Skip to main content

Exosomes as drug delivery systems in glioma immunotherapy

Abstract

Recently, the significant benefits of cancer immunotherapy for most cancers have been demonstrated in clinical and preclinical studies. However, the efficacy of these immunotherapies for gliomas is limited, owing to restricted drug delivery and insufficient immune activation. As drug carriers, exosomes offer the advantages of low toxicity, good biocompatibility, and intrinsic cell targeting, which could enhance glioma immunotherapy efficacy. However, a review of exosome-based drug delivery systems for glioma immunotherapy has not been presented. This review introduces the current problems in glioma immunotherapy and the role of exosomes in addressing these issues. Meanwhile, preparation and application strategies of exosome-based drug delivery systems for glioma immunotherapy are discussed, especially for enhancing immunogenicity and reversing the immunosuppressive tumor microenvironment. Finally, we briefly describe the challenges of exosome-based drug delivery systems in clinical translation. We anticipate that this review will guide the use of exosomes as drug carriers for glioma immunotherapy.

Graphical Abstract

Introduction

Gliomas are tumors derived from glial or neural precursor cells in the central nervous system (CNS), including astrocytoma, oligodendroglioma, ventricular meningioma, and mixed glioma. According to recent data from The Central Brain Tumor Registry of the United States, gliomas account for 24% of all CNS tumors and 80.9% of malignant tumors, making them the most prevalent intracranial malignancy [1]. The World Health Organization categorizes gliomas into four grades (grades 1–4), with grades 1–2 considered low-grade gliomas and grades 3–4 classified as high-grade gliomas [2]. Low-grade gliomas are associated with a 10-year survival rate of 39% and a median survival time of 87.3 months [3]. Conversely, high-grade gliomas, particularly glioblastoma (GBM), demonstrate high malignancy, with a median survival time not exceeding 15 months [4]. Owing to their high recurrence rate, high disability and mortality rates, and limited cure rate, gliomas are the most aggressive and refractory CNS tumors. Currently, the standard treatment for gliomas involves surgical intervention combined with chemoradiotherapy; however, this approach is inefficient in preventing tumor recurrence. Therefore, it is critical to explore innovative treatment approaches to enhance therapeutic efficacy.

In recent years, tumor immunotherapy has gained prominence for its ability to restore and enhance immune responses, through exogenous intervention with the body’s immune system. The tumor immune cycle includes the release of specific antigens by tumor cells, which are captured and processed by antigen-presenting cells (APCs) for presentation to T cells. These T cells are then activated, and they proliferate and infiltrate tumors, ultimately recognizing and eliminating tumor cells [5]. Tumor immunotherapy can specifically kill tumor cells and establish long-term immune memory by facilitating any step in this process. Common cancer immunotherapies include immune checkpoint inhibitors (ICIs), chimeric antigen receptor T cell therapy, cancer vaccines, and oncolytic viruses, and these are currently the subject of many clinical trials [6]. However, patients with glioma derive limited benefits from immunotherapy compared to those with other malignancies [7]. Several factors contribute to this, including (i) the blood–brain barrier (BBB) preventing the entry of therapeutic molecules, (ii) the low immunogenicity of gliomas limiting the initiation of immune responses, and (iii) the immunosuppressive microenvironment inhibiting the activity of effector T cells. To address these challenges, researchers are developing drug delivery systems capable of efficiently delivering therapeutic agents, such as antigens, immunomodulatory molecules, and anticancer drugs. These systems aim to penetrate the BBB precisely and enhance the immune response, thereby improving the efficacy of glioma immunotherapy.

Common drug delivery systems mainly comprise nanoformulations, including liposomes, solid lipid nanoparticles, dendritic polymers, and inorganic materials [8, 9]. However, these nanoformulations are identified as “non-self” and are susceptible to capture and rapid clearance by the immune system. Additionally, nanocarriers synthesized from inorganic materials can have potential toxicity owing to their inability to be degraded [10]. In contrast, exosomes are endogenous nanoscale vesicles secreted by living cells, with several advantages, including excellent stability, low toxicity, a long circulating half-life, ease of modification, and intrinsic cell targeting. Most notably, exosomes are vital in mediating cellular communication within the tumor immune response. In addition, they participate in presenting immune signals, regulating the tumor microenvironment (TME), and influencing immune cell responses [11]. Therefore, exosomes have great potential as carriers for drug delivery in glioma immunotherapy. However, a review on this topic has not been presented. In this review, we introduce the problems in glioma immunotherapy and the advantages of exosomes in it, and we discuss the preparation of exosomes as drug delivery systems and the strategies for their application in glioma immunotherapy to provide new ideas for realizing efficient glioma treatment.

Current problems in glioma immunotherapy

Blood–brain barrier

The BBB is a neurovascular unit consisting of brain microvascular endothelial cells (BMECs), basement membrane, astrocytes, microglia, and pericytes [12]. Among these components, BMECs, which form the primary structure of the BBB, display a flattened cell morphology, limited caveolae on the luminal surface, abundant mitochondria, and tight intercellular junctions [13]. This unique physical barrier permits only lipophilic molecules with molecular weights less than 400 Dalton and fewer than eight hydrogen bonds to traverse the BBB along the concentration gradient [14]. In addition, highly active intracellular and extracellular enzymes on the BBB prevent drugs, such as proteins and peptides, from passing through the BBB by degrading them [15]. Furthermore, various transmembrane transporter proteins, particularly ATP-binding cassette transporters, limit the accumulation of certain small-molecule drugs by expelling them into the capillary lumen [16, 17]. Given these constraints, the BBB not only protects the brain from foreign pathogens but also blocks nearly all large-molecule drugs and approximately 98% of small-molecule drugs from reaching the treatment site, while simultaneously controlling peripheral immune cell infiltration into the brain [18, 19] (Fig. 1a). As tumors progress, BBB disruption can lead to the formation of a blood–brain tumor barrier characterized by a looser vascular system, diminished tight junctions, an abnormal pericyte distribution, an altered astrocyte end-foot, and reduced neuronal connectivity [20]. In a sense, BBB disruption might facilitate the entry of drugs and immune factors into tumors. However, the blood–brain tumor barrier still expresses efflux transporters [20], and clinical data indicate that in all patients with GBM, an intact BBB is retained in the tumor region [21]. Thus, drugs that effectively penetrate the BBB are essential for glioma treatment.

Tumor low immunogenicity

The tumor mutational burden (TMB) indirectly reflects the ability of tumors to produce neoantigens and can be used to predict the therapeutic efficacy of ICIs. A higher TMB suggests that the patient is more likely to benefit from ICIs [22]. However, GBM exhibits a low TMB and neoantigen burden, resulting in limited therapeutic targets for the immune system [23, 24] (Fig. 1b). By evaluating changes in the neoantigen load and immunologic characteristics in patients with primary and recurrent gliomas, Neio et al. [25] found no difference in the number of expressed neoantigens (e-) and predicted neoantigens (p-); however, their ratio (e-/p-) was significantly decreased in patients with recurrent gliomas. They concluded that the reduced expression of mutant neoantigens and impaired antigen processing and presentation could cause tumor immune escape and recurrence. Furthermore, beyond mutated neoantigens, non-mutated shared antigens, such as IL13Rα2, HER2, WT1, and EphA2, are commonly found in most patients with glioma. However, these antigens are expressed at low and highly variable levels compared to those in malignant tumors such as melanoma [26, 27]. At the same time, these non-mutated antigens are also usually expressed in normal tissues [28], increasing the risk of complications when targeting them for treatment.

Additionally, tumor antigens necessitate the formation of mature peptide–major histocompatibility complexes (MHCs) through antigen processing and presentation machinery (APM) for T cell recognition. Previous research has shown that glioma cell migration and invasion are correlated with downregulated MHC class I and II antigen expression [29]. In developing APVAC2 for the GAPVAC-101 clinical trial, based on a vaccine derived from glioma-associated mutant peptides, researchers performed extensive immunopeptidome analysis. They identified 35,156 distinct human leukocyte antigens (HLAs) but found no mutant peptides presented by HLA in the plasma or tissue from patients with GBM [30, 31]. Moreover, Facoetti et al. observed that more than 50% of patients with GBM were deficient in HLA class I and II molecules, with a significant correlation between the loss of HLA class I and a higher tumor grade. Furthermore, the expression of tapasin, a key component of the APM that facilitates MHC class I interactions with antigen-processing transporters, was determined to be reduced in several GBM specimens. This indicates that abnormalities in HLA and APM might contribute to the limited efficacy of immunotherapy for malignant gliomas [32].

Immunosuppressive microenvironment

Tumor cell invasion into normal tissues establishes the TME, which comprises immune cells, cancer-associated fibroblasts, endothelial cells, and an extracellular matrix [33, 34]. In the TME, densely infiltrating immunosuppressive cells and cytokines, along with hypoxia and chronic inflammation, synergistically inhibit T cell responses, facilitate tumor immune escape, and adversely affect glioma immunotherapy [35, 36] (Fig. 1c). Tumor-associated macrophages (TAMs) constitute 40% of the total tumor volume and play a crucial role in the TME [37]. Accordingly, high TAM infiltration is typically linked to reduced overall survival in patients with GBM [38]. TAMs secrete TGF-β, targeting cytotoxic T lymphocytes (CTLs) and specifically suppressing the expression of perforin, granzyme A, granzyme B, Fas ligand, and IFN-γ, consequently diminishing CTL-mediated cytotoxicity [39]. Moreover, GBM-induced kynurenine activates the aryl hydrocarbon receptor in TAMs, promoting CD39 expression, which synergizes with CD73 to produce adenosine, leading to CD8+ T cell dysfunction [40]. Furthermore, TAMs disrupt T cell metabolism through the overexpression of arginase (ARG1) and indoleamine 2,3-dioxygenase (IDO), catabolizing amino acids that are vital for T cell proliferation [41]. Similarly to that observed in tumor cells, TAMs express inhibitory surface molecules, such as PD-L1, PD-L2, B7H4, and CD80/CD86, which hinder T cell activation and even trigger apoptosis [42]. Moreover, the LILRB1 inhibitory receptor on TAMs binds to MHC I on cancer cells, directly shielding them from immune-mediated phagocytosis [43].

Myeloid-derived suppressor cells (MDSCs) represent a diverse group of immature myeloid cells (IMCs) known for their immunosuppressive capabilities [44]. Typically, IMCs originating from the bone marrow rapidly differentiate into mature macrophages, granulocytes, and dendritic cells (DCs) [45, 46]. In contrast, the maturation of IMCs is hindered, leading them to transform into MDSCs with immunosuppressive functions in tumors [47]. MDSCs can pleiotropically inhibit CTL activity through the expression of IL-4Rα, PD-L1, and CD80 and the upregulation of ARG1 and inducible nitric oxide synthase (iNOS) expression [48]. Research has further shown that systemic lymphopenia following radiotherapy is correlated with an increase in MDSC numbers and that targeting MDSCs is effective for mitigating immunosuppression and lymphopenia in patients with GBM patients [49]. Moreover, MDSCs exert immunosuppressive effects by inhibiting the functions of DCs, natural killer (NK) cells, and macrophages, in addition to inducing the expansion and differentiation of regulatory T cells (Tregs).

Tregs are a subpopulation of CD4+ T cells that usually express the transcription factor Foxp3. The tumor tissues and peripheral blood of patients with glioma have a higher proportion of Tregs compared to that in healthy individuals [50], with a higher glioma grade corresponding to more significant Treg enrichment [51]. Tregs can suppress anti-tumor immune responses through a variety of mechanisms [52,53,54], including (i) the secretion of immunosuppressive cytokines, (ii) the induction of target cell lysis via cell–cell contact, (iii) the expression of inhibitory receptors to suppress APCs, (iv) competition with effector T cells for IL-2 to inhibit their growth, and (v) the promotion of adenosine accumulation and competition for nutrients to disrupt T cell metabolism. Recently, van Hooren et al. [55] reported that CD103+ Tregs with upregulated lipid metabolism are accumulated in the TME and significantly inhibit CTL activation after radiotherapy combined with ICIs. The depletion of Tregs results in tertiary lymphoid structure formation, which in turn induces responses in CTLs and improves treatment resistance. Therefore, there is an urgent need to develop therapies targeting immunosuppressive cells to reverse the immunosuppressive microenvironment.

Fig. 1
figure 1

Current problems in glioma immunotherapy (By Figdraw). a Blood-brain barrier prevents the entry of therapeutic molecules. b Low immunogenicity of gliomas restricts immune response initiation. c Immunosuppressive microenvironment inhibits T cell activity

Overview of exosomes

Biogenesis and characteristics of exosomes

Exosomes are double-membrane structured extracellular vesicles with a 40–160 nm diameter formed by outgrowth of the plasma membrane. Most cells can actively secrete exosomes, which are naturally present in various body fluids such as blood, saliva, urine, and cerebrospinal fluid [56]. Exosome generation involves double invagination of the plasma membrane and the formation of intracellular multivesicular bodies (MVBs) containing intraluminal vesicles (ILVs) (Fig. 2a(i)). Specifically, cell membranes undergo endocytosis to form early sorting endosomes, which then mature and further invaginate to form MVBs. Subsequently, MVBs can either be degraded by lysosomes or autophagosomes or fuse with the plasma membrane, thereby releasing ILVs as exosomes [57]. Exosome biogenesis generally involves two potential mechanisms: endosomal sorting complex required for transport (ESCRT) and ESCRT-independent pathway. The ESCRT pathway relies on the synergistic of the ESCRT complexes (ESCRT-0, ESCRT-I, ESCRT-II, and ESCRT-III) and the AAA ATPase Vps4 for recognition sorting of ubiquitinated proteins to maintain the specificity of the contents of ILVs [58]. The ESCRT-independent pathway is driven by partial lipids. Sphingomyelins produce ceramides via neutral sphingomyelinase 2, and ceramides form lipid raft microdomains to trigger the formation of ILVs while promoting exosome release [59].

Depending on the parental cell of origin, exosomes can carry a combination of different bioactive molecules, including proteins, nucleic acids, and lipids (Fig. 2a(ii)) [60]. Among these molecules, proteins constitute the most abundant component of exosomes and carry a wide range of functions. Synaptosomal-associated proteins, annexins, and Rab proteins are responsible for intracellular membrane fusion and trafficking. Heat shock proteins such as Hsp70 and Hsp90 are involved in antigen presentation and immunomodulation, while proteins like Alix and TSG101 are essential for MVB biogenesis. Tetratransmembrane proteins (CD9, CD36, CD81, and CD82) and integrins mediate cell infiltration, adhesion, and fusion. Transporter proteins (ATP7A, ATP7B, SLC16A1, and CL1C1) and receptors (CD46, CD55) facilitate cell communication and molecular transport [61,62,63]. Another vital cargo of exosomes is nucleic acids, including messenger RNAs (mRNAs), microRNAs (miRNAs), circular RNAs (circRNAs), and long non-coding RNAs (lnRNAs). mRNAs mediate genetic information transfer and regulate receptor cell function and protein synthesis [64]. miRNAs can inhibit the translation of target mRNAs or promote their degradation to regulate gene expression, affecting a wide range of biological processes, such as cell proliferation, differentiation, apoptosis, and disease development [65]. circRNAs and lnRNAs can be selectively packaged, secreted, and transferred between cells in exosomes to regulate cancer angiogenesis, immune escape, invasive metastasis, and epithelial-mesenchymal transition [66, 67]. Furthermore, exosomes contain diverse lipids, such as ceramides, gangliosidesphosphatidylserine, cholesterol, sphingomyelins, phosphatidylcholine, and phosphatidylserine. These lipid components not only keep the morphology and structure of exosomes stable but also play a crucial role in the production and release of exosomes [68, 69].

