Next Article in Journal
Optical and Scanning Electron Microscopy Thrombus Findings in Patients with STEMI Undergoing Primary Versus Rescue PCI
Previous Article in Journal
Sweet Relief? Short-Term Post-Traumatic High-Sucrose Intake Attenuates Acute but Not Long-Term Fear Responses in Mice
Previous Article in Special Issue
Expression Analysis of let-7a-5p and miR-21-3p in Extracellular Vesicles Derived from Serum of NSCLC Patients
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Editorial of the Special Issue: Extracellular Vesicles and Exosomes as Therapeutic Agents

by
David J. Rademacher
Department of Microbiology and Immunology and Core Microscopy Facility, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA
Biomedicines 2025, 13(9), 2234; https://doi.org/10.3390/biomedicines13092234
Submission received: 4 September 2025 / Accepted: 9 September 2025 / Published: 11 September 2025
(This article belongs to the Special Issue Extracellular Vesicles and Exosomes as Therapeutic Agents)

1. Introduction

Membrane-enclosed particles, known as extracellular vesicles (EVs), are ubiquitously present in organisms, including animals, plants, and microorganisms [1,2]. EVs are classified according to their size and origin as apoptotic bodies, microvesicles, or exosomes (EXs) [1,2,3]. EXs, the most extensively studied EVs, are enclosed in a single phospholipid bilayer and are secreted by all cell types [1,3,4,5]. EXs are usually 30 to 150 nm in diameter and are formed by fusion of the multivesicular body and the inward invagination of the endosomal membrane [1,3,4,5]. EXs are present in body fluids such as saliva, amniotic fluid, breast milk, urine, and plasma [6]. They are essential for intercellular communication because they change the functions of recipient cells by delivering a variety of biologically active cargo, including proteins, lipids, and nucleic acids [7,8].
The use of EXs as a delivery strategy for therapeutic cargo has become increasingly popular, in part, because they overcome many of the challenges encountered by other approaches. EXs exhibit a high degree of stability and biocompatibility with minimal immunogenicity [9]. The delivery-to-target efficiency is higher for EXs than conventional approaches because they can traverse biological barriers, such as the blood-brain barrier (BBB) [10,11]. EXs can selectively target tissues or cells and reduce off-target effects because of their inherent tropism based on their cells of origin [12]. EXs provide a protective environment for the encapsulated cargo, safeguarding it from degradation [13]. Furthermore, because EXs are the primary way that stem cells exert their biological effects, the use of stem cell-derived EXs preserves the therapeutic potential of stem cells while avoiding any potential harm from the cells [14,15]. Importantly, systemic administration of EXs derived from a variety of cell types often results in their primary accumulation in the liver and spleen, with less than 1% of that administered reaching the desired target [12,16]. Consequently, several engineering approaches have been developed to improve targeting and increase the accumulation of EXs in the target tissue and/or cells.

2. Recent Developments in the Use of Extracellular Vesicles and Exosomes as Therapeutic Delivery Systems

2.1. Genetic Engineering of EXs

Genetic engineering has been used to modify exosomal surface proteins to better target EXs to tissues and cell types of interest, since even slight variations in exosomal tetraspanin complexes have a significant impact on target cell selection. Transfecting genes expressing a targeting moiety with exosomal membrane components is one method of genetically altering EX-producing cells. Through the natural EX biogenesis process, the cells transfected with these vectors produce surface-modified EXs that express the targeting moieties. EXs produced from genetically engineered cells stably display the introduced targeting moiety on their surface [17]. For example, Wang and colleagues [18] genetically engineered stem cell-derived EXs loaded with a ferroptosis inhibitor to target M2 microglia. The genetically engineered EXs exhibited M2 microglia targeting specificity in vitro and in vivo, inhibited ferroptosis in M2 microglia, and improved neurological function in ischemic stroke-affected mice [18]. A fusion protein, consisting of rabies virus glycoprotein (RVG), a protein that binds nicotinic acetylcholine receptors, and lysosomal membrane-associated protein 2B, an exosomal membrane protein, was transfected into 293T cells. The cells produced EXs with RVG embedded in the exosomal membrane [19]. As neurons and glia express receptors that bind to RVG on their cell surface, systemic administration of RVG-expressing EXs resulted in a two-fold increase in brain EX accumulation compared to systemic administration of non-engineered EXs [19].

