452
Views
0
CrossRef citations to date
0
Altmetric
Novel Vaccines

Exosome may be the next generation of promising cell-free vaccines

, , , , & ORCID Icon
Article: 2345940 | Received 12 Jan 2024, Accepted 18 Apr 2024, Published online: 07 May 2024

ABSTRACT

Traditional vaccines have limits against some persistent infections and pathogens. The development of novel vaccine technologies is particularly critical for the future. Exosomes play an important role in physiological and pathological processes. Exosomes present many advantages, such as inherent capacity being biocompatible, non-toxic, which make them a more desirable candidate for vaccines. However, research on exosomes are in their infancy and the barriers of low yield, low purity, and weak targeting of exosomes limit their applications in vaccines. Accordingly, further exploration is necessary to improve these problems and subsequently facilitate the functional studies of exosomes. In this study, we reviewed the origin, classification, functions, modifications, separation and purification, and characterization methods of exosomes. Meanwhile, we focused on the role and mechanism of exosomes for cancer and COVID-19 vaccines.

Introduction

In the last century, vaccinations have been one of the greatest public health achievements. Vaccines are critical tools to reduce morbidity and mortality caused by communicable diseases. It has been reported that vaccines save at least 2–3 million lives annually around the world. Smallpox has been eradicated and polio has almost disappeared worldwide, owing to the use of vaccines.Citation1 Traditional vaccine technologies have been widely used for bacterial and viral pathogens, yet they failed to cure some persistent infections and pathogens with high sequence variability, complex viral antigens, and emerging pathogens.Citation2 It is essential for developing novel vaccine technology. Exosomes not only can effectively deliver vaccine ingredients in a range of communicable diseases,Citation3 but also transfer genes, lipids, proteins and RNA between cells, as well as antigen presentation. They also regulate physiological and pathological processes. Meanwhile, their inherent capacity being biocompatible, Citation4–8 non-toxic,Citation9 as well as possessing the ability to cross biological barriers (such as the blood–brain barrier)Citation10,Citation11 and penetrate into several tissues through their surface ligands and receptors.Citation12 The findings of these studies suggest that exosomes modified by different methods will have the potential as biomarkers, vaccines, and drug carriers.Citation13 Exosomes are superior to other nanoparticles in terms of biosafety, and currently increasing evidences suggesting that exosomes would serve as the basis for the creation of cell-free cancer vaccines in the future.Citation14,Citation15

Composition and biogenesis of exosomes

Exosomes, which are extracellular vesicles of about 30–150 nm are secreted by almost all mammalian cells.Citation16 Exosomes originate from the initial endosomes. The first invagination of the cell membrane forms the early sorting endosomes, which form late sorting endosomes under the action of endocytic sorting complexes and associated transport proteins. A second invagination of late sorting endosomes results in the formation of multivesicular bodies. Subsequently, exosomes are released into the extracellular environment once the multivesicular bodies fuse with the plasma membrane.Citation7,Citation17 Exosomes are taken up by various cells after being released into the extracellular environment and bind to target cells via receptor–ligand interactions or lipids adhesion to cells, and then they are fully internalized by endocytosis, pinocytosis, phagocytosis or fuse to the cell surface membrane,Citation18 thereby exosomes release cargo into target cells and exerting biological functions.Citation3 The cargo carried by exosomes contain abundant enzymes, transcription factors, heat shock proteins, major histocompatibility complexes, cytoskeletal components, signal transduction proteins, four transmembrane proteins, lipids, RNA, and DNA.Citation19,Citation20 It is reported that exosomes are involved in clearance of unnecessary proteins or molecules in the cells, the substance exchange between cells, intercellular communication, pathogen transmission, the regulation of immune system, and antigen presentation.Citation21

Exosomes are categorized into natural exosomes and artificial exosomes depending on whether they are artificially modified or not. Natural exosomes are subdivided into animal-derived and plant-derived. Almost all types of animal cells secrete exosomes,Citation7,Citation22 including tumor cells, mast cells, dendritic cells, mesenchymal stem cells, B cells, T cells, macrophages, natural killer cells, neurons, adipocytes, endothelial cells, and epithelial cells.Citation23,Citation24 Plant-derived exosomes emerge as a new therapeutic optionCitation25 and are currently promising,Citation7 such as ginger exosome-like nanoparticles (GELN-RNA) which improve colitis in mice via IL-22-dependent mechanisms,Citation26 and GELN microRNA which eliminates exosomesNsp12Nsp13–mediated inflammation in the lungs.Citation27 In addition, bacteria-derived exosomes are involved in the disease process. For instance, mycobacterium tuberculosis-derived exosomes may play a significant role in the pathogenesis of tuberculosis by delivering mycobacterium tuberculosis components to the recipient cells.Citation28

Exosome engineering

The appropriate properties of exosomes make them well suited as a drug delivery vehicle, but their clinical application faces several challenges, such as a limited half-life, and low solubility.Citation29 Natural exosomes lack particular cell or tissue targeting properties. In order to optimize the targeting of exosomes and overcome the limitations of using autologous, hence numerous studies showed that engineered exosomes enhanced anticancer immunogenicity by expressing specific antigenic molecules.Citation30,Citation31 The engineered exosome present multiple antigens and can be designed in two ways, parental cell-based modification and post-isolation modification.Citation31 In contrast to parental cell-based exosomes, post-isolated exosomes are technically less complex.Citation31 Typical manipulations which include electroporation,Citation32,Citation33 extrusion, ultrasound, incubation, and freeze-thawCitation34 can be directly applied to post-isolated exosome engineering.

Parental cell-based exosome modification occurs before exosomes are isolated from parental cells.Citation31 The most common modification approach is to transfer proteins to the exosome surface exploiting exosome signal peptides. Another approach is to load molecules into the lumen of the exosomes, which is accomplished by recruiting molecular sorting modules (MSMs) that bind to proteins or RNAs of interest and drives them to the exosome to sort proteins and RNA entering the lumen. Moreover, RNA loading methods are also used in parental cell-based exosome engineering. There is a device for loading mRNAs called EXOtic (Exosomal Transfer Into Cells). In the EXOtic device, there is also a cytoplasmic delivery assistance system and a targeting module in addition to the RNA packaging system. These systems work together to enable the loading of therapeutic mRNAs into exosomes.Citation31

Parental cells can be transfected by vectors to generate surface-modified exosomes that can express targeting moieties via a natural biosynthesis process.Citation35 The indirect exosome modification possesses some advantages over direct exosome modification in terms of expression and stability displayed targeting moieties of engineered exosome.Citation36 Next, we discussed some details about genetically engineered exosomes for targeting cancer. For example, the C1C2 domain of lactadherin can be used to express anti-Her2 single-chain variable fragments (scFv) on the surface of therapeutic exosomes. Parental cells were genetically engineered to generate exosomes of Her2-targeting by expressing fusion proteins of anti-Her2 scFv that was fused to the C-terminus on the C1C2 domain of lactadherin, and N-terminus used signal peptide to introduce fusion proteins into the secretory pathway thus inducing it to bind to extramembrane of the exosome. Compared to non-engineered exosomes, engineered exosomes exhibited the better uptake by breast cancer cells expressing HER2 gene, showing two-fold and two to three-fold higher accumulation in vivo.Citation37

Direct exosome gene modifications can be categorized as membrane protein-based gene modifications and lipid–protein interaction-based gene modifications. Many membrane proteins of exosomes such as Lamp2b and tetraspanins, which can be applied to display targeting moieties.Citation38 For example, a study described that exosomes modified by the insertion of ApoB between amino acids 170–171 of CD9 facilitated BBB penetration via hijacking receptor-mediated transcytosisCitation39 and the engineered exosome prolonged the residence in the brain after intravenous administration.Citation38

