Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 2257 2022-12-15 15:16:15 |
2 format corrected. + 1 word(s) 2258 2022-12-19 04:26:20 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Rech, J.;  Getinger-Panek, A.;  Gałka, S.;  Bednarek, I. Exosomes in Designing Drug Delivery Systems. Encyclopedia. Available online: https://encyclopedia.pub/entry/38924 (accessed on 02 July 2024).
Rech J,  Getinger-Panek A,  Gałka S,  Bednarek I. Exosomes in Designing Drug Delivery Systems. Encyclopedia. Available at: https://encyclopedia.pub/entry/38924. Accessed July 02, 2024.
Rech, Jakub, Agnieszka Getinger-Panek, Sabina Gałka, Ilona Bednarek. "Exosomes in Designing Drug Delivery Systems" Encyclopedia, https://encyclopedia.pub/entry/38924 (accessed July 02, 2024).
Rech, J.,  Getinger-Panek, A.,  Gałka, S., & Bednarek, I. (2022, December 17). Exosomes in Designing Drug Delivery Systems. In Encyclopedia. https://encyclopedia.pub/entry/38924
Rech, Jakub, et al. "Exosomes in Designing Drug Delivery Systems." Encyclopedia. Web. 17 December, 2022.
Exosomes in Designing Drug Delivery Systems
Edit

Exosomes are a subpopulation of extravascular vesicles with a diameter of 30–150 nm. They are cellular-communication mediators, often reaching very distant organism tissues. Information is transferred by exosomal cargo, composed of a wide variety of macromolecules such as nucleic acids, proteins, and lipids. Exosomes possess natural specific cell targeting properties that are desirable in designing targeted macromolecules (DNA and RNA) and drug delivery systems (doxorubicin, paclitaxel, and taxol). In this context, exosomes can be defined as bio-derived drug transporting and protecting devices for the treatment of bacterial (toxoplasmosis and salmonellosis), viral (AIDS and hepatitis B), and cancer (lung, pancreatic, colon, brain, and breast) diseases. Extensive research proves that exosomes’ natural cargo can double-act, both increasing and decreasing the disease severity. In this case, the exosomes need to be prepared, namely, their origin and their cargo need to be screened and known. Thus, appropriate methods for intact and price-effective exosome isolation are needed with further exosome properties description. Among many utilized isolation methods, the most common are ultracentrifugation, polymer-based precipitation, and affinity precipitation-isolation systems, but novel microfluidic methods compromising high efficacy and purity are being developed. 

exosomes drug delivery systems targeted drug delivery

1. Introduction

Exosomes can serve as a drug delivery platform for macromolecules (DNA, RNA, proteins, and lipids) and drugs in hastily developing pharmaceutical units. Currently, they are under extensive research to adjust their properties to overcome specific drug delivery obstacles [1][2].
For over the last two decades, exosomes have been isolated from many cell types including normal and cancer cells, and their impact on other cells has been studied. It is a well-known fact that their main properties come from the cargo that can vary between distinct cells. These cargoes are cell-derived particles, namely nucleic acids, proteins, and lipids, which can influence other cells’ metabolism and functionality [3][4]. Due to the similarity of exosomes to artificial lipid microvesicles, researchers started to incorporate drugs, macromolecules, and other substances, which, for clarity, are altogether stated as drugs in this research [5][6]. This attitude resulted in an improvement in therapy efficiency employing increased cell-drug response, a decrease in necessary drug concentration, and an unwanted systemic organism response. The possibility of additional targeted delivery is another advantage of exosomes [7]. Some researchers proposed advanced combined delivery strategies that improve therapeutics efficiency by additional cell expression pattern manipulation with macromolecules such as RNA or DNA [8][9][10].

