Skip to main content
MedComm logoLink to MedComm
. 2021 Feb 18;2(1):17–26. doi: 10.1002/mco2.57

Small extracellular vesicles‐based cell‐free strategies for therapy

Yeye Guo 1, Huaishan Wang 1, Lili Huang 1, Lingling Ou 1, Jinjin Zhu 1, Shujing Liu 1, Xiaowei Xu 1,
PMCID: PMC8491241  PMID: 34766134

Abstract

Small extracellular vesicles (sEVs) are extracellular nanovesicles that contain bioactive proteins, lipids, RNA, and DNA. A variety of biological process is regulated with sEVs. sEVs are an intercellular messenger regulating recipient cell function and play a role in disease initiation and progression. sEVs derived from certain cells, such as mesenchymal stem cells and immune cells, have the potential for clinical therapy as they possess the characteristics of their parental cells. With better understanding of sEVs biogenesis, their transportation properties, extended circulatory capability, and exceptional biocompatibility, sEVs emerge as a potential therapeutic tool in the clinic. Here, we summarize applications of sEVs‐based therapies in different diseases and current knowledge about the strategies in bioengineered sEVs, as well as the challenges for their use in clinical settings.

Keywords: cancer, exosome, extracellular vesicle, immunotherapy


Small extracellular vesicles (sEVs) are extracellular nanovesicles that contain bioactive proteins, lipids, RNA, and DNA. A variety of biological process is regulated with sEVs. sEVs are an intercellular messenger regulating recipient cell function and play a role in disease initiation and progression. sEVs derived from certain cells, such as mesenchymal stem cells and immune cells, have the potential for clinical therapy as they possess the characteristics of their parental cells. With better understanding of sEVs biogenesis, their transportation properties, extended circulatory capability, and exceptional biocompatibility, sEVs emerge as a potential therapeutic tool in the clinic. Here, we summarize applications of sEVs‐based therapy in different diseases and current knowledge about the strategies in bioengineered sEVs, as well as the challenges for their use in clinical settings.

graphic file with name MCO2-2-17-g002.jpg

1. INTRODUCTION

Small extracellular vesicles (sEVs) are 30–200 nm lipid bilayer‐enclosed extracellular vesicles. A heterogenous mixture of sEV subsets, including exosomes, is derived when commonly used “exosome” isolation techniques are employed. Almost all cells generate sEVs and deliver to the surrounding biological fluids. 1 , 2 Briefly, the formation of sEVs is initiated from endosomal system. Early endosomes mature into late endosomes or multivesicular bodies (MVB) by inward budding of the endosomal membrane. Constitutively, the intraluminal vesicles (ILVs) within large MVBs are formed during this process that the endosomal membrane invaginates. When fusion with the plasma membrane, most ILVs are released into the extracellular space, and turn to sEVs. 3 , 4 , 5

Small EVs were initially regarded as cellular garbage disposals as a result of cell damage, or by‐products of cell imbalance. Further studies found that those sEVs exhibited biological function. The bioactive cargo contains lots of information from their parental cell, including noncoding RNAs and mRNA, 6 free fatty acids, 7 surface receptors, and proteins 8 inside and on the surface. Therefore, sEVs mediate the short‐range and distant communications between cells. Various target cells can be stimulated by the membrane molecules on sEVs or the contents inside sEVs. 9 During tumor progression, sEVs from tumor cells may help to form a premetastatic niche for tumor metastasis. 10

Over the past decades, many studies demonstrated that sEVs were associated with various diseases, such as inflammatory diseases, 11 diabetes, 12 cardiovascular diseases, 13 central nervous system diseases, 14 tumors, 15 and so on. We recently showed that PD‐L1 on sEVs may be used to predict melanoma patient response to anti‐PD1 therapy. 16

2. ADVANTAGES OF sEVs FOR THERAPY

With better understanding of sEVs, its application in clinical treatment attracts researchers’ attention. Compared to traditional drug delivery system or cellular therapy, sEVs possess some natural advantages such as they are biocompatible and biodegradable, therefore are low cytotoxic and immunogenic. 17 Because of their tiny size, sEVs can be delivered to long‐distance sites and are able to escape from lung clearance and cross many biological barriers such as pass through the blood–brain barrier. 18 Considering the natural transportation feature and long‐term circulatory capability, sEVs are ideal for carrying drugs, proteins, nucleic acids, and other objects for therapy. 19 , 20 , 21 Another important characteristic of sEVs is the high specificity for target cells. It has been identified that sEVs origin could determine cell targeting and the transfer of chemicals toward target cells. 22 In addition, sEVs are simple to produce as most cell types can produce sEVs and they can be stored easily. 23 Taken together, these characteristics enable sEVs to be a promising candidate for therapy.

3. THE CURRENT STATE‐OF‐THE‐ART METHODS IN ISOLATION OF sEVs

Isolation of sEVs is labor intensive and is the bottleneck for using sEVs‐based diagnosis or treatment clinically. Ultracentrifugation is still the most common approach, which sometimes is combined with density gradient centrifugation. This is ideal for isolation of exosomes from large volume media. However, the process takes a long time and requires expensive instruments and good technical skill of researchers. The combination of ultracentrifugation with density gradient centrifugation helps to achieve a higher purity of sEVs. 24 Isolation kits based on size and membrane‐based affinity binding have been launched by some companies. 25 Precipitation of sEVs is another popular way for isolating sEVs especially from body fluids. 26 These approaches provide a choice to isolate sEVs easily in lab. For research requiring downstream analysis, magnetic bead‐based isolation kit is a favorable manner. 27 With these options, investigators can choose one of the methods to isolate sEVs according to their study design.