Another approach to direct exosome engineering is chemical modification, which loads various types of ligands onto the exosome membrane by means of conjugation and hydrophobic insertion.Citation29 Exosome membranes are composed of lipids and proteins, which can be chemically modified by lipid insertion, chemical ligation, affinity binding, and enzymatic conjugation. The exosome membrane allows hydrophobic insertion of lipids and lipid-labeled molecules, and targeted peptides labeled with lipid fragments and aptamers can be inserted into the exosome membrane by a simple mixing and incubation.Citation38 DSPE-PEG is a widely used module to anchor targeting molecules on the surface of exosomes. A study showed that exosomes modified with the application of DSPE-PEG overcame the BBB and targeted delivery drugs to treat glioblastoma.Citation40 Chemical ligation is a way based on reactive groups on lipids or proteins of exosome membrane that can react with reactive fragment-labeled peptides thereby allowing the target peptide to modify the exosome. Affinity binding is a method by which targeting moieties are attached to affinity molecules on proteins or lipids of exosomal membrane. Enzymatic conjugation is an approach based on protein ligase, and surface modifications of exosomes, which is achieved by enzymatic reactions between exosomal membrane proteins and target proteins/peptides.Citation38

In addition, structural alteration is another strategy to improve the effectiveness of exosomes. Exosome-liposome hybridization is used to optimize the surface characteristics of exosomes, and the hybridization strategy can modify immunogenicity, improve colloidal stability, increase circulation time in the blood, and elevate target cell uptake.Citation36,Citation41 The hybridization through PEG can avoid the attack of immune cells by forming a hydration layer, therefore, engineered exosomes achieve a higher stability and longer circulation time.Citation42 In addition, exosomes modified with magnetic compounds can be effectively applied for exosome detection, isolation and targeted drug delivery. For example, a novel therapeutic platform was established by combining magnetic targeting characteristics and drug delivery capabilities of magnetic nanoparticles with BBB penetration capabilities and siRNA encapsulation properties of engineered exosomes for the treatment of Gloiblastoma.Citation43

Although cell resources are crucial for biodistribution of exosomes in vivo, a large proportion of injected exosomes are distributed in the reticuloendothelial system, including the lungs, liver, spleen, and gastrointestinal tract, and exosomes are eliminated by them, resulting in a short half-life circulation. In addition, macrophage capture also plays a major role in exosome clearance.Citation29,Citation44 Targeting of exosomes, which not only increases the effectiveness of exosome delivery, but also reduces out-of-target side effects.Citation45 Further clinical applications of exosomes should focus on their manipulation to increase their lifetime in circulation while reducing immune clearance. A study showed that ‘invisible’ exosomes to the immune system through the incorporation of different anti-phagocytic molecules (including CD47, CD24, CD44, etc.) on the exosomes’ surface allow for better systemic bioavailability due to longer residence time in circulation.Citation46 For instance, the exosomes containing CD47 achieve protection against phagocytosis by interacting with α-ligand signal-regulating protein.Citation47

Exosome isolation and characterization

Exosome isolation

Exosome isolation techniques have developed dramatically in recent decades. Currently, methods of exosome isolation and purification usually include ultracentrifugation, ultrafiltration, size-exclusion chromatography (SEC), polymer precipitation, immunoaffinity capture techniques, and microfluidic techniques, which can be combined to achieve better results.Citation48 shows the comparative advantages and disadvantages of exosome isolation methods.

Table 1. Advantages and disadvantages of exosome isolation methods.

Ultracentrifugation is the most commonly used exosome separation techniques, which is suitable for the majority of samples and is regarded as the ‘gold standard’ for exosome isolation because of its mature technology and low-cost.Citation49 Differential ultracentrifugation (DUC) and density gradient ultracentrifugation (DGUC) are the two alternative isolation techniques.Citation50 Differential ultracentrifugation is low-cost. The drawbacks are too time-consuming, the equipment is costly, low recovery and purity,Citation51 and poor reproducibility. Furthermore, repeated centrifugation may also cause some damage to the exosome.Citation52,Citation53 The purity of extracellular products obtained by density gradient ultracentrifugation is higher than that by differential ultracentrifugation,Citation54 but its clinical applicability is confined due to the preliminary preparation required, low yield, cumbersome operation, and long centrifugation time (>16 h).Citation52,Citation55 Ultrafiltration is a separation method based on molecular size. It works by concentrating exosomes from a large volume of material to a small volume for further purification, which is simple and rapid, and the exosomes obtained are of intermediate yield.Citation50,Citation56 However, exosomes isolated by ultrafiltration may be deformed or break due to pressure. Additionally, it is prone to pore clogging.Citation53,Citation57 If this method is employed exclusively, exosomes are contaminated by non-exosomal free-floating humoral peptides such as albumin and α-1-antitrypsin.Citation57 Size-exclusion chromatography is a separation method based on variations in molecule size. This approach produces exosomes with high yield and purity,Citation58 and preserves the integrity and biological activity of exosomes since it utilizes gravity flow.Citation59 However, exosomes cannot be distinguished from vesicles of the same size. Polymer precipitation produces low purity exosomes with a high yield.Citation60 The exosomes extracted by this technique are susceptible to contamination by lipoproteins or virus particles to impact future analyses (such as proteomics and mass spectrometry). Recently, many commercial kits relying on precipitation technology have been used for exosome isolation, and the ExoQuick kit is widely used.Citation50 The immunoaffinity technique using magnetic beads offers a higher capture efficiency and greater sensitivity.Citation61 The immunoaffinity method is unaffected by exosome morphology and has the characteristics of high specificity and purity, intermediate yield.Citation60 However, the exosomes isolated by this method are only subtypes with positive markers and do not include all types of exosomes.Citation62 This method is time-consuming because of the time required for antigen–antibody binding. In addition, extra steps are required to isolate and purify the exosomes after antibody binding.Citation63 Microfluidics are considered a promising approach that integrate sample processing, analysis, monitoring, and other processes on a chip, resulting in miniaturized, high-throughput capabilities which is less time-consuming.Citation64 It is able to distinguish, capture, enrich, and separate particles of very similar shape and size.Citation65 This separation technology has the advantages of minimal feed volumes, high throughput, and quick sample processing, along with high purity and sensitivity.Citation52 Nevertheless, the procedure is costly, complexCitation50 and unsuitable for large-scale production.Citation63

Characterization of exosomes

EVs are similar in size and morphology and it is indispensable to distinguish different EVs subtypes by using some characterization methods after EVs are isolated. In particular, some certain markers are utilized to determine whether the extracted material is an exosome or not, so as to verify the method of isolation of exosomes. Based on these characteristics, the methods for characterizing exosomes can be briefly classified as quantitative exosome analysis methods, qualitative exosome characterization methods, and single vesicle characterization methods.Citation66

Total exosome count, protein count, lipid count, and DNA/RNA count are used for quantitative characterization of exosomes. Total exosome counts are frequently detected via nanoparticle tracking analysis (NTA), flow cytometry (FCM), dynamic light scattering (DLS), and tunable resistive pulse sensing (TRPS). Lipid count has been achieved by using techniques, such as fluorescence microscopy and fourier-transform infrared spectroscopy (FTIR).Citation66 Mass spectrometry, western blot (WB), and enzyme-linked immunosorbent analysis (ELISA) are used for protein count. For RNA counting, polymerase chain reaction (PCR), microarray analysis, and next-generation sequencing (NGS) are utilized.Citation67,Citation68 Protein markers, lipid markers and DNA/RNA markers are used for qualitative characterization of exosomes. Protein markers are often identified using mass spectrometry, flow cytometry (FCM), and enzyme-linked immunosorbent analysis (ELISA). For the recognition of DNA/RNA markers, PCR, microarray analysis, and next-generation sequencing (NGS) are employed. Additionally, mass spectrometry is used to detect lipid markers. When evaluating individual vesicles, structure, size, and chemical composition are prominent aspects to consider. The structure of exosomes has been visualized by applying scanning electron microscope (SEM), transmission electron microscope (TEM), cryo-electron microscope (cryo-EM), and atomic force microscope (AFM).Citation69 The size of exosome has been measured via nanoparticle tracking analysis (NTA), flow cytometry (FCM), dynamic light scatterin (DLS), and tunable resistive pulse sensing (TRPS). Raman Spectroscopy is used to determine chemical composition.Citation66 There are also a number of individual or combined methods used to perform exosome assays, such as surface plasmonic biosensors, microchip-based technologies, electrochemical techniques, fluorescence, and colorimetry.Citation56 shows the common exosome characterization methods.