2. Exosomes Natural Cargo and Structure

Exosomes are generally recognized as cell-to-cell signaling molecules containing numerous proteins, nucleic acids, cytokines, transcription factors, and other cell-derived particles (Figure 1). Exosomes may deliver information not only to the local environment but also to greatly distanced cells and tissues. This may serve as a general signaling and communication pathway, but may also take part in oncogenes spread and further tumor progression and malignancy increase [11]. To this date, exosomes presence was confirmed in body fluids such as amniotic liquid, blood serum, breast milk, epididymal fluid, saliva, urine, effusions of ascites and pleural, bronchoalveolar lavage fluid, synovial fluid, and cell culture supernatant in vitro [12][13][14][15]. Due to these facts, exosomes are proposed as novel therapeutic and diagnostic (theranostics) devices [6]. The excreted exosomes also serve as a way of unnecessary proteins and nucleic acids removal, e.g., during cell maturation (reticulocytes) or excretory system (guts and tubules of the kidney) [16][17][18].
For proper usage of exosomes as a DDS, people first have to consider their natural cargo and the routes of their formation. Then, the characteristics of exosomes affinity and cell uptake should also be described. Exosomal cargo exhibit different purposes and functions, thanks to exosomes being basic transporters presenting outstanding properties themselves. This can be utilized in cell targeting and also as an additional enhancement of drugs. To date, ExoCarta estimates about 10,000 proteins, 3500 mRNA, and over 1100 distinct lipids found in exosomes. For the systematic review, many great databases arise such as Vesiclepedia and ExoCarta where exosome isolation procedures and sources are described along with identified cargo [19][20]. The most important proteins for DDS design are those influencing exosomes transport and cell intake.
Proteins characteristic for exosomes are membrane-bound tetraspanins (CD9, CD63, CD81, and CD82), as well as EpCAM and Rab5, used routinely for isolation. Other commonly recognized proteins are receptors (CD46 and CD55), heat shock proteins (HSP; Hsc70, Hsp70, and Hsp90), proteins taking part in exosomes formation (Alix, TSG101), and membrane proteins responsible for fusion and transport (GTPases, annexins, and flotillin; ATP7A, ATP7B, MRP2, SLC1A4, SLC16A1, and CLIC1) [6][21]. Detection of these markers with ELISA is used for confirmation of exosomes presence.
There are also reports about exosomes containing other particles such as cholesterol (B lymphocytes derived), sphingolipids, phosphoglycerides, and ceramide. The presence of the latter also distinguishes the exosomes from the lysosomes [22].
Exosomes also transport morphogens (Hedgehog, Wingless, and Wingless-like) taking part in tissue patterning development described for Drosophila melanogaster [13].
Nucleic acids are another major group of particles found in exosomes, namely DNA and RNA, which onward can take the functional role in cells. About ~1300 mRNA, 121 miRNA, and >100 small RNA were found in exosomes derived from mouse and human mast cells, while no DNA and rRNA were detected [23]. Other miRNAs found in exosomes were lin-4 and let-7, as well as miR-181a and miR-155 in breast milk [23][24].
Some of the exosomal miRNAs can be used as biomarkers in diagnostics (Table 1).
Figure 1. Exosomes structure and cargo.
Table 1. Examples of miRNA-level changes proposed for diagnosing diseases.
miRNA Level Change Source of Exosomes Disease Source
↑ miR-21 Serum Esophageal squamous cell cancer [25]
↓ miR-21 Benign diseases  
↑ miR-21 Serum Hepatocellular carcinoma [26]
↓ miR-21 Chronic hepatitis B or healthy volunteers as well  
↑ miR-1246
↑ miR-4644
Saliva Pancreatobiliary tract cancer [27]
↑ bta-miR-142-5p
↑ bta-miR-223
Milk Detection of early mammary gland bacterial infection [28]
↑miR-1290
↑ miR-375
Plasma Poor overall survival of castration-resistant prostate cancer patients [29]
↑ miR-146a Urine Lupus nephritis patients. Also distinguishing between active and remission stages of the disease. [30]
↑ miR-1910-3p Serum Breast cancer patients [31]
↑ miR-423-5p Serum Lymph node metastasis of gastric cancer patients [32]
↑ miR-106b Serum Lung cancer patients [33]
To summarize, exosomal cargo can further improve its therapeutic properties when properly designed. This can be used for the fabrication of double-action exosomes, containing both therapeutic substances with the additional effect enhancement by incorporating proper nucleic acids, lipids, and proteins into exosomes. To this date, only a few concepts of this simultaneous strategy were proposed [8][34].

