mechanobiology and metastasis. Cancer Commun (Lond). 2022;42:374‐400. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Entezari M, Ghanbarirad M, Taheriazam A, et al. Long non‐coding RNAs and exosomal lncRNAs: potential functions in lung cancer progression, drug resistance and tumor microenvironment remodeling. Biomed Pharmacother. 2022;150:112963. [DOI] [PubMed] [Google Scholar]
- 83. Mun JY, Leem SH, Lee JH, Kim HS. Dual relationship between stromal cells and immune cells in the tumor microenvironment. Front Immunol. 2022;13:864739. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Liu K, Cui JJ, Zhan Y, et al. Reprogramming the tumor microenvironment by genome editing for precision cancer therapy. Mol Cancer. 2022;21:98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Zhao L, Liu Y, Zhang S, et al. Impacts and mechanisms of metabolic reprogramming of tumor microenvironment for immunotherapy in gastric cancer. Cell Death Dis. 2022;13:378. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Yang R, Yu S, Xu T, Zhang J, Wu S. Emerging role of RNA sensors in tumor microenvironment and immunotherapy. J Hematol Oncol. 2022;15:43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Xu Z, Chen Y, Ma L, et al. Role of exosomal non‐coding RNAs from tumor cells and tumor‐associated macrophages in the tumor microenvironment. Mol Ther. 2022;30:3133‐3154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Chen N, Peng C, Li D. Epigenetic underpinnings of inflammation: a key to unlock the tumor microenvironment in glioblastoma. Front Immunol. 2022;13:869307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Tamai S, Ichinose T, Tsutsui T, et al. Tumor microenvironment in glioma invasion. Brain Sci. 2022;12:505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Barthel L, Hadamitzky M, Dammann P, et al. Glioma: molecular signature and crossroads with tumor microenvironment. Cancer Metastasis Rev. 2022;41:53‐75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Hu H, Chen Y, Tan S, et al. The research Progress of antiangiogenic therapy, immune therapy and tumor microenvironment. Front Immunol. 2022;13:802846. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Codrici E, Popescu ID, Tanase C, Enciu AM. Friends with benefits: chemokines, glioblastoma‐associated microglia/macrophages, and tumor microenvironment. Int J Mol Sci. 2022;23:2509. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Xu Y, Zhang H, Sun Q, et al. Immunomodulatory effects of tryptophan metabolism in the glioma tumor microenvironment. Front Immunol. 2021;12:730289. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Berg TJ, Pietras A. Radiotherapy‐induced remodeling of the tumor microenvironment by stromal cells. Semin Cancer Biol. 2022;86:846‐856. [DOI] [PubMed] [Google Scholar]
- 95. Davis L, Recktenwald M, Hutt E, et al. Targeting HIF‐2α in the tumor microenvironment: redefining the role of HIF‐2α for solid cancer therapy. Cancers (Basel). 2022;14:1259. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Mafi S, Mansoori B, Taeb S, et al. mTOR‐mediated regulation of immune responses in cancer and tumor microenvironment. Front Immunol. 2021;12:774103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Sun S, Yu F, Xu D, Zheng H, Li M. EZH2, a prominent orchestrator of genetic and epigenetic regulation of solid tumor microenvironment and immunotherapy. Biochim Biophys Acta Rev Cancer. 2022;1877:188700. [DOI] [PubMed] [Google Scholar]
- 98. Lappano R, Todd LA, Stanic M, et al. Multifaceted interplay between hormones, growth factors and hypoxia in the tumor microenvironment. Cancers (Basel). 2022;14:539. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Takacs GP, Kreiger CJ, Luo D, et al. Glioma‐derived CCL2 and CCL7 mediate migration of immune suppressive CCR2(+)/CX3CR1(+) M‐MDSCs into the tumor microenvironment in a redundant manner. Front Immunol. 2022;13:993444. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. DiVita Dean B, Wildes T, Dean J, et al. Immunotherapy reverses glioma‐driven dysfunction of immune system homeostasis. J Immunother Cancer. 2023;11:e004805. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Ma X, Kuang L, Yin Y, et al. Tumor‐antigen activated dendritic cell membrane‐coated biomimetic nanoparticles with orchestrating immune responses promote therapeutic efficacy against glioma. ACS Nano. 2023;17:2341‐2355. [DOI] [PubMed] [Google Scholar]
