Next Article in Journal
Focused Ultrasound-Mediated Disruption of the Blood–Brain Barrier for AAV9 Delivery in a Mouse Model of Huntington’s Disease
Previous Article in Journal
Drug Development for Alzheimer’s and Parkinson’s Disease: Where Do We Go Now?
Previous Article in Special Issue
Development of 5-Fluorouracil/pH-Responsive Adjuvant-Embedded Extracellular Vesicles for Targeting αvβ3 Integrin Receptors in Tumors
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Insight into the Functional Dynamics and Challenges of Exosomes in Pharmaceutical Innovation and Precision Medicine

McGowan Institute for Regenerative Medicine, Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA
*
Author to whom correspondence should be addressed.
Pharmaceutics 2024, 16(6), 709; https://doi.org/10.3390/pharmaceutics16060709
Submission received: 21 April 2024 / Revised: 13 May 2024 / Accepted: 16 May 2024 / Published: 24 May 2024
(This article belongs to the Special Issue Extracellular Vesicle-Based Drug Delivery Systems)

Abstract

:
Of all the numerous nanosized extracellular vesicles released by a cell, the endosomal-originated exosomes are increasingly recognized as potential therapeutics, owing to their inherent stability, low immunogenicity, and targeted delivery capabilities. This review critically evaluates the transformative potential of exosome-based modalities across pharmaceutical and precision medicine landscapes. Because of their precise targeted biomolecular cargo delivery, exosomes are posited as ideal candidates in drug delivery, enhancing regenerative medicine strategies, and advancing diagnostic technologies. Despite the significant market growth projections of exosome therapy, its utilization is encumbered by substantial scientific and regulatory challenges. These include the lack of universally accepted protocols for exosome isolation and the complexities associated with navigating the regulatory environment, particularly the guidelines set forth by the U.S. Food and Drug Administration (FDA). This review presents a comprehensive overview of current research trajectories aimed at addressing these impediments and discusses prospective advancements that could substantiate the clinical translation of exosomal therapies. By providing a comprehensive analysis of both the capabilities and hurdles inherent to exosome therapeutic applications, this article aims to inform and direct future research paradigms, thereby fostering the integration of exosomal systems into mainstream clinical practice.

1. Introduction

The expanding domain of exosome research represents a significant frontier in the realm of pharmaceutical innovation and precision medicine, offering a convergence of novel opportunities and complex challenges [1,2]. Exosomes, defined as nanosized extracellular vesicles (EVs) of endosomal origin, secreted by almost all cell types, have gained prominence as potential vectors for therapeutic delivery [3,4]. Their intrinsic characteristics, such as biological stability, minimal immunogenicity, and the capability for cell-specific targeting, position them as pivotal candidates in the advancement of drug delivery systems, nucleic acid, and vaccine therapeutics [5,6]. In this review, we have used the term “exosomes” with the intent of discussing the small EVs of endosomal origin having diameters ranging from 30 to 150 nm. It is critically important to acknowledge the fact that the terms “exosome” and “small EV” are not synonymous and should be used with precision. This review seeks to dissect the multifaceted landscape of exosome therapy and show how it is more advantageous than liposomal and microvesicle-based therapies (Table 1) within the pharmaceutical innovation sphere and precision medicine, by focusing on the crucial aspects of regulatory scrutiny, particularly by the United States Food and Drug Administration (FDA). It also aims to analyze the hurdles intrinsic to translating exosome-based therapies from benchtop research to bedside clinical practices [7,8]. The contemporary strategies devised to address the challenges endemic to the therapeutic application of exosomes encompass advancements in research methodologies, data standardization, and the precision characterization of these nanovesicles, alongside innovations aimed at enhancing targeting specificity and cargo delivery efficiency [9]. It is time to critically explore the emerging techniques for single-exosome analysis and the refinement of surface modification approaches [10,11]. Such advancements underline the transition of exosomes from conceptual entities to practical therapeutic vectors, highlighting the importance of biochemical engineering in optimizing their clinical applicability [12]. By navigating through this discourse, we aim to trace the developmental trajectory of exosome-based therapeutic applications, emphasizing the dynamic synergy between scientific research and regulatory governance, as well as the commitment to ensuring patient safety and therapeutic efficacy [13]. This study not only seeks to contribute to the scholarly discourse on exosome therapy but also aspires to catalyze future advancements in therapeutic modalities, ushering in a new epoch of patient-focused care and therapeutic delivery.

2. Where Does the FDA Stand?

Within the broader domain of regenerative medicine products, the U.S. FDA categorizes therapies derived from stem cells as somatic cellular therapies. It is important to note that the FDA employs a tailored regulatory framework for stem cell-based products based on their characteristics and intended uses. Recent work by Han et al. highlighted the significant influence of paracrine factors such as exosomes on the therapeutic potential of stem cells [14]. The progression of therapeutic applications utilizing exosomes delineates an intricate balance between innovation in biomedical research and stringent oversight by regulatory entities, notably the US FDA [12]. The FDA’s comprehensive regulatory framework for extracellular vesicle-based therapies, particularly those derived from exosomes, is predicated on ensuring the transition of these modalities from theoretical constructs to practical clinical applications, adhering to the highest safety and efficacy standards [15,16]. Such a regulatory approach is predicated on the principles of scientific accuracy, robust quality control mechanisms, and adherence to regulatory compliance standards [17,18]. Exosomes, a subclass of EVs, are recognized for their promising therapeutic potential across various medical fields [19]. These vesicles provide several key advantages over conventional cell-based therapies by their inherent capability to circumvent the proliferative and differentiation-associated risks post-administration, unlike that of stem cells [20]. Furthermore, they offer the potential for enhanced consistency in quality control standards across different production batches, which is a critical consideration in the context of biological drug development [5]. However, the path to clinical application of these vesicles is fraught with complexities, necessitating a comprehensive elucidation of their biological mechanisms, the establishment of scalable production methodologies that ensure reproducibility, and a tangible enhancement of their intrinsic therapeutic potential [21]. A primary challenge in this developmental trajectory is the establishment of uniform manufacturing protocols that guarantee consistency across product batches. Given the classification of exosomes as biological drugs by the FDA [22,23], they are subjected to exhaustive evaluations to validate their safety and efficacy through extensive clinical trials [24]. This stringent evaluative process highlights the critical need for refined manufacturing processes, comprehensive human trials, and conclusive evidence demonstrating clinical benefits [25,26]. Figure 1 shows an analysis of exosome-based clinical trials, as listed on clinicaltrials.gov (20 April 2024). A search on clinicaltrials.gov using ‘exosome’, ‘exosome therapy’, and ‘exosome treatment’, as keywords generated 204 records and listed in Table 2.
To address the potential risks associated with unregulated exosome therapies, the FDA has proactively issued guidance and warnings concerning the marketing of unapproved exosome products [27]. These regulatory actions reflect the FDA’s dedication to safeguarding public health and maintaining the integrity of emerging therapeutic modalities. Through the enforcement of regulatory standards and the meticulous review of clinical trial submissions, the FDA endeavors to mitigate the risks posed by unauthorized exosome therapies. While the regulatory pathway for exosome-based therapies is devoid of specific guidance on delivery systems, applying the existing regulatory requirements for biological products remains imperative [27]. These include the submission of Investigational New Drug (IND) applications, adherence to Good Manufacturing Practice (GMP) protocols, implementation of quality control measures, and the selection of appropriate cell sources for therapeutic production [28].

3. Challenges and Prospects Associated with the Translation of Exosome Therapy

The clinical adoption of exosome-based therapeutic strategies is impeded by challenges associated with low exosomal yield and efficiency [7]. In a controlled laboratory setting, extraction from one milliliter of cell culture medium frequently results in less than one microgram of exosomal protein [29]. Strategies to augment exosome production have been explored, encompassing mechanical (e.g., three-dimensional culture systems and the application of shear stress), biochemical (e.g., treatment with lipopolysaccharide (LPS), interferon-gamma (IFN-γ), bone morphogenetic protein 2 (BMP-2), tumor necrosis factor-alpha (TNF-α), and hypoxia-inducible factor 1-alpha (HIF-1α)), and physical (e.g., thermal stress, hypoxic conditions, and nutrient deprivation) methodologies [30,31,32].
Exosome isolation is complicated by their intrinsic heterogeneity, manifested in variations in size, composition, surface markers, and biological origin [33]. Predominant isolation and purification methodologies are based on immunoaffinity, leveraging surface charge characteristics, or size and density differences [34]. Each methodology presents specific advantages and drawbacks, and no singular approach is universally applicable [35]. Ultracentrifugation, while often deemed the gold standard, is constrained in its scalability by factors such as cost, efficiency, vesicle aggregation, and the co-isolation of lipoproteins, despite minimal chemical and expertise requirements [36]. Conversely, immunoaffinity chromatography, exploiting specific antigen–antibody interactions, is noted for its high specificity, yield, and purity, albeit dependent on the presence of appropriate exosomal surface antigens [37]. Techniques predicated on size differentiation, including size-exclusion chromatography and ultrafiltration, are advantageous for scalability but are limited by issues such as pore clogging, loss of exosomal material, and reduced purity [38,39,40]. The amalgamation of disparate isolation techniques, incorporating precipitation- and microfluidics-based approaches, may offer a comprehensive solution to effectively address the multifaceted requirements for exosome isolation and purification [41]. For instance, in our research endeavors, a synergistic application of differential ultracentrifugation and immunomagnetic separation facilitated the isolation of cell-specific exosomes from murine tissue, and a parallel strategy was employed for the extraction of exosomes from human wound effluents [42,43].
Anticoagulants, notably EDTA, have been documented to markedly influence both the composition of exosomes and their isolation processes from blood specimens [44]. Subsequent investigations have elucidated that during the procedural phases of blood collection and processing, EDTA plays a stabilizing role in maintaining the integrity of platelet-derived EVs [45]. However, it has also been substantiated that the utilization of EDTA-containing tubes does not forestall alterations in the exosomal profile. The effects of EDTA substitutes on the characterization and profiles of exosomes remain underexplored, presenting a significant gap in current research [46]. This issue bears considerable importance for the reproducibility of quantifications of blood exosome concentrations and the isolation of platelet-derived exosomes, which pose considerable challenges in biomarker discovery for conditions not associated with platelets [47]. This aspect becomes critically pertinent when processing single-spun plasma samples, potentially leading to contaminations in the exosome isolates due to the presence of frozen platelets [48].
The selection of an anticoagulant that preserves the analytes of interest without compromising the integrity of the exosomes is of paramount consideration [47]. Reports indicate that blood collection tubes incorporating non-aldehyde-based stabilizers for cell-free nucleic acids are conducive for exosome handling, as they also contribute to exosome stabilization. [49]. Another essential aspect for consistent determination of exosome composition in blood involves understanding how various pre-analytical factors, such as collection, processing, and storage conditions, impact the protein corona and the co-isolation of exosomes with other blood components, such as platelets, lipoproteins, soluble protein aggregates, viruses, cell-free DNA/histones, or circulating mitochondria [50].
Furthermore, the quantification and characterization of blood-derived exosomes are also contingent upon the centrifugation parameters and storage conditions [51]. The integrity of microRNA analyses in exosome samples can be significantly compromised by hemolysis during blood sample collection [52]. Addressing these variables is crucial in the standardization of methodologies for exosome research.

4. Factors Taken into Consideration for Exosomal Function Dictation

The function of exosomes is determined by isolation techniques, molecular characterization, functional tests, and imaging approaches. Despite progress, challenges remain due to the need for standardization, as well as the limitations of current technology [53]. Understanding the methods of exosome uptake, such as endocytosis, membrane fusion, or receptor-mediated interactions, is essential for the successful delivery of functional biomolecular cargo such as proteins, RNA, or miRNA [54]. Advanced in vitro assays such as fluorescent labeling and reporter gene assays, in vivo models, molecular analysis, such as proteomics, transcriptomics, Western blotting, qPCR, functional impact studies, such as cell proliferation assays, apoptosis assays, migration and invasion assays, and immunomodulation assays, help elucidate the functional impacts of exosomes on recipient cells [55]. The issues of cell specificity, dose, safety, and scalability must be addressed to maximize therapeutic efficacy, while minimizing unwanted effects. Exosomes have intrinsic targeting capability influencing both physiological and pathological states through lipids, RNA and proteins. Not surprisingly, various biotechnology companies like Stem Cell Medicine Ltd., Evox Therapeutics, Pharmaceutics Inc. are working to create different therapies using exosomes [55]. Much research has been conducted to elucidate the functioning of exosomes following uptake into recipient cells, overriding their inherent targeting ability. Exosomes can enter a recipient cell by macropinocytosis, phagocytosis, lipid-raft-mediated uptake, and membrane fusion. The primary route of exosome uptake is clathrin/caveolin-mediated endocytosis [54,56,57]. Functional delivery requires exosome-encapsulated material to “escape” from the endosome and enter the cytoplasm [56]. To support the exosome-encapsulated content’s communicative qualities, robust functional transport to recipient cells must be shown. Endosomal escape and effective cargo delivery are desired outcomes for exosome therapeutic usage, but there is also a risk of cargo loss due to breakdown, recycling in the cell, or the re-release of intact vesicles into the extracellular environment [56].

4.1. Protein Corona

The concept of the protein corona refers to the phenomenon where proteins adhere to the surface of exosomes upon their introduction into biological fluids such as blood plasma, forming a coating (Figure 2) [58]. This process alters the surface properties, such as shape, size, and structural modifications of exosomes, significantly affecting their interactions with the cell membrane of recipient cells by modifying recognition, uptake, targeting, and the biological responses elicited, with profound implications for health and diseases like cancer, Alzheimer’s, wound healing, etc. [59]. The protein corona comprises not merely a random assortment of plasma proteins, but includes specific proteins, such as anti-thrombin III, factor V, complement C3, IgG, fibronectin, and complement factor H, which influence the biological behavior and interactions of exosomes within the bloodstream [60]. This aspect facilitates the targeted delivery of exosomes to specific cell types or tissues by promoting interactions with the receptors. Nonetheless, the presence of a protein corona can compromise therapeutic efficacy through mechanisms such as the accelerated clearance of exosomes, induction of adverse immunological responses or reduction in the efficiency of cargo delivery to target cells [61].
Strategies involving engineering the exosome surface with specific ligands or coatings can alter the protein corona composition, enhancing targeting specificity and reducing unwanted immune reactions [62]. Pre-exposing exosomes to plasma or specific proteins, such as anti-thrombin III, factor V, complement C3, IgG, fibronectin, and complement factor H, before therapeutic applications, such as cancer, nervous system diseases, and immune diseases, can induce a favorable corona formation, potentially augmenting their therapeutic efficacy in vivo. Wolfram et al. highlighted the role of nanoparticles high surface free energy in the adsorption of molecules, predominantly proteins, leading to the formation of the protein corona [61]. The binding forces facilitating such interactions encompass Van der Waals forces, hydrophobic interactions, hydrogen bonds, electrostatic attractions, and π-π stacking [61].
Conceptually, the protein corona is divided into a ‘hard’ and a ‘soft’ layer, distinguished by the proximity of biomolecules such as proteins, lipids, and sugars to the nanovesicles [63]. The soft layer is characterized by a more dynamic exchange of biomolecules. However, there is debate regarding the heterogeneity of the hard corona formed in human blood plasma, with some sources indicating fewer than 100 distinct proteins, while others suggest greater than 100 [64]. Table 3 lists protein coronas adsorbed around exosomes [58].
The composition of the protein corona is influenced by the size, shape, and surface charge of the nanovesicles, with prevalent proteins including albumin, complement proteins, apolipoprotein 1, and immunoglobulins [58]. Protein attachment to nanovesicles induces both reversible and irreversible structural changes in the proteins, occurring in stages, with the least stable conformations exhibiting the quickest misfolding kinetics [65]. Protein adsorption is enhanced, with an increase in size due to the lower curvature, facilitating more extensive surface interaction and consequent protein conformational changes. Additionally, a positive surface charge correlates with increased protein adsorption [61].
Table 3. List of protein coronas adsorbed around exosomes.
Table 3. List of protein coronas adsorbed around exosomes.
S. No.Hard and Soft PCTypes of InteractionsEffectReferences
1.AlbuminConnects to RNA/DNA on exosomesPhagocytosis of exosomesMalonga et al. 2006 [66]
2.ApoE and ApoB100Protein–protein interactionsIncreases transfer rate in peripheral tissueBertrand et al. 2017 [67]
3.Apolipoprotein A1Interacts with CD63 on Exosome surfacePhagocytosis of exosomesToth et al. 2021 [60]
4.Apolipoprotein BInteracts with CD63 on Exosome surfaceIncreases phagocytic activities and triggers secondary inflammation reactionsToth et al. 2021 [60]
5.Complement factors 3Interacts with CD63 on Exosome surfaceIncreases phagocytic activities and triggers secondary inflammation reactionsToth et al. 2021 [60]
6.Complement proteins C3b and C3ibProtein–protein interactionsProlonged chronic inflammatory conditionsConde et al. 2012 [68]
7.Immunoglobulin heavy chains of (γ2 and γ4)Protein–protein interactionsReduces transit time and affects bio-distributionToth et al. 2021 [60]
8.Mismatched MHC-I and IIOn the surface of ExosomesT-cell immune responsesHiltbrunner et al. 2016 [69]
9.S100-A8, LDL-receptor, CD14, HLA class IPhosphatidylserine and tissue factor on Exosome surfaceDynamic activityBuzas et al. 2018 [70]
The protein corona on exosomes can also influence the cellular uptake of nanovesicles by modulating their adhesion to the cell membrane, a process which can either inhibit or enhance cellular internalization [71]. Despite the consensus on the dynamic nature of the protein corona, specific binding events occur at the nano–plasma interface, and their sequential order remains underexplored. Recent studies indicate that the quantity of protein in the corona varies over time, although the types of proteins bound tend to remain stable [58,61]. Nevertheless, the variability in protein corona composition among extracellular vesicle populations presents challenges in controlling the protein corona via surface engineering strategies [61].

4.2. Interactions of Lipoproteins with Exosomes

The potential interactions between exosomes and lipoproteins have received minimal attention in research conducted over the last few decades [62]. Previous investigations have rigorously examined lipoproteins as exogenous impurities in EV samples. Notably, lipoproteins are identified at a minimum quintuple excess in comparison to exosomes in biological specimens processed for exosome isolation, posing a significant challenge due to their overlap in size with exosomes [72,73]. Both exosomes and lipoproteins are ubiquitously found in the bloodstream, sharing similar dimensions and densities. Plasma specimens comprise exosomes (40–120 nm), microvesicles (100 nm–1 μm), and apoptotic bodies (50 nm–2 μm), while lipoprotein subclasses, such as HDL (5–12 nm), LDL (18–25 nm), IDL (25–35 nm), VLDL (30–80 nm), chylomicron remnants (30–80 nm), and chylomicrons (75–1200 nm), exhibit size ranges overlapping with those of exosomes [62]. This congruence in size renders their separate isolation unfeasible [73,74,75]. Prior research has documented the presence of complexes formed between exosomes and lipoprotein-like structures in plasma from healthy human subjects [76]. Subsequent research corroborated these findings, observing exosome complexation with lipoproteins under physiological conditions [73,75,77]. Subsequent investigations, including the work of Lozano-Andres et al., utilizing cryogenic transmission electron microscopy, corroborated the association of EVs with lipoproteins and elucidated the consequential effects on EV detection and characterization [78]. Historically, the focus has been on treating lipoproteins as inert contaminants, neglecting the potential interactions between these two biological nanoparticles [39]. Proteomic analyses and immunogold transmission electron microscopy have indicated that lipoprotein components may engage in dynamic interactions with exosomes, leading to complex formation in both artificial and physiological milieus [79]. These complexes could influence exosome detection, characterization, cellular uptake, and subsequent biological effects. Techniques such as atomic force microscopy (AFM) or transmission electron microscopy (TEM) have been proposed to investigate the complexation phenomena between these. Collectively, these insights suggest that exosomes and lipoproteins can form complexes in various environments [80]. It is also paramount to acknowledge that exosome isolation techniques might foster non-physiological interactions between exosomes and lipoproteins [81]. Given these recent discoveries, it becomes imperative to explore the nature of these interactions and their potential mechanistic, therapeutic, and diagnostic ramifications at the exosome−lipoprotein interface [62].

4.3. Effects of Size, Morphology, and Surface Modification on Cellular Uptake of Exosomes

The physicochemical characteristics of nanovesicles significantly influence cellular internalization dynamics. It is pertinent to examine whether the methodologies employed for isolating enriched exosomal fractions impact their cellular uptake [82]. A comparative analysis of exosomes isolated via ultracentrifugation and polymer-based precipitation techniques demonstrated that the latter results in a narrower particle size distribution, accelerated absorption by target cells, and enhanced cellular motility [83]. Furthermore, differential efficiencies in exosome uptake were observed between vesicles of different cellular origins, highlighting the specificity of interaction between exosomes and target cells [2,84]. Endocytic pathways, including clathrin/caveolae-mediated endocytosis, phagocytosis, macropinocytosis, and pinocytosis, play critical roles in the size-dependent uptake of exosomes [84]. Smaller exosomes, typically ranging from 30 to 100 nm, are more likely internalized through clathrin-mediated endocytosis, while larger vesicles may prefer alternative pathways, such as caveolin-mediated endocytosis or macropinocytosis [85,86]. The preference for specific endocytic pathways based on size impacts the efficiency of uptake and subsequent intracellular routing, thereby affecting cargo delivery to targeted cellular compartments [87]. Additionally, the morphological characteristics of exosomes, such as their cup-shaped or spherical appearance under TEM, influence their binding affinity to cellular receptors and internalization rates [88]. Variations in preferred morphology are observed across different cell types [89]. The composition of the exosome membrane also affects its fusogenic capacity, facilitating direct cargo delivery into the cytoplasm by fusion with the cell membrane [90].
Exosomes present a diverse array of surface molecules, including proteins lipids, and glycans which can be exploited for targeted cellular interactions [91]. However, their targeting potential is sometimes limited. Innovative approaches such as surface functionalization can enhance targeting specificity [92]. For instance, Tian et al. enhanced the bioavailability of curcumin-loaded exosomes through peptide functionalization, enabling them to cross the blood-brain barrier post-intravenous injection [93]. Similarly, Liang et al. developed miR-140-loaded exosomes that, when administered intra-articularly, effectively penetrated dense extracellular matrices to alleviate osteoarthritis symptoms, suggesting a viable cell-free treatment strategy [94]. Moreover, Li et al. demonstrated the therapeutic potential of targeted engineered exosomes in diabetic wound healing [95]. Enhancements in the biofunctional engineering of exosomes could improve their targeting accuracy and circulation time, optimizing the delivery of therapeutic cargos to specific cells or tissues [95,96].