3. Exosomes Innate Functions

Exosomes function will vary depending on their origin, cargo, and recipient cells. These innate properties can serve for further development and upgrading of exosome-based DDS. Exosomes’ most fundamental and widely recognized function is to mediate cell–cell signalization, thus regulating cell physiological functions and responses. This mechanism relies on the exchange of molecules described in the previous chapter.
Additionally, to date, many great articles arise with exosome function/effect description [14][35]. Thus, in this research, the researchers only mention properties influencing DDS. Exosomes derived from bone marrow, macrophages, and tumors were described as influencing the inflammatory response, either prolonging allograft rejection time or activating inflammatory cells to the target tumor [36][37]. Exosomes also take part in the immune response and antigen presentation. Due to this fact, they can stimulate the immune response by MHC class I or II antigens presentation, which is not desirable in the case of DDS or allogenic therapeutics and can decrease their body circulation time. The current solution to this problem is exosomes isolated from mesenchymal or stromal stem cells [38].
As mentioned, the natural exosomal cargo consists of various nucleic acids, proteins, and lipids, which can directly influence cell behavior and expression pattern. Similarly, artificial siRNA, mRNA, and miRNA delivered by exosomes will regulate cell expression as reported [39]. In a similar manner, proteins can also act as procancerogenic agents, e.g., exosomes containing high levels of TGF-β and PGE2 will promote tumor growth [40]. On the other hand, dendritic-cell-derived exosomes containing MHC class I molecules and tumor antigens are able to activate T-cell anticancer response [37].
Exosomes are superior to artificial lipovesicles in terms of biocompatibility and internalization. They can also serve to reach hard-to-access and brain tissues due to the possibility of targeted delivery and blood–brain-barrier crossing, respectively [41].

4. Targeted Delivery

Another factor influencing exosomes targeted delivery is the cell origin and membrane-incorporated targeting moieties [42][43]. Isolated exosomes are proposed for targeted therapy and drug delivery to the same type of cells the exosomes were derived from. It is due to the fact that different membrane moieties are differently expressed by various cells. Surface moieties can be either expressed via genetic engineering of the endosomal sorting mechanism or fixed at the surface of released exosomes with click chemistry (Table 2) [35][43]. A similar approach is described for other DDS devices such as fibrin [44].
Table 2. Examples of moieties for exosome targeting.
Moiety Targeted Cells/Tissue Source
Rabies viral glycoprotein Brain [45]
Mannose- and sialic acid- enriched glycoproteins Cancer cells [46]
Integrins α6β4 and α6β1 Lung metastasis [47]
Integrins αvβ5 Liver metastasis [48]
Cd63 Neuronal dendrites [49]
Phosphatidylethanolamine Glioblastoma cells [50]
Sphingomyelin Tumor microenvironment [51]
Metalloproteinase 15 Breast cancer cells (integrin αvβ3) [8]
Folate Breast cancer cells (FA receptors) [52]
CpG-STAT3 ASO Glioma microenvironment [53]
There are also reports that certain particles, such as sphingolipids, can affect exosomes cell uptake [56].

4.1. Cell Origin

Exosomes properties depend on source cells as they are closely related to overall cell processes necessary for metabolism. Crude exosomes can act as a double-agents for cancer development according to their source. Exosomes derived from normal cells will act as anticancer/antimetastatic in opposition to the reports that cancer exosomes will further increase proliferation, metastasis, and angiogenesis of cancer cells, promoting both in vitro and in vivo growth [39][57][58]. Careful selection of the exosomes source is crucial for proper exosomal-based DDS development. Due to their bilateral mode of action, exosomes can both increase or restrain the proper DDS mode of action.

4.2. Cancer Cells

Targeted delivery of cancer exosomes is mediated by expressed surface proteins, e.g., tetraspanins, interacting with different cell types in a different manner [59]. Tetraspanins also seem responsible for further malignancy spread; thus, therapeutic usage of this type of exosomes is not recommended [60]. It is worth mentioning that cancerous exosomes were used for stimulating the immune response to prevent the remaining cancer cells to grow after resection. The immune response was mediated by tumor-specific antigens presented at the surface of exosomes administered to patients after surgery [61][62]. Additionally, exosomes released from a dorsally implanted cell-graft composed of HT29 and HCT116 cell lines preferably locate at the stomach and intestine. This may be due to the origin of the cells composing the implant, which were derived from colorectal and colon tumors [63]. This effect is also supported by higher autologous exosomes uptake by the colorectal cell line than allogenic cell-derived exosomes both in vitro and in vivo. Latter studies also revealed an increased accumulation of autologous exosomes in the tumor site [64]. This effect can be overcome by expressing on the vesicles surface cancer-targeting particles such as integrins. Designed in this manner, exosomes will specifically target cells other than autologous cells, also increasing their uptake ratio [65].

