1 Introduction

To maintain organisms’ homeostasis, intercellular communication is the key event to control multiple biological processes including mediator secretion, cellular proliferation, differentiation, and apoptosis. Cells located remotely communicate each other via soluble factors including neurotransmitters, hormones, cytokines/chemokines, lipid mediators, and extracellular vesicles (EVs) [1,2,3]. EVs, cell-secreted natural nanoparticles, are classified into three subtypes including exosomes, microvesicles, and apoptotic bodies, which exhibit different biological characteristics in terms of biogenesis, content, morphology and size (exosomes: 30 ~ 200 nm, microvesicles: 100 ~ 1,000 nm, and apoptotic bodies: 1 ~ 5 μm) [3, 4]. Exosomes are single-membrane lipid bilayer vesicles generated either by vesicle budding into endosomes that mature into multivesicular bodies or by direct vesicle budding from the plasma membrane [5]. Exosomes are secreted by all living cell types and have been found in various body fluids such as plasma, urine, saliva, semen, and breast milk [6,7,8,9,10,11]. Microvesicles (or ectosomes) are formed by direct outward budding of the plasma membrane with size typically ranging from 100 to 1000 nm [12]. Apoptotic bodies are relatively larger lipid vesicles released by dying cells which contain fragments of apoptotic cells such as micronuclei, chromatin remnants, and intact organelles [13]. EVs have traditionally been defined and sorted based on their different densities and sizes which enables separation by various methods such as differential centrifugation, filtration, and size exclusion chromatography [14]. It should be noted, however, that due to the overlap** size and density between EVs such as exosomes and microvesicles, current EV isolation techniques have limitation regarding precise purification without completely excluding other groups of EVs. Within the EVs, microvesicles and exosomes are considered as delivery vehicles of diverse biological molecules for intercellular communication including delivery of nucleic acid (e.g. DNAs, RNAs), proteins, lipids, and carbohydrates. Of note, in vivo circulating exosomes isolated from body fluids (e.g. urine and blood) carry biological materials (e.g. proteins and nucleic acid) and represent current physiological conditions, which suggests the diagnostic value of exosomes as novel biomarkers for multiple pathophysiological conditions including cancer [15, 16].

Due to their biological and functional characteristics, the therapeutic potential of exosomes is also being investigated as either natural or engineered form for different therapeutic purposes, including drug delivery tools, biological targeting agents, and vaccination [17,18,19,20,23,24,25].

Here, we will review the current knowledge of biodistribution and PK of systemically administered exosomes, and various active targeting strategies to improve target specificity with better clinical outcome.

2 Biodistribution and pharmacokinetics (PK) of in vivo administered exosomes

Numerous biocompatible and nontoxic nanoparticles have been employed as drug delivery systems including liposomes, polymeric nanoparticles, and exosomes. The biodistribution and PK profile of nanoparticles represent the in vivo behavior of administered nanoparticles and determining these two parameters are the key for successful nanoparticles-mediated novel therapeutics development. Whereas this review paper is mainly focused on exosomes, readers can find recent advances regarding other EVs, such as microvesicles, on review papers cited here [12, 26].

Focusing on exosome therapeutics, the major tissues distribution of systemically administered exosomes generally include liver, spleen, kidney, lung and gastrointestinal tract, which can be altered by various factors such as cellular origin of exosomes, exosomal membrane composition (e.g., protein, lipid, and glycan) and pathophysiological condition of host [27,28,29,30,31,32,33,34,35]. Exosome engineering for targeted delivery of therapeutic exosomes to various tissues including brain, placenta, heart, spinal cord, and cartilages is also being investigated [36,37,38,39,40,41]. Once the exosomes are administered systemically, they show rapid clearance from the blood with less than a few minutes of half-life in the circulation of healthy animals, which is primarily due to the rapid clearance by circulating phagocytic cells including macrophages and neutrophils [29, 32, 42, 43]. In contrast with the blood PK, exosomes display prolonged retention in the tissues such as the liver and spleen, showing sustained retention longer than 24 h [27, 29, 32]. Nonetheless, careful interpretation is needed for analyzing the tissue PK of exosomes, since most exosome imaging techniques utilize methods to label the lipid bilayer of exosomes with various imaging dyes which may lead to tracking of the cell-ingested phospholipids and not the exosome itself.

3 Factors modulating biodistribution and PK of in vivo administered exosomes

Recent studies begin to identify molecules displayed on the exosomal membrane which determine their cellular or organ tropism (Fig. 1) [44, 45]. If the molecular mechanisms generating target cell tropism of exosomes is fully decoded, the potential of exosomes as a therapeutic vehicle would be greatly expanded, especially to the most challenging disease areas including the central nervous system (CNS) related diseases [46]. In this part, various factors that determine biodistribution and targetability of exosomes are discussed.

Fig. 1
figure 1

Targeting and biodistribution/PK analysis strategies of exosome therapeutics. Targeting of exosomes to specific organs or cells could be achieved via modification of the composition of exosomal membrane proteins including tetraspanins and integrins. Exosomal surface engineering by displaying targeting peptides conjugated with exosomal membrane-associated domains such as lysosome-associated membrane glycoprotein 2b (Lamp2b) or C1C2 domain of lactadherin (LA) is another approach for active tissue targeting. Both glycan and lipid compositions of exosomal membrane also contribute to the biodistribution of administered exosomes. Biodistribution/PK analysis of administered exosomes can be conducted via various exosome labeling methods (i.e., bioluminescence, fluorescence, and radio isotope-labeling methods)