4. MESENCHYMAL STEM CELL‐DERIVED sEVs IN REGENERATIVE AND ANTI‐INFLAMMATORY THERAPIES

Mesenchymal stem cells (MSCs) are multipotent cells that reside in many adult tissues. It is well‐recognized that MSCs exert an anti‐inflammatory and regenerative function. 28 Accordingly, it has been reported that MSC‐derived sEVs held similar therapeutic effects to MSCs as some immune‐modulating factors from MSCs were highly enriched in sEVs. 29 Therefore, sEVs derived from MSCs are able to treat diseases related to dysfunctional immune reactions, such as autoimmune diseases, graft‐versus‐host disease (GVHD), and inflammatory diseases theoretically. 30 Compared to MSCs themselves, MSC‐sEVs reduce the risk for teratoma formation and embolization, which are major concerns for stem cell‐based therapy. As one of the most promising sEVs in therapy, a standardized protocol for MSC‐sEV preparation was defined by International Society for Extracellular Vesicles (ISEV) due to the heterogeneity of MSCs. 31

MSC‐sEVs therapy reduced the pro‐inflammatory cytokine in patient's PBMCs and improved the clinical GVHD symptoms through interleukin‐10 (IL‐10), transforming growth factor β (TGF‐β), and human leukocyte antigen (HLA‐G). 32 A majority of the literature demonstrated that MSC‐derived sEVs showed protective activity in several diseases such as myocardial infarction, 32 stroke, 32 fibrosis, 33 and ischemia. 34 , 35 For instance, MSC‐sEVs reduced infarct size by pro‐angiogenesis and anti‐inflammation as the mediator of tissue repair in myocardial ischemia/reperfusion injury mouse model. 36 , 37 Intra‐articular administration of MSC‐sEVs improved the repair of osteochondral defects in rat model. 38 By suppressing both the EMT of hepatocytes and collagen production through TGF‐β1/Smad pathway, sEVs derived from MSCs could ameliorate liver fibrosis in CCl4‐induced liver injury model. 39 In addition, MSC‐sEVs showed capability in promoting tissue regeneration. sEVs from MSCs could enhance cutaneous wound healing by inducing epithelial cell proliferation through Wnt/β‐catenin signaling pathway. 40 In addition, MSCs‐sEVs promote the angiogenesis in skin lesions by inducing growth factors through activating AKT/ERK/STAT3 signaling pathways. 41 MSCs‐sEVs also exhibited a therapeutic potential in traumatic brain injury by protecting neurons and promoting axonal growth via SNARE complex. 42 In general, these studies displayed the broad therapeutic effects of MSC‐derived sEVs (Figure 1).

FIGURE 1.

FIGURE 1

Application of MSC‐derived sEVs in regenerative and anti‐inflammatory therapies. MSC‐derived sEVs target various organs and cells such as astrocytes, myocytes, osteocytes, epithelial cells, smooth muscle cells, and hepatocytes

5. sEVs IN CANCER THERAPY

5.1. Cancer immunotherapy by sEVs‐based vaccines

Many evidences suggest that sEVs could be used as novel cancer vaccines to initiate immune system to identify and diminish cancer cells through antigen‐presenting cells (APCs) 43 (Figure 2). Dendritic cells (DCs) are APCs that can be activated upon stimulus. The crosstalk between DCs and T cells triggers an immune response by specific antigens. 44 It has been proven that DCs‐sEVs expressed functional MHC‐I, MHC‐II, and T‐cell costimulatory molecules such as CD86. Tumor peptide‐stimulated DCs‐sEVs initiated cytotoxic T cells (CTLs) in vivo and inhibited tumor growth in a T cell‐dependent approach in a mouse model. 45 Besides antigen presentation, DCs‐sEVs directly triggered natural killer cell proliferation and activation through surface proteins such as NKG2D ligand and IL‐15Rα. 46

FIGURE 2.

FIGURE 2

sEVs‐based vaccines in cancer therapy. sEVs derived from dendritic cells express functional MHC‐I, MHC‐II, and T‐cell costimulatory molecules to present antigen and activate T cells as well as natural killer cells. Cancer cells‐derived sEVs express tumor antigens and can be uptake by APCs, such as dendritic cells, therefore initiating immune response

In many respects, sEVs derived from cancer cells are like APCs because of the expression of the tumor‐associated antigen. Interestingly, cancer cells‐sEVs not only have antigen‐presenting molecules (MHC class‐I, HSP), but are also enriched with various tumor antigens such as HER2/neu, melan‐A, TRP, gp100, 47 CEA, 48 and others. Therefore, the uptake of cancer cells‐sEVs by DCs induced cross‐presentation to CTLs as a source of an antigen or antigens. Immunization of mice with DCs pulsed with cancer cell‐derived sEVs exerted CD8+ T cell‐dependent antitumor effects in mouse models. 49 It is noteworthy that sEVs may be better than other forms of antigen such as whole‐cell lysates or soluble antigen owing to the advantageous delivery in the form of sEVs 50 through molecules such as Mfg‐E8 (lactadherin), 51 integrins, 52 tetraspanins, and others. 53 In B16BL6 melanoma mouse model, investigators established a sEVs‐based tumor antigens‐adjuvant co‐delivery system. B16BL6‐sEVs containing tumor antigens could induce B16BL6‐specific T‐cell response and suppress tumor growth. 54

Peritoneal cavity fluid‐sEVs from cancer patients also showed antitumor effects by inducing DCs to activate T cells in an MHC I‐dependent manner therefore killing cancer cells. Besides, sEVs derived from ascites triggered the increase of IFN‐γ by peripheral blood lymphocytes. 47 , 55

5.2. sEVs cargo loading for cancer therapy

The natural characteristics of sEVs make them an ideal cargo for delivering drugs, miRNAs, siRNAs, and proteins in cancer therapy. Because of the protective structure of sEVs, certain chemicals and drugs can be loaded on sEVs. In that case, the target biochemical could be distributed to a broad range of biological fluids with a longer circulating time and possibly better efficacy. Immature DCs‐sEVs loaded with Doxorubicin (Dox) displayed a high efficiency in targeting breast cancer cells and delivering Dox, thus inhibiting tumor growth without overt toxicity. 56 In a zebrafish model, sEVs significantly increased the absorption and cytotoxicity of doxorubicin and paclitaxel, both of which are anticancer drugs, in brain cancer. 18