Biological function of exosomes

The diversity of the molecular composition of exosomes endows them with a wide range of biological functions that are important for maintaining the internal homeostasis of organisms, influencing the course of diseases, and developing new therapeutic approaches. First, intercellular signaling and communication are critical biological functions of exosomes. Exosomes are taken up by recipient cells through receptor-ligand interactions, membrane fusion, or endocytosis, delivering the bioactive molecules and thus modulating the functional and phenotypic characteristics of the recipient cells [70, 71]. For example, exosomes loaded with the hypoxia master regulator HIF1A transmit hypoxic signals to normoxic cells, thereby inducing neoangiogenesis [72]. Glioma-derived exosomes (GM-Exos) induce the conversion of normal human astrocytes to tumor-associated astrocytes via the miR-3065-5p/DLG2 signaling axis and promote malignant tumor progression [73]. Additionally, exosomes can modulate inflammation by inhibiting or promoting the activation of inflammasomes. Studies have shown that M2-like macrophage exosomes carrying miR-148a inactivate the TLR4/NF-κB/NLRP3 signaling pathway to alleviate myocardial ischemia/reperfusion injury [74], while macrophage-derived exosomes treated with lipopolysaccharides promote acute liver injury by activating the NLRP3 inflammasome [75]. The role of exosomes in immune responses has also been extensively studied. Depending on their molecular composition and the specific cellular environment in which they reside, exosomes can enhance immune responses or induce immune tolerance in their communication with immune cells, demonstrating their potential therapeutic role in cancer, autoimmune diseases, inflammatory diseases, and transplant rejection [76, 77]. Moreover, exosomes exhibit the ability to promote tissue repair and regeneration. In particular, mesenchymal stem cell-derived exosomes can stimulate vascular regeneration, resist apoptosis and fibrosis, enhance neuronal survival, and improve extracellular matrix remodeling [78, 79]. Numerous studies have demonstrated that mesenchymal stem cell-derived exosomes show protective and therapeutic benefits in wound healing, bone and cartilage regeneration, nerve repair, and myocardial injuries [80, 81]. Meanwhile, exosomes can carry specific molecules that reflect the pathophysiological state of their source cells, thus serving as biomarkers for early diagnosis and monitoring of diseases [82]. Finally, based on their molecular transfer function, good biocompatibility, stability, and targeting properties, exosomes have been widely investigated for drug delivery [83].

Status of exosomes for glioma treatment

In recent years, researchers’ in-depth exploration of exosomes’ composition and function has facilitated their remarkable progress in glioma treatment, mainly divided into four directions. First, inhibiting the release or uptake of GM-Exos to weaken their role in promoting glioma malignant progression. Several potential therapeutic targets have been identified, including ubiquitin-conjugating enzyme E2O, heparan sulfate proteoglycans, and the mammalian target of rapamycin [84, 85]. Second, exosomes’ inherent properties and contents can be utilized to treat gliomas. For example, mesenchymal stem cell-derived exosomes have shown the potential to treat gliomas independently of other therapies [86, 87]. Microglia-derived exosomes stimulated by lipopolysaccharides/INF-γ can also inhibit glioma growth by altering the phenotype of TAMs [88]. Third, exosomes can be used as biomarkers to monitor treatment efficacy and predict prognosis. Studies have demonstrated that certain bioactive molecule levels (e.g., circ_0072083, lncSBF2-AS1, and miR-1238) in the exosomes of glioma patients correlate with the level of temozolomide resistance and treatment efficacy [89,90,91]. In addition, circulating extracellular vesicle levels can reflect overall survival, progression-free survival, degree of tumor resection, and postoperative recurrence in glioma patients [92]. Finally, and more importantly, exosomes can deliver nucleic acids, drugs, and proteins to treat gliomas, including anti-angiogenesis, inhibiting tumor growth and metastasis, reversing drug resistance, and enhancing immune responses [18]. In the following subsection, we will focus on the advantages of exosomes as carriers in glioma immunotherapy.

Advantages of exosomes as drug delivery systems in glioma immunotherapy

BBB-penetration capability

The ability of exosomes to penetrate the BBB and enter the brain has been demonstrated. Banks et al. [93] found that exosomes derived from different species (human and mouse) and cell lines (cancerous and noncancerous) can penetrate the BBB, with penetration rates varying more than 10-fold. Here, lipopolysaccharide and wheatgerm agglutinin were found to regulate the transport of most exosomes, and the mannose 6-phosphate receptor was determined to be a potential receptor facilitating the movement of mouse macrophage exosomes across the BBB. In addition, the investigators outlined possible transport pathways utilized by exosomes to penetrate the BBB, such as macropinocytosis, lipid rafts or nonspecific exosome–endothelial cell interactions, and receptor-mediated transcytosis [94, 95] (Fig. 2b). Although the detailed mechanisms underlying exosome entry into the brain need to be fully elucidated, several studies have indicated that the binding of specific ligands on the surfaces of exosomes to brain endothelial cell receptors facilitates their passage across the BBB. For example, both in vitro and in vivo models have been used to confirm that naïve macrophage-derived exosomes are internalized by BMECs through interactions between surface integrin lymphocyte function-associated antigen 1 and intercellular adhesion molecule 1 (ICAM-1) [96]. Moreover, neural stem cell-derived exosomes that bind heparan sulfate proteoglycan receptors are endocytosed by BMECs and cross the BBB [97]. Further, serum exosomes exhibit natural brain-targeting abilities through transferrin–transferrin receptor interactions [98]. CD46 is one of the primary receptors utilized by BMECs to take up exosomes derived from brain metastatic cancer cell lines and promote cancer metastasis [99]. In addition, different pathologic conditions enhance the uptake of exosomes by the BBB. Inflammation causes the overexpression of ICAM-1 receptors on the BBB, and macrophage exosomes enter the inflamed brain 3.1-fold faster and accumulate 5.8-fold more than in a healthy brain [96]. Hypoxia triggers intracellular calcium release, modifies connexins to disrupt the BBB, and leads to the significant leakage of exosomes into the brain [100]. The brain-targeting capabilities of both natural and engineered exosomes have led to their excellent therapeutic efficacy in models of CNS diseases, including Alzheimer’s disease, Parkinson’s disease, stroke, and epilepsy [94]. Therefore, exosomes offer broad clinical applications for the delivery of drugs to treat CNS diseases and hold promise for overcoming the challenges to glioma immunotherapy posed by the BBB.

Fig. 2
figure 2

Exosome biogenesis, characteristics, and mechanism of crossing the BBB (By Figdraw). a Exosome biogenesis and characteristics. (i) Exosome biogenesis; (ii) Exosome characteristics. b Mechanism of exosome crossing the BBB. (i) Receptor-mediated transcytosis; (ii) Lipid rafts or nonspecific exosome-endothelial cell interactions; (iii) Macropinocytosis

Immunomodulatory capability

Exosomes derived from tumor and immune cells have different essential functions in terms of shaping the cancer immune cycle, including antigen processing and presentation, antigen transfer, innate and adaptive immune activation, and immune suppression [101] (Fig. 3). Clarifying the intrinsic immunomodulatory capacity of exosomes derived from different cells and using exosomes as carriers to initiate anticancer immune responses or deliver anticancer drugs could help to further improve drug efficacy.

Tumor-derived exosomes (TEXs) carry specific tumor antigens, antigen-presenting molecules, and co-stimulatory molecules that activate anti-tumor immune responses. For example, TEXs carrying HSP70 induce NK cell-specific migration and cytolytic activity [102]. Meanwhile, TEXs expressing Rab27 enhance the expression of MHC II and the co-stimulatory molecules CD80 and CD86 in DCs, inducing DC maturation and promoting T cell proliferation [103]. However, TEXs encapsulate different contents, contributing to the complexity of their functions. TEXs can disrupt host immunity by inhibiting antigen recognition and presentation by DCs [104], promoting MDSC activation and differentiation [105], inducing the polarization of TAMs toward an immunosuppressive phenotype [106], and interfering with T cell proliferation and activation [107, 108]. Furthermore, in addition to promoting tumor immune escape, TEXs are also involved in various aspects of tumor progression by communicating with adjacent or distant cells, including the establishment of pre-metastatic niches, anti-apoptosis, angiogenesis, promotion of inflammatory response, tumor growth, and drug resistance [109, 110]. Consequently, the safety of utilizing TEXs as carriers warrants further examination.

Exosomes derived from immune cells serve as crucial mediators of both innate and adaptive immune responses. DC-derived exosomes (DEXs), enriched in MHC I, MHC II, CD80, CD86, ICAM-1, HSP70, and HSP90, play a key role in antigen presentation, thereby triggering immune responses based on both CD4+ and CD8+ T cells [111]. Additionally, DEXs transport functional peptide–MHC complexes to adjacent unresponsive DCs, enhancing the proliferation of DCs with specific peptides to facilitate widespread T cell activation and amplify immune responses [112]. Furthermore, DEXs express IL-15Rα, NKG2D ligand, and the TNF superfamily ligand and can directly bind to NK cell surface receptors and induce NK cell activation [113, 114]. T cell-derived exosomes have different roles in immunomodulation depending on their phenotype. For example, exosomes secreted by antigenically fully activated CD8+ T cells induce the activation of low-affinity CD8+ T cells, which are involved in tumor cell killing [115]. Moreover, CD4+ T cell-derived exosomes mediate anti-tumor cellular immunity, by activating CD8+ T cells, and enhance humoral immunity by promoting B cell proliferation, activation, and antibody production [116, 117]. In contrast, Treg exosomes express CD73 [118], carry specific miRNAs and iNOS [119], and synergize with cytokines to suppress the immune response [120]. Another study showed that exosomes from M1-like macrophages partially deliver phagocytosed antigens to DCs via a ceramide-dependent pathway, thus enhancing T cell responses [121]. NK cell exosomes carrying several specific miRNAs (miR-10b-5p, miR-92a-3p, and miR-155-5p) target molecules involved in the Th1 immune response and increase antigen presentation and co-stimulation through monocyte and monocyte-derived DC activation [122]. In addition, the roles of exosomes from B cells, mast cells, and neutrophils in the tumor immune response have been established [11, 123].

Fig. 3
figure 3

Immunomodulatory capability of tumor cell and immune cell-derived exosomes (By Figdraw)

Preparation of exosomes as drug delivery systems

Isolation and purification

Given the characteristics of exosomes, such as their mass density, particle size, and surface protein markers, various strategies are available to isolate them from biofluids or cell culture supernatants. These include ultracentrifugation (UC), density gradient centrifugation (DG), ultrafiltration (UF), size exclusion chromatography (SEC), immunoaffinity capture, polymer precipitation, and microfluidics (Table 1).

UC is the most commonly used exosome isolation method and is the “gold standard” [124]. It involves two steps, as follows: (1) low-to-medium-speed centrifugation to remove cells, cellular debris, and large vesicles, and (2) ultra-high-speed centrifugation at 100,000 × g to obtain exosomes. UC is simple, technologically established, and suitable for processing large-dose samples. However, exosomes can be damaged by the high centrifugal forces, and impurities of a similar size and density might be co-separated with them [125,126,127]. Consequently, to enhance exosome purity, researchers developed DG through the introduction of separation media, such as sucrose and iodixanol. However, this method is cumbersome and time-consuming, limiting its efficiency [128]. Based on a difference in particle size, exosomes can be separated via UF or SEC. UF selectively separates samples through ultrafiltration membranes with different molecular-weight cutoffs. Moreover, it is a simple and low-cost method, but the membranes are susceptible to clogging, which causes exosome loss, and shear forces can damage the exosomes [129]. The principle of SEC is that large molecules cannot enter the gel pores and are eluted along the porous gel voids, while small molecules enter the gel pores and are eluted for a longer time. SEC is based on the principle of gravity flow, and the resulting exosomes retain a high degree of structural integrity and biological activity, with better recovery and purity than those with UC [130, 131]. However, the co-elution of similar-sized lipoproteins and the need for additional exosome enrichment pose challenges for the large-scale commercial application of SEC [132]. Immunoaffinity capture can be used to isolate exosomes based on interactions between their marker proteins (e.g., CD9, CD63, CD81, and ALIX, etc.) and specific antibodies. In this method, only exosomes that express proteins recognized by the antibody can be captured, and thus, the yield is low and isolation depends on the quality and specificity of the antibody. However, compared to other methods, immunoaffinity capture yields exosomes with a higher purity and can be used to isolate specific subtypes of exosomes [133, 134]. In addition, the binding of polyethylene glycol to water molecules and the formation of a hydrophobic microenvironment reduces exosome solubility and allows them to precipitate [135]. This polymer precipitation method is simple, but the exosomes obtained have more impurities and the polymer is difficult to remove [136]. Finally, microfluidics comprise a micro-sized horizontal separation technique based on the physicochemical characteristics of exosomes, and this has advantages over conventional methods, including its automated operation, precise control of flow conditions, high purity, and short time. However, microfluidics approaches require expensive and sophisticated equipment and are not suitable for large-scale applications [137, 138]. In conclusion, we should flexibly choose one or more methods for exosome isolation depending on the sample types, sources, volumes, and downstream applications, in addition to considering the yield, purity, cost, and time.

Table 1 Comparison of exosome isolation and purification methods

Drug loading

The process of loading drugs into exosomes is crucial for the development of exosome-based drug delivery systems. Maintaining drug activity and effective loading without compromising exosome integrity is the primary concern when selecting the optimal loading method. Drug loading strategies for exosomes can be primarily divided into endogenous and exogenous. Subsequently, the characteristics of these different loading methods will be discussed.

Endogenous loading refers to therapeutic molecules being imported into the source cells via transfection, co-incubation, or electroporation and then loaded into exosomes through a cell sorting mechanism. Endogenous loading has a wide range of applications and does not destroy exosomes, but the process is complex and time-consuming [139, 140]. Crucially, with endogenous loading, control over exosome yields and therapeutic molecule encapsulation is lacking, generally resulting in low loading efficiency [141]. Recently, Yang et al. [142] introduced a cellular-nanoporation technique for the large-scale production of exosomes encapsulating therapeutic mRNAs and targeted peptides. Compared to results with electroporation and other exosome production strategies, this method can be used to produce up to 50-fold more exosomes, increase mRNA transcripts by more than 103-fold, and result in better therapeutic efficacy based on glioma models.

In exogenous loading, the exosome is first isolated, and then, the therapeutic molecule is directly loaded onto the exosome membrane or into the lumen. This method offers controllable encapsulation efficiency and loading capacity, generally outperforming endogenous loading [143]. Various loading methods have been developed, such as incubation, freeze–thaw, sonication, extrusion, electroporation, and permeabilization. Incubation is used to passively load drugs via concentration gradients without compromising the integrity of the exosome membrane. However, this still mainly applies to hydrophobic small molecules, and the loading efficiency is affected by the drug concentration, pH, and temperature, among other factors [144, 145]. For freeze–thaw, rapid temperature changes are employed for active drug loading into exosomes; it is simple but has a low loading rate and can lead to exosome aggregation [143, 146]. In contrast, sonication and extrusion result in higher loading efficiencies [147, 148], but they can damage the exosome membrane and reduce drug activity [149]. Similarly, electroporation causes the formation of several temporary hydrophilic pores on the exosome membrane using an applied electric field, permitting the entry of drugs into the exosome. Studies have shown that the loading efficiency of electroporation is three times that of incubation but much less than that of sonication and that it causes exosome damage and aggregation [145, 148, 150]. Surfactants, such as saponins, enhance exosome membrane permeability for drug loading, which is known as the permeabilization method. However, surfactants can alter drug activity and are associated with a risk of hemolysis and cytotoxicity, necessitating further purification [144, 146, 151]. In addition, dialysis [152] and in situ synthesis [153] are also employed for drug loading, albeit less commonly than other methods. Table 2 outlines the principles, advantages, disadvantages, and applicable types of therapeutic molecules of commonly used drug loading methods.

Table 2 Comparison of exosome drug loading methods

Exosome-based immunotherapy strategies for gliomas

To address the challenges associated with glioma immunotherapy, we propose strategies using exosome delivery systems to boost immune efficacy by enhancing immunogenicity and reversing the immunosuppressive microenvironment (Fig. 4). Additionally, we provide a summary of the types and effectiveness of current exosome-based drug delivery systems for glioma immunotherapy (Table 3).