3.1 Cellular origin

One of the main factors that determine the biodistribution of exosomes is its cellular origin. Exosomes from different cellular sources were observed to have an asymmetric biodistribution [27], and subsequent studies found an inclination that exosomes tend to have different tropism based on their cells of origin, which could be further utilized for organ-targeted delivery. For instance, targeted delivery of exosomes to the brain could be achieved by using neural stem cell (NSC)-derived EVs, as it showed preferential brain targeting compared to mesenchymal stem cell (MSC)-derived EVs in a murine stroke model [47]. Tumor-derived exosomes were shown to be efficient in targeting its parental tumor for delivering anti-cancer drugs [29, 30, 48,49,50]. In a HT1080 xenograft mouse model, systemically injected HT1080-derived exosomes were targeted more efficiently to HT1080 tumor burden compared to HeLa-derived exosomes, with HT1080-derived exosomes delivered twice as much as HeLa-derived exosomes [30]. In zebrafish model, exosomes derived from either brain endothelial cells or brain tumor cells crossed the blood–brain barrier (BBB) and successfully deliver anti-cancer drugs to the brain tumors, with 4 nl of 0.2 mg/ml doxorubicin loaded in 200 μg/ml of brain endothelial cell-derived exosomes inhibiting expression of VEGF RNA more than half compared to doxorubicin-only injected brain tumor model of zebrafish [48]. Moreover, prostate cancer cell lines LNcaP- and PC-3-derived EVs loaded with Paclitaxel (PTX) have been shown to be effective carriers for delivering PTX to their parental cells [49]. However, Smyth et al. suggested that tumor-derived exosomes showed the tumor targeting property only when the exosomes were injected locally into the tumors [29]. In their study, they observed a minimal tumor targeting of systemically injected breast and prostate tumor-derived exosomes [29]. Jung et al. also showed that hypoxic cancer cell-derived exosomes are targeted to hypoxic cancer cells only in vitro but not in vivo [50]. More in-depth mechanistic studies are required regarding targeting ability of tumor cell-derived exosomes to their parental cancers. Even with tumor-targeting benefits for utilizing tumor-derived exosomes, they may have safety issues when administered systemically: tumor-derived exosomes may deliver tumorigenic factors to healthy cells and moreover, promote tumor metastasis by initiating pre-metastatic niche formation in healthy tissues [51,52,53]. Therefore, utilizing tumor-derived exosomes for tumor therapeutics may not be feasible. Instead, mechanistic insight for understanding tumor tropism of tumor-derived exosome can be applied to design targeting approach for tumor therapeutics with exosomes.

3.2 Membrane composition of exosomes (e.g., proteins, lipids, and glycans)

Cellular or organ targeting of exosomes is influenced by various membrane compositions of exosomes, such as proteins, lipids, and glycans. Membrane protein composition of exosomes is determined by their cellular origin as well as the physiological state of parental cells during exosome biogenesis [54]. For instance, exosomes originated from antigen-presenting cells, including dendritic cells, macrophages, and B cells, tend to display immune regulatory proteins and antigens similar to that of their parental cells [54]. Exosomes from mature dendritic cells were found to express mature phenotype markers such as major histocompatibility complex (MHC) class I, class II molecules, CD40, CD86 and ICAM-1/CD54 [55]. Similarly, exosomes released by B cells or T cells carry B-cell or T-cell receptor subunits, respectively [56, 57], and those derived from natural killer cells contain the NK cell marker CD56 [58], which partially resemble the features of the originated cells.

The major proteins that constitute the exosomal membranes are proteins such as tetraspanins (e.g., CD9, CD63, CD81, CD82), integrins and MHC molecules, of which various composition of these proteins could influence organotropism of exosomes [5]. For instance, exosomes expressing integrin α6β4 and α6β1 are targeted to laminin-enriched lung microenvironments, especially to the S100-A4-positive fibroblasts and surfactant protein C-positive epithelial cells in the lungs [59]. In contrast, exosomes displaying integrin αvβ5 preferentially interacts with fibronectin in the liver microenvironments, which is specifically targeted to F4/80 positive Kupffer cells [59]. Also, Qiao et al. identified eight different integrins (integrin αv, α3, α5, α6, β1, β4, β5, β6) in tumor-derived exosomes by proteome profiler array with receptor proteins, suggesting that these integrins contribute to the tumor tropism of tumor-derived exosomes [30]. Tetraspanins, which are abundant on the membrane of exosomes, also contribute to the organotropism of exosomes by forming a complex with other tetraspanins and integrins: exosomes with tetraspanin Tspan-8 and integrin α4 complex were readily targeted to endothelial and pancreas cells [60]. CD47 is the ligand for signal regulatory protein alpha (SIRPα), which upon binding initiates the ‘don’t eat me’ signal that inhibits phagocytosis [61]. Kamerkar et al. showed that CD47 expressed on the exosomes mediated protection from phagocytosis by monocytes and macrophages, which showed that engineering surface of exosomes with CD47 could induce prolonged circulation time [62].

The lipid and glycan composition of the surface of exosomes may also contribute to tissue tropism by modulating cellular uptake of exosomes [33, 63]. In vivo administered exosomes are rapidly up-taken by circulating phagocytic cells within several minutes after systemic administration [42, 43], and Matsumoto et al. found that the rapid uptake of intravenously administered B16-BL6 melanoma cell-derived exosomes by macrophages is mediated via recognizing negatively charged phosphatidylserine (PS) displayed on the membrane of exosomes [33]. Exosomal uptake could also be mediated by glycans on the membrane of exosomes. The uptake of glioblastoma (GBM) cell-derived EVs to the recipient GBM cells were shown to involve a triple interaction between the chemokine receptor CCR8 on the cells, glycans exposed on EVs and the soluble ligand CCL18, which in turn promoted GBM cell proliferation and resistance to the alkylating agent temozolomide [63].