4.3. Normal Cells

Exosomes isolated from immune cells are recognized as safe, serving as delivery vehicles, and were proposed for use as vehicles for drugs, vaccines, and immunotherapy. The most common types of cells are dendritic cells, macrophages, monocytes, B cells, and T cells. These cells’ native function regulates the immunologic response and, thus, exosomes derived from them will also target cells recognized as hostile, e.g., cancer cells. Dendritic-cells-derived exosomes contain numerous membrane moieties that can induce cancer cells’ death themself. Macrophage culture purity needs to be maintained at a high profile due to the potential influence of pathogens on exosomes properties and mode of action [66]. These exosomes also convert M2 macrophages to M1 type, increasing phagocytosis of tumor cells. Additionally, macrophage-derived exosomes stimulate the inflammatory response by the release of Th-1 type cytokines, which can be utilized in cancer vaccines, where exosomes may play the role of an adjuvant [67]. Human dermal fibroblasts are also used as an exosomes source [68].

4.4. Stem Cells

MSC-derived exosomes seem to be the most suitable immunologically for DDS design. These cells were isolated from various kinds of tissues such as adipose, liver, amniotic fluid, placenta, umbilical cord, and menstrual blood stem cells [68][69]. They do not express MHC antigens at the surface, thus evading phagocytosis and fast clearance from organisms [70]. Another advantage of MSCs is the production of exosomes in greater amounts than other cells. Currently, MSCs and dendritic cells are favored for use in clinical studies [71].

4.5. Plants, Fruits, and Milk

Due to the high cost and problems with the isolation of large amounts of exosomes from the mentioned cells, plants and fruits are currently recognized as promising exosomes sources [72]. In a similar manner, bovine milk is also an easily accessible, cost-effective source, containing significant amounts of microvesicles [73]. They are generally stable in acidic environments; thus, they are great candidates for oral drugs development [69]. There was no record of modifying plants or bovines in order to obtain modified exosomes.

References

  1. Ferreira, D.; Moreira, J.N.; Rodrigues, L.R. New advances in exosome-based targeted drug delivery systems. Crit. Rev. Oncol. Hematol. 2022, 172, 103628.
  2. Amiri, A.; Bagherifar, R.; Ansari Dezfouli, E.; Kiaie, S.H.; Jafari, R.; Ramezani, R. Exosomes as bio-inspired nanocarriers for RNA delivery: Preparation and applications. J. Transl. Med. 2022, 20, 125.
  3. Zhang, Y.; Bi, J.; Huang, J.; Tang, Y.; Du, S.; Li, P. Exosome: A review of its classification, isolation techniques, storage, diagnostic and targeted therapy applications. Int. J. Nanomed. 2020, 15, 6917–6934.
  4. Zhang, Y.; Liu, Y.; Liu, H.; Tang, W.H. Exosomes: Biogenesis, biologic function and clinical potential. Cell Biosci. 2019, 9, 19.
  5. Bunggulawa, E.J.; Wang, W.; Yin, T.; Wang, N.; Durkan, C.; Wang, Y.; Wang, G. Recent advancements in the use of exosomes as drug delivery systems. J. Nanobiotechnol. 2018, 16, 81.
  6. Patil, S.M.; Sawant, S.S.; Kunda, N.K. Exosomes as drug delivery systems: A brief overview and progress update. Eur. J. Pharm. Biopharm. 2020, 154, 259–269.
  7. Liang, Y.; Duan, L.; Lu, J.; Xia, J. Engineering exosomes for targeted drug delivery. Theranostics 2021, 11, 3183–3195.
  8. Gong, C.; Tian, J.; Wang, Z.; Gao, Y.; Wu, X.; Ding, X.; Qiang, L.; Li, G.; Han, Z.; Yuan, Y.; 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.
  9. Qi, H.; Liu, C.; Long, L.; Ren, Y.; Zhang, S.; Chang, X.; Qian, X.; Jia, H.; Zhao, J.; Sun, J.; et al. Blood exosomes endowed with magnetic and targeting properties for cancer therapy. ACS Nano 2016, 10, 3323–3333.
  10. Liang, G.; Zhu, Y.; Ali, D.J.; Tian, T.; Xu, H.; Si, K.; Sun, B.; Chen, B.; Xiao, Z. Engineered exosomes for targeted co-delivery of mir-21 inhibitor and chemotherapeutics to reverse drug resistance in colon cancer. J. Nanobiotechnol. 2020, 18, 10.
  11. Al-Nedawi, K.; Meehan, B.; Micallef, J.; Lhotak, V.; May, L.; Guha, A.; Rak, J. Intercellular transfer of the oncogenic receptor EGFRvIII by microvesicles derived from tumour cells. Nat. Cell Biol. 2008, 10, 619–624.
  12. van Niel, G.; Porto-Carreiro, I.; Simoes, S.; Raposo, G. Exosomes: A common pathway for a specialized function. J. Biochem. 2006, 140, 13–21.
  13. Lakkaraju, A.; Rodriguez-Boulan, E. Itinerant exosomes: Emerging roles in cell and tissue polarity. Trends Cell Biol. 2008, 18, 199–209.
  14. Schorey, J.S.; Bhatnagar, S. Exosome function: From tumor immunology to pathogen biology. Traffic 2008, 9, 871–881.
  15. Sharma, S.; Rasool, H.I.; Palanisamy, V.; Mathisen, C.; Schmidt, M.; Wong, D.T.; Gimzewski, J.K. Structural-mechanical characterization of nanoparticle exosomes in human saliva, using correlative afm, fesem, and force spectroscopy. ACS Nano 2010, 4, 1921–1926.
  16. Pan, B.T.; Teng, K.; Wu, C.; Adam, M.; Johnstone, R.M. Electron microscopic evidence for externalization of the transferrin receptor in vesicular form in sheep reticulocytes. J. Cell Biol. 1985, 101, 942–948.
  17. Johnstone, R.M. Exosomes biological significance: A concise review. Blood Cells Mol. Dis. 2006, 36, 315–321.
  18. Pisitkun, T.; Shen, R.-F.; Knepper, M.A. Identification and proteomic profiling of exosomes in human urine. Proc. Natl. Acad. Sci. USA 2004, 101, 13368–13373.
  19. Exocarta. Available online: http://www.exocarta.org/ (accessed on 8 September 2022).
  20. Vesiclepedia. Available online: http://microvesicles.org/ (accessed on 8 September 2022).
  21. Vlassov, A.V.; Magdaleno, S.; Setterquist, R.; Conrad, R. Exosomes: Current knowledge of their composition, biological functions, and diagnostic and therapeutic potentials. Biochim. Biophys. Acta—Gen. Subj. 2012, 1820, 940–948.
  22. Trajkovic, K.; Hsu, C.; Chiantia, S.; Rajendran, L.; Wenzel, D.; Wieland, F.; Schwille, P.; Brügger, B.; Simons, M. Ceramide triggers budding of exosome vesicles into multivesicular endosomes. Science 2008, 319, 1244–1247.
  23. Valadi, H.; Ekström, K.; Bossios, A.; Sjöstrand, M.; Lee, J.J.; Lötvall, J.O. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat. Cell Biol. 2007, 9, 654–659.
  24. Kosaka, N.; Izumi, H.; Sekine, K.; Ochiya, T. MicroRNA as a new immune-regulatory agent in breast milk. Silence 2010, 1, 7.
  25. Tanaka, Y.; Kamohara, H.; Kinoshita, K.; Kurashige, J.; Ishimoto, T.; Iwatsuki, M.; Watanabe, M.; Baba, H. Clinical impact of serum exosomal microRNA-21 as a clinical biomarker in human esophageal squamous cell carcinoma. Cancer 2013, 119, 1159–1167.
  26. Wang, H.; Hou, L.; Li, A.; Duan, Y.; Gao, H.; Song, X. Expression of serum exosomal microRNA-21 in human hepatocellular carcinoma. Biomed Res. Int. 2014, 2014, 864894.
  27. Machida, T.; Tomofuji, T.; Maruyama, T.; Yoneda, T.; Ekuni, D.; Azuma, T.; Miyai, H.; Mizuno, H.; Kato, H.; Tsutsumi, K.; et al. MiR-1246 and miR-4644 in salivary exosome as potential biomarkers for pancreatobiliary tract cancer. Oncol. Rep. 2016, 36, 2375–2381.
  28. Sun, J.; Aswath, K.; Schroeder, S.G.; Lippolis, J.D.; Reinhardt, T.A.; Sonstegard, T.S. MicroRNA expression profiles of bovine milk exosomes in response to staphylococcus aureus infection. BMC Genom. 2015, 16, 806.
  29. Huang, X.; Yuan, T.; Liang, M.; Du, M.; Xia, S.; Dittmar, R.; Wang, D.; See, W.; Costello, B.A.; Quevedo, F.; et al. Exosomal miR-1290 and miR-375 as prognostic markers in castration-resistant prostate cancer. Eur. Urol. 2015, 67, 33–41.
  30. Perez-Hernandez, J.; Martinez-Arroyo, O.; Ortega, A.; Galera, M.; Solis-Salguero, M.A.; Chaves, F.J.; Redon, J.; Forner, M.J.; Cortes, R. Urinary exosomal miR-146a as a marker of albuminuria, activity changes and disease flares in lupus nephritis. J. Nephrol. 2021, 34, 1157–1167.
  31. Wang, B.; Mao, J.; Wang, B.; Wang, L.; Wen, H.; Xu, L.; Fu, J.; Yang, H. Exosomal miR-1910-3p promotes proliferation, metastasis, and autophagy of breast cancer cells by targeting MTMR3 and activating the NF-κB signaling pathway. Cancer Lett. 2020, 489, 87–99.
  32. Yang, H.; Fu, H.; Wang, B.; Zhang, X.; Mao, J.; Li, X.; Wang, M.; Sun, Z.; Qian, H.; Xu, W. Exosomal miR-423-5p targets SUFU to promote cancer growth and metastasis and serves as a novel marker for gastric cancer. Mol. Carcinog. 2018, 57, 1223–1236.
  33. Sun, S.; Chen, H.; Xu, C.; Zhang, Y.; Zhang, Q.; Chen, L.; Ding, Q.; Deng, Z. Exosomal miR-106b serves as a novel marker for lung cancer and promotes cancer metastasis via targeting PTEN. Life Sci. 2020, 244, 117297.
  34. Zhan, Q.; Yi, K.; Cui, X.; Li, X.; Yang, S.; Wang, Q.; Fang, C.; Tan, Y.; Li, L.; Xu, C.; et al. Blood exosomes-based targeted delivery of cPLA2 siRNA and metformin to modulate glioblastoma energy metabolism for tailoring personalized therapy. Neuro Oncol. 2022, 24, 1871–1883.
  35. He, J.; Ren, W.; Wang, W.; Han, W.; Jiang, L.; Zhang, D.; Guo, M. Exosomal targeting and its potential clinical application. Drug Deliv. Transl. Res. 2021, 12, 2385–2402.
  36. Pêche, H.; Heslan, M.; Usal, C.; Amigorena, S.; Cuturi, M.C. Presentation of donor major histocompatibility complex antigens by bone marrow dendritic cell-derived exosomes modulates allograft rejection. Transplantation 2003, 76, 1503–1510.
  37. Hao, S.; Bai, O.; Yuan, J.; Qureshi, M.; Xiang, J. Dendritic cell-derived exosomes stimulate stronger CD8+ CTL responses and antitumor immunity than tumor cell-derived exosomes. Cell. Mol. Immunol. 2006, 3, 205–211.
  38. Yi, Y.W.; Lee, J.H.; Kim, S.-Y.; Pack, C.-G.; Ha, D.H.; Park, S.R.; Youn, J.; Cho, B.S. Advances in analysis of biodistribution of exosomes by molecular imaging. Int. J. Mol. Sci. 2020, 21, 665.
  39. Fabbri, M.; Paone, A.; Calore, F.; Galli, R.; Croce, C.M. A new role for microRNAs, as ligands of Toll-like receptors. RNA Biol. 2013, 10, 169–174.
  40. Xiang, X.; Poliakov, A.; Liu, C.; Liu, Y.; Deng, Z.; Wang, J.; Cheng, Z.; Shah, S.V.; Wang, G.-J.; Zhang, L.; et al. Induction of myeloid-derived suppressor cells by tumor exosomes. Int. J. Cancer 2009, 124, 2621–2633.
  41. Mittal, R.; Bencie, N.; Langlie, J.; Mittal, J.; Eshraghi, A.A. Exosomes as drug delivery vehicles and biomarkers for neurological and auditory systems. J. Cell. Physiol. 2021, 236, 8035–8049.
  42. Wiklander, O.P.B.; Nordin, J.Z.; O’Loughlin, A.; Gustafsson, Y.; Corso, G.; Mäger, I.; Vader, P.; Lee, Y.; Sork, H.; Seow, Y.; et al. Extracellular vesicle in vivo biodistribution is determined by cell source, route of administration and targeting. J. Extracell. Vesicles 2015, 4, 26316.
  43. 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, 754–763.
  44. Rech, J.; Rogóż, W.; Borecka, A.; Turek, A. Application of fibrin in tissue engineering. Achievements and perspectives. Postep. Hig. Med. Dosw. 2021, 75, 749–761.
  45. Cui, G.; Guo, H.; Li, H.; Zhai, Y.; Gong, Z.; Wu, J.; Liu, J.; Dong, Y.; Hou, S.; Liu, J. RVG-modified exosomes derived from mesenchymal stem cells rescue memory deficits by regulating inflammatory responses in a mouse model of Alzheimer’s disease. Immun. Ageing 2019, 16, 10.
  46. Escrevente, C.; Keller, S.; Altevogt, P.; Costa, J. Interaction and uptake of exosomes by ovarian cancer cells. BMC Cancer 2011, 11, 108.
  47. Hoshino, A.; Costa-Silva, B.; Shen, T.-L.; Rodrigues, G.; Hashimoto, A.; Tesic Mark, M.; Molina, H.; Kohsaka, S.; Di Giannatale, A.; Ceder, S.; et al. Tumour exosome integrins determine organotropic metastasis. Nature 2015, 527, 329–335.
  48. Zhao, L.; Gu, C.; Gan, Y.; Shao, L.; Chen, H.; Zhu, H. Exosome-mediated siRNA delivery to suppress postoperative breast cancer metastasis. J. Control. Release 2020, 318, 1–15.
  49. Laulagnier, K.; Javalet, C.; Hemming, F.J.; Chivet, M.; Lachenal, G.; Blot, B.; Chatellard, C.; Sadoul, R. Amyloid precursor protein products concentrate in a subset of exosomes specifically endocytosed by neurons. Cell. Mol. Life Sci. 2018, 75, 757–773.
  50. Toda, Y.; Takata, K.; Nakagawa, Y.; Kawakami, H.; Fujioka, S.; Kobayashi, K.; Hattori, Y.; Kitamura, Y.; Akaji, K.; Ashihara, E. Effective internalization of U251-MG-secreted exosomes into cancer cells and characterization of their lipid components. Biochem. Biophys. Res. Commun. 2015, 456, 768–773.
  51. Parolini, I.; Federici, C.; Raggi, C.; Lugini, L.; Palleschi, S.; De Milito, A.; Coscia, C.; Iessi, E.; Logozzi, M.; Molinari, A.; et al. Microenvironmental pH is a key factor for exosome traffic in tumor cells. J. Biol. Chem. 2009, 284, 34211–34222.
  52. Yu, M.; Gai, C.; Li, Z.; Ding, D.; Zheng, J.; Zhang, W.; Lv, S.; Li, W. Targeted exosome-encapsulated erastin induced ferroptosis in triple negative breast cancer cells. Cancer Sci. 2019, 110, 3173–3182.
  53. Adamus, T.; Hung, C.-Y.; Yu, C.; Kang, E.; Hammad, M.; Flores, L.; Nechaev, S.; Zhang, Q.; Gonzaga, J.M.; Muthaiyah, K.; et al. Glioma-targeted delivery of exosome-encapsulated antisense oligonucleotides using neural stem cells. Mol. Ther.—Nucleic Acids 2022, 27, 611–620.
  54. Parada, N.; Romero-Trujillo, A.; Georges, N.; Alcayaga-Miranda, F. Camouflage strategies for therapeutic exosomes evasion from phagocytosis. J. Adv. Res. 2021, 31, 61–74.
  55. Haney, M.J.; Klyachko, N.L.; Zhao, Y.; Gupta, R.; Plotnikova, E.G.; He, Z.; Patel, T.; Piroyan, A.; Sokolsky, M.; Kabanov, A.V.; et al. Exosomes as drug delivery vehicles for Parkinson’s disease therapy. J. Control. Release 2015, 207, 18–30.
  56. Charoenviriyakul, C.; Takahashi, Y.; Morishita, M.; Matsumoto, A.; Nishikawa, M.; Takakura, Y. Cell type-specific and common characteristics of exosomes derived from mouse cell lines: Yield, physicochemical properties, and pharmacokinetics. Eur. J. Pharm. Sci. 2017, 96, 316–322.