2.2. Chemical Modifications of EXs

To make therapeutic EXs more targetable, their surface can be directly engineered through chemical modifications. One such approach is click chemistry. Click chemistry utilizes covalent interactions between an alkyne and an azide residue to form a stable triazole linkage, which can be applied to attach targeting moieties on the surface of EXs [20,21]. For example, Jia and colleagues [22] used click chemistry to conjugate the RGERPPR peptide (RGE) to the exosomal membrane. The engineered EXs delivered therapeutic cargo to intracranial tumors because RGE binds specifically to neuropilin-1 (NRP-1), which is overexpressed in glioma cells and is either not expressed at all or expressed at a low level in normal neurons and other tissues [22]. Chen and coworkers [23] used click chemistry to attach a targeting peptide to the surface of macrophage-derived EXs. These engineered EXs targeted bone tissue and released anti-microbial peptides, thereby relieving osteomyelitis without adverse effects in a rat model [23].

2.3. Combinatorial Approaches

Hybrid Nanoparticles

As an alternative to genetic engineering of parent cells or EXs and chemical modification of EXs, targeting ligands conjugated to polyethylene glycol (PEG) have been used to produce EXs in which the targeting ligand is expressed in the exosomal membrane. For example, nanobodies specific for epidermal growth factor receptor (EGFR), upregulated in a variety of solid tumors, were conjugated to phospholipid-PEG derivatives to produce nanobody-PEG-micelles [24]. Nanobody-PEG-micelles were expressed in the exosomal membrane when they were incubated with EXs; this had no effect on the size distribution, protein composition, or morphology of EXs. The PEGylated and targeted EXs increased the binding to tumor cells overexpressing EGFR and were retained in the circulation for six times longer than unmodified EXs [24].
Diacyl-chain phospholipids self-assemble in aqueous solutions to form spherical vesicles called liposomes [25]. Liposomes expressing tLyp-1 (CGNKRTR), which has a high affinity to NRP-1 to target glioma cells, were created and fused with EXs, which have an enhanced ability to cross the BBB, in part because of the expression of transferrin receptors on the EX’s surface, to create hybrid liposome-EX nanovesicles with an enhanced ability to cross the BBB and target brain tumors [26,27]. Compared with liposomes and EXs, the hybrid nanovesicles demonstrated an increased ability to cross the BBB and accumulate in glioma in mice [26].
In a fascinating study, adipose-derived mesenchymal stem cell (ADSC) EXs and gelatine nanoparticle (GNP) hydrogel combined to form a stable meshwork because of the opposing charges of the two substances. In a rat skull defect model, the skull bone formation produced by the GNP hydrogel-ADSC EX mixture was substantially greater than that of the GNP- and sham-treated groups [28].
Liu and colleagues [29] created a nanocomplex, referred to as REXO-C/ANP/S, which was comprised of a core and a shell. The core, referred to as C/ANP/S, was comprised of: (a) curcumin, a small molecule that disrupts α-synuclein aggregates; (b) small interfering RNA targeting SNCA to downregulate α-synuclein synthesis; and (c) a reactive oxygen species (ROS)-responsive polymer, poly(2-(dimethylamino)ethyl acrylate, which encapsulates curcumin and enables ROS-responsive drug release. The shell, termed REXO, was comprised of: (a) immature dendritic cell-derived EXs; and (b) RVG peptide, a targeting peptide embedded in the exosomal membrane to enhance delivery across the BBB and target neurons. In a mouse model of Parkinson’s disease, the REXO-C/ANP/S nanoparticles decreased α-synuclein aggregates in diseased neurons [29].