4.4. Cellular Environment

Paracrine intercellular communication via exosomes utilizes the cellular milieu as a conduit and modulates exosome–cell interactions to varying extents [97]. This milieu includes not only the extracellular matrix (ECM) but also external biophysical variables such as pH, temperature, and oxidative or hypoxic conditions [98]. For instance, ECM rigidity may influence exosome uptake, while its mechanical properties can dictate exosomal transit by interacting with water permeability [99]. Additionally, the extracellular environment’s pH and extracellular ion concentrations can significantly affect the stability, absorption, and release of exosomes [100]. Variations in ion levels might alter the composition and structural integrity of exosomal membranes, thereby modifying their interactions with recipient cells [101]. A low pH environment, commonly encountered in tumor settings, may enhance the malignancy of cancer cells by modulating exosome release and uptake [102].
Temperature also profoundly affects the cellular uptake of exosomes, with reduced temperatures markedly impeding this process, indicating its energy-dependent nature [103]. Pathological conditions such as cancer, inflammation, or infection led cells to alter their exosome secretion and composition [104]. For example, cancer cells typically secrete more exosomes containing oncogenic factors that facilitate tumor growth, angiogenesis, and metastatic site preparation [105].
Under conditions of oxidative stress or hypoxia, cells might emit exosomes laden with stress-response proteins or RNAs, influencing the survival and functionality of recipient cells [106]. The local tissue environment, which encompasses components of the ECM, cell–cell interactions, growth factors, and cytokines, also plays a crucial role in modulating exosome activity. Specific receptors within the tissue microenvironment can enhance the selectivity of exosome uptake by recipient cells [107]. Furthermore, oxidative stress can alter the protein content of exosomes from amnion-epithelial cells, promoting the release of inflammatory mediators that trigger inflammation [106]. Additionally, external factors such as ionizing radiation have been shown to enhance exosomal secretion pathways in breast cancer cells, as demonstrated by Jabbari et al., suggesting a potential mechanism for developing treatment resistance [108].
Exosomes play an important function in immunological regulation. They are released by antigen-presenting cells and can provide therapeutic advantages by suppressing or increasing the immune response [109,110]. Exosomes from antigen-presenting cells include major histocompatibility peptide complexes and costimulatory molecules that influence antigen-specific CD8+ and CD4+ responses. Antigen-specific T cells directly contact MHC–peptide complexes on exosomes, activating T cells. Exosomes produced by dendritic cells (DCs) can stimulate T and B cells and have been studied for their immunostimulatory characteristics in cancer treatment. Exosomes generated by dendritic cells (DCs) can activate T and B cells and have been investigated for immunostimulatory properties in cancer therapy [111,112]. They can increase immunological responses in vivo by transferring MHC–peptide complexes from DCs that have been exposed to an antigen to another DC that has not encountered the antigen [113].

4.5. Entry and Departure of Exosomes from the Circulation

Exosomes function as pivotal carriers of bioactive molecules, facilitating intercellular communication and playing a significant role in both paracrine and endocrine-signaling processes within the tissue interstitium and circulatory system [114,115]. Nevertheless, the precise biological mechanisms facilitating their ingress and egress from the circulatory system have yet to be fully elucidated. Prior research has delineated the traversal of exosomes through both paracellular and transcellular pathways, with paracellular transport assuming significance in pathological contexts, such as inflammation [116]. The transcellular pathway entails the endocytosis of exosomes by endothelial cells, followed by their transport across the cellular body for subsequent release on the opposite side [117]. Moreover, the lymphatic system is implicated in the transit of exosomes from various organs into the bloodstream, suggesting its critical role in the systemic dissemination of exosomes [114].
Endothelial cells are known to internalize exosomes through diverse mechanisms, including clathrin-dependent endocytosis, caveolin-mediated uptake, macropinocytosis, and lipid raft-mediated internalization [54]. These uptake pathways are influenced by the exosomes biophysical attributes, which may, in turn, modulate the endothelial internalization profile [117]. This inherent variability in exosome properties could be consequential in determining their cellular uptake dynamics, a phenomenon similarly observed with synthetic nanoparticles [54,117]. For instance, larger particles are predominantly internalized through phagocytosis or macropinocytosis, whereas smaller counterparts are primarily subject to alternative endocytic pathways [54,118]. This intricate interplay between exosomes and endothelial cells highlights the complexity of cellular internalization mechanisms and the potential for differential pathway engagement based on exosomal characteristics [114].

4.6. Functional Delivery of Exosomal Cargo

Cells encapsulate specific molecular cargoes within exosomes, which are then released into the extracellular milieu [88]. The lipid bilayer membrane of exosomes serves a critical protective role, shielding the encapsulated cargo from enzymatic degradation in the extracellular environment [1,119]. This process of paracrine signaling involves the direct transfer of this cargo into recipient cells, thereby exerting control over multiple levels, including genetic, signaling pathways, and overall cellular activities [97]. A key aspect of the therapeutic application of exosomes is ensuring the efficient release of the encapsulated content from the endosome before its internal environment becomes acidic and degrades the cargo [120]. However, challenges such as cargo degradation, intracellular recycling, or re-release of intact vesicles into the extracellular space can impede effective therapeutic delivery [12].
Once internalized by target cells, the exosomal cargoes are released, impacting various cellular processes based on the nature of the cargo and the physiological state of the donor cell [121]. For instance, mRNA and miRNA within exosomes can modify gene expression patterns in recipient cells, potentially leading to the synthesis of new proteins. Similarly, proteins and lipids carried by exosomes can also activate or inhibit specific signaling pathways [32], while antigens delivered via exosomes may modulate immune responses, enhancing or suppressing them as required. For example, cargos carried by exosomes activate the Wnt/β-catenin pathway for collagen deposition; the PI3K/AKT/mTOR pathway can activate endothelial cells or fibroblast functions, while the VEGF pathway promotes angiogenesis in wound healing processes [122].
The specificity with which exosomes target recipient cells offers a significant advantage in designing targeted therapeutic strategies, thereby increasing efficacy, and minimizing side effects [119]. This targeting capability also allows for the in vitro manipulation of exosomes to load them with specific therapeutic agents, including drugs such as curcumin, doxorubicin, paclitaxel [123], RNA interference molecules, or other bioactive compounds. Due to their natural origin and biocompatibility, exosomes generally evade immediate immune detection, which is particularly advantageous in drug delivery applications. For example, in oncological therapies, exosomes derived from immune cells can be engineered to carry tumor-suppressive agents such as miRNA, miR-199a-3p [124], cisplatin, doxorubicin [124,125] directly to cancer cells, thus inhibiting tumor growth and progression [126]. This nuanced understanding of exosomal functions and their potential applications underscores the transformative possibilities of exosomes in medical science, particularly in targeted and precision therapies [127].

5. Current Attempts to Overcome Challenges Associated with the Therapeutic Application of Exosomes

To address these concerns, a task force from the International Society for Extracellular Vesicles (ISEV) Rigor and Standardization Committee has outlined the pre-analytical factors essential for the research on blood-derived exosomes, aiming to enhance the reproducibility of exosome isolation from blood samples.

5.1. Standardization of Research

Efforts to standardize clinical research on exosomes have predominantly concentrated on the optimization of isolation and characterization protocols [53]. However, the preanalytical phases, encompassing specimen handling, storage, and collection protocols, substantially influence the reproducibility and integrity of research outcomes [128,129]. Among the critical preanalytical variables, the transportation duration of biological specimens, such as blood, wound exudates, urine, and other bodily fluids, exhibits significant variability in terms of temperature and transit time, even within identical specimen types [130]. This variability can markedly affect experimental results and should be meticulously considered in data interpretation, especially when samples originate from multiple collection sites. Explicit documentation of these preanalytical variables in the methods section is imperative to evaluate their impact on the research findings and to facilitate cross-study comparisons.
The present market offers a variety of commercial kits, such as exosome isolation kits by System Biosciences, Thermo Fischer Scientific, Qiagen, Miletnyi, Norgen Biotek, designed to isolate exosomes from specific biological sources [130]. These kits employ various methodologies, including polymer precipitation, membrane affinity, antibody capture, and filtration to separate or concentrate exosomes. These kits, while beneficial under certain conditions, must be used with caution, as they lack comprehensive documentation on the principles of exosome isolation and enrichment [130,131,132]. The absence of detailed methodological information can lead to the introduction of unknown impurities, such as polyethylene glycol, which can compromise the purity of the exosome preparations and potentially skew the experimental results. Such contaminants could mislead the interpretation of the data, affecting the validity of the research findings [37].
Therefore, it is recommended to select commercial exosome isolation kits that provide detailed procedural information for reproducibility. Standardizing these procedures not only enhances the reliability of the research but also supports the broader scientific community in achieving consistent and verifiable results in exosome-based studies.

5.2. Reporting Standards

The integrity of the data within Vesiclepedia (http://www.microvesicles.org/), derived from both curated and author-submitted publications, is directly linked to the quality of the corresponding EV research [133,134]. The field of exosomes is characterized by a lack of stringent nomenclature and variability in the exosome isolation protocols across different studies and laboratories, significantly affecting the purity and type of the isolated exosomes, as well as their associated cargoes [1]. Furthermore, it is imperative to note that the recurrent identification of certain proteins in exosome studies does not necessarily qualify them as definitive exosome markers or proteins enriched in exosomes. Consequently, users are advised to exercise caution when employing Vesiclepedia data for further analysis, paying particular attention to the isolation methods and meta-annotations to select high-quality datasets pertinent to their research [135]. As of the latest update, Vesiclepedia comprises data from 3533 EV-related studies, marking an over twofold increase in the database’s catalog since the 2019 update. This expansion includes contributions of 56,691 proteins, 50,550 RNA, 3839 lipids, 192 metabolites, and 167 DNA entries [133]. Quantitative data, now available for 62,822 entries derived from 47 studies, represent a significant augmentation of Vesiclepedia’s utility. The database presently catalogs 252 sample sources from 56 distinct organisms. The latest enhancement to Vesiclepedia introduces EVQUANT, a novel feature facilitating the relative quantification of extracellular vesicle (EV) proteins, RNA, and lipid cargoes within individual studies [133]. Given the diversity in experimental methodologies and sample processing across studies, it is currently not feasible to perform quantitative cross-comparisons. Nevertheless, the advent of high-throughput data generation and the potential for establishing uniform analytical pipelines herald the possibility of cross-study comparisons, contingent upon the standardization of experimental procedures.

5.3. Accurately Defining the Vesicles

As per the MISEV 2023 guidelines [136], exosomes are defined as endosomal-derived intraluminal vesicles that form a specific subtype of small EVs with diameters less than 200 nm. This classification distinguishes them from other small EVs, such as ectosomes, which also form part of the broader EV population, but originate through different biogenetic processes. The terms “exosome” and “small EV” are not synonymous and should be used with precision. The lack of universal molecular markers for exosomes, ectosomes, and other EV subtypes presents a significant challenge in definitively characterizing these entities based on their origin. Consequently, much of the current research focuses broadly on mixed EV populations rather than exclusively on exosomes, pending clear evidence of their specific cellular origins [136].

5.4. Single-Exosome Studies

In the context of therapeutic applications, the characterization and validation of exosomes necessitate a comprehensive assessment, focusing on their size, shape, and molecular content [137]. Current methodologies for exosome characterization are broadly categorized into three primary domains: morphological analysis, size determination, and cargo profiling [138].
Morphological analysis employs techniques such as Scanning Electron Microscopy (SEM) and TEM, which facilitate the direct visualization of exosomal internal structures and surface topography, respectively [138]. Despite the detailed resolution offered by TEM, its intricate operation requirements [139] and labor-intensive sample preparation procedures limit its suitability for high-throughput analysis [140]. High-throughput analysis enables the simultaneous profiling of thousands of exosomes, facilitating the categorization of these vesicles based on their molecular signatures, such as lipids, proteins, and mRNA. This technology assists in identifying the origins and potential functions of exosomes [141]. Additionally, it can be employed to engineer exosomes and to screen them from various biological fluids, including blood, urine, and cerebrospinal fluid, enhancing their utility in disease diagnostics and prognostics [142]. This approach is instrumental in advancing our understanding of exosome-mediated processes and their implications in health and disease [143]. For size determination, methodologies such as nanoparticle tracking analysis (NTA), Dynamic Light Scattering (DLS), and tunable resistive pulse sensing (TRPS) are employed [144]. Among these, NTA stands out for its ability to provide high-resolution measurements, enabling the rapid identification and real-time observation of exosomes [145]. However, NTA’s capability to distinguish between exosomal particles and protein contaminants remains a challenge [146]. To overcome the limitations inherent in each characterization technique, it is a common practice to employ a multifaceted approach, integrating methods from each of the three domains, such as a combination of TEM, NTA, and protein-marker studies. This integrated strategy ensures a comprehensive characterization of exosomes, balancing the advantages and disadvantages of each method [137].
The diversity in biophysical and biochemical characteristics of exosomes is significantly influenced by the originating cell line and the employed isolation technique, leading to variability in the outcomes of biological analyses such as Western blots, PCR, and Dynamic Light Scattering (DLS) [147]. While traditional ensemble analysis methods have provided insights into the general biological and physical properties of exosomes, they offer limited resolution regarding the heterogeneity and individual characteristics of these vesicles. Consequently, these ensemble approaches, encompassing Western blot, enzyme-linked immunosorbent assay (ELISA), and Polymerase Chain Reaction (PCR), have been recognized for their limitations in accurately reflecting the complexity and diversity of exosomal populations [148].
In response to these limitations, advancements in analytical technologies have facilitated the shift towards single-particle analyses, enabling detailed investigation into the unique roles and properties of individual exosomes [149]. The guidelines proposed by the Minimal Information for Studies of Extracellular Vesicles (MISEV) advocate for the implementation of at least two single-particle analysis techniques to evaluate both the shape and biological attributes of single exosomes, emphasizing the importance of characterizing these vesicles at the individual level [150]. The development and refinement of single-exosome analysis techniques aim to address critical challenges associated with exosome research, including their intrinsic heterogeneity, measurement precision, complex biochemical composition, and nanoscale dimensions [11]. The increasing recognition of the need for precise sorting and phenotyping of specific exosome subpopulations has spurred the development of over twenty innovative single-vesicle methodologies [151]. Despite these advancements, the direct visualization of exosomes remains challenging due to their size, which often approaches the diffraction limit of standard optical microscopy. This limitation hampers the detailed examination of extracellular vesicle interactions and behaviors within cellular environments [152]. However, the advent of super-resolution imaging techniques, such as direct stochastic optical reconstruction microscopy (dSTORM), has opened new avenues for understanding the formation, function, and intracellular dynamics of extracellular vesicles [153]. dSTORM offers unprecedented sensitivity from free dyes or dye aggregations and resolution, enabling the precise determination of individual exosome arrangement, localization, and clustering through the tracking of vesicles in fluorescence mode with ultra-high single-molecule sensitivity [154,155,156]. Total internal reflection fluorescence (TIRF) microscopy, in synergy with single-molecule localization techniques, significantly enhances the signal-to-noise ratio and reduces the duration of imaging sessions [157]. However, super-resolution fluorescence microscopy demands sophisticated instrumentation and powerful data processing software, often resulting in low throughput [158]. While fluorescence microscopy techniques are pivotal for elucidating the mechanisms of exosome secretion, characterization, and uptake, the potential of fluorescent markers to interfere with their localization, activity, and functionality cannot be overlooked [150]. Additionally, lipid markers may exhibit non-specificity in their labeling properties or might aggregate [159].
In this context, flow cytometry (FCM) and nanoparticle tracking analysis (NTA) emerge as alternative approaches, leveraging the analysis of scattered light patterns from single particles and the tracking of light-scattering signals from particle diffusion, respectively, to derive physical insights at sub-wavelength scales [160]. Furthermore, fluorescence-based methodologies harness the interactions between light and molecules, employing fluorophore-conjugated target markers for labeling and detection [161]. The innovation of nano-flow cytometry (nFCM) by Prof. Yan Xiaomei’s laboratory represents a significant leap forward. By amalgamating Rayleigh light scattering with sheath flow single-molecule fluorescence detection technology, nFCM markedly enhances the sensitivity of both scattering and fluorescence detection relative to conventional FCM [162]. Capable of analyzing up to 10,000 particles per minute, this technology permits the multi-parametric quantitative measurement of single vesicles, down to a minimum particle size of 40 nm [150]. However, the advanced equipment and expertise required for nFCM impose limitations on its widespread clinical application. Furthermore, electron microscopy (EM) and atomic force microscopy (AFM) offer high-resolution imaging capabilities that allow for the direct visualization of exosomes, thus facilitating the characterization of their morphological and particle size attributes [163]. The application of these diverse nanotechnologies enables the comprehensive examination and cross-validation of the distinct traits and biological functions of individual extracellular vesicles (EVs) [164]. Consequently, researchers must judiciously select a combination of these techniques to effectively conduct their analyses. Therefore, the strategic integration of single-exosome analysis methodologies is a critical prerequisite for advancing our understanding of the physiological and pathological roles of exosomes [165]. This integrative approach not only elucidates the mechanisms underlying exosomal-mediated cell-to-cell communication but also paves the way for establishing a novel paradigm in exosome research [10,166].

5.5. Exosome Storage

Cryopreservation, lyophilization, and spray-drying are the predominant methodologies for the extended preservation of therapeutic exosomes, primarily relying on controlled temperature regulation and the inclusion of cryoprotective agents [167]. Although it is imperative to analyze exosomes in their native state post-isolation, for therapeutic purposes, extended shelf life is typically necessary. The stability of exosomes can vary, with some remaining intact without the need for freezing, depending on their biochemical composition and source of origin. Previous research has demonstrated that for long-term storage, a standard temperature of −80 °C is often used [168]. Specific studies have demonstrated that urinary exosomes can be preserved for up to four years at −20 °C, while saliva-derived exosomes may retain their protein composition and membrane integrity for up to 20 months when stored at 4 °C, although storage at this temperature can potentially diminish their protein content and biological activity [169].
The process of lyophilization offers an alternative storage solution, allowing exosomes to be easily preserved and reconstituted with the addition of an appropriate buffered solvent [170]. Recent studies suggest that lyophilization, particularly with the inclusion of cryoprotectants, can maintain the functionality of exosomal proteins and RNA for approximately four weeks, even at ambient temperatures [169]. The application of cryoprotectants is crucial in mitigating the adverse effects of freeze–thaw cycles, preventing cryodamage and aggregation of exosomes [171]. Formulations such as phosphate-buffered saline enriched with human albumin and the non-permeable disaccharide trehalose (PBS-HAT) have been found to enhance both short- and long-term stability and enhance both the short- and long-term exosome stability of therapeutic exosomes at −80 °C and across multiple freeze–thaw cycles [172,173]. The specifics of the freezing protocol (e.g., snap-freezing in liquid nitrogen vs. gradual freezing), the composition of the suspension buffer, storage duration, thawing techniques, and the number of freeze–thaw cycles are critical variables that need documentation, since optimal storage conditions can vary based on the exosomes’ origin and therapeutic composition [174]. To minimize detrimental freeze–thaw cycles, careful aliquoting is recommended, and it is crucial to recognize that samples subjected to varying numbers of freeze–thaw cycles may not be directly comparable. Proper labeling and documentation of storage containers are also essential to prevent the loss of exosomes due to adhesion to the container surfaces [174,175].