3.3 Pathophysiological conditions of host

The biodistribution and PK parameters of exosomes could be affected by the pathophysiological conditions of host. Grange et al. observed the biodistribution of MSC-derived EVs in a model of acute kidney injury (AKI) after intravenous injection and found significant accumulation of EVs in the kidney of AKI-induced mouse 15 min after exosome injection, whereas accumulation of EVs in kidney appeared 5 h after injection in healthy mouse [64]. BBB crossing can also be achieved at certain pathological conditions. Under the circumstances of brain inflammation, Yuan et al. showed that macrophage-derived exosomes expressing LFA-1 and C-type lectin receptors can penetrate the BBB by interacting with inflamed brain microvascular endothelial cells, with showing over three times more accumulation of exosomes in brain of inflamed mice compared to healthy mice [65]. An in vitro trans-well assay study demonstrated that unmodified exosomes can cross the BBB through endocytosis by brain microvascular endothelial cells which occurred only under stroke-like, inflamed conditions induced by TNF-α [66]. Mirzaaghasi et al. investigated the biodistribution and blood PK of HEK293T cell-derived exosomes in sepsis-induced mouse. They found that substantial number of exosomes were delivered to the lung compared to healthy mouse after intravenous injection, of which more than 30% of exosomes were delivered to the lung in sepsis-induced mouse after 1 h of injection whereas almost none were detected in the lung of healthy mouse. Also, prolonged retention of exosomes in the blood circulation were observed due to liver dysfunction [28]. In other disease models, clearance of fluorophore labeled exosomes (10 nmol, intravenous) from blood in normal mice were 0.0054–0.0154 mL/min [67], whereas Gaussia luciferase (gLuc)-lactadherin (LA) labeled exosomes (5 µg, intravenous) in macrophage-depleted mice [42] and 125I labeled exosomes (4 × 105 cpm, intravenous) in Parkinson’s disease mouse model [65] were 0.651 ± 0.157 mL/h and 0.016 mL/min, respectively.

4 Imaging methods for determining biodistribution and PK of exosomes

Currently, bioluminescence and fluorescence imaging are the most commonly use methods for monitoring in vivo behavior of administered exosomes. However, with the recent technological advances for deep tissue penetration imaging, other clinical imaging methods including magnetic resonance imaging (MRI), positron emission tomography (PET) and single photon emission computed tomography (SPECT) are also being utilized for biodistribution and PK studies of exosomes (Fig. 1) [27, 62,

References

  1. Fradelizi D. Cytokines: soluble factors in intercellular communication. Med Trop (Mars). 1998;58:427–32.

    CAS  Google Scholar 

  2. Lauder JM. Neurotransmitters as growth regulatory signals: role of receptors and second messengers. Trends Neurosci. 1993;16:233–40.

    Article  CAS  PubMed  Google Scholar 

  3. Raposo G, Stoorvogel W. Extracellular vesicles: exosomes, microvesicles, and friends. J Cell Biol. 2013;200:373–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Thery C, Zitvogel L, Amigorena S. Exosomes: composition, biogenesis and function. Nat Rev Immunol. 2002;2:569–79.

    Article  CAS  PubMed  Google Scholar 

  5. Pegtel DM, Gould SJ. Exosomes. Annu Rev Biochem. 2019;88:487–514.

    Article  CAS  PubMed  Google Scholar 

  6. Peng H, Ji W, Zhao R, Yang J, Lu Z, Li Y, et al. Exosome: a significant nano-scale drug delivery carrier. J Mater Chem B. 2020;8:7591–608.

    Article  CAS  PubMed  Google Scholar 

  7. Caby MP, Lankar D, Vincendeau-Scherrer C, Raposo G, Bonnerot C. Exosomal-like vesicles are present in human blood plasma. Int Immunol. 2005;17:879–87.

    Article  CAS  PubMed  Google Scholar 

  8. Pisitkun T, Shen RF, Knepper MA. Identification and proteomic profiling of exosomes in human urine. Proc Natl Acad Sci U S A. 2004;101:13368–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Michael A, Bajracharya SD, Yuen PS, Zhou H, Star RA, Illei GG, et al. Exosomes from human saliva as a source of microRNA biomarkers. Oral Dis. 2010;16:34–8.

    Article  CAS  PubMed  Google Scholar 

  10. Admyre C, Johansson SM, Qazi KR, Filen JJ, Lahesmaa R, Norman M, et al. Exosomes with immune modulatory features are present in human breast milk. J Immunol. 2007;179:1969–78.

    Article  CAS  PubMed  Google Scholar 

  11. Vojtech L, Woo S, Hughes S, Levy C, Ballweber L, Sauteraud RP, et al. Exosomes in human semen carry a distinctive repertoire of small non-coding RNAs with potential regulatory functions. Nucleic Acids Res. 2014;42:7290–304.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Doyle LM, Wang MZ. Overview of extracellular vesicles, their origin, composition, purpose, and methods for exosome isolation and analysis. Cells. 2019;8:727.

    Article  CAS  PubMed Central  Google Scholar 

  13. Battistelli M, Falcieri E. Apoptotic bodies: particular extracellular vesicles involved in intercellular communication. Biology (Basel). 2020;9:21.

    CAS  Google Scholar 

  14. Yang D, Zhang W, Zhang H, Zhang F, Chen L, Ma L, et al. Progress, opportunity, and perspective on exosome isolation - efforts for efficient exosome-based theranostics. Theranostics. 2020;10:3684–707.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Choi DS, Choi DY, Hong BS, Jang SC, Kim DK, Lee J, et al. Quantitative proteomics of extracellular vesicles derived from human primary and metastatic colorectal cancer cells. J Extracell Vesicles. 2012;1:18704.

    Article  CAS  Google Scholar 

  16. Moon PG, Lee JE, You S, Kim TK, Cho JH, Kim IS, et al. Proteomic analysis of urinary exosomes from patients of early IgA nephropathy and thin basement membrane nephropathy. Proteomics. 2011;11:2459–75.

    Article  CAS  PubMed  Google Scholar 

  17. Andre F, Chaput N, Schartz NE, Flament C, Aubert N, Bernard J, et al. Exosomes as potent cell-free peptide-based vaccine. I. Dendritic cell-derived exosomes transfer functional MHC class I/peptide complexes to dendritic cells. J Immunol. 2004;172:2126–36.

    Article  CAS  PubMed  Google Scholar 

  18. Kalluri R, LeBleu VS. The biology, function, and biomedical applications of exosomes. Science. 2020;367(6478):eaau6977.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Kim YK, Choi Y, Nam GH, Kim IS. Functionalized exosome harboring bioactive molecules for cancer therapy. Cancer Lett. 2020;489:155–62.

    Article  CAS  PubMed  Google Scholar 

  20. Wahlund CJE, Gucluler G, Hiltbrunner S, Veerman RE, Naslund TI, Gabrielsson S. Exosomes from antigen-pulsed dendritic cells induce stronger antigen-specific immune responses than microvesicles in vivo. Sci Rep. 2017;7:17095.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  21. **ao L, Erb U, Zhao K, Hackert T, Zoller M. Efficacy of vaccination with tumor-exosome loaded dendritic cells combined with cytotoxic drug treatment in pancreatic cancer. Oncoimmunology. 2017;6:e1319044.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Song Y, Kim Y, Ha S, Sheller-Miller S, Yoo J, Choi C, et al. The emerging role of exosomes as novel therapeutics: biology, technologies, clinical applications, and the next. Am J Reprod Immunol. 2021;85:e13329.

    Article  PubMed  Google Scholar 

  23. Jafari D, Shajari S, Jafari R, Mardi N, Gomari H, Ganji F, et al. Designer exosomes: a new platform for biotechnology therapeutics. BioDrugs. 2020;34:567–86.

    Article  PubMed  PubMed Central  Google Scholar 

  24. Zipkin M. Exosome redux. Nat Biotechnol. 2019;37:1395–400.

    Article  CAS  PubMed  Google Scholar 

  25. Kim J, Song Y, Park CH, Choi C. Platform technologies and human cell lines for the production of therapeutic exosomes. Extracell Vesicles Circ Nucleic Acid. 2021;2:3–17.

    Google Scholar 

  26. Agrahari V, Agrahari V, Burnouf PA, Chew CH, Burnouf T. Extracellular microvesicles as new industrial therapeutic frontiers. Trends Biotechnol. 2019;37:707–29.

    Article  CAS  PubMed  Google Scholar 

  27. Wiklander OP, Nordin JZ, O’Loughlin A, Gustafsson Y, Corso G, Mager I, et al. Extracellular vesicle in vivo biodistribution is determined by cell source, route of administration and targeting. J Extracell Vesicles. 2015;4:26316.

    Article  PubMed  Google Scholar 

  28. Mirzaaghasi A, Han Y, Ahn SH, Choi C, Park JH. Biodistribution and pharmacokinectics of liposomes and exosomes in a mouse model of sepsis. Pharmaceutics. 2021;13:427.

    Article  PubMed  PubMed Central  Google Scholar 

  29. Smyth T, Kullberg M, Malik N, Smith-Jones P, Graner MW, Anchordoquy TJ. Biodistribution and delivery efficiency of unmodified tumor-derived exosomes. J Control Release. 2015;199:145–55.

    Article  CAS  PubMed  Google Scholar 

  30. Qiao L, Hu SQ, Huang K, Su T, Li ZH, Vandergriff A, et al. Tumor cell-derived exosomes home to their cells of origin and can be used as Trojan horses to deliver cancer drugs. Theranostics. 2020;10:3474–87.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Rashid MH, Borin TF, Ara R, Angara K, Cai J, Achyut BR, et al. Differential in vivo biodistribution of 131I-labeled exosomes from diverse cellular origins and its implication for theranostic application. Nanomedicine. 2019;21:102072.

    Article  CAS  PubMed  Google Scholar 

  32. Faruqu FN, Wang JT, Xu L, McNickle L, Chong EM, Walters A, et al. Membrane radiolabelling of exosomes for comparative biodistribution analysis in immunocompetent and immunodeficient mice - a novel and universal approach. Theranostics. 2019;9:1666–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Matsumoto A, Takahashi Y, Nishikawa M, Sano K, Morishita M, Charoenviriyakul C, et al. Role of phosphatidylserine-derived negative surface charges in the recognition and uptake of intravenously injected B16BL6-derived exosomes by macrophages. J Pharm Sci. 2017;106:168–75.

    Article  CAS  PubMed  Google Scholar 

  34. Morishita M, Takahashi Y, Nishikawa M, Takakura Y. Pharmacokinetics of exosomes-an important factor for elucidating the biological roles of exosomes and for the development of exosome-based therapeutics. J Pharm Sci. 2017;106:2265–9.

    Article  CAS  PubMed  Google Scholar 

  35. Kim S, Lee SA, Yoon H, Kim MY, Yoo JK, Ahn SH, et al. Exosome-based delivery of super-repressor IκBα ameliorates kidney ischemia-reperfusion injury. Kidney Int. 2021. https://doi.org/10.1016/j.kint.2021.04.039

  36. 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;29:341–5.

    Article  CAS  PubMed  Google Scholar 

  37. Sheller-Miller S, Radnaa E, Yoo JK, Kim E, Choi K, Kim Y, et al. Exosomal delivery of NF-κB inhibitor delays LPS-induced preterm birth and modulates fetal immune cell profile in mouse models. Sci Adv. 2021;7:eabd3865.

    Article  CAS  PubMed  Google Scholar 

  38. Sheller-Miller S, Choi K, Choi C, Menon R. Cyclic-recombinase-reporter mouse model to determine exosome communication and function during pregnancy. Am J Obstet Gynecol. 2019;221:502.e1–12.

  39. Wang N, Chen C, Yang D, Liao Q, Luo H, Wang X, et al. Mesenchymal stem cells-derived extracellular vesicles, via miR-210, improve infarcted cardiac function by promotion of angiogenesis. Biochim Biophys Acta Mol Basis Dis. 2017;1863:2085–92.

    Article  CAS  PubMed  Google Scholar 

  40. Li D, Zhang P, Yao X, Li H, Shen H, Li X, et al. Exosomes derived from mir-133b-modified mesenchymal stem cells promote recovery after spinal cord injury. Front Neurosci. 2018;12:845.

    Article  PubMed  PubMed Central  Google Scholar 

  41. Tao SC, Yuan T, Zhang YL, Yin WJ, Guo SC, Zhang CQ. Exosomes derived from miR-140-5p-overexpressing human synovial mesenchymal stem cells enhance cartilage tissue regeneration and prevent osteoarthritis of the knee in a rat model. Theranostics. 2017;7:180–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Imai T, Takahashi Y, Nishikawa M, Kato K, Morishita M, Yamashita T, et al. Macrophage-dependent clearance of systemically administered B16BL6-derived exosomes from the blood circulation in mice. J Extracell Vesicles. 2015;4:26238.

    Article  PubMed  Google Scholar 

  43. Choi H, Kim Y, Mirzaaghasi A, Heo J, Kim YN, Shin JH, et al. Exosome-based delivery of super-repressor I kappa B alpha relieves sepsis-associated organ damage and mortality. Sci Adv. 2020;6:eaaz6980.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Murphy DE, de Jong OG, Brouwer M, Wood MJ, Lavieu G, Schiffelers RM, et al. Extracellular vesicle-based therapeutics: natural versus engineered targeting and trafficking. Exp Mol Med. 2019;51:1–12.

    Article  CAS  PubMed  Google Scholar 

  45. Yi YW, Lee JH, Kim SY, Pack CG, Ha DH, Park SR, et al. Advances in analysis of biodistribution of exosomes by molecular imaging. Int J Mol Sci. 2020;21:665.

    Article  CAS  PubMed Central  Google Scholar 

  46. Druzhkova TA, Yakovlev AA. Exosome drug delivery through the blood-brain barrier: experimental approaches and potential applications. Neurochem J. 2018;12:195–204.

    Article  CAS  Google Scholar 

  47. Webb RL, Kaiser EE, Scoville SL, Thompson TA, Fatima S, Pandya C, et al. Human neural stem cell extracellular vesicles improve tissue and functional recovery in the murine thromboembolic stroke model. Transl Stroke Res. 2018;9:530–9.

    Article  CAS  PubMed  Google Scholar 

  48. Yang TZ, Martin P, Fogarty B, Brown A, Schurman K, Phipps R, et al. Exosome delivered anticancer drugs across the blood-brain barrier for brain cancer therapy in danio rerio. Pharm Res-Dordr. 2015;32:2003–14.

    Article  CAS  Google Scholar 

  49. Saari H, Lazaro-Ibanez E, Viitala T, Vuorimaa-Laukkanen E, Siljander P, Yliperttula M. Microvesicle- and exosome-mediated drug delivery enhances the cytotoxicity of Paclitaxel in autologous prostate cancer cells. J Control Release. 2015;220:727–37.

    Article  CAS  PubMed  Google Scholar 

  50. Jung KO, Jo H, Yu JH, Gambhir SS, Pratx G. Development and MPI tracking of novel hypoxia-targeted theranostic exosomes. Biomaterials. 2018;177:139–48.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Costa-Silva B, Aiello NM, Ocean AJ, Singh S, Zhang H, Thakur BK, et al. Pancreatic cancer exosomes initiate pre-metastatic niche formation in the liver. Nat Cell Biol. 2015;17:816–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Guo Y, Ji X, Liu J, Fan D, Zhou Q, Chen C, et al. Effects of exosomes on pre-metastatic niche formation in tumors. Mol Cancer. 2019;18:39.

    Article  PubMed  PubMed Central  Google Scholar 

  53. Suetsugu A, Honma K, Saji S, Moriwaki H, Ochiya T, Hoffman RM. Imaging exosome transfer from breast cancer cells to stroma at metastatic sites in orthotopic nude-mouse models. Adv Drug Deliv Rev. 2013;65:383–90.

    Article  CAS  PubMed  Google Scholar 

  54. Hu Q, Su H, Li J, Lyon C, Tang W, Wan M, et al. Clinical applications of exosome membrane proteins. Precis Clin Med. 2020;3:54–66.

    Article  PubMed  PubMed Central  Google Scholar 

  55. Besse B, Charrier M, Lapierre V, Dansin E, Lantz O, Planchard D, et al. Dendritic cell-derived exosomes as maintenance immunotherapy after first line chemotherapy in NSCLC. Oncoimmunology. 2016;5:e1071008.

    Article  PubMed  CAS  Google Scholar 

  56. Rialland P, Lankar D, Raposo G, Bonnerot C, Hubert P. BCR-bound antigen is targeted to exosomes in human follicular lymphoma B-cells. Biol Cell. 2006;98:491–501.

    Article  CAS  PubMed  Google Scholar 

  57. Blanchard N, Lankar D, Faure F, Regnault A, Dumont C, Raposo G, et al. TCR activation of human T cells induces the production of exosomes bearing the TCR/CD3/zeta complex. J Immunol. 2002;168:3235–41.