2.4. Large-Scale EX Production Approaches

The quality and quantity of secreted EXs are reduced in conventional two-dimensional, static cultures, as these cultures cannot replicate the in vivo environment, including fluidic stimuli [30]. To overcome these limitations, Huang and coworkers [31] developed a method of culturing genetically engineered mesenchymal stromal cells to increase the production of EXs rich in hepatocyte growth factor (HGF) for improved wound healing. Two structured polydimethylsiloxane layers were used to construct a microfluidic chip, which was a crucial part of their culturing technique. The top layer had herringbone grooves, and the bottom layer had a micropillar array. By disrupting the laminar flow, these structures produced a turbulent vortex that improved waste removal and nutrient delivery. In addition, the bottom micropillar layer provided a large number of cell adhesion sites. The EX yield obtained from cells cultured on the microfluidic chip was ~14 times that obtained from cells cultured in a conventional, two-dimensional static cell culture flask. Furthermore, the EXs derived from microfluidic chip-cultured cells were abundant in HGF and aided in wound healing [31].
Contrary to conventional, static cell culture conditions, a flat-plate bioreactor was used to increase EX production. The chamber in the flat-plate bioreactor was seeded with cells. The culture medium was delivered to the chamber via a peristaltic pump, which produced laminar flow conditions. The flow introduced shear stress to the cells, which opened calcium channels, leading to increased calcium influx [32]. Prior studies have shown that increasing intracellular calcium concentration increases EX yield [33]. A closed-loop system was used to collect and recirculate the culture medium, which ensured a continuous exchange of oxygen and nutrients while eliminating waste. The bioreactor produced ~7 times more EXs than static culture conditions, while preserving the quality and therapeutic efficacy of the EXs [32].

2.5. Milk and Plant EXs

Milk- and plant-derived EXs have gained popularity recently despite advancements in the mass production of EXs derived from cultured cells. Milk is a particularly dense source of EXs, with concentrations in milk exceeding 1012 EXs per milliliter [34]. Due to their stability, scalability, ability to cross biological barriers, and between-species biocompatibility, recent research has focused on the development of milk EXs as a drug delivery platform. Milk EXs are non-immunologic oral delivery vehicles for gene editing agents, peptides, small molecules, and bioactive RNA cargo [35,36,37]. However, the isolation and characterization of milk EXs is complicated by the fact that milk contains a variety of nanoparticles besides EXs [38]. Continued research is crucial to harness the full potential of milk EXs in clinical and therapeutic contexts.
Similar to milk-derived EXs, plant-derived EXs represent a potentially rich source of useful EXs. Plant-derived EXs are thought to be used as a means of intercellular communication and immune regulation to protect plants from pathogenic attacks [39]. Notably, the size distribution, density, morphology, and surface electric charge of EXs derived from plants are comparable to those derived from mammals [40]. Like mammalian cell-derived EXs, they contain lipids, proteins, and RNAs, which reflect their plant sources, such as grapes, ginger, and apples [41,42,43]. The techniques used to isolate EXs from plants are not very different from those used to isolate EXs produced by cultured mammalian cells [44]. Plant EXs are safe [42] and their production is more scalable and cost-effective than EXs derived from mammalian cells [45]. Ongoing research is needed to establish standardized plant EX extraction protocols and evaluate long-term safety for clinical applications.

3. Contributions to This Special Issue

In this Special Issue, Mecocci and colleagues [46] described the anti-microbial and anti-inflammatory effects of cow colostrum-derived EXs in an in vitro model of neonatal calf diarrhea caused by Escherichia coli infection. Their results present the fascinating prospect that colostrum-derived EXs may provide an alternative to antibiotics in combating drug-resistant bacterial infections. Schepici and coworkers [47] provided a comprehensive review of the regenerative effects of mesenchymal stromal/stem cell-derived EXs in spinal cord injury and a summary of the preclinical studies on their therapeutic potential. Mohammad and colleagues [48] examined the effects of exercise-induced EXs on trophoblasts in vitro, highlighting their potential role in maternal–fetal communication during pregnancy. Rademacher [49] described the dual role of EXs in Parkinson’s disease, as both a contributor to disease progression and a potential therapeutic tool. In Parkinson’s disease, EXs act as mediators of disease progression by spreading toxic proteins and promoting neuroinflammation. However, their natural role in intercellular communication and ability to deliver therapeutic agents make them a promising tool for developing next-generation therapies [49]. Wang and coworkers [50] investigated the role of EXs derived from osteogenic-differentiated human bone marrow-derived mesenchymal cells (hBMSCs) in the rescue of the osteogenic ability of hBMSCs impaired by hypoxia. They discovered that EXs derived from hBMSCs cultured in normoxia restored the osteogenic capacity of hBMSCs that had been compromised by hypoxia, offering a possible treatment approach for bone regeneration in hypoxic conditions [50]. The literature on the effects of physical cues on stem cell-derived EXs to treat peripheral neuropathy was reviewed by Berry and colleagues [51]. They concluded that physical cues such as electrical stimulation, mechanical agitation, and conductive materials greatly improved the production, quality, and therapeutic potential of stem cell-derived EXs. These improvements make EXs more effective in treating peripheral neuropathy [51]. The efficiency and quality of artificial EXs produced by T cells using two different induction techniques—cytochalasin B, a chemical inducer, and ultrasonication, a physical inducer—were compared by Zmievskaya and colleagues [52]. Garaeva and colleagues [53] discovered that EXs loaded with exogenous, recombinant heat shock protein 70 derived from grapefruit triggered an anti-tumor immune response in both in vitro and in vivo models of colorectal cancer, highlighting the potential of plant-derived EXs as effective therapeutic cargo delivery vehicles. Jones and coworkers [54] determined the efficacy of lyophilized EXs derived from human adipose stem cells in promoting healing from traumatic brain injury, underlining their potential for storage at room temperature and clinical application. Finally, Salih and colleagues [55] investigated the expression of the microRNAs let-7a-5p and miR-21-3p in EXs derived from the serum of non-small-cell lung cancer patients, highlighting their potential as diagnostic biomarkers.