5.6. Modification of Exosomes for Specific Targeting

Exosomes, with their natural stability, low immunogenicity, and innate ability to target specific recipient cells represent an ideal vehicle for drug delivery [8]. These nanovesicles present a versatile platform for the bioengineering of therapeutic agents with minimal biochemical modification to enhance, broaden, or alter their therapeutic potentials [176]. The techniques for cargo loading into exosomes are categorized into pre- and post-biogenesis methodologies [177,178]. Pre-production strategies encompass methods such as transfection, co-incubation, and electroporation, implemented before the biogenesis of exosomes [179]. Post-production approaches include freeze–thaw cycles, incubation, sonication, extrusion, and hypotonic dialysis, applied after exosome formation [31,179,180]. For instance, Kim et al. utilized incubation and sonication to incorporate paclitaxel into exosomes derived from RAW 264.7 cells, aiming to counteract multidrug resistance in cancer therapy [181]. Similarly, Ohno et al. employed transfection to load anti-cancer let-7a miRNA into exosomes from HEK293 cells for breast cancer treatment [182].
The surface modification of exosomes represents another critical area of interest, achieved through the genetic manipulation of the exosomal membrane or the parental cells, the chemical conjugation of targeted ligands, electrostatic interactions, and the incorporation of magnetic nanoparticles [183]. The primary objective of these modifications is to achieve the targeted delivery of exosomes to specific cell types for precise therapeutic intervention [93]. Alvarez-Erviti et al. demonstrated targeted delivery to the central nervous system (CNS) by genetically engineering dendritic cells (DCs) to express Lamp2b fused with rabies viral glycoprotein (RVG) peptides [184]. In a similar vein, Zhu et al. chemically integrated c(RGDyK) tumor-targeting peptides onto exosomal surfaces to home in on glioblastoma cells [185].
Further innovations include the integration of α-EGFR, α-mCherry, and α-HER2 nanobodies onto the exosomal surface via phospholipid conjugation, effectively altering their targeting specificity in vitro [186]. This approach parallels strategies leveraging native exosomal membrane proteins, such as Lamp2b and platelet-derived growth factor receptors, as fusion partners for targeting ligands [186]. Additionally, chemical engineering methods such as click chemistry have been explored for surface modification, though the impact of these alterations on exosome–cell interaction dynamics and delivery efficacy remain under investigation [187]. Current research efforts are focused on elucidating the relationship between modified exosomal surface properties and their functional outcomes, with a view towards optimizing exosome-based therapeutic delivery systems [188,189].
Exosomes have pronounced membrane curvature as a consequence of their diminutive radius. This curvature arises through two principal mechanisms: first, ESCRT-mediated endosomal membrane deformation, leading to intraluminal vesicle budding within multivesicular endosomes [190]; second, neutral sphingomyelinase-induced ceramide production [191]. Based on these characteristics, an innovative approach to target lipid membranes effectively involves the design of peptides mimicking membrane-interacting proteins [82]. This approach could pioneer a new category of peptide sensors capable of simultaneous detection of phosphatidylserine (PS) and membrane curvature. Research findings indicate that the binding efficiency of the myristoylated alanine-rich C kinase substrate (MARCKS) effector domain (ED) is markedly reduced in the mutants MARCKSmut1 and MARCKSmut2, suggesting that MARCKS-ED can recognize PS-enriched, curved membranes [192]. This recognition is facilitated by phenylalanine (Phe) residues adapting to asymmetrically stretched bilayers and filling structural gaps in highly curved vesicles, thereby stabilizing membrane irregularities [193]. It was found that MARCKS-ED can differentiate between the sizes of lipid vesicles derived from both an animal model (rats) and synthetic phospholipid models, binding preferentially to highly curved membrane surfaces [193]. The effectiveness of curvature sensing is primarily dependent on the integration of aromatic Phe residues from the ED region into the lipid bilayers, as well as on the electrostatic interactions between the cationic residues (e.g., lysine, arginine) in the ED region and the negatively charged PS [194]. These lipid-targeting strategies offer new possibilities for probing exosomes in vivo or ex vivo, enhancing our ability to study critical biological phenomena such as apoptosis and vesicular shedding. Furthermore, MARCKS-ED-conjugated therapeutics could potentially achieve targeted delivery by binding to specific exosomes, followed by endocytosis, although the mechanisms of exosome cellular uptake remain under exploration [195].
In the context of infectious diseases, exosome-sized vesicles interconnecting red blood cells infected with malaria could enhance parasite survival under stress conditions [196]. MARCKS-ED targeting could disrupt intercellular communication via exosome-like vesicles between red blood cells infected with malaria, potentially delaying the development of drug resistance [197]. Advancing our understanding of exosomal functions in pathological conditions necessitates systematic peptide truncation to identify the minimal active sequence and essential residues for curvature detection by MARCKS-ED [194]. Enhancements in peptide affinity or specificity for specific exosomal populations could be attained through further residue modification and chemical optimization, including deletion, alanine scanning, and cyclization [198]. Investigating the membrane-binding capabilities of peptides derived from MARCKS-related proteins, which share nearly identical effector domains but differ by minor residue substitutions, could provide insights into how such modifications impact curvature sensing [194].
Further research involving systematic truncation is essential to determine the smallest active sequence and the minimal residues necessary for curvature sensing by MARCKS-ED [195]. Enhancements in binding affinity or specificity of MARCKS-derived peptides for cell-specific exosomes could be achieved through residue analysis and chemical optimization, including deletion, alanine scanning, and cyclization [199]. Additionally, evaluating the membrane-binding capabilities of peptide derivatives from a MARCKS homolog, which features a serine-to-proline substitution in the ED, could clarify whether this residue’s structural rigidity impacts curvature sensing [200]. An advanced method for visualizing the lipid composition and membrane curvature of exosomes would involve the application of bioorthogonal “click” chemistry to label the curvature probes with small molecule fluorophores, avoiding the use of large fluorescent proteins [201].
Elucidating the pharmacokinetic behavior of exosomes within biological systems and selecting an optimal delivery mechanism are paramount for determining the in vivo fate of these nanovesicles [202]. For the clinical advancement of exosome-based therapies (Figure 3) [26], it is imperative to consider several critical factors in the modification of exosome surfaces [203]. These include the selection of an appropriate targeting ligand or labeling agent, which requires a comprehensive understanding of the target site and the intended therapeutic application, all the while ensuring the absence of undesired immunogenic responses (Figure 4) [204]. Additionally, maintaining the physicochemical stability of exosomes necessitates the prevention of inadvertent alterations to their surface charge characteristics [176,204].
While covalent modification methods are frequently employed to affix labeling or targeting agents across the exosome surface, they present several challenges [205]. These techniques are limited to the attachment of genetically encodable proteins and peptides, excluding the possibility of targeting a broader range of molecules [206]. Moreover, the implementation of genetic modifications must not disrupt the intrinsic functions of proteins constituting the exosome membrane, given its complex nature [207].
The application of click chemistry for targeting specific proteins within the exosome membrane for modification is met with difficulties due to the membrane’s complexity [89]. However, advancements in the metabolic engineering of exosome parental cells have offered partial solutions to these challenges. This innovative approach involves the incorporation of artificially modified lipids, amino acids, and glycans to facilitate the expression of modified proteins on the exosome surface [208,209]. After this expression, additional chemical treatments are required to attach the desired labeling or targeting moieties, thus enabling the precision modification of exosomes for therapeutic purposes [209,210]. The utilization of click chemistry for exosome surface modification inherently poses a risk of chemical residue retention, which may elicit safety concerns regarding the modified exosomes and potentially complicate the regulatory approval process [211]. Additionally, the stability of aptamers under experimental conditions presents another significant challenge. Given these limitations, there is a growing preference for more efficient methods of exosome surface modification that circumvent these drawbacks [26,212].
An innovative approach emerging in this context involves the encapsulation of exosomes within phenol-metal-based nanofilms. This technique employs a coordination complex formed between tannic acid (TA) and Fe3+, onto which a targeting ligand can be functionalized via Michael addition to free amine groups [213]. For instance, this method has been applied to fabricate glutathione-capped gold nanoparticles by anchoring them to FA-functionalized, DOX-loaded exosomes (Exos-DOX-TA-Fe3+-FA), demonstrating the versatility and efficacy of this approach [213,214].
The TA-Fe3+ nanofilm offers commendable resistance against oxidants, heat, and ultraviolet (UV) light, presenting a robust platform for exosome modification [215]. However, it is crucial to assess the impact of such modifications on the structural integrity of the exosomes and the natural functions of their lipid, carbohydrate, and surface protein components [216]. Addressing these concerns meticulously is vital for harnessing the full potential of exosomes as an effective drug delivery system. Looking forward, the development of single-exosome analysis and the broader application of these novel technologies in a clinical setting will necessitate further clinical validation and the establishment of reproducibility [8]. This progression is essential for confirming the safety, efficacy, and reliability of exosome-based therapeutic delivery vehicles, paving the way for their successful integration into clinical practice [217].

6. Bio-Engineered Exosome Therapy in Precision Medicine

Exosome-based methodologies in precision medicine embody an integrative approach, combining molecular biology, nanotechnology, and individualized healthcare. They are poised to significantly enhance the prognosis, treatment, and monitoring of diseases in the future [218]. The foundational aspect of precision and personalized medicine involves the collection of patient-specific information, including lifestyle factors and genetic predispositions [219]. This information is crucial for devising tailored therapeutic strategies. Exosomes play a pivotal role in both the diagnosis of diseases and, potentially, preemptive therapeutic interventions, should early biomarkers be identified [220]. The advent of advanced “next generation” diagnostic techniques has facilitated the identification of myriad mutations and has enriched our understanding of the pathogenesis of various conditions, including cancers, neurological disorders, and infectious diseases [219,221]. These developments enable the formulation of individualized treatment regimens based on patient-specific biomarkers identified through exosomal analysis. A quantum leap in medical innovation is the concept of the “liquid biopsy,” a non-tissue-based biopsy method long-awaited for its minimal invasiveness [222,223]. This approach allows for the non-invasive screening, evaluation, monitoring, and diagnosis of diseases, garnering considerable interest due to its low-risk profile [219]. Recent advancements in liquid biopsy technologies have further streamlined the profiling of tumoral databases, encompassing methylation patterns and DNA/chromatin modifications [223]. Exosomes are integral to liquid biopsies, serving as carriers of biomarkers reflective of cellular conditions and being present across all body fluids [224]. The principal objective of precision medicine is the targeted treatment, or “ablation,” of pathological states with minimal collateral damage to healthy tissues, contingent upon the availability of comprehensive, personalized data sets [225]. Exosomes are particularly valuable in liquid biopsies for conditions such as pancreatic cancer, which are challenging to detect in their early, asymptomatic stages, and in circumstances where traditional tissue biopsies pose risks, such as during pregnancy [226].
Exosomes play a dual role in homeostatic processes; they can promote diseases, such as neuroinflammation and diabetes, and protect against them, facilitating immune homeostasis, tissue regeneration, and repair [227]. This dual functionality is pivotal as the utilization of exosomes as biomarkers and therapeutic agents in personalized medicine progresses [228]. Through interdisciplinary strategies, it is possible to load exosomes with specific cargoes, including DNA, RNA, oligonucleotides, proteins, and pharmaceuticals, drawing from extensive databases to maximize therapeutic efficacy. These modularized exosomes can be engineered with customizable surface molecules, enhancing their targeting capabilities, and minimizing damage to healthy tissues [224]. However, a significant challenge in the development of exosome-based personalized and precision medicine is the inherent heterogeneity of exosomes [8,229]. To address this complexity, a unified exosome profiling strategy that combines intradisciplinary expertise, advanced nanotechnology, and a multi-omics approach is essential. Additionally, comprehensive toxicological analysis is required to ensure the safety and effectiveness of exosome-based therapies. In this context, modularized exosomes, with their tailored compositions and targeting abilities, hold great promise for advancing the fields of precision and personalized medicine.
The bioengineering of exosomes is an excellent approach to develop proper production standards and quality, but it may impair the outcomes by inaccurate targeting due to over-modification and leads to unintended side effects [230]. In addition, these manipulations could trigger adverse immune reactions, causing inflammation. Complex modifications could complicate the regulatory approval process, delaying clinical application [231]. Scaling up the production of bioengineered EVs, including exosomes, for large-scale applications presents several challenges and requires optimizations [29]. The isolation method includes ultracentrifugation, the density-gradient method, immunoprecipitation, which are labor-intensive and not easily scalable. By developing high-throughput methods for rapid and accurate characterizations and ensuring batch-to-batch consistency in surface modifications, it is possible to scale up the production of bio-engineered exosomes for clinical and commercial applications. Furthermore, there is a concern related to safety, which arises from the co-isolation of proteins, lipids, and other cellular debris, which can contaminate exosome preparations [230]. This can be overcome by implementing multi-step purification processes, such as combining size-exclusion chromatography with affinity capture, which can enhance purity. Additionally, there are risks associated with the high shear forces during isolation and processing, which can damage exosomes. Therefore, using gentle processing techniques such as tangential flow filtration can minimize the damage and maintain exosome integrity by reducing shear stress and efficiently separating exosomes from other vesicles [230].

7. Future Perspective and Conclusions

As the field of exosome research continues to evolve, the horizon for their application in therapeutic contexts appears increasingly promising. The future of exosome-based therapies is poised at the confluence of technological innovation, deeper biological understanding, and regulatory refinement. Key areas of focus will likely include the enhancement of exosome isolation and purification techniques to achieve higher yield and purity, which is critical for clinical applications. Furthermore, the development of scalable manufacturing processes that can maintain the functional integrity of exosomes will be essential to meet the demands of clinical trials and subsequent therapeutic use. Advancements in the genetic and biochemical surface modification of exosomes will enable more precise targeting and cargo delivery, opening new avenues for the treatment of a wide range of diseases, from cancer to neurodegenerative disorders. The integration of cutting-edge technologies, such as CRISPR-Cas9 for genome editing within exosomes, could further enhance their therapeutic potential. Moreover, the exploration of synthetic and biomimetic exosomes presents an intriguing frontier that may overcome some of the limitations associated with natural exosomal systems. Regulatory considerations will remain at the forefront, with a need for standardized protocols and benchmarks that can streamline the approval process for exosome-based therapies. Collaborative efforts between researchers, clinicians, and regulatory agencies will be crucial to establishing a framework that ensures safety and efficacy without stifling innovation. It is necessary to conduct a thorough and time-consuming safety examination. When evaluating the regulatory needs for exosome therapeutic uses, the purity of exosomes is crucial. For approval, the FDA and other international regulatory bodies look for safety, effectiveness, potency, and purity. For therapeutic application, a pure product, devoid of impurities such proteins, peptides, cell-free DNA, and other cell detritus, is necessary [55]. Large animals or primates are now being used by more researchers to examine the safety and effectiveness of exosome products. Human safety regarding exosomes is becoming more widely acknowledged. Although the FDA has not yet authorized any exosome products for therapeutic use [22,23], they have provided specific guidelines to sponsors on how to supply the necessary chemistry, manufacturing, and control (CMC) data to ensure the safety of their products [232]. Furthermore, biologics intended for the mitigation, treatment, cure, or prevention of illness must meet strict regulations, set out by the FDA and other international regulatory bodies. Exosomes must be isolated and purified with strict quality, purity, potency, and repeatability standards. More controls are needed for the exosome alterations that follow. Exosome standards are probably going to include both the contents and the cells from where they originated. There needs to be more standardization and review of exosome release criteria. The exosome-based product age is almost upon us, thanks to advancements in separation technology and our growing understanding of exosomes [233]. Furthermore, the integration of computational biology, machine learning, and artificial intelligence in exosome research holds the potential to unravel complex biological interactions, predict therapeutic outcomes, and optimize treatment regimens. This multidisciplinary approach will not only enhance our understanding of exosomal functions but also streamline the development of exosome-based diagnostics and therapeutics.
In summary, the exploration of exosomes as vehicles for therapeutic delivery embodies a significant leap toward the realization of precision medicine and pharmaceutical innovation. Exosome-based therapies from bench to bedside are fraught with challenges, but they are undeniably paved with significant potential [234]. The journey from conceptual understanding to clinical application of exosome-based therapies encapsulates the collaborative effort of researchers, clinicians, and regulatory authorities, underscored by the commitment to scientific rigor and patient safety. The future of exosome-based therapies is not without its uncertainties, yet the foundation laid by current research efforts provides a robust basis for optimism. Continued exploration and innovation within this domain hold the promise of revolutionizing the way we approach disease treatment, offering hope for patient-centric, precision medical solutions that could fundamentally alter the therapeutic landscape. As we stand on the cusp of this promising frontier, the path forward is complex and demands a multidisciplinary effort. The evolution of exosome research stands as a testament to the relentless pursuit of knowledge and the quest for therapeutic innovation. The collective efforts of the scientific community, clinicians, and regulatory bodies are crucial for navigating the complex regulatory landscape, ultimately facilitating the delivery of these innovative therapies to patients, and heralding new paradigms in medical treatment and patient care. As we peer into the future, exosomes may well be at the heart of the next wave of breakthroughs in pharmaceutical sciences and medicine, offering a beacon of hope for patients around the globe.

Author Contributions

Conceptualization, S.G.; research and writing—original draft preparation, A.S., A.Y., A.N., and S.G.; supervision, S.G.; funding, S.G. All authors have read and agreed to the published version of the manuscript.

Funding

This study was primarily supported by the NIH R01 Grant DK129592 to S.G.

Conflicts of Interest

The authors declare no conflicts of interest. HDL (5–12 nm), LDL (18–25 nm), IDL (25–35 nm), VLDL.

Abbreviations

AFMatomic force microscopy
BMP-2bone morphogenetic protein 2
CMCchemistry, manufacturing, and control
CNScentral nervous system
DNAdeoxyribonucleic acid
dSTORMdirect stochastic optical reconstruction microscopy
DCsdendritic cells
DLSdynamic light scattering
EDeffector domain
EMelectron microscopy
ELISAenzyme-linked immunosorbent assay
ECMextracellular matrix
EVextracellular vesicle
FCMflow cytometry
FDAfood and drug administration
GMPgood manufacturing practice
HDLhigh density lipoproteins
HIF-1αhypoxia-inducible factor 1-alpha
IFN-γinterferon-gamma
ISEVinternational society for extracellular vesicles
INDinvestigational new drug
IDLintermediate-density lipoproteins
LDLlow density lipoproteins
LPSlipopolysaccharide
miRNAmicroRNA
MISEVminimal information for studies of extracellular vesicles
MARCKSmyristoylated alanine-rich C kinase substrate
NTAnanoparticle tracking analysis
nFCMnano-flow cytometry
PCRpolymerase chain reaction
PBS-HATphosphate buffered saline-human albumin and trehalose
PSphosphatidylserine
Phephenylalanine
qPCRquantitative polymerase chain reaction
RNAribonucleic acid
RVGrabies viral glycoprotein
SEMscanning electron microscopy
TNF-αtumor necrosis factor-alpha
TEMtransmission electron microscopy
TRPStunable resistive pulse sensing
TIRFtotal internal reflection fluorescence
TAtannic acid
UVultraviolet light
VLDLvery low-density lipoproteins