Figure 1. The common exosome characterization techniques.

ELISA, enzyme-linked immunosorbent analysis; PCR, polymerase chain reaction; NGS, next-generation sequencing; SEM, scanning electron microscope; TEM, transmission electron microscope; cryo-EM, cryo-electron microscope; AFM, atomic force microscope; NTA, nanoparticle tracking analysis; DLS, dynamic light scatterin; TRPS, tunable resistive pulse sensing; WB, western blot; FTIR, fourier-transform infrared spectroscopy; ddPCR, droplet digital PCR.
Figure 1. The common exosome characterization techniques.

Exosomes and vaccines

Exosomes have attracted great interest for their ability to stimulate dendritic cells recognizing and killing cancer cells.Citation14 In addition, exosomes transfer antigens into DC for action, causing the initiation and amplification of specific immune responses.Citation70 These characteristics make it a desirable candidate for cancer vaccines. In order to improve the targeting of exosomes, many studies have genetically engineered exosomes to express specific antigenic molecules or target cancer cells.Citation30 To achieve genetic modification of exosomes, a novel exosome platform named Synthetic Multivalent Antibody Retargeted Exosome (SMART-Exo) was designed and developed.Citation71,Citation72 describes the benefits and disadvantages of available vaccines and exosome-based vaccines.

Table 2. Advantages and disadvantages of vaccine types.

Currently, there are two ways to load proteins onto exosomes. One way is feeding antigens to dendritic cells (DCs) and then collecting the supernatants to separate exosomes for indirect loading of peptides, which ensures that antigen peptide binds to MHC molecules. MHC molecules are crucial for the interaction between DCs and T cells. However, the limitation of this strategy is that the type of antigen must be recognized by the DCs.Citation73 The other way is that any soluble antigen can be attached to the exosome and can be used to modify exosomes with additional functions, such as adding adhesion molecules to exosomes, which can be targeted to certain sites.Citation74

Exosomes and cancer vaccines

Exosomes can not only stimulate anti-tumor immune responses, but also inhibit anti-tumor immune responses to promote tumor growth through the ability of intercellular signaling and antigen presentation.Citation75 An appealing topic of study is how the tumor microenvironment is impacted by normal-derived and tumor-derived exosomes, which provides chances for the enhancement of antitumor immune responses, the development of exosomal vaccines, and the use of exosomes as drug nanocarriers.Citation17

In cancer immunotherapy, tumor-derived exosomes (TEX) which are rich in tumor antigens can be presented by APC to induce T and B cell responses. Tumor-derived exosomes (TEX) have emerged as a promising cell-free therapeutic vaccine in cancer immunotherapy.Citation76 TEXs are enriched with proteins such as major histocompatibility complex I and II (MHCI and MHCII), CD81, CD54, and CD63, which can be used for exosome binding and uptake by related proteins on DCs.Citation76 Meanwhile, heat shock proteins are presented on TEX, including 71 kDa heat shock protein, 70 kDa heat shock protein 4, and HSP90 α & β.Citation77 The heat shock protein (HSP) targets and activates antigen-presenting cells including dendritic cells (DCs), thus providing a natural link between the innate and adaptive immune responses.Citation78 HSPs have powerful adjuvant capabilities that enhance the immunogenicity of TEXs and improve the effectiveness of cancer vaccines.Citation76

The design protocol of the effective cancer vaccine must be able to trigger strong and sustained immunity mediated by T cells.Citation79 TEX contains tumor-associated antigens and participates in antigen presentation either directly or indirectly, triggering CD8+ T cell responses for cross-initiating immunotherapy.Citation80,Citation81 Nonetheless, TEXs-induced immunosuppression and limited TEXs immunogenicity, and frequent application of TEXs alone in vivo leads to unsatisfactory antitumor immune effects. Therefore, several strategies have been used to improve the effectiveness of TEXs vaccines. TEXs can be genetically or non-genetically modified to enrich tumor antigens, microRNAs, and immunostimulatory molecules on TEXs to directly enhance tumor cell death or to be killed by immune cells. Another strategy is to improve TEX vaccination by DC loading, TEX-loaded DCs promote the transformation of naïve CD8+T cells into mature antigen-specific CTL cells, induce the activation of NF-κB in macrophages, and participate in tumor cytotoxicity via the release of tumor necrosis factor (TNF).Citation76

In addition to tumor-derived exosomes, dendritic cell-derived exosomes (DCexos) also have the potential to be cell-free therapeutic vaccines due to their resistance to tumor immunosuppression, significant bioavailability, and biostability.Citation82 DCexos which are inert vesicles have a longer half-life in vivo and may be stored for a longer period of time. It is reported that DCexos are also more resistant to immunomodulation by the tumor or tumor microenvironment (TME) than DCs,Citation83 thus supporting their clinical application as tumor vaccines.Citation83–86

DCexos are enriched in MHC class I and II complexes, heat shock proteins (HSPs), and costimulatory molecules such as CD86, which stimulate the activation of CD4+ helper T cells and CD8+ CTLs and induce potent antitumor effects.Citation87–89 Some functional molecules such as MHC-I, MHC-II, CD40, CD80, CD86, and the FasL, TRAIL, and NKG2D (natural killer group 2D) ligands on the surface of DCexos contribute to the enhancement of both innate and adaptive anti-tumor immune responses.Citation90 In the antitumor response, DCexos also induce other immune cells such as NK cells.Citation91 NK cell effector function enhanced in clinical trials using modified DCexos to treat cancer, which suggest that DCexos may induce NK cell function in vivo.Citation91

It is reported that DCexos dramatically increased CD8+ T lymphocytes, resulting in elevating IFN-γ and IL-2, decreasing CD25+Foxp3+ Treg cells, and decreasing IL-10 and TGF-β at tumor site, thus improving the tumor microenvironment.Citation92 While in the tumor microenvironment, DCexos contain TNF, Fas ligand (FasL) and TNF-related apoptotic ligand (TRAIL) can cause apoptosis in tumor cells.Citation93 Meanwhile, DCexos activate natural killer cells through TNF superfamily ligands, thereby presenting an inhibitory effect on tumors.Citation94

Despite the fact that we have concentrated on DCexos as a tumor vaccine, exosomes derived from other cell such as macrophages, NK cells, B cells, T cells, mesenchymal stem cells, and tumor cells, have also been employed in the study of tumor vaccines.Citation93,Citation95 Normally, exosomes derived from immune cells promote the proliferation of T cells.Citation96 Exosomes are usually presented by mature DCs to elicit T cell responses because the antigens on exosomes must be taken up by DCs to activate T lymphocytes, which avoids the use of adjuvants to some extent, and exosomes can directly activate T lymphocytes.Citation97

During antigen presentation, B cell-derived exosomes first interact with antigen and then regulate T cell activation and cytokine secretion.Citation98,Citation99 Additionally, it is reported that cytotoxic T lymphocyte (CTL)-derived exosomes contain CTL surface membrane molecules (CD3, CD8 and TCR) that cause tumor cell death through TCR-MHC/antigen interactions.Citation100 NK cell-derived exosomes play a significant therapeutic function, which produce cytotoxic effects on melanomas by presenting perforin and FasL and participate in apoptosis in vivo and in vitro.Citation101 Exosomes from M1-polarized macrophage can enhance the activity of anti-tumor vaccines and promote CTL-related immune responses.Citation102,Citation103 Exosomes secreted by M1-polarized macrophages can be taken up by local macrophages and DCs in lymph nodes (LNs), increasing the release of Th1 cytokines and enhancing the CTL response. In melanoma, macrophage-derived exosomes enhance the efficacy of tumor polypeptide vaccines and significantly inhibit tumor growth.Citation104 shows the mechanism of action of tumor-derived exosomes and immune cell-derived exosomes on tumor cells.