    Article  CAS  PubMed  Google Scholar 

  58. Lugini L, Cecchetti S, Huber V, Luciani F, Macchia G, Spadaro F, et al. Immune surveillance properties of human NK cell-derived exosomes. J Immunol. 2012;189:2833–42.

    Article  CAS  PubMed  Google Scholar 

  59. Hoshino A, Costa-Silva B, Shen TL, Rodrigues G, Hashimoto A, Mark MT, et al. Tumour exosome integrins determine organotropic metastasis. Nature. 2015;527:329.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Rana S, Yue SJ, Stadel D, Zoller M. Toward tailored exosomes: the exosomal tetraspanin web contributes to target cell selection. Int J Biochem Cell B. 2012;44:1574–84.

    Article  CAS  Google Scholar 

  61. Chao MP, Weissman IL, Majeti R. The CD47-SIRPalpha pathway in cancer immune evasion and potential therapeutic implications. Curr Opin Immunol. 2012;24:225–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Kamerkar S, LeBleu VS, Sugimoto H, Yang S, Ruivo CF, Melo SA, et al. Exosomes facilitate therapeutic targeting of oncogenic KRAS in pancreatic cancer. Nature. 2017;546:498–503.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Berenguer J, Lagerweij T, Zhao XW, Dusoswa S, van der Stoop P, Westerman B, et al. Glycosylated extracellular vesicles released by glioblastoma cells are decorated by CCL18 allowing for cellular uptake via chemokine receptor CCR8. J Extracell Vesicles. 2018;7(1):1446660.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  64. Grange C, Tapparo M, Bruno S, Chatterjee D, Quesenberry PJ, Tetta C, et al. Biodistribution of mesenchymal stem cell-derived extracellular vesicles in a model of acute kidney injury monitored by optical imaging. Int J Mol Med. 2014;33:1055–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Yuan D, Zhao Y, Banks WA, Bullock KM, Haney M, Batrakova E, et al. Macrophage exosomes as natural nanocarriers for protein delivery to inflamed brain. Biomaterials. 2017;142:1–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Chen CC, Liu LN, Ma FX, Wong CW, Guo XNE, Chacko JV, et al. Elucidation of exosome migration across the blood-brain barrier model in vitro. Cell Mol Bioeng. 2016;9:509–29.

    Article  CAS  PubMed  Google Scholar 

  67. Hwang DW, Jo MJ, Lee JH, Kang H, Bao K, Hu S, et al. Chemical modulation of bioengineered exosomes for tissue-specific biodistribution. Adv Ther. 2019;2:1900111.

    Article  CAS  Google Scholar 

  68. Fu S, Wang Y, **a X, Zheng JC. Exosome engineering: Current progress in cargo loading and targeted delivery. NanoImpact. 2020;20:100261.

    Article  Google Scholar 

  69. Wen SW, Sceneay J, Lima LG, Wong CSF, Becker M, Krumeich S, et al. The biodistribution and immune suppressive effects of breast cancer-derived exosomes. Can Res. 2016;76:6816–27.

    Article  CAS  Google Scholar 

  70. Munagala R, Aqil F, Jeyabalan J, Gupta RC. Bovine milk-derived exosomes for drug delivery. Cancer Lett. 2016;371:48–61.

    Article  CAS  PubMed  Google Scholar 

  71. Mendt M, Kamerkar S, Sugimoto H, McAndrews KM, Wu C-C, Gagea M, et al. Generation and testing of clinical-grade exosomes for pancreatic cancer. JCI Insight. 2018;3:e99263.

  72. Peinado H, Alečković M, Lavotshkin S, Matei I, Costa-Silva B, Moreno-Bueno G, et al. Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat Med. 2012;18:883–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Takahashi Y, Nishikawa M, Shinotsuka H, Matsui Y, Ohara S, Imai T, et al. Visualization and in vivo tracking of the exosomes of murine melanoma B16-BL6 cells in mice after intravenous injection. J Biotechnol. 2013;165:77–84.

    Article  CAS  PubMed  Google Scholar 

  74. Lai CP, Mardini O, Ericsson M, Prabhakar S, Maguire CA, Chen JW, et al. Dynamic biodistribution of extracellular vesicles in vivo using a multimodal imaging reporter. ACS Nano. 2014;8:483–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. González MI, Martín-Duque P, Desco M, Salinas B. Radioactive labeling of milk-derived exosomes with 99mTc and in vivo tracking by SPECT imaging. Nanomaterials. 2020;10:1062.

    Article  PubMed Central  CAS  Google Scholar 

  76. Jung KO, Kim YH, Chung SJ, Lee CH, Rhee S, Pratx G, et al. Identification of lymphatic and hematogenous routes of rapidly labeled radioactive and fluorescent exosomes through highly sensitive multimodal imaging. Int J Mol Sci. 2020;21:7850.

    Article  CAS  PubMed Central  Google Scholar 

  77. Busato A, Bonafede R, Bontempi P, Scambi I, Schiaffino L, Benati D, et al. Magnetic resonance imaging of ultrasmall superparamagnetic iron oxide-labeled exosomes from stem cells: a new method to obtain labeled exosomes. Int J Nanomed. 2016;11:2481–90.

    CAS  Google Scholar 

  78. Hu L, Wickline SA, Hood JL. Magnetic resonance imaging of melanoma exosomes in lymph nodes. Magn Reson Med. 2015;74:266–71.

    Article  CAS  PubMed  Google Scholar 

  79. Dabrowska S, Del Fattore A, Karnas E, Frontczak-Baniewicz M, Kozlowska H, Muraca M, et al. Imaging of extracellular vesicles derived from human bone marrow mesenchymal stem cells using fluorescent and magnetic labels. Int J Nanomed. 2018;13:1653–64.