- 102. Bu N, Wu H, Zhang G, et al. Exosomes from dendritic cells loaded with chaperone‐rich cell lysates elicit a potent T cell immune response against intracranial glioma in mice. J Mol Neurosci. 2015;56:631‐643. [DOI] [PubMed] [Google Scholar]
- 103. Wang M, Cai Y, Peng Y, Xu B, Hui W, Jiang Y. Exosomal LGALS9 in the cerebrospinal fluid of glioblastoma patients suppressed dendritic cell antigen presentation and cytotoxic T‐cell immunity. Cell Death Dis. 2020;11:896. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Cao Y, Liu B, Cai L, et al. G9a promotes immune suppression by targeting the Fbxw7/Notch pathway in glioma stem cells. CNS Neurosci Ther. 2023. doi: 10.1111/cns.14191. Online ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. Sica A, Strauss L. Energy metabolism drives myeloid‐derived suppressor cell differentiation and functions in pathology. J Leukoc Biol. 2017;102:325‐334. [DOI] [PubMed] [Google Scholar]
- 106. Zöller M. Janus‐faced myeloid‐derived suppressor cell exosomes for the good and the bad in cancer and autoimmune disease. Front Immunol. 2018;9:137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107. Domenis R, Cesselli D, Toffoletto B, et al. Systemic T cells immunosuppression of glioma stem cell‐derived exosomes is mediated by Monocytic myeloid‐derived suppressor cells. PLoS One. 2017;12:e0169932. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Guo X, Qiu W, Liu Q, et al. 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. [DOI] [PubMed] [Google Scholar]
- 109. Zhang X, Liu Y, Dai L, et al. BATF2 prevents glioblastoma multiforme progression by inhibiting recruitment of myeloid‐derived suppressor cells. Oncogene. 2021;40:1516‐1530. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Qi Y, Jin C, Qiu W, et al. The dual role of glioma exosomal microRNAs: glioma eliminates tumor suppressor miR‐1298‐5p via exosomes to promote immunosuppressive effects of MDSCs. Cell Death Dis. 2022;13:426. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Wang M, Zhou Z, Wang X, Zhang C, Jiang X. Natural killer cell awakening: unleash cancer‐immunity cycle against glioblastoma. Cell Death Dis. 2022;13:588. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Ran GH, Lin YQ, Tian L, et al. Natural killer cell homing and trafficking in tissues and tumors: from biology to application. Signal Transduct Target Ther. 2022;7:205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Zhang W, Zhao Z, Li F. Natural killer cell dysfunction in cancer and new strategies to utilize NK cell potential for cancer immunotherapy. Mol Immunol. 2022;144:58‐70. [DOI] [PubMed] [Google Scholar]
- 114. Shah D, Comba A, Faisal SM, et al. A novel miR1983‐TLR7‐IFNβ circuit licenses NK cells to kill glioma cells, and is under the control of galectin‐1. Onco Targets Ther. 2021;10:1939601. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Granhøj JS, Witness Præst Jensen A, Presti M, Met Ö, Svane IM, Donia M. Tumor‐infiltrating lymphocytes for adoptive cell therapy: recent advances, challenges, and future directions. Expert Opin Biol Ther. 2022;22:627‐641. [DOI] [PubMed] [Google Scholar]
- 116. Hiwa R, Brooks JF, Mueller JL, Nielsen HV, Zikherman J. NR4A nuclear receptors in T and B lymphocytes: gatekeepers of immune tolerance(). Immunol Rev. 2022;307:116‐133. [DOI] [PubMed] [Google Scholar]
- 117. Loi S, Michiels S, Adams S, et al. The journey of tumor‐infiltrating lymphocytes as a biomarker in breast cancer: clinical utility in an era of checkpoint inhibition. Ann Oncol. 2021;32:1236‐1244. [DOI] [PubMed] [Google Scholar]
- 118. Paijens ST, Vledder A, de Bruyn M, Nijman HW. Tumor‐infiltrating lymphocytes in the immunotherapy era. Cell Mol Immunol. 2021;18:842‐859. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Zouali M. B lymphocytes, the gastrointestinal tract and autoimmunity. Autoimmun Rev. 2021;20:102777. [DOI] [PubMed] [Google Scholar]