4. Future Research

High production costs, a high level of technical expertise, and challenges differentiating genetically engineered EXs from naturally occurring EXs in biological fluids are some of the drawbacks of genetically engineered EXs, despite their many benefits such as improved targeting precision, effective therapeutic delivery, and decreased off-target effects. Further research and optimization are needed to make them more feasible for clinical applications. Chemically modified EXs offer several advantages, including enhanced targeting, versatility, retention of the natural properties of the EXs, and the ability to cross biological barriers. The technical difficulties of both covalent and non-covalent EX modifications, the possible loss of native protein function in the EX membrane, and the difficulty in scaling up chemical modification techniques to large-scale production are the drawbacks of chemical modification of EXs. Future studies should focus on addressing these issues. Potential disruption of the exosomal membrane during production, the possibility of an immunological reaction, and regulatory ambiguity surrounding their classification—which makes approval more difficult—are some of the difficulties with hybrid nanoparticles. Clear regulatory frameworks, scalable production methods, and improved engineering techniques are essential to tackle these challenges. Future developments in isolation technologies, engineering methods, and standardization protocols are required to enable scalable EX production for clinical applications, even though recent advancements in the large-scale production of EXs, such as the use of microfluidic chips and bioreactors, are encouraging. Despite the fact that milk EXs are non-immunologic oral delivery vehicles for a range of therapeutic substances, a major obstacle is the transfer of isolation techniques from the laboratory to the industrial production process. In addition, there are no detailed pharmacokinetic studies, and the complex composition of milk makes it difficult to isolate high-purity milk EXs. To address these issues, improvements in isolation techniques, scalable production methods, and thorough safety and efficacy studies are needed. Despite the fact that plant EXs are safe [42] and can be produced more cheaply and efficiently than EXs produced by mammalian cells [45], preclinical and clinical research are required to confirm their therapeutic efficacy, optimize dosages, and assess long-term effects before they can be used successfully in clinical settings.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The author declares no conflicts of interest.