References

  1. Li, X.; Corbett, A.L.; Taatizadeh, E.; Tasnim, N.; Little, J.P.; Garnis, C.; Daugaard, M.; Guns, E.; Hoorfar, M.; Li, I.T.S. Challenges and opportunities in exosome research-Perspectives from biology, engineering, and cancer therapy. APL Bioeng. 2019, 3, 011503. [Google Scholar] [CrossRef] [PubMed]
  2. Zou, Z.; Li, H.; Xu, G.; Hu, Y.; Zhang, W.; Tian, K. Current Knowledge and Future Perspectives of Exosomes as Nanocarriers in Diagnosis and Treatment of Diseases. Int. J. Nanomed. 2023, 18, 4751–4778. [Google Scholar] [CrossRef] [PubMed]
  3. Rajput, A.; Varshney, A.; Bajaj, R.; Pokharkar, V. Exosomes as New Generation Vehicles for Drug Delivery: Biomedical Applications and Future Perspectives. Molecules 2022, 27, 7289. [Google Scholar] [CrossRef] [PubMed]
  4. Butreddy, A.; Kommineni, N.; Dudhipala, N. Exosomes as Naturally Occurring Vehicles for Delivery of Biopharmaceuticals: Insights from Drug Delivery to Clinical Perspectives. Nanomaterials 2021, 11, 1481. [Google Scholar] [CrossRef]
  5. Herrmann, I.K.; Wood, M.J.A.; Fuhrmann, G. Extracellular vesicles as a next-generation drug delivery platform. Nat. Nanotechnol. 2021, 16, 748–759. [Google Scholar] [CrossRef] [PubMed]
  6. Yang, L.; Yang, Y.; Chen, Y.; Xu, Y.; Peng, J. Cell-based drug delivery systems and their in vivo fate. Adv. Drug Deliv. Rev. 2022, 187, 114394. [Google Scholar] [CrossRef] [PubMed]
  7. Rezaie, J.; Feghhi, M.; Etemadi, T. A review on exosomes application in clinical trials: Perspective, questions, and challenges. Cell Commun. Signal. 2022, 20, 145. [Google Scholar] [CrossRef] [PubMed]
  8. Koh, H.; Kim, H.; Kang, S.; Yoo, T.-H. Exosome-Based Drug Delivery: Translation from Bench to Clinic. Pharmaceutics 2023, 15, 2042. [Google Scholar] [CrossRef] [PubMed]
  9. Sousa, P.; Lopes, B.; Sousa, A.C.; Moreira, A.; Coelho, A.; Alvites, R.; Alves, N.; Geuna, S.; Maurício, A.C. Advancements and Insights in Exosome-Based Therapies for Wound Healing: A Comprehensive Systematic Review (2018-June 2023). Biomedicines 2023, 11, 2099. [Google Scholar] [CrossRef]
  10. Gao, J.; Li, A.; Hu, J.; Feng, L.; Liu, L.; Shen, Z. Recent developments in isolating methods for exosomes. Front. Bioeng. Biotechnol. 2022, 10, 1100892. [Google Scholar] [CrossRef]
  11. Kwon, Y.; Park, J. Methods to analyze extracellular vesicles at single particle level. Micro Nano Syst. Lett. 2022, 10, 14. [Google Scholar] [CrossRef]
  12. Song, Y.; Kim, Y.; Ha, S.; Sheller-Miller, S.; Yoo, J.; Choi, C.; Park, C.H. The emerging role of exosomes as novel therapeutics: Biology, technologies, clinical applications, and the next. Am. J. Reprod. Immunol. 2021, 85, e13329. [Google Scholar] [CrossRef] [PubMed]
  13. Wang, X.; Xia, J.; Yang, L.; Dai, J.; He, L. Recent progress in exosome research: Isolation, characterization and clinical applications. Cancer Gene Ther. 2023, 30, 1051–1065. [Google Scholar] [CrossRef] [PubMed]
  14. Han, C.; Sun, X.; Liu, L.; Jiang, H.; Shen, Y.; Xu, X.; Li, J.; Zhang, G.; Huang, J.; Lin, Z.; et al. Exosomes and Their Therapeutic Potentials of Stem Cells. Stem Cells Int. 2016, 2016, 7653489. [Google Scholar] [CrossRef] [PubMed]
  15. Hong, P.; Yang, H.; Wu, Y.; Li, K.; Tang, Z. The functions and clinical application potential of exosomes derived from adipose mesenchymal stem cells: A comprehensive review. Stem Cell Res. Ther. 2019, 10, 242. [Google Scholar] [CrossRef] [PubMed]
  16. Tan, F.; Li, X.; Wang, Z.; Li, J.; Shahzad, K.; Zheng, J. Clinical applications of stem cell-derived exosomes. Signal Transduct. Target. Ther. 2024, 9, 17. [Google Scholar] [CrossRef]
  17. Asadpour, A.; Yahaya, B.H.; Bicknell, K.; Cottrell, G.S.; Widera, D. Uncovering the gray zone: Mapping the global landscape of direct-to-consumer businesses offering interventions based on secretomes, extracellular vesicles, and exosomes. Stem Cell Res. Ther. 2023, 14, 111. [Google Scholar] [CrossRef] [PubMed]
  18. Khan, A.Q.; Akhtar, S.; Prabhu, K.S.; Zarif, L.; Khan, R.; Alam, M.; Buddenkotte, J.; Ahmad, A.; Steinhoff, M.; Uddin, S. Exosomes: Emerging Diagnostic and Therapeutic Targets in Cutaneous Diseases. Int. J. Mol. Sci. 2020, 21, 9264. [Google Scholar] [CrossRef] [PubMed]
  19. Colao, I.L.; Corteling, R.; Bracewell, D.; Wall, I. Manufacturing Exosomes: A Promising Therapeutic Platform. Trends Mol. Med. 2018, 24, 242–256. [Google Scholar] [CrossRef]
  20. Li, M.; Fang, F.; Sun, M.; Zhang, Y.; Hu, M.; Zhang, J. Extracellular vesicles as bioactive nanotherapeutics: An emerging paradigm for regenerative medicine. Theranostics 2022, 12, 4879–4903. [Google Scholar] [CrossRef]
  21. Wang, Z.; Wang, X.; Xu, W.; Li, Y.; Lai, R.; Qiu, X.; Chen, X.; Chen, Z.; Mi, B.; Wu, M.; et al. Translational Challenges and Prospective Solutions in the Implementation of Biomimetic Delivery Systems. Pharmaceutics 2023, 15, 2623. [Google Scholar] [CrossRef] [PubMed]
  22. FDA. Public Safety Notification on Exosome Products. 2019. Available online: https://www.fda.gov/vaccines-blood-biologics/safety-availability-biologics/public-safety-notification-exosome-products (accessed on 20 April 2024).
  23. FDA. Consumer Alert on Regenerative Medicine Products Including Stem Cells and Exosomes. 2020. Available online: https://www.fda.gov/vaccines-blood-biologics/consumers-biologics/consumer-alert-regenerative-medicine-products-including-stem-cells-and-exosomes (accessed on 20 April 2024).
  24. Ahn, S.H.; Ryu, S.W.; Choi, H.; You, S.; Park, J.; Choi, C. Manufacturing Therapeutic Exosomes: From Bench to Industry. Mol. Cells 2022, 45, 284–290. [Google Scholar] [CrossRef] [PubMed]
  25. Thomas, S.C.; Kim, J.W.; Pauletti, G.M.; Hassett, D.J.; Kotagiri, N. Exosomes: Biological Pharmaceutical Nanovectors for Theranostics. Front. Bioeng. Biotechnol. 2021, 9, 808614. [Google Scholar] [CrossRef] [PubMed]
  26. Hussen, B.M.; Faraj, G.S.H.; Rasul, M.F.; Hidayat, H.J.; Salihi, A.; Baniahmad, A.; Taheri, M.; Ghafouri-Frad, S. Strategies to overcome the main challenges of the use of exosomes as drug carrier for cancer therapy. Cancer Cell Int. 2022, 22, 323. [Google Scholar] [CrossRef] [PubMed]
  27. Vyas, K.S.; Kaufman, J.; Munavalli, G.S.; Robertson, K.; Behfar, A.; Wyles, S.P. Exosomes: The latest in regenerative aesthetics. Regen. Med. 2023, 18, 181–194. [Google Scholar] [CrossRef] [PubMed]
  28. Chen, Y.-S.; Lin, E.-Y.; Chiou, T.-W.; Harn, H.-J. Exosomes in clinical trial and their production in compliance with good manufacturing practice. Tzu Chi Med. J. 2020, 32, 113–120. [Google Scholar] [CrossRef]
  29. Syromiatnikova, V.; Prokopeva, A.; Gomzikova, M. Methods of the Large-Scale Production of Extracellular Vesicles. Int. J. Mol. Sci. 2022, 23, 10522. [Google Scholar] [CrossRef] [PubMed]
  30. Qu, Q.; Fu, B.; Long, Y.; Liu, Z.Y.; Tian, X.H. Current Strategies for Promoting the Large-scale Production of Exosomes. Curr. Neuropharmacol. 2023, 21, 1964–1979. [Google Scholar] [CrossRef]
  31. Huang, G.; Lin, G.; Zhu, Y.; Duan, W.; Jin, D. Emerging technologies for profiling extracellular vesicle heterogeneity. Lab Chip 2020, 20, 2423–2437. [Google Scholar] [CrossRef]
  32. Yang, B.; Lin, Y.; Huang, Y.; Zhu, N.; Shen, Y.-Q. Extracellular vesicles modulate key signalling pathways in refractory wound healing. Burns Trauma 2023, 11, tkad039. [Google Scholar] [CrossRef]
  33. Willms, E.; Cabañas, C.; Mäger, I.; Wood, M.J.A.; Vader, P. Extracellular Vesicle Heterogeneity: Subpopulations, Isolation Techniques, and Diverse Functions in Cancer Progression. Front. Immunol. 2018, 9, 738. [Google Scholar] [CrossRef] [PubMed]
  34. Meggiolaro, A.; Moccia, V.; Brun, P.; Pierno, M.; Mistura, G.; Zappulli, V.; Ferraro, D. Microfluidic Strategies for Extracellular Vesicle Isolation: Towards Clinical Applications. Biosensors 2022, 13, 50. [Google Scholar] [CrossRef] [PubMed]
  35. Konoshenko, M.Y.; Lekchnov, E.A.; Vlassov, A.V.; Laktionov, P.P. Isolation of Extracellular Vesicles: General Methodologies and Latest Trends. Biomed. Res. Int. 2018, 2018, 8545347. [Google Scholar] [CrossRef] [PubMed]
  36. Coughlan, C.; Bruce, K.D.; Burgy, O.; Boyd, T.D.; Michel, C.R.; Garcia-Perez, J.E.; Adame, V.; Anton, P.; Bettcher, B.M.; Chial, H.J.; et al. Exosome Isolation by Ultracentrifugation and Precipitation and Techniques for Downstream Analyses. Curr. Protoc. Cell Biol. 2020, 88, e110. [Google Scholar] [CrossRef] [PubMed]
  37. Chen, J.; Li, P.; Zhang, T.; Xu, Z.; Huang, X.; Wang, R.; Du, L. Review on Strategies and Technologies for Exosome Isolation and Purification. Front. Bioeng. Biotechnol. 2022, 9, 811971. [Google Scholar] [CrossRef]
  38. Sidhom, K.; Obi, P.O.; Saleem, A. A Review of Exosomal Isolation Methods: Is Size Exclusion Chromatography the Best Option? Int. J. Mol. Sci. 2020, 21, 6466. [Google Scholar] [CrossRef] [PubMed]
  39. Yang, D.; Zhang, W.; Zhang, H.; Zhang, F.; Chen, L.; Ma, L.; Larcher, L.M.; Chen, S.; Liu, N.; Zhao, Q.; et al. Progress, opportunity, and perspective on exosome isolation—Efforts for efficient exosome-based theranostics. Theranostics 2020, 10, 3684–3707. [Google Scholar] [CrossRef] [PubMed]
  40. Liangsupree, T.; Multia, E.; Riekkola, M.-L. Modern isolation and separation techniques for extracellular vesicles. J. Chromatogr. A 2021, 1636, 461773. [Google Scholar] [CrossRef]
  41. Gholizadeh, S.; Shehata Draz, M.; Zarghooni, M.; Sanati-Nezhad, A.; Ghavami, S.; Shafiee, H.; Akbari, M. Microfluidic approaches for isolation, detection, and characterization of extracellular vesicles: Current status and future directions. Biosens. Bioelectron. 2017, 91, 588–605. [Google Scholar] [CrossRef]
  42. Zhou, X.; Brown, B.A.; Siegel, A.P.; El Masry, M.S.; Zeng, X.; Song, W.; Das, A.; Khandelwal, P.; Clark, A.; Singh, K.; et al. Exosome-Mediated Crosstalk between Keratinocytes and Macrophages in Cutaneous Wound Healing. ACS Nano 2020, 14, 12732–12748. [Google Scholar] [CrossRef]
  43. Guda, P.R.; Sharma, A.; Anthony, A.J.; El Masry, M.S.; Couse, A.D.; Ghatak, P.D.; Das, A.; Timsina, L.; Trinidad, J.C.; Roy, S.; et al. Nanoscopic and functional characterization of keratinocyte-originating exosomes in the wound fluid of non-diabetic and diabetic chronic wound patients. Nano Today 2023, 52, 101954. [Google Scholar] [CrossRef] [PubMed]
  44. Clayton, A.; Boilard, E.; Buzas, E.I.; Cheng, L.; Falcón-Perez, J.M.; Gardiner, C.; Gustafson, D.; Gualerzi, A.; Hendrix, A.; Hoffman, A.; et al. Considerations towards a roadmap for collection, handling and storage of blood extracellular vesicles. J. Extracell. Vesicles 2019, 8, 1647027. [Google Scholar] [CrossRef] [PubMed]
  45. Venturella, M.; Carpi, F.M.; Zocco, D. Standardization of Blood Collection and Processing for the Diagnostic Use of Extracellular Vesicles. Curr. Pathobiol. Rep. 2019, 7, 1–8. [Google Scholar] [CrossRef]
  46. Coumans, F.A.W.; Brisson, A.R.; Buzas, E.I.; Dignat-George, F.; Drees, E.E.E.; El-Andaloussi, S.; Emanueli, C.; Gasecka, A.; Hendrix, A.; Hill, A.F.; et al. Methodological Guidelines to Study Extracellular Vesicles. Circ. Res. 2017, 120, 1632–1648. [Google Scholar] [CrossRef] [PubMed]
  47. Lacroix, R.; Judicone, C.; Poncelet, P.; Robert, S.; Arnaud, L.; Sampol, J.; Dignat-George, F. Impact of pre-analytical parameters on the measurement of circulating microparticles: Towards standardization of protocol. J. Thromb. Haemost. 2012, 10, 437–446. [Google Scholar] [CrossRef] [PubMed]
  48. Buntsma, N.C.; Gąsecka, A.; Roos, Y.; van Leeuwen, T.G.; van der Pol, E.; Nieuwland, R. EDTA stabilizes the concentration of platelet-derived extracellular vesicles during blood collection and handling. Platelets 2022, 33, 764–771. [Google Scholar] [CrossRef] [PubMed]
  49. Lacroix, R.; Judicone, C.; Mooberry, M.; Boucekine, M.; Key, N.S.; Dignat-George, F. Standardization of pre-analytical variables in plasma microparticle determination: Results of the International Society on Thrombosis and Haemostasis SSC Collaborative workshop. J. Thromb. Haemost. 2013, 11, 1190–1193. [Google Scholar] [CrossRef] [PubMed]
  50. Holcar, M.; Kandušer, M.; Lenassi, M. Blood Nanoparticles—Influence on Extracellular Vesicle Isolation and Characterization. Front. Pharmacol. 2021, 12, 773844. [Google Scholar] [CrossRef] [PubMed]
  51. Vila-Liante, V.; Sánchez-López, V.; Martínez-Sales, V.; Ramón-Nuñez, L.A.; Arellano-Orden, E.; Cano-Ruiz, A.; Rodríguez-Martorell, F.J.; Gao, L.; Otero-Candelera, R. Impact of sample processing on the measurement of circulating microparticles: Storage and centrifugation parameters. Clin. Chem. Lab. Med. 2016, 54, 1759–1767. [Google Scholar]
  52. Aguilera-Rojas, M.; Sharbati, S.; Stein, T.; Candela Andrade, M.; Kohn, B.; Einspanier, R. Systematic analysis of different degrees of haemolysis on miRNA levels in serum and serum-derived extracellular vesicles from dogs. BMC Vet. Res. 2022, 18, 355. [Google Scholar] [CrossRef]
  53. Gurunathan, S.; Kang, M.H.; Jeyaraj, M.; Qasim, M.; Kim, J.H. Review of the Isolation, Characterization, Biological Function, and Multifarious Therapeutic Approaches of Exosomes. Cells 2019, 8, 307. [Google Scholar] [CrossRef] [PubMed]
  54. Mulcahy, L.A.; Pink, R.C.; Carter, D.R. Routes and mechanisms of extracellular vesicle uptake. J. Extracell. Vesicles 2014, 3, 24641. [Google Scholar] [CrossRef] [PubMed]
  55. Klyachko, N.L.; Arzt, C.J.; Li, S.M.; Gololobova, O.A.; Batrakova, E.V. Extracellular Vesicle-Based Therapeutics: Preclinical and Clinical Investigations. Pharmaceutics 2020, 12, 1171. [Google Scholar] [CrossRef] [PubMed]
  56. O’Brien, K.; Ughetto, S.; Mahjoum, S.; Nair, A.V.; Breakefield, X.O. Uptake, functionality, and re-release of extracellular vesicle-encapsulated cargo. Cell Rep. 2022, 39, 110651. [Google Scholar] [CrossRef] [PubMed]
  57. Mathieu, M.; Martin-Jaular, L.; Lavieu, G.; Théry, C. Specificities of secretion and uptake of exosomes and other extracellular vesicles for cell-to-cell communication. Nat. Cell Biol. 2019, 21, 9–17. [Google Scholar] [CrossRef] [PubMed]
  58. Heidarzadeh, M.; Zarebkohan, A.; Rahbarghazi, R.; Sokullu, E. Protein corona and exosomes: New challenges and prospects. Cell Commun. Signal. 2023, 21, 64. [Google Scholar] [CrossRef]
  59. Capolla, S.; Colombo, F.; De Maso, L.; Mauro, P.; Bertoncin, P.; Kähne, T.; Engler, A.; Núñez, L.; Spretz, R.; Larsen, G.; et al. Surface antibody changes protein corona both in human and mouse serum but not final opsonization and elimination of targeted polymeric nanoparticles. J. Nanobiotechnol. 2023, 21, 376. [Google Scholar] [CrossRef] [PubMed]
  60. Tóth, E.; Turiák, L.; Visnovitz, T.; Cserép, C.; Mázló, A.; Sódar, B.W.; Försönits, A.I.; Petővári, G.; Sebestyén, A.; Komlósi, Z.; et al. Formation of a protein corona on the surface of extracellular vesicles in blood plasma. J. Extracell. Vesicles 2021, 10, e12140. [Google Scholar] [CrossRef]
  61. Wolfram, J.; Yang, Y.; Shen, J.; Moten, A.; Chen, C.; Shen, H.; Ferrari, M.; Zhao, Y. The nano-plasma interface: Implications of the protein corona. Colloids Surf B Biointerfaces 2014, 124, 17–24. [Google Scholar] [CrossRef]
  62. Ghebosu, R.E.; Pendiuk Goncalves, J.; Wolfram, J. Extracellular Vesicle and Lipoprotein Interactions. Nano Lett. 2024, 24, 1–8. [Google Scholar] [CrossRef]
  63. Nguyen, V.H.; Lee, B.J. Protein corona: A new approach for nanomedicine design. Int. J. Nanomed. 2017, 12, 3137–3151. [Google Scholar] [CrossRef] [PubMed]
  64. Tenzer, S.; Docter, D.; Rosfa, S.; Wlodarski, A.; Kuharev, J.; Rekik, A.; Knauer, S.K.; Bantz, C.; Nawroth, T.; Bier, C.; et al. Nanoparticle size is a critical physicochemical determinant of the human blood plasma corona: A comprehensive quantitative proteomic analysis. ACS Nano 2011, 5, 7155–7167. [Google Scholar] [CrossRef] [PubMed]
  65. Shang, L.; Wang, Y.; Jiang, J.; Dong, S. pH-dependent protein conformational changes in albumin:gold nanoparticle bioconjugates: A spectroscopic study. Langmuir 2007, 23, 2714–2721. [Google Scholar] [CrossRef] [PubMed]
  66. Malonga, H.; Neault, J.F.; Tajmir-Riahi, H.A. Transfer RNA binding to human serum albumin: A model for protein-RNA interaction. DNA and Cell Biol. 2006, 25, 393–398. [Google Scholar] [CrossRef] [PubMed]
  67. Bertrand, N.; Grenier, P.; Mahmoudi, M.; Lima, E.M.; Appel, E.A.; Dormont, F.; Lim, J.M.; Karnik, R.; Langer, R.; Farokhzad, O.C. Mechanistic understanding of in vivo protein corona formation on polymeric nanoparticles and impact on pharmacokinetics. Nat Commun. 2017, 8, 777. [Google Scholar] [CrossRef]
  68. Carrillo-Conde, B.R.; Ramer-Tait, A.E.; Wannemuehler, M.J.; Narasimhan, B. Chemistry-dependent adsorption of serum proteins onto polyanhydride microparticles diferentially influences dendritic cell uptake and activation. Acta Biomater 2012, 8, 3618–3628. [Google Scholar] [CrossRef]
  69. Hiltbrunner, S.; Larssen, P.; Eldh, M.; Martinez-Bravo, M.-J.; Wagner, A.K.; Karlsson, M.C.; Gabrielsson, S. Exosomal cancer immunotherapy is independent of MHC molecules on exosomes. Oncotarget 2016, 7, 387. [Google Scholar] [CrossRef] [PubMed]
  70. Buzás, E.I.; Tóth, E.Á.; Sódar, B.W.; Szabó-Taylor, K.É. Molecular interactions at the surface of extracellular vesicles. Semin Immunopathol 2018, 40, 453–464. [Google Scholar] [CrossRef] [PubMed]
  71. Dietz, L.; Oberländer, J.; Mateos-Maroto, A.; Schunke, J.; Fichter, M.; Krämer-Albers, E.M.; Landfester, K.; Mailänder, V. Uptake of extracellular vesicles into immune cells is enhanced by the protein corona. J. Extracell. Vesicles 2023, 12, e12399. [Google Scholar] [CrossRef]
  72. Botha, J.; Handberg, A.; Simonsen, J.B. Lipid-based strategies used to identify extracellular vesicles in flow cytometry can be confounded by lipoproteins: Evaluations of annexin V, lactadherin, and detergent lysis. J. Extracell. Vesicles 2022, 11, e12200. [Google Scholar] [CrossRef]
  73. Busatto, S.; Walker, S.A.; Grayson, W.; Pham, A.; Tian, M.; Nesto, N.; Barklund, J.; Wolfram, J. Lipoprotein-based drug delivery. Adv. Drug Deliv. Rev. 2020, 159, 377–390. [Google Scholar] [CrossRef] [PubMed]