- 120. Pacaud M, Colas L, Brouard S. Microbiota and immunoregulation: a focus on regulatory B lymphocytes and transplantation. Am J Transplant. 2021;21:2341‐2347. [DOI] [PubMed] [Google Scholar]
- 121. Liu ZM, Wang YB, Yuan XH. Exosomes from murine‐derived GL26 cells promote glioblastoma tumor growth by reducing number and function of CD8+T cells. Asian Pac J Cancer Prev. 2013;14:309‐314. [DOI] [PubMed] [Google Scholar]
- 122. Wang M, Jia J, Cui Y, Peng Y, Jiang Y. CD73‐positive extracellular vesicles promote glioblastoma immunosuppression by inhibiting T‐cell clonal expansion. Cell Death Dis. 2021;12:1065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123. Huang S, Xue P, Han X, et al. Exosomal miR‐130b‐3p targets SIK1 to inhibit medulloblastoma tumorigenesis. Cell Death Dis. 2020;11:408. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124. Xue P, Huang S, Han X, et al. Exosomal miR‐101‐3p and miR‐423‐5p inhibit medulloblastoma tumorigenesis through targeting FOXP4 and EZH2. Cell Death Differ. 2022;29:82‐95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Zhang XH, Song YC, Qiu F, Wang ZC, Li N, Zhao FB. Hypoxic glioma cell‐secreted exosomal circ101491 promotes the progression of glioma by regulating miR‐125b‐5p/EDN1. Brain Res Bull. 2023;195:55‐65. [DOI] [PubMed] [Google Scholar]
- 126. Raskov H, Orhan A, Gaggar S, Gögenur I. Neutrophils and polymorphonuclear myeloid‐derived suppressor cells: an emerging battleground in cancer therapy. Oncogenesis. 2022;11:22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127. Langiu M, Palacios‐Acedo AL, Crescence L, Mege D, Dubois C, Panicot‐Dubois L. Neutrophils, cancer and thrombosis: the new Bermuda triangle in cancer research. Int J Mol Sci. 2022;23:1257. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128. Wang J, Tang W, Yang M, et al. Inflammatory tumor microenvironment responsive neutrophil exosomes‐based drug delivery system for targeted glioma therapy. Biomaterials. 2021;273:120784. [DOI] [PubMed] [Google Scholar]
- 129. Liu Z, Yang Z, He L. Effect of miR‐29a‐3p in exosomes on glioma cells by regulating the PI3K/AKT/HIF‐1alpha pathway. Mol Med Rep. 2023;27:72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130. Gao J, Liang Y, Wang L. Shaping polarization of tumor‐associated macrophages In cancer immunotherapy. Front Immunol. 2022;13:888713. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131. Wu D, Liu X, Mu J, Yang J, Wu F, Zhou H. Therapeutic approaches targeting proteins in tumor‐associated macrophages and their applications in cancers. Biomolecules. 2022;12:392. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132. Kumari N, Choi SH. Tumor‐associated macrophages in cancer: recent advancements in cancer nanoimmunotherapies. J Exp Clin Cancer Res. 2022;41:68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133. Baradaran A, Asadzadeh Z, Hemmat N, et al. The cross‐talk between tumor‐associated macrophages and tumor endothelium: recent advances in macrophage‐based cancer immunotherapy. Biomed Pharmacother. 2022;146:112588. [DOI] [PubMed] [Google Scholar]
- 134. Li M, He L, Zhu J, Zhang P, Liang S. Targeting tumor‐associated macrophages for cancer treatment. Cell Biosci. 2022;12:85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135. Pu Y, Ji Q. Tumor‐associated macrophages regulate PD‐1/PD‐L1 immunosuppression. Front Immunol. 2022;13:874589. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136. van der Vos KE, Abels ER, Zhang X, et al. Directly visualized glioblastoma‐derived extracellular vesicles transfer RNA to microglia/macrophages in the brain. Neuro Oncol. 2016;18:58‐69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137. Yang JK, Liu HJ, Wang Y, et al. Exosomal miR‐214‐5p released from glioblastoma cells modulates inflammatory response of microglia after lipopolysaccharide stimulation through targeting CXCR5. CNS Neurol Disord Drug Targets. 2019;18:78‐87. [DOI] [PubMed] [Google Scholar]