References

  1. Zaborowski, M.P.; Balaj, L.; Breakefield, X.O.; Lai, C.P. Extracellular Vesicles: Composition, Biological Relevance, and Methods of Study. Bioscience 2015, 65, 783–797. [Google Scholar] [CrossRef] [PubMed]
  2. Yáñez-Mó, M.; Siljander, P.R.; Andreu, Z.; Zavec, A.B.; Borràs, F.E.; Buzas, E.I.; Buzas, K.; Casal, E.; Cappello, F.; Carvalho, J.; et al. Biological properties of extracellular vesicles and their physiological functions. J. Extracell. Vesicles 2015, 4, 27066. [Google Scholar] [CrossRef] [PubMed]
  3. Borges, F.T.; Reis, L.A.; Schor, N. Extracellular vesicles: Structure, function, and potential clinical uses in renal diseases. Braz. J. Med. Biol. Res. 2013, 46, 824–830. [Google Scholar] [CrossRef]
  4. Bebelman, M.P.; Smit, M.J.; Pegtel, D.M.; Baglio, S.R. Biogenesis and function of extracellular vesicles in cancer. Pharmacol. Ther. 2018, 188, 1–11. [Google Scholar] [CrossRef]
  5. Raposo, G.; Stoorvogel, W. Extracellular vesicles: Exosomes, microvesicles, and friends. J. Cell Biol. 2013, 200, 373–383. [Google Scholar] [CrossRef]
  6. Tabatabai, T.S.; Alizadeh, M.; Rezakhani, L.; Tabatabai, T.S.; Ehterami, A.; Kloucheh, S.G.; Kebria, M.M.; Vaez, A.; Salehi, M. Unlocking the potential of EXOs in regenerative medicine: A comprehensive review. Tissue Cell 2025, 97, 103068. [Google Scholar] [CrossRef]
  7. Cocucci, E.; Meldolesi, J. Ectosomes and exosomes: Shedding the confusion between extracellular vesicles. Trends Cell Biol. 2015, 25, 364–372. [Google Scholar] [CrossRef]
  8. Meldolesi, J. Exosomes and Ectosomes in Intercellular Communication. Curr. Biol. 2018, 28, R435–R444. [Google Scholar] [CrossRef]
  9. Kim, H.; Jang, H.; Cho, H.; Choi, J.; Hwang, K.Y.; Choi, Y.; Kim, S.H.; Yang, Y. Recent Advances in Exosome-Based Drug Delivery for Cancer Therapy. Cancers 2021, 13, 4435. [Google Scholar] [CrossRef]
  10. Samanta, S.; Rajasingh, S.; Drosos, N.; Zhou, Z.; Dawn, B.; Rajasingh, J. Exosomes: New molecular targets of diseases. Acta Pharmacol. Sin. 2018, 39, 501–513. [Google Scholar] [CrossRef]
  11. Gomez-Molina, C.; Sandoval, M.; Henzi, R.; Ramirez, J.P.; Varas-Godoy, M.; Luarte, A.; Lafourcade, C.A.; Lopez-Verrilli, A.; Smalla, K.H.; Kaehne, T.; et al. Small Extracellular Vesicles in Rat Serum Contain Astrocyte-Derived Protein Biomarkers of Repetitive Stress. Int. J. Neuropsychopharmacol. 2019, 22, 232–246. [Google Scholar] [CrossRef]
  12. Wiklander, O.P.; Nordin, J.Z.; O’Loughlin, A.; Gustafsson, Y.; Corso, G.; Mager, 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. [Google Scholar] [CrossRef]
  13. Akuma, P.; Okagu, O.D.; Udenigwe, C.C. Naturally Occurring Exosome Vesicles as Potential Delivery Vehicle for Bioactive Compounds. Front. Sustain. Food Syst. 2019, 3, 23. [Google Scholar] [CrossRef]
  14. Yousefi Dehbidi, M.; Goodarzi, N.; Azhdari, M.H.; Doroudian, M. Mesenchymal stem cells and their derived exosomes to combat Covid-19. Rev. Med. Virol. 2022, 32, e2281. [Google Scholar] [CrossRef]
  15. Phinney, D.G.; Pittenger, M.F. Concise Review: MSC-Derived Exosomes for Cell-Free Therapy. Stem Cells 2017, 35, 851–858. [Google Scholar] [CrossRef]
  16. Mirzaaghasi, A.; Han, Y.; Ahn, S.H.; Choi, C.; Park, J.H. Biodistribution and Pharmacokinectics of Liposomes and Exosomes in a Mouse Model of Sepsis. Pharmaceutics 2021, 13, 427. [Google Scholar] [CrossRef] [PubMed]
  17. Zhang, M.; Zang, X.; Wang, M.; Li, Z.; Qiao, M.; Hu, H.; Chen, D. Exosome-based nanocarriers as bio-inspired and versatile vehicles for drug delivery: Recent advances and challenges. J. Mater. Chem. B 2019, 7, 2421–2433. [Google Scholar] [CrossRef] [PubMed]
  18. Wang, Y.; Liu, Z.; Li, L.; Zhang, Z.; Zhang, K.; Chu, M.; Liu, Y.; Mao, X.; Wu, D.; Xu, D.; et al. Anti-ferroptosis exosomes engineered for targeting M2 microglia to improve neurological function in ischemic stroke. J. Nanobiotechnol. 2024, 22, 291. [Google Scholar] [CrossRef] [PubMed]
  19. Kim, M.; Kim, G.; Hwang, D.W.; Lee, M. Delivery of High Mobility Group Box-1 siRNA Using Brain-Targeting Exosomes for Ischemic Stroke Therapy. J. Biomed. Nanotechnol. 2019, 15, 2401–2412. [Google Scholar] [CrossRef]
  20. Smyth, T.; Petrova, K.; Payton, N.M.; Persaud, I.; Redzic, J.S.; Graner, M.W.; Smith-Jones, P.; Anchordoquy, T.J. Surface functionalization of exosomes using click chemistry. Bioconjug. Chem. 2014, 25, 1777–1784. [Google Scholar] [CrossRef]
  21. Nwe, K.; Brechbiel, M.W. Growing applications of “click chemistry” for bioconjugation in contemporary biomedical research. Cancer Biother. Radiopharm. 2009, 24, 289–302. [Google Scholar] [CrossRef]
  22. 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. [Google Scholar] [CrossRef] [PubMed]
  23. Chen, Y.; Dong, J.; Li, J.; Li, J.; Lu, Y.; Dong, W.; Zhang, D.; Dang, X. Engineered macrophage-derived exosomes via click chemistry for the treatment of osteomyelitis. J. Mater. Chem. B 2024, 12, 10593–10604. [Google Scholar] [CrossRef] [PubMed]
  24. Kooijmans, S.A.A.; Fliervoet, L.A.L.; van der Meel, R.; Fens, 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. Control. Release 2016, 224, 77–85. [Google Scholar] [CrossRef]
  25. Wu, X.; Dai, X.; Liao, Y.; Sheng, M.; Shi, X. Investigation on drug entrapment location in liposomes and transfersomes based on molecular dynamics simulation. J. Mol. Model. 2021, 27, 111. [Google Scholar] [CrossRef]
  26. Wang, R.; Wang, X.; Zhao, H.; Li, N.; Li, J.; Zhang, H.; Di, L. Targeted delivery of hybrid nanovesicles for enhanced brain penetration to achieve synergistic therapy of glioma. J. Control. Release 2024, 365, 331–347. [Google Scholar] [CrossRef]
  27. Ma, Y.; Zhang, J.; Rui, Y.; Rolle, J.; Xu, T.; Qian, Z.; Gu, Y.; Li, S. Depletion of glioma stem cells by synergistic inhibition of mTOR and c-Myc with a biological camouflaged cascade brain-targeting nanosystem. Biomaterials 2021, 268, 120564. [Google Scholar] [CrossRef]
  28. Li, R.; Li, D.; Wang, H.; Chen, K.; Wang, S.; Xu, J.; Ji, P. Exosomes from adipose-derived stem cells regulate M1/M2 macrophage phenotypic polarization to promote bone healing via miR-451a/MIF. Stem Cell Res. Ther. 2022, 13, 149. [Google Scholar] [CrossRef] [PubMed]
  29. Liu, L.; Li, Y.; Peng, H.; Liu, R.; Ji, W.; Shi, Z.; Shen, J.; Ma, G.; Zhang, X. Targeted exosome coating gene-chem nanocomplex as “nanoscavenger” for clearing alpha-synuclein and immune activation of Parkinson’s disease. Sci. Adv. 2020, 6, eaba3967. [Google Scholar] [CrossRef]
  30. Cao, J.; Wang, B.; Tang, T.; Lv, L.; Ding, Z.; Li, Z.; Hu, R.; Wei, Q.; Shen, A.; Fu, Y.; et al. Three-dimensional culture of MSCs produces exosomes with improved yield and enhanced therapeutic efficacy for cisplatin-induced acute kidney injury. Stem Cell Res. Ther. 2020, 11, 206. [Google Scholar] [CrossRef]
  31. Huang, J.; Chen, H.; Luo, Z.; Nie, M.; Wang, J.; Lu, L.; Zhao, Y. Genetically Engineered Stromal Cell Exosomes from High-Throughput Herringbone Microfluidics. ACS Nano 2025, 19, 10568–10577. [Google Scholar] [CrossRef] [PubMed]
  32. Kang, H.; Bae, Y.H.; Kwon, Y.; Kim, S.; Park, J. Extracellular Vesicles Generated Using Bioreactors and their Therapeutic Effect on the Acute Kidney Injury Model. Adv. Healthc. Mater. 2022, 11, e2101606. [Google Scholar] [CrossRef]
  33. Taylor, J.; Azimi, I.; Monteith, G.; Bebawy, M. Ca2+ mediates extracellular vesicle biogenesis through alternate pathways in malignancy. J. Extracell. Vesicles 2020, 9, 1734326. [Google Scholar] [CrossRef]
  34. Marsh, S.R.; Pridham, K.J.; Jourdan, J.; Gourdie, R.G. Novel Protocols for Scalable Production of High Quality Purified Small Extracellular Vesicles from Bovine Milk. Nanotheranostics 2021, 5, 488–498. [Google Scholar] [CrossRef]
  35. Munagala, R.; Aqil, F.; Jeyabalan, J.; Gupta, R.C. Bovine milk-derived exosomes for drug delivery. Cancer Lett. 2016, 371, 48–61. [Google Scholar] [CrossRef]
  36. Aqil, F.; Munagala, R.; Jeyabalan, J.; Agrawal, A.K.; Kyakulaga, A.H.; Wilcher, S.A.; Gupta, R.C. Milk exosomes—Natural nanoparticles for siRNA delivery. Cancer Lett. 2019, 449, 186–195. [Google Scholar] [CrossRef] [PubMed]
  37. Yan, C.; Chen, J.; Wang, C.; Yuan, M.; Kang, Y.; Wu, Z.; Li, W.; Zhang, G.; Machens, H.G.; Rinkevich, Y.; et al. Milk exosomes-mediated miR-31-5p delivery accelerates diabetic wound healing through promoting angiogenesis. Drug Deliv. 2022, 29, 214–228. [Google Scholar] [CrossRef] [PubMed]
  38. Holt, C.; Carver, J.A.; Ecroyd, H.; Thorn, D.C. Invited review: Caseins and the casein micelle: Their biological functions, structures, and behavior in foods. J. Dairy Sci. 2013, 96, 6127–6146. [Google Scholar] [CrossRef]
  39. Malhotra, H.; Sheokand, N.; Kumar, S.; Chauhan, A.S.; Kumar, M.; Jakhar, P.; Boradia, V.M.; Raje, C.I.; Raje, M. Exosomes: Tunable Nano Vehicles for Macromolecular Delivery of Transferrin and Lactoferrin to Specific Intracellular Compartment. J. Biomed. Nanotechnol. 2016, 12, 1101–1114. [Google Scholar] [CrossRef]
  40. Iriawati, I.; Vitasasti, S.; Rahmadian, F.N.A.; Barlian, A. Isolation and characterization of plant-derived exosome-like nanoparticles from Carica papaya L. fruit and their potential as anti-inflammatory agent. PLoS ONE 2024, 19, e0304335. [Google Scholar] [CrossRef]
  41. Cai, Q.; Qiao, L.; Wang, M.; He, B.; Lin, F.M.; Palmquist, J.; Huang, S.D.; Jin, H. Plants send small RNAs in extracellular vesicles to fungal pathogen to silence virulence genes. Science 2018, 360, 1126–1129. [Google Scholar] [CrossRef]
  42. Rutter, B.D.; Innes, R.W. Extracellular Vesicles Isolated from the Leaf Apoplast Carry Stress-Response Proteins. Plant Physiol. 2017, 173, 728–741. [Google Scholar] [CrossRef]
  43. Alzahrani, F.A.; Khan, M.I.; Kameli, N.; Alsahafi, E.; Riza, Y.M. Plant-Derived Extracellular Vesicles and Their Exciting Potential as the Future of Next-Generation Drug Delivery. Biomolecules 2023, 13, 839. [Google Scholar] [CrossRef]
  44. Mun, J.G.; Song, D.H.; Kee, J.Y.; Han, Y. Recent Advances in the Isolation Strategies of Plant-Derived Exosomes and Their Therapeutic Applications. Curr. Issues Mol. Biol. 2025, 47, 144. [Google Scholar] [CrossRef] [PubMed]
  45. Wang, Q.; Zhuang, X.; Mu, J.; Deng, Z.B.; Jiang, H.; Zhang, L.; Xiang, X.; Wang, B.; Yan, J.; Miller, D.; et al. Delivery of therapeutic agents by nanoparticles made of grapefruit-derived lipids. Nat. Commun. 2013, 4, 1867. [Google Scholar] [CrossRef] [PubMed]
  46. Mecocci, S.; De Paolis, L.; Zoccola, R.; Fruscione, F.; De Ciucis, C.G.; Chiaradia, E.; Moccia, V.; Tognoloni, A.; Pascucci, L.; Zoppi, S.; et al. Antimicrobial and Immunomodulatory Potential of Cow Colostrum Extracellular Vesicles (ColosEVs) in an Intestinal In Vitro Model. Biomedicines 2022, 10, 3264. [Google Scholar] [CrossRef]