  74. Wu, M.; Chen, C.; Wang, Z.; Bachman, H.; Ouyang, Y.; Huang, P.H.; Sadovsky, Y.; Huang, T.J. Separating extracellular vesicles and lipoproteins via acoustofluidics. Lab Chip 2019, 19, 1174–1182. [Google Scholar] [CrossRef] [PubMed]
  75. Karimi, N.; Cvjetkovic, A.; Jang, S.C.; Crescitelli, R.; Feizi, M.; Nieuwland, R.; Lotvall, J.; Lässer, C. Detailed analysis of the plasma extracellular vesicle proteome after separation from lipoproteins. Cell. Mol. Life Sci. 2018, 75, 2873–2886. [Google Scholar] [CrossRef]
  76. Mo, Z.-C.; Ren, K.; Liu, X.; Tang, Z.-L.; Yi, G.-H. A high-density lipoprotein-mediated drug delivery system. Adv. Drug Deliv. Rev. 2016, 106, 132–147. [Google Scholar] [CrossRef] [PubMed]
  77. Park, H.J.; Kuai, R.; Jeon, E.J.; Seo, Y.; Jung, Y.; Moon, J.J.; Schwendeman, A.; Cho, S.W. High-density lipoprotein-mimicking nanodiscs carrying peptide for enhanced therapeutic angiogenesis in diabetic hindlimb ischemia. Biomaterials 2018, 161, 69–80. [Google Scholar] [CrossRef] [PubMed]
  78. Lozano-Andrés, E.; Enciso-Martinez, A.; Gijsbers, A.; Ridolfi, A.; Van Niel, G.; Libregts, S.; Pinheiro, C.; van Herwijnen, M.J.C.; Hendrix, A.; Brucale, M.; et al. Physical association of low density lipoprotein particles and extracellular vesicles unveiled by single particle analysis. J. Extracell. Vesicles 2023, 12, e12376. [Google Scholar] [CrossRef] [PubMed]
  79. Nguyen, P.; Le, A.; Pek, J.; Pham, T.; Jayasinghe, M.; Do, D.; Cao Dai, P.; Le, M. Extracellular vesicles and lipoproteins—Smart messengers of blood cells in the circulation. J. Extracell. Biol. 2022, 1, e49. [Google Scholar] [CrossRef]
  80. Ridolfi, A.; Conti, L.; Brucale, M.; Frigerio, R.; Cardellini, J.; Musicò, A.; Romano, M.; Zendrini, A.; Polito, L.; Bergamaschi, G.; et al. Particle profiling of EV-lipoprotein mixtures by AFM nanomechanical imaging. J. Extracell. Vesicles 2023, 12, e12349. [Google Scholar] [CrossRef] [PubMed]
  81. Donoso-Quezada, J.; Ayala-Mar, S.; González-Valdez, J. The role of lipids in exosome biology and intercellular communication: Function, analytics and applications. Traffic 2021, 22, 204–220. [Google Scholar] [CrossRef]
  82. Salatin, S.; Maleki Dizaj, S.; Yari Khosroushahi, A. Effect of the surface modification, size, and shape on cellular uptake of nanoparticles. Cell Biol. Int. 2015, 39, 881–890. [Google Scholar] [CrossRef]
  83. Caponnetto, F.; Manini, I.; Skrap, M.; Palmai-Pallag, T.; Di Loreto, C.; Beltrami, A.P.; Cesselli, D.; Ferrari, E. Size-dependent cellular uptake of exosomes. Nanomed. Nanotechnol. Biol. Med. 2017, 13, 1011–1020. [Google Scholar] [CrossRef] [PubMed]
  84. Gurung, S.; Perocheau, D.; Touramanidou, L.; Baruteau, J. The exosome journey: From biogenesis to uptake and intracellular signalling. Cell Commun. Signal. 2021, 19, 47. [Google Scholar] [CrossRef] [PubMed]
  85. Kim, H.I.; Park, J.; Zhu, Y.; Wang, X.; Han, Y.; Zhang, D. Recent advances in extracellular vesicles for therapeutic cargo delivery. Exp. Mol. Med. 2024, 56, 836–849. [Google Scholar] [CrossRef] [PubMed]
  86. van der Pol, E.; Coumans, F.A.; Grootemaat, A.E.; Gardiner, C.; Sargent, I.L.; Harrison, P.; Sturk, A.; van Leeuwen, T.G.; Nieuwland, R. Particle size distribution of exosomes and microvesicles determined by transmission electron microscopy, flow cytometry, nanoparticle tracking analysis, and resistive pulse sensing. J. Thromb. Haemost. 2014, 12, 1182–1192. [Google Scholar] [CrossRef] [PubMed]
  87. Gong, J.; Wang, H.X.; Leong, K.W. Determination of Cellular Uptake and Endocytic Pathways. Bio-protocol 2019, 9, e3169. [Google Scholar] [CrossRef] [PubMed]
  88. Di Bella, M.A. Overview and Update on Extracellular Vesicles: Considerations on Exosomes and Their Application in Modern Medicine. Biology 2022, 11, 804. [Google Scholar] [CrossRef] [PubMed]
  89. Raposo, G.; Stoorvogel, W. Extracellular vesicles: Exosomes, microvesicles, and friends. J. Cell Biol. 2013, 200, 373–383. [Google Scholar] [CrossRef] [PubMed]
  90. Karmacharya, M.; Kumar, S.; Cho, Y.K. Tuning the Extracellular Vesicles Membrane through Fusion for Biomedical Applications. J. Funct. Biomater. 2023, 14, 117. [Google Scholar] [CrossRef] [PubMed]
  91. Chen, H.; Wang, L.; Zeng, X.; Schwarz, H.; Nanda, H.S.; Peng, X.; Zhou, Y. Exosomes, a New Star for Targeted Delivery. Front. Cell Dev. Biol. 2021, 9, 751079. [Google Scholar] [CrossRef]
  92. Salunkhe, S.; Dheeraj; Basak, M.; Chitkara, D.; Mittal, A. Surface functionalization of exosomes for target-specific delivery and in vivo imaging & tracking: Strategies and significance. J. Control Release 2020, 326, 599–614. [Google Scholar]
  93. Tian, T.; Zhang, H.X.; He, C.P.; Fan, S.; Zhu, Y.L.; Qi, C.; Huang, N.P.; Xiao, Z.D.; Lu, Z.H.; Tannous, B.A.; et al. Surface functionalized exosomes as targeted drug delivery vehicles for cerebral ischemia therapy. Biomaterials 2018, 150, 137–149. [Google Scholar] [CrossRef] [PubMed]
  94. 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. [Google Scholar] [CrossRef] [PubMed]
  95. Li, Q.; Hu, W.; Huang, Q.; Yang, J.; Li, B.; Ma, K.; Wei, Q.; Wang, Y.; Su, J.; Sun, M.; et al. MiR146a-loaded engineered exosomes released from silk fibroin patch promote diabetic wound healing by targeting IRAK1. Signal Transduct. Target. Ther. 2023, 8, 62. [Google Scholar] [CrossRef] [PubMed]
  96. Shi, L.; Song, D.; Meng, C.; Cheng, Y.; Wang, B.; Yang, Z. Opportunities and challenges of engineered exosomes for diabetic wound healing. Giant 2024, 18, 100251. [Google Scholar] [CrossRef]
  97. Gartz, M.; Strande, J.L. Examining the Paracrine Effects of Exosomes in Cardiovascular Disease and Repair. J. Am. Heart Assoc. 2018, 7, e007954. [Google Scholar] [CrossRef]
  98. Yue, B. Biology of the extracellular matrix: An overview. J. Glaucoma 2014, 23 (Suppl. 1), S20–S23. [Google Scholar] [CrossRef] [PubMed]
  99. Wu, B.; Liu, D.A.; Guan, L.; Myint, P.K.; Chin, L.; Dang, H.; Xu, Y.; Ren, J.; Li, T.; Yu, Z.; et al. Stiff matrix induces exosome secretion to promote tumour growth. Nat. Cell Biol. 2023, 25, 415–424. [Google Scholar] [CrossRef] [PubMed]
  100. Logozzi, M.; Mizzoni, D.; Angelini, D.F.; Di Raimo, R.; Falchi, M.; Battistini, L.; Fais, S. Microenvironmental pH and Exosome Levels Interplay in Human Cancer Cell Lines of Different Histotypes. Cancers 2018, 10, 370. [Google Scholar] [CrossRef] [PubMed]
  101. Palomar-Alonso, N.; Lee, M.; Kim, M. Exosomes: Membrane-associated proteins, challenges and perspectives. Biochem. Biophys. Rep. 2024, 37, 101599. [Google Scholar] [CrossRef]
  102. Nakase, I.; Ueno, N.; Matsuzawa, M.; Noguchi, K.; Hirano, M.; Omura, M.; Takenaka, T.; Sugiyama, A.; Bailey Kobayashi, N.; Hashimoto, T.; et al. Environmental pH stress influences cellular secretion and uptake of extracellular vesicles. FEBS Open Bio. 2021, 11, 753–767. [Google Scholar] [CrossRef]
  103. Mahmood, A.; Otruba, Z.; Weisgerber, A.W.; Palay, M.D.; Nguyen, M.T.; Bills, B.L.; Knowles, M.K. Exosome secretion kinetics are controlled by temperature. Biophys. J. 2023, 122, 1301–1314. [Google Scholar] [CrossRef]
  104. Wang, Y.; Xiao, T.; Zhao, C.; Li, G. The Regulation of Exosome Generation and Function in Physiological and Pathological Processes. Int. J. Mol. Sci. 2023, 25, 255. [Google Scholar] [CrossRef]
  105. Whiteside, T.L. Tumor-Derived Exosomes and Their Role in Cancer Progression. Adv. Clin. Chem. 2016, 74, 103–141. [Google Scholar] [PubMed]
  106. Chiaradia, E.; Tancini, B.; Emiliani, C.; Delo, F.; Pellegrino, R.M.; Tognoloni, A.; Urbanelli, L.; Buratta, S. Extracellular Vesicles under Oxidative Stress Conditions: Biological Properties and Physiological Roles. Cells 2021, 10, 1763. [Google Scholar] [CrossRef] [PubMed]
  107. Kolesnikoff, N.; Chen, C.H.; Samuel, M.S. Interrelationships between the extracellular matrix and the immune microenvironment that govern epithelial tumour progression. Clin. Sci. 2022, 136, 361–377. [Google Scholar] [CrossRef]
  108. Jabbari, N.; Akbariazar, E.; Feqhhi, M.; Rahbarghazi, R.; Rezaie, J. Breast cancer-derived exosomes: Tumor progression and therapeutic agents. J. Cell Physiol. 2020, 235, 6345–6356. [Google Scholar] [CrossRef]
  109. Greening, D.W.; Gopal, S.K.; Xu, R.; Simpson, R.J.; Chen, W. Exosomes and their roles in immune regulation and cancer. Semin. Cell Dev. Biol. 2015, 40, 72–81. [Google Scholar] [CrossRef] [PubMed]
  110. Chaput, N.; Théry, C. Exosomes: Immune properties and potential clinical implementations. Semin. Immunopathol. 2011, 33, 419–440. [Google Scholar] [CrossRef] [PubMed]
  111. Shenoda, B.B.; Ajit, S.K. Modulation of Immune Responses by Exosomes Derived from Antigen-Presenting Cells. Clin. Med. Insights Pathol. 2016, 9 (Suppl. 1), 1–8. [Google Scholar] [CrossRef]
  112. Théry, C.; Amigorena, S. The cell biology of antigen presentation in dendritic cells. Curr. Opin. Immunol. 2001, 13, 45–51. [Google Scholar] [CrossRef]
  113. Théry, C.; Ostrowski, M.; Segura, E. Membrane vesicles as conveyors of immune responses. Nat. Rev. Immunol. 2009, 9, 581–593. [Google Scholar] [CrossRef] [PubMed]
  114. Iannotta, D.; Kijas, A.W.; Rowan, A.E.; Wolfram, J. Entry and exit of extracellular vesicles to and from the blood circulation. Nat. Nanotechnol. 2024, 19, 13–20. [Google Scholar] [CrossRef] [PubMed]
  115. Wang, Z.; Zhao, Z.; Gao, B.; Zhang, L. Exosome mediated biological functions within skeletal microenvironment. Front. Bioeng. Biotechnol. 2022, 10, 953916. [Google Scholar] [CrossRef] [PubMed]
  116. Wolfram, J.; Ferrari, M. Clinical Cancer Nanomedicine. Nano Today 2019, 25, 85–98. [Google Scholar] [CrossRef] [PubMed]
  117. 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. [Google Scholar] [CrossRef]
  118. Rudt, S.; Müller, R.H. In vitro phagocytosis assay of nano- and microparticles by chemiluminescence. III. Uptake of differently sized surface-modified particles, and its correlation to particle properties and in vivo distribution. Eur. J. Pharm. Sci. 1993, 1, 31–39. [Google Scholar]
  119. Kalluri, R. The biology and function of exosomes in cancer. J. Clin. Investig. 2016, 126, 1208–1215. [Google Scholar] [CrossRef]
  120. Dai, J.; Su, Y.; Zhong, S.; Cong, L.; Liu, B.; Yang, J.; Tao, Y.; He, Z.; Chen, C.; Jiang, Y. Exosomes: Key players in cancer and potential therapeutic strategy. Signal Transduct. Target. Ther. 2020, 5, 145. [Google Scholar] [CrossRef]
  121. Wei, H.; Chen, Q.; Lin, L.; Sha, C.; Li, T.; Liu, Y.; Yin, X.; Xu, Y.; Chen, L.; Gao, W.; et al. Regulation of exosome production and cargo sorting. Int. J. Biol. Sci. 2021, 17, 163–177. [Google Scholar] [CrossRef]
  122. Essola, J.M.; Zhang, M.; Yang, H.; Li, F.; Xia, B.; Mavoungou, J.F.; Hussain, A.; Huang, Y. Exosome regulation of immune response mechanism: Pros and cons in immunotherapy. Bioact. Mater. 2024, 32, 124–146. [Google Scholar] [CrossRef]
  123. Ha, D.; Yang, N.; Nadithe, V. Exosomes as therapeutic drug carriers and delivery vehicles across biological membranes: Current perspectives and future challenges. Acta Pharm. Sin. B 2016, 6, 287–296. [Google Scholar] [CrossRef]
  124. Kobayashi, M.; Sawada, K.; Miyamoto, M.; Shimizu, A.; Yamamoto, M.; Kinose, Y.; Nakamura, K.; Kawano, M.; Kodama, M.; Hashimoto, K.; et al. Exploring the potential of engineered exosomes as delivery systems for tumor-suppressor microRNA replacement therapy in ovarian cancer. Biochem. Biophys. Res. Commun. 2020, 527, 153–161. [Google Scholar] [CrossRef] [PubMed]
  125. Raguraman, R.; Bhavsar, D.; Kim, D.; Ren, X.; Sikavitsas, V.; Munshi, A.; Ramesh, R. Tumor-targeted exosomes for delivery of anticancer drugs. Cancer Lett. 2023, 558, 216093. [Google Scholar] [CrossRef] [PubMed]
  126. Kase, Y.; Uzawa, K.; Wagai, S.; Yoshimura, S.; Yamamoto, J.-I.; Toeda, Y.; Okubo, M.; Eizuka, K.; Ando, T.; Nobuchi, T.; et al. Engineered exosomes delivering specific tumor-suppressive RNAi attenuate oral cancer progression. Sci. Rep. 2021, 11, 5897. [Google Scholar] [CrossRef]
  127. Perocheau, D.; Touramanidou, L.; Gurung, S.; Gissen, P.; Baruteau, J. Clinical applications for exosomes: Are we there yet? Br. J. Pharmacol. 2021, 178, 2375–2392. [Google Scholar] [CrossRef] [PubMed]
  128. Agrawal, L.; Engel, K.B.; Greytak, S.R.; Moore, H.M. Understanding preanalytical variables and their effects on clinical biomarkers of oncology and immunotherapy. Semin. Cancer Biol. 2018, 52 Pt 2, 26–38. [Google Scholar] [CrossRef]
  129. Nieuwland, R.; Siljander, P.R.M.; Falcón-Pérez, J.M.; Witwer, K.W. Reproducibility of extracellular vesicle research. Eur. J. Cell Biol. 2022, 101, 151226. [Google Scholar] [CrossRef]
  130. Yakubovich, E.I.; Polischouk, A.G.; Evtushenko, V.I. Principles and Problems of Exosome Isolation from Biological Fluids. Biochem. Suppl. Ser. A Membr. Cell Biol. 2022, 16, 115–126. [Google Scholar] [CrossRef]
  131. Helwa, I.; Cai, J.; Drewry, M.D.; Zimmerman, A.; Dinkins, M.B.; Khaled, M.L.; Seremwe, M.; Dismuke, W.M.; Bieberich, E.; Stamer, W.D.; et al. A Comparative Study of Serum Exosome Isolation Using Differential Ultracentrifugation and Three Commercial Reagents. PLoS ONE 2017, 12, e0170628. [Google Scholar] [CrossRef]
  132. Soares Martins, T.; Catita, J.; Martins Rosa, I.; AB da Cruz e Silva, O.; Henriques, A.G. Exosome isolation from distinct biofluids using precipitation and column-based approaches. PLoS ONE 2018, 13, e0198820. [Google Scholar] [CrossRef]
  133. Chitti, S.V.; Gummadi, S.; Kang, T.; Shahi, S.; Marzan, A.L.; Nedeva, C.; Sanwlani, R.; Bramich, K.; Stewart, S.; Petrovska, M.; et al. Vesiclepedia 2024: An extracellular vesicles and extracellular particles repository. Nucleic Acids Res. 2023, 52, D1694–D1698. [Google Scholar] [CrossRef] [PubMed]
  134. Kalra, H.; Simpson, R.J.; Ji, H.; Aikawa, E.; Altevogt, P.; Askenase, P.; Bond, V.C.; Borràs, F.E.; Breakefield, X.; Budnik, V.; et al. Vesiclepedia: A compendium for extracellular vesicles with continuous community annotation. PLoS Biol. 2012, 10, e1001450. [Google Scholar] [CrossRef] [PubMed]
  135. Witwer, K.W.; Buzás, E.I.; Bemis, L.T.; Bora, A.; Lässer, C.; Lötvall, J.; Nolte-’t Hoen, E.N.; Piper, M.G.; Sivaraman, S.; Skog, J.; et al. Standardization of sample collection, isolation and analysis methods in extracellular vesicle research. J. Extracell. Vesicles 2013, 2, 20360. [Google Scholar] [CrossRef]
  136. Welsh, J.A.; Goberdhan, D.C.I.; O’Driscoll, L.; Buzas, E.I.; Blenkiron, C.; Bussolati, B.; Cai, H.; Di Vizio, D.; Driedonks, T.A.P.; Erdbrügger, U.; et al. Minimal information for studies of extracellular vesicles (MISEV2023): From basic to advanced approaches. J. Extracell. Vesicles 2024, 13, e12404. [Google Scholar] [CrossRef] [PubMed]
  137. Lai, J.J.; Chau, Z.L.; Chen, S.Y.; Hill, J.J.; Korpany, K.V.; Liang, N.W.; Lin, L.H.; Lin, Y.H.; Liu, J.K.; Liu, Y.C.; et al. Exosome Processing and Characterization Approaches for Research and Technology Development. Adv. Sci. 2022, 9, e2103222. [Google Scholar] [CrossRef]
  138. Hartjes, T.A.; Mytnyk, S.; Jenster, G.W.; van Steijn, V.; van Royen, M.E. Extracellular Vesicle Quantification and Characterization: Common Methods and Emerging Approaches. Bioengineering 2019, 6, 7. [Google Scholar] [CrossRef] [PubMed]
  139. Corona, M.L.; Hurbain, I.; Raposo, G.; van Niel, G. Characterization of Extracellular Vesicles by Transmission Electron Microscopy and Immunolabeling Electron Microscopy. Methods Mol. Biol. 2023, 2668, 33–43. [Google Scholar]
  140. Tanasi, I.; Adamo, A.; Kamga, P.T.; Bazzoni, R.; Krampera, M. High-throughput analysis and functional interpretation of extracellular vesicle content in hematological malignancies. Comput. Struct. Biotechnol. J. 2020, 18, 2670–2677. [Google Scholar] [CrossRef]
  141. Jiang, C.; Fu, Y.; Liu, G.; Shu, B.; Davis, J.; Tofaris, G.K. Multiplexed Profiling of Extracellular Vesicles for Biomarker Development. Nano-Micro Lett. 2021, 14, 3. [Google Scholar] [CrossRef]
  142. He, A.; Wang, M.; Li, X.; Chen, H.; Lim, K.; Lu, L.; Zhang, C. Role of Exosomes in the Pathogenesis and Theranostic of Alzheimer’s Disease and Parkinson’s Disease. Int. J. Mol. Sci. 2023, 24, 11054. [Google Scholar] [CrossRef]
  143. D’Argenio, V. The High-Throughput Analyses Era: Are We Ready for the Data Struggle? High Throughput 2018, 7, 8. [Google Scholar] [CrossRef] [PubMed]
  144. Huber, M.J.; Ivleva, N.P.; Booth, A.M.; Beer, I.; Bianchi, I.; Drexel, R.; Geiss, O.; Mehn, D.; Meier, F.; Molska, A.; et al. Physicochemical characterization and quantification of nanoplastics: Applicability, limitations and complementarity of batch and fractionation methods. Anal. Bioanal. Chem. 2023, 415, 3007–3031. [Google Scholar] [CrossRef] [PubMed]
  145. Omrani, M.; Beyrampour-Basmenj, H.; Jahanban-Esfahlan, R.; Talebi, M.; Raeisi, M.; Serej, Z.A.; Akbar-Gharalari, N.; Khodakarimi, S.; Wu, J.; Ebrahimi-Kalan, A. Global trend in exosome isolation and application: An update concept in management of diseases. Mol. Cell Biochem. 2024, 479, 679–691. [Google Scholar] [CrossRef] [PubMed]
  146. Allelein, S.; Medina-Perez, P.; Lopes, A.L.H.; Rau, S.; Hause, G.; Kölsch, A.; Kuhlmeier, D. Potential and challenges of specifically isolating extracellular vesicles from heterogeneous populations. Sci. Rep. 2021, 11, 11585. [Google Scholar] [CrossRef] [PubMed]
  147. Kurian, T.K.; Banik, S.; Gopal, D.; Chakrabarti, S.; Mazumder, N. Elucidating Methods for Isolation and Quantification of Exosomes: A Review. Mol. Biotechnol. 2021, 63, 249–266. [Google Scholar] [CrossRef] [PubMed]
  148. Rastogi, S.; Sharma, V.; Bharti, P.S.; Rani, K.; Modi, G.P.; Nikolajeff, F.; Kumar, S. The Evolving Landscape of Exosomes in Neurodegenerative Diseases: Exosomes Characteristics and a Promising Role in Early Diagnosis. Int. J. Mol. Sci. 2021, 22, 440. [Google Scholar] [CrossRef] [PubMed]
  149. Hilton, S.H.; White, I.M. Advances in the analysis of single extracellular vesicles: A critical review. Sens. Actuators Rep. 2021, 3, 100052. [Google Scholar] [CrossRef]
  150. Zhu, J.; Wu, F.; Li, C.; Mao, J.; Wang, Y.; Zhou, X.; Xie, H.; Wen, C. Application of Single Extracellular Vesicle Analysis Techniques. Int. J. Nanomed. 2023, 18, 5365–5376. [Google Scholar] [CrossRef]
  151. Groot Kormelink, T.; Arkesteijn, G.J.; Nauwelaers, F.A.; van den Engh, G.; Nolte-’t Hoen, E.N.; Wauben, M.H. Prerequisites for the analysis and sorting of extracellular vesicle subpopulations by high-resolution flow cytometry. Cytometry A 2016, 89, 135–147. [Google Scholar] [CrossRef]
  152. McNamara, R.P.; Zhou, Y.; Eason, A.B.; Landis, J.T.; Chambers, M.G.; Willcox, S.; Peterson, T.A.; Schouest, B.; Maness, N.J.; MacLean, A.G.; et al. Imaging of surface microdomains on individual extracellular vesicles in 3-D. J. Extracell. Vesicles 2022, 11, e12191. [Google Scholar] [CrossRef]
  153. Nizamudeen, Z.; Markus, R.; Lodge, R.; Parmenter, C.; Platt, M.; Chakrabarti, L.; Sottile, V. Rapid and accurate analysis of stem cell-derived extracellular vesicles with super resolution microscopy and live imaging. Biochim. Biophys. Acta Mol. Cell Res. 2018, 1865, 1891–1900. [Google Scholar] [CrossRef]
  154. Bond, C.; Santiago-Ruiz, A.N.; Tang, Q.; Lakadamyali, M. Technological advances in super-resolution microscopy to study cellular processes. Mol. Cell 2022, 82, 315–332. [Google Scholar] [CrossRef]
  155. Endesfelder, U.; Heilemann, M. Direct Stochastic Optical Reconstruction Microscopy (dSTORM). Methods Mol. Biol. 2015, 1251, 263–276. [Google Scholar]
  156. Lehmann, M.; Gottschalk, B.; Puchkov, D.; Schmieder, P.; Schwagerus, S.; Hackenberger, C.P.; Haucke, V.; Schmoranzer, J. Multicolor Caged dSTORM Resolves the Ultrastructure of Synaptic Vesicles in the Brain. Angew. Chem. Int. Ed. 2015, 54, 13230–13235. [Google Scholar] [CrossRef]
  157. Kudalkar, E.M.; Davis, T.N.; Asbury, C.L. Single-Molecule Total Internal Reflection Fluorescence Microscopy. Cold Spring Harb. Protoc. 2016, 2016, pdb.top077800. [Google Scholar] [CrossRef]
  158. Ter-Ovanesyan, D.; Kowal, E.J.K.; Regev, A.; Church, G.M.; Cocucci, E. Imaging of Isolated Extracellular Vesicles Using Fluorescence Microscopy. Methods Mol. Biol. 2017, 1660, 233–241. [Google Scholar]
  159. Skotland, T.; Hessvik, N.P.; Sandvig, K.; Llorente, A. Exosomal lipid composition and the role of ether lipids and phosphoinositides in exosome biology. J. Lipid Res. 2019, 60, 9–18. [Google Scholar] [CrossRef]
  160. Gardiner, C.; Shaw, M.; Hole, P.; Smith, J.; Tannetta, D.; Redman, C.W.; Sargent, I.L. Measurement of refractive index by nanoparticle tracking analysis reveals heterogeneity in extracellular vesicles. J. Extracell. Vesicles 2014, 3, 25361. [Google Scholar] [CrossRef]
  161. Hickey, S.M.; Ung, B.; Bader, C.; Brooks, R.; Lazniewska, J.; Johnson, I.R.D.; Sorvina, A.; Logan, J.; Martini, C.; Moore, C.R.; et al. Fluorescence Microscopy-An Outline of Hardware, Biological Handling, and Fluorophore Considerations. Cells 2021, 11, 35. [Google Scholar] [CrossRef] [PubMed]
  162. Zhu, S.; Ma, L.; Wang, S.; Chen, C.; Zhang, W.; Yang, L.; Hang, W.; Nolan, J.P.; Wu, L.; Yan, X. Light-scattering detection below the level of single fluorescent molecules for high-resolution characterization of functional nanoparticles. ACS Nano 2014, 8, 10998–11006. [Google Scholar] [CrossRef] [PubMed]
  163. Sharma, S.; LeClaire, M.; Gimzewski, J.K. Ascent of atomic force microscopy as a nanoanalytical tool for exosomes and other extracellular vesicles. Nanotechnology 2018, 29, 132001. [Google Scholar] [CrossRef]
  164. Shao, H.; Im, H.; Castro, C.M.; Breakefield, X.; Weissleder, R.; Lee, H. New Technologies for Analysis of Extracellular Vesicles. Chem. Rev. 2018, 118, 1917–1950. [Google Scholar] [CrossRef]
  165. Li, X.H.; Zhang, J.; Li, D.F.; Wu, W.; Xie, Z.W.; Liu, Q. Physiological and pathological insights into exosomes in the brain. Zool. Res. 2020, 41, 365–372. [Google Scholar] [CrossRef]
  166. Liu, S.-L.; Sun, P.; Li, Y.; Liu, S.-S.; Lu, Y. Exosomes as critical mediators of cell-to-cell communication in cancer pathogenesis and their potential clinical application. Transl. Cancer Res. 2019, 8, 298–311. [Google Scholar] [CrossRef]
  167. Bahr, M.M.; Amer, M.S.; Abo-El-Sooud, K.; Abdallah, A.N.; El-Tookhy, O.S. Preservation techniques of stem cells extracellular vesicles: A gate for manufacturing of clinical grade therapeutic extracellular vesicles and long-term clinical trials. Int. J. Vet. Sci. Med. 2020, 8, 1–8. [Google Scholar] [CrossRef]
  168. Wu, J.Y.; Li, Y.J.; Hu, X.B.; Huang, S.; Xiang, D.X. Preservation of small extracellular vesicles for functional analysis and therapeutic applications: A comparative evaluation of storage conditions. Drug Deliv. 2021, 28, 162–170. [Google Scholar] [CrossRef]
  169. Jeyaram, A.; Jay, S.M. Preservation and Storage Stability of Extracellular Vesicles for Therapeutic Applications. AAPS J. 2017, 20, 1–7. [Google Scholar] [CrossRef]
  170. Qazi, R.E.M.; Sajid, Z.; Zhao, C.; Hussain, I.; Iftikhar, F.; Jameel, M.; Rehman, F.U.; Mian, A.A. Lyophilization Based Isolation of Exosomes. Int. J. Mol. Sci. 2023, 24, 10477. [Google Scholar] [CrossRef]
  171. Cottle, C.; Porter, A.P.; Lipat, A.; Turner-Lyles, C.; Nguyen, J.; Moll, G.; Chinnadurai, R. Impact of Cryopreservation and Freeze-Thawing on Therapeutic Properties of Mesenchymal Stromal/Stem Cells and Other Common Cellular Therapeutics. Curr. Stem Cell Rep. 2022, 8, 72–92. [Google Scholar] [CrossRef] [PubMed]
  172. Görgens, A.; Corso, G.; Hagey, D.W.; Jawad Wiklander, R.; Gustafsson, M.O.; Felldin, U.; Lee, Y.; Bostancioglu, R.B.; Sork, H.; Liang, X.; et al. Identification of storage conditions stabilizing extracellular vesicles preparations. J. Extracell. Vesicles 2022, 11, e12238. [Google Scholar] [CrossRef] [PubMed]
  173. Kusuma, G.D.; Barabadi, M.; Tan, J.L.; Morton, D.A.V.; Frith, J.E.; Lim, R. To Protect and to Preserve: Novel Preservation Strategies for Extracellular Vesicles. Front. Pharmacol. 2018, 9, 417722. [Google Scholar] [CrossRef]
  174. Gelibter, S.; Marostica, G.; Mandelli, A.; Siciliani, S.; Podini, P.; Finardi, A.; Furlan, R. The impact of storage on extracellular vesicles: A systematic study. J. Extracell. Vesicles 2022, 11, e12162. [Google Scholar] [CrossRef]
  175. Sivanantham, A.; Jin, Y. Impact of Storage Conditions on EV Integrity/Surface Markers and Cargos. Life 2022, 12, 697. [Google Scholar] [CrossRef] [PubMed]
  176. Sadeghi, S.; Tehrani, F.R.; Tahmasebi, S.; Shafiee, A.; Hashemi, S.M. Exosome engineering in cell therapy and drug delivery. Inflammopharmacology 2023, 31, 145–169. [Google Scholar] [CrossRef] [PubMed]
  177. Zeng, H.; Guo, S.; Ren, X.; Wu, Z.; Liu, S.; Yao, X. Current Strategies for Exosome Cargo Loading and Targeting Delivery. Cells 2023, 12, 1416. [Google Scholar] [CrossRef]
  178. Chen, H.; Yao, H.; Chi, J.; Li, C.; Liu, Y.; Yang, J.; Yu, J.; Wang, J.; Ruan, Y.; Pi, J.; et al. Engineered exosomes as drug and RNA co-delivery system: New hope for enhanced therapeutics? Front. Bioeng. Biotechnol. 2023, 11, 1254356. [Google Scholar] [CrossRef] [PubMed]
  179. Patel, G.; Agnihotri, T.G.; Gitte, M.; Shinde, T.; Gomte, S.S.; Goswami, R.; Jain, A. Exosomes: A potential diagnostic and treatment modality in the quest for counteracting cancer. Cell Oncol. 2023, 46, 1159–1179. [Google Scholar] [CrossRef] [PubMed]
  180. Xu, M.; Yang, Q.; Sun, X.; Wang, Y. Recent Advancements in the Loading and Modification of Therapeutic Exosomes. Front. Bioeng. Biotechnol. 2020, 8, 586130. [Google Scholar] [CrossRef]
  181. Kim, M.S.; Haney, M.J.; Zhao, Y.; Mahajan, V.; Deygen, I.; Klyachko, N.L.; Inskoe, E.; Piroyan, A.; Sokolsky, M.; Okolie, O.; et al. Development of exosome-encapsulated paclitaxel to overcome MDR in cancer cells. Nanomedicine 2016, 12, 655–664. [Google Scholar] [CrossRef]
  182. 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. 2013, 21, 185–191. [Google Scholar] [CrossRef]
  183. Kučuk, N.; Primožič, M.; Knez, Ž.; Leitgeb, M. Exosomes Engineering and Their Roles as Therapy Delivery Tools, Therapeutic Targets, and Biomarkers. Int. J. Mol. Sci. 2021, 22, 9543. [Google Scholar] [CrossRef]
  184. Xu, M.; Feng, T.; Liu, B.; Qiu, F.; Xu, Y.; Zhao, Y.; Zheng, Y. Engineered exosomes: Desirable target-tracking characteristics for cerebrovascular and neurodegenerative disease therapies. Theranostics 2021, 11, 8926–8944. [Google Scholar] [CrossRef]
  185. Zhu, Q.; Ling, X.; Yang, Y.; Zhang, J.; Li, Q.; Niu, X.; Hu, G.; Chen, B.; Li, H.; Wang, Y.; et al. Embryonic Stem Cells-Derived Exosomes Endowed with Targeting Properties as Chemotherapeutics Delivery Vehicles for Glioblastoma Therapy. Adv. Sci. 2019, 6, 1801899. [Google Scholar] [CrossRef]
  186. Pham, T.C.; Jayasinghe, M.K.; Pham, T.T.; Yang, Y.; Wei, L.; Usman, W.M.; Chen, H.; Pirisinu, M.; Gong, J.; Kim, S.; et al. Covalent conjugation of extracellular vesicles with peptides and nanobodies for targeted therapeutic delivery. J. Extracell. Vesicles 2021, 10, e12057. [Google Scholar] [CrossRef]
  187. Wang, J.; Wang, M.; Jiang, N.; Ding, S.; Peng, Q.; Zheng, L. Emerging chemical engineering of exosomes as “bioscaffolds” in diagnostics and therapeutics. Genes Dis. 2023, 10, 1494–1512. [Google Scholar] [CrossRef]
  188. Sharma, V.; Mukhopadhyay, C.D. Exosome as drug delivery system: Current advancements. Extracell. Vesicle 2024, 3, 100032. [Google Scholar] [CrossRef]
  189. Li, T.; Li, X.; Han, G.; Liang, M.; Yang, Z.; Zhang, C.; Huang, S.; Tai, S.; Yu, S. The Therapeutic Potential and Clinical Significance of Exosomes as Carriers of Drug Delivery System. Pharmaceutics 2022, 15, 21. [Google Scholar] [CrossRef]
  190. Krylova, S.V.; Feng, D. The Machinery of Exosomes: Biogenesis, Release, and Uptake. Int. J. Mol. Sci. 2023, 24, 1337. [Google Scholar] [CrossRef]
  191. Ghadami, S.; Dellinger, K. The lipid composition of extracellular vesicles: Applications in diagnostics and therapeutic delivery. Front. Mol. Biosci. 2023, 10, 1198044. [Google Scholar] [CrossRef] [PubMed]
  192. Morton, L.A.; Yang, H.; Saludes, J.P.; Fiorini, Z.; Beninson, L.; Chapman, E.R.; Fleshner, M.; Xue, D.; Yin, H. MARCKS-ED peptide as a curvature and lipid sensor. ACS Chem. Biol. 2013, 8, 218–225. [Google Scholar] [CrossRef] [PubMed]
  193. Unterreitmeier, S.; Fuchs, A.; Schäffler, T.; Heym, R.G.; Frishman, D.; Langosch, D. Phenylalanine promotes interaction of transmembrane domains via GxxxG motifs. J. Mol. Biol. 2007, 374, 705–718. [Google Scholar] [CrossRef]
  194. Yan, L.; de Jesus, A.J.; Tamura, R.; Li, V.; Cheng, K.; Yin, H. Curvature sensing MARCKS-ED peptides bind to membranes in a stereo-independent manner. J. Pept. Sci. 2015, 21, 577–585. [Google Scholar] [CrossRef] [PubMed]
  195. Flynn, A.D.; Yin, H. Lipid-Targeting Peptide Probes for Extracellular Vesicles. J. Cell Physiol. 2016, 231, 2327–2332. [Google Scholar] [CrossRef] [PubMed]
  196. Babatunde, K.A.; Yesodha Subramanian, B.; Ahouidi, A.D.; Martinez Murillo, P.; Walch, M.; Mantel, P.Y. Role of Extracellular Vesicles in Cellular Cross Talk in Malaria. Front. Immunol. 2020, 11, 22. [Google Scholar] [CrossRef] [PubMed]
  197. Regev-Rudzki, N.; Wilson, D.W.; Carvalho, T.G.; Sisquella, X.; Coleman, B.M.; Rug, M.; Bursac, D.; Angrisano, F.; Gee, M.; Hill, A.F.; et al. Cell-cell communication between malaria-infected red blood cells via exosome-like vesicles. Cell 2013, 153, 1120–1133. [Google Scholar] [CrossRef] [PubMed]
  198. Ngambenjawong, C.; Pineda, J.M.; Pun, S.H. Engineering an Affinity-Enhanced Peptide through Optimization of Cyclization Chemistry. Bioconjug. Chem. 2016, 27, 2854–2862. [Google Scholar] [CrossRef] [PubMed]
  199. Xu, K.; Jin, Y.; Li, Y.; Huang, Y.; Zhao, R. Recent Progress of Exosome Isolation and Peptide Recognition-Guided Strategies for Exosome Research. Front. Chem. 2022, 10, 844124. [Google Scholar] [CrossRef] [PubMed]
  200. Morton, L.A.; Tamura, R.; de Jesus, A.J.; Espinoza, A.; Yin, H. Biophysical investigations with MARCKS-ED: Dissecting the molecular mechanism of its curvature sensing behaviors. Biochim. Biophys. Acta 2014, 1838, 3137–3144. [Google Scholar] [CrossRef] [PubMed]
  201. Quadri, Z.; Elsherbini, A.; Bieberich, E. Extracellular vesicles in pharmacology: Novel approaches in diagnostics and therapy. Pharmacol. Res. 2022, 175, 105980. [Google Scholar] [CrossRef]
  202. 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, 2265–2269. [Google Scholar] [CrossRef]
  203. Zhang, Y.; Liu, Q.; Zhang, X.; Huang, H.; Tang, S.; Chai, Y.; Xu, Z.; Li, M.; Chen, X.; Liu, J.; et al. Recent advances in exosome-mediated nucleic acid delivery for cancer therapy. J. Nanobiotechnol. 2022, 20, 279. [Google Scholar] [CrossRef]
  204. Bao, C.; Xiang, H.; Chen, Q.; Zhao, Y.; Gao, Q.; Huang, F.; Mao, L. A Review of Labeling Approaches Used in Small Extracellular Vesicles Tracing and Imaging. Int. J. Nanomed. 2023, 18, 4567–4588. [Google Scholar] [CrossRef] [PubMed]
  205. Choi, H.; Choi, Y.; Yim, H.Y.; Mirzaaghasi, A.; Yoo, J.K.; Choi, C. Biodistribution of Exosomes and Engineering Strategies for Targeted Delivery of Therapeutic Exosomes. Tissue Eng. Regen. Med. 2021, 18, 499–511. [Google Scholar] [CrossRef]
  206. Bruno, B.J.; Miller, G.D.; Lim, C.S. Basics and recent advances in peptide and protein drug delivery. Ther. Deliv. 2013, 4, 1443–1467. [Google Scholar] [CrossRef]
  207. 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] [PubMed]
  208. Susa, F.; Limongi, T.; Dumontel, B.; Vighetto, V.; Cauda, V. Engineered Extracellular Vesicles as a Reliable Tool in Cancer Nanomedicine. Cancers 2019, 11, 1979. [Google Scholar] [CrossRef]
  209. Kooijmans, S.A.A.; de Jong, O.G.; Schiffelers, R.M. Exploring interactions between extracellular vesicles and cells for innovative drug delivery system design. Adv. Drug Deliv. Rev. 2021, 173, 252–278. [Google Scholar] [CrossRef] [PubMed]
  210. Santelices, J.; Ou, M.; Hui, W.W.; Maegawa, G.H.B.; Edelmann, M.J. Fluorescent Labeling of Small Extracellular Vesicles (EVs) Isolated from Conditioned Media. Bio-protocol 2022, 12, e4447. [Google Scholar] [CrossRef] [PubMed]
  211. N’Diaye, E.R.; Orefice, N.S.; Ghezzi, C.; Boumendjel, A. Chemically Modified Extracellular Vesicles and Applications in Radiolabeling and Drug Delivery. Pharmaceutics 2022, 14, 653. [Google Scholar] [CrossRef]
  212. Szymanowski, W.; Szymanowska, A.; Bielawska, A.; Lopez-Berestein, G.; Rodriguez-Aguayo, C.; Amero, P. Aptamers as Potential Therapeutic Tools for Ovarian Cancer: Advancements and Challenges. Cancers 2023, 15, 5300. [Google Scholar] [CrossRef]
  213. Chen, Z.; Xiong, M.; Tian, J.; Song, D.; Duan, S.; Zhang, L. Encapsulation and assessment of therapeutic cargo in engineered exosomes: A systematic review. J. Nanobiotechnol. 2024, 22, 18. [Google Scholar] [CrossRef]
  214. Zhang, Z.; Jia, J.; Lai, Y.; Ma, Y.; Weng, J.; Sun, L. Conjugating folic acid to gold nanoparticles through glutathione for targeting and detecting cancer cells. Bioorganic Med. Chem. 2010, 18, 5528–5534. [Google Scholar] [CrossRef] [PubMed]
  215. Xu, Z.; Wang, X.; Liu, X.; Cui, Z.; Yang, X.; Yeung, K.W.K.; Chung, J.C.; Chu, P.K.; Wu, S. Tannic Acid/Fe3+/Ag Nanofilm Exhibiting Superior Photodynamic and Physical Antibacterial Activity. ACS Appl. Mater. Interfaces 2017, 9, 39657–39671. [Google Scholar] [CrossRef] [PubMed]
  216. 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. [Google Scholar] [CrossRef]
  217. Muthu, S.; Bapat, A.; Jain, R.; Jeyaraman, N.; Jeyaraman, M. Exosomal therapy-a new frontier in regenerative medicine. Stem Cell Investig. 2021, 8, 7. [Google Scholar] [CrossRef] [PubMed]
  218. Huda, M.N.; Nafiujjaman, M.; Deaguero, I.G.; Okonkwo, J.; Hill, M.L.; Kim, T.; Nurunnabi, M. Potential Use of Exosomes as Diagnostic Biomarkers and in Targeted Drug Delivery: Progress in Clinical and Preclinical Applications. ACS Biomater. Sci. Eng. 2021, 7, 2106–2149. [Google Scholar] [CrossRef]
  219. Beetler, D.J.; Di Florio, D.N.; Bruno, K.A.; Ikezu, T.; March, K.L.; Cooper, L.T.; Jr Wolfram, J.; Fairweather, D. Extracellular vesicles as personalized medicine. Mol. Aspects Med. 2023, 91, 101155. [Google Scholar] [CrossRef] [PubMed]
  220. Mosquera-Heredia, M.I.; Morales, L.C.; Vidal, O.M.; Barceló, E.; Silvera-Redondo, C.; Vélez, J.I.; Garavito-Galofre, P. Exosomes: Potential Disease Biomarkers and New Therapeutic Targets. Biomedicines 2021, 9, 1061. [Google Scholar] [CrossRef]
  221. Del Bene, M.; Osti, D.; Faletti, S.; Beznoussenko, G.V.; DiMeco, F.; Pelicci, G. Extracellular vesicles: The key for precision medicine in glioblastoma. Neuro-Oncology 2021, 24, 184–196. [Google Scholar] [CrossRef]
  222. Shang, M.; Ji, J.S.; Song, C.; Gao, B.J.; Jin, J.G.; Kuo, W.P.; Kang, H. Extracellular Vesicles: A Brief Overview and Its Role in Precision Medicine, in Extracellular Vesicles: Methods and Protocols; Kuo, W.P., Jia, S., Eds.; Springer: New York, NY, USA, 2017; pp. 1–14. [Google Scholar]
  223. Lone, S.N.; Nisar, S.; Masoodi, T.; Singh, M.; Rizwan, A.; Hashem, S.; El-Rifai, W.; Bedognetti, D.; Batra, S.K.; Haris, M.; et al. Liquid biopsy: A step closer to transform diagnosis, prognosis and future of cancer treatments. Mol. Cancer 2022, 21, 79. [Google Scholar] [CrossRef]
  224. Areny-Balagueró, A.; Solé-Porta, A.; Camprubí-Rimblas, M.; Campaña-Duel, E.; Ceccato, A.; Roig, A.; Closa, D.; Artigas, A. Bioengineered extracellular vesicles: Future of precision medicine for sepsis. Intensive Care Med. Exp. 2023, 11, 11. [Google Scholar] [CrossRef] [PubMed]
  225. Mora, E.M.; Álvarez-Cubela, S.; Oltra, E. Biobanking of Exosomes in the Era of Precision Medicine: Are We There Yet? Int. J. Mol. Sci. 2015, 17, 13. [Google Scholar] [CrossRef] [PubMed]
  226. Yang, J.; Shin, T.-S.; Kim, J.S.; Jee, Y.-K.; Kim, Y.-K. A new horizon of precision medicine: Combination of the microbiome and extracellular vesicles. Exp. Mol. Med. 2022, 54, 466–482. [Google Scholar] [CrossRef] [PubMed]
  227. Desdín-Micó, G.; Mittelbrunn, M. Role of exosomes in the protection of cellular homeostasis. Cell Adhes. Migr. 2017, 11, 127–134. [Google Scholar] [CrossRef] [PubMed]
  228. Chutipongtanate, S.; Pattanapanyasat, K.; Meller, J.; Serrano López, J. Editorial: Driving extracellular vesicles toward applications in precision medicine. Front. Med. 2022, 9, 1049697. [Google Scholar] [CrossRef]
