Next Article in Journal
pH-Responsive Hybrid Nanoassemblies for Cancer Treatment: Formulation Development, Optimization, and In Vitro Therapeutic Performance
Next Article in Special Issue
The Effect of Super-Repressor IkB-Loaded Exosomes (Exo-srIκBs) in Chronic Post-Ischemia Pain (CPIP) Models
Previous Article in Journal
Optimisation of AAV-NDI1 Significantly Enhances Its Therapeutic Value for Correcting Retinal Mitochondrial Dysfunction
Previous Article in Special Issue
Quantitative Biodistribution and Pharmacokinetics Study of GMP-Grade Exosomes Labeled with 89Zr Radioisotope in Mice and Rats
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Small Extracellular Vesicles as a New Class of Medicines

1
KU-KIST Graduate School of Converging Science and Technology, Korea University, 145, Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
2
Department of Research and Development, SHIFTBIO Inc., Seoul 02751, Republic of Korea
3
Chemical & Biological Integrative Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
4
Department of Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul 02841, Republic of Korea
5
Department of Otolaryngology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
*
Authors to whom correspondence should be addressed.
Pharmaceutics 2023, 15(2), 325; https://doi.org/10.3390/pharmaceutics15020325
Submission received: 10 December 2022 / Revised: 13 January 2023 / Accepted: 17 January 2023 / Published: 18 January 2023
(This article belongs to the Special Issue Exosome-Based Drug Delivery: Translation from Bench to Clinic)

Abstract

:
Extracellular vesicles (EVs) are nanovesicles that are naturally released from cells in a lipid bilayer-bound form. A subset population with a size of 200 nm, small EVs (sEVs), is enticing in many ways. Initially perceived as mere waste receptacles, sEVs have revealed other biological functions, such as cell-to-cell signal transduction and communication. Besides their notable biological functions, sEVs have profound advantages as future drug modalities: (i) excellent biocompatibility, (ii) high stability, and (iii) the potential to carry undruggable macromolecules as cargo. Indeed, many biopharmaceutical companies are utilizing sEVs, not only as diagnostic biomarkers but as therapeutic drugs. However, as all inchoate fields are challenging, there are limitations and hindrances in the clinical translation of sEV therapeutics. In this review, we summarize different types of sEV therapeutics, future improvements, and current strategies in large-scale production.

1. Introduction

sEVs are known to exert various functions, from aiding in the excretion of waste within cells to participating in cell-to-cell signal transduction [1]. Surprisingly, but importantly, sEVs have recently been found to play roles related to human diseases [2]. They disseminate diseases by transferring pathological cargo from abnormally altered cells to other cells. In this manner, sEVs derived from cancer cells can determine the aggressiveness of cancer or be associated with metastasis [2]. Furthermore, they have been found to be closely related to the pathophysiology of neurodegenerative and cardiovascular diseases [3,4]. Therefore, many studies are being conducted to exploit sEVs as diagnostic biomarkers and therapeutic targets by analyzing the proteins and nucleic acids in sEVs related to diseases [5].
Extracellular vesicles (EVs) are particles surrounded by lipid bilayers, naturally released from cells [2]. Although they are demarcated by lipid bilayers like cells, EVs cannot replicate and do not have a functional nucleus. Classifying EVs to certain biogenesis routes, such as the endosomal system (exosomes) or plasma membrane (ectosomes, microvesicles, microparticles), is challenging due to the lack of specific markers regarding each cell compartment [1]. Instead, EV subtypes are classified according to their physical properties (size or density), biochemical compositions (specific biomarkers), or recognizable conditions (cellular origin) [2]. For example, EVs with a size of up to 200 nm in diameter are classified as small EVs, and EVs larger than 200 nm are classified as medium/large EVs (m/lEVs). The Minimal Information for Studies of Extracellular Vesicles (MISEV or MISEV2018) encourages the use of small EVs (sEVs) instead of exosomes or microvesicles, with their size determined by appropriate methods [1]. Furthermore, EVs can be classified as low, middle, or high EVs according to their defined density range, or they can be distinguished through specific biomarkers, such as CD81+/CD9+ EVs. Lastly, nomenclatures based on cellular origins are often used, such as apoptotic bodies.
sEVs are currently receiving a great amount of attention as a promising therapeutic tool for diseases with high unmet medical needs due to their (i) excellent biocompatibility begetting low immunogenicity, (ii) high stability for the in vivo transport of substances, and (iii) the potential for loading a myriad of macromolecules as cargo [2]. Furthermore, sEVs perform the unique process of cellular signaling and uptake—they provide the optimal microenvironment for surface ligands to signal and display the intracellular delivery of therapeutic cargos to an extent [6]. Therefore, many scientists and drug developers are interested in the use of sEVs as a delivery tool for proteins and genes. In addition, sEVs show an advantage in cell-free therapies, as they can overcome the current safety concerns and challenges regarding the injection of viable cells and scalable manufacturing [2,6].
Although sEVs have shown versatility and high potential as a disease treatment at the pre-clinical level, limitations of the practical application in clinical practice remain [2]. Therefore, in this review, we introduce the methods and studies of sEVs being used as therapeutic agents for human diseases and discuss problems and notable points to be overcome in future clinical applications regarding sEV-based drugs.

2. Types of sEV Therapeutics

The current sEV therapeutics include utilizing naïve/engineered sEVs and suppressing the secretion/uptake of sEVs, as shown in Figure 1. Many researchers and biotech companies have been aiming to develop a new class of medicines harnessing sEVs harboring macromolecules [2,6]. They have used sEVs for cell-free therapy and as efficient delivery tools for therapeutic cargo, targeting high unmet medical needs. Furthermore, since various diseases have been found to progress through the communication of sEVs produced from transformed cells, some studies have suggested that the inhibition of the production and uptake of pathological sEVs could be a promising therapeutic strategy [2]. In this section, we address several sEV therapeutic strategies.

2.1. Inhibition of the Release and Uptake of sEVs

Emerging evidence points to sEVs having roles in human diseases. sEVs disseminate diseases by transferring pathological cargo from diseased donor cells to normal cells. For instance, cancer cell-derived sEVs are associated with tumor progression and metastasis [7,8,9] Therefore, it is important to understand the biogenesis of sEVs and strategies to inhibit the release and uptake of sEVs.
Intracellular sEV production is predominantly based on two pathways: endosomal sorting complexes required for transport machinery (ESCRT)-dependent and -independent pathways. In the former case, multi-vesicular bodies (MVBs) are formed by ESCRT, and intraluminal vesicles (ILVs) contained therein are released in the form of EVs outside the cell. In the ESCRT-independent pathway, MVBs-ILVs are formed by neutral sphingomyelinase 2 (nSmase2) through sphingomyelinase hydrolysis and ceramide formation [10]. Accordingly, drugs that inhibit sEV release mainly target ESCRT and nSmase2. ESCRT is recognized to be closely related to the rat sarcoma virus (Ras) family protein. Drugs such as manumycin A and tipifarnib are often used as therapeutic agents to reduce sEV secretion by inhibiting ESCRT due to their specific inhibition of farnesyltransferase, one of the major enzymes in the lipid metabolism pathway of Ras. Datta et al. reported that inhibiting Ras/Raf/MEK/ERK1/2 signaling using Manumacin A can suppress the biogenesis of sEVs in castration-resistant prostate cancer cells, and Greenberg et al. highlighted the reduction in sEV secretion in sunitinib-sensitive renal cell carcinoma when treated with Tipifarnib [11,12]. The nSMase2 is a ubiquitous enzyme that generates a bioactive lipid ceramide through the hydrolysis of the membrane lipid sphingomyelin. A potent and specific nSMase2 inhibitor, such as GW4869, can prevent the formation of ILVs and consequently diminish sEV production [13]. Similarly, Imipramine also affects the lipid metabolism of the secreting cells, thereby reducing micropinocytosis activity and decreasing the secretion of sEVs. Hekmatirad et al. reported that the prevention of sEV release by GW4869, the nSMase2 inhibitor, can enhance doxorubicin sensitivity U937 cells by the inhibition of expelling doxorubicin via sEVs [14]. Other drugs known to inhibit sEV release are summarized in Table 1.
Another strategy to inhibit the propagation of sEVs is to inhibit uptake in recipient cells. The primary mechanism of sEV uptake is associated with the endocytosis pathway, which is divided into clathrin-dependent and -independent mechanisms [15]. Furthermore, other sEV uptake processes are mediated through membrane fusion, phagocytosis, and micropinocytosis [15]. Dynasore, a dynamin inhibitor, was found to inhibit sEV uptake by interfering with the transferrin internalization through the clathrin-dependent pathway, and it has been reported to inhibit the angiogenesis associated with the propagation of sEVs derived from malignant melanoma cells [16]. In addition, Nanbo et al. reported that dynasore could hamper virus spread by inhibiting the uptake of exosomes derived from Epstein–Barr virus-infected B cells into uninfected B cells [17]. Heparin was reported to hamper the sEV delivery between doner and recipient cells via competitive binding to the receptors of recipient cells [18]. Therefore, the uptake of sEVs that involve heparin sulfate proteoglycan (HSPG) coreceptors can be prevented by pretreatment with heparin on the recipient cells [19,20]. Unfortunately, a therapeutic strategy to inhibit sEV uptake is not readily applicable due to the convoluted mechanisms and the unfeasibility of the selective inhibition and visual verification of sEV uptake in specific cells [21]. For instance, a method for selectively inhibiting pathological sEVs by distinguishing them from healthy sEVs essential to maintaining normal physiological functions has not been demonstrated to date.
Table 1. Therapeutic approaches via inhibition of the release and uptake of sEVs.
Table 1. Therapeutic approaches via inhibition of the release and uptake of sEVs.
StrategyReagentDiseaseMode of ActionTreatmentResultReference
Exosome
biogenesis and release
inhibition
Manumycin AC42B prostate cancerERK-dependent inhibition of hnRNP H1250 nM treatment for 48 hDecreased Ras activation by GTPγS[11]
GW4869U937 acute myeloblastic leukemiaEnhance doxorubicin sensitivity on resistant cells by inhibition doxorubicin expulsionvia sEVs20 µM GW4869 + 0.5–2 uM PEGylated liposomal doxorubicin for 24 hEnhanced cytotoxicity[14]
TipifarnibMetastatic renal cell carcinomaDisrupt ESCRT-dependent and ESCRT-independent functional proteins (Alix, nSMase, and Rab27a)0.25–1 µM treatment for 48 h at 37 °CReduced sEV load in sunitinib-sensitive renal cell carcinomas[12]
5-(N-ethyl-N-isopropyl)
amiloride
A431 human epidermoid carcinomaDisrupt Rac1 activation and assembly of actin50–100 µM pretreatment for 30 minInhibited clathrin-independent endocytosis and macropinocytosis of sEVs[22]
ImipraminePC3 prostate cancerReduce macropinocytosis25 µM treatmentReduced total EV release by 77% in PC3[23]
4T1 mammary carcinoma5 μM treatment for 1 hInhibited membrane ruffle formation[24]
CalpeptinWormsInhibit calpain80 μM treatmentPrevented the secretion of miRNAs from adult worms[25]
Exosome
endocytosis
inhibition
DynasoreHUVECInterfere with the internalization of transferrin through the clathrin-dependent pathwayPretreatment on HUVECs with 20 µM dynasore for 30 min at 37 °CPrevented pancreatic cancer cell-derived sEVs[16]
B cellsPretreatment on uninfected B cells with 150 μM dynasor for 30 min at 37 °CPrevented virus spread by inhibiting the uptake of Epstein–Barr virus-infected B cell-derived exosomes[17]
Human bone marrow stromal cellsPretreatment on bone marrow stromal cells with 50 μM dynasore for 30 minSuppressed the effects of multiple myeloma cell-derived exosome uptake[26]
HeparinU-87 MG glioblastomaCompetitively inhibit cancer cell surface receptors depending on heparin sulfate proteoglycan coreceptors for the uptake of sEVsPretreatment on U-87 MG with 10 μg/mL heparin for 1 hReduced sEV uptake in U87 cells by 55%[19]
SW780 human bladder cancer cell linePretreatment on SW780 with 10 μg/mL heparin at 4 °C for 4 hInhibited receptors on recipient cells[20]

2.2. Naïve sEV Therapeutics

Naïve sEVs, or native sEVs, reflect diverse characteristics, such as membrane proteins or contents, of their parental cells. By leveraging this property, various studies reported the potential of naïve sEVs’ therapeutic efficacy. Depending on the origin of cells, sEVs can be utilized in appropriate diseases. In this section, we explore various therapeutic effects of naïve sEVs derived from stem cells, immune cells, and other cells, such as red blood cells or platelets.