Figure 2. The mechanism of action of tumor-derived exosomes and immune cell-derived exosomes on tumor cells.

(a) TEXs is modified to enrich tumor antigens, microRNAs, and immunostimulatory molecules on TEXs to enhance tumor cell death or to be killed by immune cells. (b) TEX is presented to CD8+ T cells by DC, ultimately leading to tumor cell death through cytotoxicity. (c) TEX activates the B cell response to produce antibodies, which eventually causes the tumor cells are phagocytosed by macrophages. (d) TEX-loaded DCs with activate macrophages, which participate in cytotoxicity by releasing TNF. (e) DCexo induces NK cell function and NK cell-derived exosome presents cytotoxic effects via perforin and FasL. (f) CTL cell-derived exosomes cause tumor cell death through TCR-MHC/antigen interactions.
Figure 2. The mechanism of action of tumor-derived exosomes and immune cell-derived exosomes on tumor cells.

Exosomes and COVID-19 vaccines

At present, the outbreak of the COVID-19 pandemic has accelerated the research on EVs, especially the development of EV-based vaccines.Citation105 As exosomes are essential intercellular communication molecules with immunogenicity, high biocompatibility and inherent cargo-loading capacity, they offer new opportunities for the development of COVID-19 vaccines and may be crucial in restricting the COVID-19 pandemic.Citation106 Extracellular vesicles (EVs) have several biological advantages over virus-vectors, such as endogenous origin, the ability to cross biological barriers, high stability, immunogenicity, and low toxicity. Several companies are experimenting and testing COVID-19 vaccines based on the EVs platform.Citation107 For example, a biologic company had researched two different COVID-19 vaccines.Citation106 One type of vaccine comprises human HEK293 cells transfected with vectors expressing four structural proteins (S, E, M and N proteins) of the SARS-CoV-2. The other type is an mRNA vaccine. EVs are loaded with five mRNAs encoding modified SARS-CoV-2 spike, nucleocapsid protein, membrane protein, cytosolic proteins, and full-length spike protein from the wuhan-1 isolate.Citation105 Another biologic company developed a vaccine in which induced pluripotent stem cell line were transfected with different mRNAs encoding SARS-CoV-2 antigen proteins. These cells produce a large amount of EVs that carry the viral mRNAs and corresponding antigen proteins. This type of vaccine possesses multiple mRNAs and is better than single mRNA vaccines. Another exoVACCTM vaccine is a modular vaccine system that takes advantages of EVs, which can simultaneously deliver specific antigens and immune-stimulating adjuvants to antigen-presenting cells (APCs) to elicit cellular and humoral immune responses. Recent reports have shown that exosomal vaccines loaded with mRNAs encoding SARS-CoV-2 spikes and nucleocapsid proteins induced long-lasting cellular and humoral immune responses and produced fewer adverse effects than the currently available COVID-19 vaccines.Citation108 Accoding to Kuate et al., the mice were initially immunized with an exosomal vaccine containing the SARS-S protein and then were immunized with an adenovirus vector vaccine expressing the S protein, their neutralizing antibody titers were higher than those in the serum of SARS patients. Moreover, both vaccines induced neutralizing antibody titers in mice.Citation109 Therefore, EV-based vaccine design protocols can be applied to the treatment of SARS-CoV-2.

Conclusions

Although EV-based vaccines present many advantages, there are still a large number of obstacles needed to be overcome in clinical applications. For example, production and isolation of exosomes on a large scale remains a great challenge. In terms of targeted therapies, the original structure of exosomes can be modified in a variety of methods, such as genetic engineering, chemical procedures, physical techniques, and microfluidic technology, which make additional cargo load on exosomes to expand the biomedical applications of exosomes.Citation110 In conclusion, the development of exosome modification techniques, automated manufacturing processes and strict quality control systems are pivotal to produce engineered exosomes of high purity on a large scale. A large number of cells are required to secrete sufficient exosomes for large-scale production. Bioreactors produce an enormous number of cells in a short period of time and the most popular bioreactors currently available are hollow fiber bioreactors and stirred tank bioreactors.Citation63 Exosome isolation can be achieved by different methods. Though conventional centrifugation technology is low-cost, it takes a long time. In addition, exosome extracted by this method are low purity. Ultrafiltration does not require expensive equipment and can be utilized for large-scale production. Nevertheless, isolation of exosomes from impurity proteins needs to be optimized. Size-exclusion chromatography technique preserves the integrity of exosomes, while it cannot distinguish between vesicles of the same density and size. Polymer precipitation method is simple to operate and easy to scale up. However, the polymer precipitation method is prone to be contaminated. Immunoaffinity isolation technique is highly specific, but requires extra isolation and purification steps after binding the antibody and is not suitable for large-scale production. In contrast, microfluidic technology, which takes the advantage of precise nanoscale liquid and particle control capabilities is faster and more efficient than the current techniques for exosome isolation. However, it is not appropriate for large-scale production. Methods of exosome isolation should ensure the integrity, sensitivity and specificity. The methods of exosome isolation or combinations should be optimized to obtain high-purity exosomes.

Exosome-based vaccines demonstrate impressive outcomes in the cancer and viral infectious diseases. Inactivated vaccines have no active ingredients and are technically mature, but a single dose is not sufficient to induce long-lasting immunity. Adenoviral vector vaccines induce multiple epitopes and can be produced on a large scale, but pre-existing anti-adenovirus immune responses have potential adverse events. DNA vaccines allow the construction of vectors to encode different antigens in a single vaccine, which can be mass-produced with high stability in production and storage. However, DNA vaccines can only elicit a weak immune response and require a delivery agent to reach the nucleus. RNA vaccines can quickly assemble novel mRNA sequences into existing vaccine formulations and there is no risk of integration with host cell genes. However, RNA vaccines are prone to rare and severe allergic reactions. Recombinant protein vaccines require adjuvants and are easy to produce on a large scale, however, they can only express protein fragments. Conversely, exosomes are effective carriers of viral antigens, which present antigens in their natural state and are able to cross the blood–brain barrier and generate a protective immune response. Compared to other carriers, such as lipid-based nanoparticles and viral carriers, exosomes have lower immunogenicity and higher absorptivity and are safely and efficiently employed in vaccines.Citation106

This review summarizes methods for large-scale production, isolation, and characterization of exosomes, and focuses on the anti-tumor mechanism of tumor-derived exosomes and immune cell-derived exosomes. These studies provide a pivotal basis for the development of exosome-based vaccines for cancer. As cell-free vaccines, exosomes have limitations in terms of therapeutic efficacy. Tumor-derived exosomes may carry immunosuppressive molecules and induce pro-tumor effect, which limits their role in anti-tumor immunotherapy. To address this issue, tumor-derived exosomes need to be modified to improve their anti-tumor immune response. More studies are needed to evaluate different modification techniques, screen out the optimal combinations of antigens and adjuvants, and evaluate the adverse molecular responses of EV in recipient cells, which will benefit to improving therapeutic efficacy and safety of exosomes in vaccines. In the future, exosome-based vaccines are expected to become a significant breakthrough in the field of cancer immunization and infectious diseases with increased research and technological advances.