Fig. 4
figure 4

Exosome-based immunotherapy strategies for gliomas (By Figdraw). a Improvement of tumor low immunogenicity. (i) Exosomes precisely deliver chemotherapeutic drugs to promote antigen release and trigger immunogenic cell death; (ii) Exosomes from tumor and immune cells as cancer vaccines to enhance antigen expression and presentation. b Improvement of the immunosuppressive microenvironment, including reprogramming TAMs, targeting MDSCs/Tregs, and improving TME physicochemical properties

Cascade modulation to improve low tumor immunogenicity

Targeted delivery of chemotherapeutic agents to promote antigen release

Tumor cell death resulting in the release of sufficient antigens is the basis for triggering specific immune responses. Chemotherapy can kill tumor cells, produce large amounts of antigens, and induce immunogenic cell death (ICD). However, the inefficient targeted delivery of chemotherapeutic agents impairs their cytotoxic effects on tumors, leading to reduced antigen release and limiting the effects of ICD, which can even cause other severe side effects in organs. Therefore, exosome-based drug delivery systems could be a promising strategy for enhancing the efficacy of chemotherapeutic agents.

Transmembrane proteins on the surfaces of exosomes (such as integrins, tetraspanins, and ICAM-1) confer intrinsic cell- or tissue-targeting properties to exosomes [101]. For example, exosome-encapsulated doxorubicin (DOX) shows faster cellular uptake and accumulates at higher concentrations than free DOX and liposomal DOX in multiple cell lines [154]. Moreover, brain endothelial cell-derived exosomes carrying DOX nanoparticles can be used to deliver drugs to the tumor region and cause their accumulation, inducing tumor cell apoptosis and ICD, leading to DC maturation and CTL infiltration, and enhancing immune responses in GBM mouse models [155]. Based on the intrinsic inflammatory chemotactic capacity of neutrophils, Wang et al. [156] constructed a neutrophil-exosome system (NEs-Exos) to deliver DOX. Substantial evidence suggested that NEs-Exos can cross the BBB and respond chemotactically to inflammatory stimuli, targeting the inflammatory sites of gliomas. Genetic engineering or chemical modification to introduce tissue- or cell-specific ligands onto the exosome surface can further enhance targeting. Angiopep-2 (ANG) has a high affinity for low-density lipoprotein receptor-related protein-1, which is highly expressed in glioma and BBB endothelial cells. Exosomes co-modified using ANG and trans-activator of transcription peptides, highly potent cell-penetrating peptides, can significantly increase DOX penetration into the tumor region through dual targeting for optimal glioma therapy [157]. Moreover, c(RGDyk) modifications on the surfaces of embryonic stem cell-derived exosomes and simultaneous paclitaxel loading were used to treat GBM; here, the modified exosomes significantly improved the therapeutic efficacy of paclitaxel through enhanced targeting compared to that with unmodified exosomes and free paclitaxel [158]. Other tumor-targeting molecules, such as neuropilin-1-targeting peptide [159], heme oxygenase-1 specific short peptide [160], and transferrin receptor-binding peptide [161], have also been experimentally used for exosome surface modification to enhance drug delivery. Thus, exosomes, based on intrinsic targeting or obtained through engineered modifications contribute to chemotherapeutic agent delivery, enhance tumor cell killing, and ultimately facilitate the release of many tumor antigens to enhance immune efficacy.

Mono-chemotherapy often results in resistance and causes insufficient antigen release [162]. Therapies such as radiotherapy, photodynamic therapy, photodynamic therapy (PDT), and oncolytic viruses can also cause apoptosis in tumor cells, resulting in the release of antigens and triggering ICD [163, 164]. Therefore, combination therapy might be a practical anti-tumor approach. Wang et al. [165] fabricated Rg3-modified homologous engineered exosomes for co-delivery of the chemotherapeutic agent arsenic trioxide (ATO) and the photosensitizer chlorin e6 (Ce6). Specifically, it was determined that ATO and Ce6 could be delivered to the tumor area. Under laser irradiation, Ce6-triggered PDT destroys tumor cells and results in the release of more tumor antigens, synergistically improving ATO efficacy. Similarly, a recent study reported a novel strategy for multilevel cascade GBM therapy with M1 macrophage-derived extracellular vesicles (M1EVs) co-loaded with the chemo excitation source CPPO, Ce6, and hypoxia-activated prodrug AQ4N. In this system, M1EVs were found to modulate the TME to increase the level of hydrogen peroxide, which reacts with CPPO to generate chemical energy to activate Ce6 and produce reactive oxygen species for PDT. At the same time, this reaction consumes oxygen to activate the AQ4N toxicity [166]. In addition, in lung cancer-model mice, exosome-mediated combination therapy with oncolytic viruses and paclitaxel significantly induced ICD and T cell infiltration [167], and its application to gliomas might also have significant potential.

Increased antigen expression and presentation with cancer vaccines

TEXs can be used as antigenic vehicles for cancer immunotherapy, for example as cancer vaccines, to elicit immune responses. Harshyne et al. [168] injected different doses of GM-Exos into mice to establish an immune model. They found that high doses of GM-Exos induced protective immunity in mice transplanted with GL261 cells, causing less tumor growth in vivo. This protective effect was associated with enhanced T cell activity and specific antibody production induced by the GM-Exos. However, the immune response elicited by TEXs is relatively weak, requiring the preparation of vaccines with higher immunogenicity. Vaccines comprising DCs loaded with tumor-associated antigens or tumor cell lysates are being widely developed. TEXs have many advantages over traditional antigens, and these include the enrichment of tumor antigens, a strong antigen-presenting phenotype, and easy uptake by DCs and guidance to the antigen-processing region; therefore, these could serve as antigenic vectors for DC vaccines [169]. One study demonstrated that DCs co-incubated with TEXs stimulate the differentiation of T lymphocytes into antigen-specific CTLs, with better glioma cell-killing ability than DCs loaded with cell lysates [170]. Based on these findings, Liu et al. [171] constructed a novel DC vaccine using TEXs in combination with an invariant natural killer T cell adjuvant, which could modulate the release of immunosuppressive/immunostimulatory factors, deregulate the precancer immunosuppressive microenvironment, and induce a solid antigen-specific CTL response against GBM cells.

In addition, the artificial modification of TEXs to enrich tumor antigens, immunostimulatory molecules, and miRNAs enables the fabrication of cell-free cancer vaccines with good immunogenicity and immunostimulatory capacity [169]. To date, engineered TEX vaccines for gliomas have been less studied, but several strategies used to modify TEXs have been developed and have shown good efficacy for other cancers [172]. For example, leukemia cells were transfected with lentiviral vectors encoding B7 co-stimulatory molecules, and the exosomes (LEX-8086) obtained expressed high levels of CD80 and CD86. LEX-8086 was effective in inducing CD4+ T cell proliferation, Th1 cytokine secretion, and a CTL response [173]. Moreover, heat-treated tumor cells can produce exosomes that are highly enriched in HSP70 (HS-TEXs). HS-TEXs, an MHC-independent vaccine, increase IgG2a and IFN-γ production, triggering a strong Th1-type immune response [174]. INF-γ-modified exosome vaccines (INF-γ-TEXs), obtained via protein-anchoring, increase the number of M1-like macrophages and the proportion of IFN-γ+ CD8+ T cells and decrease the proportion of Tregs [175]. In conclusion, these studies suggest that fully deciphering the immunostimulatory/immunosuppressive mechanisms of TEXs and modifying them appropriately could be a valuable immunotherapeutic approach for gliomas.

DEXs express more MHC antigens than DCs and are easy to modify genetically, have a long half-life, and can be used as a biological nano-vaccine to activate the immune response, instead of DCs [111, 176]. Bu et al. [177] found that DEXs containing chaperonin-rich cell lysates induce a strong anti-tumor immune response in mice with glioma by maintaining T cell activation. Moreover, DEXs loaded with neoantigens significantly inhibit tumor growth, preventively delay tumor occurrence, and establish long-term immune memory-mediated protection by inducing broad-spectrum T and B cell immune responses [178]. Recently, Zuo et al. co-loaded hepatocellular carcinoma-targeting peptides, α-fetoprotein epitopes, and an immune adjuvant on the surface of DEXs to prepare a novel bio-nano-vaccine with universal applicability for DC recruitment and activation. The vaccine was found to promote the recruitment and activation of endogenous DCs, as well as tumor neoantigen cross-presentation, to induce T cell responses and activate the innate immune response, providing an innovative approach for developing personalized cancer vaccines [179]. In addition, M1-like macrophage exosomes can be taken up by local DCs and macrophages and cause the release of Th1 cytokines, inducing more robust CTL responses and synergistically enhancing the efficacy of nanoparticle vaccines, suggesting that they have the potential to serve as adjuvants for cancer vaccines [180].

Although the applications of exosome vaccines in glioma are still limited, there is a relatively solid theoretical foundation and reliable methods to prepare exosome vaccines for glioma based on their widespread use in other cancers. In the future, we should continue to explore new approaches to increase exosome immunogenicity and immune activation capacity and overcome host immunosuppression to obtain effective exosome vaccines for glioma.

Multiple targets to synergistically improve the immunosuppressive microenvironment

Reprogramming TAMs

TAMs have two phenotypes, specifically M1-like TAMs obtained via INF-γ and Toll-like receptor 4 stimulation and M2-like macrophages derived from IL-4, IL-10, and IL-13 stimulation [181]. M1-like TAMs secrete TNF-α, IL-6, and IL-12 to promote anti-tumor immune responses. However, the M2 phenotype is predominant among TAMs and is associated with local and systemic immunosuppression in glioma patients [182]. Therefore, reprogramming M2 tumor-promoting phenotypes into M1 anti-tumor phenotypes is a promising strategy for cancer therapy. However, owing to potential side effects, most approaches do not allow for the in situ reprogramming of TAMs. Based on the advantages of exosomes as transporter vehicles, attempts have been made to precisely deliver various therapeutic agents to reprogram TAMs and reduce immunosuppression.

Li et al. [183] constructed a dual delivery system for GBM immunotherapy and evaluated its effect on tumor-bearing mice. First, they prepared T7 peptide-modified exosomes (T7-Exos) and loaded Galectin‑9 siRNA (siGalectin-9) into the exosomes via electroporation. The results showed that T7-Exos could effectively deliver siGalectin-9 to GBM cells. Moreover, siGalectin-9 was dependent on activation of the TLR7–IRF5 pathway, which promoted macrophage polarization to the M1 phenotype, increased macrophage phagocytosis of glioma cells, and activated the immune response. Signal transducer and activator of transcription 3 (STAT3) is a crucial transcription factor that induces macrophage polarization towards the M2 phenotype [181]. Cui et al. [184] reported the self-assembly of tanshinone IIA (TanIIA) and glycyrrhizic acid (GL), which are the inhibitors of STAT3, to form TanIIA-GL nanogel micelles (TGM). Serum-derived exosomes encapsulated with TGM and surface-modified with the immunoadjuvant CpG oligonucleotides were further used to construct a bionic nanodelivery system (CpG-EXO/TGM). CpG-EXO/TGM could be effectively taken up by glioma cells, promoting TAM reprogramming, DC maturation, and CTL activation and activating immune memory cells to prevent tumor recurrence in glioma recurrence models. Similarly, the encapsulation of CpG-STAT3 antisense oligonucleotide using neural stem cell-derived exosomes was found to stimulate macrophage immunoactivity, induce NF-κB signaling and IL-12 production, and inhibit glioma growth [185]. High-molecular weight hyaluronic acid (HMW-HA) inhibits M1-like macrophage polarization and enhances M2-like macrophage polarization [186]. Human hyaluronidase delivered by folic acid-modified exosomes can enhance antitumor efficacy by degrading HMW-HA to promote the polarization of TAMs to the M1 phenotype [187]. In addition, ginseng-derived exosome-like nanoparticles (GENs) carrying multiple chemical cargoes can penetrate the BBB and target glioma cells, recruit M1-like macrophages, and downregulate the M2-like macrophage phenotype, suggesting that GENs are promising candidates for the development of nanocarriers for glioma immunotherapy [188].

Further, M1-like macrophages can secrete the necessary molecules to consistently drive an M2-to-M1 phenotypic conversion, suggesting the feasibility of exploiting M1-like macrophage-derived exosomes to mediate TAM reprogramming [189]. Researchers found that exosome-mimetic nanovesicles from M1-like macrophages can induce the repolarization of M2-like TAMs to the M1 phenotype by modulating the expression profile of miRNAs and mRNAs, which in turn results in the release of pro-inflammatory cytokines to induce anti-tumor immune responses and synergistically enhance the efficacy of ICIs in glioma models [190]. Compared to those on M1-like TAMs, IL4 receptor (IL4R) is expressed at higher levels on M2-like TAMs and is involved in M2 macrophage polarization in response to the cytokine IL-4 [191]. M1 exosomes were transfected with NF-κB p50 siRNA and miR-511-3p to enhance M1 polarization and were surface-modified using IL-4R-binding peptides to target the IL4 receptor (IL4R-Exos(si/mi)). IL4R-Exos(si/mi) can be efficiently internalized by M2-like TAMs and reprogrammed to M1-like macrophages to enhance anti-tumor immunity [192]. In conclusion, exosome-based drug delivery systems can reverse the immunosuppressive microenvironment by reprogramming TAMs into anti-tumor phenotypes to achieve efficient therapy for gliomas.

Targeting MDSCs and Tregs

Based on the immunosuppressive role of MDSCs and Tregs in the TME, researchers have focused on developing immunotherapies targeting these cells to restore the anti-tumor immune response. This includes (i) blocking the recruitment of MDSCs/Tregs, (ii) deregulating the immunosuppressive activity of MDSCs/Tregs, and (iii) depleting MDSCs/Tregs. Here, we present the current strategies used to target MDSCs/Tregs in glioma therapy, to load these therapeutic agents into exosomes and achieve precision therapy.

Targeting MDSCs

The CCL2–CCR2 axis is an essential target for blocking the migration of MDSCs to tumors. CCX872, an antagonist of CCR2, reduces MDSC infiltration and enhances the efficiency of ICIs in glioma-bearing mice [193]. Moreover, a macrophage migration inhibitory factor inhibitor targets a monocytic subset of MDSCs via the CD47 receptor, suppressing downstream MCP-1 signaling and inhibiting MDSC recruitment and expansion [194]. In addition, other chemokines that block MDSC recruitment, such as CCR5 antagonists [195], VEGF antagonists [196], and CXCR2 antagonists [197], are also being actively studied. Developing exosomes containing these blockers for glioma therapy might result in excellent efficacy.

Promoting the differentiation of MDSCs to mature myeloid cells is the most direct and effective way to reverse their immunosuppressive activity. Vitamin A and its metabolites, vitamin D, STAT3 inhibitors, and TLR agonists can mediate the differentiation of MDSCs [44]. All-trans retinoic acid is a vitamin A derivative that can effectively inhibit glioma growth when applied alone [198] or in combination with a DNA demethylating agent [199] and interferon-gamma [200]. Aberrant activation of the cyclooxygenase 2 (COX2)/prostaglandin E2 (PGE2)/PGE2 receptor axis is vital for maintaining the immunosuppressive activity of MDSCs. Researchers found that the COX2 inhibitors acetylsalicylic acid or celecoxib inhibit MDSC activation by decreasing PGE2 production and retard the growth of glioma [201]. Currently, combination therapeutic strategies are under investigation, and exosome-based specific delivery systems could offer a promising means to inhibit MDSCs.

In addition, MDSCs can be exhausted by specific antibodies targeting MDSCs and chemotherapeutic agents. An anti-CD33 antibody, as well as an agonistic DR5 antibody, resulted in the favorable induction of apoptosis in MDSCs [202]. Moreover, some low-dose chemotherapeutic agents (5-fluorouracil and gemcitabine, etc.) can directly exert cytotoxic effects, mediating the depletion of MDSCs. A phase 0/I clinical trial found that the combination of capecitabine, a prodrug of 5-fluorouracil, and bevacizumab reduces circulating MDSC levels in GBM patients [203].