2.2.1. Stem Cell-Derived Naïve sEVs

Stem cells are undifferentiated but can be multilineage differentiated cells with self-renewal capability. Accordingly, stem cells have been frequently and widely used in clinics, especially in regenerative medicine, regarding their pleiotropic differentiating potential and immunomodulatory properties [27]. Generally, stem cells are divided into embryonic stem cells, adult stem cells that originate from diverse mesenchymal/stromal tissues and bone marrow, and induced pluripotent stem cells. A large body of work has demonstrated that mesenchymal stem cells (MSCs) can modulate inflammation and immune responses [27]. MSCs can be harvested from diverse tissues, including bone marrow, the umbilical cord, adipose tissue, and brain tissue. Notably, MSCs activated by inflammatory cytokines have tropism to ischemia, injury, or inflammation sites [28]. Furthermore, MSCs can suppress pro-inflammatory processes by releasing an array of factors [29]. These factors consist of interleukin-10 and growth factors (GFs), such as trans-forming GF-β (TGF-β), hepatocyte GF (HGF), stromal-cell-derived factor-1 (SDF-1), epidermal GF (EGF), keratinocyte GF (KGF)-1, fibroblast GF (FGF), vascular endothelial GF (VEGF), plate-derived GF (PDGF), and insulin GF (IGF)-1.
Nonetheless, stem cell therapies show several notable qualities regarding large-scale production, quality control, and off-the-shelf medicines [30]. Moreover, MSC-based therapeutics have been reported to elicit tumorigenicity as a severe side effect [30]. Interestingly, recent studies discovered that stem cells’ abilities arise from the secretion of paracrine factors, and sEVs comprise the primary mediators [31,32]. Therefore, MSC-derived sEVs offer the possibility of an alternative approach to stem cell therapeutics for treating various diseases, including cardiovascular, neurodegenerative, and immunologic diseases, as shown in Table 2. Zhao et al. reported that micro-RNA-182-containing MSC-derived sEVs could attenuate myocardial ischemia–reperfusion injury via changing M1-like polarized macrophages into M2 phenotypes [33]. Alzheimer’s disease (AD) is among the most problematic and frequently investigated human diseases in terms of neurogenerative disorders, and therefore, a number of attempts have been made to mitigate the progression of Altzheimer’s disease using MSC-derived sEVs. A recent study demonstrated that intranasally inhaled human MSC-derived sEVs could slow down AD-related pathogenesis [34]. Furthermore, MSC-derived sEVs could mitigate autoimmune-related demyelinating processes and influence neuroprotective mechanisms via systemically modulating regulatory T cells and peripheral blood mononuclear cells [35]. The normalization of kidney function can be achieved by the administration of stem cell-derived sEVs on chronic kidney disease or diabetic nephropathy [36,37]. These findings suggest the potential of MSC-derived sEVs as an alternative approach to stem cell therapies in diverse diseases.

2.2.2. Immune Cell-Derived Naïve sEVs

According to previous reports, tumor cell-derived EVs (TEVs) have shown conflicting characteristics in terms of both promoting the aggressiveness of tumors and initiating anticancer immunity cycles. Zitvogel et al. reported the promising vaccine effects of TEVs as sources of tumor antigens for the first time [52]. This study found that TEVs induced better anti-tumor immune responses than tumor cell lysates. Moreover, TEVs can directly activate antigen-presenting cells (APCs) to release pro-inflammatory cytokines [53]. Notably, TEVs can elicit prophylactic cancer vaccine effects but not therapeutic ones [54]. Contrary to the anti-tumor immunity of TEVs, TEVs have been found to provoke heterogenous pro-tumorigenic effects dependent on the tumor type and stage, such as initiating cell transformation, modulating the tumor and metastatic microenvironment, and fostering tumor progression [55,56,57,58]. These results collectively indicate the ambivalence of TEVs in cancer vaccines, which represents a hurdle to their use as transformative medicines. Therefore, researchers have developed alternative methods, such as immune cell-derived sEVs, for the anti-tumor therapeutic approach.
sEVs derived from tumor antigen-exposed dendritic cells (DEVs, sometimes referred to as ‘dexosomes’) have been considered [59] to overcome the limitations of TEVs for cancer vaccines. This approach takes advantage of the molecular properties of DEVs, which contain the adhesion/docking molecules and immunostimulatory factors presented on DCs. Specifically, peptide–MHC complexes can be formed spontaneously and loaded at the external surface of DEVs, thereby provoking additional immunostimulatory effects [60]. Interestingly, DEVs produced more effective anticancer immunity than microvesicles from DCs [61]. Since DEVs show the efficient induction of tumor-specific immunity, their potential to be utilized as an anticancer vaccine has been investigated extensively, concurrent with multiple ongoing clinical trials, as recently reviewed [62]. The completed clinical trials on DEV-based therapy have reported mild to moderate side effects, with relatively milder responses such as low levels of T cell responses, with NK cell stimulation, showing tolerability among cancer patients [62]. According to the clinical trial on metastatic melanoma patients treated with DEVs containing MHC class II peptides as a vaccination, the majority of patients showed a minimal response, and only one patient showed the recruitment and activation of T cell response in the tumor area. Similarly, a modest response was reported on patients with non-small-cell lung cancer, with two patients showing enhanced activity in NK cells. Overall, the results from completed clinical trials imply that DEVs bear the potential to be utilized as cancer vaccines. Studies need to be further implemented or engineered to maximize and guarantee immune response enhancement.
Other than DCs, some studies have evaluated the anti-tumor effects of sEVs from immune cells. For instance, NK cell line-derived sEVs (NK-EVs) have been reported to eradicate specific cancer cells through cytotoxic molecules such as tumor necrosis factor-α, perforin, granzyme, and the Fas ligand [63,64,65]. Although the mechanism of M1-type macrophage-derived sEVs (M1-EVs) in increasing anticancer immunity is still unclear, several reports demonstrated that M1-EVs induced anti-tumor immune responses through activating APCs, including macrophages and dendritic cells [66,67]. Furthermore, sEVs derived from effector chimeric antigen receptor (CAR) T cells (CAR-EVs) have been shown to mainly carry CAR and sufficient cytotoxic molecules, without expressing programmed cell death protein-1 (PD-1) on their surfaces [68]. Notably, CAR-EVs substantially inhibited tumor growth without cytokine release syndrome, which is the primary side effect of CAR-T cell therapy, and the tumor-inhibiting efficacy did not decrease with the treatment of PD-1:PD-L1 blockade [69]. Collectively, these studies demonstrate the wide versatility of immune cell-derived sEVs as a promising therapeutic strategy for cancer treatment (Table 3).

2.2.3. Other Cell-Derived Naïve sEVs

Platelets, or thrombocytes, are anuclear cells produced in the bone marrow. They were once regarded as mere fragments of megakaryocytes. However, accumulated research has pointed out the important biological roles of platelets, including angiogenesis, hemostasis, and thrombosis [74]. Platelet-derived EVs (pEVs or PLT-EVs) have been found to take the lead in these biological roles through intercellular communication. Most EVs isolated from human serum are pEVs, and they participate in both regenerative responses, such as wound healing or tissue regeneration, and pathological processes, including inflammation and tumor progression [75]. During the normal wound-healing process, it is unequivocal that keratinocytes and fibroblasts orchestrate most of the repair process [76]. Previous reports have demonstrated, in vitro, that growth factor cargos of pEVs influence these repair cells to migrate and accumulate in wound sites. These growth factors include platelet-derived growth factor (PDGF), basic fibroblast growth factors (FGF2), transforming growth factor-β (TGF-β), and vascular endothelial growth factor (VEGF) [76]. Guo et al. observed wound-healing effects of pEVs, enhancing cell proliferation and migration through an angiogenesis-promoting effect by stimulating HMEC-1 growth factor in diabetic rat models [76]. Furthermore, previous studies have shown another interesting property of pEVs having neuroregenerative effects [77]. Protein cargos such as PDGF and VEGF in pEVs influenced the neurogenesis of neural stem cells. Hayon et al. used permanent middle cerebral artery occlusion (MCAO) rat models to show a dosage-dependent increase in neuroregenerative effects of pEVs [77]. On the other hand, many researchers have studied pEVs that aggravate cancer progression or inflammation. Ironically, the critical crosstalk between MAPK and YAP during the wound-repair process arranged through pEVs is hijacked in tumor cells. Labelle et al. showed exacerbated metastasis through platelet-derived TGFB that activates the NF-kB pathway [78].
Red blood cell (RBC)-derived EVs (RBCEVs) have gained attention due to their safety and biocompatibility in clinical applications. For instance, RBCEVs have a lower risk of horizontal gene transfer, because they lack nuclear DNA and mitochondria. RBCEVs participate in important biological processes, such as nitric oxide homeostasis, redox balance, immunomodulation, and coagulation [79]. Hitherto, there is no gold standard on which cell type-derived EVs should be used for drug delivery systems due to their unique property of reflecting proteins of their donor cells. For instance, EVs derived from tumor cells pose tumorigenicity, and EVs from nucleated cells have the risk of horizontal gene transfer. However, cumulative research points to the potential and strong efficacy of RBCEVs as a drug delivery system. Zhang et al. showed the feasibility of RBCEVs as a drug delivery system in acute liver failure (ALF) mouse models by loading antisense oligonucleotides of miR-155 (miR155-ASO) [80]. Although the downregulation of miR-155 through miR155-ASO has been reported to alleviate liver injury, efficient drug delivery was a major challenge. RBCEVs loaded with miR155-ASO were specifically delivered to the liver and demonstrated potent therapeutic effects [80]. Despite these noticeable advantages, RBCEVs show some challenges, such as side effects or scalability, to be overcome for further clinical applications [81].