Authors’ contribution

Conceptualization: Ruiru Cai; Hong Zeng; Hailian Zhu; Youwei Dou

Design: Shibo Sun

Writing-original draft: Zelan Dai

Writing-review: Shibo Sun

Disclosure statement

No potential conflict of interest was reported by the author(s).

Data availability statement

Data sharing is not applicable to this article as no datasets were generated or analyzed during the current study.

Additional information

Funding

This work was supported by the National Natural Science Foundation of China [grant no. 82160007], the Yunnan Provincial Science and Technology Department [grant no. 2022201AY070001-074], Yunnan Province young and middle-aged academic and technical leaders reserve talent project [grant no. 202305AC160017], 535 Talent Project of First Affiliated Hospital of Kunming Medical University [grant no. 2023535D12], and Student Innovation Fund project of Kunming Medical University [grant no. 2023CXD077, 2023CXD091, 2023CXD195, 2023CXD247, 2023CXD257, 2023CXD270].

References

  • Delany I, Rappuoli R, De Gregorio E. Vaccines for the 21st century. EMBO Mol Med. 2014;6(6):708–11. doi:10.1002/emmm.201403876.
  • Gebre MS, Brito LA, Tostanoski LH, Edwards DK, Carfi A, Barouch DH. Novel approaches for vaccine development. Cell. 2021;184(6):1589–603. doi:10.1016/j.cell.2021.02.030.
  • Sun YF, Pi J, Xu JF. Emerging role of exosomes in tuberculosis: from immunity regulations to vaccine and immunotherapy. Front Immunol. 2021;12:628973. doi:10.3389/fimmu.2021.628973.
  • Tang Y, Zhou Y, Li HJ. Advances in mesenchymal stem cell exosomes: a review. Stem Cell Res Ther. 2021;12(1):71. doi:10.1186/s13287-021-02138-7.
  • Lässer C. Exosomes in diagnostic and therapeutic applications: biomarker, vaccine and RNA interference delivery vehicle. Expert Opin Biol Ther. 2014;15(1):103–17. doi:10.1517/14712598.2015.977250.
  • Amiri A, Bagherifar R, Ansari Dezfouli E, Kiaie SH, Jafari R, Ramezani R. Exosomes as bio-inspired nanocarriers for RNA delivery: preparation and applications. J Transl Med. 2022;20(1):125. doi:10.1186/s12967-022-03325-7.
  • 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. doi:10.2147/IJN.S264498.
  • Greening DW, Xu R, Ji H, Tauro BJ, Simpson RJ. A protocol for exosome isolation and characterization: evaluation of ultracentrifugation, density-gradient separation, and immunoaffinity capture methods. Methods Mol Biol. 2015;1295:179–209. doi:10.1007/978-1-4939-2550-6_15.
  • Shahabipour F, Banach M, Sahebkar A. Exosomes as nanocarriers for siRNA delivery: paradigms and challenges. Arch Med Sci. 2016;12(6):1324–6. doi:10.5114/aoms.2016.62911.
  • Haney MJ, Klyachko NL, Zhao Y, Gupta R, Plotnikova EG, He Z, Patel T, Piroyan A, Sokolsky M, Kabanov AV, et al. Exosomes as drug delivery vehicles for Parkinson’s disease therapy. J Control Release. 2015;207:18–30. doi:10.1016/j.jconrel.2015.03.033.
  • El Andaloussi S, Lakhal S, Mäger I, Wood MJ. Exosomes for targeted siRNA delivery across biological barriers. Adv Drug Deliv Rev. 2013;65(3):391–7. doi:10.1016/j.addr.2012.08.008.
  • Headland SE, Jones HR, Norling LV, Kim A, Souza PR, Corsiero E, Gil CD, Nerviani A, Dell’accio F, Pitzalis C, et al. Neutrophil-derived microvesicles enter cartilage and protect the joint in inflammatory arthritis. Sci Transl Med. 2015;7(315):315ra190. doi:10.1126/scitranslmed.aac5608.
  • Heusermann W, Hean J, Trojer D, Steib E, von Bueren S, Graff-Meyer A, Genoud C, Martin K, Pizzato N, Voshol J, et al. Exosomes surf on filopodia to enter cells at endocytic hot spots, traffic within endosomes, and are targeted to the ER. J Cell Biol. 2016;213(2):173–84. doi:10.1083/jcb.201506084.
  • Kanlikilicer P. Exosome-related methods and potential use as vaccines. Methods Mol Biol. 2022;2435:35–41. doi:10.1007/978-1-0716-2014-4_4.
  • Natasha G, Gundogan B, Tan A, Farhatnia Y, Wu W, Rajadas J, Seifalian AM. Exosomes as immunotheranostic nanoparticles. Cli Ther. 2014;36(6):820–9. doi:10.1016/j.clinthera.2014.04.019.
  • El Safadi D, Mokhtari A, Krejbich M, Lagrave A, Hirigoyen U, Lebeau G, Viranaicken W, Krejbich-Trotot P. Exosome-mediated antigen delivery: unveiling novel strategies in viral infection control and vaccine design. Nato Adv Sci Inst Se. 2024;12(3):280. doi:10.3390/vaccines12030280.
  • Bae S, Brumbaugh J, Bonavida B. Exosomes derived from cancerous and non-cancerous cells regulate the anti-tumor response in the tumor microenvironment. Genes Cancer. 2018;9(3–4):87–100. doi:10.18632/genesandcancer.172.
  • Hendrix A, Hume AN. Exosome signaling in mammary gland development and cancer. Int J Dev Biol. 2011;55(7–9):879–87. doi:10.1387/ijdb.113391ah.
  • McAndrews KM, Kalluri R. Mechanisms associated with biogenesis of exosomes in cancer. Mol Cancer. 2019;18(1):52. doi:10.1186/s12943-019-0963-9.
  • Milane L, Singh A, Mattheolabakis G, Suresh M, Amiji MM. Exosome mediated communication within the tumor microenvironment. J Control Release. 2015;219:278–94. doi:10.1016/j.jconrel.2015.06.029.
  • Beach A, Zhang HG, Ratajczak MZ, Kakar SS. Exosomes: an overview of biogenesis, composition and role in ovarian cancer. J Ovarian Res. 2014;7(1):14. doi:10.1186/1757-2215-7-14.
  • He C, Zheng S, Luo Y, Wang B. Exosome theranostics: biology and translational medicine. Theranostics. 2018;8(1):237–55. doi:10.7150/thno.21945.
  • 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. doi:10.3389/fimmu.2020.00221.
  • Théry C, Zitvogel L, Amigorena S. Exosomes: composition, biogenesis and function. Nat Rev Immunol. 2002;2(8):569–79. doi:10.1038/nri855.
  • Kantarcioglu M. New therapeutic players on the horizon: edible plant derived exosomes. Hepatol Forum. 2021;2(3):89–90. doi:10.14744/hf.2021.2021.ed0002.
  • Teng Y, Ren Y, Sayed M, Hu X, Lei C, Kumar A, Hutchins E, Mu J, Deng Z, Luo C, et al. Plant-derived exosomal MicroRNAs shape the gut microbiota. Cell Host Microbe. 2018;24(5):637–52.e8. doi:10.1016/j.chom.2018.10.001.
  • Teng Y, Xu F, Zhang X, Mu J, Sayed M, Hu X, Lei C, Sriwastva M, Kumar A, Sundaram K, et al. Plant-derived exosomal microRnas inhibit lung inflammation induced by exosomes SARS-CoV-2 Nsp12. Mol Ther. 2021;29(8):2424–40. doi:10.1016/j.ymthe.2021.05.005.
  • Biadglegne F, König B, Rodloff AC, Dorhoi A, Sack U. Composition and clinical significance of exosomes in tuberculosis: a systematic literature review. J Clin Med. 2021;10(1):145. doi:10.3390/jcm10010145.
  • Malekian F, Shamsian A, Kodam SP, Ullah M. Exosome engineering for efficient and targeted drug delivery: current status and future perspective. J Physiol. 2022;601(22):4853–72. doi:10.1113/JP282799.
  • Gehrmann U, Näslund TI, Hiltbrunner S, Larssen P, Gabrielsson S. Harnessing the exosome-induced immune response for cancer immunotherapy. Semin Cancer Biol. 2014;28:58–67. doi:10.1016/j.semcancer.2014.05.003.
  • Jafari D, Shajari S, Jafari R, Mardi N, Gomari H, Ganji F, Forouzandeh Moghadam M, Samadikuchaksaraei A. Designer Exosomes: a new platform for biotechnology therapeutics. BioDrugs. 2020;34(5):567–86. doi:10.1007/s40259-020-00434-x.
  • Hood JL. Post isolation modification of exosomes for nanomedicine applications. Nanomedicine. 2016;11(13):1745–56. doi:10.2217/nnm-2016-0102.
  • Naseri Z, Oskuee RK, Jaafari MR, Forouzandeh Moghadam M. Exosome-mediated delivery of functionally active miRNA-142-3p inhibitor reduces tumorigenicity of breast cancer in vitro and in vivo. Int J Nanomed. 2018;13:7727–47. doi:10.2147/IJN.S182384.
  • Kooijmans SAA, Aleza CG, Roffler SR, van Solinge WW, Vader P, Schiffelers RM. Display of GPI-anchored anti-EGFR nanobodies on extracellular vesicles promotes tumour cell targeting. J Extracell Vesicles. 2016;5:31053. doi:10.3402/jev.v5.31053.
  • Zhang M, Zang X, Wang M, Li Z, Qiao M, Hu H, Chen D. Exosome-based nanocarriers as bio-inspired and versatile vehicles for drug delivery: recent advances and challenges. J Mater Chem B. 2019;7(15):2421–33. doi:10.1039/c9tb00170k.
  • Choi H, Choi Y, Yim HY, Mirzaaghasi A, Yoo J-K, Choi C. Biodistribution of exosomes and engineering strategies for targeted delivery of therapeutic exosomes. Tissue Eng Regen Med. 2021;18(4):499–511. doi:10.1007/s13770-021-00361-0.
  • Longatti A, Schindler C, Collinson A, Jenkinson L, Matthews C, Fitzpatrick L, Blundy M, Minter R, Vaughan T, Shaw M, et al. High affinity single-chain variable fragments are specific and versatile targeting motifs for extracellular vesicles. Nanoscale. 2018;10(29):14230–44. doi:10.1039/c8nr03970d.
  • Liu Q, Li D, Pan X, Liang Y. Targeted therapy using engineered extracellular vesicles: principles and strategies for membrane modification. J Nanobiotechnol. 2023;21(1). doi:10.1186/s12951-023-02081-0.
  • Choi H, Choi K, Kim D-H, Oh B-K, Yim H, Jo S, Choi C. Strategies for targeted delivery of exosomes to the brain: advantages and challenges. Pharmaceutics. 2022;14(3):672. doi:10.3390/pharmaceutics14030672.
  • Wu JY, Li YJ, Wang J, Hu XB, Huang S, Luo S, Xiang D-X. Multifunctional exosome-mimetics for targeted anti-glioblastoma therapy by manipulating protein corona. J Nanobiotechnol. 2021;19(1). doi:10.1186/s12951-021-01153-3.
  • Sato YT, Umezaki K, Sawada S, Mukai SA, Sasaki Y, Harada N, Shiku H, Akiyoshi K. Engineering hybrid exosomes by membrane fusion with liposomes. Sci Rep. 2016;6(1). doi:10.1038/srep21933.
  • Weng Z, Zhang B, Wu C, Yu F, Han B, Li B, Li L. Therapeutic roles of mesenchymal stem cell-derived extracellular vesicles in cancer. J Hematol Oncol. 2021;14(1). doi:10.1186/s13045-021-01141-y.
  • Li B, Chen X, Qiu W, Zhao R, Duan J, Zhang S, Pan Z, Zhao S, Guo Q, Qi Y, et al. Synchronous disintegration of ferroptosis defense axis via engineered exosome‐conjugated magnetic nanoparticles for glioblastoma therapy. Adv Sci. 2022;9(17). doi:10.1002/advs.202105451.
  • Chen H, Wang L, Zeng X, Schwarz H, Nanda HS, Peng X, Zhou Y. Exosomes, a new star for targeted delivery. Front Cell Dev Biol. 2021;9:751079. doi:10.3389/fcell.2021.751079.
  • Sadeghi S, Tehrani FR, Tahmasebi S, Shafiee A, Hashemi SM. Exosome engineering in cell therapy and drug delivery. Inflammopharmacology. 2023;31(1):145–69. doi:10.1007/s10787-022-01115-7.
  • Parada N, Romero-Trujillo A, Georges N, Alcayaga-Miranda F. Camouflage strategies for therapeutic exosomes evasion from phagocytosis. J Adv Res. 2021;31:61–74. doi:10.1016/j.jare.2021.01.001.
  • Ferrero-Andrés A, Closa D, Roselló-Catafau J, Folch-Puy E. Polyethylene glycol 35 (PEG35) modulates exosomal uptake and function. Polymers. 2020;12(12):3044. doi:10.3390/polym12123044.
  • Witwer KW, Buzás EI, Bemis LT, Bora A, Lässer C, Lötvall J, Nolte‐‘t Hoen EN, Piper MG, Sivaraman S, Skog J, et al. Standardization of sample collection, isolation and analysis methods in extracellular vesicle research. J Extracell Vesicles. 2013;2(1). doi:10.3402/jev.v2i0.20360.
  • Théry C, Amigorena S, Raposo G, Clayton A. Isolation and characterization of exosomes from cell culture supernata nts and biological fluids. Curr Protoc Cell Biol. 2006;30(1). Chapter 3:Unit 3.22. doi:10.1002/0471143030.cb0322s30.
  • Li J, Zhang Y, Dong PY, Yang GM, Gurunathan S. A comprehensive review on the composition, biogenesis, purification, and multifunctional role of exosome as delivery vehicles for cancer therapy. Biomed Pharmacother. 2023;165:115087. doi:10.1016/j.biopha.2023.115087.
  • Lin S, Yu Z, Chen D, Wang Z, Miao J, Li Q, Zhang D, Song J, Cui D. Progress in microfluidics-based exosome separation and detection technologies for diagnostic applications. Small. 2020;16(9):e1903916. doi:10.1002/smll.201903916.
  • Chen J, Li P, Zhang T, Xu Z, Huang X, Wang R, Du L. Review on strategies and technologies for exosome isolation and purification. Front Bioeng Biotechnol. 2022;9:811971. doi:10.3389/fbioe.2021.811971.
  • Jiang Z, Liu G, Li J. Recent progress on the isolation and detection methods of exosomes. Chem Asian J. 2020;15(23):3973–82. doi:10.1002/asia.202000873.
  • Konoshenko MY, Lekchnov EA, Vlassov AV, Laktionov PP. Isolation of extracellular vesicles: general methodologies and latest trends. Biomed Res Int. 2018;2018:8545347. doi:10.1155/2018/8545347.
  • Kamerkar S, LeBleu VS, Sugimoto H, Yang S, Ruivo CF, Melo SA, Lee JJ, Kalluri R. Exosomes facilitate therapeutic targeting of oncogenic KRAS in pancreatic cancer. Nature. 2017;546(7659):498–503. doi:10.1038/nature22341.
  • Haraszti RA, Miller R, Stoppato M, Sere YY, Coles A, Didiot MC, Wollacott R, Sapp E, Dubuke ML, Li X, et al. Exosomes produced from 3D cultures of MSCs by tangential flow filtration show higher yield and improved activity. Mol Ther. 2018;26(12):2838–47. doi:10.1016/j.ymthe.2018.09.015.
  • Sidhom K, Obi PO, Saleem A. A review of exosomal isolation methods: is size exclusion chromatography the best option? Int J Mol Sci. 2020;21(18):6466. doi:10.3390/ijms21186466.
  • 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(8):1051–65. doi:10.1038/s41417-023-00617-y.
  • Batrakova EV, Kim MS. Using exosomes, naturally-equipped nanocarriers, for drug delivery. J Control Release. 2015;219:396–405. doi:10.1016/j.jconrel.2015.07.030.
  • Coumans FAW, Brisson AR, Buzas EI, Dignat-George F, Drees EEE, El-Andaloussi S, Emanueli C, Gasecka A, Hendrix A, Hill AF, et al. Methodological guidelines to study extracellular vesicles. Circ Res. 2017;120(10):1632–48. doi:10.1161/circresaha.117.309417.
  • Gurunathan S, Kang MH, Jeyaraj M, Qasim M, Kim JH. Review of the isolation, characterization, biological function, and multifarious therapeutic approaches of exosomes. Cells. 2019;8(4):307. doi:10.3390/cells8040307.
  • Fitzgerald J, Leonard P, Darcy E, Sharma S, O’Kennedy R. Immunoaffinity chromatography: concepts and applications. Methods Mol Biol. 2017;1485:27–51. doi:10.1007/978-1-4939-6412-3_3.
  • Kimiz-Gebologlu I, Oncel SS. Exosomes: large-scale production, isolation, drug loading efficiency, and biodistribution and uptake. J Control Release. 2022;347:533–43. doi:10.1016/j.jconrel.2022.05.027.
  • Jiang X, Jing W, Zheng L, Liu S, Wu W, Sui G. A continuous-flow high-throughput microfluidic device for airborne bacteria PCR detection. Lab Chip. 2014;14(4):671–6. doi:10.1039/c3lc50977j.
  • Tschuschke M, Kocherova I, Bryja A, Mozdziak P, Angelova Volponi A, Janowicz K, Sibiak R, Piotrowska-Kempisty H, Iżycki D, Bukowska D, et al. Inclusion biogenesis, methods of isolation and clinical application of human cellular exosomes. J Clin Med. 2020;9(2):436. doi:10.3390/jcm9020436.
  • Lai JJ, Chau ZL, Chen SY, Hill JJ, Korpany KV, Liang NW, Lin L-H, Lin Y-H, Liu JK, Liu Y-C, et al. Exosome processing and characterization approaches for research and technology development. Adv Sci. 2022;9(15). doi:10.1002/advs.202103222.
  • Yu D, Li Y, Wang M, Gu J, Xu W, Cai H, Fang X, Zhang X. Exosomes as a new frontier of cancer liquid biopsy. Mol Cancer. 2022;21(1):56. doi:10.1186/s12943-022-01509-9.
  • Butreddy A, Kommineni N, Dudhipala N. Exosomes as naturally occurring vehicles for delivery of biopharmaceuticals: insights from drug delivery to clinical perspectives. Nanomaterials. 2021;11(6):1481. doi:10.3390/nano11061481.
  • Yang B, Chen Y, Shi J. Exosome biochemistry and advanced nanotechnology for next‐generation theranostic platforms. Adv Mater. 2019;31(2). doi:10.1002/adma.201802896.
  • Chaput N, Schartz NEC, Andre F, Zitvogel L. Exosomes for immunotherapy of cancer. Adv Exp Med Biol. 2003;532:215–21. doi:10.1007/978-1-4615-0081-0_17.
  • Cheng Q, Shi X, Han M, Smbatyan G, Lenz HJ, Zhang Y. Reprogramming exosomes as nanoscale controllers of cellular immunity. J Am Chem Soc. 2018;140(48):16413–17. doi:10.1021/jacs.8b10047.
  • Shi X, Cheng Q, Hou T, Han M, Smbatyan G, Lang JE, Epstein AL, Lenz H-J, Zhang Y. Genetically engineered cell-derived nanoparticles for targeted breast cancer immunotherapy. Mol Ther. 2020;28(2):536–47. doi:10.1016/j.ymthe.2019.11.020.
  • Qazi KR, Gehrmann U, Domange Jordö E, Karlsson MC, Gabrielsson S. Antigen-loaded exosomes alone induce Th1-type memory through a B-cell-dependent mechanism. Blood. 2009;113(12):2673–83. doi:10.1182/blood-2008-04-153536.
  • Delcayre A, Estelles A, Sperinde J, Roulon T, Paz P, Aguilar B, Villanueva J, Khine S, Le Pecq JB. Exosome display technology: applications to the development of new diagnostics and therapeutics. Blood Cells Mol Dis. 2005;35(2):158–68. doi:10.1016/j.bcmd.2005.07.003.
  • Greening DW, Gopal SK, Xu R, Simpson RJ, Chen W. Exosomes and their roles in immune regulation and cancer. Semin Cell Dev Biol. 2015;40:72–81. doi:10.1016/j.semcdb.2015.02.009.
  • 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(1):1779991. doi:10.1080/2162402x.2020.1779991.
  • Burke M, Choksawangkarn W, Edwards N, Ostrand-Rosenberg S, Fenselau C. Exosomes from myeloid-derived suppressor cells carry biologically active proteins. J Proteome Res. 2014;13(2):836–43. doi:10.1021/pr400879c.
  • McNulty S, Colaco CA, Blandford LE, Bailey CR, Baschieri S, Todryk S. Heat-shock proteins as dendritic cell-targeting vaccines–getting warmer. Immunology. 2013;139(4):407–15. doi:10.1111/imm.12104.
  • Nie X, Shi C, Chen X, Yu C, Jiang Z, Xu G, Lin Y, Tang M, Luan Y. A single-shot prophylactic tumor vaccine enabled by an injectable biomembrane hydrogel. Acta Biomater. 2023;169:306–16. doi:10.1016/j.actbio.2023.08.010.
  • Mears R, Craven RA, Hanrahan S, Totty N, Upton C, Young SL, Patel P, Selby PJ, Banks RE. Proteomic analysis of melanoma-derived exosomes by two-dimensional polyacrylamide gel electrophoresis and mass spectrometry. Proteomics. 2004;4(12):4019–31. doi:10.1002/pmic.200400876.
  • Chaput N, Théry C. Exosomes: immune properties and potential clinical implementations. Semin Immunopathol. 2011;33(5):419–40. doi:10.1007/s00281-010-0233-9.
  • Zitvogel L, Regnault A, Lozier A, Wolfers J, Flament C, Tenza D, Ricciardi-Castagnoli P, Raposo G, Amigorena S. Eradication of established murine tumors using a novel cell-free vaccine: dendritic cell-derived exosomes. Nat Med. 1998;4(5):594–600. doi:10.1038/nm0598-594.
  • Pitt JM, André F, Amigorena S, Soria JC, Eggermont A, Kroemer G, Zitvogel L. Dendritic cell–derived exosomes for cancer therapy. J Clin Invest. 2016;126(4):1224–32. doi:10.1172/jci81137.
  • Kowal J, Tkach M. Dendritic cell extracellular vesicles. Int Rev Cell Mol Biol. 2019;349:213–49. doi:10.1016/bs.ircmb.2019.08.005.
  • Zhang B, Yin Y, Lai RC, Lim SK. Immunotherapeutic potential of extracellular vesicles. Front Immunol. 2014;5:518. doi:10.3389/fimmu.2014.00518.
  • Lindenbergh MFS, Stoorvogel W. Antigen presentation by extracellular vesicles from professional antigen-presenting cells. Annu Rev Immunol. 2018;36(1):435–59. doi:10.1146/annurev-immunol-041015-055700.
  • Hsu DH, Paz P, Villaflor G, Rivas A, Mehta-Damani A, Angevin E, Zitvogel L, Le Pecq J-B. Exosomes as a tumor vaccine: enhancing potency through direct loading of antigenic peptides. J Immunother. 2003;26(5):440–50. doi:10.1097/00002371-200309000-00007.
  • Viaud S, Théry C, Ploix S, Tursz T, Lapierre V, Lantz O, Zitvogel L, Chaput N. Dendritic cell-derived exosomes for cancer immunotherapy: what’s next? Cancer Research. 2010;70(4):1281–5. doi:10.1158/0008-5472.Can-09-3276.
  • Chaput N, Taïeb J, Schartz NEC, André F, Angevin E, Zitvogel L. Exosome-based immunotherapy. Cancer Immunol Immunother. 2004;53(3):234–9. doi:10.1007/s00262-003-0472-x.
  • Syn NL, Wang L, Chow EKH, Lim CT, Goh BC. Exosomes in cancer nanomedicine and immunotherapy: prospects and challenges. Trends Biotechnol. 2017;35(7):665–76. doi:10.1016/j.tibtech.2017.03.004.
  • Viaud S, Terme M, Flament C, Taieb J, André F, Novault S, Escudier B, Robert C, Caillat-Zucman S, Tursz T, et al. Dendritic cell-derived exosomes promote natural killer cell activation and proliferation: a role for NKG2D ligands and IL-15Rα. PLOS ONE. 2009;4(3):e4942. doi:10.1371/journal.pone.0004942.
  • Lu Z, Zuo B, Jing R, Gao X, Rao Q, Liu Z, Qi H, Guo H, Yin H. Dendritic cell-derived exosomes elicit tumor regression in autochthonous hepatocellular carcinoma mouse models. J Hepatol. 2017;67(4):739–48. doi:10.1016/j.jhep.2017.05.019.
  • Veerman RE, Güçlüler Akpinar G, Eldh M, Gabrielsson S. Immune cell-derived extracellular vesicles – functions and therapeutic applications. Trends Mol Med. 2019;25(5):382–94. doi:10.1016/j.molmed.2019.02.003.
  • 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(7):1074–83. doi:10.4161/onci.20897.
  • Robbins PD, Morelli AE. Regulation of immune responses by extracellular vesicles. Nat Rev Immunol. 2014;14(3):195–208. doi:10.1038/nri3622.
  • Wieckowski E, Whiteside TL. Human tumor-derived vs dendritic cell-derived exosomes have distinct biologic roles and molecular profiles. Immunol Res. 2006;36(1–3):247–54. doi:10.1385/ir:36:1:247.
  • Inamdar S, Nitiyanandan R, Rege K. Emerging applications of exosomes in cancer therapeutics and diagnostics. Bioeng Transl Med. 2017;2(1):70–80. doi:10.1002/btm2.10059.
  • Gutiérrez-Vázquez C, Villarroya-Beltri C, Mittelbrunn M, Sánchez-Madrid F. Transfer of extracellular vesicles during immune cell-cell interactions. Immunol Rev. 2013;251(1):125–42. doi:10.1111/imr.12013.
  • Wen C, Seeger RC, Fabbri M, Wang L, Wayne AS, Jong AY. Biological roles and potential applications of immune cell-derived extracellular vesicles. J Extracell Vesicles. 2017;6(1):1400370. doi:10.1080/20013078.2017.1400370.
  • Peters PJ, Geuze HJ, Van der Donk HA, Slot JW, Griffith JM, Stam NJ, Clevers HC, Borst J. Molecules relevant for T cell-target cell interaction are present in cytolytic granules of human T lymphocytes. Eur J Immunol. 1989;19(8):1469–75. doi:10.1002/eji.1830190819.
  • Zhu L, Kalimuthu S, Gangadaran P, Oh JM, Lee HW, Baek SH, Jeong SY, Lee S-W, Lee J, Ahn B-C, et al. Exosomes derived from natural killer cells exert therapeutic effect in melanoma. Theranostics. 2017;7(10):2732–45. doi:10.7150/thno.18752.
  • 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(7):1665–75. doi:10.1016/j.ymthe.2017.02.007.
  • Wang P, Wang H, Huang Q, Peng C, Yao L, Chen H, Qiu Z, Wu Y, Wang L, Chen W, et al. Exosomes from M1-polarized macrophages enhance paclitaxel antitumor activity by activating macrophages-mediated inflammation. Theranostics. 2019;9(6):1714–27. doi:10.7150/thno.30716.
  • Behzadi E, Hosseini HM, Halabian R, Fooladi AAI. Macrophage cell-derived exosomes/staphylococcal enterotoxin B against fibrosarcoma tumor. Microb Pathog. 2017;111:132–8. doi:10.1016/j.micpath.2017.08.027.
  • Sabanovic B, Piva F, Cecati M, Giulietti M. Promising extracellular vesicle-based vaccines against viruses, including SARS-CoV-2. Biology. 2021;10(2):94. doi:10.3390/biology10020094.
  • Yoo KH, Thapa N, Kim BJ, Lee JO, Jang YN, Chwae YJ, Kim J. Possibility of exosome‑based coronavirus disease 2019 vaccine (review). Mol Med Rep. 2022;25(1):26. doi:10.3892/mmr.2021.12542.
  • Zani-Ruttenstock E, Antounians L, Khalaj K, Figueira RL, Zani A. The role of exosomes in the treatment, prevention, diagnosis, and pathogenesis of COVID-19. Eur J Pediatr Surg. 2021;31(4):326–34. doi:10.1055/s-0041-1731294.
  • Tsai SJ, Atai NA, Cacciottolo M, Nice J, Salehi A, Guo C, Sedgwick A, Kanagavelu S, Gould SJ. Exosome-mediated mRNA delivery in vivo is safe and can be used to induce SARS-CoV-2 immunity. J Biol Chem. 2021;297(5):101266. doi:10.1016/j.jbc.2021.101266.
  • Kuate S, Cinatl J, Doerr HW, Uberla K. Exosomal vaccines containing the S protein of the SARS coronavirus induce high levels of neutralizing antibodies. Virology. 2007;362(1):26–37. doi:10.1016/j.virol.2006.12.011.
  • Mondal J, Pillarisetti S, Junnuthula V, Saha M, Hwang SR, Park IK, Lee Y-K. Hybrid exosomes, exosome-like nanovesicles and engineered exosomes for therapeutic applications. J Control Release. 2023;353:1127–49. doi:10.1016/j.jconrel.2022.12.027.