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 -- 1554 2022-09-27 07:31:24 |
2 format correct Meta information modification 1554 2022-09-27 07:59:16 |

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.
Frolova, L.;  Li, I.T.S. Engineering Extracellular Vesicles for Targeted Drug Delivery. Encyclopedia. Available online: https://encyclopedia.pub/entry/27627 (accessed on 06 July 2024).
Frolova L,  Li ITS. Engineering Extracellular Vesicles for Targeted Drug Delivery. Encyclopedia. Available at: https://encyclopedia.pub/entry/27627. Accessed July 06, 2024.
Frolova, Liubov, Isaac T. S. Li. "Engineering Extracellular Vesicles for Targeted Drug Delivery" Encyclopedia, https://encyclopedia.pub/entry/27627 (accessed July 06, 2024).
Frolova, L., & Li, I.T.S. (2022, September 27). Engineering Extracellular Vesicles for Targeted Drug Delivery. In Encyclopedia. https://encyclopedia.pub/entry/27627
Frolova, Liubov and Isaac T. S. Li. "Engineering Extracellular Vesicles for Targeted Drug Delivery." Encyclopedia. Web. 27 September, 2022.
Engineering Extracellular Vesicles for Targeted Drug Delivery
Edit

Extracellular vesicles (EVs) are membranous nanosized particles produced by nearly all cell types, including eukaryotic and prokaryotic cells, and they carry their parent cell’s cytosolic components in their lumen, including RNA and various proteins. EVs can be broadly classified into three types by their biogenesis pathway: exosomes, microvesicles (ectosomes) and apoptotic bodies. EVs can be decorated with surface molecules to enhance their targeting abilities. This can be accomplished by directly attaching targeting moieties to the EV surface or modifying EV-producing cells.

extracellular vesicle exosome drug delivery endocytosis drug targeting

1. Targeting EVs by Modifying Parent Cells

Modifying parent cells is a common method to obtain EVs with particular targeting properties and is superior to direct modification of exosomal surface in terms of the stability of the targeting moiety. In this method, a gene encoding the targeting proteins is inserted into donor cells, and the cells then release EVs carrying those proteins via the natural biogenesis pathways. If certain targeting moieties are not naturally found on EV membranes, cells can be made to express the desired moiety fused to an EV membrane component.
Lysosomal-associated membrane protein 2b (Lamp2b) is one of the most commonly used exosomal pedestals to attach guiding moieties [1][2][3][4]. For example, Tian et al. utilized Lamp2b fused to αv integrin-specific iRGD peptide to target αv integrin-positive breast cancer cells in vitro and in vivo [2]. A dramatic increase in cellular uptake was observed for iRGD-decorated exosomes compared to control exosomes (95.4% vs. 35.0%) [2]. Designer EVs can be used to overcome one of the main challenges faced by traditional therapeutics—crossing the BBB to deliver drugs to the brain. Again, Lamp2b acted as an exosomal pedestal to fuse brain targeting moiety in a study by Alvarez-Erviti et al. They accomplished the targeting of short interfering (si)RNA to the brain in mice by engineering dendritic cells to express Lamp2b conjugated to the neuron-specific rabies viral glycoprotein (RVG) peptide [1].
The designs of protein-targeting constructs that get incorporated into therapeutic EVs can become rather complicated. Ohno et al. used cloned tumour-targeting peptides (EGF and its less mitogenic alternative GE11) into pDisplay vector, which already contained hemagglutinin, myc-tag and platelet-derived growth factor receptor (PDGFR) [5]. Human embryonic kidney cell line 293 (HEK293) cells were then transfected with the pDisplay vector, thus allowing for the incorporation of targeting peptides into exosomes with the PDGFR acting as the carrying pedestal [5]. The targeting modifications with EGF and GE11 peptides increased the uptake of exosomes by breast cancer cells which typically overexpress EGF receptors [5]. At the same time, anti-hemagglutinin and anti-Myc-tag antibodies were used to confirm the expression of EGF and GE11 in exosomes [5]. Johnsen et al. have published a very comprehensive review of EVs as drug delivery vehicles, covering the use of targeting peptides to enhance the precision of EV-based therapeutics [6].
Besides targeting peptides, antibodies and nanobodies have been installed on EV surfaces to improve targeting. In one study, EV-producing cells were transfected with a vector encoding for anti-EGFR nanobodies fused to glycosylphosphatidylinositol (GPI) peptides to target cancer cells [7]. Lipid raft-associated lipids and proteins, including GPI, are naturally enriched on EV membranes, making them ideal for conjugating a targeting ligand [8]. Lactadherin was also used as an anchor for cancer-targeting moiety—single chain variable fragments (scFv) with an affinity to human epidermal growth factor receptor (EGFR) 2 overexpressed in breast cancer cells [9]. This approach was justified as lactadherin associates with the phosphatidylserine enriched in EV membranes.
While peptides and antibodies/nanobodies are the most popular choices for EV target guidance, other more imaginative approaches have also been employed. For example, pseudotyping—a method to change the tropism of viruses by packaging the genetic components of one virus into the envelope proteins of a different virus—has been applied to engineering targeted exosomes [10]. Meyer et al. expressed vesicular stomatitis virus glycoprotein (VSVG), which is frequently used for pseudotyping retroviruses and known for its broad tropism, in HEK293 cells [10]. The resulting VSVG-pseudotyped exosomes were incubated with multiple cell lines, and enhanced uptake by those cells was shown compared to controls [10].
An interesting direction in EV targeting is to use targeting moieties that also have a therapeutic effect. Jiang et al. induced overexpression of tumour necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) in the membranes of donor cells which subsequently got incorporated into exosomes loaded with an anti-cancer agent triptolide [11]. In this study, TRAIL acted not only as a targeting ligand for death receptor 5, which is abundant in cancer cells, but also induced apoptosis in cancer cells, thus amplifying the therapeutic effect of triptolide loaded into exosomes [11]. In a different study, MHC-II, a major histocompatibility complex molecule normally only present on professional antigen-presenting cells, was overexpressed in murine melanoma cells [12]. The resulting MHC-II-enriched exosomes not only showed increased targeting towards T cells, but also enhanced the immunological response of the type 1 T helper cell (TH1) against cancer cells [12].
Supplying EVs with targeting properties via genetic modification of parent cells is an effective approach. However, it is not suitable for personalized medicine applications as it is difficult to apply this approach to patients’ own cells. It is also time-consuming, hard to scale for mass production and limiting as only genetically encodable targeting moieties may be used. Moreover, some targeting moieties tend to be expressed incorrectly and are quickly degraded in producer cells, affecting the resulting EVs’ targeting efficiency [13].