2.3. Engineered sEV Therapeutics

sEVs hold tremendous advantages in drug modalities, and many studies have leveraged engineered sEVs to deliver potent macromolecules, including proteins and genes, as shown in Table 4. Due to phospholipid bilayer membranes, EVs provide the optimal microenvironment to therapeutic proteins and allow proteins to perform their original functions as if on cell membranes [82]. Furthermore, therapeutic proteins have been shown to cluster and be enriched in the lipid rafts of sEV membranes, leading to high avidity of the targeted ligand [83]. Therefore, the expression levels of therapeutic proteins are positively correlated with their therapeutic efficacies. Many researchers and biomedical companies have created diverse engineering strategies to effectively display proteins of interest on the surfaces of sEVs to maximize the therapeutic effects [84,85]. For example, Evox Therapeutics found that the genetic engineering of sEV-producing cells to use an N-terminal fragment of syntenin (a cytoplasmic adaptor of syndecan) enhanced the expression efficiency and therapeutic activity of proteins [86]. They created sEVs expressing decoy receptors of inflammatory cytokines to ameliorate inflammatory diseases, such as neuroinflammation, intestinal inflammation, and systemic inflammation. Codiak Biosciences suggested one scaffold protein, PTGFRN (prostaglandin F2 receptor negative regulator), preferentially sorted into sEVs to facilitate the high-level surface expression of proteins of interest [87]. They presented the preclinical and clinical data of sEVs expressing IL-12 via PTGFRN for cancer treatment, verifying the development of a promising technology platform. Recently, Kai Hu et al. observed that the presentation of antigens on sEVs utilizing the transmembrane domain of viral glycoproteins efficiently increased antigen-specific humoral and cellular immunity [88]. Although there are still limitations in expressing a controlled number of proteins on heterogenous EV populations, these efforts represent a substantial advance toward realizing the full therapeutic potential of sEVs.
The intracellular delivery of therapeutic cargo, including genes and proteins, is often unable to surpass the cell membrane. The usage of lipid nanoparticles (LNPs) received attention after the COVID-19 pandemic due to LNPs containing mRNA passing the lipid bilayer of the cell membrane [89]. However, LNPs are artificial nanoparticles that can provoke unexpected immune responses in the body and engender significant long-term toxicity. Moreover, LNPs cannot evade the endolysosomal pathway, which can quickly degrade therapeutic cargo [90]. Surprisingly, a previous study demonstrated that 30% of sEVs evaded the endolysosomal pathway to transfer their cargo into the cell cytosol through a mechanism involving fusion between exosomal and endosomal membranes [91,92]. This unique property is one profound advantage of using sEVs for the intracellular transfer of macromolecules, making undruggable targets druggable. However, the major obstacle to sEV-based intracellular delivery is that efficient methods of loading cargo into sEVs are still under development. Some researchers and biopharmaceutical companies have suggested exogenous loading strategies, including sonication, extrusion, surfactant treatment, dialysis, freeze–thaw treatment, or electroporation, but these methods show very low efficiency. Consequently, endogenous loading strategies using the genetic engineering of EV-producing cells have emerged as alternatives to these methods [93]. For example, Tian et al. reported that incubating CD4+ T cell-derived sEVs with anti-VEGF antibodies can suppress angiogenesis and inflammation on choroidal neovascularization [94]. The application of adipose- and stem cell-derived EVs primed by IFNγ helped to repair tendon injury [95]. EVs with transfection of IL-10 overexpressing vector plasmids can modulate T cell immunity in autoimmune uveitis [96]. The chemical engineering of EVs to express a collagen-binding peptide could reduce inflammation and induce muscle regeneration in ischemic disease [97]. The incubation of EVs with anti-angiogenic peptide KV11-anchoring peptide CP05 with EVs could suppress neovascularization in the retina [98]. The loading of therapeutic reagents in sEVs by transfection, incubation, and sonication is widely being tried to utilize EVs as carriers of therapeutic cargo. The loading of paclitaxel into EVs using sonication or electroporation could suppress the neoplastic effect in pulmonary metastasis [99], as well as in Lewis lung carcinoma in an in vivo model [100] and in breast cancer [66], and it prevented alveolar bone loss in periodontitis [101]. The transfection of miRNA containing plasmid after EV isolation could ameliorate diabetic wounds [102], acute lung injury [103], or prevent tumor growth in the brain tumor model [104] and the A549 non-small-cell lung cancer model [105]. However, further research is required to develop efficient methods for the preferential sorting of cargo into sEVs and the loading of the detached cargo from the membranes of sEVs.

2.4. sEV Therapeutics in Clinical Trials

Since sEVs can recapitulate the comprehensive therapeutic potential of the donor cell, clinical trials utilizing MSC-derived sEVs are being extensively researched to evaluate the safety of treatment and efficacy on various diseases. The therapeutic dosage widely ranges from 1.2 × 1012 to 1.22 × 106 sEV particles per injection, and sEV source cells are diverse, such as adipose-MSCs, bone-marrow-MSCs, and synovial fluid-MSCs.
Recently, sEV treatments for COVID-19 and its complications are also being tested. Since most complications involve respiratory diseases, such as pneumonia and acute respiratory distress syndrome, not only intravenous injection but also the inhalation of sEVs is actively being tested (NCT04969172, NCT04389385, NCT04276987, NCT04747574, NCT04602104). Hitherto, EVs across diverse cellular origins are undergoing clinical trials for a wide range of diseases—from non-life-threatening hair loss to complex neoplasm diseases such as cancer, as shown in Table 5.
Table 4. Therapeutics using engineered sEVs.
Table 4. Therapeutics using engineered sEVs.
StrategySource cellsEV Purification and EngineeringLoaded Cargo AmountDiseaseTherapeutic ScheduleResultReference
Surface
engineering of sEVs
Mouse spleen-derived CD4+ T cellsUltracentrifugation and incubation with anti-VEGF10 anti VEGF molecules per sEVChoroidal neovascularization10 μg sEVs, intravitreal injection per eyesSuppressed angiogenesis and inflammation[94]
Adipose-derived stem cellsUltracentrifugation and primed with 100 ng/mL IFNγ overnightN/ATendon injuryEV-laden collagen sheet containing 5–6 × 109 EVs applied around the injury siteAmeliorated the repair site’s inflammatory response, regenerated tendon matrix[95]
Human umbilical cord MSCsUltracentrifugation and transfection of IL-10-overexpressing vector plasmidsN/AAutoimmune uveitis50 μg sEVs, intravenous injection on day 11 post-immunizationModulated T cell immunity[96]
Human placental MSCsUltracentrifugation and expression of collagen-binding peptide using click chemistryN/AIschemic disease1 × 1010 sEVs, intramuscular injection into four different sites around the hind limb ischemic regionReduced inflammation and increased muscle regeneration[97]
Human umbilical vascular endothelial cells Ultracentrifugation and incubation with anti-angiogenic peptide KV11-anchoring peptide CP05 83.1% EVs anchoring KV11 peptides Retina neovascularization 50 μg sEVs, retro-orbital injection on day 12Suppressed neovascularization[98]
Loading
cargo into sEVs
Mouse bone marrow-derived macrophagesExoQuick-TC™ Kit and incubation and sonication with Paclitaxel10 mg/mL Paclitaxel mixed with 1011 sEVsPulmonary metastases4 × 1011 particles of sEVs, systemic injection, three times on day 1, 4, and 7Suppressed neoplastic effect and increased survival period[99]
Rat marrow stromal cells ExoQuick-TC™ Kit and transfection of miR-67 or 146b plasmidN/ABrain tumor50 μg sEV, intratumoral injection on day 5Reduced glioma growth[104]
Human adipose-derived stem cellsUltracentrifugation and loading of miR-21-5p into sEVs by electroporationN/ADiabetic wound5 μg/200 μL solution applied to the wound bed every 3 days for 15 daysAmeliorated diabetic wounds[102]
HEK293exoEasy Maxi Kit and incubation for loading of curcumin into sEVs1 μg curcumin in 2.04–2.46 × 109 sEVsAcute lung injury15 μg sEVs, intratracheal instillation into the lungsReduced proinflammatory cytokines[103]
A549Centrifugation and transfection of TAT and TAR-miR-449a plasmidsN/ANon-small-cell lung cancer2 mg/kg sEVs, intravenous injectionSuppressed tumor growth[105]
RAW 264.7ExoQuick-TC™ Kit and incubation, electroporation, and sonication with PaclitaxelN/ALung metastasis1 × 107 particles, intravenous injection every other day, 7 timesAnticancer effect in Lewis lung carcinoma[100]
RAW 264.7Ultracentrifugation and incubation, electroporation, and sonication with catalase1376 ± 64.1 U of catalase activity in 4 × 1011 sEV/mLParkinson’s disease1.2 × 109 sEVs, injection into each nostril, 10 times every other dayReduced microgliosis and protected neurons[106]
RAW 264.7 M1 macrophageUltracentrifugation and sonication with PaclitaxelN/ABreast cancer0.1 mg sEVs, intravenous injection every 3 days for 27 daysAnti-tumor effects[66]
Mouse bone marrow-derived dendritic cells Ultracentrifugation and sonication with Paclitaxel5 μg TGFB1 and 5 μg IL10 in 1 × 109Periodontitis200 µL sEVs, intravenous or palatal tissue local injection on day 3 Prevented cytokines from proteolytic attack and alveolar bone loss[101]
Table 5. sEV therapeutics in clinical trials.
Table 5. sEV therapeutics in clinical trials.
Applied EVsDiseasesDosing SchedulesExpected ResultsPatientsPhaseRecruitment StatusIdentifier
Amniotic MSC-sEVHair loss, alopecia1012 sEVs administered through an interval of 14 days over two monthsAnticipation of growth factors contained in stem cell-sEV to improve hair loss20N/ARecruitingNCT05658094
Placenta MSC-sEVComplex anal fistulaN/A Anticipation of anal fistula treatment effect using stem cell sEVs80Phase 1, Phase 2RecruitingNCT05402748
MSC-sEVCerebrovascular disordersN/AEvaluation of improvement in acute ischemic stroke patients receiving MSC-sEVs5Phase 1, Phase 2UnknownNCT03384433
Embryonic kidney T-REx™-293 cell-sEVCOVID-191010 sEVs/4 mL normal saline administered through inhalation, once a day for 5 daysEvaluation of the safety and efficacy of sEVs overexpressing CD24155Phase 2Active, not recruitingNCT04969172
Adipose tissue stem cell-sEVPeriodontitisN/AEvaluation of regeneration effect10Early Phase 1UnknownNCT04270006
Plasma-sEVUlcerN/AAnticipation of skin wound-healing efficacy of plasma sEVs5Early Phase 1UnknownNCT02565264
sEVNeuralgiaN/AEvaluation of safety and efficacy of sEVs in patients with craniofacial neuralgia100N/ASuspendedNCT04202783
MSC-sEVMyocardial infarction,
myocardial ischemia,
myocardial stunning
100 μg/mL sEVs administered through intracoronary and intra-myocardial injection Anticipation of improvement in patient outcomes from the detrimental effects of ischemia and reperfusion injury20Phase 1, Phase 2RecruitingNCT05669144
sEVRefractory depression, anxiety disorders, neurodegenerative diseases2.1 × 107 allogenic sEVs/15 mL administered through intravenous injectionEvaluation of efficacy of sEVs in patients with neurodegenerative dementia300N/ASuspendedNCT04202770
MSC-sEVSARS-CoV-2 infectionsEV administered through intravenous injection twice, in day 1 and day 7 of 2 weeksEvaluation of efficacy of MSC-sEVs in reducing hyper-inflammation in patients with moderate COVID-1960Phase 2, Phase 3RecruitingNCT05216562
MSC-sEVMultiple organ failure180 mg sEV administered through intravenous injection once a day for 14 daysEvaluation of efficacy of MSC-sEVs for multiple organ dysfunction syndrome60N/ANot yet recruitingNCT04356300
Mesenchymal progenitor cell-sEVDrug-resistant(8 or 16) × 103 sEVs/3 mL administered through inhalation 7 times, day 1 to 7Evaluation of efficacy of sEV treatment for pulmonary infection caused by carbapenem-resistant gram-negative bacilli60Phase 1, Phase 2RecruitingNCT04544215
Wharton jelly MSC-sEVRetinitis pigmentosasEVs administered through single subtenon injectionEvaluation of efficacy of umbilical cord MSC-sEVs in the treatment of retinitis pigmentosa135Phase 2, Phase 3RecruitingNCT05413148
COVID-19-specific T cell-sEVCorona virus infection
pneumonia
2.0 × 103 sEVs/3 mL administered once a day for 5 daysEvaluation of efficacy after targeted delivery of T cell sEVs by metered-dose inhaler60Phase 1UnknownNCT04389385
MSC-sEVPsoriasis100 µg MSC sEV/g of ointment was dripped once a day for 20 days Anticipation of psoriasis treatment efficacy using MSC-sEV ointment10Phase 1CompletedNCT05523011
Adipose MSC-sEVCorona virus2.0 × 108 sEVs/3 mL administered through aerosol inhalation 5 times for 5 daysAnticipation of safety and efficacy of stem cell sEVs in treatment of severely ill patients hospitalized with novel coronavirus pneumonia24Phase 1CompletedNCT04276987
MSC-sEVCOVID-19, novel coronavirus pneumonia, acute respiratory distress syndrome(2, 4, and 8 × 109 or 8, 4, and 8 × 109) sEVs/mL administered through injection every other day for 5 days or 8 × 109 sEV administered through injection every other day for 5 daysAnticipation of treatment effects of stem cell sEVs in treatments of patients with acute respiratory distress syndrome and novel coronavirus pneumonia55Phase 1, Phase 2Not yet recruitingNCT04798716
Neonatal stem cell-sEVNeuralgiaN/AAnticipation of treatment in neuralgia patients using neonatal sEVs100N/ASuspendedNCT04202783
MSC-sEVOsteoarthritis(3–5) × 1011 sEVs administered through single dose injectionAnticipation of knee arthritis treatment effect using stem cell sEVs10Phase 1Not yet recruitingNCT05060107
T-REx™-293 cell-sEVSARS-CoV-2(1, 5, 10, 100) × 108 sEVs/2 mL saline) administered through QD inhalation deviceAnticipation of therapeutic effect in moderate/severe COVID-19 patients with sEV-CD2435Phase 1RecruitingNCT04747574
BM MSC-sEVFamilial hypercholesterolemia(0.044, 0.088, 0.145, 0.220, 0.295, or 0.394 mg/kg) sEV administered through single dose injectionAnticipation of treatment of hypercholesterolemia patients with sEV-based LDLR mRNA nano platform30Phase 1Not yet recruitingNCT05043181
Synovial fluid MSC-sEVDegenerative meniscal injury1 × 106 sEVs/kg administered through intra-articular injectionAnticipation of efficacy of synovial MSC-sEVs in patients with degenerative meniscal cartilage damage 30Phase 2RecruitingNCT05261360
Cell free umbilical cord-blood-MSC-sEVDiabetes mellitus type 1(1.22–1.51) × 106 sEVs/kg administered through intravenous injectionAnticipation of cord blood-sEVs to treat type 1 diabetes patients20Phase 2, Phase 3UnknownNCT02138331
Umbilical MSC-sEVDry eye10 ug/drop sEV administered through eye drops 4 times a day for 14 daysAnticipation of MSC-sEVs to alleviate dry eye symptoms in patients with chronic graft versus host diseases27Phase 1, Phase 2RecruitingNCT04213248
Adipose-sEVWounds and injuriesN/AAnticipation of sEVs to promote wound healing5N/ANot yet recruitingNCT05475418
MSC-sEVCOVID-19 SARS-CoV-2 pneumonia(0.5–2 × 1010) sEVs of the first or second type/3 mL administered through inhalation twice a day for 10 days Anticipation of safety and efficacy of sEV inhalation method for COVID-19-associated pneumonia90Phase 2UnknownNCT04602442
sEVCOVID-19(1 or 10) × 109 sEVs administered once a day for 5 daysAnticipation of safety and efficacy assessment of CD24 overexpressing sEVs for patients with severe COVID-1990Phase 2RecruitingNCT04902183
MSC-sEVAcute respiratory distress syndrome(2, 8, 16) × 108 sEVs administered through inhalation 7 times 1/4 MTD/day, or MTD/day at day 1 to 7Anticipation of treatment effects of MSC-sEVs in acute respiratory distress syndrome169Phase 1, Phase 2UnknownNCT04602104
MSC-sEVMacular hole50 or 20 μg/10 μL sEV was dripped into vitreous cavity around macular holesEvaluation of safety and efficacy evaluation of MSC-sEVs for promoting healing of large and refractory macular holes44Early Phase 1Active, not recruitingNCT03437759
Adipose MSC-sEVAlzheimer’s disease(5, 10, or 20 μg/1 mL) sEV administered through nasal drip twice a week for 12 weeksEvaluation of safety and efficacy evaluation of allogeneic adipose MSC-sEVs in patients with Alzheimer’s disease9Phase 1, Phase 2UnknownNCT04388982
MSC-sEVDystrophic epidermolysis bullosaN/AEvaluation of the safety and efficacy of sEVs in the treatment of lesions in patients with epidermolysis bullosa10Phase 1, Phase 2Not yet recruitingNCT04173650
ASO-STAT6-sEVAdvanced hepatocellular carcinoma, gastric cancer metastatic to liver colorectal cancer metastatic to liverN/AEvaluation of treatment efficacy for advanced hepatocellular carcinoma and primary gastric cancer with ASO-STAT6 sEVs (CDK-004)30Phase 1RecruitingNCT05375604
sEVCutaneous T-cell lymphomaN/AEvaluation of safety, tolerability, pharmacokinetics, and pharmacodynamic effects of CDK-0032Phase 1TerminatedNCT05156229
BM MSC-sEVCOVID-19 acute respiratory distress syndrome1.2 × 1012 sEVs/85 mL, 8 × 1011 sEVs/90 mL administered through intravenous injectionEvaluation of safety and efficacy of acute respiratory distress syndrome treatment using DB-0018 SEV120Phase 2CompletedNCT04493242
DC-sEVNon-small-cell lung cancer1.3 × 1013 MHC class II molecules administered through injection, four doses at weekly intervalsEvaluation of the safety, feasibility, and efficacy of administering tumor antigen-loaded autologous dexosomes to patients with advanced non-small-cell lung cancer (NSCLC)13Phase 1Completed
Turmeric-sEVColon cancer3.6 g curcumin-conjugated sEV tablets taken daily for 7 daysEvaluation of the ability of sEVs to deliver curcumin more effectively to normal colon tissues and colon tumors7Phase 1Active, not recruitingNCT01294072