However, when using these therapeutic strategies, problems such as high toxicity, low permeability, and delivery efficiency could be generally encountered. Exosomes, as a powerful drug-loading system, are expected to achieve better therapeutic effects. In fact, researchers have already made some attempts to implement this strategy. Qiu et al. found that modified DEXs loaded with miR-21 inhibitors synergistically improve the therapeutic efficacy of ICIs and prolong the survival of glioma-bearing mice. This was associated with its ability to target tumor mesenchymal stem cells and disrupt the miR-21/SP1/DNMT1 positive feedback loop and reduce MDSC activation by decreasing CD73 expression on MDSCs [204]. Furthermore, glioma-derived extracellular vesicles overexpressing basic leucine zipper ATF-like transcription factor 2 inhibit the recruitment of MDSCs to limit glioma growth [205]. Therefore, based on the aforementioned studies, developing exosome delivery systems targeting MDSCs could be a promising approach.

Targeting Tregs

Similarly, treatment strategies based on Tregs have been extensively studied. Targeting the glucocorticoid-induced TNFR-related receptor (GITR) in Tregs using αGITR promotes Treg differentiation into effector T cells and attenuates treatment resistance in GBM models [206]. Loading an anti-GITR antibody and catalase into nanoparticles consisting of a photothermal agent and photosensitizer induces a 4.3-fold reduction in Tregs and exerts a strong antitumor effect [207]. Moreover, heat stress-treated TEXs stimulate DCs to secrete IL-6 to block the TGF-β1-induced differentiation of Tregs and promote Treg conversion to Th17 cells [208]. A detailed summary of cancer therapeutic strategies targeting Tregs has been presented [209], which is an important guide for the development of exosome-based glioma nano-immunotherapies.

Improving TME physicochemical properties

Hypoxia is a crucial trigger of glioma invasion, anti-apoptotic processes, angiogenesis, and resistance to radiotherapy and chemotherapy [210]. Hypoxic glioma niches attract and sequester TAMs and CTLs and reprogram them toward an immunosuppressed state [211]. Liu et al. [212] developed a glioma-targeted transport system to enhance sonodynamic therapy by alleviating tumor hypoxia. They first encapsulated catalase into silica nanoparticles (CAT@SiO2) and then loaded the sonosensitizer indocyanine green to prepare a biodegradable nano platform (CSIs). The CSIs were further encapsulated with AS1411 aptamer-modified macrophage exosomes to form a delivery system (termed CSI@ Ex-A) with highly efficient BBB-infiltration and tumor-targeting capabilities. At the tumor site, highly expressed glutathione triggered the biodegradation of CSIs, and the catalase that was released catalyzed the production of O2 from H2O2 in the tumor, thus alleviating the hypoxic microenvironment and improving the efficiency of sonodynamic therapy. In addition, CSI@Ex-A effectively inhibited glioma metastasis in vivo, which could be related to the alleviation of tumor hypoxia and the suppression of the activation of hypoxia-inducible factor 1α. Moreover, chronic persistent inflammation confers immune-escape ability in glioma, inducing immune tolerance to numerous therapies. Therefore, glioma therapy can be achieved by suppressing the inflammatory state in the TME. Extracellular vesicles overexpressing esophageal cancer related gene-4 significantly inhibit the expression of inflammatory cytokines (IL-1β, IL-6, IL-8) and activate the p38–AMPK signaling pathway to suppress the inflammatory response and induce anti-tumor effects in vitro and in vivo [213]. Exosomes loaded with STAT3 inhibitors that enter the brain by crossing the BBB, after nasal administration, are selectively taken up by microglia and inhibit the expression of inflammatory cytokines, such as IL-1β and IL-6, ultimately retarding glioma cell growth [214]. In summary, improving hypoxia and chronic inflammation in the TME could also be a strategy to reverse immunosuppression and enhance therapeutic efficacy.

Table 3 Exosome-based drug delivery systems for glioma immunotherapy

Clinical translation and challenge

According to Clinicaltrials.gov (https://clinicaltrials.gov/) [215], there are four clinical trials based on exosomes as drug delivery systems for cancer treatment (Table 4), all of which are at an early stage. Although exosomes for drug delivery have been extensively studied, they still face challenges in clinical translation. First, there is an urgent need to develop standardized and industrialized methods for exosome production, isolation, and purification for the large-scale preparation of clinical-grade exosome products. Exosomes are heterogeneous, and small changes in the production process can affect their yield and activity. However, a gold standard for exosome preparation is lacking, and relatively low yields limit the further clinical applications of exosomes. Researchers have already enhanced exosome secretion in preclinical studies using physical stimulation, molecular interference, environmental factors, and external inducers [216], but extending these findings to the clinic still requires the consideration of various aspects, such as safety, bioactivity, efficiency, and stability. In addition, the exosomes collected using existing separation and purification techniques do not meet the standards for clinical use. Second, the safety of exosomes should be considered. Exosomes of different cell origins have different functions in cancer immunomodulation, and the mechanism underlying the effects of exosomes on cancer immunity should be thoroughly clarified. Especially, TEXs, despite their homing properties and immunogenicity, also contribute to tumor progression through epithelial–mesenchymal transition, angiogenesis, and immune escape. Mesenchymal stem cells can be used for safe and stable exosome production [217]. However, these exosomes lack antigens and antigen-presenting molecules, when compared to exosomes derived from immune and tumor cells, which limits their use in cancer vaccines. In addition, serious safety issues, such as cytokine release syndrome, could arise from the use of exosome drugs [218]. Third, there are still some limitations to the engineering and modification of exosomes. Genetic engineering or chemical modifications can further enhance exosome-specific targeting capabilities. However, genetic engineering is limited to targeting motifs that can be genetically encoded. Moreover, chemical modifications often affect the functions of exosomes, and the complexity of the exosome structure reduces the efficiency of the reaction [219]. In addition, there is no standardized drug-loading technique. Several commonly used drug-loading strategies have different disadvantages and cannot simultaneously combine the properties of high loading efficiency, limited exosome damage, and easy operation. Finally, exosome storage is also a pressing issue. Different storage conditions will affect the exosome morphological structure, protein content, and biological activity. Currently, it is believed that exosomes are more stable at − 80 °C and can maintain clinical availability after 5 months [220, 221]. However, high storage and transportation costs can restrict generalization. In summary, we still need to improve on existing technologies or develop new ones to overcome the problems associated with exosome-based drug delivery systems in terms of their clinical translation, large-scale production, stable preparation and storage, and quality control.

Table 4 Clinical trials of exosomes as drug delivery systems

Conclusions

As a natural delivery system, exosomes have the unique advantages of low toxicity, targeting ability, and immunomodulatory properties, which can be exploited for cancer immunotherapy. Exosomes for the precise delivery of chemotherapeutic drugs, antigens, antigen-presenting molecules, and immunomodulatory molecules to tumors can overcome certain obstacles to existing glioma immunotherapy approaches, including the BBB, low immunogenicity, and an immunosuppressive microenvironment. Exosome-based immunotherapeutic strategies, including increasing antigen release, expression, and presentation, reprogramming TAMs, targeting Tregs and MDSCs, and improving the physicochemical properties of the TME, offer new hopes for effectively enhancing glioma treatment. Although the clinical translation of exosomes still faces many challenges, exosome-based drug delivery systems hold great promise for the glioma immunotherapy. With the continuous development and innovation of new technologies, exosome therapy will soon be at the forefront of glioma treatment.

Data availability

No datasets were generated or analysed during the current study.

Abbreviations

ANG:

Angiopep-2

APC:

Antigen-presenting cell

APM:

Antigen processing and presentation machinery

ARG1:

Arginase

ATO:

Arsenic trioxide

BBB:

Blood–brain barrier

BMEC:

Brain microvascular endothelial cell

Ce6:

Chlorin e6

circRNAs:

Circular RNAs

CNS:

Central nervous system

COX2:

Cyclooxygenase 2

CRCL:

Chaperone-rich cell lysates

CTL:

Cytotoxic T lymphocyte

DC:

Dendritic cell

DEX:

Dendritic cell-derived exosomes

DG:

Density gradient centrifugation

DOX:

Doxorubicin

e-:

Expressed neoantigens

ECRG4:

Esophageal cancer related gene-4

ESCRT:

Endosomal sorting complex required for transport

GBM:

Glioblastoma

GEN:

Ginseng-derived exosome-like nanoparticle

GITR:

Glucocorticoid-induced TNFR-related receptor

GL:

Glycyrrhizic acid

GM-Exos:

Glioma-derived exosomes

HIF-1α:

Hypoxia-inducible factor 1α

HLA:

Human leukocyte antigen

HMW-HA:

High-molecular weight hyaluronic acid

HS-TEXs:

Exosomes that are highly enriched in HSP70

ICAM-1:

Intercellular adhesion molecule

ICD:

Immunogenic cell death

ICG:

Indocyanine green

ICI:

Immune checkpoint inhibitor

INF-γ-TEXs:

INF-γ-modified exosome vaccines

ILV:

Intraluminal vesicle

IMC:

Immature myeloid cell

iNOS:

Inducible nitric oxide synthase

LEX8086:

Exosomes enriched for B7 co-stimulatory molecules

lnRNAs:

Long non-coding RNAs

M1EV:

M1 macrophage-derived extracellular vesicle

MDSC:

Myeloid-derived suppressor cell

MHC:

Major histocompatibility complex

MVB:

Multivesicular body

miRNAs:

microRNAs

mRNAs:

Messenger RNAs

NEs-Exos:

Neutrophil–exosome system

NK:

Natural killer

p-:

Predicted neoantigens

PGE2:

Prostaglandin E2

PDT:

Photodynamic therapy

PDX:

Patient-derived xenograft

SEC:

Size exclusion chromatography

siGalectin-9:

Galectin‑9 siRNA

STAT3:

Signal transducer and activator of transcription 3

T7-Exos:

T7 peptide-modified exosomes

TAM:

Tumor-associated macrophage

TanIIA:

Tanshinone IIA

TEX:

Tumor-derived exosome

TGMs:

TanIIA-GL nanogel micelles

TMB:

Tumor mutational burden

TLR:

Toll-like receptor

TME:

Tumor microenvironment

Treg:

Regulatory T cell

UC:

Ultracentrifugation

UF:

Ultrafiltration

References

  1. Ostrom QT, Price M, Neff C, Cioffi G, Waite KA, Kruchko C, et al. CBTRUS Statistical Report: primary brain and other Central Nervous System tumors diagnosed in the United States in 2015–2019. Neurooncology. 2022;24:v1–95. https://doi.org/10.1093/neuonc/noac202.

    Article  CAS  Google Scholar 

  2. Berger TR, Wen PY, Lang-Orsini M, Chukwueke UN. World Health Organization 2021 Classification of Central Nervous System Tumors and implications for Therapy for adult-type gliomas: a review. JAMA Oncol. 2022;8:1493–501. https://doi.org/10.1001/jamaoncol.2022.2844.

    Article  PubMed  Google Scholar 

  3. Okamoto Y, Di Patre PL, Burkhard C, Horstmann S, Jourde B, Fahey M, et al. Population-based study on incidence, survival rates, and genetic alterations of low-grade diffuse astrocytomas and oligodendrogliomas. Acta Neuropathol. 2004;108:49–56. https://doi.org/10.1007/s00401-004-0861-z.

    Article  PubMed  Google Scholar 

  4. Jiang T, Nam DH, Ram Z, Poon WS, Wang J, Boldbaatar D, et al. Clinical practice guidelines for the management of adult diffuse gliomas. Cancer Lett. 2021;499:60–72. https://doi.org/10.1016/j.canlet.2020.10.050.

    Article  CAS  PubMed  Google Scholar 

  5. Fan X, Wang K, Lu Q, Lu Y, Sun J. Cell-based drug Delivery systems participate in the Cancer immunity cycle for Improved Cancer Immunotherapy. Small. 2023;19:e2205166. https://doi.org/10.1002/smll.202205166.

    Article  CAS  PubMed  Google Scholar 

  6. Yasinjan F, Xing Y, Geng H, Guo R, Yang L, Liu Z, et al. Immunotherapy: a promising approach for glioma treatment. Front Immunol. 2023;14:1255611. https://doi.org/10.3389/fimmu.2023.1255611.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Andersen BM, Reardon DA. Immunotherapy approaches for adult glioma: knowledge gained from recent clinical trials. Curr Opin Neurol. 2022;35:803–13. https://doi.org/10.1097/wco.0000000000001118.

    Article  CAS  PubMed  Google Scholar 

  8. Zhao M, van Straten D, Broekman MLD, Préat V, Schiffelers RM. Nanocarrier-based drug combination therapy for glioblastoma. Theranostics. 2020;10:1355–72. https://doi.org/10.7150/thno.38147.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Aghebati-Maleki A, Dolati S, Ahmadi M, Baghbanzhadeh A, Asadi M, Fotouhi A, et al. Nanoparticles and cancer therapy: perspectives for application of nanoparticles in the treatment of cancers. J Cell Physiol. 2020;235:1962–72. https://doi.org/10.1002/jcp.29126.

    Article  CAS  PubMed  Google Scholar 

  10. Yan H, Xue Z, Xie J, Dong Y, Ma Z, Sun X, et al. Toxicity of Carbon nanotubes as Anti-tumor Drug Carriers. Int J Nanomed. 2019;14:10179–94. https://doi.org/10.2147/ijn.S220087.

    Article  CAS  Google Scholar 

  11. Xu Z, Zeng S, Gong Z, Yan Y. Exosome-based immunotherapy: a promising approach for cancer treatment. Mol Cancer. 2020;19:160. https://doi.org/10.1186/s12943-020-01278-3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Zhao Z, Nelson AR, Betsholtz C, Zlokovic BV. Establishment and dysfunction of the blood-brain barrier. Cell. 2015;163:1064–78. https://doi.org/10.1016/j.cell.2015.10.067.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Kadry H, Noorani B, Cucullo L. A blood-brain barrier overview on structure, function, impairment, and biomarkers of integrity. Fluids Barriers CNS. 2020;17:69. https://doi.org/10.1186/s12987-020-00230-3.

    Article  PubMed  PubMed Central  Google Scholar 

  14. Rui Y, Green JJ. Overcoming delivery barriers in immunotherapy for glioblastoma. Drug Deliv Transl Res. 2021;11:2302–16. https://doi.org/10.1007/s13346-021-01008-2.

    Article  PubMed  PubMed Central  Google Scholar 

  15. Miranda A, Blanco-Prieto M, Sousa J, Pais A, Vitorino C. Breaching barriers in glioblastoma. Part I: molecular pathways and novel treatment approaches. Int J Pharm. 2017;531:372–88. https://doi.org/10.1016/j.ijpharm.2017.07.056.

    Article  CAS  PubMed  Google Scholar 

  16. Tsvankin V, Hashizume R, Katagi H, Herndon JE, Lascola C, Venkatraman TN, et al. ABC Transporter Inhibition Plus Dexamethasone enhances the efficacy of Convection enhanced delivery in H3.3K27M mutant diffuse intrinsic pontine glioma. Neurosurgery. 2020;86:742–51. https://doi.org/10.1093/neuros/nyz212.

    Article  PubMed  Google Scholar 

  17. Wijaya J, Fukuda Y, Schuetz JD. Obstacles to Brain Tumor Therapy: Key ABC transporters. Int J Mol Sci. 2017;18. https://doi.org/10.3390/ijms18122544.

  18. Macedo-Pereira A, Martins C, Lima J, Sarmento B. Digging the intercellular crosstalk via extracellular vesicles: may exosomes be the drug delivery solution for target glioblastoma? J Controlled Release: Official J Controlled Release Soc. 2023;358:98–115. https://doi.org/10.1016/j.jconrel.2023.04.038.

    Article  CAS  Google Scholar 

  19. Sanders S, Debinski W. Challenges to successful implementation of the Immune checkpoint inhibitors for treatment of Glioblastoma. Int J Mol Sci. 2020;21. https://doi.org/10.3390/ijms21082759.

  20. Arvanitis CD, Ferraro GB, Jain RK. The blood-brain barrier and blood-tumour barrier in brain tumours and metastases. Nat Rev Cancer. 2020;20:26–41. https://doi.org/10.1038/s41568-019-0205-x.