2. Modifying EV Surface for Improved Targeting

Direct chemical modification of EVs allows for a wider selection of targeting ligands and is more suited to personalized medicine applications, as EVs can be isolated from patient’s own body fluids and later decorated with chosen guiding moieties. Further, targeting ligands can be attached to isolated EVs in a very controlled manner which is not achievable with the parental cell modification approach.
Targeting peptides and antibodies/nanobodies are most commonly used to achieve efficient targeting of EVs post-isolation. An interesting approach was suggested by Ye et al., who used a multifunctional peptide to target EVs to glioblastoma cells [14]. The peptide contained a sequence which targeted it to the low-density lipoprotein receptor expressed on the glioblastoma cells, as well as an apoptosis-inducing sequence [14]. This therapeutic/targeting peptide amplified the effectiveness of the chemotherapy agent loaded into EVs and ensured successful delivery over the BBB in a mouse model [14]. Glioma cells were also targeted by Jia et al. via the conjugation of neuropilin-1-targeted peptide to exosomes via click chemistry [15]. The exosomes were loaded with superparamagnetic iron oxide nanoparticles and curcumin to enable both glioma imaging and treatment [15].
Anti-EGFR nanobodies fused to lactadherin, which bind to the phosphatidylserine in EVs post-isolation, have been used to target EGFR-positive tumour cells [16]. Similarly, anti-Her2 single-chain variable fragment fused to lactadherin was conjugated to phosphatidylserine-enriched EVs to target Her2-positive cancer cells [17]. A different approach to surface modification of EVs with nanobodies was adopted by Koojmans et al. who mixed EVs with micelles containing PEG and EGFR nanobodies [18]. This resulted in the emergence of PEGylated EVs targeted to EGFR-positive cancer cells [18].
More out-of-the-box approaches to direct modification of EVs for targeted drug delivery have also been tested. For instance, both DNA and RNA aptamers (single strands of nucleic acid that can bind to specific targets) have been used to direct EVs towards tumours. Cholesterol, naturally present in EV membranes, was used to conjugate AS1411, a DNA aptamer with a high affinity to nucleolin typically overexpressed in breast cancer cells [19]. AS1411-decorated EVs displayed better tumour-targeting abilities and had an added therapeutic effect as AS1411 is known to inhibit tumour activity [19]. An RNA aptamer targeted at prostate-specific membrane antigen (PSMA) has also been loaded into EV membranes to gain directional control [20]. Interestingly, the orientation of this arrow-shaped aptamer could be altered (with either the arrowhead or the arrow tail facing the membrane-anchoring cholesterol) to achieve either better targeting or better cargo loading into EVs [20].
Even actual magnets have been used to direct exosomes towards tumours. Qi at al. decorated transferrin receptors of blood-derived exosomes with magnetic nanoparticles and then guided them towards murine tumours using external magnets [21].
A niche and underexplored method for functionalizing EVs with targeting moieties is their fusion with liposomes decorated with targeting peptides or antibodies [22]. For example, Li et al. fused cancer exosomes with liposomes decorated with tumour-targeting peptide cRGD and loaded the resulting hybrid with a chemotherapy drug. The addition of the targeting peptide amplified the natural homing ability of cancer exosomes and resulted in the efficient delivery of the drug in vitro and in vivo [23]. A different study used an exosome-liposome hybrid to treat diabetic peripheral neuropathy. Singh et al. fused exosomes from bone marrow mesenchymal stromal cells with liposomes containing polypyrrole nanoparticles, thus combining stem cell therapy with electrical stimulation to achieve a therapeutic effect. Polypyrrole was selected because it is a conducting polymer and served to target the neurons’ electrical stimulation [24]. Despite some success, there are very few studies using fusion liposomes to achieve the targeting of exosomes.
Overall, the direct EV modification approach allows for a wider selection of targeting ligands and greater freedom in choosing chemical tools for their conjugation. These targeting moieties often have therapeutic and even diagnostic modalities that amplify the effect of loaded cargo drugs. Still, direct modification of EVs is challenging as the reaction conditions must be adapted to preserve EV membranes, prevent aggregation and ensure sufficient density of targeting ligands on EV surfaces.