  229. Arraud, N.; Linares, R.; Tan, S.; Gounou, C.; Pasquet, J.M.; Mornet, S.; Brisson, A.R. Extracellular vesicles from blood plasma: Determination of their morphology, size, phenotype and concentration. J. Thromb. Haemost. 2014, 12, 614–627. [Google Scholar] [CrossRef] [PubMed]
  230. Yang, G.; Waheed, S.; Wang, C.; Shekh, M.; Li, Z.; Wu, J. Exosomes and Their Bioengineering Strategies in the Cutaneous Wound Healing and Related Complications: Current Knowledge and Future Perspectives. Int. J. Biol. Sci. 2023, 19, 1430–1454. [Google Scholar] [CrossRef] [PubMed]
  231. Cao, J.; Lv, G.; Wei, F. Engineering exosomes to reshape the immune microenvironment in breast cancer: Molecular insights and therapeutic opportunities. Clin. Transl. Med. 2024, 14, e1645. [Google Scholar] [CrossRef] [PubMed]
  232. FDA. Public Safety Alert Due to Marketing of Unapproved Stem Cell and Exosome Products. 2019. Available online: https://www.fda.gov/safety/medical-product-safety-information/public-safety-alert-due-marketing-unapproved-stem-cell-and-exosome-products (accessed on 20 April 2024).
  233. Cheng, K.; Kalluri, R. Guidelines for clinical translation and commercialization of extracellular vesicles and exosomes based therapeutics. Extracell. Vesicle 2023, 2, 100029. [Google Scholar] [CrossRef]
  234. Tzng, E.; Bayardo, N.; Yang, P.C. Current challenges surrounding exosome treatments. Extracell. Vesicle 2023, 2, 100023. [Google Scholar] [CrossRef]
Figure 1. Exosome-based therapy in clinical trials (as listed on clinicaltrials.gov as accessed on 20 April 2024).
Figure 1. Exosome-based therapy in clinical trials (as listed on clinicaltrials.gov as accessed on 20 April 2024).
Pharmaceutics 16 00709 g001
Figure 2. Illustration of the protein corona formation around exosomes.
Figure 2. Illustration of the protein corona formation around exosomes.
Pharmaceutics 16 00709 g002
Figure 3. Timeline outlining the advancement in exosome-based therapy.
Figure 3. Timeline outlining the advancement in exosome-based therapy.
Pharmaceutics 16 00709 g003
Figure 4. A schematic representation of the different strategies used for modifying the exosomal surface for targeted therapy and imaging applications. These strategies include labelling with antibodies, fluorescent dyes, radiolabeling, and other engineering techniques to enhance delivery specificity.
Figure 4. A schematic representation of the different strategies used for modifying the exosomal surface for targeted therapy and imaging applications. These strategies include labelling with antibodies, fluorescent dyes, radiolabeling, and other engineering techniques to enhance delivery specificity.
Pharmaceutics 16 00709 g004
Table 1. Advantages and disadvantages of exosome, microvesicle, and liposome-based therapies.
Table 1. Advantages and disadvantages of exosome, microvesicle, and liposome-based therapies.
Vesicle TypeAdvantagesDisadvantages
Exosomes
-
Small and stable structures; have a high capacity for carrying and protecting therapeutic cargoes such as miRNAs, mRNAs, proteins, and drugs
-
Ability to cross biological barriers, including the blood–brain barrier
-
Ability to deliver therapeutics to difficult-to-reach tissues due to small size
-
Active packaging of cargo
-
Fast uptake
-
More targeting capacity
-
Lower toxicity
-
Limited clinical trials
-
Scalability for clinical use
-
Need to standardize the isolation techniques
-
Difficult to perform modifications because of complex structure
Microvesicles
-
Nonimmunogenic
-
Lower toxicity
-
Easily targetable
-
Can carry a more diverse array of cargo, including larger molecules and organelles
-
Passive packaging of cargo
-
Need to standardize the isolation techniques
-
Potential safety concerns related to the heterogeneous nature of microvesicles
-
Limited clinical trials
-
Scalability for clinical use
Liposomes
-
Diversity of lipid compositions
-
Scalable synthesis
-
Ability to load with desired cargo
-
Surface and cargo can be easily modified
-
Active clinical research
-
Not easily targetable
-
Low cellular uptake and potential complement activation
-
Premature drug release
-
Higher toxicity
Table 2. List of exosome-based clinical trials (as of 10 May 2024, listed on clinicaltrials.gov).
Table 2. List of exosome-based clinical trials (as of 10 May 2024, listed on clinicaltrials.gov).
NCT NumberStudy TitleConditionsPhases
NCT05218759Exosomes Detection for the Prediction of the Efficacy and Adverse Reactions of Anlotinib in Patients with Advanced NSCLCNon-Small-Cell Lung CancerNA
NCT02822131Phosphate in Blood Pressure RegulationHypertensionNA
NCT02579460Reflux-Induced Oxidative Stress in Barrett’s Esophagus: Response, Repair, and Epithelial-Mesenchymal-TransitionBarrett’s Esophagus|Gastroesophageal Reflux DiseaseNA
NCT06072794A Proof-of-Concept Study to Evaluate Exosomes from Human Mesenchymal Stem Cells in Women with Premature Ovarian Insufficiency (POI)Premature Ovarian Insufficiency|Diminished Ovarian ReservePhase 1
NCT05658094Exosome Effect on Prevention of Hair lossHair Loss|AlopeciaNA
NCT02649465SGLT2 Inhibitor Versus Sulfonylurea on Type 2 Diabetes With NAFLDNon-alcoholic Fatty Liver DiseasePhase 4
NCT03109873Metformin Hydrochloride in Affecting Cytokines and Exosomes in Patients with Head and Neck CancerLarynx|Lip|Oral Cavity|PharynxEarly Phase 1
NCT04184076Randomized Controlled Trial of Time-Restricted Feeding (TRF) in Acute Ischemic Stroke PatientsFastingNA
NCT05490173The Pilot Experimental Study of the Neuroprotective Effects of Exosomes in Extremely Low Birth Weight InfantsPremature Birth|Extreme Prematurity|Preterm Intraventricular Hemorrhage|Hypoxia-Ischemia, Cerebral|Neurodevelopmental DisordersNA
NCT02138331Effect of Microvesicles and Exosomes Therapy on β-cell Mass in Type I Diabetes Mellitus (T1DM)Diabetes Mellitus Type 1Phase 2|Phase 3
NCT04459182Circulating Exosomes and Endothelial Dysfunction in Patients with Obstructive Sleep Apneas Hypopneas SyndromeEndothelial DysfunctionNA
NCT03608631iExosomes in Treating Participants with Metastatic Pancreas Cancer with KrasG12D MutationKRAS NP_004976.2: p. G12D|Metastatic Pancreatic Adenocarcinoma Pancreatic Ductal Adenocarcinoma Stage IV Pancreatic Cancer AJCC v8Phase 1
NCT04388982the Safety and the Efficacy Evaluation of Allogenic Adipose MSC-Exos in Patients with Alzheimer’s DiseaseAlzheimer’s DiseasePhase 1|Phase 2
NCT01104220Role of Immune System in Obesity-related Inflammation and Cardiometabolic RiskNon-alcoholic Fatty Liver Disease|Metabolic Syndrome|Metabolically Abnormal Obesity|Metabolically Normal Obesity|ObesityNA
NCT05073627The Effect of Dicloxacillin on Oral Absorption of DrugsHealthy Volunteers|Drug–Drug InteractionPhase 1
NCT04153539Acute Health Effects of Traffic-Related Air Pollution ExposureCardiovascular System|Respiratory SystemNA
NCT05871463Effect of Mesenchymal Stem Cells-derived Exosomes in Decompensated Liver CirrhosisDecompensated Liver CirrhosisPhase 2
NCT05692635Reducing the Incidence of Symptomatic Brain Metastases with MRI SurveillanceBrain Metastases|Non-Small-Cell Lung Cancer Stage IIIPhase 2
NCT05163626Combined Aerobic Exercise and Cognitive Training in Seniors at Increased Risk for Alzheimer’s DiseaseAlzheimer’s DiseaseNA
NCT04131166Precision Nutrition and Metabolic FunctionObesity|Insulin ResistanceNA
NCT03217266Navtemadlin and Radiation Therapy in Treating Patients with Soft Tissue SarcomaResectable Soft Tissue Sarcoma|Soft Tissue SarcomaPhase 1
NCT03027726Prevention of Diabetes in Overweight/Obese Preadolescent ChildrenOverweight Children with Type 2 Diabetes RiskNA
NCT06259435Modulating Energy Density in Time-Restricted EatingInsulin Resistance|Body WeightNA
NCT04134676Therapeutic Potential of Stem Cell Conditioned Medium on Chronic Ulcer WoundsChronic UlcerPhase 1
NCT05777876Early Identification of TRD and Construction and Clinical Validation of NTBS Precision TechnologyTreatment-Resistant DepressionNA
NCT05563766A Phase II Trial to Evaluate the Effect of Itraconazole on Pathologic Complete Response Rates in Resectable Esophageal CancerEsophageal Adenocarcinoma|Esophageal Squamous Cell Carcinoma|Gastroesophageal Junction CarcinomaPhase 2
NCT04460963Role of Adrenomedullin in Leukemic Endosteal/Vascular NichesAcute Myeloid LeukemiaNA
NCT04258735Genetic Characteristics of Metastatic Breast Cancer PatientsMetastatic Breast CancerNA
NCT03410030Trial of Ascorbic Acid (AA) + Nanoparticle Paclitaxel Protein Bound + Cisplatin + Gemcitabine (AA NABPLAGEM)Pancreatic Cancer|Pancreas Cancer|Pancreatic Adenocarcinoma Resectable|Pancreatic Ductal Adenocarcinoma|Pancreas MetastasesPhase 1|Phase 2
NCT04747574Evaluation of the Safety of CD24-Exosomes in Patients With COVID-19 InfectionSARS-CoV-2Phase 1
NCT05969717Induced Pluripotent Stem Cell Derived Exosomes for the Treatment of Atopic DermatitisAtopic DermatitisEarly Phase 1
NCT02653339Effectiveness of Qufeng Shengshi Fang on Treatment of Allergic Rhinitis.Rhinitis, Allergic, PerennialNA
NCT04266639Rheo-Erythrocrine Dysfunction as a Biomarker for RIC Treatment in Acute Ischemic StrokeStroke, Acute|Ischemic Stroke|Cerebrovascular Disorders|Central Nervous System DiseasesNA
NCT05354141Extracellular Vesicle Treatment for Acute Respiratory Distress Syndrome (ARDS) (EXTINGUISH ARDS)Acute Respiratory Distress Syndrome|ARDSPhase 3
NCT05616234Exercise-induced Changes in ExosomesExosomes|Connective Tissue|ExerciseNA
NCT06279039Clinical Observation of Exosomes in Patients After Q-switched Laser SurgeryExosome|Skin Regeneration|LaserNA
NCT03230019Two-Lumen Catheterization for Lung Wedge ResectionThoracic Surgery, Video-AssistedNA
NCT03392441Insulin Deprivation on Brain Structure and Function in Humans with Type 1 DiabetesDiabetes Mellitus, Type 1|Diabetes|Diabetes ComplicationsNA
NCT04483219Tyrosine Kinase Inhibitor (TKI) + Anti-PD-1 Antibody in TKI-responded Microsatellite Stability/Proficient Mismatch Repair (MSS/pMMR) Metastatic Colorectal Adenocarcinoma.MSS|pMMR|Metastatic Colorectal AdenocarcinomaPhase 2
NCT05475418Pilot Study of Human Adipose Tissue Derived Exosomes Promoting Wound HealingWounds and InjuriesNA
NCT06110130Effect of Empagliflozin on Podocyte Specific Proteins in African American Veterans With NDKDChronic Kidney DiseasesPhase 4
NCT05413148The Effect of Stem Cells and Stem Cell Exosomes on Visual Functions in Patients with Retinitis PigmentosaRetinitis PigmentosaPhase 2|Phase 3
NCT04213248Effect of UMSCs Derived Exosomes on Dry Eye in Patients With cGVHDDry Eye DiseasePhase 1|Phase 2
NCT05402748Safety and Efficacy of Injection of Human Placenta Mesenchymal Stem Cells Derived Exosomes for Treatment of Complex Anal FistulaFistula PerianalPhase 1|Phase 2
NCT03985696Exosomes and Immunotherapy in Non-Hodgkin B-cell LymphomasLymphoma, B-cell, Aggressive Non-Hodgkin (B-NHL)NA
NCT05166616Minnelide and Osimertinib for the Treatment of Advanced EGFR Mutated Non-Small Cell Lung CancerAdvanced Lung Non-Small-Cell Carcinoma|Locally Advanced Lung Non-Small-Cell Carcinoma|Stage III Lung Cancer AJCC v8|Stage IIIA Lung Cancer AJCC v8|Stage IIIB Lung Cancer AJCC v8|Stage IIIC Lung Cancer AJCC v8|Stage IV Lung Cancer AJCC v8|Stage IVA Lung Cancer AJCC v8|Stage IVB Lung Cancer AJCC v8|Unresectable Lung Non-Small-Cell CarcinomaPhase 1
NCT04556916Early Detection of Prostate CancerProstate CancerNA
NCT05815524Physical Activity in Patients with Parkinson’s Disease: A “Disease Modifying” Intervention?Parkinson’s DiseaseNA
NCT04998058Autogenous Mesenchymal Stem Cell Culture-Derived Signaling Molecules as Enhancers of Bone Formation in Bone GraftingBone Loss, Osteoclastic|Bone Loss, Alveolar|Alveolar Bone Loss|Alveolar Bone Atrophy|Grafting BonePhase 1|Phase 2
NCT02892734Ipilimumab and Nivolumab in Treating Patients with Recurrent Stage IV HER2 Negative Inflammatory Breast CancerHER2/Neu Negative|Recurrent Inflammatory Breast Carcinoma|Stage IV Breast Cancer|Stage IV Inflammatory Breast CarcinomaPhase 2
NCT04493242Extracellular Vesicle Infusion Treatment for COVID-19 Associated ARDSCOVID-19|ARDSPhase 2
NCT05698524A Study of Temodar with Abexinostat (PCI-24781) for Patients with Recurrent GliomaRecurrent High-Grade Glioma|Anaplastic Astrocytoma|Anaplastic Oligodendroglioma|Glioblastoma|GliosarcomaPhase 1
NCT04798716The Use of Exosomes for the Treatment of Acute Respiratory Distress Syndrome or Novel Coronavirus Pneumonia Caused by COVID-19COVID-19|Novel Coronavirus Pneumonia|Acute Respiratory Distress SyndromePhase 1|Phase 2
NCT04939324Molecular Profiling of Exosomes in Tumor-draining Vein of Early-staged Lung CancerLung Cancer|Exosomes|Non-Small-Cell Lung CancerNA
NCT05326724The Role of Acupuncture-induced Exosome in Treating Post-stroke DementiaExosome|Post-stroke Dementia|AcupunctureNA
NCT05621109PRE-Pregnancy Weight Loss and the Reducing Effect on Childhood Overweight—CopenhagenOverweight and Obesity|Weight Loss|Pregnancy RelatedNA
NCT04641585Brugada Syndrome and Artificial Intelligence Applications to DiagnosisBrugada Syndrome 1NA
NCT05610332Clinical Efficacy in Neoadjuvant Treatment of Locally Advanced Gastric Cancer with Different ImmunotypesLocally Advanced Gastric CancerPhase 3
NCT02869685Consistency Analysis of PD-L1s in Advanced NSCLC Tissues and in Plasma Exosomes Before and After RadiotherapyNSCLCNA
NCT06361485Safety and Feasibility of Umbilical Cord Wharton’s Jelly Allograft Injections for Lumbar PainLow Back PainPhase 1
NCT02928432SWITCH: Study of the Prednisone to Dexamethasone Change in mCRPC Patients Treated with AbirateroneProstate CancerPhase 2
NCT04765137Evaluate the Effect of Atorvastatin on Cerebrovascular Reactivity in MCIMild Cognitive ImpairmentPhase 2
NCT03854032Nivolumab and BMS986205 in Treating Patients with Stage II-IV Squamous Cell Cancer of the Head and NeckLip|Oral Cavity Squamous Cell Carcinoma|Pharynx|Larynx|Squamous Cell CarcinomaPhase 2
NCT04585932Androgen Deprivation Therapy and Apalutamide With or Without Radiation Therapy for the Treatment of Biochemically Recurrent Prostate Cancer, RESTART StudyBiochemically Recurrent Prostate Carcinoma|Metastatic Prostate Carcinoma|Oligometastatic Prostate Carcinoma|Stage IV Prostate Cancer AJCC v8|Stage IVA Prostate Cancer AJCC v8|Stage IVB Prostate Cancer AJCC v8Phase 2
NCT05669144Co-transplantation of Mesenchymal Stem Cell Derived Exosomes and Autologous Mitochondria for Patients Candidate for CABG SurgeryMyocardial Infarction|Myocardial Ischemia|Myocardial StunningPhase 1 |Phase 2
NCT04029740Exosomal microRNAs as a Biomarker in Panic Disorder and in Response to CBTPanic DisorderNA
NCT06339840The Impact of Lifestyle Intervention on Weight and Fertility in Obese MalesObesity|Weight Loss|Male Fertility|Artificial Insemination|IVF-ETNA
NCT05192694Evaluation of Fapi-pet in Prostate Cancer.Prostate CancerNA
NCT04781062Development of a Horizontal Data Integration Classifier for Noninvasive Early Diagnosis of Breast CancerBreast CancerNA
NCT03762629Exercise and Diet Restriction on Cardiovascular Function in Obese Children and AdolescentsChildhood Obesity|Adolescent ObesityNA
NCT04849429Intra-discal Injection of Platelet-rich Plasma (PRP) Enriched with Exosomes in Chronic Low Back PainChronic Low Back Pain|Degenerative Disc DiseasePhase 1
NCT05927129Research on the Biological Mechanism of the Efficacy of Psychotherapy for Depression Based on the fNIRSDepressive DisorderNA
NCT05738629Safety and Efficacy of Pluripotent Stem Cell-derived Mesenchymal Stem Cell Exosome (PSC-MSC-Exo) Eye Drops Treatment for Dry Eye Diseases Post Refractive Surgery and Associated with BlepharospasmDry Eye DiseasePhase 1|Phase 2
NCT05798494PRE-Pregnancy Weight Loss and the Reducing Effect on Childhood Overweight - AarhusOverweight and Obesity|Weight Loss|Pregnancy Related|Child Nutrition Sciences|Body CompositionNA
NCT05261360Clinical Efficacy of Exosome in Degenerative Meniscal InjuryKnee; Injury, Meniscus (Lateral) (Medial)|Meniscus Tear|Meniscus Lesion|Meniscus; Degeneration|Meniscus; Laceration|Meniscus Injury, Tibial|Knee Injuries|Knee Pain Swelling|ArthralgiaPhase 2
NCT03406780A Study of CAP-1002 in Ambulatory and Non-Ambulatory Patients with Duchenne Muscular DystrophyMuscular Dystrophies|Muscular Dystrophy, Duchenne|Muscular Disorders, Atrophic|Muscular Diseases|Neuromuscular diseases|Nervous System Diseases|Genetic Diseases, X-Linked|Genetic Diseases, InbornPhase 2
NCT04334603Exercise Intervention in Heart FailureHeart FailureNA
NCT04595903Treatment of SARS-CoV-2 Virus Disease (COVID-19) in Humans with Hemopurifier® DeviceCOVID-19NA
NCT03711890Ultra-High Resolution Optical Coherence Tomography in Detecting Micrometer Sized Early-Stage Pancreatic Cancer in Participants With Pancreatic CancerPancreatic Carcinoma|Pancreatic Intraductal Papillary Mucinous Neoplasm, Pancreatobiliary-TypeNA
NCT05955521Exosome as the Prognostic and Predictive Biomarker in EBC PatientsTriple Negative Breast Cancer|HER2-positive Breast CancerNA
NCT03493984Plant Exosomes and Patients Diagnosed with Polycystic Ovary Syndrome (PCOS) 17Polycystic Ovary SyndromeNA
NCT03260179Study to Evaluate the Safety, Preliminary Efficacy and Pharmacokinetics of 3810Advanced Solid Tumor|Advanced/Metastatic Colorectal CancerPhase 1
NCT05286684Feasibility of Exosome Analysis in Cerebrospinal Fluid During the Diagnostic Workup of Metastatic Meningitis (Exo-LCR)Breast CancerNA
NCT04173650MSC EVs in Dystrophic Epidermolysis BullosaDystrophic Epidermolysis BullosaPhase 1|Phase 2
NCT05243368Evaluation of Personalized Nutritional Intervention on Wound Healing of Cutaneous Ulcers in DiabeticsFoot, DiabeticsNA
NCT04421872The Disorder of Circadian Clock Gene and Early Cognitive Dysfunction After General AnesthesiaPostoperative Delirium|General Anesthesia|Exosomes|Clock Gene Circadian Rhythm DisordersNA
NCT04969172A Phase II Randomized, Double-blind, Placebo-controlled Study to Evaluate the Safety and Efficacy of Exosomes Overexpressing CD24 to Prevent Clinical Deterioration in Patients with Moderate or Severe COVID-19 InfectionCOVID-19 DiseasePhase 2