    Article  CAS  PubMed  Google Scholar 

  21. Sarkaria JN, Hu LS, Parney IF, Pafundi DH, Brinkmann DH, Laack NN, et al. Is the blood-brain barrier really disrupted in all glioblastomas? A critical assessment of existing clinical data. Neurooncology. 2018;20:184–91. https://doi.org/10.1093/neuonc/nox175.

    Article  CAS  Google Scholar 

  22. Anagnostou V, Bardelli A, Chan TA, Turajlic S. The status of tumor mutational burden and immunotherapy. Nat Cancer. 2022;3:652–6. https://doi.org/10.1038/s43018-022-00382-1.

    Article  PubMed  Google Scholar 

  23. Alexandrov LB, Nik-Zainal S, Wedge DC, Aparicio SA, Behjati S, Biankin AV, et al. Signatures of mutational processes in human cancer. Nature. 2013;500:415–21. https://doi.org/10.1038/nature12477.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Yarchoan M, Hopkins A, Jaffee EM. Tumor mutational burden and response rate to PD-1 inhibition. N Engl J Med. 2017;377:2500–1. https://doi.org/10.1056/NEJMc1713444.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Nejo T, Matsushita H, Karasaki T, Nomura M, Saito K, Tanaka S, et al. Reduced Neoantigen expression revealed by Longitudinal Multiomics as a possible Immune Evasion mechanism in Glioma. Cancer Immunol Res. 2019;7:1148–61. https://doi.org/10.1158/2326-6066.Cir-18-0599.

    Article  CAS  PubMed  Google Scholar 

  26. Dutoit V, Migliorini D, Dietrich PY, Walker PR. Immunotherapy of malignant tumors in the brain: how different from other sites? Front Oncol. 2016;6:256. https://doi.org/10.3389/fonc.2016.00256.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Nejo T, Yamamichi A, Almeida ND, Goretsky YE, Okada H. Tumor antigens in glioma. Semin Immunol. 2020;47:101385. https://doi.org/10.1016/j.smim.2020.101385.

    Article  CAS  Google Scholar 

  28. Press MF, Cordon-Cardo C, Slamon DJ. Expression of the HER-2/neu proto-oncogene in normal human adult and fetal tissues. Oncogene. 1990;5:953–62.

    CAS  PubMed  Google Scholar 

  29. Zagzag D, Salnikow K, Chiriboga L, Yee H, Lan L, Ali MA, et al. Downregulation of major histocompatibility complex antigens in invading glioma cells: stealth invasion of the brain. Lab Invest. 2005;85:328–41. https://doi.org/10.1038/labinvest.3700233.

    Article  CAS  PubMed  Google Scholar 

  30. Hilf N, Kuttruff-Coqui S, Frenzel K, Bukur V, Stevanović S, Gouttefangeas C, et al. Actively personalized vaccination trial for newly diagnosed glioblastoma. Nature. 2019;565:240–5. https://doi.org/10.1038/s41586-018-0810-y.

    Article  CAS  PubMed  Google Scholar 

  31. Shraibman B, Barnea E, Kadosh DM, Haimovich Y, Slobodin G, Rosner I, et al. Identification of Tumor antigens among the HLA peptidomes of Glioblastoma Tumors and plasma. Mol Cell Proteom. 2019;18:1255–68. https://doi.org/10.1074/mcp.RA119.001524.

    Article  CAS  Google Scholar 

  32. Facoetti A, Nano R, Zelini P, Morbini P, Benericetti E, Ceroni M, et al. Human leukocyte antigen and antigen processing machinery component defects in astrocytic tumors. Clin Cancer Res. 2005;11:8304–11. https://doi.org/10.1158/1078-0432.Ccr-04-2588.

    Article  CAS  PubMed  Google Scholar 

  33. Hanahan D, Monje M. Cancer hallmarks intersect with neuroscience in the tumor microenvironment. Cancer Cell. 2023;41:573–80. https://doi.org/10.1016/j.ccell.2023.02.012.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Bikfalvi A, da Costa CA, Avril T, Barnier JV, Bauchet L, Brisson L, et al. Challenges in glioblastoma research: focus on the tumor microenvironment. Trends cancer. 2023;9:9–27. https://doi.org/10.1016/j.trecan.2022.09.005.

    Article  CAS  PubMed  Google Scholar 

  35. Li L, Yu R, Cai T, Chen Z, Lan M, Zou T, et al. Effects of immune cells and cytokines on inflammation and immunosuppression in the tumor microenvironment. Int Immunopharmacol. 2020;88:106939. https://doi.org/10.1016/j.intimp.2020.106939.

    Article  CAS  PubMed  Google Scholar 

  36. Liu Z, Zhou Z, Dang Q, Xu H, Lv J, Li H, et al. Immunosuppression in tumor immune microenvironment and its optimization from CAR-T cell therapy. Theranostics. 2022;12:6273–90. https://doi.org/10.7150/thno.76854.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Buonfiglioli A, Hambardzumyan D. Macrophages and microglia: the cerberus of glioblastoma. Acta Neuropathol Commun. 2021;9:54. https://doi.org/10.1186/s40478-021-01156-z.

    Article  PubMed  PubMed Central  Google Scholar 

  38. Urbantat RM, Jelgersma C, Brandenburg S, Nieminen-Kelhä M, Kremenetskaia I, Zollfrank J, et al. Tumor-Associated Microglia/Macrophages as a predictor for Survival in Glioblastoma and Temozolomide-Induced changes in CXCR2 Signaling with New Resistance Overcoming Strategy by Combination Therapy. Int J Mol Sci. 2021;22. https://doi.org/10.3390/ijms222011180.

  39. Thomas DA, Massagué J. TGF-beta directly targets cytotoxic T cell functions during tumor evasion of immune surveillance. Cancer Cell. 2005;8:369–80. https://doi.org/10.1016/j.ccr.2005.10.012.

    Article  CAS  PubMed  Google Scholar 

  40. Takenaka MC, Gabriely G, Rothhammer V, Mascanfroni ID, Wheeler MA, Chao CC, et al. Control of tumor-associated macrophages and T cells in glioblastoma via AHR and CD39. Nat Neurosci. 2019;22:729–40. https://doi.org/10.1038/s41593-019-0370-y.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Tu S, Lin X, Qiu J, Zhou J, Wang H, Hu S, et al. Crosstalk between Tumor-Associated Microglia/Macrophages and CD8-Positive T cells plays a key role in Glioblastoma. Front Immunol. 2021;12:650105. https://doi.org/10.3389/fimmu.2021.650105.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Mantovani A, Marchesi F, Malesci A, Laghi L, Allavena P. Tumour-associated macrophages as treatment targets in oncology. Nat Rev Clin Oncol. 2017;14:399–416. https://doi.org/10.1038/nrclinonc.2016.217.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Barkal AA, Weiskopf K, Kao KS, Gordon SR, Rosental B, Yiu YY, et al. Engagement of MHC class I by the inhibitory receptor LILRB1 suppresses macrophages and is a target of cancer immunotherapy. Nat Immunol. 2018;19:76–84. https://doi.org/10.1038/s41590-017-0004-z.

    Article  CAS  PubMed  Google Scholar 

  44. Li K, Shi H, Zhang B, Ou X, Ma Q, Chen Y, et al. Myeloid-derived suppressor cells as immunosuppressive regulators and therapeutic targets in cancer. Signal Transduct Target Ther. 2021;6:362. https://doi.org/10.1038/s41392-021-00670-9.

    Article  PubMed  PubMed Central  Google Scholar 

  45. Mi Y, Guo N, Luan J, Cheng J, Hu Z, Jiang P, et al. The emerging role of myeloid-derived suppressor cells in the Glioma Immune Suppressive Microenvironment. Front Immunol. 2020;11:737. https://doi.org/10.3389/fimmu.2020.00737.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Lakshmanachetty S, Cruz-Cruz J, Hoffmeyer E, Cole AP, Mitra SS. New insights into the multifaceted role of myeloid-derived suppressor cells (MDSCs) in high-Grade gliomas: from metabolic reprograming, immunosuppression, and therapeutic resistance to current strategies for Targeting MDSCs. Cells. 2021. https://doi.org/10.3390/cells10040893.

    Article  PubMed  PubMed Central  Google Scholar 

  47. Vetsika EK, Koukos A, Kotsakis A. Myeloid-derived suppressor cells: Major figures that shape the immunosuppressive and angiogenic network in Cancer. Cells. 2019;8. https://doi.org/10.3390/cells8121647.

  48. Kamran N, Kadiyala P, Saxena M, Candolfi M, Li Y, Moreno-Ayala MA, et al. Immunosuppressive myeloid cells’ blockade in the Glioma Microenvironment enhances the efficacy of Immune-Stimulatory Gene Therapy. Mol Ther. 2017;25:232–48. https://doi.org/10.1016/j.ymthe.2016.10.003.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Ghosh S, Huang J, Inkman M, Zhang J, Thotala S, Tikhonova E, et al. Radiation-induced circulating myeloid-derived suppressor cells induce systemic lymphopenia after chemoradiotherapy in patients with glioblastoma. Sci Transl Med. 2023;15:eabn6758. https://doi.org/10.1126/scitranslmed.abn6758.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. El Andaloussi A, Lesniak MS. An increase in CD4 + CD25 + FOXP3 + regulatory T cells in tumor-infiltrating lymphocytes of human glioblastoma multiforme. Neurooncology. 2006;8:234–43. https://doi.org/10.1215/15228517-2006-006.

    Article  Google Scholar 

  51. Heimberger AB, Abou-Ghazal M, Reina-Ortiz C, Yang DS, Sun W, Qiao W, et al. Incidence and prognostic impact of FoxP3 + regulatory T cells in human gliomas. Clin Cancer Res. 2008;14:5166–72. https://doi.org/10.1158/1078-0432.Ccr-08-0320.

    Article  CAS  PubMed  Google Scholar 

  52. Nishikawa H, Koyama S. Mechanisms of regulatory T cell infiltration in tumors: implications for innovative immune precision therapies. J Immunother Cancer. 2021;9. https://doi.org/10.1136/jitc-2021-002591.

  53. Li C, Jiang P, Wei S, Xu X, Wang J. Regulatory T cells in tumor microenvironment: new mechanisms, potential therapeutic strategies and future prospects. Mol Cancer. 2020;19:116. https://doi.org/10.1186/s12943-020-01234-1.

    Article  Google Scholar 

  54. Yan Y, Huang L, Liu Y, Yi M, Chu Q, Jiao D, et al. Metabolic profiles of regulatory T cells and their adaptations to the tumor microenvironment: implications for antitumor immunity. J Hematol Oncol. 2022;15:104. https://doi.org/10.1186/s13045-022-01322-3.

    Article  CAS  Google Scholar 

  55. van Hooren L, Handgraaf SM, Kloosterman DJ, Karimi E, van Mil L, Gassama AA, et al. CD103(+) regulatory T cells underlie resistance to radio-immunotherapy and impair CD8(+) T cell activation in glioblastoma. Nat Cancer. 2023;4:665–81. https://doi.org/10.1038/s43018-023-00547-6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Shao H, Im H, Castro CM, Breakefield X, Weissleder R, Lee H. New Technologies for Analysis of Extracellular Vesicles. Chem Rev. 2018;118:1917–50. https://doi.org/10.1021/acs.chemrev.7b00534.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Kalluri R, LeBleu VS. The biology, function, and biomedical applications of exosomes. New York, N.Y.: Science; 2020;367. https://doi.org/10.1126/science.aau6977.

    Book  Google Scholar 

  58. Juan T, Fürthauer M. Biogenesis and function of ESCRT-dependent extracellular vesicles. Semin Cell Dev Biol. 2018;74:66–77. https://doi.org/10.1016/j.semcdb.2017.08.022.

    Article  CAS  PubMed  Google Scholar 

  59. Lee YJ, Shin KJ, Chae YC. Regulation of cargo selection in exosome biogenesis and its biomedical applications in cancer. Exp Mol Med. 2024. https://doi.org/10.1038/s12276-024-01209-y.

    Article  PubMed  PubMed Central  Google Scholar 

  60. Gurunathan S, Kang MH, Kim JH. A comprehensive review on factors influences Biogenesis, functions, therapeutic and clinical implications of Exosomes. Int J Nanomed. 2021;16:1281–312. https://doi.org/10.2147/ijn.S291956.

    Article  Google Scholar 

  61. Rajput A, Varshney A, Bajaj R, Pokharkar V. Exosomes as New Generation vehicles for Drug Delivery: Biomedical Applications and Future perspectives. Molecules. 2022;27. https://doi.org/10.3390/molecules27217289.

  62. Patil SM, Sawant SS, Kunda NK. Exosomes as drug delivery systems: a brief overview and progress update. Eur J Pharm Biopharmaceutics: Official J Arbeitsgemeinschaft fur Pharmazeutische Verfahrenstechnik e V. 2020;154:259–69. https://doi.org/10.1016/j.ejpb.2020.07.026.

    Article  CAS  Google Scholar 

  63. Yuan YG, Wang JL, Zhang YX, Li L, Reza A, Gurunathan S. Biogenesis, composition and potential therapeutic applications of mesenchymal stem cells derived exosomes in various diseases. Int J Nanomed. 2023;18:3177–210. https://doi.org/10.2147/ijn.S407029.

    Article  CAS  Google Scholar 

  64. Skog J, Würdinger T, van Rijn S, Meijer DH, Gainche L, Sena-Esteves M, et al. Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers. Nat Cell Biol. 2008;10:1470–6. https://doi.org/10.1038/ncb1800.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell. 2004;116:281–97. https://doi.org/10.1016/s0092-8674(04)00045-5.

    Article  CAS  PubMed  Google Scholar 

  66. Yun BD, Choi YJ, Son SW, Cipolla GA, Berti FCB, Malheiros D, et al. Oncogenic Role of Exosomal Circular and long noncoding RNAs in gastrointestinal cancers. Int J Mol Sci. 2022;23. https://doi.org/10.3390/ijms23020930.

  67. Cheng J, Meng J, Zhu L, Peng Y. Exosomal noncoding RNAs in glioma: biological functions and potential clinical applications. Mol Cancer. 2020;19:66. https://doi.org/10.1186/s12943-020-01189-3.

    Article  CAS  Google Scholar 

  68. Skotland T, Sagini K, Sandvig K, Llorente A. An emerging focus on lipids in extracellular vesicles. Adv Drug Deliv Rev. 2020;159:308–21. https://doi.org/10.1016/j.addr.2020.03.002.

    Article  CAS  PubMed  Google Scholar 

  69. Chen H, Wang L, Zeng X, Schwarz H, Nanda HS, Peng X, et al. Exosomes, a New Star for targeted delivery. Front cell Dev Biology. 2021;9:751079. https://doi.org/10.3389/fcell.2021.751079.

    Article  Google Scholar 

  70. Tkach M, Théry C. Communication by Extracellular vesicles: where we are and where we need to go. Cell. 2016;164:1226–32. https://doi.org/10.1016/j.cell.2016.01.043.

    Article  CAS  PubMed  Google Scholar 

  71. Paskeh MDA, Entezari M, Mirzaei S, Zabolian A, Saleki H, Naghdi MJ, et al. Emerging role of exosomes in cancer progression and tumor microenvironment remodeling. J Hematol Oncol. 2022;15:83. https://doi.org/10.1186/s13045-022-01305-4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Ferreira JV, da Rosa Soares A, Ramalho J, Máximo Carvalho C, Cardoso MH, Pintado P, et al. LAMP2A regulates the loading of proteins into exosomes. Sci Adv. 2022;8:eabm1140. https://doi.org/10.1126/sciadv.abm1140.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Li H, Zhu J, Liu X, Liu L, Huang S, Wu A, et al. Glioma stem cell-derived exosomes induce the transformation of astrocytes via the miR-3065-5p/DLG2 signaling axis. Glia. 2024;72:857–71. https://doi.org/10.1002/glia.24506.

    Article  CAS  PubMed  Google Scholar 

  74. Dai Y, Wang S, Chang S, Ren D, Shali S, Li C, et al. M2 macrophage-derived exosomes carry microRNA-148a to alleviate myocardial ischemia/reperfusion injury via inhibiting TXNIP and the TLR4/NF-κB/NLRP3 inflammasome signaling pathway. J Mol Cell Cardiol. 2020;142:65–79. https://doi.org/10.1016/j.yjmcc.2020.02.007.