  57. Keller, S.; König, A.-K.; Marmé, F.; Runz, S.; Wolterink, S.; Koensgen, D.; Mustea, A.; Sehouli, J.; Altevogt, P. Systemic presence and tumor-growth promoting effect of ovarian carcinoma released exosomes. Cancer Lett. 2009, 278, 73–81.
  58. Umezu, T.; Tadokoro, H.; Azuma, K.; Yoshizawa, S.; Ohyashiki, K.; Ohyashiki, J.H. Exosomal miR-135b shed from hypoxic multiple myeloma cells enhances angiogenesis by targeting factor-inhibiting HIF-1. Blood 2014, 124, 3748–3757.
  59. Rana, S.; Yue, S.; Stadel, D.; Zöller, M. Toward tailored exosomes: The exosomal tetraspanin web contributes to target cell selection. Int. J. Biochem. Cell Biol. 2012, 44, 1574–1584.
  60. Zomer, A.; Maynard, C.; Verweij, F.J.; Kamermans, A.; Schäfer, R.; Beerling, E.; Schiffelers, R.M.; de Wit, E.; Berenguer, J.; Ellenbroek, S.I.J.; et al. In vivo imaging reveals extracellular vesicle-mediated phenocopying of metastatic behavior. Cell 2015, 161, 1046–1057.
  61. Wolfers, J.; Lozier, A.; Raposo, G.; Regnault, A.; Théry, C.; Masurier, C.; Flament, C.; Pouzieux, S.; Faure, F.; Tursz, T.; et al. Tumor-derived exosomes are a source of shared tumor rejection antigens for CTL cross-priming. Nat. Med. 2001, 7, 297–303.
  62. ClinicalTrials. Available online: https://clinicaltrials.gov/ct2/show/NCT01550523 (accessed on 15 November 2022).
  63. Hikita, T.; Miyata, M.; Watanabe, R.; Oneyama, C. Sensitive and rapid quantification of exosomes by fusing luciferase to exosome marker proteins. Sci. Rep. 2018, 8, 14035.
  64. Emam, S.E.; Abu Lila, A.S.; Elsadek, N.E.; Ando, H.; Shimizu, T.; Okuhira, K.; Ishima, Y.; Mahdy, M.A.; Ghazy, F.S.; Ishida, T. Cancer cell-type tropism is one of crucial determinants for the efficient systemic delivery of cancer cell-derived exosomes to tumor tissues. Eur. J. Pharm. Biopharm. 2019, 145, 27–34.
  65. Nie, H.; Xie, X.; Zhang, D.; Zhou, Y.; Li, B.; Li, F.; Li, F.; Cheng, Y.; Mei, H.; Meng, H.; et al. Use of lung-specific exosomes for miRNA-126 delivery in non-small cell lung cancer. Nanoscale 2020, 12, 877–887.
  66. Arteaga-Blanco, L.A.; Bou-Habib, D.C. The role of extracellular vesicles from human macrophages on host-pathogen interaction. Int. J. Mol. Sci. 2021, 22, 10262.
  67. Xia, Y.; Rao, L.; Yao, H.; Wang, Z.; Ning, P.; Chen, X. Engineering macrophages for cancer immunotherapy and drug delivery. Adv. Mater. 2020, 32, 2002054.
  68. Rosenberger, L.; Ezquer, M.; Lillo-Vera, F.; Pedraza, P.L.; Ortúzar, M.I.; González, P.L.; Figueroa-Valdés, A.I.; Cuenca, J.; Ezquer, F.; Khoury, M.; et al. Stem cell exosomes inhibit angiogenesis and tumor growth of oral squamous cell carcinoma. Sci. Rep. 2019, 9, 663.
  69. Meng, W.; He, C.; Hao, Y.; Wang, L.; Li, L.; Zhu, G. Prospects and challenges of extracellular vesicle-based drug delivery system: Considering cell source. Drug Deliv. 2020, 27, 585–598.
  70. Moon, B.; Chang, S. Exosome as a delivery vehicle for cancer therapy. Cells 2022, 11, 316.
  71. Rezaie, J.; Feghhi, M.; Etemadi, T. A review on exosomes application in clinical trials: Perspective, questions, and challenges. Cell Commun. Signal. 2022, 20, 145.
  72. Roshancheshm, S.; Asadi, A.; Khoshnazar, S.M.; Abdolmaleki, A.; Khudhur, O.O.; Sh, W.S. Application of natural and modified exosomes as a drug delivery system. Nanomed. J. 2022, 9, 192–204.
  73. Mehryab, F.; Rabbani, S.; Shahhosseini, S.; Shekari, F.; Fatahi, Y.; Baharvand, H.; Haeri, A. Exosomes as a next-generation drug delivery system: An update on drug loading approaches, characterization, and clinical application challenges. Acta Biomater. 2020, 113, 42–62.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , ,
View Times: 505
Entry Collection: Biopharmaceuticals Technology
Revisions: 2 times (View History)
Update Date: 19 Dec 2022
1000/1000
Video Production Service