References

  1. Alvarez-Erviti, L.; Seow, Y.; Yin, H.; Betts, C.; Lakhal, S.; Wood, M.J.A. Delivery of SiRNA to the Mouse Brain by Systemic Injection of Targeted Exosomes. Nat. Biotechnol. 2011, 29, 341–345.
  2. Tian, Y.; Li, S.; Song, J.; Ji, T.; Zhu, M.; Anderson, G.J.; Wei, J.; Nie, G. A Doxorubicin Delivery Platform Using Engineered Natural Membrane Vesicle Exosomes for Targeted Tumor Therapy. Biomaterials 2014, 35, 2383–2390.
  3. Bai, J.; Duan, J.; Liu, R.; Du, Y.; Luo, Q.; Cui, Y.; Su, Z.; Xu, J.; Xie, Y.; Lu, W. Engineered Targeting TLyp-1 Exosomes as Gene Therapy Vectors for Efficient Delivery of SiRNA into Lung Cancer Cells. Asian J. Pharm. Sci. 2020, 15, 461–471.
  4. Liang, Y.; Xu, X.; Li, X.; Xiong, J.; Li, B.; Duan, L.; Wang, D.; Xia, J. Chondrocyte-Targeted MicroRNA Delivery by Engineered Exosomes toward a Cell-Free Osteoarthritis Therapy. ACS Appl. Mater. Interfaces 2020, 12, 36938–36947.
  5. Ohno, S.; Takanashi, M.; Sudo, K.; Ueda, S.; Ishikawa, A.; Matsuyama, N.; Fujita, K.; Mizutani, T.; Ohgi, T.; Ochiya, T.; et al. Systemically Injected Exosomes Targeted to EGFR Deliver Antitumor MicroRNA to Breast Cancer Cells. Mol. Ther. J. Am. Soc. Gene Ther. 2013, 21, 185–191.
  6. Johnsen, K.B.; Gudbergsson, J.M.; Skov, M.N.; Pilgaard, L.; Moos, T.; Duroux, M. A Comprehensive Overview of Exosomes as Drug Delivery Vehicles—Endogenous Nanocarriers for Targeted Cancer Therapy. Biochim. Biophys. Acta BBA-Rev. Cancer 2014, 1846, 75–87.
  7. Kooijmans, S.A.A.; Aleza, C.G.; Roffler, S.R.; van Solinge, W.W.; Vader, P.; Schiffelers, R.M. Display of GPI-Anchored Anti-EGFR Nanobodies on Extracellular Vesicles Promotes Tumour Cell Targeting. J. Extracell. Vesicles 2016, 5, 31053.
  8. de Gassart, A.; Geminard, C.; Fevrier, B.; Raposo, G.; Vidal, M. Lipid Raft-Associated Protein Sorting in Exosomes. Blood 2003, 102, 4336–4344.
  9. Longatti, A.; Schindler, C.; Collinson, A.; Jenkinson, L.; Matthews, C.; Fitzpatrick, L.; Blundy, M.; Minter, R.; Vaughan, T.; Shaw, M.; et al. High Affinity Single-Chain Variable Fragments Are Specific and Versatile Targeting Motifs for Extracellular Vesicles. Nanoscale 2018, 10, 14230–14244.
  10. 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.
  11. Jiang, L.; Gu, Y.; Du, Y.; Tang, X.; Wu, X.; Liu, J. Engineering Exosomes Endowed with Targeted Delivery of Triptolide for Malignant Melanoma Therapy. ACS Appl. Mater. Interfaces 2021, 13, 42411–42428.
  12. Lee, Y.S.; Kim, S.H.; Cho, J.A.; Kim, C.W. Introduction of the CIITA Gene into Tumor Cells Produces Exosomes with Enhanced Anti-Tumor Effects. Exp. Mol. Med. 2011, 43, 281–290.
  13. Hung, M.E.; Leonard, J.N. Stabilization of Exosome-Targeting Peptides via Engineered Glycosylation. J. Biol. Chem. 2015, 290, 8166–8172.
  14. Ye, Z.; Zhang, T.; He, W.; Jin, H.; Liu, C.; Yang, Z.; Ren, J. Methotrexate-Loaded Extracellular Vesicles Functionalized with Therapeutic and Targeted Peptides for the Treatment of Glioblastoma Multiforme. ACS Appl. Mater. Interfaces 2018, 10, 12341–12350.
  15. Jia, G.; Han, Y.; An, Y.; Ding, Y.; He, C.; Wang, X.; Tang, Q. NRP-1 Targeted and Cargo-Loaded Exosomes Facilitate Simultaneous Imaging and Therapy of Glioma in Vitro and in Vivo. Biomaterials 2018, 178, 302–316.
  16. Kooijmans, S.A.A.; Gitz-Francois, J.J.J.M.; Schiffelers, R.M.; Vader, P. Recombinant Phosphatidylserine-Binding Nanobodies for Targeting of Extracellular Vesicles to Tumor Cells: A Plug-and-Play Approach. Nanoscale 2018, 10, 2413–2426.
  17. Wang, J.-H.; Forterre, A.V.; Zhao, J.; Frimannsson, D.O.; Delcayre, A.; Antes, T.J.; Efron, B.; Jeffrey, S.S.; Pegram, M.D.; Matin, A.C. Anti-HER2 ScFv-Directed Extracellular Vesicle-Mediated MRNA-Based Gene Delivery Inhibits Growth of HER2-Positive Human Breast Tumor Xenografts by Prodrug Activation. Mol. Cancer Ther. 2018, 17, 1133–1142.
  18. Kooijmans, S.A.A.; Fliervoet, L.A.L.; van der Meel, R.; Fens, M.H.A.M.; Heijnen, H.F.G.; van Bergen en Henegouwen, P.M.P.; Vader, P.; Schiffelers, R.M. PEGylated and Targeted Extracellular Vesicles Display Enhanced Cell Specificity and Circulation Time. J. Controlled Release 2016, 224, 77–85.
  19. Wang, Y.; Chen, X.; Tian, B.; Liu, J.; Yang, L.; Zeng, L.; Chen, T.; Hong, A.; Wang, X. Nucleolin-Targeted Extracellular Vesicles as a Versatile Platform for Biologics Delivery to Breast Cancer. Theranostics 2017, 7, 1360–1372.
  20. Pi, F.; Binzel, D.W.; Lee, T.J.; Li, Z.; Sun, M.; Rychahou, P.; Li, H.; Haque, F.; Wang, S.; Croce, C.M.; et al. Nanoparticle Orientation to Control RNA Loading and Ligand Display on Extracellular Vesicles for Cancer Regression. Nat. Nanotechnol. 2018, 13, 82–89.
  21. 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.
  22. Sato, Y.T.; Umezaki, K.; Sawada, S.; Mukai, S.; Sasaki, Y.; Harada, N.; Shiku, H.; Akiyoshi, K. Engineering Hybrid Exosomes by Membrane Fusion with Liposomes. Sci. Rep. 2016, 6, 21933.
  23. Li, L.; He, D.; Guo, Q.; Zhang, Z.; Ru, D.; Wang, L.; Gong, K.; Liu, F.; Duan, Y.; Li, H. Exosome-Liposome Hybrid Nanoparticle Codelivery of TP and MiR497 Conspicuously Overcomes Chemoresistant Ovarian Cancer. J. Nanobiotechnology 2022, 20, 50.
  24. Singh, A.; Raghav, A.; Shiekh, P.A.; Kumar, A. Transplantation of Engineered Exosomes Derived from Bone Marrow Mesenchymal Stromal Cells Ameliorate Diabetic Peripheral Neuropathy under Electrical Stimulation. Bioact. Mater. 2021, 6, 2231–2249.
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: 457
Revisions: 2 times (View History)
Update Date: 27 Sep 2022
1000/1000
Video Production Service