    Article  CAS  PubMed  Google Scholar 

  75. Wang G, Jin S, Ling X, Li Y, Hu Y, Zhang Y, et al. Proteomic profiling of LPS-Induced macrophage-derived exosomes indicates their involvement in Acute Liver Injury. Proteomics. 2019;19:e1800274. https://doi.org/10.1002/pmic.201800274.

    Article  CAS  PubMed  Google Scholar 

  76. Zhang X, Xu D, Song Y, He R, Wang T. Research Progress in the application of exosomes in Immunotherapy. Front Immunol. 2022;13:731516. https://doi.org/10.3389/fimmu.2022.731516.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Gołębiewska JE, Wardowska A, Pietrowska M, Wojakowska A, Dębska-Ślizień A. Small extracellular vesicles in transplant rejection. Cells. 2021. https://doi.org/10.3390/cells10112989.

    Article  PubMed  PubMed Central  Google Scholar 

  78. Kou M, Huang L, Yang J, Chiang Z, Chen S, Liu J, et al. Mesenchymal stem cell-derived extracellular vesicles for immunomodulation and regeneration: a next generation therapeutic tool? Cell Death Dis. 2022;13:580. https://doi.org/10.1038/s41419-022-05034-x.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Varderidou-Minasian S, Lorenowicz MJ. Mesenchymal stromal/stem cell-derived extracellular vesicles in tissue repair: challenges and opportunities. Theranostics. 2020;10:5979–97. https://doi.org/10.7150/thno.40122.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Tsiapalis D, O’Driscoll L. Mesenchymal stem cell derived Extracellular vesicles for tissue Engineering and Regenerative Medicine Applications. Cells. 2020;9. https://doi.org/10.3390/cells9040991.

  81. Matsuzaka Y, Yashiro R. Therapeutic strategy of mesenchymal-stem-cell-derived extracellular vesicles as Regenerative Medicine. Int J Mol Sci. 2022;23. https://doi.org/10.3390/ijms23126480.

  82. Ma YS, Yang XL, Xin R, Liu JB, Fu D. Power and promise of exosomes as clinical biomarkers and therapeutic vectors for liquid biopsy and cancer control. Biochimica et biophysica acta. Reviews cancer. 2021;1875:188497. https://doi.org/10.1016/j.bbcan.2020.188497.

    Article  CAS  Google Scholar 

  83. Moon B, Chang S. Exosome as a delivery vehicle for Cancer Therapy. Cells. 2022;11. https://doi.org/10.3390/cells11030316.

  84. Liguori GL. Challenges and Promise for Glioblastoma Treatment through Extracellular Vesicle Inquiry. Cells. 2024;13. https://doi.org/10.3390/cells13040336.

  85. Cen X, Chen Q, Wang B, Xu H, Wang X, Ling Y, et al. UBE2O ubiquitinates PTRF/CAVIN1 and inhibits the secretion of exosome-related PTRF/CAVIN1. Cell communication and signaling. CCS. 2022;20:191. https://doi.org/10.1186/s12964-022-00996-z.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Parsaei H, Moosavifar MJ, Eftekharzadeh M, Ramezani R, Barati M, Mirzaei S, et al. Exosomes to control glioblastoma multiforme: investigating the effects of mesenchymal stem cell-derived exosomes on C6 cells in vitro. Cell Biol Int. 2022;46:2028–40. https://doi.org/10.1002/cbin.11884.

    Article  CAS  PubMed  Google Scholar 

  87. Hao SC, Ma H, Niu ZF, Sun SY, Zou YR, Xia HC. hUC-MSCs secreted exosomes inhibit the glioma cell progression through PTENP1/miR-10a-5p/PTEN pathway. Eur Rev Med Pharmacol Sci. 2019;23:10013–23. https://doi.org/10.26355/eurrev_201911_19568.

    Article  PubMed  Google Scholar 

  88. Grimaldi A, Serpe C, Chece G, Nigro V, Sarra A, Ruzicka B, et al. Microglia-Derived Microvesicles affect Microglia phenotype in Glioma. Front Cell Neurosci. 2019;13:41. https://doi.org/10.3389/fncel.2019.00041.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Ding C, Yi X, Chen X, Wu Z, You H, Chen X, et al. Warburg effect-promoted exosomal circ_0072083 releasing up-regulates NANGO expression through multiple pathways and enhances temozolomide resistance in glioma. J Experimental Clin cancer Research: CR. 2021;40:164. https://doi.org/10.1186/s13046-021-01942-6.

    Article  CAS  Google Scholar 

  90. Zhang Z, Yin J, Lu C, Wei Y, Zeng A, You Y. Exosomal transfer of long non-coding RNA SBF2-AS1 enhances chemoresistance to temozolomide in glioblastoma. J Experimental Clin cancer Research: CR. 2019;38:166. https://doi.org/10.1186/s13046-019-1139-6.

    Article  PubMed Central  Google Scholar 

  91. Yin J, Zeng A, Zhang Z, Shi Z, Yan W, You Y. Exosomal transfer of miR-1238 contributes to temozolomide-resistance in glioblastoma. EBioMedicine. 2019;42:238–51. https://doi.org/10.1016/j.ebiom.2019.03.016.

    Article  PubMed  PubMed Central  Google Scholar 

  92. Ricklefs FL, Wollmann K, Salviano-Silva A, Drexler R, Maire CL, Kaul MG, et al. Circulating extracellular vesicles as biomarker for diagnosis, prognosis and monitoring in glioblastoma patients. Neurooncology. 2024. https://doi.org/10.1093/neuonc/noae068.

    Article  Google Scholar 

  93. Banks WA, Sharma P, Bullock KM, Hansen KM, Ludwig N, Whiteside TL. Transport of Extracellular vesicles across the blood-brain barrier: Brain Pharmacokinetics and effects of inflammation. Int J Mol Sci. 2020;21. https://doi.org/10.3390/ijms21124407.

  94. Rehman FU, Liu Y, Zheng M, Shi B. Exosomes based strategies for brain drug delivery. Biomaterials. 2023;293:121949. https://doi.org/10.1016/j.biomaterials.2022.121949.

    Article  CAS  PubMed  Google Scholar 

  95. Chen CC, Liu L, Ma F, Wong CW, Guo XE, Chacko JV, et al. Elucidation of Exosome Migration across the blood-brain barrier model in Vitro. Cell Mol Bioeng. 2016;9:509–29. https://doi.org/10.1007/s12195-016-0458-3.

    Article  CAS  PubMed  Google Scholar 

  96. Yuan D, Zhao Y, Banks WA, Bullock KM, Haney M, Batrakova E, et al. Macrophage exosomes as natural nanocarriers for protein delivery to inflamed brain. Biomaterials. 2017;142:1–12. https://doi.org/10.1016/j.biomaterials.2017.07.011.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Joshi BS, Zuhorn IS. Heparan sulfate proteoglycan-mediated dynamin-dependent transport of neural stem cell exosomes in an in vitro blood-brain barrier model. Eur J Neurosci. 2021;53:706–19. https://doi.org/10.1111/ejn.14974.

    Article  CAS  PubMed  Google Scholar 

  98. Qu M, Lin Q, Huang L, Fu Y, Wang L, He S, et al. Dopamine-loaded blood exosomes targeted to brain for better treatment of Parkinson’s disease. J Controlled Release: Official J Controlled Release Soc. 2018;287:156–66. https://doi.org/10.1016/j.jconrel.2018.08.035.

    Article  CAS  Google Scholar 

  99. Kuroda H, Tachikawa M, Yagi Y, Umetsu M, Nurdin A, Miyauchi E, et al. Cluster of differentiation 46 is the major receptor in human blood-brain barrier endothelial cells for Uptake of exosomes derived from brain-metastatic melanoma cells (SK-Mel-28). Mol Pharm. 2019;16:292–304. https://doi.org/10.1021/acs.molpharmaceut.8b00985.

    Article  CAS  PubMed  Google Scholar 

  100. Abdelsalam M, Ahmed M, Osaid Z, Hamoudi R, Harati R. Insights into Exosome Transport through the blood-brain barrier and the potential therapeutical applications in Brain diseases. Pharmaceuticals (Basel). 2023;16. https://doi.org/10.3390/ph16040571.

  101. Ruan S, Greenberg Z, Pan X, Zhuang P, Erwin N, He M. Extracellular vesicles as an Advanced Delivery Biomaterial for Precision Cancer Immunotherapy. Adv Healthc Mater. 2022;11:e2100650. https://doi.org/10.1002/adhm.202100650.

    Article  CAS  PubMed  Google Scholar 

  102. Gastpar R, Gehrmann M, Bausero MA, Asea A, Gross C, Schroeder JA, et al. Heat shock protein 70 surface-positive tumor exosomes stimulate migratory and cytolytic activity of natural killer cells. Cancer Res. 2005;65:5238–47. https://doi.org/10.1158/0008-5472.Can-04-3804.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Li W, Mu D, Tian F, Hu Y, Jiang T, Han Y, et al. Exosomes derived from Rab27a–overexpressing tumor cells elicit efficient induction of antitumor immunity. Mol Med Rep. 2013;8:1876–82. https://doi.org/10.3892/mmr.2013.1738.

    Article  CAS  PubMed  Google Scholar 

  104. 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. https://doi.org/10.1038/s41419-020-03042-3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Qiu W, Guo X, Li B, Wang J, Qi Y, Chen Z, et al. Exosomal miR-1246 from glioma patient body fluids drives the differentiation and activation of myeloid-derived suppressor cells. Mol Ther. 2021;29:3449–64. https://doi.org/10.1016/j.ymthe.2021.06.023.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Pan Z, Zhao R, Li B, Qi Y, Qiu W, Guo Q, et al. EWSR1-induced circNEIL3 promotes glioma progression and exosome-mediated macrophage immunosuppressive polarization via stabilizing IGF2BP3. Mol Cancer. 2022;21:16. https://doi.org/10.1186/s12943-021-01485-6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. 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. https://doi.org/10.1038/s41419-021-04359-3.

    Article  CAS  Google Scholar 

  108. Ricklefs FL, Alayo Q, Krenzlin H, Mahmoud AB, Speranza MC, Nakashima H, et al. Immune evasion mediated by PD-L1 on glioblastoma-derived extracellular vesicles. Sci Adv. 2018;4:eaar2766. https://doi.org/10.1126/sciadv.aar2766.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Yang S, Sun Y, Liu W, Zhang Y, Sun G, Xiang B, et al. Exosomes in Glioma: unraveling their roles in progression, diagnosis, and Therapy. Cancers. 2024;16. https://doi.org/10.3390/cancers16040823.

  110. Guo X, Sui R, Piao H. Tumor-derived small extracellular vesicles: potential roles and mechanism in glioma. J Nanobiotechnol. 2022;20:383. https://doi.org/10.1186/s12951-022-01584-6.

    Article  CAS  Google Scholar 

  111. Pitt JM, André F, Amigorena S, Soria JC, Eggermont A, Kroemer G, et al. Dendritic cell-derived exosomes for cancer therapy. J Clin Invest. 2016;126:1224–32. https://doi.org/10.1172/jci81137.

    Article  PubMed  PubMed Central  Google Scholar 

  112. Théry C, Duban L, Segura E, Véron P, Lantz O, Amigorena S. Indirect activation of naïve CD4 + T cells by dendritic cell-derived exosomes. Nat Immunol. 2002;3:1156–62. https://doi.org/10.1038/ni854.

    Article  CAS  PubMed  Google Scholar 

  113. Munich S, Sobo-Vujanovic A, Buchser WJ, Beer-Stolz D, Vujanovic NL. Dendritic cell exosomes directly kill tumor cells and activate natural killer cells via TNF superfamily ligands. Oncoimmunology. 2012;1:1074–83. https://doi.org/10.4161/onci.20897.

    Article  PubMed  PubMed Central  Google Scholar 

  114. Viaud S, Terme M, Flament C, Taieb J, André F, Novault S, et al. Dendritic cell-derived exosomes promote natural killer cell activation and proliferation: a role for NKG2D ligands and IL-15Ralpha. PLoS ONE. 2009;4:e4942. https://doi.org/10.1371/journal.pone.0004942.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Wu SW, Li L, Wang Y, Xiao Z. CTL-Derived exosomes enhance the activation of CTLs stimulated by Low-Affinity Peptides. Front Immunol. 2019;10:1274. https://doi.org/10.3389/fimmu.2019.01274.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Shin S, Jung I, Jung D, Kim CS, Kang SM, Ryu S, et al. Novel antitumor therapeutic strategy using CD4(+) T cell-derived extracellular vesicles. Biomaterials. 2022;289:121765. https://doi.org/10.1016/j.biomaterials.2022.121765.

    Article  CAS  PubMed  Google Scholar 

  117. Lu J, Wu J, Xie F, Tian J, Tang X, Guo H, et al. CD4(+) T cell-released extracellular vesicles potentiate the efficacy of the HBsAg vaccine by enhancing B cell responses. Adv Sci (Weinh). 2019;6:1802219. https://doi.org/10.1002/advs.201802219.

    Article  CAS  PubMed  Google Scholar 

  118. Smyth LA, Ratnasothy K, Tsang JY, Boardman D, Warley A, Lechler R, et al. CD73 expression on extracellular vesicles derived from CD4 + CD25 + Foxp3 + T cells contributes to their regulatory function. Eur J Immunol. 2013;43:2430–40. https://doi.org/10.1002/eji.201242909.

    Article  CAS  PubMed  Google Scholar 

  119. Aiello S, Rocchetta F, Longaretti L, Faravelli S, Todeschini M, Cassis L, et al. Extracellular vesicles derived from T regulatory cells suppress T cell proliferation and prolong allograft survival. Sci Rep. 2017;7:11518. https://doi.org/10.1038/s41598-017-08617-3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Okoye IS, Coomes SM, Pelly VS, Czieso S, Papayannopoulos V, Tolmachova T, et al. MicroRNA-containing T-regulatory-cell-derived exosomes suppress pathogenic T helper 1 cells. Immunity. 2014;41:89–103. https://doi.org/10.1016/j.immuni.2014.05.019.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Xu Y, Liu Y, Yang C, Kang L, Wang M, Hu J, et al. Macrophages transfer antigens to dendritic cells by releasing exosomes containing dead-cell-associated antigens partially through a ceramide-dependent pathway to enhance CD4(+) T-cell responses. Immunology. 2016;149:157–71. https://doi.org/10.1111/imm.12630.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Dosil SG, Lopez-Cobo S, Rodriguez-Galan A, Fernandez-Delgado I, Ramirez-Huesca M, Milan-Rois P, et al. Natural killer (NK) cell-derived extracellular-vesicle shuttled microRNAs control T cell responses. Elife. 2022;11. https://doi.org/10.7554/eLife.76319.

  123. Hazrati A, Soudi S, Malekpour K, Mahmoudi M, Rahimi A, Hashemi SM, et al. Immune cells-derived exosomes function as a double-edged sword: role in disease progression and their therapeutic applications. Biomark Res. 2022;10:30. https://doi.org/10.1186/s40364-022-00374-4.

    Article  PubMed  PubMed Central  Google Scholar 

  124. Saad MG, Beyenal H, Dong WJ. Exosomes as powerful engines in Cancer: isolation, characterization and detection techniques. Biosens (Basel). 2021;11. https://doi.org/10.3390/bios11120518.

  125. Yang D, Zhang W, Zhang H, Zhang F, Chen L, Ma L, et al. Progress, opportunity, and perspective on exosome isolation - efforts for efficient exosome-based theranostics. Theranostics. 2020;10:3684–707. https://doi.org/10.7150/thno.41580.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Batrakova EV, Kim MS. Using exosomes, naturally-equipped nanocarriers, for drug delivery. J Controlled Release: Official J Controlled Release Soc. 2015;219:396–405. https://doi.org/10.1016/j.jconrel.2015.07.030.

    Article  CAS  Google Scholar 

  127. Wang X, Xia J, Yang L, Dai J, He L. Recent progress in exosome research: isolation, characterization and clinical applications. Cancer Gene Ther. 2023;30:1051–65. https://doi.org/10.1038/s41417-023-00617-y.

    Article  CAS  PubMed  Google Scholar 

  128. Zhang Y, Bi J, Huang J, Tang Y, Du S, Li P, Exosome. A review of its classification, isolation techniques, Storage, Diagnostic and targeted therapy applications. Int J Nanomed. 2020;15:6917–34. https://doi.org/10.2147/ijn.S264498.

    Article  CAS  Google Scholar 

  129. Doyle LM, Wang MZ. Overview of Extracellular vesicles, their origin, composition, purpose, and methods for Exosome isolation and analysis. Cells. 2019;8. https://doi.org/10.3390/cells8070727.

  130. Guan S, Yu H, Yan G, Gao M, Sun W, Zhang X. Characterization of urinary exosomes purified with size Exclusion Chromatography and Ultracentrifugation. J Proteome Res. 2020;19:2217–25. https://doi.org/10.1021/acs.jproteome.9b00693.

    Article  CAS  PubMed  Google Scholar 

  131. Sidhom K, Obi PO, Saleem A. A review of Exosomal isolation methods: is size Exclusion Chromatography the best option? Int J Mol Sci. 2020;21. https://doi.org/10.3390/ijms21186466.

  132. Liu DSK, Upton FM, Rees E, Limb C, Jiao LR, Krell J, et al. Size-exclusion chromatography as a technique for the investigation of Novel Extracellular vesicles in Cancer. Cancers. 2020;12. https://doi.org/10.3390/cancers12113156.

  133. Zarovni N, Corrado A, Guazzi P, Zocco D, Lari E, Radano G, et al. Integrated isolation and quantitative analysis of exosome shuttled proteins and nucleic acids using immunocapture approaches. Methods. 2015;87:46–58. https://doi.org/10.1016/j.ymeth.2015.05.028.

    Article  CAS  PubMed  Google Scholar 

  134. Chen J, Li P, Zhang T, Xu Z, Huang X, Wang R, et al. Review on strategies and technologies for Exosome isolation and purification. Front Bioeng Biotechnol. 2021;9:811971. https://doi.org/10.3389/fbioe.2021.811971.

    Article  PubMed  Google Scholar 

  135. Weng Y, Sui Z, Shan Y, Hu Y, Chen Y, Zhang L, et al. Effective isolation of exosomes with polyethylene glycol from cell culture supernatant for in-depth proteome profiling. Analyst. 2016;141:4640–6. https://doi.org/10.1039/c6an00892e.

    Article  CAS  PubMed  Google Scholar 

  136. Huang LH, Rau CS, Wu SC, Wu YC, Wu CJ, Tsai CW, et al. Identification and characterization of hADSC-derived exosome proteins from different isolation methods. J Cell Mol Med. 2021;25:7436–50. https://doi.org/10.1111/jcmm.16775.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Shirejini SZ, Inci F. The Yin and Yang of exosome isolation methods: conventional practice, microfluidics, and commercial kits. Biotechnol Adv. 2022;54:107814. https://doi.org/10.1016/j.biotechadv.2021.107814.

    Article  CAS  PubMed  Google Scholar 

  138. Lin B, Lei Y, Wang J, Zhu L, Wu Y, Zhang H, et al. Microfluidic-based Exosome Analysis for Liquid Biopsy. Small Methods. 2021;5:e2001131. https://doi.org/10.1002/smtd.202001131.

    Article  CAS  PubMed  Google Scholar 

  139. Fu Z, Zhang X, Zhou X, Ur-Rehman U, Yu M, Liang H, et al. In vivo self-assembled small RNAs as a new generation of RNAi therapeutics. Cell Res. 2021;31:631–48. https://doi.org/10.1038/s41422-021-00491-z.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Silva AM, Lázaro-Ibáñez E, Gunnarsson A, Dhande A, Daaboul G, Peacock B, et al. Quantification of protein cargo loading into engineered extracellular vesicles at single-vesicle and single-molecule resolution. J Extracell Vesicles. 2021;10:e12130. https://doi.org/10.1002/jev2.12130.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Dang XTT, Kavishka JM, Zhang DX, Pirisinu M, Le MTN. Extracellular vesicles as an efficient and versatile system for drug delivery. Cells. 2020;9. https://doi.org/10.3390/cells9102191.

  142. Yang Z, Shi J, Xie J, Wang Y, Sun J, Liu T, et al. Large-scale generation of functional mRNA-encapsulating exosomes via cellular nanoporation. Nat Biomed Eng. 2020;4:69–83. https://doi.org/10.1038/s41551-019-0485-1.

    Article  CAS  PubMed  Google Scholar 

  143. Burgio S, Noori L, Marino Gammazza A, Campanella C, Logozzi M, Fais S, et al. Extracellular vesicles-based drug Delivery systems: a New Challenge and the Exemplum of Malignant Pleural Mesothelioma. Int J Mol Sci. 2020;21. https://doi.org/10.3390/ijms21155432.

  144. Herrmann IK, Wood MJA, Fuhrmann G. Extracellular vesicles as a next-generation drug delivery platform. Nat Nanotechnol. 2021;16:748–59. https://doi.org/10.1038/s41565-021-00931-2.

    Article  CAS  PubMed  Google Scholar 

  145. Kimiz-Gebologlu I, Oncel SS, Exosomes. Large-scale production, isolation, drug loading efficiency, and biodistribution and uptake. J Controlled Release: Official J Controlled Release Soc. 2022;347:533–43. https://doi.org/10.1016/j.jconrel.2022.05.027.

    Article  CAS  Google Scholar 

  146. Ortega A, Martinez-Arroyo O, Forner MJ, Cortes R. Exosomes as Drug Delivery systems: endogenous nanovehicles for treatment of systemic Lupus Erythematosus. Pharmaceutics. 2020;13. https://doi.org/10.3390/pharmaceutics13010003.

  147. Haney MJ, Klyachko NL, Zhao Y, Gupta R, Plotnikova EG, He Z, et al. Exosomes as drug delivery vehicles for Parkinson’s disease therapy. J Controlled Release: Official J Controlled Release Soc. 2015;207:18–30. https://doi.org/10.1016/j.jconrel.2015.03.033.

    Article  CAS  Google Scholar 

  148. Kim MS, Haney MJ, Zhao Y, Mahajan V, Deygen I, Klyachko NL, et al. Development of exosome-encapsulated paclitaxel to overcome MDR in cancer cells. Nanomedicine. 2016;12:655–64. https://doi.org/10.1016/j.nano.2015.10.012.

    Article  CAS  PubMed  Google Scholar 

  149. Susa F, Limongi T, Dumontel B, Vighetto V, Cauda V. Engineered Extracellular vesicles as a Reliable Tool in Cancer Nanomedicine. Cancers. 2019;11. https://doi.org/10.3390/cancers11121979.

  150. Toffoli G, Hadla M, Corona G, Caligiuri I, Palazzolo S, Semeraro S, et al. Exosomal doxorubicin reduces the cardiac toxicity of doxorubicin. Nanomed (Lond). 2015;10:2963–71. https://doi.org/10.2217/nnm.15.118.

    Article  CAS  Google Scholar 

  151. Luan X, Sansanaphongpricha K, Myers I, Chen H, Yuan H, Sun D. Engineering exosomes as refined biological nanoplatforms for drug delivery. Acta Pharmacol Sin. 2017;38:754–63. https://doi.org/10.1038/aps.2017.12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Jeyaram A, Lamichhane TN, Wang S, Zou L, Dahal E, Kronstadt SM, et al. Enhanced loading of functional miRNA Cargo via pH gradient modification of Extracellular vesicles. Mol Ther. 2020;28:975–85. https://doi.org/10.1016/j.ymthe.2019.12.007.

    Article  CAS  PubMed  Google Scholar 

  153. Sancho-Albero M, Rubio-Ruiz B, Pérez-López AM, Sebastián V, Martín-Duque P, Arruebo M, et al. Cancer-derived exosomes loaded with ultrathin palladium nanosheets for targeted bioorthogonal catalysis. Nat Catal. 2019;2:864–72. https://doi.org/10.1038/s41929-019-0333-4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Schindler C, Collinson A, Matthews C, Pointon A, Jenkinson L, Minter RR, et al. Exosomal delivery of doxorubicin enables rapid cell entry and enhanced in vitro potency. PLoS ONE. 2019;14:e0214545. https://doi.org/10.1371/journal.pone.0214545.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Zhang C, Song J, Lou L, Qi X, Zhao L, Fan B, et al. Doxorubicin-loaded nanoparticle coated with endothelial cells-derived exosomes for immunogenic chemotherapy of glioblastoma. Bioeng Transl Med. 2021;6:e10203. https://doi.org/10.1002/btm2.10203.

    Article  CAS  PubMed  Google Scholar 

  156. Wang J, Tang W, Yang M, Yin Y, Li H, Hu F, et al. Inflammatory tumor microenvironment responsive neutrophil exosomes-based drug delivery system for targeted glioma therapy. Biomaterials. 2021;273:120784. https://doi.org/10.1016/j.biomaterials.2021.120784.

    Article  CAS  PubMed  Google Scholar 

  157. Zhu Z, Zhai Y, Hao Y, Wang Q, Han F, Zheng W, et al. Specific anti-glioma targeted-delivery strategy of engineered small extracellular vesicles dual-functionalised by Angiopep-2 and TAT peptides. J Extracell Vesicles. 2022;11:e12255. https://doi.org/10.1002/jev2.12255.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Zhu Q, Ling X, Yang Y, Zhang J, Li Q, Niu X, et al. Embryonic stem cells-derived exosomes endowed with Targeting properties as Chemotherapeutics Delivery vehicles for Glioblastoma Therapy. Adv Sci (Weinh). 2019;6:1801899. https://doi.org/10.1002/advs.201801899.

    Article  CAS  PubMed  Google Scholar 

  159. Jia G, Han Y, An Y, Ding Y, He C, Wang X, et al. NRP-1 targeted and cargo-loaded exosomes facilitate simultaneous imaging and therapy of glioma in vitro and in vivo. Biomaterials. 2018;178:302–16. https://doi.org/10.1016/j.biomaterials.2018.06.029.

    Article  CAS  PubMed  Google Scholar 

  160. Rehman FU, Liu Y, Yang Q, Yang H, Liu R, Zhang D, et al. Heme Oxygenase-1 targeting exosomes for temozolomide resistant glioblastoma synergistic therapy. J Controlled Release: Official J Controlled Release Soc. 2022;345:696–708. https://doi.org/10.1016/j.jconrel.2022.03.036.

    Article  CAS  Google Scholar 

  161. Liu X, Cao Z, Liu N, Gao G, Du M, Wang Y, et al. Kill two birds with one stone: Engineered exosome-mediated delivery of cholesterol modified YY1-siRNA enhances chemoradiotherapy sensitivity of glioblastoma. Front Pharmacol. 2022;13:975291. https://doi.org/10.3389/fphar.2022.975291.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Ou A, Yung WKA, Majd N. Molecular mechanisms of Treatment Resistance in Glioblastoma. Int J Mol Sci. 2020;22. https://doi.org/10.3390/ijms22010351.

  163. Decraene B, Yang Y, De Smet F, Garg AD, Agostinis P, De Vleeschouwer S. Immunogenic cell death and its therapeutic or prognostic potential in high-grade glioma. Genes Immun. 2022;23:1–11. https://doi.org/10.1038/s41435-021-00161-5.

    Article  PubMed  PubMed Central  Google Scholar 

  164. Wang T, Peng W, Du M, Chen Z. Immunogenic sonodynamic therapy for inducing immunogenic cell death and activating antitumor immunity. Front Oncol. 2023;13:1167105. https://doi.org/10.3389/fonc.2023.1167105.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. Li J, Wang X, Guo Y, Zhang Y, Zhu A, Zeng W, et al. Ginsenoside Rg3-engineered exosomes as effective delivery platform for potentiated chemotherapy and photoimmunotherapy of glioblastoma. Chem Eng J. 2023;471:144692. https://doi.org/10.1016/j.cej.2023.144692.

    Article  CAS  Google Scholar 

  166. Wang X, Ding H, Li Z, Peng Y, Tan H, Wang C, et al. Exploration and functionalization of M1-macrophage extracellular vesicles for effective accumulation in glioblastoma and strong synergistic therapeutic effects. Signal Transduct Target Ther. 2022;7:74. https://doi.org/10.1038/s41392-022-00894-3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  167. Garofalo M, Villa A, Rizzi N, Kuryk L, Rinner B, Cerullo V, et al. Extracellular vesicles enhance the targeted delivery of immunogenic oncolytic adenovirus and paclitaxel in immunocompetent mice. J Controlled Release: Official J Controlled Release Soc. 2019;294:165–75. https://doi.org/10.1016/j.jconrel.2018.12.022.

    Article  CAS  Google Scholar 

  168. Harshyne LA, Hooper KM, Andrews EG, Nasca BJ, Kenyon LC, Andrews DW, et al. Glioblastoma exosomes and IGF-1R/AS-ODN are immunogenic stimuli in a translational research immunotherapy paradigm. Cancer Immunol Immunother. 2015;64:299–309. https://doi.org/10.1007/s00262-014-1622-z.

    Article  CAS  PubMed  Google Scholar 

  169. Naseri M, Bozorgmehr M, Zöller M, Ranaei Pirmardan E, Madjd Z. Tumor-derived exosomes: the next generation of promising cell-free vaccines in cancer immunotherapy. Oncoimmunology. 2020;9:1779991. https://doi.org/10.1080/2162402x.2020.1779991.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Bu N, Wu H, Sun B, Zhang G, Zhan S, Zhang R, et al. Exosome-loaded dendritic cells elicit tumor-specific CD8 + cytotoxic T cells in patients with glioma. J Neurooncol. 2011;104:659–67. https://doi.org/10.1007/s11060-011-0537-1.

    Article  CAS  PubMed  Google Scholar 

  171. Liu H, Chen L, Liu J, Meng H, Zhang R, Ma L, et al. Co-delivery of tumor-derived exosomes with alpha-galactosylceramide on dendritic cell-based immunotherapy for glioblastoma. Cancer Lett. 2017;411:182–90. https://doi.org/10.1016/j.canlet.2017.09.022.

    Article  CAS  PubMed  Google Scholar 

  172. Taghikhani A, Farzaneh F, Sharifzad F, Mardpour S, Ebrahimi M, Hassan ZM. Engineered Tumor-Derived Extracellular vesicles: potentials in Cancer Immunotherapy. Front Immunol. 2020;11:221. https://doi.org/10.3389/fimmu.2020.00221.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  173. Hu W, Huang F, Ning L, Hao J, Wan J, Hao S. Enhanced immunogenicity of leukemia-derived exosomes via transfection with lentiviral vectors encoding costimulatory molecules. Cell Oncol (Dordr). 2020;43:889–900. https://doi.org/10.1007/s13402-020-00535-3.

    Article  CAS  PubMed  Google Scholar 

  174. Cho JA, Lee YS, Kim SH, Ko JK, Kim CW. MHC independent anti-tumor immune responses induced by Hsp70-enriched exosomes generate tumor regression in murine models. Cancer Lett. 2009;275:256–65. https://doi.org/10.1016/j.canlet.2008.10.021.

    Article  CAS  PubMed  Google Scholar 

  175. Shi X, Sun J, Li H, Lin H, Xie W, Li J, et al. Antitumor efficacy of interferon-γ-modified exosomal vaccine in prostate cancer. Prostate. 2020;80:811–23. https://doi.org/10.1002/pros.23996.

    Article  CAS  PubMed  Google Scholar 

  176. Ghorbaninezhad F, Alemohammad H, Najafzadeh B, Masoumi J, Shadbad MA, Shahpouri M, et al. Dendritic cell-derived exosomes: a new horizon in personalized cancer immunotherapy? Cancer Lett. 2023;562:216168. https://doi.org/10.1016/j.canlet.2023.216168.