The small interfering RNAs (siRNAs) represented a powerful strategy for inhibition of gene expression and were applied to gene therapy. 57 sEVs‐delivered siRNA caused effective posttranscriptional gene silencing in recipient cells and also exhibited a therapeutic potential in various cancers. 58 , 59 Restore of the expression level of tumor‐inhibitory miRNAs in cancer cells are identified to effectively suppress cancer progression. 60 B cells infected with Epstein‐barr virus (EBV) that encodes miRNAs could secrete sEVs containing miRNAs. Then sEVs were transferred to target cells and repressed the target immunoregulatory genes in EBV‐associated lymphomas. 61 sEVs derived from miR‐146a overexpressing HEK293 cells inhibited cell growth in prostate cancer by suppressing target gene. 62

Besides delivery of drugs and nucleic acids, proteins are in great attention as they serve crucial functions in essentially all biological processes. sEVs can not only deliver tumor antigens 63 as discussed above, but also apoptosis‐related proteins such as caspase‐1, 64 peptides, 65 and other cancer‐associated proteins into target cells for cancer therapy. When donor cells harbor a dominant‐negative mutant of Survivin (Survivin‐T34A) that blocks the inhibition of apoptosis, the sEVs increased the apoptotic cell death and enhanced the Gemcitabine sensitivity in pancreatic adenocarcinoma. 66 More recently, sEVs from CAR‐T cells showed promising therapeutic efficacy with low toxicity. 67

5.3. Clinical trials of sEVs‐based therapy

In view of the promising results achieved both in vitro and in vivo, sEVs‐based therapy has been considered to be an encouraging approach for cancer treatment. Quite a few early phase clinical trials have already been performed or are ongoing (Table 1). The first phase I sEVs‐based clinical trial was launched in metastatic melanoma patients through vaccination with autologous DCs‐sEVs and loaded with the MAGE tumor antigens for 4 weeks. The outcome confirmed the feasibility and safety of DC‐derived sEVs‐based vaccination in melanoma patients for the first time. 68 Nearly the same time, another phase I trial of DCs‐derived sEVs immunotherapy was performed on nonsmall cell lung cancer (NSCLC) patients. The vaccination was also well‐tolerated and exhibited an activated immune response in some patients. 69

TABLE 1.

Clinical trials of sEVs‐based therapy

Year (Completed/Updated) Disease Phase sEVs source Status Reference
2005 Metastatic melanoma I DCs Completed 62
2005 NSCLC I DCs Completed 63
2008 Colorectal cancer I Autologous ascites Completed 49
2016 Advanced NSCLC II DCs Completed 65
2018 Malignant glioma I Glioma cells Completed NCT01550523
2019 Head and neck cancer I Grape Ongoing NCT01668849
2019 Colorectal cancer I Plant Ongoing NCT01294072
2020 Metastatic pancreas cancer I Mesenchymal stromal cells Ongoing NCT03608631

Abbreviation: NSCLC, nonsmall cell lung cancer.

Evidence suggested that sEVs isolated from melanoma patients’ ascites delivered Mart‐1 and tumor antigens to DCs for cross‐presentation to CTLs. 47 Thus, ascites‐derived sEVs combined with GM‐CSF treatment were used in a phase I clinical trial for colorectal cancer. The study indicated that ascites‐derived sEVs in combination with GM‐CSF are safe, nontoxic, and could induce antitumor CTLs response. 55 It has been reported that sEVs treated with IFN‐γ induced immune activation and tumor suppression. 70 Based on these, a phase II trial of IFN‐γ‐DC‐derived sEVs was performed on advanced NSCLC patients. The result showed that IFN‐γ‐DC‐derived sEVs boosted NK cell‐mediated antitumor immunity. 71

A recently completed phase I clinical trial was performed in malignant glioma patients. Glioma cells‐sEVs were loaded with an antisense molecule drug and re‐implanted to patients. These sEVs were expected to stimulate the immune system and induce a T cell‐mediated antitumor response. The result has not been published yet and is of great interest (NCT01550523).

Some other clinical trials concerning plant‐origin sEVs in cancer therapy are currently being investigated. One clinical trial is designed to determine the effects of grape‐derived sEVs‐like nanoparticles on reliving oral mucositis and related pain in head and neck cancers patients (NCT01668849). Another phase I clinical trial was using plant‐derived sEVs to deliver curcumin in colorectal cancer. The efficacy of curcumin‐loaded sEVs in the treatment of cancer patients is going to be detected (NCT01294072). Recently, a group in M.D. Anderson Cancer Center found that sEVs carrying siRNA specific to oncogenic KrasG12D, a common mutation in pancreatic cancer, suppressed cancer progression in pancreatic cancer mouse models and significantly increased overall survival. 72 Based on this, they are conducting a phase I clinical trial regarding the sEVs‐based therapy in metastatic pancreas cancer with KrasG12D Mutation (NCT03608631).

All the above studies (both completed and ongoing) suggested the fact that sEVs‐based therapies are safe in a clinical setting, whereas their therapeutic efficacy is still under evaluation.

6. STRATEGIES FOR ENGINEERING sEVs

To better translate sEVs‐based treatments to clinical setting, there are still many obstacles needed to be overcame. Such as how to increase the carrying capacity of drugs, RNAs, and proteins on sEVs and how to improve the specificity of sEVs to target cells. Therefore, various strategies were designed to bioengineer sEVs.