3. Issues to Overcome for Realization of sEV Therapeutics

3.1. Biodistribution

An sEV biodistribution (BD) evaluation should be conducted to create a new, effective class of medicines and to begin the first in-human studies. Some questions still need to be answered regarding the BD of sEVs. For example, how do the different administration routes affect sEVs’ BD? What is the best labeling method for sEVs? To date, the most popular administration route for sEVs in preclinical studies is intravenous injection, occupying more than half of the total [107]. Much evidence has shown that the primary accumulation organs of intravenously injected sEVs are the liver and spleen, the reticuloendothelial systems [107]. However, studies comparing the efficacy of sEVs’ BD using different injection routes are scarce [108]; therefore, further studies of sEVs’ BD for the delivery of therapeutic cargo using sEVs to specific organs, including the lungs and brain, are required.
Despite many studies attempting to accurately assess the BD of sEVs using diverse labeling methods, the gold standard for labeling EVs has yet to be determined. The most widely utilized labeling approach is lipophilic fluorescent dye, including PKH, and diakylcarbocyanine dyes (DiD, Dil, Dio, and DiR), which can be readily integrated into the membranes of sEVs. However, these lipophilic dyes may aggregate sEVs and cause background/pseudo signals. Moreover, these dyes eventually affect the composition of the surfaces of sEVs, leading to effects on the biological activity of sEVs [109]. Several other methods have been attempted, such as encapsulation, metabolic labeling (e.g., click chemistry), and surface modification by genetic engineering (luciferase). However, these approaches showed limitations in measuring the accurate pharmacokinetics (PK) of sEVs [109]. Some papers have shown a covalent binding method using Cy dyes (Cy5.5 and Cy 7) or radioisotopes (64Cu, 68Ga, 125I, 99mTc, and 89Zr) to sEV surfaces [110]. Although this covalent binding may also affect the interaction of sEVs and targeted cells, this method has a low risk of eliciting pseudo-signals caused by the release of free-from dyes from dye-conjugated sEVs [110]. Moreover, radioisotope-based imaging with positron emission tomography (PET) and single-photon emission computed tomography (SPECT) were shown to be highly sensitive over in vivo tracking compared with fluorescence or luminescence imaging [111]. Taken together, we can begin to understand the BD and PK of sEVs with recent advancements in labeling methods, but further work is required to reveal accurate data.

3.2. Large-Scale Manufacturing

Compared to conventional therapeutics such as protein drugs, antibodies, and cell or gene therapeutics, there is no state of the art for the large-scale production of EV products, and there is also no concrete regulation or guidance from a regulatory board such as the FDA or EMA. Nonetheless, current EV therapeutics are actively being developed; thus, the establishment of manufacturing protocols and regulatory guidelines is needed. Since EVs are retrieved from naïve or engineered cells, the overall production process is perceived as similar to that of cell or gene therapy products. The master cell banking process is required to collect sEVs to maintain the cell homology, such as surface molecule expression, intracellular content, and engineered traits. In terms of engineered EVs, ex vivo manipulation is the leading strategy. For instance, the cells can be transduced with a retrovirus, adenovirus, or lentivirus to express the desired EVs stably [112]. The transduced stable cells are stored and managed in cryopreserved form, often referred to as a master cell bank (MCB), which can be utilized to assess product quality. EV manufacturing processes can be divided into two main types: upstream process development (USP) and downstream process development (DSP).
The MCBs or banked source cells are used for USP. The banked cells are thawed and expanded through the culture process. Cells are seeded on an appropriate culture dish, depending on the cell type. Alternative culture systems, such as 3D fiber cell systems, cell stacks, or seed trains, are used to increase the production of sEVs per cell, since these platforms improve cell viability and enable high-density cell growth. After adequate expansion, cells are transferred to a 3D bioreactor, an automated system optimized for cell growth. Once the cells are fully expanded, the culture medium is exchanged for serum-free media to inhibit soluble protein contamination and secure the purity of the final product. During downstream process development, serum-free medium is collected, and purified EVs are isolated. There are no set standard procedures, but most DSP resembles that of cell or gene therapeutic development. DSP focuses on collecting high-purity EVs with desired yields, appropriate for commercialization. Since the yield of the product and purity are trade-offs, the key in DSP development is to optimize both variables for a high-quality product with a practical outcome. The initial step is to remove the potential contaminants and collect small-sized EVs by depth filtration. Serial filtration, or depth filtration, sorts desired EVs from non-desired EVs through size cut-offs. This step is similar to the serial centrifugation process during the lab-scale production of EVs. Once the filtered EVs are retrieved, the product undergoes tangential flow filtration (TFF) to minimize the damage of EVs, maximize the purity, and concentrate the media into higher concentrations. The concentrated product undergoes a chromatography step to enhance the purity. Different types of chromatography columns are used, such as size exclusion chromatography or ion exchange chromatography, depending on the physical and chemical features of the EVs. Since most chromatography steps result in the dilution of the samples, the retrieved EVs often undergo TFF once more for concentration. The final drug products are packaged through fill-and-finish procedures. Though the gold standard of this step is also not yet established, many CDMOs and biopharmaceuticals lyophilize acquired EVs for higher stability and to facilitate storage and transport.

3.3. Quality Control

Quality control (QC) tests are crucial for the clinical translation of sEV therapeutics. Protein- and small-chemical-based medicines should be verified as a homogenous population through several robust QC tests. However, the heterogeneous populations of sEVs cannot be converted into a homogenous population. Instead, the batch-to-batch consistency demonstrated by appropriate QC tests is the major priority for the GMP-grade manufacturing of sEV therapeutics. To prove the batch-to-batch consistency, we must establish a list of QC tests on final products (sEVs) with a sufficient scientific rationale to persuade the FDA or other regulatory authorities to approve human clinical trials. The International Society for Extracellular Vesicles (ISEV) suggested minimal requirements in the MISEV2018 guidelines for the quality control of sEVs [1]. According to their guidelines, QC items of sEVs include the quantification (particle number, protein, and lipid), size (<200 nm), identification (the positive and negative markers), and purity (ratios of proteins or lipids: particles or proteins: lipids).
With the advancement of single-particle analysis technologies, quantifying the amount of therapeutic cargo loaded into a single particle is becoming feasible. Developed by NanoView Biosciences, the Exoview R200 automatically analyzes the EVs’ number and size data through probed tetraspanin markers, including CD63, CD81, and CD9, by taking micro-biochip-based fluorescent images [113]. Furthermore, by customizing the biochip according to the experimenter’s needs, the amount of therapeutic cargo loaded in EVs can be measured for each particle. Similarly, NanoAnalyzer, developed by NanoFCM, is a device that distinguishes between sEVs with a diameter of 40 nm and protein aggregates, enabling the evaluation of the characteristics of single sEV using antibodies, such as flow cytometry, which analyzes cells [114]. NanoAnalyzer is cost-effective, since additional consumables other than the device are not required. Moreover, it is familiar to users due to the interface’s similarity to existing flow cytometry methods. These devices for single EV analysis can assess not only the shape and size of a single EV, but also EV markers, enabling the more accurate measurement of the purity of sEVs. These advanced developments in QC tests are rapidly accelerating the clinical realization of sEV therapeutics. Therefore, an essential task is to develop a rigorous assay suitably customized for each sEV therapeutic.

4. Conclusions and Perspectives

sEVs are naturally produced in our bodies and play vital roles in biological functions, with numerous advantages as a new class of medicines. Developing platform technologies and establishing therapeutic strategies that maximize the advantages of sEVs are considered the most significant paradigm shifts in creating new treatments. Although sEV therapeutics have not yet been approved and used in patients, numerous clinical trials based on sEVs have recently been attempted, and the numbers are constantly increasing. The large-scale manufacturing and QC of sEVs, which were previously inconceivable, have also made much progress in convincing regulatory authorities. There are still issues to be solved, but we expect that continuous technological development and research will establish innovative sEV-based treatments as promising therapeutic options to solve existing high unmet medical needs.