    Article  CAS  PubMed  Google Scholar 

  177. Bu N, Wu H, Zhang G, Zhan S, Zhang R, Sun H, 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;56:631–43. https://doi.org/10.1007/s12031-015-0506-9.

    Article  CAS  PubMed  Google Scholar 

  178. Li J, Li J, Peng Y, Du Y, Yang Z, Qi X. Dendritic cell derived exosomes loaded neoantigens for personalized cancer immunotherapies. J Controlled Release: Official J Controlled Release Soc. 2023;353:423–33. https://doi.org/10.1016/j.jconrel.2022.11.053.

    Article  CAS  Google Scholar 

  179. Zuo B, Zhang Y, Zhao K, Wu L, Qi H, Yang R, et al. Universal immunotherapeutic strategy for hepatocellular carcinoma with exosome vaccines that engage adaptive and innate immune responses. J Hematol Oncol. 2022;15:46. https://doi.org/10.1186/s13045-022-01266-8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  180. Cheng L, Wang Y, Huang L. Exosomes from M1-Polarized macrophages Potentiate the Cancer Vaccine by creating a pro-inflammatory microenvironment in the Lymph Node. Mol Ther. 2017;25:1665–75. https://doi.org/10.1016/j.ymthe.2017.02.007.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  181. Wei J, Chen P, Gupta P, Ott M, Zamler D, Kassab C, et al. Immune biology of glioma-associated macrophages and microglia: functional and therapeutic implications. Neurooncology. 2020;22:180–94. https://doi.org/10.1093/neuonc/noz212.

    Article  CAS  Google Scholar 

  182. Vidyarthi A, Agnihotri T, Khan N, Singh S, Tewari MK, Radotra BD, et al. Predominance of M2 macrophages in gliomas leads to the suppression of local and systemic immunity. Cancer Immunol Immunother. 2019;68:1995–2004. https://doi.org/10.1007/s00262-019-02423-8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  183. Li C, Guan N, Liu F. T7 peptide-decorated exosome-based nanocarrier system for delivery of Galectin-9 siRNA to stimulate macrophage repolarization in glioblastoma. J Neurooncol. 2023;162:93–108. https://doi.org/10.1007/s11060-023-04257-y.

    Article  CAS  PubMed  Google Scholar 

  184. Cui J, Wang X, Li J, Zhu A, Du Y, Zeng W, et al. Immune exosomes Loading Self-assembled Nanomicelles Traverse the blood-brain barrier for chemo-immunotherapy against Glioblastoma. ACS Nano. 2023. https://doi.org/10.1021/acsnano.2c10219.

    Article  PubMed  PubMed Central  Google Scholar 

  185. Adamus T, Hung CY, Yu C, Kang E, Hammad M, Flores L, et al. Glioma-targeted delivery of exosome-encapsulated antisense oligonucleotides using neural stem cells. Mol Ther Nucleic Acids. 2022;27:611–20. https://doi.org/10.1016/j.omtn.2021.12.029.

    Article  CAS  PubMed  Google Scholar 

  186. Shi Q, Zhao L, Xu C, Zhang L, Zhao H. High Molecular Weight Hyaluronan suppresses macrophage M1 polarization and enhances IL-10 production in PM(2.5)-Induced Lung inflammation. Molecules. 2019;24. https://doi.org/10.3390/molecules24091766.

  187. Feng C, Xiong Z, Wang C, Xiao W, Xiao H, Xie K, et al. Folic acid-modified Exosome-PH20 enhances the efficiency of therapy via modulation of the tumor microenvironment and directly inhibits tumor cell metastasis. Bioact Mater. 2021;6:963–74. https://doi.org/10.1016/j.bioactmat.2020.09.014.

    Article  CAS  PubMed  Google Scholar 

  188. Kim J, Zhu Y, Chen S, Wang D, Zhang S, Xia J, et al. Anti-glioma effect of ginseng-derived exosomes-like nanoparticles by active blood-brain-barrier penetration and tumor microenvironment modulation. J Nanobiotechnol. 2023;21:253. https://doi.org/10.1186/s12951-023-02006-x.

    Article  CAS  Google Scholar 

  189. Nie W, Wu G, Zhang J, Huang LL, Ding J, Jiang A, et al. Responsive Exosome Nano-bioconjugates for synergistic Cancer therapy. Angew Chem Int Ed Engl. 2020;59:2018–22. https://doi.org/10.1002/anie.201912524.

    Article  CAS  PubMed  Google Scholar 

  190. Choo YW, Kang M, Kim HY, Han J, Kang S, Lee JR, et al. M1 macrophage-derived nanovesicles potentiate the Anticancer efficacy of Immune Checkpoint inhibitors. ACS Nano. 2018;12:8977–93. https://doi.org/10.1021/acsnano.8b02446.

    Article  CAS  PubMed  Google Scholar 

  191. Movahedi K, Laoui D, Gysemans C, Baeten M, Stangé G, Van den Bossche J, et al. Different tumor microenvironments contain functionally distinct subsets of macrophages derived from Ly6C(high) monocytes. Cancer Res. 2010;70:5728–39. https://doi.org/10.1158/0008-5472.Can-09-4672.

    Article  CAS  PubMed  Google Scholar 

  192. Gunassekaran GR, Poongkavithai Vadevoo SM, Baek MC, Lee B. M1 macrophage exosomes engineered to foster M1 polarization and target the IL-4 receptor inhibit tumor growth by reprogramming tumor-associated macrophages into M1-like macrophages. Biomaterials. 2021;278:121137. https://doi.org/10.1016/j.biomaterials.2021.121137.

    Article  CAS  PubMed  Google Scholar 

  193. Flores-Toro JA, Luo D, Gopinath A, Sarkisian MR, Campbell JJ, Charo IF, et al. CCR2 inhibition reduces tumor myeloid cells and unmasks a checkpoint inhibitor effect to slow progression of resistant murine gliomas. Proc Natl Acad Sci U S A. 2020;117:1129–38. https://doi.org/10.1073/pnas.1910856117.

    Article  CAS  PubMed  Google Scholar 

  194. Alban TJ, Bayik D, Otvos B, Rabljenovic A, Leng L, Jia-Shiun L, et al. Glioblastoma myeloid-derived suppressor cell Subsets Express Differential Macrophage Migration Inhibitory factor receptor profiles that can be targeted to reduce Immune suppression. Front Immunol. 2020;11:1191. https://doi.org/10.3389/fimmu.2020.01191.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  195. Ban Y, Mai J, Li X, Mitchell-Flack M, Zhang T, Zhang L, et al. Targeting Autocrine CCL5-CCR5 Axis reprograms Immunosuppressive myeloid cells and reinvigorates Antitumor Immunity. Cancer Res. 2017;77:2857–68. https://doi.org/10.1158/0008-5472.Can-16-2913.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  196. Koinis F, Vetsika EK, Aggouraki D, Skalidaki E, Koutoulaki A, Gkioulmpasani M, et al. Effect of First-Line treatment on myeloid-derived suppressor cells’ subpopulations in the Peripheral blood of patients with Non-small Cell Lung Cancer. J Thorac Oncol. 2016;11:1263–72. https://doi.org/10.1016/j.jtho.2016.04.026.

    Article  PubMed  Google Scholar 

  197. Greene S, Robbins Y, Mydlarz WK, Huynh AP, Schmitt NC, Friedman J, et al. Inhibition of MDSC trafficking with SX-682, a CXCR1/2 inhibitor, enhances NK-Cell Immunotherapy in Head and Neck Cancer models. Clin Cancer Res. 2020;26:1420–31. https://doi.org/10.1158/1078-0432.Ccr-19-2625.

    Article  CAS  PubMed  Google Scholar 

  198. Jones T, Zhang B, Major S, Webb A. All-trans retinoic acid eluting poly(diol citrate) wafers for treatment of glioblastoma. J Biomed Mater Res B Appl Biomater. 2020;108:619–28. https://doi.org/10.1002/jbm.b.34416.

    Article  CAS  PubMed  Google Scholar 

  199. da Costa Rosa M, Yamashita AS, Riggins GJ. Evaluation of a DNA demethylating agent in combination with all-trans retinoic acid for IDH1-mutant gliomas. Neurooncology. 2022;24:711–23. https://doi.org/10.1093/neuonc/noab263.

    Article  CAS  Google Scholar 

  200. Haque A, Banik NL, Ray SK. Emerging role of combination of all-trans retinoic acid and interferon-gamma as chemoimmunotherapy in the management of human glioblastoma. Neurochem Res. 2007;32:2203–9. https://doi.org/10.1007/s11064-007-9420-z.

    Article  CAS  PubMed  Google Scholar 

  201. Fujita M, Kohanbash G, Fellows-Mayle W, Hamilton RL, Komohara Y, Decker SA, et al. COX-2 blockade suppresses gliomagenesis by inhibiting myeloid-derived suppressor cells. Cancer Res. 2011;71:2664–74. https://doi.org/10.1158/0008-5472.Can-10-3055.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  202. Veglia F, Sanseviero E, Gabrilovich DI. Myeloid-derived suppressor cells in the era of increasing myeloid cell diversity. Nat Rev Immunol. 2021;21:485–98. https://doi.org/10.1038/s41577-020-00490-y.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  203. Peereboom DM, Alban TJ, Grabowski MM, Alvarado AG, Otvos B, Bayik D, et al. Metronomic capecitabine as an immune modulator in glioblastoma patients reduces myeloid-derived suppressor cells. JCI Insight. 2019;4. https://doi.org/10.1172/jci.insight.130748.

  204. Qiu W, Guo Q, Guo X, Wang C, Li B, Qi Y, et al. Mesenchymal stem cells, as glioma exosomal immunosuppressive signal multipliers, enhance MDSCs immunosuppressive activity through the miR-21/SP1/DNMT1 positive feedback loop. J Nanobiotechnol. 2023;21:233. https://doi.org/10.1186/s12951-023-01997-x.

    Article  CAS  Google Scholar 

  205. Zhang X, Liu Y, Dai L, Shi G, Deng J, Luo Q, et al. BATF2 prevents glioblastoma multiforme progression by inhibiting recruitment of myeloid-derived suppressor cells. Oncogene. 2021;40:1516–30. https://doi.org/10.1038/s41388-020-01627-y.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  206. Amoozgar Z, Kloepper J, Ren J, Tay RE, Kazer SW, Kiner E, et al. Targeting Treg cells with GITR activation alleviates resistance to immunotherapy in murine glioblastomas. Nat Commun. 2021;12:2582. https://doi.org/10.1038/s41467-021-22885-8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  207. Sun Q, Yang Z, Lin M, Peng Y, Wang R, Du Y, et al. Phototherapy and anti-GITR antibody-based therapy synergistically reinvigorate immunogenic cell death and reject established cancers. Biomaterials. 2021;269:120648. https://doi.org/10.1016/j.biomaterials.2020.120648.

    Article  CAS  PubMed  Google Scholar 

  208. Guo D, Chen Y, Wang S, Yu L, Shen Y, Zhong H, et al. Exosomes from heat-stressed tumour cells inhibit tumour growth by converting regulatory T cells to Th17 cells via IL-6. Immunology. 2018;154:132–43. https://doi.org/10.1111/imm.12874.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  209. Tay C, Tanaka A, Sakaguchi S. Tumor-infiltrating regulatory T cells as targets of cancer immunotherapy. Cancer Cell. 2023;41:450–65. https://doi.org/10.1016/j.ccell.2023.02.014.

    Article  CAS  PubMed  Google Scholar 

  210. Sharma P, Aaroe A, Liang J, Puduvalli VK. Tumor microenvironment in glioblastoma: current and emerging concepts. Neurooncol Adv. 2023;5:vdad009. https://doi.org/10.1093/noajnl/vdad009.

    Article  PubMed  PubMed Central  Google Scholar 

  211. Sattiraju A, Kang S, Giotti B, Chen Z, Marallano VJ, Brusco C, et al. Hypoxic niches attract and sequester tumor-associated macrophages and cytotoxic T cells and reprogram them for immunosuppression. Immunity. 2023;56:1825–43. https://doi.org/10.1016/j.immuni.2023.06.017.

    Article  CAS  PubMed  Google Scholar 

  212. Wu T, Liu Y, Cao Y, Liu Z. Engineering Macrophage Exosome disguised biodegradable nanoplatform for enhanced Sonodynamic Therapy of Glioblastoma. Adv Mater. 2022;34:e2110364. https://doi.org/10.1002/adma.202110364.

    Article  CAS  PubMed  Google Scholar 

  213. Huo H, Yang S, Wu H, Sun Y, Zhao R, Ye R, et al. Brain endothelial cells-derived extracellular vesicles overexpressing ECRG4 inhibit glioma proliferation through suppressing inflammation and angiogenesis. J Tissue Eng Regen Med. 2021;15:1162–71. https://doi.org/10.1002/term.3244.

    Article  CAS  PubMed  Google Scholar 

  214. Zhuang X, Xiang X, Grizzle W, Sun D, Zhang S, Axtell RC, et al. Treatment of brain inflammatory diseases by delivering exosome encapsulated anti-inflammatory drugs from the nasal region to the brain. Mol Ther. 2011;19:1769–79. https://doi.org/10.1038/mt.2011.164.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  215. National Library of Medicine. https://clinicaltrials.gov/. Accessed 4 March 2024.

  216. Debbi L, Guo S, Safina D, Levenberg S. Boosting extracellular vesicle secretion. Biotechnol Adv. 2022;59:107983. https://doi.org/10.1016/j.biotechadv.2022.107983.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  217. Kalimuthu S, Gangadaran P, Rajendran RL, Zhu L, Oh JM, Lee HW, et al. A New Approach for Loading Anticancer drugs into mesenchymal stem cell-derived Exosome Mimetics for Cancer Therapy. Front Pharmacol. 2018;9:1116. https://doi.org/10.3389/fphar.2018.01116.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  218. Shimabukuro-Vornhagen A, Gödel P, Subklewe M, Stemmler HJ, Schlößer HA, Schlaak M, et al. Cytokine release syndrome. J Immunother Cancer. 2018;6:56. https://doi.org/10.1186/s40425-018-0343-9.

    Article  PubMed  PubMed Central  Google Scholar 

  219. Liang Y, Duan L, Lu J, Xia J. Engineering exosomes for targeted drug delivery. Theranostics. 2021;11:3183–95. https://doi.org/10.7150/thno.52570.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  220. Kalra H, Adda CG, Liem M, Ang CS, Mechler A, Simpson RJ, et al. Comparative proteomics evaluation of plasma exosome isolation techniques and assessment of the stability of exosomes in normal human blood plasma. Proteomics. 2013;13:3354–64. https://doi.org/10.1002/pmic.201300282.

    Article  CAS  PubMed  Google Scholar 

  221. Mendt M, Kamerkar S, Sugimoto H, McAndrews KM, Wu CC, Gagea M, et al. Generation and testing of clinical-grade exosomes for pancreatic cancer. JCI Insight. 2018;3. https://doi.org/10.1172/jci.insight.99263.

Download references

Acknowledgements

We would like to thank Yang Jiao for valuable suggestions, Editage (www.editage.cn) for English language editing, and Figdraw (www.figdraw.com) for figure drawing.

Funding

This work was supported by the National Natural Science Foundation of China (No. 82072953, 81471308 and 81802819), the Liaoning Province Science and Technology Project (No. 2021JH2/10300135), Dalian Science and Technology Talent Innovation Support Project (No.2022RG18 and 2023RY034), and Dalian Science and Technology Innovation Fund (No. 2018J11CY025 and 2022JJ13SN064).

Author information

Authors and Affiliations

Authors

Contributions

XQH wrote and revised the original draft. SMW drew figures and tables. LW and JQL reviewed and edited the draft. YL and JL performed project management, provided resources, and confirmed the final draft. All authors reviewed the manuscript.

Corresponding authors

Correspondence to Ying Li or Jing Liu.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hao, X., Wang, S., Wang, L. et al. Exosomes as drug delivery systems in glioma immunotherapy. J Nanobiotechnol 22, 340 (2024). https://doi.org/10.1186/s12951-024-02611-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12951-024-02611-4

Keywords