6.1. Origins of sEVs and factors promoting the production

The origin of sEVs determines its natural characteristics. Immature DCs‐derived sEVs are low immunogenic because of the lack of surface markers such as MHC‐I, MHC‐II, CD40, and CD86. sEVs from MSCs are able to mediate the immunosuppressive effect. 73 CD8+ T lymphocytes‐derived sEVs express cytotoxic molecules such as granzyme B and perforin, therefore harboring the cytotoxic potential. 67 HEK293T cells and BJ cells are easy to expand and stable for transfection. Therefore, they are capable of large‐scale production of sEVs. Besides cells, agricultural products such as plants, fruits, and milk can be used as other sources for isolating sEVs that are easy to produce and safe to use by not activating host immune response. 23 , 74

Several factors are found to improve the production of sEVs, such as increased intracellular calcium level 75 ; external stress including thermal stress, 76 anoxia, 77 radiation, 78 and lower pH 78 ; cytoskeletal blocking 79 ; drugs such as sitafloxacin, forskolin, fenoterol, nitrefazole, and pentetrazol 80 ; and expression of certain genes, for instance, nSMase2, 81 CD9, 82 and HIF‐1α. 83 It is noteworthy that immortalized cell line is more stable and convenient than primary cells in preparing sEVs. A myc‐mediated immortalization of MSCs was developed to produce sEVs and exhibited therapeutic effect in cardiovascular diseases. 84 Immortalized adipose‐derived Mesenchymal stem cell line (ATCC® SCRC‐4000) has currently been used and proposed as one of the strategies for scale up and mass production of sEVs in a reproducible manner.

6.2. Loading nucleic acids into sEVs

Electroporation is the most common way to load RNAs into sEVs. TRPP2 siRNA was loaded into 293T‐derived sEVs by electroporation, and then the siRNA loaded sEVs‐targeted FaDu cells, a cell line of human pharyngeal squamous cell carcinoma, leading to the inhibition of the EMT. 85

Some transfection reagents have also been used in RNA loading. The purified sEVs could be incubated with siRNA mixed with lipofectamine at room temperature for 30 min, followed by three to five times wash and ultrafiltration. When culturing the recipient cells with engineered sEVs, the uptake of sEVs enabled exogenous siRNA transferring to recipient cells. 58

There are other novel ways to actively packaging nucleic acids into sEVs. One is binding specific RNA sequences to proteins and the RNA packaging device in sEVs producing cells could package specific mRNAs into sEVs. By using this system, the therapeutic mRNA that significantly reduces the neuroinflammation and neurotoxicity could be successfully delivered to the brain in the Parkinson's disease mouse models. 86 The conserved sequence of sEVs‐enriched RNAs (eRNAs) carries a certain sequence that targets RNAs into sEVs as a cis‐element. Therefore, the use of eRNAs is another approach to deliver RNA into sEVs. 87 A couple of proteins such as MVP, GW182, AGO2, and Myoferlin were reported to be associated with the delivery of nucleic acid sequences into the sEVs. However, the specific ability for packaging remains to be explored in the future. 88

6.3. Loading proteins into sEVs

One practical method is to overexpress the proteins of interest in donor cells by transfecting the vectors containing the specific gene. Then donor cells produce the proteins encoded by the inserted genes and during the natural packaging processes, the proteins are going to be secreted into sEVs. 89 However, the potential cytotoxicity to donor cells, the complicated interactions in cells, and the low loading efficiency are the main concerns to make that. Hence, several efforts have been made to promote the loading efficiency of target proteins into sEVs.

The fusion of target proteins with the constitutive protein in sEVs and the specific modification of target proteins are valuable ways to engineer sEVs. A group of proteins that are constitutively expressed on sEVs, such as CD9, CD63, and TSG101, 90 can enhance the specificity of target proteins. Other confirmed proteins including HIV‐1 Nef (mut), 91 VSVG, 92 lactadherin C1C2 domain, 93 LAMP2B, 94 and PDGFR TM domain 95 are demonstrated to package certain proteins into or on the surface of sEVs.

It has been demonstrated that specific modifications of target protein contribute to protein transport. Ubiquitination of target proteins at the C‐terminus led to a 10‐fold increase of expression in sEVs with biological function. 96 In addition, myristoylation or palmitoylation tag or a transmembrane motif fusion on the N‐terminus of target proteins enabled the fusion proteins to target the plasma membrane. 97 A major concern of the modifications on proteins is the potential alteration of protein function, especially the ubiquitination modification that may result in the degradation of the target protein by the proteasome.

Several mechanical methods were tried to load proteins to sEVs: sonication, mechanical extrusion, saponin permeabilization, incubation at room temperature, and repeated freeze‐thaw. 19 Among these, the first three ways displayed high efficiency in vitro. When using mechanical methods, it requires high sEVs purification and careful maintenance of the integrity of sEVs and biological activity.

7. CHALLENGES FOR DEVELOPING sEVs‐BASED THERAPY

Small EVs are an exceptionally qualified biological vehicle for clinical therapies. However, there are still several challenges and obstacles needed to be overcome. The first problem to be solved is how to generate a sufficient number of sEVs. Therefore, developing a scalable and reproducible protocol is of great importance. In addition, the criteria for purification and storage of sEVs, as well as the quality analysis, are necessary to attain compliance with good manufacturing practice guidelines. 98 sEVs from nucleated cells present some level of risk for horizontal gene transfer because it is unpredictable that which cells already harbor dangerous DNA. Bioengineering of red blood cells that do not have nuclei is a potential safer alternative. There are several parameters require to be considered when treating a patient with therapeutic sEVs: dose of sEVs, dosage regimen, and way to administration. These parameters are hard to define as previous studies were mainly performed by empiric therapy without a standard. 99 In that case, an appropriate way to assess the biological effect caused by sEVs in vitro and in vivo is of the essence. Besides these, optimizing approaches to enhance the sEVs loading efficiency determine the therapeutic activity. Meanwhile, further work still remains to be accomplished to elucidate the exact mechanisms of the interaction between sEVs and targeted cells.

8. CONCLUSION AND PERSPECTIVE

Small EVs have gained increasing attention in understanding disease etiology. The robust biocompatibility enables sEVs to be optimum vehicles for delivering chemicals and other therapeutic molecules. Meanwhile, some natural characteristics of sEVs obtained from parental cells make it a promising novel approach for treating a diverse variety of diseases. Because almost all the cells produce sEVs, the donor cells could be selectively chosen based on the clinical needs.

Even though sEVs‐based therapy showed an extensive application prospect, further studies are still needed for in vivo studies and clinical trials. Besides the challenges we discussed above, it is also urgent to investigate the exact components from sEVs that may facilitate the progress of diseases. In the future, more engineered methods are needed to remove the disease‐supporting content from sEVs and enhance the cargo loading efficiency of sEVs. Nanosized sEVs may have immense applications in future clinical therapies.