Author Contributions

Conceptualization, G.-H.N. and M.K. (Minsu Kwon); writing—original draft preparation, I.L., Y.C., D.-U.S., M.K. (Minjeong Kwon), S.K., H.J. and G.-H.N.; writing—review and editing, I.L., G.-H.N. and M.K. (Minsu Kwon); supervision, G.-H.N. and M.K. (Minsu Kwon); funding acquisition, G.-H.N. and M.K. (Minsu Kwon); G.-H.N. and M.K. (Minsu Kwon) contributed equally to this work. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by SHIFTBIO Inc., a Korea University Grant (K2225811), and a National Research Foundation of Korea (NRF) grant funded by the Ministry of Science and ICT of Korea, grant number 2020R1C1C1003539.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available in this article.

Conflicts of Interest

G.-H.N. is the co-founder and stockholder, and I.L., Y.C., and S.K. are employees, of SHIFTIBIO Inc. The other authors declare no conflicts of interest.

References

  1. Théry, C.; Witwer, K.W.; Aikawa, E.; Alcaraz, M.J.; Anderson, J.D.; Andriantsitohaina, R.; Antoniou, A.; Arab, T.; Archer, F.; Atkin-Smith, G.K.; et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): A position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J. Extracell. Vesicles 2018, 7, 1535750. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Nam, G.; Choi, Y.; Kim, G.B.; Kim, S.; Kim, S.A.; Kim, I. Emerging Prospects of Exosomes for Cancer Treatment: From Conventional Therapy to Immunotherapy. Adv. Mater. 2020, 32, e2002440. [Google Scholar] [CrossRef] [PubMed]
  3. Hill, A.F. Extracellular Vesicles and Neurodegenerative Diseases. J. Neurosci. 2019, 39, 9269–9273. [Google Scholar] [CrossRef] [PubMed]
  4. Fu, S.; Zhang, Y.; Li, Y.; Luo, L.; Zhao, Y.; Yao, Y. Extracellular vesicles in cardiovascular diseases. Cell Death Discov. 2020, 6, 68. [Google Scholar] [CrossRef]
  5. Sahoo, S.; Adamiak, M.; Mathiyalagan, P.; Kenneweg, F.; Kafert-Kasting, S.; Thum, T. Therapeutic and Diagnostic Translation of Extracellular Vesicles in Cardiovascular Diseases: Roadmap to the Clinic. Circulation 2021, 143, 1426–1449. [Google Scholar] [CrossRef] [PubMed]
  6. Kim, Y.K.; Choi, Y.; Nam, G.-H.; Kim, I.-S. Functionalized exosome harboring bioactive molecules for cancer therapy. Cancer Lett. 2020, 489, 155–162. [Google Scholar] [CrossRef]
  7. Bai, S.; Wang, Z.; Wang, M.; Li, J.; Wei, Y.; Xu, R.; Du, J. Tumor-Derived Exosomes Modulate Primary Site Tumor Metastasis. Front. Cell Dev. Biol. 2022, 10, 752818. [Google Scholar] [CrossRef]
  8. Tian, W.; Liu, S.; Li, B. Potential Role of Exosomes in Cancer Metastasis. BioMed Res. Int. 2019, 2019, 4649705. [Google Scholar] [CrossRef] [Green Version]
  9. Huang, S.; Dong, M.; Chen, Q. Tumor-Derived Exosomes and Their Role in Breast Cancer Metastasis. Int. J. Mol. Sci. 2022, 23, 13993. [Google Scholar] [CrossRef]
  10. Hessvik, N.P.; Llorente, A. Current knowledge on exosome biogenesis and release. Cell. Mol. Life Sci. 2018, 75, 193–208. [Google Scholar] [CrossRef]
  11. Datta, A.; Kim, H.; Lal, M.; McGee, L.; Johnson, A.; Moustafa, A.A.; Jones, J.C.; Mondal, D.; Ferrer, M.; Abdel-Mageed, A.B. Manumycin A suppresses exosome biogenesis and secretion via targeted inhibition of Ras/Raf/ERK1/2 signaling and hnRNP H1 in castration-resistant prostate cancer cells. Cancer Lett. 2017, 408, 73–81. [Google Scholar] [CrossRef] [PubMed]
  12. Greenberg, J.W.; Kim, H.; Ahn, M.; Moustafa, A.A.; Zhou, H.; Barata, P.C.; Boulares, A.H.; Abdel-Mageed, A.B.; Krane, L.S. Combination of Tipifarnib and Sunitinib Overcomes Renal Cell Carcinoma Resistance to Tyrosine Kinase Inhibitors via Tumor-Derived Exosome and T Cell Modulation. Cancers 2022, 14, 903. [Google Scholar] [CrossRef]
  13. Kosaka, N.; Iguchi, H.; Hagiwara, K.; Yoshioka, Y.; Takeshita, F.; Ochiya, T. Neutral Sphingomyelinase 2 (nSMase2)-dependent Exosomal Transfer of Angiogenic MicroRNAs Regulate Cancer Cell Metastasis. J. Biol. Chem. 2013, 288, 10849–10859. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Hekmatirad, S.; Moloudizargari, M.; Moghadamnia, A.A.; Kazemi, S.; Mohammadnia-Afrouzi, M.; Baeeri, M.; Moradkhani, F.; Asghari, M.H. Inhibition of Exosome Release Sensitizes U937 Cells to PEGylated Liposomal Doxorubicin. Front. Immunol. 2021, 12, 692654. [Google Scholar] [CrossRef]
  15. Tian, T.; Zhu, Y.-L.; Zhou, Y.-Y.; Liang, G.-F.; Wang, Y.-Y.; Hu, F.-H.; Xiao, Z.-D. Exosome Uptake through Clathrin-mediated Endocytosis and Macropinocytosis and Mediating miR-21 Delivery. J. Biol. Chem. 2014, 289, 22258–22267. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Chiba, M.; Kubota, S.; Sato, K.; Monzen, S. Exosomes released from pancreatic cancer cells enhance angiogenic activities via dynamin-dependent endocytosis in endothelial cells in vitro. Sci. Rep. 2018, 8, 11972. [Google Scholar] [CrossRef] [Green Version]
  17. Nanbo, A.; Kawanishi, E.; Yoshida, R.; Yoshiyama, H. Exosomes Derived from Epstein-Barr Virus-Infected Cells Are Internalized via Caveola-Dependent Endocytosis and Promote Phenotypic Modulation in Target Cells. J. Virol. 2013, 87, 10334–10347. [Google Scholar] [CrossRef] [Green Version]
  18. Atai, N.A.; Balaj, L.; Van Veen, H.; Breakefield, X.O.; Jarzyna, P.A.; Van Noorden, C.J.F.; Skog, J.; Maguire, C.A. Heparin blocks transfer of extracellular vesicles between donor and recipient cells. J. Neurooncol. 2013, 115, 343–351. [Google Scholar] [CrossRef] [Green Version]
  19. Christianson, H.C.; Svensson, K.J.; van Kuppevelt, T.H.; Li, J.-P.; Belting, M. Cancer cell exosomes depend on cell-surface heparan sulfate proteoglycans for their internalization and functional activity. Proc. Natl. Acad. Sci. USA 2013, 110, 17380–17385. [Google Scholar] [CrossRef] [Green Version]
  20. Franzen, C.A.; Simms, P.E.; Van Huis, A.F.; Foreman, K.E.; Kuo, P.C.; Gupta, G.N. Characterization of Uptake and Internalization of Exosomes by Bladder Cancer Cells. BioMed Res. Int. 2014, 2014, 619829. [Google Scholar] [CrossRef] [PubMed]
  21. McKelvey, K.J.; Powell, K.L.; Ashton, A.W.; Morris, J.M.; McCracken, S.A. Exosomes: Mechanisms of Uptake. J. Circ. Biomark. 2015, 4, 7. [Google Scholar] [CrossRef] [Green Version]
  22. Verdera, H.C.; Gitz-Francois, J.J.; Schiffelers, R.M.; Vader, P. Cellular uptake of extracellular vesicles is mediated by clathrin-independent endocytosis and macropinocytosis. J. Control Release 2017, 266, 100–108. [Google Scholar] [CrossRef] [PubMed]
  23. Lim, E.Y.; Park, J.; Kim, Y.T.; Kim, M.J. Imipramine Inhibits Migration and Invasion in Metastatic Castration-Resistant Prostate Cancer PC-3 Cells via AKT-Mediated NF-κB Signaling Pathway. Molecules 2020, 25, 4619. [Google Scholar] [CrossRef] [PubMed]
  24. Lin, H.-P.; Singla, B.; Ghoshal, P.; Faulkner, J.L.; Cherian-Shaw, M.; O’Connor, P.M.; She, J.-X.; de Chantemele, E.J.B.; Csányi, G. Identification of novel macropinocytosis inhibitors using a rational screen of Food and Drug Administration-approved drugs. Br. J. Pharmacol. 2018, 175, 3640–3655. [Google Scholar] [CrossRef] [Green Version]
  25. Kumagai, T.; Shimogawara, R.; Ichimura, K.; Iwanaga, S. Calpain inhibitor suppresses both extracellular vesicle-mediated secretion of miRNAs and egg production from paired adults of Schistosoma japonicum. Parasitol. Int. 2022, 87, 102540. [Google Scholar] [CrossRef] [PubMed]
  26. Zheng, Y.; Tu, C.; Zhang, J.; Wang, J. Inhibition of multiple myeloma-derived exosomes uptake suppresses the functional response in bone marrow stromal cell. Int. J. Oncol. 2019, 54, 1061–1070. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Kimbrel, E.A.; Lanza, R. Next-generation stem cells—Ushering in a new era of cell-based therapies. Nat. Rev. Drug Discov. 2020, 19, 463–479. [Google Scholar] [CrossRef]
  28. Yagi, H.; Soto-Gutierrez, A.; Parekkadan, B.; Kitagawa, Y.; Tompkins, R.G.; Kobayashi, N.; Yarmush, M.L. Mesenchymal Stem Cells: Mechanisms of Immunomodulation and Homing. Cell Transplant. 2010, 19, 667–679. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Nikfarjam, S.; Rezaie, J.; Zolbanin, N.M.; Jafari, R. Mesenchymal stem cell derived-exosomes: A modern approach in translational medicine. J. Transl. Med. 2020, 18, 449. [Google Scholar] [CrossRef]
  30. Gowen, A.; Shahjin, F.; Chand, S.; Odegaard, K.E.; Yelamanchili, S.V. Mesenchymal Stem Cell-Derived Extracellular Vesicles: Challenges in Clinical Applications. Front. Cell Dev. Biol. 2020, 8, 149. [Google Scholar] [CrossRef]
  31. Lai, R.C.; Arslan, F.; Lee, M.M.; Sze, N.S.K.; Choo, A.; Chen, T.S.; Salto-Tellez, M.; Timmers, L.; Lee, C.N.; El Oakley, R.M.; et al. Exosome secreted by MSC reduces myocardial ischemia/reperfusion injury. Stem Cell Res. 2010, 4, 214–222. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Hsieh, J.-Y.; Wang, H.-W.; Chang, S.-J.; Liao, K.-H.; Lee, I.-H.; Lin, W.-S.; Wu, C.-H.; Lin, W.-Y.; Cheng, S.-M. Mesenchymal Stem Cells from Human Umbilical Cord Express Preferentially Secreted Factors Related to Neuroprotection, Neurogenesis, and Angiogenesis. PLoS ONE 2013, 8, e72604. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Zhao, J.; Li, X.; Hu, J.; Chen, F.; Qiao, S.; Sun, X.; Gao, L.; Xie, J.; Xu, B. Mesenchymal stromal cell-derived exosomes attenuate myocardial ischaemia-reperfusion injury through miR-182-regulated macrophage polarization. Cardiovasc. Res. 2019, 115, 1205–1216. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Cone, A.S.; Yuan, X.; Sun, L.; Duke, L.C.; Vreones, M.P.; Carrier, A.N.; Kenyon, S.M.; Carver, S.R.; Benthem, S.D.; Stimmell, A.C.; et al. Mesenchymal stem cell-derived extracellular vesicles ameliorate Alzheimer’s disease-like phenotypes in a preclinical mouse model. Theranostics 2021, 11, 8129–8142. [Google Scholar] [CrossRef]