    Article  CAS  Google Scholar 

  80. Liu T, Zhu Y, Zhao R, Wei X, **n X. Visualization of exosomes from mesenchymal stem cells in vivo by magnetic resonance imaging. Magn Reson Imaging. 2020;68:75–82.

    Article  CAS  PubMed  Google Scholar 

  81. Zhu Q, Heon M, Zhao Z, He M. Microfluidic engineering of exosomes: editing cellular messages for precision therapeutics. Lab Chip. 2018;18:1690–703.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Rayamajhi S, Aryal S. Surface functionalization strategies of extracellular vesicles. J Mater Chem B. 2020;8:4552–69.

    Article  CAS  PubMed  Google Scholar 

  83. Baek G, Choi H, Kim Y, Lee HC, Choi C. Mesenchymal stem cell-derived extracellular vesicles as therapeutics and as a drug delivery platform. Stem Cells Transl Med. 2019;8:880–6.

    Article  PubMed  PubMed Central  Google Scholar 

  84. Smyth T, Petrova K, Payton NM, Persaud I, Redzic JS, Graner MW, et al. Surface functionalization of exosomes using click chemistry. Bioconjug Chem. 2014;25:1777–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Ramasubramanian L, Kumar P, Wang A. Engineering extracellular vesicles as nanotherapeutics for regenerative medicine. Biomolecules. 2019;10:48.

    Article  PubMed Central  CAS  Google Scholar 

  86. Algar WR, Prasuhn DE, Stewart MH, Jennings TL, Blanco-Canosa JB, Dawson PE, et al. The controlled display of biomolecules on nanoparticles: a challenge suited to bioorthogonal chemistry. Bioconjug Chem. 2011;22:825–58.

    Article  CAS  PubMed  Google Scholar 

  87. Villata S, Canta M, Cauda V. EVs and bioengineering: from cellular products to engineered nanomachines. Int J Mol Sci. 2020;21:6048.

    Article  CAS  PubMed Central  Google Scholar 

  88. Nwe K, Brechbiel MW. Growing applications of “click chemistry” for bioconjugation in contemporary biomedical research. Cancer Biother Radiopharm. 2009;24:289–302.

    CAS  PubMed  PubMed Central  Google Scholar 

  89. Susa F, Limongi T, Dumontel B, Vighetto V, Cauda V. Engineered extracellular vesicles as a reliable tool in cancer nanomedicine. Cancers (Basel). 2019;11:1979.

    Article  CAS  Google Scholar 

  90. Kim MS, Haney MJ, Zhao Y, Yuan D, Deygen I, Klyachko NL, et al. Engineering macrophage-derived exosomes for targeted paclitaxel delivery to pulmonary metastases: in vitro and in vivo evaluations. Nanomedicine. 2018;14:195–204.

    Article  CAS  PubMed  Google Scholar 

  91. Jia G, Han Y, An Y, Ding Y, He C, Wang X, et al. NRP-1 targeted and cargo-loaded exosomes facilitate simultaneous imaging and therapy of glioma in vitro and in vivo. Biomaterials. 2018;178:302–16.

    Article  CAS  PubMed  Google Scholar 

  92. Chen L, Miao W, Tang X, Zhang H, Wang S, Luo F, et al. The expression and significance of neuropilin-1 (NRP-1) on glioma cell lines and glioma tissues. J Biomed Nanotechnol. 2013;9:559–63.

    Article  CAS  PubMed  Google Scholar 

  93. Tian T, Zhang HX, He CP, Fan S, Zhu YL, Qi C, et al. Surface functionalized exosomes as targeted drug delivery vehicles for cerebral ischemia therapy. Biomaterials. 2018;150:137–49.

    Article  CAS  PubMed  Google Scholar 

  94. Armstrong JP, Holme MN, Stevens MM. Re-engineering extracellular vesicles as smart nanoscale therapeutics. ACS Nano. 2017;11:69–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Nel AE, Madler L, Velegol D, **a T, Hoek EM, Somasundaran P, et al. Understanding biophysicochemical interactions at the nano-bio interface. Nat Mater. 2009;8:543–57.

    Article  CAS  PubMed  Google Scholar 

  96. Qi H, Liu C, Long L, Ren Y, Zhang S, Chang X, et al. Blood exosomes endowed with magnetic and targeting properties for cancer therapy. ACS Nano. 2016;10:3323–33.

    Article  CAS  PubMed  Google Scholar 

  97. Nakase I, Futaki S. Combined treatment with a pH-sensitive fusogenic peptide and cationic lipids achieves enhanced cytosolic delivery of exosomes. Sci Rep. 2015;5:10112.

    Article  PubMed  PubMed Central  Google Scholar 

  98. Leroueil PR, Berry SA, Duthie K, Han G, Rotello VM, McNerny DQ, et al. Wide varieties of cationic nanoparticles induce defects in supported lipid bilayers. Nano Lett. 2008;8:420–4.

    Article  CAS  PubMed  Google Scholar 

  99. Biscans A, Haraszti RA, Echeverria D, Miller R, Didiot MC, Nikan M, et al. Hydrophobicity of lipid-conjugated siRNAs predicts productive loading to small extracellular vesicles. Mol Ther. 2018;26:1520–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Vandergriff A, Huang K, Shen D, Hu S, Hensley MT, Caranasos TG, et al. Targeting regenerative exosomes to myocardial infarction using cardiac homing peptide. Theranostics. 2018;8:1869–78.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Sato YT, Umezaki K, Sawada S, Mukai SA, Sasaki Y, Harada N, et al. Engineering hybrid exosomes by membrane fusion with liposomes. Sci Rep. 2016;6:21933.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Mentkowski KI, Snitzer JD, Rusnak S, Lang JK. Therapeutic potential of engineered extracellular vesicles. AAPS J. 2018;20:50.

    Article  PubMed  CAS  Google Scholar 

  103. Liang Y, Duan L, Lu J, **a J. Engineering exosomes for targeted drug delivery. Theranostics. 2021;11:3183–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Luan X, Sansanaphongpricha K, Myers I, Chen H, Yuan H, Sun D. Engineering exosomes as refined biological nanoplatforms for drug delivery. Acta Pharmacol Sin. 2017;38:754–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Vakhshiteh F, Atyabi F, Ostad SN. Mesenchymal stem cell exosomes: a two-edged sword in cancer therapy. Int J Nanomed. 2019;14:2847–59.

    Article  CAS  Google Scholar 

  106. Zhang M, Zang X, Wang M, Li Z, Qiao M, Hu H, et al. Exosome-based nanocarriers as bio-inspired and versatile vehicles for drug delivery: recent advances and challenges. J Mater Chem B. 2019;7:2421–33.

    Article  CAS  PubMed  Google Scholar 

  107. Wang J, Zheng Y, Zhao M. Exosome-based cancer therapy: implication for targeting cancer stem cells. Front Pharmacol. 2016;7:533.

    PubMed  Google Scholar 

  108. Kooijmans SA, Aleza CG, Roffler SR, van Solinge WW, Vader P, Schiffelers RM. Display of GPI-anchored anti-EGFR nanobodies on extracellular vesicles promotes tumour cell targeting. J Extracell Vesicles. 2016;5:31053.

    Article  PubMed  CAS  Google Scholar 

  109. de Gassart A, Geminard C, Fevrier B, Raposo G, Vidal M. Lipid raft-associated protein sorting in exosomes. Blood. 2003;102:4336–44.

    Article  PubMed  CAS  Google Scholar 

  110. Longatti A, Schindler C, Collinson A, Jenkinson L, Matthews C, Fitzpatrick L, et al. High affinity single-chain variable fragments are specific and versatile targeting motifs for extracellular vesicles. Nanoscale. 2018;10:14230–44.

    Article  CAS  PubMed  Google Scholar 

  111. Tian Y, Li S, Song J, Ji T, Zhu M, Anderson GJ, et al. A doxorubicin delivery platform using engineered natural membrane vesicle exosomes for targeted tumor therapy. Biomaterials. 2014;35:2383–90.

    Article  CAS  PubMed  Google Scholar 

  112. Bai J, Duan J, Liu R, Du Y, Luo Q, Cui Y, et al. Engineered targeting tLyp-1 exosomes as gene therapy vectors for efficient delivery of siRNA into lung cancer cells. Asian J Pharm Sci. 2020;15:461–71.

    Article  PubMed  Google Scholar 

  113. Gomari H, Forouzandeh Moghadam M, Soleimani M. Targeted cancer therapy using engineered exosome as a natural drug delivery vehicle. Onco Targets Ther. 2018;11:5753–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Liang G, Kan S, Zhu Y, Feng S, Feng W, Gao S. Engineered exosome-mediated delivery of functionally active miR-26a and its enhanced suppression effect in HepG2 cells. Int J Nanomed. 2018;13:585–99.

    Article  CAS  Google Scholar 

  115. Gong C, Tian J, Wang Z, Gao Y, Wu X, Ding X, et al. Functional exosome-mediated co-delivery of doxorubicin and hydrophobically modified microRNA 159 for triple-negative breast cancer therapy. J Nanobiotechnol. 2019;17:93.

    Article  CAS  Google Scholar 

  116. Albelda SM, Mette SA, Elder DE, Stewart R, Damjanovich L, Herlyn M, et al. Integrin distribution in malignant melanoma: association of the beta 3 subunit with tumor progression. Cancer Res. 1990;50:6757–64.

    CAS  PubMed  Google Scholar 

  117. Gingras MC, Roussel E, Bruner JM, Branch CD, Moser RP. Comparison of cell adhesion molecule expression between glioblastoma multiforme and autologous normal brain tissue. J Neuroimmunol. 1995;57:143–53.

    Article  CAS  PubMed  Google Scholar 

  118. Natali PG, Hamby CV, Felding-Habermann B, Liang B, Nicotra MR, Di Filippo F, et al. Clinical significance of alpha(v)beta3 integrin and intercellular adhesion molecule-1 expression in cutaneous malignant melanoma lesions. Cancer Res. 1997;57:1554–60.

    CAS  PubMed  Google Scholar 

  119. Yang Z, Shi J, **e J, Wang Y, Sun J, Liu T, et al. Large-scale generation of functional mRNA-encapsulating exosomes via cellular nanoporation. Nat Biomed Eng. 2020;4:69–83.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgement

This work was supported by a grant (2016M3A9B6945931) from Ministry of Science and ICT, Republic of Korea.

Author information

Authors and Affiliations

Authors

Contributions

HC, YC, HY and AM wrote, and edited the manuscript. JY and CC supervised, reviewed, edited, and approved the manuscript.

Corresponding authors

Correspondence to Jae-Kwang Yoo or Chulhee Choi.

Ethics declarations

Conflict of interest

CC is the founder and shareholder, and JY, HC, HY are minor shareholders of ILIAS Biologics Inc. The authors have no additional financial interests.

Ethical statement

There were no animal or human subject experiments carried out for this article.

Additional information

Publisher's Note

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

Rights and permissions

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

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Choi, H., Choi, Y., Yim, H.Y. et al. Biodistribution of Exosomes and Engineering Strategies for Targeted Delivery of Therapeutic Exosomes. Tissue Eng Regen Med 18, 499–511 (2021). https://doi.org/10.1007/s13770-021-00361-0

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s13770-021-00361-0

Keywords