AUTHOR CONTRIBUTIONS

Yeye Guo wrote the initial draft of the manuscript. Huaishan Wang, Lili Huang, Lingling Ou, Jinjin Zhu, and Shujing Liu provided the resources for this work. Xiaowei Xu conceptualized the study, edited the manuscript, and provided the funding for this work.

CONFLICT OF INTEREST

The authors declare no conflict of interest.

ETHICS APPROVAL

Not applicable.

ACKNOWLEDGMENT

This work was supported by National Institutes of Health (CA170340, CA114046, CA210944 and CA224070).

Guo Y, Wang H, Huang L, et al. Small extracellular vesicles‐based cell‐free strategies for therapy. MedComm. 2021;2:17–26. 10.1002/mco2.57

DATA AVAILABILITY STATEMENT

Not applicable.

REFERENCES

  • 1. Li M, Zeringer E, Barta T, Schageman J, Cheng A, Vlassov AV. Analysis of the RNA content of the exosomes derived from blood serum and urine and its potential as biomarkers. Philos Trans R Soc Lond B Biol Sci. 2014;369(1652):20130502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Théry C, Witwer KW, Aikawa E, 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(1):1535750. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Raposo G, Stoorvogel W. Extracellular vesicles: exosomes, microvesicles, and friends. J Cell Biol. 2013;200(4):373‐383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Hessvik NP, Llorente A. Current knowledge on exosome biogenesis and release. Cell Mol Life Sci. 2018;75(2):193‐208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Zhang Y, Liu Y, Liu H, Tang WH. Exosomes: biogenesis, biologic function and clinical potential. Cell Biosci. 2019;9:19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Valadi H, Ekström K, Bossios A, Sjöstrand M, Lee JJ, Lötvall JO. Exosome‐mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol. 2007;9(6):654‐659. [DOI] [PubMed] [Google Scholar]
  • 7. Vidal M, Sainte‐Marie J, Philippot JR, Bienvenue A. Asymmetric distribution of phospholipids in the membrane of vesicles released during in vitro maturation of guinea pig reticulocytes: evidence precluding a role for “aminophospholipid translocase. J Cell Physiol. 1989;140(3):455‐462. [DOI] [PubMed] [Google Scholar]
  • 8. Simpson RJ, Lim JW, Moritz RL, Mathivanan S. Exosomes: proteomic insights and diagnostic potential. Expert Rev Proteomics. 2009;6(3):267‐283. [DOI] [PubMed] [Google Scholar]
  • 9. Camussi G, Deregibus MC, Bruno S, Grange C, Fonsato V, Tetta C. Exosome/microvesicle‐mediated epigenetic reprogramming of cells. Am J Cancer Res. 2011;1(1):98‐110. [PMC free article] [PubMed] [Google Scholar]
  • 10. Chalmin F, Ladoire S, Mignot G, et al. Membrane‐associated Hsp72 from tumor‐derived exosomes mediates STAT3‐dependent immunosuppressive function of mouse and human myeloid‐derived suppressor cells. J Clin Invest. 2010;120(2):457‐471. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Chan BD, Wong WY, Lee MM, et al. Exosomes in inflammation and inflammatory disease. Proteomics. 2019;19(8):e1800149. [DOI] [PubMed] [Google Scholar]
  • 12. Castaño C, Novials A, Párrizas M. Exosomes and diabetes. Diabetes Metab Res Rev. 2019;35(3):e3107. [DOI] [PubMed] [Google Scholar]
  • 13. Bellin G, Gardin C, Ferroni L, et al. Exosome in cardiovascular diseases: a complex world full of hope. Cells. 2019;8(2):166. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Liu W, Bai X, Zhang A, Huang J, Xu S, Zhang J. Role of exosomes in central nervous system diseases. Front Mol Neurosci. 2019;12:240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Seo N, Akiyoshi K, Shiku H. Exosome‐mediated regulation of tumor immunology. Cancer Sci. 2018;109(10):2998‐3004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Chen G, Huang AC, Zhang W, et al. Exosomal PD‐L1 contributes to immunosuppression and is associated with anti‐PD‐1 response. Nature. 2018;560(7718):382‐386. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Ha D, Yang N, Nadithe V. Exosomes as therapeutic drug carriers and delivery vehicles across biological membranes: current perspectives and future challenges. Acta Pharm Sin B. 2016;6(4):287‐296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Yang T, Martin P, Fogarty B, et al. Exosome delivered anticancer drugs across the blood‐brain barrier for brain cancer therapy in Danio rerio. Pharm Res. 2015;32(6):2003‐2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Haney MJ, Klyachko NL, Zhao Y, et al. Exosomes as drug delivery vehicles for Parkinson's disease therapy. J Control Release. 2015;207:18‐30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Alvarez‐Erviti L, Seow Y, Yin H, Betts C, Lakhal S, Wood MJ. Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes. Nat Biotechnol. 2011;29(4):341‐345. [DOI] [PubMed] [Google Scholar]
  • 21. Yim N, Ryu SW, Choi K, et al. Exosome engineering for efficient intracellular delivery of soluble proteins using optically reversible protein‐protein interaction module. Nat Commun. 2016;7:12277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Sancho‐Albero M, Navascués N, Mendoza G, et al. Exosome origin determines cell targeting and the transfer of therapeutic nanoparticles towards target cells. J Nanobiotechnol. 2019;17(1):16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Munagala R, Aqil F, Jeyabalan J, Gupta RC. Bovine milk‐derived exosomes for drug delivery. Cancer Lett. 2016;371(1):48‐61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Théry C, Amigorena S, Raposo G, Clayton A. Isolation and characterization of exosomes from cell culture supernatants and biological fluids. Curr Protoc Cell Biol. 2006;30(1):3.22.1‐3.22.29. [DOI] [PubMed] [Google Scholar]
  • 25. Zeringer E, Barta T, Li M, Vlassov AV. Strategies for isolation of exosomes. Cold Spring Harb Protoc. 2015;2015(4):319‐323. [DOI] [PubMed] [Google Scholar]
  • 26. Peng Q, Zhang J, Zhou G. Comparison of plasma exosomes by differential ultracentrifugation and solvent precipitation methods. Clin Lab. 2018;64(6):991‐998. [DOI] [PubMed] [Google Scholar]
  • 27. Oksvold MP, Neurauter A, Pedersen KW. Magnetic bead‐based isolation of exosomes. Methods Mol Biol. 2015;1218:465‐481. [DOI] [PubMed] [Google Scholar]
  • 28. Chamberlain G, Fox J, Ashton B, Middleton J. Concise review: mesenchymal stem cells: their phenotype, differentiation capacity, immunological features, and potential for homing. Stem Cells. 2007;25(11):2739‐2749. [DOI] [PubMed] [Google Scholar]
  • 29. Yamashita T, Takahashi Y, Takakura Y. Possibility of exosome‐based therapeutics and challenges in production of exosomes eligible for therapeutic application. Biol Pharm Bull. 2018;41(6):835‐842. [DOI] [PubMed] [Google Scholar]
  • 30. Lai P, Weng J, Guo L, Chen X, Du X. Novel insights into MSC‐EVs therapy for immune diseases. Biomark Res. 2019;7:6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Witwer KW, Van Balkom BWM, Bruno S, et al. Defining mesenchymal stromal cell (MSC)‐derived small extracellular vesicles for therapeutic applications. J Extracell Vesicles. 2019;8(1):1609206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Kordelas L, Rebmann V, Ludwig AK, et al. MSC‐derived exosomes: a novel tool to treat therapy‐refractory graft‐versus‐host disease. Leukemia. 2014;28(4):970‐973. [DOI] [PubMed] [Google Scholar]
  • 33. Li T, Yan Y, Wang B, et al. Exosomes derived from human umbilical cord mesenchymal stem cells alleviate liver fibrosis. Stem Cells Dev. 2013;22(6):845‐854. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Zhang Y, Chopp M, Meng Y, et al. Effect of exosomes derived from multipluripotent mesenchymal stromal cells on functional recovery and neurovascular plasticity in rats after traumatic brain injury. J Neurosurg. 2015;122(4):856‐867. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Hu GW, Li Q, Niu X, et al. Exosomes secreted by human‐induced pluripotent stem cell‐derived mesenchymal stem cells attenuate limb ischemia by promoting angiogenesis in mice. Stem Cell Res Ther. 2015;6(1):10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Lai RC, Arslan F, Lee MM, et al. Exosome secreted by MSC reduces myocardial ischemia/reperfusion injury. Stem Cell Res. 2010;4(3):214‐222. [DOI] [PubMed] [Google Scholar]
  • 37. Teng X, Chen L, Chen W, Yang J, Yang Z, Shen Z. Mesenchymal stem cell‐derived exosomes improve the microenvironment of infarcted myocardium contributing to angiogenesis and anti‐inflammation. Cell Physiol Biochem. 2015;37(6):2415‐2424. [DOI] [PubMed] [Google Scholar]
  • 38. Zhang S, Chu WC, Lai RC, Lim SK, Hui JH, Toh WS. Exosomes derived from human embryonic mesenchymal stem cells promote osteochondral regeneration. Osteoarthritis Cartilage. 2016;24(12):2135‐2140. [DOI] [PubMed] [Google Scholar]
  • 39. Lou G, Chen Z, Zheng M, Liu Y. Mesenchymal stem cell‐derived exosomes as a new therapeutic strategy for liver diseases. Exp Mol Med. 2017;49(6):e346. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Zhang B, Wang M, Gong A, et al. HucMSC‐exosome mediated‐Wnt4 signaling is required for cutaneous wound healing. Stem Cells. 2015;33(7):2158‐2168. [DOI] [PubMed] [Google Scholar]
  • 41. Shabbir A, Cox A, Rodriguez‐Menocal L, Salgado M, Van Badiavas E. Mesenchymal stem cell exosomes induce proliferation and migration of normal and chronic wound fibroblasts, and enhance angiogenesis in vitro. Stem Cells Dev. 2015;24(14):1635‐1647. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Zhang Y, Chopp M, Liu XS, et al. Exosomes derived from mesenchymal stromal cells promote axonal growth of cortical neurons. Mol Neurobiol. 2017;54(4):2659‐2673. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Tan A, De La Peña H, Seifalian AM. The application of exosomes as a nanoscale cancer vaccine. Int J Nanomedicine. 2010;5:889‐900. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Banchereau J, Briere F, Caux C, et al. Immunobiology of dendritic cells. Annu Rev Immunol. 2000;18:767‐811. [DOI] [PubMed] [Google Scholar]
  • 45. Zitvogel L, Regnault A, Lozier A, et al. Eradication of established murine tumors using a novel cell‐free vaccine: dendritic cell‐derived exosomes. Nat Med. 1998;4(5):594‐600. [DOI] [PubMed] [Google Scholar]
  • 46. Viaud S, Terme M, Flament C, et al. Dendritic cell‐derived exosomes promote natural killer cell activation and proliferation: a role for NKG2D ligands and IL‐15Ralpha. PLoS One. 2009;4(3):e4942. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Andre F, Schartz NE, Movassagh M, et al. Malignant effusions and immunogenic tumour‐derived exosomes. Lancet. 2002;360(9329):295‐305. [DOI] [PubMed] [Google Scholar]
  • 48. Dai S, Wan T, Wang B, et al. More efficient induction of HLA‐A*0201‐restricted and carcinoembryonic antigen (CEA)‐specific CTL response by immunization with exosomes prepared from heat‐stressed CEA‐positive tumor cells. Clin Cancer Res. 2005;11(20):7554‐7563. [DOI] [PubMed] [Google Scholar]
  • 49. Wolfers J, Lozier A, Raposo G, et al. Tumor‐derived exosomes are a source of shared tumor rejection antigens for CTL cross‐priming. Nat Med. 2001;7(3):297‐303. [DOI] [PubMed] [Google Scholar]