  35. Shen, Z.; Huang, W.; Liu, J.; Tian, J.; Wang, S.; Rui, K. Effects of Mesenchymal Stem Cell-Derived Exosomes on Autoimmune Diseases. Front. Immunol. 2021, 12, 749192. [Google Scholar] [CrossRef] [PubMed]
  36. Kholia, S.; Sanchez, M.B.H.; Cedrino, M.; Papadimitriou, E.; Tapparo, M.; Deregibus, M.C.; Brizzi, M.F.; Tetta, C.; Camussi, G. Human Liver Stem Cell-Derived Extracellular Vesicles Prevent Aristolochic Acid-Induced Kidney Fibrosis. Front. Immunol. 2018, 9, 1639. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Jiang, Z.-Z.; Liu, Y.-M.; Niu, X.; Yin, J.-Y.; Hu, B.; Guo, S.-C.; Fan, Y.; Wang, Y.; Wang, N.-S. Exosomes secreted by human urine-derived stem cells could prevent kidney complications from type I diabetes in rats. Stem Cell Res. Ther. 2016, 7, 24. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Fujii, S.; Miura, Y.; Fujishiro, A.; Shindo, T.; Shimazu, Y.; Hirai, H.; Tahara, H.; Takaori-Kondo, A.; Ichinohe, T.; Maekawa, T. Graft-Versus-Host Disease Amelioration by Human Bone Marrow Mesenchymal Stromal/Stem Cell-Derived Extracellular Vesicles Is Associated with Peripheral Preservation of Naive T Cell Populations. Stem Cells 2017, 36, 434–445. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. An, J.-H.; Li, Q.; Bhang, D.-H.; Song, W.-J.; Youn, H.-Y. TNF-α and INF-γ primed canine stem cell-derived extracellular vesicles alleviate experimental murine colitis. Sci. Rep. 2020, 10, 2115. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Bruno, S.; Collino, F.; Deregibus, M.C.; Grange, C.; Tetta, C.; Camussi, G. Microvesicles Derived from Human Bone Marrow Mesenchymal Stem Cells Inhibit Tumor Growth. Stem Cells Dev. 2013, 22, 758–771. [Google Scholar] [CrossRef]
  41. Mathew, B.; Ravindran, S.; Liu, X.; Torres, L.; Chennakesavalu, M.; Huang, C.-C.; Feng, L.; Zelka, R.; Lopez, J.; Sharma, M.; et al. Mesenchymal stem cell-derived extracellular vesicles and retinal ischemia-reperfusion. Biomaterials 2019, 197, 146–160. [Google Scholar] [CrossRef] [PubMed]
  42. Li, D.; Zhang, J.; Liu, Z.; Gong, Y.; Zheng, Z. Human umbilical cord mesenchymal stem cell-derived exosomal miR-27b attenuates subretinal fibrosis via suppressing epithelial–mesenchymal transition by targeting HOXC6. Stem Cell Res. Ther. 2021, 12, 42. [Google Scholar] [CrossRef] [PubMed]
  43. He, G.-H.; Zhang, W.; Ma, Y.-X.; Yang, J.; Chen, L.; Song, J.; Chen, S. Mesenchymal stem cells-derived exosomes ameliorate blue light stimulation in retinal pigment epithelium cells and retinal laser injury by VEGF-dependent mechanism. Int. J. Ophthalmol. 2018, 11, 559–566. [Google Scholar] [CrossRef] [PubMed]
  44. Joshi, B.S.; Youssef, S.A.; Bron, R.; de Bruin, A.; Kampinga, H.H.; Zuhorn, I.S. DNAJB6b-enriched small extracellular vesicles decrease polyglutamine aggregation in in vitro and in vivo models of Huntington disease. Iscience 2021, 24, 103282. [Google Scholar] [CrossRef]
  45. Chen, Y.-A.; Lu, C.-H.; Ke, C.-C.; Chiu, S.-J.; Jeng, F.-S.; Chang, C.-W.; Yang, B.-H.; Liu, R.-S. Mesenchymal Stem Cell-Derived Exosomes Ameliorate Alzheimer’s Disease Pathology and Improve Cognitive Deficits. Biomedicines 2021, 9, 594. [Google Scholar] [CrossRef]
  46. Ding, M.; Shen, Y.; Wang, P.; Xie, Z.; Xu, S.; Zhu, Z.; Wang, Y.; Lyu, Y.; Wang, D.; Xu, L.; et al. Exosomes Isolated From Human Umbilical Cord Mesenchymal Stem Cells Alleviate Neuroinflammation and Reduce Amyloid-Beta Deposition by Modulating Microglial Activation in Alzheimer’s Disease. Neurochem. Res. 2018, 43, 2165–2177. [Google Scholar] [CrossRef]
  47. Cui, G.-H.; Guo, H.-D.; Li, H.; Zhai, Y.; Gong, Z.-B.; Wu, J.; Liu, J.-S.; Dong, Y.-R.; Hou, S.-X. RVG-modified exosomes derived from mesenchymal stem cells rescue memory deficits by regulating inflammatory responses in a mouse model of Alzheimer’s disease. Immun. Ageing 2019, 16, 10. [Google Scholar] [CrossRef] [Green Version]
  48. Wang, S.-S.; Jia, J.; Wang, Z. Mesenchymal Stem Cell-Derived Extracellular Vesicles Suppresses iNOS Expression and Ameliorates Neural Impairment in Alzheimer’s Disease Mice. J. Alzheimer’s Dis. 2018, 61, 1005–1013. [Google Scholar] [CrossRef]
  49. Cui, G.-H.; Wu, J.; Mou, F.-F.; Xie, W.-H.; Wang, F.-B.; Wang, Q.-L.; Fang, J.; Xu, Y.-W.; Dong, Y.-R.; Liu, J.-R.; et al. Exosomes derived from hypoxia-preconditioned mesenchymal stromal cells ameliorate cognitive decline by rescuing synaptic dysfunction and regulating inflammatory responses in APP/PS1 mice. FASEB J. 2018, 32, 654–668. [Google Scholar] [CrossRef] [Green Version]
  50. Elia, C.A.; Tamborini, M.; Rasile, M.; Desiato, G.; Marchetti, S.; Swuec, P.; Mazzitelli, S.; Clemente, F.; Anselmo, A.; Matteoli, M.; et al. Intracerebral Injection of Extracellular Vesicles from Mesenchymal Stem Cells Exerts Reduced Aβ Plaque Burden in Early Stages of a Preclinical Model of Alzheimer’s Disease. Cells 2019, 8, 1059. [Google Scholar] [CrossRef]
  51. Yang, L.; Zhai, Y.; Hao, Y.; Zhu, Z.; Cheng, G. The Regulatory Functionality of Exosomes Derived from hUMSCs in 3D Culture for Alzheimer’s Disease Therapy. Small 2019, 16, e1906273. [Google Scholar] [CrossRef] [PubMed]
  52. Wolfers, J.; Lozier, A.; Raposo, G.; Regnault, A.; Théry, C.; Masurier, C.; Flament, C.; Pouzieux, S.; Faure, F.; Tursz, T.; et al. Tumor-derived exosomes are a source of shared tumor rejection antigens for CTL cross-priming. Nat. Med. 2001, 7, 297–303. [Google Scholar] [CrossRef] [PubMed]
  53. Droste, M.; Thakur, B.K.; Eliceiri, B.P. Tumor-Derived Extracellular Vesicles and the Immune System—Lessons From Immune-Competent Mouse-Tumor Models. Front. Immunol. 2020, 11, 606859. [Google Scholar] [CrossRef] [PubMed]
  54. Santos, P.; Almeida, F. Exosome-Based Vaccines: History, Current State, and Clinical Trials. Front. Immunol. 2021, 12, 711565. [Google Scholar] [CrossRef]
  55. Stefanius, K.; Servage, K.A.; Santos, M.D.S.; Gray, H.F.; Toombs, J.E.; Chimalapati, S.; Kim, M.S.; Malladi, V.S.; Brekken, R.A.; Orth, K. Human pancreatic cancer cell exosomes, but not human normal cell exosomes, act as an initiator in cell transformation. eLife 2019, 8, e40226. [Google Scholar] [CrossRef]
  56. Kucharzewska, P.; Christianson, H.C.; Welch, J.E.; Svensson, K.J.; Fredlund, E.; Ringnér, M.; Mörgelin, M.; Bourseau-Guilmain, E.; Bengzon, J.; Belting, M. Exosomes reflect the hypoxic status of glioma cells and mediate hypoxia-dependent activation of vascular cells during tumor development. Proc. Natl. Acad. Sci. USA 2013, 110, 7312–7317. [Google Scholar] [CrossRef] [Green Version]
  57. Capello, M.; Vykoukal, J.V.; Katayama, H.; Bantis, L.E.; Wang, H.; Kundnani, D.L.; Aguilar-Bonavides, C.; Aguilar, M.; Tripathi, S.C.; Dhillon, D.S.; et al. Exosomes harbor B cell targets in pancreatic adenocarcinoma and exert decoy function against complement-mediated cytotoxicity. Nat. Commun. 2019, 10, 254. [Google Scholar] [CrossRef] [Green Version]
  58. Hoshino, A.; Costa-Silva, B.; Shen, T.-L.; Rodrigues, G.; Hashimoto, A.; Mark, M.T.; Molina, H.; Kohsaka, S.; Di Giannatale, A.; Ceder, S.; et al. Tumour exosome integrins determine organotropic metastasis. Nature 2015, 527, 329–335. [Google Scholar] [CrossRef] [Green Version]
  59. Romagnoli, G.G.; Zelante, B.B.; Toniolo, P.A.; Migliori, I.K.; Barbuto, J.A.M. Dendritic Cell-Derived Exosomes may be a Tool for Cancer Immunotherapy by Converting Tumor Cells into Immunogenic Targets. Front. Immunol. 2015, 5, 692. [Google Scholar] [CrossRef] [Green Version]
  60. Markov, O.; Oshchepkova, A.; Mironova, N. Immunotherapy Based on Dendritic Cell-Targeted/-Derived Extracellular Vesicles—A Novel Strategy for Enhancement of the Anti-tumor Immune Response. Front. Pharmacol. 2019, 10, 1152. [Google Scholar] [CrossRef]
  61. Wahlund, C.J.E.; Güclüler, G.; Hiltbrunner, S.; Veerman, R.E.; Näslund, T.I.; Gabrielsson, S. Exosomes from antigen-pulsed dendritic cells induce stronger antigen-specific immune responses than microvesicles in vivo. Sci. Rep. 2017, 7, 17095. [Google Scholar] [CrossRef] [Green Version]
  62. Xia, J.; Miao, Y.; Wang, X.; Huang, X.; Dai, J. Recent progress of dendritic cell-derived exosomes (Dex) as an anti-cancer nanovaccine. Biomed. Pharmacother. 2022, 152, 113250. [Google Scholar] [CrossRef]
  63. Lugini, L.; Cecchetti, S.; Huber, V.; Luciani, F.; Macchia, G.; Spadaro, F.; Paris, L.; Abalsamo, L.; Colone, M.; Molinari, A.; et al. Immune surveillance properties of human NK cell-derived exosomes. J. Immunol. 2012, 189, 2833–2842. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Zhu, L.; Kalimuthu, S.; Gangadaran, P.; Oh, J.M.; Lee, H.W.; Baek, S.H.; Jeong, S.Y.; Lee, S.-W.; Lee, J.; Ahn, B.-C. Exosomes derived from natural killer cells exert therapeutic effect in melanoma. Theranostics 2017, 7, 2732–2745. [Google Scholar] [CrossRef] [PubMed]
  65. Shoae-Hassani, A.; Hamidieh, A.A.; Behfar, M.; Mohseni, R.; Mortazavi-Tabatabaei, S.A.; Asgharzadeh, S. NK Cell–derived Exosomes From NK Cells Previously Exposed to Neuroblastoma Cells Augment the Antitumor Activity of Cytokine-activated NK Cells. J. Immunother. 2017, 40, 265–276. [Google Scholar] [CrossRef] [PubMed]
  66. Wang, P.; Wang, H.; Huang, Q.; Peng, C.; Yao, L.; Chen, H.; Qiu, Z.; Wu, Y.; Wang, L.; Chen, W. Exosomes from M1-Polarized Macrophages Enhance Paclitaxel Antitumor Activity by Activating Macrophages-Mediated Inflammation. Theranostics 2019, 9, 1714–1727. [Google Scholar] [CrossRef] [PubMed]
  67. 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, 1665–1675. [Google Scholar] [CrossRef] [Green Version]