  47. Schepici, G.; Silvestro, S.; Mazzon, E. Regenerative Effects of Exosomes-Derived MSCs: An Overview on Spinal Cord Injury Experimental Studies. Biomedicines 2023, 11, 201. [Google Scholar] [CrossRef]
  48. Mohammad, S.; Bhattacharjee, J.; Tzaneva, V.; Hutchinson, K.A.; Shaikh, M.; Fernandes da Silva, D.; Burger, D.; Adamo, K.B. The Influence of Exercise-Associated Small Extracellular Vesicles on Trophoblasts In Vitro. Biomedicines 2023, 11, 857. [Google Scholar] [CrossRef]
  49. Rademacher, D.J. Potential for Therapeutic-Loaded Exosomes to Ameliorate the Pathogenic Effects of alpha-Synuclein in Parkinson’s Disease. Biomedicines 2023, 11, 1187. [Google Scholar] [CrossRef]
  50. Wang, C.; Stockl, S.; Pattappa, G.; Schulz, D.; Hofmann, K.; Ilic, J.; Reinders, Y.; Bauer, R.J.; Sickmann, A.; Grassel, S. Extracellular Vesicles Derived from Osteogenic-Differentiated Human Bone Marrow-Derived Mesenchymal Cells Rescue Osteogenic Ability of Bone Marrow-Derived Mesenchymal Cells Impaired by Hypoxia. Biomedicines 2023, 11, 2804. [Google Scholar] [CrossRef]
  51. Berry, D.; Ene, J.; Nathani, A.; Singh, M.; Li, Y.; Zeng, C. Effects of Physical Cues on Stem Cell-Derived Extracellular Vesicles toward Neuropathy Applications. Biomedicines 2024, 12, 489. [Google Scholar] [CrossRef] [PubMed]
  52. Zmievskaya, E.A.; Mukhametshin, S.A.; Ganeeva, I.A.; Gilyazova, E.M.; Siraeva, E.T.; Kutyreva, M.P.; Khannanov, A.A.; Yuan, Y.; Bulatov, E.R. Artificial Extracellular Vesicles Generated from T Cells Using Different Induction Techniques. Biomedicines 2024, 12, 919. [Google Scholar] [CrossRef] [PubMed]
  53. Garaeva, L.; Komarova, E.; Emelianova, S.; Putevich, E.; Konevega, A.L.; Margulis, B.; Guzhova, I.; Shtam, T. Grapefruit-Derived Vesicles Loaded with Recombinant HSP70 Activate Antitumor Immunity in Colon Cancer In Vitro and In Vivo. Biomedicines 2024, 12, 2759. [Google Scholar] [CrossRef]
  54. Jones, B.; Patel, R.; Wang, B.; Evans-Nguyen, T.; Patel, N.A. Lyophilized Small Extracellular Vesicles (sEVs) Derived from Human Adipose Stem Cells Maintain Efficacy to Promote Healing in Neuronal Injuries. Biomedicines 2025, 13, 275. [Google Scholar] [CrossRef]
  55. Salih, D.J.; Reiners, K.S.; Loizzi, D.; Ardò, N.P.; Santantonio, T.A.; Sollitto, F.; Hartmann, G. Expression Analysis of let-7a-5p and miR-21-3p in Extracellular Vesicles Derived from Serum of NSCLC Patients. Biomedicines 2025, 13, 2060. [Google Scholar] [CrossRef]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Rademacher, D.J. Editorial of the Special Issue: Extracellular Vesicles and Exosomes as Therapeutic Agents. Biomedicines 2025, 13, 2234. https://doi.org/10.3390/biomedicines13092234

AMA Style

Rademacher DJ. Editorial of the Special Issue: Extracellular Vesicles and Exosomes as Therapeutic Agents. Biomedicines. 2025; 13(9):2234. https://doi.org/10.3390/biomedicines13092234

Chicago/Turabian Style

Rademacher, David J. 2025. "Editorial of the Special Issue: Extracellular Vesicles and Exosomes as Therapeutic Agents" Biomedicines 13, no. 9: 2234. https://doi.org/10.3390/biomedicines13092234

APA Style

Rademacher, D. J. (2025). Editorial of the Special Issue: Extracellular Vesicles and Exosomes as Therapeutic Agents. Biomedicines, 13(9), 2234. https://doi.org/10.3390/biomedicines13092234

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

Article Metrics

Back to TopTop