- 138. Qian M, Wang S, Guo X, 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. [DOI] [PubMed] [Google Scholar]
- 139. Azambuja JH, Ludwig N, Yerneni SS, Braganhol E, Whiteside TL. Arginase‐1+ exosomes from reprogrammed macrophages promote glioblastoma progression. Int J Mol Sci. 2020;21:3990. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140. Li X, Guan J, Jiang Z, et al. Microglial exosome miR‐7239‐3p promotes glioma progression by regulating circadian genes. Neurosci Bull. 2021;37:497‐510. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141. Xu J, Zhang J, Zhang Z, et al. Hypoxic glioma‐derived exosomes promote M2‐like macrophage polarization by enhancing autophagy induction. Cell Death Dis. 2021;12:373. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142. Hong S, You JY, Paek K, et al. Inhibition of tumor progression and M2 microglial polarization by extracellular vesicle‐mediated microRNA‐124 in a 3D microfluidic glioblastoma microenvironment. Theranostics. 2021;11:9687‐9704. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143. Pan Z, Zhao R, Li B, et al. EWSR1‐induced circNEIL3 promotes glioma progression and exosome‐mediated macrophage immunosuppressive polarization via stabilizing IGF2BP3. Mol Cancer. 2022;21:16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144. Kim HJ, Ji YR, Lee YM. Crosstalk between angiogenesis and immune regulation in the tumor microenvironment. Arch Pharm Res. 2022;45:401‐416. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145. Xu S, Ye C, Chen R, Li Q, Ruan J. The landscape and clinical application of the tumor microenvironment in Gastroenteropancreatic neuroendocrine neoplasms. Cancers (Basel). 2022;14:2911. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146. Kim SK, Cho SW. The evasion mechanisms of cancer immunity and drug intervention in the tumor microenvironment. Front Pharmacol. 2022;13:868695. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147. Wang L, Chard Dunmall LS, Cheng Z, Wang Y. Remodeling the tumor microenvironment by oncolytic viruses: beyond oncolysis of tumor cells for cancer treatment. J Immunother Cancer. 2022;10:e004167. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148. Zhang A, Miao K, Sun H, Deng CX. Tumor heterogeneity reshapes the tumor microenvironment to influence drug resistance. Int J Biol Sci. 2022;18:3019‐3033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149. Li G, Lan Q. Exosome‐mediated transfer of circ‐GLIS3 enhances temozolomide resistance in glioma cells through the miR‐548m/MED31 axis. Cancer Biother Radiopharm. 2023;38:62‐73. [DOI] [PubMed] [Google Scholar]
- 150. Guo X, Qiu W, Wang J, 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. [DOI] [PubMed] [Google Scholar]
- 151. Qiu W, Guo X, Li B, et al. Exosomal miR‐1246 from glioma patient body fluids drives the differentiation and activation of myeloid‐derived suppressor cells. Mol Ther. 2021;29:3449‐3464. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152. Zhu L, Kalimuthu S, Oh JM, et al. Enhancement of antitumor potency of extracellular vesicles derived from natural killer cells by IL‐15 priming. Biomaterials. 2019;190‐191:38‐50. [DOI] [PubMed] [Google Scholar]
- 153. Zhang Z, Xu J, Chen Z, et al. Transfer of MicroRNA via macrophage‐derived extracellular vesicles promotes proneural‐to‐mesenchymal transition in glioma stem cells, cancer. Immunol Res. 2020;8:966‐981. [DOI] [PubMed] [Google Scholar]
- 154. Yao J, Wang Z, Cheng Y, et al. M2 macrophage‐derived exosomal microRNAs inhibit cell migration and invasion in gliomas through PI3K/AKT/mTOR signaling pathway. J Transl Med. 2021;19:99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155. Li Z, Meng X, Wu P, et al. Glioblastoma cell‐derived lncRNA‐containing exosomes induce microglia to produce complement C5, promoting chemotherapy resistance. Cancer Immunol Res. 2021;9:1383‐1399. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
The data in the current study are available from the corresponding authors upon reasonable request.