  68. Aharon, A.; Horn, G.; Bar-Lev, T.H.; Yohay, M.E.Z.; Waks, M.T.; Levin, M.; Unger, N.D.; Avivi, I.; Levin, A.G. Extracellular Vesicles Derived from Chimeric Antigen Receptor-T Cells: A Potential Therapy for Cancer. Hum. Gene Ther. 2021, 32, 1224–1241. [Google Scholar] [CrossRef]
  69. Fu, W.; Lei, C.; Liu, S.; Cui, Y.; Wang, C.; Qian, K.; Li, T.; Shen, Y.; Fan, X.; Lin, F.; et al. CAR exosomes derived from effector CAR-T cells have potent antitumour effects and low toxicity. Nat. Commun. 2019, 10, 4355. [Google Scholar] [CrossRef] [Green Version]
  70. 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, 594–600. [Google Scholar] [CrossRef]
  71. Qiu, Y.; Yang, Y.; Yang, R.; Liu, C.; Hsu, J.-M.; Jiang, Z.; Sun, L.; Wei, Y.; Li, C.-W.; Yu, D.; et al. Activated T cell-derived exosomal PD-1 attenuates PD-L1-induced immune dysfunction in triple-negative breast cancer. Oncogene 2021, 40, 4992–5001. [Google Scholar] [CrossRef] [PubMed]
  72. Wang, G.; Hu, W.; Chen, H.; Shou, X.; Ye, T.; Xu, Y. Cocktail Strategy Based on NK Cell-Derived Exosomes and Their Biomimetic Nanoparticles for Dual Tumor Therapy. Cancers 2019, 11, 1560. [Google Scholar] [CrossRef] [Green Version]
  73. Yang, P.; Cao, X.; Cai, H.; Feng, P.; Chen, X.; Zhu, Y.; Yang, Y.; An, W.; Yang, Y.; Jie, J. The exosomes derived from CAR-T cell efficiently target mesothelin and reduce triple-negative breast cancer growth. Cell. Immunol. 2020, 360, 104262. [Google Scholar] [CrossRef] [PubMed]
  74. Gurbel, P.A.; Jeong, Y.-H.; Navarese, E.P.; Tantry, U.S. Platelet-Mediated Thrombosis: From Bench to Bedside. Circ. Res. 2016, 118, 1380–1391. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Tao, S.-C.; Guo, S.-C.; Zhang, C.-Q. Platelet-derived Extracellular Vesicles: An Emerging Therapeutic Approach. Int. J. Biol. Sci. 2017, 13, 828–834. [Google Scholar] [CrossRef] [Green Version]
  76. Guo, S.-C.; Tao, S.-C.; Yin, W.-J.; Qi, X.; Yuan, T.; Zhang, C.-Q. Exosomes derived from platelet-rich plasma promote the re-epithelization of chronic cutaneous wounds via activation of YAP in a diabetic rat model. Theranostics 2017, 7, 81–96. [Google Scholar] [CrossRef] [Green Version]
  77. Hayon, Y.; Dashevsky, O.; Shai, E.; Brill, A.; Varon, D.; Leker, R.R. Platelet Microparticles Induce Angiogenesis and Neurogenesis after Cerebral Ischemia. Curr. Neurovascular Res. 2012, 9, 185–192. [Google Scholar] [CrossRef]
  78. Labelle, M.; Begum, S.; Hynes, R.O. Direct signaling between platelets and cancer cells induces an epithelial-mesenchymal-like transition and promotes metastasis. Cancer Cell 2011, 20, 576–590. [Google Scholar] [CrossRef] [Green Version]
  79. Thangaraju, K.; Neerukonda, S.N.; Katneni, U.; Buehler, P.W. Extracellular Vesicles from Red Blood Cells and Their Evolving Roles in Health, Coagulopathy and Therapy. Int. J. Mol. Sci. 2020, 22, 153. [Google Scholar] [CrossRef]
  80. Zhang, G.; Huang, X.; Xiu, H.; Sun, Y.; Chen, J.; Cheng, G.; Song, Z.; Peng, Y.; Shen, Y.; Wang, J.; et al. Extracellular vesicles: Natural liver-accumulating drug delivery vehicles for the treatment of liver diseases. J. Extracell. Vesicles 2020, 10, e12030. [Google Scholar] [CrossRef]
  81. Chiangjong, W.; Netsirisawan, P.; Hongeng, S.; Chutipongtanate, S. Red Blood Cell Extracellular Vesicle-Based Drug Delivery: Challenges and Opportunities. Front. Med. 2021, 8, 761362. [Google Scholar] [CrossRef] [PubMed]
  82. Cho, E.; Nam, G.-H.; Hong, Y.; Kim, Y.K.; Kim, D.-H.; Yang, Y.; Kim, I.-S. Comparison of exosomes and ferritin protein nanocages for the delivery of membrane protein therapeutics. J. Control Release 2018, 279, 326–335. [Google Scholar] [CrossRef]
  83. Van Niel, G.; D’Angelo, G.; Raposo, G. Shedding light on the cell biology of extracellular vesicles. Nat. Rev. Mol. Cell Biol. 2018, 19, 213–228. [Google Scholar] [CrossRef]
  84. Kim, G.B.; Nam, G.-H.; Hong, Y.; Woo, J.; Cho, Y.; Kwon, I.C.; Yang, Y.; Kim, I.-S. Xenogenization of tumor cells by fusogenic exosomes in tumor microenvironment ignites and propagates antitumor immunity. Sci. Adv. 2020, 6, eaaz2083. [Google Scholar] [CrossRef]
  85. Koh, E.; Lee, E.J.; Nam, G.-H.; Hong, Y.; Cho, E.; Yang, Y.; Kim, I.-S. Exosome-SIRPα, a CD47 blockade increases cancer cell phagocytosis. Biomaterials 2017, 121, 121–129. [Google Scholar] [CrossRef]
  86. Conceição, M.; Forcina, L.; Wiklander, O.P.; Gupta, D.; Nordin, J.Z.; Vrellaku, B.; McClorey, G.; Mäger, I.; Görgens, A.; Lundin, P.; et al. Engineered extracellular vesicle decoy receptor-mediated modulation of the IL6 trans-signalling pathway in muscle. Biomaterials 2020, 266, 120435. [Google Scholar] [CrossRef] [PubMed]
  87. Dooley, K.; McConnell, R.E.; Xu, K.; Lewis, N.D.; Haupt, S.; Youniss, M.R.; Martin, S.; Sia, C.L.; McCoy, C.; Moniz, R.J.; et al. A versatile platform for generating engineered extracellular vesicles with defined therapeutic properties. Mol. Ther. 2021, 29, 1729–1743. [Google Scholar] [CrossRef] [PubMed]
  88. Hu, K.; McKay, P.F.; Samnuan, K.; Najer, A.; Blakney, A.K.; Che, J.; O’Driscoll, G.; Cihova, M.; Stevens, M.M.; Shattock, R.J. Presentation of antigen on extracellular vesicles using transmembrane domains from viral glycoproteins for enhanced immunogenicity. J. Extracell. Vesicles 2022, 11, e12199. [Google Scholar] [CrossRef] [PubMed]
  89. Schoenmaker, L.; Witzigmann, D.; Kulkarni, J.A.; Verbeke, R.; Kersten, G.; Jiskoot, W.; Crommelin, D.J. mRNA-lipid nanoparticle COVID-19 vaccines: Structure and stability. Int. J. Pharm. 2021, 601, 120586. [Google Scholar] [CrossRef] [PubMed]
  90. Sahay, G.; Querbes, W.; Alabi, C.; Eltoukhy, A.; Sarkar, S.; Zurenko, C.; Karagiannis, E.; Love, K.; Chen, D.; Zoncu, R.; et al. Efficiency of siRNA delivery by lipid nanoparticles is limited by endocytic recycling. Nat. Biotechnol. 2013, 31, 653–658. [Google Scholar] [CrossRef]
  91. Bonsergent, E.; Grisard, E.; Buchrieser, J.; Schwartz, O.; Théry, C.; Lavieu, G. Quantitative characterization of extracellular vesicle uptake and content delivery within mammalian cells. Nat. Commun. 2021, 12, 1864. [Google Scholar] [CrossRef] [PubMed]
  92. Yao, Z.; Qiao, Y.; Li, X.; Chen, J.; Ding, J.; Bai, L.; Shen, F.; Shi, B.; Liu, J.; Peng, L.; et al. Exosomes Exploit the Virus Entry Machinery and Pathway To Transmit Alpha Interferon-Induced Antiviral Activity. J. Virol. 2018, 92, e01578-18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Liang, Y.; Duan, L.; Lu, J.; Xia, J. Engineering exosomes for targeted drug delivery. Theranostics 2021, 11, 3183–3195. [Google Scholar] [CrossRef]
  94. Tian, Y.; Zhang, F.; Qiu, Y.; Wang, S.; Li, F.; Zhao, J.; Pan, C.; Tao, Y.; Di Yu, D.; Wei, W. Reduction of choroidal neovascularization via cleavable VEGF antibodies conjugated to exosomes derived from regulatory T cells. Nat. Biomed. Eng. 2021, 5, 968–982. [Google Scholar] [CrossRef]
  95. Shen, H.; Yoneda, S.; Abu-Amer, Y.; Guilak, F.; Gelberman, R.H. Stem cell-derived extracellular vesicles attenuate the early inflammatory response after tendon injury and repair. J. Orthop. Res. 2019, 38, 117–127. [Google Scholar] [CrossRef]
  96. Li, Y.; Ren, X.; Zhang, Z.; Duan, Y.; Li, H.; Chen, S.; Shao, H.; Li, X.; Zhang, X. Effect of small extracellular vesicles derived from IL-10-overexpressing mesenchymal stem cells on experimental autoimmune uveitis. Stem Cell Res. Ther. 2022, 13, 100. [Google Scholar] [CrossRef]
  97. Hao, D.; Lu, L.; Song, H.; Duan, Y.; Chen, J.; Carney, R.; Li, J.J.; Zhou, P.; Nolta, J.; Lam, K.S.; et al. Engineered extracellular vesicles with high collagen-binding affinity present superior in situ retention and therapeutic efficacy in tissue repair. Theranostics 2022, 12, 6021–6037. [Google Scholar] [CrossRef] [PubMed]
  98. Dong, X.; Lei, Y.; Yu, Z.; Wang, T.; Liu, Y.; Han, G.; Zhang, X.; Li, Y.; Song, Y.; Xu, H.; et al. Exosome-mediated delivery of an anti-angiogenic peptide inhibits pathological retinal angiogenesis. Theranostics 2021, 11, 5107–5126. [Google Scholar] [CrossRef]
  99. Kim, M.S.; Haney, M.J.; Zhao, Y.; Yuan, D.; Deygen, I.; Klyachko, N.L.; Kabanov, A.V.; Batrakova, E.V. Engineering macrophage-derived exosomes for targeted paclitaxel delivery to pulmonary metastases: In vitro and in vivo evaluations. Nanomedicine 2018, 14, 195–204. [Google Scholar] [CrossRef]
  100. Kim, M.S.; Haney, M.J.; Zhao, Y.; Mahajan, V.; Deygen, I.; Klyachko, N.L.; Inskoe, E.; Piroyan, A.; Sokolsky, M.; Okolie, O.; et al. Development of exosome-encapsulated paclitaxel to overcome mdr in cancer cells. Nanomedicine 2016, 12, 655–664. [Google Scholar] [CrossRef]
  101. Elashiry, M.; Elashiry, M.M.; Elsayed, R.; Rajendran, M.; Auersvald, C.; Zeitoun, R.; Rashid, M.H.; Ara, R.; Meghil, M.M.; Liu, Y.; et al. Dendritic cell derived exosomes loaded with immunoregulatory cargo reprogram local immune responses and inhibit degenerative bone disease in vivo. J. Extracell. Vesicles 2020, 9, 1795362. [Google Scholar] [CrossRef]
  102. Lv, Q.; Deng, J.; Chen, Y.; Wang, Y.; Liu, B.; Liu, J. Engineered Human Adipose Stem-Cell-Derived Exosomes Loaded with miR-21-5p to Promote Diabetic Cutaneous Wound Healing. Mol. Pharm. 2020, 17, 1723–1733. [Google Scholar] [CrossRef]
  103. Kim, G.; Lee, Y.; Ha, J.; Han, S.; Lee, M. Engineering exosomes for pulmonary delivery of peptides and drugs to inflammatory lung cells by inhalation. J. Control Release 2021, 330, 684–695. [Google Scholar] [CrossRef]
  104. Katakowski, M.; Buller, B.; Zheng, X.; Lu, Y.; Rogers, T.; Osobamiro, O.; Shu, W.; Jiang, F.; Chopp, M. Exosomes from marrow stromal cells expressing miR-146b inhibit glioma growth. Cancer Lett. 2013, 335, 201–204. [Google Scholar] [CrossRef] [Green Version]