NCT01294072Study Investigating the Ability of Plant Exosomes to Deliver Curcumin to Normal and Colon Cancer TissueColon CancerNA
NCT04879810Plant Exosomes +/− Curcumin to Abrogate Symptoms of Inflammatory Bowel DiseaseIrritable Bowel DiseaseNA
NCT01550523Pilot Immunotherapy Trial for Recurrent Malignant GliomasMalignant Glioma of BrainPhase 1
NCT02439008Early Biomarkers of Tumor Response in High Dose Hypo Fractionated Radiotherapy Word Package 3: Immune ResponseCarcinoma, Hepatocellular|Colorectal Neoplasms|Melanoma|Kidney NeoplasmsNA
NCT03974204Analyses of Exosomes in the Cerebrospinal Fluid for Breast Cancer Patients with Suspicion of Leptomeningeal Metastasis.Breast Cancer|Leptomeningeal MetastasisNA
NCT05913960Accelerated Intermittent Theta-Burst Stimulation Ameliorate Major Depressive Disorder by Regulating CAMKII PathwayMajor Depressive Disorder|Major Depressive Disorder, RecurrentNA
NCT05375604A Study of exoASO-STAT6 (CDK-004) in Patients with Advanced Hepatocellular Carcinoma (HCC) and Patients with Liver Metastases from Either Primary Gastric Cancer or Colorectal Cancer (CRC)Advanced Hepatocellular Carcinoma (HCC)|Gastric Cancer Metastatic to Liver|Colorectal Cancer Metastatic to LiverPhase 1
NCT02706262Complex Effects of Dietary Manipulation on Metabolic Function, Inflammation and HealthObesity|Insulin ResistanceNA
NCT05216562Efficacy and Safety of EXOSOME-MSC Therapy to Reduce Hyper-inflammation In Moderate COVID-19 PatientsSARS-CoV2 InfectionPhase 2|Phase 3
NCT05775146SBRT of Metastases Following Neo-Adjuvant Treatment for Colorectal Cancer with Synchronous Liver MetastasesColorectal Cancer|Liver Metastasis Colon CancerPhase 2
NCT02737267Development of a Nutrigenetic Test for Personalized Prescription of Body Weight Loss Diets (Obekit)Body Weight ChangesNA
NCT05524974Clinical Study of Camrelizumab, Apatinib Mesylate and Nab-paclitaxel Combined with Oxplatin and S-1 in the Neoadjuvant Treatment of Locally Advanced Gastric Cancer With Different GenotypesLocally Advanced Gastric CancerPhase 2
NCT06116903Clinical Relevance of Detecting Molecular Abnormalities in Glial Tumor ExosomesGliomaNA
NCT04664738PEP on a Skin Graft Donor Site WoundSkin GraftPhase 1
NCT04530890Interest of Circulating Tumor DNA in Digestive and Gynecologic/Breast CancerBreast Cancer|Digestive Cancer|Gynecologic Cancer|Circulating Tumor DNA|ExosomesNA
NCT02565264Effect of Plasma Derived Exosomes on Cutaneous Wound HealingUlcerEarly Phase 1
NCT04542902Non-coding RNAs Analysis of Eosinophil Subtypes in AsthmaAllergic Asthma|Severe Eosinophilic AsthmaNA
NCT02662621Pilot Study with the Aim to Quantify a Stress Protein in the Blood and in the Urine for the Monitoring and Early Diagnosis of Malignant Solid TumorsCancerNA
NCT05813379Mesenchymal Stem Cells Derived Exosomes in Skin RejuvenationAnti-AgingPhase 1|Phase 2
NCT03255408Cerebral Blood Flow and Ventilatory Responses During Sleep in Normoxia and Intermittent HypoxiaObstructive Sleep Apnea of Adult|Hypoxia, Brain|Sleep Apnea|Sleep Disorder|Stroke|Blood Pressure|Endothelial Dysfunction|Oxidative StressPhase 1|Phase 2
NCT05043181Exosome-based Nanoplatform for Ldlr mRNA Delivery in FHFamilial HypercholesterolemiaPhase 1
NCT05110781Atezolizumab Before Surgery for the Treatment of Regionally Metastatic Head and Neck Squamous Cell Cancer with an Unknown or Historic Primary SiteLocally Advanced Cutaneous Squamous Cell Carcinoma of the Head and Neck|Resectable Cutaneous Squamous-Cell Carcinoma of the Head and Neck|Stage III Cutaneous Squamous Cell Carcinoma of the Head and Neck AJCC v8Phase 2
NCT05243381Inflammation, NK Cells, Antisense Protein and Exosomes, and Correlation with Immune Response During HIV InfectionHIV InfectionsNA
NCT05373381The KetoGlioma (Ketogenic Glioma) StudyGliomaNA
NCT03202212Effect of Mixed On-line Hemodiafiltration on Circulating Markers of Inflammation and Vascular DysfunctionChronic Kidney Failure|Dialysis-Related ComplicationPhase 1|Phase 2
NCT01811381Curcumin and Yoga Therapy for Those at Risk for Alzheimer’s DiseaseMild Cognitive ImpairmentPhase 2
NCT03459703Effect of Time-Restricted Feeding on Fat Loss and Cardiometabolic Risk Factors in Overweight AdultsObesityNA
NCT05624203Clinical Efficacy of Extracorporeal Cardiac Shock Wave Therapy in Patients with Ischemia-reperfusion InjuryMyocardial Reperfusion Injury|Treatment Outcome|Prognosis|ST Elevation Myocardial InfarctionNA
NCT04235023PTP1B Implication in the Vascular Dysfunction Associated with Obstructive Sleep ApneaSleep Apnea|Inflammation|AtherosclerosisNA
NCT05480150Chinese Longitudinal and Systematic Study of Bioplar DisorderMajor Depressive Disorder|Bipolar Disorder, Mixed|Affective; Disorder, OrganicNA
NCT05832255An Investigation of Psilocybin on Behavioural and Cognitive Symptoms of Adults with Fragile X SyndromeFragile X Syndrome|Behavior|Cognitive DysfunctionPhase 2
NCT06245746UCMSC-Exo for Chemotherapy-induced Myelosuppression in Acute Myeloid LeukemiaAcute Myeloid Leukemia|Neutropenia|Anemia|Thrombocytopenia|Infections|BleedingPhase 1
NCT04453046Hemopurifier Plus Pembrolizumab in Head and Neck CancerSquamous Cell Carcinoma of the Head and NeckNA
NCT04142138When the Kidney Reacts to Nutritional ChangesPrehypertensionNA
NCT05833568Five-day 20-min 10-Hz tACS in Patients with a Disorder of ConsciousnessBrain Injury Traumatic Severe|Disorder of Consciousness|Brain InjuriesNA
NCT02977468Effects of MK-3475 (Pembrolizumab) on the Breast Tumor Microenvironment in Triple Negative Breast CancerTriple Negative Breast CancerPhase 1
NCT06236568Impact of Graft Reconditioning with Hypothermic Machine Perfusion on HCC Recurrence After Liver TransplantationHepatocellular Carcinoma|Liver Transplant; ComplicationsNA
NCT02748369In vivo Assessment of Cellular Metabolism in HumansNormal Cellular MetabolismPhase 1
NCT04500769Training Induced Muscle Exosome ReleaseMetabolismNA
NCT02051101Pathogenic Mechanisms of Port Wine Stain and Repository of Port Wine Stain Biopsy SamplesPort-Wine StainNA
NCT04602104A Clinical Study of Mesenchymal Stem Cell Exosomes Nebulizer for the Treatment of ARDSAcute Respiratory Distress SyndromePhase 1|Phase 2
NCT04491240Evaluation of Safety and Efficiency of Method of Exosome Inhalation in SARS-CoV-2 Associated Pneumonia.COVID-19|SARS-CoV-2 PNEUMONIA|COVID-19Phase 1|Phase 2
NCT04653740Omic Technologies to Track Resistance to Palbociclib in Metastatic Breast CancerAdvanced Breast CancerNA
NCT03096340Safety and Pharmacokinetic Study of IT-141 in Monotherapy in Patients with Advanced CancerCancer|Neoplasms|Tumors|Refractory Solid Tumors|Recurrent Solid TumorsPhase 1
NCT03478410Role of Exosomes Derived from Epicardial Fat in Atrial FibrillationAtrial FibrillationNA
NCT06138210The Effect of GD-iExo-003 in Acute Ischemic StrokeAcute Ischemic StrokePhase 1
NCT05787288A Clinical Study on Safety and Effectiveness of Mesenchymal Stem Cell Exosomes for the Treatment of COVID-19.COVID-19 PneumoniaEarly Phase 1
NCT04965961The Effect of Micro-doses Erytropoietin on Exercise Capacity in Male and FemalesSports Drug AbuseNA
NCT03384433Allogenic Mesenchymal Stem Cell Derived Exosome in Patients with Acute Ischemic StrokeCerebrovascular DisordersPhase 1|Phase 2
NCT06319287Phase 2a Multi-Center Prospective, Randomized Trial to Evaluate the Safety & Efficacy of Topical PEP-TISSEEL for Diabetic Foot Ulcers (DFU)Diabetic Foot UlcerPhase 2
NCT04276987A Pilot Clinical Study on Inhalation of Mesenchymal Stem Cells Exosomes Treating Severe Novel Coronavirus PneumoniaCoronavirusPhase 1
NCT06051461Deciphering the Role of Dietary Fatty Acids on Extracellular Vesicles-mediated Intercellular CommunicationObesity|Metabolic Syndrome|Metabolic Disorder|Inflammation|Immune System and Related DisordersNA
NCT06239207Efficacy and Safety of Exosomes Versus Platelet Rich Plasma in Patients of Androgenetic AlopeciaAndrogenetic AlopeciaPhase 2
NCT05933707Small Extracellular Vesicles and Insulin ActionObesity|Insulin Resistance|Metabolically Healthy Obesity|Obesity, Metabolically BenignNA
NCT01159288Trial of a Vaccination with Tumor Antigen-loaded Dendritic Cell-derived ExosomesNon-Small-Cell Lung CancerPhase 2
NCT04536688A Study of RGLS4326 in Patients with Autosomal Dominant Polycystic Kidney DiseasePolycystic Kidney Disease, Autosomal DominantPhase 1
NCT05060107Intra-articular Injection of MSC-derived Exosomes in Knee Osteoarthritis (ExoOA-1)Osteoarthritis, KneePhase 1
NCT04250493Insulin Resistance in Multiple System AtrophyMultiple System AtrophyNA
NCT05523011Safety and Tolerability Study of MSC Exosome OintmentPsoriasisPhase 1
NCT05669261Treatment of Long COVID Symptoms Utilizing Autologous Stem Cells Following COVID-19 InfectionLong COVIDPhase 1
NCT05499156Safety of Injection of Placental Mesenchymal Stem Cell Derived Exosomes for Treatment of Resistant Perianal Fistula in Crohn’s PatientsPerianal Fistula in Patients with Crohn’s DiseasePhase 1|Phase 2
NCT02507583Antisense102: Pilot Immunotherapy for Newly Diagnosed Malignant GliomaMalignant Glioma|NeoplasmsPhase 1
NCT03660683Effect of Saxagliptin and Dapagliflozin on Endothelial Progenitor Cell in Patients with Type 2 Diabetes MellitusDiabetes Mellitus, Type 2|Cardiovascular diseasesPhase 4
NCT05318898Effect of Dietary Protein on the Regulation of Exosome microRNA Expression in Patients with Insulin Resistance.Insulin ResistanceNA
NCT06244849Toward a Better Understanding of the Autophagy Machinery for the Identification of Potential Novel Biomarkers and Therapeutic Targets in Crohn’s Disease - TOPIC StudyCrohn’s DiseaseNA
NCT0382427518F-DCFPyL Positron Emission Tomography (PET)/Computed Tomography (CT) in Men with Prostate CancerProstatic NeoplasmsPhase 2|Phase 3
NCT05864534Phase 2a Immune Modulation with Ultrasound for Newly Diagnosed GlioblastomaNewly Diagnosed Glioblastoma|Glioblastoma, Isocitric Dehydrogenase (IDH)-Wildtype|Gliosarcoma|Glioblastoma MultiformePhase 2
NCT04202770Focused Ultrasound and Exosomes to Treat Depression, Anxiety, and DementiasRefractory Depression|Anxiety disorders|neurodegenerative diseasesNA
NCT01668849Edible Plant Exosome Ability to Prevent Oral Mucositis Associated with Chemoradiation Treatment of Head and Neck CancerHead and Neck Cancer|Oral MucositisPhase 1
NCT05843799A Study to Evaluate the Safety and Tolerability of ILB-202HealthyPhase 1
NCT05228899Zofin to Treat COVID-19 Long HaulersCOVID-19Phase 1|Phase 2
NCT04270006Evaluation of Adipose Derived Stem Cells Exo.in Treatment of PeriodontitisPeriodontitisEarly Phase 1
NCT05259449Efficacy of Educational Nutrition and Exercise on the Regulation of Appetite Through Exosomes in Type 2 DiabeticsDiabetes Mellitus, Type 2NA
NCT02890849Clinical Research for the Consistency Analysis of PD-L1 in Cancer Tissue and Plasma ExosomeNSCLCNA
NCT02823613The Influence of High and Low Salt on Exosomes in the UrineHealthyNA
NCT05871359Transcranial Direct Current Stimulation and Dual TasksParkinson’s DiseaseNA
NCT03437759MSC-Exos Promote Healing of MHsMacular HolesEarly Phase 1
NCT03419000Circulating microRNAs as Biomarkers of Respiratory Dysfunction in Patients with Refractory EpilepsyDrug-Resistant EpilepsyNA
NCT04427475Prediction of Immunotherapeutic Effect of Advanced Non-small Cell Lung CancerNSCLC PatientsNA
NCT04602442Safety and Efficiency of Method of Exosome Inhalation in COVID-19 Associated PneumoniaCOVID-19|SARS-CoV-2 PNEUMONIA|COVID-19Phase 2
NCT03837470Evaluation of Renal Sodium Excretion After Salt Loading in Heart Failure with Preserved Ejection FractionHeart Failure with Preserved Ejection FractionEarly Phase 1
NCT03083678Afatinib in Locally Advanced and Metastatic ChordomaChordomaPhase 2
NCT05977088Prediction of Effectiveness of rTMS Application in Alzheimer’s PatientsAlzheimer’s DiseaseNA
NCT02706288Effects of a Calorie Restricted, Very Low-Fat Plant-based Diet and Multi-component Exercise Program on Metabolic HealthObesity|Insulin ResistanceNA
NCT06391307The Role of Mesenchymal Stem Cell and Exosome in Treating Pilonidal Sinus Disease in ChildrenPilonidal Sinus|Pilonidal DiseaseNA
NCT04636788Circulating Extracellular Exosomal Small RNA as Potential Biomarker for Human Pancreatic CancerPancreas AdenocarcinomaNA
NCT03700515Exosome Proteomics to Detect EPOHealthyNA
NCT05387278Safety and Effectiveness of Placental Derived Exosomes and Umbilical Cord Mesenchymal Stem Cells in Moderate to Severe Acute Respiratory Distress Syndrome (ARDS) Associated with the Novel Corona Virus Infection (COVID-19)COVID-19 Acute Respiratory Distress Syndrome|Respiratory Distress SyndromePhase 1
NCT05834413Clinical Study on the Prevention of Driver Gene Negative II-IIIa Lung Cancer Recurrence and Metastasis by Staged Chinese Herbal Medicine Combined With Chemotherapy and Immune Checkpoint InhibitorsLung CancerNA
NCT04389385COVID-19 Specific T Cell Derived Exosomes (CSTC-Exo)Corona Virus Infection|PneumoniaPhase 1
NCT05808400Safety and Efficacy of Umbilical Cord Mesenchymal Stem Cell Exosomes in Treating Chronic Cough After COVID-19Long COVID-19 SyndromeEarly Phase 1
NCT04356300Exosome of Mesenchymal Stem Cells for Multiple Organ Dysfunction Syndrome After Surgical Repair of Acute Type A Aortic DissectionMultiple Organ FailureNA
NCT03537599Daratumumab and Donor Lymphocyte Infusion in Treating Participants with Relapsed Acute Myeloid Leukemia After Stem Cell TransplantMinimal Residual Disease|Recurrent Acute Myeloid Leukemia with Myelodysplasia-Related Changes|Recurrent Adult Acute Myeloid Leukemia|Recurrent Childhood Acute Myeloid LeukemiaPhase 1|Phase 2
NCT04544215A Clinical Study of Mesenchymal Progenitor Cell Exosomes Nebulizer for the Treatment of Pulmonary InfectionDrug-resistantPhase 1|Phase 2
NCT03627611Identification of Non-responders to Levothyroxine TherapyHypothyroidism|Biomarkers|Endocrine System DiseasesPhase 2
NCT04202783The Use of Exosomes in Craniofacial NeuralgiaNeuralgiaNA
NCT05886205Induced Pluripotent Stem Cell Derived Exosomes Nasal Drops for the Treatment of Refractory Focal EpilepsyRefractory Focal EpilepsyEarly Phase 1
NCT04902183Safety and Efficacy of Exosomes Overexpressing CD24 in Two Doses for Patients with Moderate or Severe COVID-19COVID-19Phase 2
NCT02535247Study of MK-3475 Alone or in Combination with Copanlisib in Relapsed or Refractory NK and T-cell Non-Hodgkin LymphomaLymphoma, T-Cell, PeripheralPhase 1|Phase 2
NCT03228277Olmutinib Trial in T790M (+) NSCLC Patients Detected by Liquid Biopsy Using BALF Extracellular Vesicular DNANon-Small-Cell Lung CancerPhase 2
NCT04313647A Tolerance Clinical Study on Aerosol Inhalation of Mesenchymal Stem Cells Exosomes in Healthy VolunteersHealthyPhase 1
NCT04623671Intravenous Infusion of CAP-1002 in Patients With COVID-19COVID-19Phase 2
NCT00285805The Influence of Rosiglitazone on the Diuretic Effect of Furosemide and AmilorideInsulin ResistanceNA
NCT04177498Rigosertib in Patients with Recessive Dystrophic Epidermolysis Bullosa Associated SCCRecessive Dystrophic Epidermolysis BullosaEarly Phase 1
NCT01629498Image-Guided, Intensity-Modulated Photon or Proton Beam Radiation Therapy in Treating Patients with Stage II-IIIB Non-small Cell Lung CancerRecurrent Lung Non-Small-Cell Carcinoma|Stage II Non-Small-Cell Lung Cancer AJCC v7|Stage IIA Non-Small-Cell Lung Carcinoma AJCC v7|Stage IIB Non-Small-Cell Lung Carcinoma AJCC v7|Stage IIIA Non-Small-Cell Lung Cancer AJCC v7|Stage IIIB Lung Non-Small-Cell Cancer AJCC v7Phase 1|Phase 2
NCT03927898Phase II Study of Toripalimab Plus Stereotactic Body Radiotherapy in Colorectal Cancer Patients with OligometastasisMetastatic Colorectal CancerPhase 2
NCT05947747Safety and Efficacy of EXO-CD24 in Preventing Clinical Deterioration in Patients with Mild-Moderate ARDSARDSPhase 2
NCT02594345Effect of Exosomes Derived from Red Blood Cell Units on Platelet Function and Blood CoagulationBlood Coagulation|Platelet FunctionNA
NCT04384445Zofin (Organicell Flow) for Patients With COVID-19Corona Virus Infection|COVID-19|SARS|Acute Respiratory Distress SyndromePhase 1|Phase2
NCT04298398Impact of Group Psychological Interventions on Extracellular Vesicles in People Who Had CancerCancerNA
NCT03109847Metformin Hydrochloride in Mitigating Side Effects of Radioactive Iodine Treatment in Patients with Differentiated Thyroid CancerThyroidPhase 2
NCT04350177A Study to Assess Single and Multiple Doses of IkT-148009 in Healthy Elderly Participants and Parkinson’s PatientsHealthy Elderly|Parkinson’s DiseasePhase 1
NCT05559177An Open, Dose-escalation Clinical Study of Chimeric Exosomal Tumor Vaccines for Recurrent or Metastatic Bladder CancerRecurrent or Metastatic Bladder CancerEarly Phase 1
NCT05607654The Mechanisms of Underlying Accelerated Repetitive Transcranial Magnetic Stimulation for Treatment-resistant DepressionTreatment-Resistant DepressionNA
NCT06221787Stem Cell Derived Exosomes in the Treatment of Melasma and Its Percutaneous PenetrationMelasmaNA
NCT02921854Detection of Circulating Biomarkers of Immunogenic Cell DeathNon-Small-Cell Lung CancerNA
NCT02310451Study of Molecular Mechanisms Implicated in the Pathogenesis of Melanoma. Role of ExosomesMetastatic MelanomaNA
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

Sharma, A.; Yadav, A.; Nandy, A.; Ghatak, S. Insight into the Functional Dynamics and Challenges of Exosomes in Pharmaceutical Innovation and Precision Medicine. Pharmaceutics 2024, 16, 709. https://doi.org/10.3390/pharmaceutics16060709

AMA Style

Sharma A, Yadav A, Nandy A, Ghatak S. Insight into the Functional Dynamics and Challenges of Exosomes in Pharmaceutical Innovation and Precision Medicine. Pharmaceutics. 2024; 16(6):709. https://doi.org/10.3390/pharmaceutics16060709

Chicago/Turabian Style

Sharma, Anu, Anita Yadav, Aparajita Nandy, and Subhadip Ghatak. 2024. "Insight into the Functional Dynamics and Challenges of Exosomes in Pharmaceutical Innovation and Precision Medicine" Pharmaceutics 16, no. 6: 709. https://doi.org/10.3390/pharmaceutics16060709

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