  • 50. Zeelenberg IS, Ostrowski M, Krumeich S, et al. Targeting tumor antigens to secreted membrane vesicles in vivo induces efficient antitumor immune responses. Cancer Res. 2008;68(4):1228‐1235. [DOI] [PubMed] [Google Scholar]
  • 51. Véron P, Segura E, Sugano G, Amigorena S, Théry C. Accumulation of MFG‐E8/lactadherin on exosomes from immature dendritic cells. Blood Cells Mol Dis. 2005;35(2):81‐88. [DOI] [PubMed] [Google Scholar]
  • 52. Clayton A, Turkes A, Dewitt S, Steadman R, Mason MD, Hallett MB. Adhesion and signaling by B cell‐derived exosomes: the role of integrins. FASEB J. 2004;18(9):977‐979. [DOI] [PubMed] [Google Scholar]
  • 53. Morelli AE, Larregina AT, Shufesky WJ, et al. Endocytosis, intracellular sorting, and processing of exosomes by dendritic cells. Blood. 2004;104(10):3257‐3266. [DOI] [PubMed] [Google Scholar]
  • 54. Morishita M, Takahashi Y, Matsumoto A, Nishikawa M, Takakura Y. Exosome‐based tumor antigens‐adjuvant co‐delivery utilizing genetically engineered tumor cell‐derived exosomes with immunostimulatory CpG DNA. Biomaterials. 2016;111:55‐65. [DOI] [PubMed] [Google Scholar]
  • 55. Dai S, Wei D, Wu Z, et al. Phase I clinical trial of autologous ascites‐derived exosomes combined with GM‐CSF for colorectal cancer. Mol Ther. 2008;16(4):782‐790. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Tian Y, Li S, Song J, et al. A doxorubicin delivery platform using engineered natural membrane vesicle exosomes for targeted tumor therapy. Biomaterials. 2014;35(7):2383‐2390. [DOI] [PubMed] [Google Scholar]
  • 57. Urban‐Klein B, Werth S, Abuharbeid S, Czubayko F, Aigner A. RNAi‐mediated gene‐targeting through systemic application of polyethylenimine (PEI)‐complexed siRNA in vivo. Gene Ther. 2005;12(5):461‐466. [DOI] [PubMed] [Google Scholar]
  • 58. Shtam TA, Kovalev RA, Varfolomeeva EY, Makarov EM, Kil YV, Filatov MV. Exosomes are natural carriers of exogenous siRNA to human cells in vitro. Cell Commun Signal. 2013;11:88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Pan Q, Ramakrishnaiah V, Henry S, et al. Hepatic cell‐to‐cell transmission of small silencing RNA can extend the therapeutic reach of RNA interference (RNAi). Gut. 2012;61(9):1330‐1339. [DOI] [PubMed] [Google Scholar]
  • 60. Kosaka N, Takeshita F, Yoshioka Y, et al. Exosomal tumor‐suppressive microRNAs as novel cancer therapy: “exocure” is another choice for cancer treatment. Adv Drug Deliv Rev. 2013;65(3):376‐382. [DOI] [PubMed] [Google Scholar]
  • 61. Pegtel DM, Cosmopoulos K, Thorley‐Lawson DA, et al. Functional delivery of viral miRNAs via exosomes. Proc Natl Acad Sci USA. 2010;107(14):6328‐6333. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Kosaka N, Iguchi H, Yoshioka Y, Takeshita F, Matsuki Y, Ochiya T. Secretory mechanisms and intercellular transfer of microRNAs in living cells. J Biol Chem. 2010;285(23):17442‐17452. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Cho JA, Yeo DJ, Son HY, et al. Exosomes: a new delivery system for tumor antigens in cancer immunotherapy. Int J Cancer. 2005;114(4):613‐622. [DOI] [PubMed] [Google Scholar]
  • 64. Hall J, Prabhakar S, Balaj L, Lai CP, Cerione RA, Breakefield XO. Delivery of therapeutic proteins via extracellular vesicles: review and potential treatments for parkinson's disease, glioma, and schwannoma. Cell Mol Neurobiol. 2016;36(3):417‐427. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Hung ME, Leonard JN. Stabilization of exosome‐targeting peptides via engineered glycosylation. J Biol Chem. 2015;290(13):8166‐8172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Aspe JR, Diaz Osterman CJ, Jutzy JM, Deshields S, Whang S, Wall NR. Enhancement of Gemcitabine sensitivity in pancreatic adenocarcinoma by novel exosome‐mediated delivery of the Survivin‐T34A mutant. J Extracell Vesicles. 2014;3:23244. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Fu W, Lei C, Liu S, et al. CAR exosomes derived from effector CAR‐T cells have potent antitumour effects and low toxicity. Nat Commun. 2019;10(1):4355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Escudier B, Dorval T, Chaput N, et al. Vaccination of metastatic melanoma patients with autologous dendritic cell (DC) derived‐exosomes: results of thefirst phase I clinical trial. J Transl Med. 2005;3(1):10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Morse MA, Garst J, Osada T, et al. A phase I study of dexosome immunotherapy in patients with advanced non‐small cell lung cancer. J Transl Med. 2005;3(1):9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Viaud S, Ploix S, Lapierre V, et al. Updated technology to produce highly immunogenic dendritic cell‐derived exosomes of clinical grade: a critical role of interferon‐γ. J Immunother. 2011;34(1):65‐75. [DOI] [PubMed] [Google Scholar]
  • 71. Besse B, Charrier M, Lapierre V, et al. Dendritic cell‐derived exosomes as maintenance immunotherapy after first line chemotherapy in NSCLC. Oncoimmunology. 2015;5(4):e1071008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Kamerkar S, LeBleu VS, Sugimoto H, et al. Exosomes facilitate therapeutic targeting of oncogenic KRAS in pancreatic cancer. Nature. 2017;546(7659):498‐503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Yeo RW, Lai RC, Zhang B, et al. Mesenchymal stem cell: an efficient mass producer of exosomes for drug delivery. Adv Drug Deliv Rev. 2013;65(3):336‐341. [DOI] [PubMed] [Google Scholar]
  • 74. Ju S, Mu J, Dokland T, et al. Grape exosome‐like nanoparticles induce intestinal stem cells and protect mice from DSS‐induced colitis. Mol Ther. 2013;21(7):1345‐1357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Savina A, Furlán M, Vidal M, Colombo MI. Exosome release is regulated by a calcium‐dependent mechanism in K562 cells. J Biol Chem. 2003;278(22):20083‐20090. [DOI] [PubMed] [Google Scholar]
  • 76. Hedlund M, Nagaeva O, Kargl D, Baranov V, Mincheva‐Nilsson L. Thermal‐ and oxidative stress causes enhanced release of NKG2D ligand‐bearing immunosuppressive exosomes in leukemia/lymphoma T and B cells. PLoS One. 2011;6(2):e16899. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. King HW, Michael MZ, Gleadle JM. Hypoxic enhancement of exosome release by breast cancer cells. BMC Cancer. 2012;12:421. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Wysoczynski M, Ratajczak MZ. Lung cancer secreted microvesicles: underappreciated modulators of microenvironment in expanding tumors. Int J Cancer. 2009;125(7):1595‐1603. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Zaborowski MP, Balaj L, Breakefield XO, Lai CP. Extracellular vesicles: composition, biological relevance, and methods of study. Bioscience. 2015;65(8):783‐797. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Datta A, Kim H, McGee L, et al. High‐throughput screening identified selective inhibitors of exosome biogenesis and secretion: a drug repurposing strategy for advanced cancer. Sci Rep. 2018;8(1):8161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Zhu J, Lu K, Zhang N, et al. Myocardial reparative functions of exosomes from mesenchymal stem cells are enhanced by hypoxia treatment of the cells via transferring microRNA‐210 in an nSMase2‐dependent way. Artif Cells Nanomed Biotechnol. 2018;46(8):1659‐1670. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Böker KO, Lemus‐Diaz N, Rinaldi Ferreira R, Schiller L, Schneider S, Gruber J. The impact of the CD9 tetraspanin on lentivirus infectivity and exosome secretion. Mol Ther. 2018;26(2):634‐647. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Gonzalez‐King H, García NA, Ontoria‐Oviedo I, Ciria M, Montero JA, Sepúlveda P. Hypoxia inducible factor‐1α potentiates jagged 1‐mediated angiogenesis by mesenchymal stem cell‐derived exosomes. Stem Cells. 2017;35(7):1747‐1759. [DOI] [PubMed] [Google Scholar]
  • 84. Chen TS, Arslan F, Yin Y, et al. Enabling a robust scalable manufacturing process for therapeutic exosomes through oncogenic immortalization of human ESC‐derived MSCs. J Transl Med. 2011;9:47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Wang C, Chen L, Huang Y, et al. Exosome‐delivered TRPP2 siRNA inhibits the epithelial‐mesenchymal transition of FaDu cells. Oncol Lett. 2019;17(2):1953‐1961. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Kojima R, Bojar D, Rizzi G, et al. Designer exosomes produced by implanted cells intracerebrally deliver therapeutic cargo for Parkinson's disease treatment. Nat Commun. 2018;9(1):1305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Batagov AO, Kuznetsov VA, Kurochkin IV. Identification of nucleotide patterns enriched in secreted RNAs as putative cis‐acting elements targeting them to exosome nano‐vesicles. BMC Genomics. 2011;12(Suppl 3):S18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Liu C, Su C. Design strategies and application progress of therapeutic exosomes. Theranostics. 2019;9(4):1015‐1028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Rivoltini L, Chiodoni C, Squarcina P, et al. TNF‐related apoptosis‐inducing ligand (TRAIL)‐armed exosomes deliver proapoptotic signals to tumor site. Clin Cancer Res. 2016;22(14):3499‐3512. [DOI] [PubMed] [Google Scholar]
  • 90. Simpson RJ, Jensen SS, Lim JW. Proteomic profiling of exosomes: current perspectives. Proteomics. 2008;8(19):4083‐4099. [DOI] [PubMed] [Google Scholar]
  • 91. Manfredi F, Di Bonito P, Arenaccio C, Anticoli S, Federico M. Incorporation of heterologous proteins in engineered exosomes. Methods Mol Biol. 2016;1448:249‐260. [DOI] [PubMed] [Google Scholar]
  • 92. Meyer C, Losacco J, Stickney Z, Li L, Marriott G, Lu B. Pseudotyping exosomes for enhanced protein delivery in mammalian cells. Int J Nanomedicine. 2017;12:3153‐3170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Hartman ZC, Wei J, Glass OK, et al. Increasing vaccine potency through exosome antigen targeting. Vaccine. 2011;29(50):9361‐9367. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Mentkowski KI, Lang JK. Exosomes engineered to express a cardiomyocyte binding peptide demonstrate improved cardiac retention in vivo. Sci Rep. 2019;9(1):10041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. Ohno S, Takanashi M, Sudo K, et al. Systemically injected exosomes targeted to EGFR deliver antitumor microRNA to breast cancer cells. Mol Ther. 2013;21(1):185‐191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. Cheng Y, Schorey JS. Targeting soluble proteins to exosomes using a ubiquitin tag. Biotechnol Bioeng. 2016;113(6):1315‐1324. [DOI] [PubMed] [Google Scholar]
  • 97. Shen B, Wu N, Yang JM, Gould SJ. Protein targeting to exosomes/microvesicles by plasma membrane anchors. J Biol Chem. 2011;286(16):14383‐14395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Gimona M, Pachler K, Laner‐Plamberger S, Schallmoser K, Rohde E. Manufacturing of human extracellular vesicle‐based therapeutics for clinical use. Int J Mol Sci. 2017;18(6):1190. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Lener T, Gimona M, Aigner L, et al. Applying extracellular vesicles based therapeutics in clinical trials ‐ an ISEV position paper. J Extracell Vesicles. 2015;4:30087. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

Not applicable.


Articles from MedComm are provided here courtesy of Wiley

RESOURCES