  105. Zhou, W.; Xu, M.; Wang, Z.; Yang, M. Engineered exosomes loaded with miR-449a selectively inhibit the growth of homologous non-small cell lung cancer. Cancer Cell Int. 2021, 21, 485. [Google Scholar] [CrossRef]
  106. Haney, M.J.; Klyachko, N.L.; Zhao, Y.; Gupta, R.; Plotnikova, E.G.; He, Z.; Patel, T.; Piroyan, A.; Sokolsky, M.; Kabanov, A.V.; et al. Exosomes as drug delivery vehicles for Parkinson’s disease therapy. J. Control Release 2015, 207, 18–30. [Google Scholar] [CrossRef] [Green Version]
  107. Skotland, T.; Iversen, T.G.; Llorente, A.; Sandvig, K. Biodistribution, pharmacokinetics and excretion studies of intravenously injected nanoparticles and extracellular vesicles: Possibilities and challenges. Adv. Drug Deliv. Rev. 2022, 186, 114326. [Google Scholar] [CrossRef]
  108. Kang, M.; Jordan, V.; Blenkiron, C.; Chamley, L.W. Biodistribution of extracellular vesicles following administration into animals: A systematic review. J. Extracell. Vesicles 2021, 10, e12085. [Google Scholar] [CrossRef]
  109. Shimomura, T.; Seino, R.; Umezaki, K.; Shimoda, A.; Ezoe, T.; Ishiyama, M.; Akiyoshi, K. New Lipophilic Fluorescent Dyes for Labeling Extracellular Vesicles: Characterization and Monitoring of Cellular Uptake. Bioconjug. Chem. 2021, 32, 680–684. [Google Scholar] [CrossRef]
  110. Goh, W.J.; Zou, S.; Ong, W.Y.; Torta, F.; Alexandra, A.F.; Schiffelers, R.M.; Storm, G.; Wang, J.-W.; Czarny, B.; Pastorin, G. Bioinspired Cell-Derived Nanovesicles versus Exosomes as Drug Delivery Systems: A Cost-Effective Alternative. Sci. Rep. 2017, 7, 14322. [Google Scholar] [CrossRef]
  111. Khan, A.A.; de Rosales, R.T.M. Radiolabelling of Extracellular Vesicles for PET and SPECT imaging. Nanotheranostics 2021, 5, 256–274. [Google Scholar] [CrossRef]
  112. Joshi, B.; Ortiz, D.; Zuhorn, I. Converting extracellular vesicles into nanomedicine: Loading and unloading of cargo. Mater. Today Nano 2021, 16, 100148. [Google Scholar] [CrossRef]
  113. An, H.J.; Cho, H.-K.; Song, D.H.; Kee, C. Quantitative analysis of exosomes in the aqueous humor of Korean patients with pseudoexfoliation glaucoma. Sci. Rep. 2022, 12, 12875. [Google Scholar] [CrossRef]
  114. Yang, J.; Zhang, Y.; Gao, X.; Yuan, Y.; Zhao, J.; Zhou, S.; Wang, H.; Wang, L.; Xu, G.; Li, X.; et al. Plasma-Derived Exosomal ALIX as a Novel Biomarker for Diagnosis and Classification of Pancreatic Cancer. Front. Oncol. 2021, 11, 628346. [Google Scholar] [CrossRef]
Figure 1. Types and methods of EV-mediated therapeutics. (a) Inhibition of sEV release of secreting cells or sEV uptake by recipient cells can be utilized to prevent the progression of diverse diseases. (b) sEVs show diverse therapeutic functions depending on the functionality of source cells. (c) sEVs can be engineered before and/or after isolation from the cell media. The most well-known methods are the transfection of engineered vectors to embody desired traits or electroporation to load therapeutic proteins into isolated sEVs.
Figure 1. Types and methods of EV-mediated therapeutics. (a) Inhibition of sEV release of secreting cells or sEV uptake by recipient cells can be utilized to prevent the progression of diverse diseases. (b) sEVs show diverse therapeutic functions depending on the functionality of source cells. (c) sEVs can be engineered before and/or after isolation from the cell media. The most well-known methods are the transfection of engineered vectors to embody desired traits or electroporation to load therapeutic proteins into isolated sEVs.
Pharmaceutics 15 00325 g001
Table 2. Therapeutics using stem cell-derived naïve sEVs.
Table 2. Therapeutics using stem cell-derived naïve sEVs.
Source CellsEV PurificationDiseaseTherapeutic ScheduleResultReference
Mouse bone marrow MSCsUltracentrifugationAcute myocardial infarction50 μg MSC-sEVs in 25 μL PBS, intramyocardial injectionChanged M1 macrophages to M2 by delivery of miR-182[33]
Human liver stem cellsUltracentrifugation, purification by iodixanolChronic kidney disease1 × 1010 particles/mL MSC-sEVs, intravenous injection weekly for 4 weeksAnti-fibrosis and improvement in kidney function[36]
Human bone marrow MSCsFiltration, Total Exosome Isolation ReagentGVHD2 × 10⁶ particles/kg MSC-sEVs in 200 μL saline, intravenous injectionImmunomodulatory effects on T cells by delivering miRNA[38]
Rat urine stem cellsUltracentrifugation, purification by 30% sucrose/D2O cushionDiabetic nephropathy100 μg MSC-sEVs in 200 μL of PBS, intravenous injection weekly for 4 weeksAmeliorated kidney impairment[37]
Canine adipose tissue MSCsUltracentrifugationInflammatory bowel disease100 µg MSC-sEVs from either naïve or primed cASCs, in 200 µL PBS, intraperitoneally injected at days 1, 3, and 5Increased the immune modulatory effect[39]
Human bone marrow MSCsUltracentrifugationTumorFirst, 100 μg MSC-sEVs; followed by 50 μg MSC-sEVs, in a volume of 20 mL of PBS, intravenous injection weekly for 4 weeksInhibited tumor growth[40]
Human bone marrow MSCs100 kDa ultra-filtration, Exo Quick-TCTM KitRetinal ischemia4 μL of 1 × 109 particles/mL MSC-sEVs, intravitreous humor injectionNeuroprotection and regeneration[41]
Human umbilical cord MSCsUltracentrifugationEye subretinal fibrosis2 μL MSC-sEVs, intravitreous tumor injectionAmeliorated subretinal fibrosis by delivering miR-27b[42]
Human umbilical cord blood MSCsUltracentrifugationChoroidal neovascularization1 μg, 2 μg, 3 μg of 50 μg/Ml MSC-sEVs, intravitreal humor injectionAmeliorated RPE cells and retina via downregulation of VEGF-A[43]
Mouse neural stem cellsUltracentrifugationHuntington’s disease10 μL of 5 mg/mL MSC-sEVs, injection into area between the first and second lumbar vertebrae, twice after a 7-day interval eachReduced mutant HTT aggregation in the brain[44]
Wharton’s jellyUltracentrifugation, Exo-Prep kitAlzheimer’s disease50 µg MSC-sEVs, intravenous injection, weekly for 4 weeksDownregulated HDAC4, improved AD pathology[45]
Human bone marrow MSCsCentrifugation, purification by PEG solutionAlzheimer’s disease2 × 109 MSC-sEVs in 5 µL, intranasal injection every 4 days for 4 monthsImproved in cognitive tests[34]
Human umbilical cord blood MSCsExo Quick-TCTM kitAlzheimer’s disease30 μg MSC-sEVs, intravenous injection, every 2 weeks, four timesReduced neuroinflammation and Aβ deposition[46]
Murine bone marrow MSCsUltracentrifugationAlzheimer’s disease5 × 1011 MSC-sEVs, intravenous injection, monthly for 4 monthsLessened plaque deposition, restored inflammatory cytokine levels[47]
Human bone marrow MSCsUltracentrifugationAlzheimer’s disease100 μg MSC-sEVs, intracerebroventricular injection, once every 2 days for 2 weeksReduced iNOS expression, relieved synaptic impairment, and long-term potentiation[48]
Mouse bone marrow-derived MSCsExoQuickAlzheimer’s disease150 μg MSC-sEVs, intravenous injection, biweekly for 4 monthsRecovered learning and memory capabilities and synaptic dysfunction[49]
Mouse bone marrow MSCsUltracentrifugationAlzheimer’s disease22.4 μg MSC-sEVs, single intracerebral injectionAmeliorated Aβ burden and dystrophic neurites[50]
Human umbilical cord MSCsUltracentrifugationAlzheimer’s disease2 mg/mL intracerebroventricular injectionReduced Aβ generation and oxidative stress, prevented microglia activity[51]
Human umbilical cord MSCsCentrifugationAlzheimer’s disease5 × 105 MSC-sEVs, intravenous injection, at weeks 2 and 3Improved neurogenesis and neuroinflammation properties[51]
Table 3. Therapeutics using immune cell-derived naïve sEVs.
Table 3. Therapeutics using immune cell-derived naïve sEVs.
Source CellsEV PurificationDiseaseTherapeutic ScheduleResultReference
Murine bone marrow-derived DCsUltracentrifugationMastocytoma, mammary adenocarcinoma3–5 μg sEVs, single intradermal injectionSuppressed tumor growth, primed tumor-specific cytotoxic T cells[70]
NK-92MI human NK cellsUltracentrifugationMelanoma20 μg sEVs, intertumoral injection for two daysSuppressed tumor growth[64]
Peripheral blood mononuclear cell-derived T cellsUltracentrifugationTriple-negative breast cancer240 μg sEVs, intraperitoneal injection, every 3 days for 27 daysDecreased tumor cell-induced T cell dysfunction[71]
NK cellsUltracentrifugationSK-N-SH neuroblastomaTreated with sEVs, intraperitoneal injection, repeated three times with a 7-day interval until death Increased survival time[65]
NK cellsUltracentrifugationAnti-tumor effect100 µg sEV injection Suppression of tumor growth[72]
CAR-T cellsUltracentrifugationMDA-MB-231, HCC827, SK-BR-325–125 µg sEV injection, every week for 40 daysSuppression of dose-dependent tumor growth [69]
CAR-T cellsUltracentrifugationBreast cancer100–500 μg sEVs, intravenous injection on days 3, 6, 9, 12, and 15Inhibited tumor growth, low toxicity[73]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Lee, I.; Choi, Y.; Shin, D.-U.; Kwon, M.; Kim, S.; Jung, H.; Nam, G.-H.; Kwon, M. Small Extracellular Vesicles as a New Class of Medicines. Pharmaceutics 2023, 15, 325. https://doi.org/10.3390/pharmaceutics15020325

AMA Style

Lee I, Choi Y, Shin D-U, Kwon M, Kim S, Jung H, Nam G-H, Kwon M. Small Extracellular Vesicles as a New Class of Medicines. Pharmaceutics. 2023; 15(2):325. https://doi.org/10.3390/pharmaceutics15020325

Chicago/Turabian Style

Lee, Inkyu, Yoonjeong Choi, Dong-U Shin, Minjeong Kwon, Seohyun Kim, Hanul Jung, Gi-Hoon Nam, and Minsu Kwon. 2023. "Small Extracellular Vesicles as a New Class of Medicines" Pharmaceutics 15, no. 2: 325. https://doi.org/10.3390/pharmaceutics15020325

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop