Next Article in Journal
Chitosan-Based Scaffolds Incorporated with Silver Nanoparticles for the Treatment of Infected Wounds
Previous Article in Journal
Formulation Development of Solid Self-Nanoemulsifying Drug Delivery Systems of Quetiapine Fumarate via Hot-Melt Extrusion Technology: Optimization Using Central Composite Design
Previous Article in Special Issue
Development of Ciprofloxacin-Loaded Electrospun Yarns of Application Interest as Antimicrobial Surgical Suture Materials
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Research Progress of Extracellular Vesicles-Loaded Microneedle Technology

by
Xue Wang
1,†,
Wei Cheng
2,† and
Jiandong Su
1,*
1
Department of Burn and Plastic Surgery, Suzhou Hospital Affiliated to Nanjing Medical University, Suzhou 215000, China
2
Jiangsu Key Laboratory of Thin Films, School of Physical Science and Technology, Soochow University, Suzhou 215006, China
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Pharmaceutics 2024, 16(3), 326; https://doi.org/10.3390/pharmaceutics16030326
Submission received: 30 January 2024 / Revised: 19 February 2024 / Accepted: 23 February 2024 / Published: 26 February 2024
(This article belongs to the Special Issue Novel Technologies for Buccal and Transdermal Drug Delivery)

Abstract

:
Microneedles (MNs), renowned for their painless and minimally invasive qualities, exhibit significant potential for facilitating effective drug delivery, vaccination, and targeted sample extraction. Extracellular vesicles (EVs), serving as cargo for MNs, are naturally occurring nanovesicles secreted by cells and characterized by novel biomarkers, low immunogenicity, and cell-source-specific traits. MNs prove instrumental in extracting EVs from the sample fluid, thereby facilitating a promising diagnostic and prognostic tool. To harness the therapeutic potential of EVs in tissue repair, MNs with sustained delivery of EVs leverage micron-sized channels to enhance targeted site concentration, demonstrating efficacy in treating various diseases, such as Achillea tendinopathy, hair loss, spinal cord injury, and diabetic ulcers. EV-loaded MNs emerge as a promising platform for repair applications of skin, cardiac, tendon, hair, and spinal cord tissues. This review commences with an overview of MNs, subsequently delving into the role of EVs as cargo for MNs. The paper then synthesizes the latest advancements in the use of EV-loaded MNs for tissue regenerative repair, extending to research progress in extracting EVs from MNs for disease diagnosis and prognostic evaluations. It aims to offer valuable insights and forecast future research trajectories with the hope of inspiring innovative ideas among researchers in this field.

1. Introduction

Microneedles (MNs) are microscopic needle-like structures, typically fashioned in a pyramid shape, with heights ranging from 150 to 1500 μm, widths between 50 and 250 μm, and tip thickness between 1 and 25 μm [1,2]. They are specifically engineered to penetrate the skin’s stratum corneum, creating micron-sized mechanical channels on the surface, thereby contributing to local effective drug concentration and facilitating various medical applications [3]. In routine transdermal drug delivery, MNs are distinguished for painless puncture [4], scarless healing [5], and a few gastrointestinal degradations and hepatic first-pass metabolisms [6]. In addition, recent advancements have seen the development of MNs for targeted drug delivery to various non-cutaneous organs and tissues, such as facilitating drug entry into the oral mucosa [7], eyeballs [8,9], cardiovascular tissues [10,11], spinal cord [12,13], scalp hair follicles [14], endometrium [15], etc. Using MNs to deliver drugs to different anatomical sites needs to include sensitive biosensor design and miniature flexible and biocompatible electronics in the assessment [12]. MNs can also be integrated with multiple sensors for detecting skin interstitial fluid (ISF), consistently monitoring ion concentrations, glucose, uric acid, insulin, and serotonin, providing a novel approach for disease diagnosis and prognosis monitoring [16,17,18]. Considering factors such as material composition, shape, structure, mechanical strength, and biodegradability, MNs can be custom-designed for various therapeutic applications [19]. For instance, MNs’ dressings equipped with multiple drug loads and enhanced mechanical strength not only minimize mechanical damage to non-healing wounds but also significantly expedite the wound-healing process [20]. The programmed core–shell structure of MNs can be used to regulate the inflammatory microenvironment according to the healing stages of chronic wounds by releasing drugs in a sequential response [21]. Extracellular vesicles (EVs), not only as messengers of cell-to-cell interstitial signaling exchange but also as carriers of multifarious bioactive molecules, nutrients, and garbage, exert various cellular life activities depending on different cellular origins [22,23]. A specialized type of extracellular vesicles, called matrix vesicles, can induce mineral formation in bone tissues due to their abundant calcium and phosphorus content [24]. The properties and cargo-carrying of EVs are decided by primitive cells [22]. For instance, depending on various cell types, EVs carry cell-type-specific proteins displaying specific fates and functions [25]. However, challenges in their application arise, as EVs repeatedly administered through intravenous injection lead to accelerated clearance in the circulatory system [26]. Additionally, EVs applied locally often face degradation and a consequent loss of their therapeutic activity [27]. To mitigate the accumulation of EVs in non-target organs and prevent their premature clearance, EVs are engineered to be modified by targeting their membrane components or contents [28,29]. It is also necessary to develop continuous delivery of EVs [30]. Thus, the availability of MNs for delivery has also been proposed as a relatively alternative method [31]. Researchers have recently explored a range of applications utilizing MNs for the delivery of EVs to intradermal or other non-cutaneous tissues, as well as employing MNs for the detection of EVs in specific bodily or interstitial fluids [32,33,34,35,36]. These advancements underscore the considerable potential of MN-mediated EVs in therapeutic and diagnostic applications for prognostic purposes in clinical settings [37].
This paper begins by presenting the current state of MN technology, delving into the sources, extraction methods, and status of applications involving loaded EVs. Subsequently, a comprehensive literature review was conducted on MNs and EVs from the past decade, utilizing databases such as PubMed, OVID, and Web of Science. The focus is on highlighting the most recent advancements in MNs loaded with EVs for both percutaneous and non-percutaneous delivery. Moreover, the paper outlines effective strategies for the extraction of EVs using MNs, encompassing diagnostic prognosis as well as purification and isolation techniques. Finally, it discusses the limitations encountered in the application of EV-loaded MNs and anticipates future research directions in this burgeoning field.

2. Types and Materials of Microneedles

The skin, serving as a vital protective barrier for the human body, often impedes the penetration of effective drug concentrations into the deeper tissues of wounds, particularly in cases of chronic bacterial or fungal infections caused by external injuries or immunosuppression [38,39,40]. MNs, as an innovative tool in the realm of drug delivery, are designed to inject drugs locally into the epidermis, superficial dermis, or deep dermis by penetrating the skin’s stratum corneum [41]. Research indicates that the insertion of MNs, particularly those under 1000 μm in size, avoids contact with nerve tissue and dermal blood vessels [42]. This feature allows for the achievement of painless injections, making MNs a highly effective and patient-friendly option for localized drugs [43]. In addition, MNs can be administered with less discomfort than conventional injections, increasing patient compliance, fastening puncture area healing, and avoiding the first-pass metabolism of orally administered drugs [44,45,46]. Some MNs are prepared using specially designed materials, such as PN-Si, chitosan metal nanocomposites, etc., which are naturally antimicrobial, and thus, MNs themselves can be used as therapeutic agents to promote tissue repair [47,48,49]. In addition, MN-based biosensors effectively capture dermal interstitial fluid (ISF) [50] and have been shown in preclinical experiments to be effective in monitoring blood glucose [51], electrolyte levels [52], Ph level [53], and biomarkers such as epidermal growth factor receptor 2 [54], carcinoembryonic antigen [55], cystatin C, etc. [56]. According to different drug release mechanisms, MNs are mainly categorized as coated, solid, hollow, dissolved, and soluble [57]. As shown in Figure 1, they have a wide range of tissue administrations.
The resealing time of human skin following MN treatment varies from 3 to 40 h, which can effectively enhance skin permeability for a viable duration, thereby facilitating improved drug absorption [58]. Coated MNs are applied by puncturing the skin and delivering the drug coated on their surface to the puncture site. However, an MN patch with a size of about 10–20 cm2 can only be coated with up to 1 mg of the drug, which constrains its ability to deliver effective drug concentrations for certain applications [59,60]. Enhancing the coating process to achieve a uniform coating can increase the maximum drug-loading dose of MNs without compromising the drug’s activity [61]. This improvement offers promising prospects for the large-scale production and utilization of MN patches [62,63]. The application of solid MNs involves a two-step process: initially, they pierce the skin to create a channel, followed by the application of a topical preparation [64]. However, solid MNs, along with hollow MNs, are susceptible to needle tip breakage, with the potential for the broken needle fragments to remain embedded in the skin tissue, posing biocompatibility issues [59,65]. Research indicates that the use of solid MN rollers to puncture the skin, followed by the application of EVs derived from human umbilical cord mesenchymal stem cells (HUC-MSCs), can effectively alleviate the symptoms of melasma patients, offering an enhanced experience for the patient [66]. Hollow MNs are similar in principle to the routine use of syringes and have clinical advantages in simple steps for doctors to follow [67]. As the name suggests, hollow MNs have holes in the tip and utilize a mechanism of “poke-and-flow” to deliver the drug to the skin tissue, which allows for high-dose administration compared to other types of MNs. However, a notable disadvantage is that these needles possess relatively weak mechanical strength, making them prone to breakage, and the rate of drug delivery can lead to obstructions within the lumen [3,68]. Dissolving MNs dissolve after insertion into the skin, and the drug is released with hydrolyzed or enzymatically dissolved polymers, which sustainably maintain the drug concentration and show promise toward wounds requiring prolonged healing [69]. Additionally, Han and colleagues have employed digital light processing and 3D printing techniques to fabricate polymeric MNs with barbed tips aimed at enhancing tissue adhesion and maintaining the duration of drug release [70]. The swelling MNs consist of crosslinked hydrogels that swell upon insertion into the skin by absorbing water, and the hydrogels have the ability to adapt to and mimic mechanical changes over time with a high drug loading capacity and adjustable drug release rate, overcoming the limitations of conventional MNs [71,72]. You and their team have demonstrated that the delivery of human dermal fibroblast-derived EVs using hyaluronic acid MN patches effectively reduces skin wrinkles [34]. Additionally, the loading of these patches with EVs-encapsulated collagen mRNAs leads to sustained collagen implantation in the dermis, further enhancing the improvement of skin wrinkles [34].
Materials used to prepare MNs include silicon [73], metal [6], ceramics [74], silica glass [2], silk proteins [75], polymers [76], etc., which have been summarized in detail in many reviews [3,27,77]. Polymer MNs have received more attention in recent years, and here, we focus on new advances in the preparation of MNs from this material.
A diverse range of polymers is utilized in the fabrication of MNs, commonly encompassing soluble polymers such as sodium hyaluronate, sodium carboxymethylcellulose, polyvinyl alcohol, polyvinylpyrrolidone, and degradable polymers such as polylactic acid, chitosan, poly or polylactic acid-hydroxyacetic acid copolymer, etc. [78]. In comparison to MNs made from other materials, the mechanical properties and drug release characteristics of polymer-based MNs can be tailored by modifying factors such as the cross-linking density, concentration, molecular weight, and charge properties of the polymers [79]. Soluble MNs are predominantly used in in vivo settings and recognized for their capability to quickly release drugs or vaccines within the body [80]. An example of this is noted in a study by Ito et al., where it was observed that using dextrin as a matrix material for MNs facilitated the release of almost the entire quantity of formulated insulin within just one hour [81,82]. Designing MNs with a dual-layer structure, comprising a matrix layer and a backing layer, enhances their drug delivery capabilities [83]. The matrix layer allows for rapid administration to achieve the effective dose, while the backing layer continuously replenishes the drug [84]. This design supports prolonged, effective drug release, contributing to a stable therapeutic environment [85]. Patricia G-V and colleagues developed dissolvable MNs for treating neonatal sepsis, utilizing sodium hyaluronate and polyvinylpyrrolidone as the materials for the matrix and the backing layers of the MNs, respectively [86]. In this study, gentamicin was initially administered from the needle’s tip, followed by the sustained release of the antibiotic from the backing layer through the created pores, ensuring continuous drug delivery [86]. Zhang et al., prepared hydrogel-structured modular MNs for multiple delivery of antibiotics, IL-4, and TGF-β for the treatment of periodontitis, in which the basement membrane of the MNs rapidly dissolved to release the antibiotics, and silica particles and biodegradable nanoparticles acted as the carriers for the drugs and cytokines in the hydrogel structure of the MNs, slowly releasing the encapsulated contents [87]. Similarly, the encapsulation of successfully isolated EVs derived from human adipose stem cells into the tips of MNs crafted from polymeric materials has been shown not only to preserve their molecular activity over an extended period but also to effectively regulate the release of these EVs [88].
Hydrogels are three-dimensional porous polymer networks prepared by the physical or chemical cross-linking of hydrophilic molecules, and one of the most commonly used polymers is poly methyl vinyl ether-co-maleic acid cross-linked with polyethylene glycol [89]. The hydrogels undergo swelling upon insertion into the skin without dissolving. Further, they have chemical and mechanical properties similar to those of human tissues, are biocompatible, soft, and stretchable, and can be self-healing [90,91,92,93,94]. Hydrogels find extensive applications in fields such as tissue engineering, sensor fabrication, drug delivery, and biological research. It is capable of providing an intelligent drug delivery system that responds to various types of stimuli, such as temperature [95,96,97], light [98], Ph [99,100], glucose concentration [101], active oxygen species [102], etc. Compared with other types of MNs, hydrogel MNs have the outstanding advantages of significantly higher drug loading and intelligent control of drug release rate [103,104]. Guo et al., prepared glucose-responsive hydrogel-based MN dressings with strong adhesion to diabetic wounds and responsive release of insulin to different concentrations of glucose, accelerating the diabetic wound healing process, reducing inflammatory response, and realizing intelligent drug release [105]. In addition, utilizing the water absorption and swelling characteristics, Razzaghi et al., designed 3D-printed hydrogel MNs arrays of polyethylene glycol diacrylate material integrated with multiple sensors, which allowed for the extraction of biomarkers from interstitial fluids and colorimetric diagnostic assays in a few minutes [106]. The multifunctionality and unique properties of hydrogels make them ideal systems for biomedical applications, and the preparation of soft dressings from hydrogel MNs combined with EVs is currently a research hotspot for wound repair [107]. While polymers and hydrogel materials demonstrate promising application prospects, current research often transcends the use of a single, simple polymer for MN fabrication [108]. Instead, researchers are exploring related modifications or combinations of various materials to achieve enhanced application outcomes [109]. For instance, the development of multifunctional MNs based on a magnesium metal–organic framework combined with hydrogel has shown significantly improved wound healing in diabetic mice [110]. Soluble MNs were prepared by integrating textured polysaccharides into diphenyl carbonate cross-linked cyclodextrin metal–organic frameworks, which exhibited higher mechanical strength and better physical stability than MNs made of single hyaluronic acid [111].

3. Loaded Cargos of Microneedles

On the one hand, MNs can effortlessly puncture tissues, change the local stress environment, induce skin collagen deposition and reorganization, and provide natural mechanical stimulation for tissue regeneration and wound repair [112,113]. On the other hand, MNs fully demonstrate their potential in diabetes, superficial tumors, Alzheimer’s disease, infected wounds, contraception, and other therapeutically diverse applications by carrying a wide range of drugs, including small or large molecules, vaccines, nucleic acids, nanoparticles, EVs, and cells, among others [12,59,114,115,116].
Given that this review concentrates on the application of EV-loaded MNs, our focus will be on the isolation methods and applications of EVs. EVs were initially identified in mature reticulocytes and peripheral blood platelets as globular membrane vesicles, distinct from the small cellular fragments shed by dying and damaged cells [117,118]. Almost simultaneously, matrix vesicles (MVs) were observed as electron-dense ‘leaf-like’ particles with ‘needle-like’ projections within an ossifying cartilaginous matrix [119]. Thanks to the more widespread physiological contributions of EVs, advancements in our understanding of MVs have occurred mostly in parallel with associated developments in EV biology [120]. A notable study shows that EVs from MSCs presented a clinical benefit to patients suffering from Menière’s disease by acting as a local adjuvant treatment, which is of great significance for EVs to become clinical therapeutic agents [121]. Extensive research has revealed that EVs are abundantly present in various biological fluids, including plasma, intercellular fluid, cerebrospinal fluid, urine, sperm, bile, synovial fluid, saliva, and breast milk, as well as in malignant fluids in pathological conditions, effectively facilitating cellular communication by transmitting signals [122,123]. Of note, EVs are considered to be one of the structural and functional components of the extracellular matrix [23]. EVs are now characterized as bilipid membrane structures carrying cell-specific nucleic acids, proteins, lipids, and other bioactive molecules. These molecules bind specifically to target cells, altering the structure and function of the recipient cell [124]. Not only EVs but also non-vesicular nanoparticles can carry nucleic acids, proteins, and other bioactive molecules. Based on their size, biogenesis, secretion mechanisms, surface markers, and physiological functions, EVs are classically categorized into microvesicles, apoptotic vesicles, and exosomes (exos) [125]. Recent studies have discovered new EVs, such as autophagic EVs, stressed EVs, and matrix vesicles [126]. Additional special types of EVs include membrane granules, exos-like vesicles, neutrophil-derived microvesicles, and prostasomes [127,128]. High-resolution imaging and tracking of EVs are challenging, and specific subgroups are difficult to identify with biomarkers with 100% accuracy, leading to the potential misinterpretation of the overall effect of EVs as a heterogeneous presentation of a subgroup [125,129]. Despite these challenges, EVs have leveraged existing isolation techniques, demonstrating significant therapeutic and companion diagnostic potential. Here, we offer a detailed description of their isolation methods prior to microneedling and their applications.

3.1. Isolation Methods of EVs

Gámez-Valero et al., emphasized the importance of selecting appropriate EV isolation methods tailored to the specific characteristics of EVs [130]. Most of the current isolation methods for EVs have been developed based on the biophysical principles of size separation, immunoaffinity capture, and density precipitation and can be broadly categorized into four groups with their notable properties summarized [131] (listed in Table 1). Among these, ultracentrifugation, the most commonly employed technique for EV isolation, is straightforward but fails to completely segregate EVs from other vesicular structures or proteins [132]. Moreover, high-speed centrifugation may compromise the integrity and activity of EVs [133]. Ultrafiltration-based EV isolation can shorten processing time and obtain a comparable yield to the ultracentrifugation method [112]. Several commercially developed methods for EV enrichment and isolation leverage a combination of separation techniques for efficient retrievals, such as simple dilution filtration through ligand-based EV affinity purification column chromatography, followed by centrifugal filters to procure purified platelet-derived EVs [134]. Based on polymer precipitation and aqueous two-phase system separation, the polymer precipitation method creates a hydrophobic microenvironment for obtaining EVs, which is faster and easier than ultracentrifugation methods [135]. Additionally, Nwokwu et al., employed the immunoaffinity principle for adhering EVs to porous MN structures and then isolated EVs directly from biological fluids [136]. This approach offers a broad dynamic range of temperature and incubation parameters, enhancing flexibility for laboratory and clinical applications [136]. Although the immunoaffinity method can distinguish between subgroups of vesicles, it only allows the nonspecific isolation of the EVs [137]. Recent advances in microfluidic technologies show promise for clinical utility due to their low yield and high sensitivity, particularly in diagnostic applications [138]. However, standardized protocols for EV isolation and identification techniques are lacking. Isolation techniques that enable access to high-quality, large quantities of EVs at a low cost are crucial for further research to facilitate mass production and expand medical applications.

3.2. Applications of Extracellular Vesicles (Preclinical Studies and Clinical Trials)

As research on EVs intensifies, there is growing interest in utilizing EVs as sub-cellular therapies for various diseases and as prognostic markers in disease diagnosis [147,148]. To date, the majority of EV products have not yet received approval for commercial production or clinical use due to challenges in quality control and safety concerns, though clinical and preclinical studies are on the rise annually [149]. Compared to the direct application of stem cells for disease treatment, EVs offer numerous advantages, such as high biosafety, ease of storage and transportation, rapid efficiency, and a broad range of sources [150]. The heterogeneity of EVs is attributed to the diversity of target cell receptor phenotypes and the variety of source tissues or organs. However, due to constraints related to cell activity, stimulation conditions, isolation techniques, and storage conditions, even EVs secreted by the same type of cell may exhibit significant functional differences [151,152].
EVs perform physiological functions akin to their cells of origin but exhibit relatively low immunogenicity and tumorigenicity risks and may traverse the blood–brain barrier, offering a highly effective cell-free therapy [153,154]. In fact, much of the evidence for EV transfer across the blood–brain barrier is indirect. A study reported that EVs administered intravenously in mice were detectable in cerebrospinal fluid [26]. Studies have demonstrated that EVs play roles in prostate cancer therapy and tissue engineering by being incorporated into biomaterials and bone regeneration [155,156,157]. On one hand, hepatocellular carcinoma cell-derived EVs can promote tumor formation and metastasis by inducing changes in surrounding non-cancerous cells [158]. On the other hand, they can also play an anti-cancer role by activating immune responses through the presentation of neoantigens and/or MHC–peptide complexes [159]. EVs play diverse or sequential roles in the cancer process, indicating the need for further research into their mechanisms of action for restorative purposes [160]. Yuan et al., demonstrated that adipose stem cells (ADSCs)-EV promote fibrosis and dermal regeneration in the early stages of wound healing and reduce scar formation in later stages [150]. Free EVs, lacking a controlled release mechanism, can be utilized in scaffolds not only to replace damaged or non-functional tissues but also to enhance the bioavailability of EVs at the site of action [161]. EVs are also widely used in tissue regeneration, such as improving myocardial fibrosis in arrhythmogenic cardiomyopathy [162], regenerating the retina [163], repairing damaged liver [164], improving nerve regeneration in neurological disorders [165], and promoting bone and cartilage regeneration and differentiation [166] (all proven effective in clinical or animal experiments). Currently, MSC-derived EVs have become a hot research topic and are widely used as ideal drugs or carriers for drug delivery [167]. Many studies have shown that MSC-derived EVs can promote wound healing, normalize skin structure, and reduce scar formation in various conditions like burn wounds, diabetic wounds, systemic sclerosis, radiation dermatitis, and excisional wounds [168,169]. Moreover, modifying parental MSCs or pretreatment with certain components can enhance the therapeutic effects of MSC-derived EVs. For instance, lipopolysaccharide-pretreated EVs showed a better ability to regulate macrophage homeostasis by upregulating the expression of anti-inflammatory cytokines and promoting the activation of M2 macrophages compared to untreated MSC-derived EVs [170]. This modification or pretreatment of EVs opens new horizons for further exploration of their mechanisms of action.
EVs emerging as effective biomarkers are capable of delivering specific proteins, lipids, messenger RNAs, non-coding RNAs, and other bioactive compounds [171,172]. Therefore, at present, EVs can only serve as a valid reference in disease diagnosis, and further large-scale studies are required to establish them as a diagnostic standard. A meta-analysis incorporating the detection of positive EV biomarkers of exoDNA KRASmut, ExmiR 451 a, ExmiR 200 b, ExmiR 222, and so on in 634 patients indicated that the presence of positive EV biomarkers in the blood varied in correlation with mortality at different stages of pancreatic ductal adenocarcinoma treatment [173]. This illustrates the effectiveness of EVs in disease prognosis, as endothelium-derived EVs expressing CD146 and CD105 were significantly predictive of overall survival in metastatic colorectal cancer [174]. Additionally, investigators have found significant variability in EVs between patients with castration-resistant prostate cancer who are sensitive or resistant to docetaxel, making it a valuable source of prognostic biomarkers [175].

4. Extracellular Vesicles-Loaded Microneedles: A Promising Direction

EVs are administered through various methods, including intravenous injection, intraperitoneal injection, local injection, and nebulized inhalation [176,177]. While intravenous injection is less commonly utilized, EVs have demonstrated a homing ability akin to that of parental cells and can efficiently accumulate in wound areas to aid in the healing process [178]. However, they are rapidly degraded in the systemic circulation [178]. Utilizing MNs for the target delivery of EVs to intradermal or other non-cutaneous tissues facilitates sustaining the local effective drug concentration of trauma [179]. In contrast, nebulized inhalation of EVs allows direct delivery to the fine bronchioles and alveoli, optimizing drug concentration for pulmonary applications [177]. Studies have shown that fluorescent-labeled human adipose-derived MSC-EVs reached peak concentrations in the lungs of mice with severe pneumonia 24 h post-nebulization, with a gradual decline over 28 days [180]. Local injection, though direct, is hindered by secondary wound injury and fluid flow in the body, limiting EV penetration and retention at the wound site, thus diminishing treatment efficacy [13]. Cao et al., developed a photoaging model on hairless mice to compare the effects of MN rollers alone versus a combination of topical application of ADSCs aqueous EVs [32]. Findings revealed that the combination treatment significantly reduced wrinkles, enhanced collagen fiber density and organization, and promoted inflammation clearance [32]. However, this administration method is somewhat cumbersome and carries a risk of infection. Further research is necessary to optimize EV therapy, not only in the areas of isolation methods and engineering but also in determining the optimal dose, timing, administration, and frequency of EV therapy [181]. Maintaining a sustained, effective EV concentration is crucial for effective wound healing [181].
To preserve the biological activity of EVs and control their release during long-term damage repair, the use of biocompatible biomaterials or biological devices for EV delivery has been extensively explored [182]. MNs emerge as a promising strategy for efficient EV delivery, combining minimally invasive and intelligent modulation properties with the pro-angiogenic, pro-tissue regeneration, and anti-inflammatory properties of EVs, presenting a promising avenue for therapeutic applications [13,183,184].

4.1. In Vivo Study

Notably, EV-loaded MNs have shown effectiveness in various damage models, as illustrated in Figure 2. This approach, through combining human ADSC-derived EVs with hyaluronic acid-solubilizing MNs, ensures sustained release of EVs into the dorsal skin of SKH1 mice, stimulating collagen and elastin synthesis and promoting fibroblast proliferation [88]. These MNs, under mild storage conditions, can maintain the bioactivity of EVs for over six months by proving effective on human dermal fibroblasts and enabling precise transdermal delivery [88]. In a rat diabetic wound model, MSC-derived EVs encapsulated at the tips of porous methacrylate gelatin hydrogel-based MNs were used. This innovative design facilitated the delivery of anti-inflammatory and pro-angiogenic MSC-EVs directly to the wound bed, significantly accelerating wound healing [185]. Furthermore, bilayer MNs incorporating M2 macrophage-derived EVs and polydopamine demonstrated efficacy in enhancing rat diabetic wound repair and healing [186]. In another pivotal study by Fang et al., MSC-EVs and MSCs were encapsulated in porous gelatin methacryloyl material to create an MN patch (approximately 4 mm × 4 mm, containing 45 needles) [179]. Applied to a rat T10 spinal cord injury model, the patch showed a rapid initial release of MSC-EVs within the first four days, whereas the release of MSC-EVs from MSC-inoculated MNs persisted for at least a week, aligning with the optimal treatment window for spinal cord injuries (SCI) [179]. This suggests that strategically designed MNs, with their excellent biocompatibility, can enhance stem cell survival, ensuring sustained and efficient delivery of MSC-EVs [187]. A parallel study demonstrated that EVs derived from human amniotic MSCs, attached to peptide-modified hyaluronic acid hydrogels, and locally administered in spinal cord tissues significantly reduced inducible nitric oxide synthase levels [188]. This intervention improved the inflammatory microenvironment, effectively promoting neural tissue repair and functional recovery [188]. These findings underscore the potential of EV-based therapies in regenerative medicine.
Separable MN patches composed of chitosan lactic acid (CL) and ADSC-derived EVs offer a safe and highly effective strategy for treating hair loss [14]. The inherent antimicrobial properties of CL prevent potential infection, while the sustained release of EVs promotes dermal cell proliferation and growth, thereby aiding hair regeneration [14]. In a similar design, Liu and the team prepared an array of MNs loaded with nitric oxide-modified EVs [189]. Accompanied by local nitric oxide production, these EVs passively accumulate at the injury site, significantly inhibiting the inflammatory response in Achilles tendinopathy rats, promoting tendon cell proliferation, and facilitating healing [189].
Researchers have also utilized EVs as drug carriers to enhance the protection and delivery of therapeutic agents, thereby improving therapeutic efficiency [190]. For instance, curcumin and albumin were encapsulated into EVs, which were then integrated into the tips of dissolvable MN arrays [191]. The results indicated that the encapsulated curcumin exhibited enhanced and prolonged stability, significantly and effectively alleviating skin inflammation in vivo in a mouse model [191]. Additionally, MNs were developed by fusing liposomes with EVs derived from MSCs to efficiently load and deliver ziconotide [192]. This approach offers an alternative administration route to intrathecal injection for ziconotide while also providing an effective drug delivery method for analgesia in multiple mouse pain models [192].
Figure 2. EV-loaded MNs for different therapeutic applications. (I) Schematic diagram of the fabrication of EV@MN and skincare applications. Reprinted with permission from [88] (Copyright {2023} Elsevier). (II) Illustration of the MSC-EV loaded MN patch for promoting diabetic wound healing. Reprinted with permission from [185] (Copyright {2023} Elsevier). (III) MN-mediated delivery of ziconotide-loaded liposomes fused with EVs for analgesia. Reprinted with permission from [192] (Copyright {2023} Elsevier). (IV) Schematic illustration of MN-MSC patch implantation on the injury site of the spinal cord. Reprinted with permission from [179]. (V) The manufacturing procedure of EXO is modified by a nitric oxide nanomotor-loaded MN array, and the healing process of Achilles tendinopathy occurs after the application of the MN array. Reprinted with permission from [189] (Copyright {2023} American Chemical Society). (VI) Schematic diagram of EV-encapsulated curcumin using dissolvable MN for promoting wound healing. Reprinted with permission from [191] (Copyright {2023} Elsevier).
Figure 2. EV-loaded MNs for different therapeutic applications. (I) Schematic diagram of the fabrication of EV@MN and skincare applications. Reprinted with permission from [88] (Copyright {2023} Elsevier). (II) Illustration of the MSC-EV loaded MN patch for promoting diabetic wound healing. Reprinted with permission from [185] (Copyright {2023} Elsevier). (III) MN-mediated delivery of ziconotide-loaded liposomes fused with EVs for analgesia. Reprinted with permission from [192] (Copyright {2023} Elsevier). (IV) Schematic illustration of MN-MSC patch implantation on the injury site of the spinal cord. Reprinted with permission from [179]. (V) The manufacturing procedure of EXO is modified by a nitric oxide nanomotor-loaded MN array, and the healing process of Achilles tendinopathy occurs after the application of the MN array. Reprinted with permission from [189] (Copyright {2023} American Chemical Society). (VI) Schematic diagram of EV-encapsulated curcumin using dissolvable MN for promoting wound healing. Reprinted with permission from [191] (Copyright {2023} Elsevier).
Pharmaceutics 16 00326 g002

4.2. In Clinical Study

EVs can be obtained from various cellular sources, including mesenchymal stem cells, fibroblasts, and macrophages, as well as somatic fluids [193]. However, explicit standards for isolation assays and efficacy assessments are currently lacking [193]. Although there are some limitations related to cell-based strategies due to the low survival rate, immune rejection, and tumor formation, using EVs is a helpful strategy to overcome these challenges [194]. In a first-in-human, phase I clinical trial of healthy volunteer adults, platelet EVs demonstrated safe injection as a potential wound-healing treatment [134]. Despite the absence of formal ratification for clinical use, numerous preclinical trials have substantiated the efficacy and safety of EV therapy in applications related to wound healing and tissue regeneration [195,196,197,198]. EVs present a promising cell-free therapeutic approach, particularly when pretreated and combined with a diverse array of novel materials for loading into MNs [179,199]. This approach enables efficient utilization of EVs and drug-loaded EVs while minimizing spinal cord damage [179]. Polymeric materials, chosen for their reliable biocompatibility and smart response, are commonly utilized in current studies for loading EVs [188,200]. Smart hydrogel-forming microneedle arrays as a technique for transdermal drug delivery with stimuli response show great potential in delivering EVs [201,202].

4.3. In Extraction Application

Currently, the extraction of EVs from ISF using MNs is widespread [35,37,203]. MNs obtain EVs by researching the skin of colorectal cancer-induced model mice and patient-derived xenograft models of melanoma, thereby facilitating the early detection of melanoma and colorectal cancer and providing a novel liquid biopsy method with both diagnostic and prognostic value [35,204]. In a notable study, researchers utilized MNs functionalized with anti-CD63 antibodies to co-culture with astrocyte-derived EV suspension and capture CD63+ EV subpopulations [136]. The method of MNs extracting EVs is expected to enable parallel, high-throughput isolation of various EV classes, thereby providing a direct analysis of biological fluids [136].
MN delivery of EVs addresses some of the current challenges in storage and delivery, offering a potential platform for the clinical application of EVs. Slow-release MNs loaded with EVs also provide a self-regulating drug delivery system for long-acting formulations [205]. Furthermore, the extraction of EVs from interstitial fluid post-MN acquisition for biomarker detection can effectively contribute to the early diagnosis of diseases [35]. The EV-loaded MN delivery system represents a significant advancement in drug delivery technology [154]. However, more specific testing and development are required in the future to further refine and improve this technology.

5. Conclusions

EV-loaded MNs offer a promising strategy for targeted and sustained EV concentration [192]. Extracting EVs through MNs for diagnosis and prognosis shows promise for effectively monitoring and controlling disease progression [206]. However, the impact of MNs’ material, structure, and other design factors on the bioactivity and storage time of loaded EVs requires further investigation. In the future, longer time points and larger animal models should be used for further EV-loaded MN clinical translation to test the optimal time, biosafety, and effectiveness. To promote diffusion and improve delivery efficiency, a precise porous MN structure constructed with two-photon 3D printing technology might have higher precision in controlling the size of the MNs [179]. Up to now, there has been little research on EV-loaded MNs in clinical trials. Although the journey from experimental animal models to clinical patient applications is long and challenging, the importance of EV-loaded MNs in designing efficient delivery systems has become clear. Subsequent research will concentrate on needle-loaded EVs, engineered assembly, functionalized loading, and intelligently regulated drug delivery systems to maintain the biological activity of drugs for more effective, regulated, and targeted delivery.

Author Contributions

Investigation and writing—original draft preparation, X.W. and W.C.; writing—review and editing, supervision, and funding acquisition, J.S. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Suzhou Burn Clinical Medical Center Project (Szlcyxzx202105) and the Suzhou Key Clinical Diseases Project (LCZX202315).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data sharing does not apply to this article, as no new data were created or analyzed in this study.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Akhtar, N. Microneedles: An Innovative Approach to Transdermal Delivery—A Review. Int. J. Pharm. Pharm. Sci. 2014, 6, 18–25. [Google Scholar]
  2. Larrañeta, E.; Lutton, R.E.M.; Woolfson, A.D.; Donnelly, R.F. Microneedle Arrays as Transdermal and Intradermal Drug Delivery Systems: Materials Science, Manufacture and Commercial Development. Mater. Sci. Eng. R Rep. 2016, 104, 1–32. [Google Scholar] [CrossRef]
  3. Waghule, T.; Singhvi, G.; Dubey, S.K.; Pandey, M.M.; Gupta, G.; Singh, M.; Dua, K. Microneedles: A Smart Approach and Increasing Potential for Transdermal Drug Delivery System. Biomed. Pharmacother. 2019, 109, 1249–1258. [Google Scholar] [CrossRef] [PubMed]
  4. Abdullah, H.; Phairatana, T.; Jeerapan, I. Tackling the Challenges of Developing Microneedle-Based Electrochemical Sensors. Mikrochim. Acta 2022, 189, 440. [Google Scholar] [CrossRef] [PubMed]
  5. Nguyen, T.T.; Nguyen, T.T.D.; Tran, N.-M.-A.; Vo, G.V. Advances of Microneedles in Hormone Delivery. Biomed. Pharmacother. 2022, 145, 112393. [Google Scholar] [CrossRef] [PubMed]
  6. Donnelly, R.F.; Singh, T.R.R.; Woolfson, A.D. Microneedle-Based Drug Delivery Systems: Microfabrication, Drug Delivery, and Safety. Drug Deliv. 2010, 17, 187–207. [Google Scholar] [CrossRef] [PubMed]
  7. Creighton, R.L.; Woodrow, K.A. Microneedle-Mediated Vaccine Delivery to the Oral Mucosa. Adv. Healthc. Mater. 2019, 8, 1801180. [Google Scholar] [CrossRef]
  8. Than, A.; Liu, C.; Chang, H.; Duong, P.K.; Cheung, C.M.G.; Xu, C.; Wang, X.; Chen, P. Self-Implantable Double-Layered Micro-Drug-Reservoirs for Efficient and Controlled Ocular Drug Delivery. Nat. Commun. 2018, 9, 4433. [Google Scholar] [CrossRef] [PubMed]
  9. Shi, H.; Zhou, J.; Wang, Y.; Zhu, Y.; Lin, D.; Lei, L.; Vakal, S.; Wang, J.; Li, X. A Rapid Corneal Healing Microneedle for Efficient Ocular Drug Delivery. Small 2022, 18, 2104657. [Google Scholar] [CrossRef]
  10. Huang, L.; Fang, H.; Zhang, T.; Hu, B.; Liu, S.; Lv, F.; Zeng, Z.; Liu, H.; Zhou, W.; Wang, X. Drug-Loaded Balloon with Built-in NIR Controlled Tip-Separable Microneedles for Long-Effective Arteriosclerosis Treatment. Bioact. Mater. 2023, 23, 526–538. [Google Scholar] [CrossRef]
  11. Chen, H.; Fan, L.; Peng, N.; Yin, Y.; Mu, D.; Wang, J.; Meng, R.; Xie, J. Galunisertib-Loaded Gelatin Methacryloyl Hydrogel Microneedle Patch for Cardiac Repair after Myocardial Infarction. ACS Appl. Mater. Interfaces 2022, 14, 40491. [Google Scholar] [CrossRef]
  12. Lyu, S.; Dong, Z.; Xu, X.; Bei, H.-P.; Yuen, H.-Y.; James Cheung, C.-W.; Wong, M.-S.; He, Y.; Zhao, X. Going below and beyond the Surface: Microneedle Structure, Materials, Drugs, Fabrication, and Applications for Wound Healing and Tissue Regeneration. Bioact. Mater. 2023, 27, 303–326. [Google Scholar] [CrossRef]
  13. Han, M.; Yang, H.; Lu, X.; Li, Y.; Liu, Z.; Li, F.; Shang, Z.; Wang, X.; Li, X.; Li, J.; et al. Three-Dimensional-Cultured MSC-Derived Exosome-Hydrogel Hybrid Microneedle Array Patch for Spinal Cord Repair. Nano Lett. 2022, 22, 6391–6401. [Google Scholar] [CrossRef]
  14. Shi, Y.; Zhao, J.; Li, H.; Yu, M.; Zhang, W.; Qin, D.; Qiu, K.; Chen, X.; Kong, M. A Drug-Free, Hair Follicle Cycling Regulatable, Separable, Antibacterial Microneedle Patch for Hair Regeneration Therapy. Adv. Healthc. Mater. 2022, 11, e2200908. [Google Scholar] [CrossRef] [PubMed]
  15. Zhu, Y.; Li, S.; Li, Y.; Tan, H.; Zhao, Y.; Sun, L. Antioxidant Nanozyme Microneedles with Stem Cell Loading for in Situ Endometrial Repair. Chem. Eng. J. 2022, 449, 137786. [Google Scholar] [CrossRef]
  16. Donnelly, R.F.; Mooney, K.; Caffarel-Salvador, E.; Torrisi, B.M.; Eltayib, E.; McElnay, J.C. Microneedle-mediated minimally invasive patient monitoring. Ther. Drug Monit. 2014, 36, 10–17. [Google Scholar] [CrossRef] [PubMed]
  17. Huang, X.; Zheng, S.; Liang, B.; He, M.; Wu, F.; Yang, J.; Chen, H.-J.; Xie, X. 3D-Assembled Microneedle Ion Sensor-Based Wearable System for the Transdermal Monitoring of Physiological Ion Fluctuations. Microsyst. Nanoeng. 2023, 9, 25. [Google Scholar] [CrossRef]
  18. Keyvani, F.; Zheng, H.; Kaysir, M.R.; Mantaila, D.F.; Ghavami Nejad, P.; Rahman, F.A.; Quadrilatero, J.; Ban, D.; Poudineh, M. A Hydrogel Microneedle Assay Combined with Nucleic Acid Probes for On-Site Detection of Small Molecules and Proteins. Angew. Chem. Int. Ed. Engl. 2023, 62, e202301624. [Google Scholar] [CrossRef]
  19. Khalid, R.; Mahmood, S.; Mohamed Sofian, Z.; Hilles, A.R.; Hashim, N.M.; Ge, Y. Microneedles and Their Application in Transdermal Delivery of Antihypertensive Drugs—A Review. Pharmaceutics 2023, 15, 2029. [Google Scholar] [CrossRef]
  20. Wang, Y.; Lu, H.; Guo, M.; Chu, J.; Gao, B.; He, B. Personalized and Programmable Microneedle Dressing for Promoting Wound Healing. Adv. Healthc. Mater. 2022, 11, 2101659. [Google Scholar] [CrossRef] [PubMed]
  21. Zhang, Y.; Wang, S.; Yang, Y.; Zhao, S.; You, J.; Wang, J.; Cai, J.; Wang, H.; Wang, J.; Zhang, W.; et al. Scarless Wound Healing Programmed by Core-Shell Microneedles. Nat. Commun. 2023, 14, 3431. [Google Scholar] [CrossRef]
  22. Whitford, W.; Guterstam, P. Exosome Manufacturing Status. Future Med. Chem. 2019, 11, 1225–1236. [Google Scholar] [CrossRef] [PubMed]
  23. Rilla, K.; Mustonen, A.-M.; Arasu, U.T.; Härkönen, K.; Matilainen, J.; Nieminen, P. Extracellular Vesicles Are Integral and Functional Components of the Extracellular Matrix. Matrix Biol. 2019, 75–76, 201–219. [Google Scholar] [CrossRef] [PubMed]
  24. Su, G.; Zhang, D.; Li, T.; Pei, T.; Yang, J.; Tu, S.; Liu, S.; Ren, J.; Zhang, Y.; Duan, M.; et al. Annexin A5 Derived from Matrix Vesicles Protects against Osteoporotic Bone Loss via Mineralization. Bone Res. 2023, 11, 60. [Google Scholar] [CrossRef] [PubMed]
  25. Colombo, M.; Raposo, G.; Théry, C. Biogenesis, Secretion, and Intercellular Interactions of Exosomes and Other Extracellular Vesicles. Annu. Rev. Cell Dev. Biol. 2014, 30, 255–289. [Google Scholar] [CrossRef] [PubMed]
  26. Driedonks, T.; Jiang, L.; Carlson, B.; Han, Z.; Liu, G.; Queen, S.E.; Shirk, E.N.; Gololobova, O.; Liao, Z.; Nyberg, L.H.; et al. Pharmacokinetics and Biodistribution of Extracellular Vesicles Administered Intravenously and Intranasally to Macaca Nemestrina. J. Extracell. Biol. 2022, 1, e59. [Google Scholar] [CrossRef] [PubMed]
  27. Lim, D.-J.; Kim, H.-J. Microneedles in Action: Microneedling and Microneedles-Assisted Transdermal Delivery. Polymers 2022, 14, 1608. [Google Scholar] [CrossRef]
  28. Subra, C.; Grand, D.; Laulagnier, K.; Stella, A.; Lambeau, G.; Paillasse, M.; De Medina, P.; Monsarrat, B.; Perret, B.; Silvente-Poirot, S.; et al. Exosomes Account for Vesicle-Mediated Transcellular Transport of Activatable Phospholipases and Prostaglandins. J. Lipid Res. 2010, 51, 2105–2120. [Google Scholar] [CrossRef]
  29. Zhang, F.; Guo, J.; Zhang, Z.; Duan, M.; Wang, G.; Qian, Y.; Zhao, H.; Yang, Z.; Jiang, X. Application of engineered extracellular vesicles for targeted tumor therapy. J. Biomed. Sci. 2022, 29, 14. [Google Scholar] [CrossRef]
  30. Yang, C.; Xue, Y.; Duan, Y.; Mao, C.; Wan, M. Extracellular Vesicles and Their Engineering Strategies, Delivery Systems, and Biomedical Applications. J. Control. Release Off. J. Control. Release Soc. 2024, 365, 1089–1112. [Google Scholar] [CrossRef]
  31. Fathi-Karkan, S.; Heidarzadeh, M.; Narmi, M.T.; Mardi, N.; Amini, H.; Saghati, S.; Abrbekoh, F.N.; Saghebasl, S.; Rahbarghazi, R.; Khoshfetrat, A.B. Exosome-Loaded Microneedle Patches: Promising Factor Delivery Route. Int. J. Biol. Macromol. 2023, 243, 125232. [Google Scholar] [CrossRef]
  32. Cao, Z.; Jin, S.; Wang, P.; He, Q.; Yang, Y.; Gao, Z.; Wang, X. Microneedle Based Adipose Derived Stem Cells-derived Extracellular Vesicles Therapy Ameliorates UV-induced Photoaging in SKH-1 Mice. J. Biomed. Mater. Res. 2021, 109, 1849–1857. [Google Scholar] [CrossRef]
  33. Yuan, M.; Liu, K.; Jiang, T.; Li, S.; Chen, J.; Wu, Z.; Li, W.; Tan, R.; Wei, W.; Yang, X.; et al. GelMA/PEGDA microneedles patch loaded with HUVECs-derived exosomes and Tazarotene promote diabetic wound healing. J. Nanobiotechnology 2022, 20, 147. [Google Scholar] [CrossRef]
  34. You, Y.; Tian, Y.; Yang, Z.; Shi, J.; Kwak, K.J.; Tong, Y.; Estania, A.P.; Cao, J.; Hsu, W.-H.; Liu, Y.; et al. Intradermally Delivered mRNA-Encapsulating Extracellular Vesicles for Collagen-Replacement Therapy. Nat. Biomed. Eng. 2023, 7, 887–900. [Google Scholar] [CrossRef] [PubMed]
  35. Park, W.; Maeng, S.-W.; Mok, J.W.; Choi, M.; Cha, H.J.; Joo, C.-K.; Hahn, S.K. Hydrogel Microneedles Extracting Exosomes for Early Detection of Colorectal Cancer. Biomacromolecules 2023, 24, 1445–1452. [Google Scholar] [CrossRef]
  36. Cao, Y.; Xu, Y.; Chen, C.; Xie, H.; Lu, H.; Hu, J. Local Delivery of USC-Derived Exosomes Harboring ANGPTL3 Enhances Spinal Cord Functional Recovery after Injury by Promoting Angiogenesis. Stem Cell Res. Ther. 2021, 12, 20. [Google Scholar] [CrossRef]
  37. Taylor, R.M.; Miller, P.R.; Ebrahimi, P.; Polsky, R.; Baca, J.T. Minimally-Invasive, Microneedle-Array Extraction of Interstitial Fluid for Comprehensive Biomedical Applications: Transcriptomics, Proteomics, Metabolomics, Exosome Research, and Biomarker Identification. Lab. Anim. 2018, 52, 526–530. [Google Scholar] [CrossRef] [PubMed]
  38. Esposito, S.; Ascione, T.; Pagliano, P. Management of Bacterial Skin and Skin Structure Infections with Polymicrobial Etiology. Expert Rev. Anti-Infect. Ther. 2019, 17, 17–25. [Google Scholar] [CrossRef]
  39. Savin, J.A.; Noble, W.C. Immunosuppression and Skin Infection. Br. J. Dermatol. 1975, 93, 115–120. [Google Scholar] [CrossRef] [PubMed]
  40. Russell, L.M.; Wiedersberg, S.; Delgado-Charro, M.B. The Determination of Stratum Corneum Thickness: An Alternative Approach. Eur. J. Pharm. Biopharm. 2008, 69, 861–870. [Google Scholar] [CrossRef]
  41. Jiang, X.; Zhao, H.; Li, W. Microneedle-Mediated Transdermal Delivery of Drug-Carrying Nanoparticles. Front. Bioeng. Biotechnol. 2022, 10, 840395. [Google Scholar] [CrossRef] [PubMed]
  42. Gill, H.S.; Denson, D.D.; Burris, B.A.; Prausnitz, M.R. Effect of Microneedle Design on Pain in Human Volunteers. Clin. J. Pain 2008, 24, 585. [Google Scholar] [CrossRef]
  43. Miller, P.R.; Taylor, R.M.; Tran, B.Q.; Boyd, G.; Glaros, T.; Chavez, V.H.; Krishnakumar, R.; Sinha, A.; Poorey, K.; Williams, K.P.; et al. Extraction and Biomolecular Analysis of Dermal Interstitial Fluid Collected with Hollow Microneedles. Commun. Biol. 2018, 1, 173. [Google Scholar] [CrossRef]
  44. Narayan, R.J. Transdermal delivery of insulin via microneedles. J. Biomed. Nanotechnol. 2014, 10, 2244–2260. [Google Scholar] [CrossRef]
  45. Bariya, S.H.; Gohel, M.C.; Mehta, T.A.; Sharma, O.P. Microneedles: An Emerging Transdermal Drug Delivery System. J. Pharm. Pharmacol. 2012, 64, 11–29. [Google Scholar] [CrossRef]
  46. Jeong, H.R.; Lee, H.S.; Choi, I.J.; Park, J.H. Considerations in the Use of Microneedles: Pain, Convenience, Anxiety and Safety. J. Drug Target. 2017, 25, 29–40. [Google Scholar] [CrossRef] [PubMed]
  47. Ni, M.; Li, W.; Yuan, B.; Zou, S.; Cheng, W.; Yang, K.; Su, J.; Sun, B.; Su, X. Micro-Structured P-N Junction Surfaces: Large-Scale Preparation, Antifouling Properties, and a Synergistic Antibacterial Mechanism. J. Mater. Chem. B 2023, 11, 1312–1319. [Google Scholar] [CrossRef]
  48. Mohandas, A.; Deepthi, S.; Biswas, R.; Jayakumar, R. Chitosan Based Metallic Nanocomposite Scaffolds as Antimicrobial Wound Dressings. Bioact. Mater. 2018, 3, 267–277. [Google Scholar] [CrossRef]
  49. Ni, M.; Jiang, C.; Cheng, W.; Yang, K.; Dai, L.; Zeng, Y.; Su, J.; Lu, Z.; Zou, S.; Su, X. A Solid Strategy to Realize Efficient Antibacterial Activity on the Shade Surface of Bulk Silicon under Natural or Indoor Lighting. Chem. Eng. J. 2024, 479, 147734. [Google Scholar] [CrossRef]
  50. Erdem, Ö.; Eş, I.; Akceoglu, G.A.; Saylan, Y.; Inci, F. Recent Advances in Microneedle-Based Sensors for Sampling, Diagnosis and Monitoring of Chronic Diseases. Biosensors 2021, 11, 296. [Google Scholar] [CrossRef]
  51. Zhang, B.L.; Yang, Y.; Zhao, Z.Q.; Guo, X.D. A gold nanoparticles deposited polymer microneedle enzymatic biosensor for glucose sensing. Electrochim. Acta 2020, 358, 136917. [Google Scholar] [CrossRef]
  52. Molinero-Fernández, Á.; Casanova, A.; Wang, Q.; Cuartero, M.; Crespo, G.A. In Vivo Transdermal Multi-Ion Monitoring with a Potentiometric Microneedle-Based Sensor Patch. ACS Sens. 2023, 8, 158–166. [Google Scholar] [CrossRef]
  53. Dervisevic, M.; Dervisevic, E.; Esser, L.; Easton, C.D.; Cadarso, V.J.; Voelcker, N.H. Wearable Microneedle Array-Based Sensor for Transdermal Monitoring of pH Levels in Interstitial Fluid. Biosens. Bioelectron. 2023, 222, 114955. [Google Scholar] [CrossRef] [PubMed]
  54. Dervisevic, M.; Alba, M.; Adams, T.E.; Prieto-Simon, B.; Voelcker, N.H. Electrochemical Immunosensor for Breast Cancer Biomarker Detection Using High-Density Silicon Microneedle Array. Biosens. Bioelectron. 2021, 192, 113496. [Google Scholar] [CrossRef] [PubMed]
  55. Chen, L.; Zhang, C.; Xiao, J.; You, J.; Zhang, W.; Liu, Y.; Xu, L.; Liu, A.; Xin, H.; Wang, X. Local Extraction and Detection of Early Stage Breast Cancers through a Microneedle and Nano-Ag/MBL Film Based Painless and Blood-Free Strategy. Mater. Sci. Eng. C 2020, 109, 110402. [Google Scholar] [CrossRef] [PubMed]
  56. Tran, B.Q.; Miller, P.R.; Taylor, R.M.; Boyd, G.; Mach, P.M.; Rosenzweig, C.N.; Baca, J.T.; Polsky, R.; Glaros, T. Proteomic Characterization of Dermal Interstitial Fluid Extracted Using a Novel Microneedle-Assisted Technique. J. Proteome Res. 2018, 17, 479–485. [Google Scholar] [CrossRef]
  57. Ita, K. Transdermal Delivery of Drugs with Microneedles: Strategies and Outcomes. J. Drug Deliv. Sci. Technol. 2015, 29, 16–23. [Google Scholar] [CrossRef]
  58. Gupta, J.; Gill, H.S.; Andrews, S.N.; Prausnitz, M.R. Kinetics of Skin Resealing after Insertion of Microneedles in Human Subjects. J. Control Release 2011, 154, 148–155. [Google Scholar] [CrossRef]
  59. Yang, J.; Liu, X.; Fu, Y.; Song, Y. Recent Advances of Microneedles for Biomedical Applications: Drug Delivery and Beyond. Acta Pharm. Sin. B 2019, 9, 469–483. [Google Scholar] [CrossRef]
  60. Gill, H.S.; Prausnitz, M.R. Coating Formulations for Microneedles. Pharm. Res. 2007, 24, 1369–1380. [Google Scholar] [CrossRef]
  61. Gidlöf, Z. Coating of Bioceramic Microneedles. 2017. Available online: https://www.diva-portal.org/smash/get/diva2:1091657/FULLTEXT01.pdf (accessed on 15 February 2024).
  62. Camović, M.; Biščević, A.; Brčić, I.; Borčak, K.; Bušatlić, S.; Ćenanović, N.; Dedović, A.; Mulalić, A.; Osmanlić, M.; Sirbubalo, M.; et al. Coated 3D Printed PLA Microneedles as Transdermal Drug Delivery Systems. In Proceedings of the CMBEBIH 2019, Banja Luka, Bosnia and Herzegovina, 16–18 May 2019; Springer International Publishing: Cham, Switzerland, 2020; Volume 73, pp. 735–742. [Google Scholar]
  63. Tarbox, T.N.; Watts, A.B.; Cui, Z.; Williams, R.O., III. An update on coating/manufacturing techniques of microneedles. Drug Deliv. Transl. Res. 2018, 8, 1828–1843. [Google Scholar] [CrossRef]
  64. Khare, N.; Shende, P. Microneedle System: A Modulated Approach for Penetration Enhancement. Drug Dev. Ind. Pharm. 2021, 47, 1183–1192. [Google Scholar] [CrossRef] [PubMed]
  65. Van Der Maaden, K.; Jiskoot, W.; Bouwstra, J. Microneedle Technologies for (Trans)Dermal Drug and Vaccine Delivery. J. Control. Release 2012, 161, 645–655. [Google Scholar] [CrossRef]
  66. Wang, T.; Gao, H.; Wang, D.; Zhang, C.; Hu, K.; Zhang, H.; Lin, J.; Chen, X. Stem cell-derived exosomes in the treatment of melasma and its percutaneous penetration. Lasers Surg. Med. 2023, 55, 178–189. [Google Scholar] [CrossRef] [PubMed]
  67. Badnikar, K.; Jayadevi, S.N.; Pahal, S.; Vemula, P.K.; Nayak, M.M.; Subramanyam, D.N. Microscale Engineering of Hollow Microneedle Tips: Design, Manufacturing, Optimization and Validation. Drug Deliv. Transl. Res. 2022, 12, 350–367. [Google Scholar] [CrossRef] [PubMed]
  68. Wang, P.M.; Cornwell, M.; Hill, J.; Prausnitz, M.R. Precise Microinjection into Skin Using Hollow Microneedles. J. Investig. Dermatol. 2006, 126, 1080–1087. [Google Scholar] [CrossRef] [PubMed]
  69. Aghabegi Moghanjoughi, A.; Khoshnevis, D.; Zarrabi, A. A Concise Review on Smart Polymers for Controlled Drug Release. Drug Deliv. Transl. Res. 2016, 6, 333–340. [Google Scholar] [CrossRef] [PubMed]
  70. Han, D.; Morde, R.S.; Mariani, S.; La Mattina, A.A.; Vignali, E.; Yang, C.; Barillaro, G.; Lee, H. 4D Printing of a Bioinspired Microneedle Array with Backward-Facing Barbs for Enhanced Tissue Adhesion. Adv. Funct. Mater. 2020, 30, 1909197. [Google Scholar] [CrossRef]
  71. El-Husseiny, H.M.; Mady, E.A.; Hamabe, L.; Abugomaa, A.; Shimada, K.; Yoshida, T.; Tanaka, T.; Yokoi, A.; Elbadawy, M.; Tanaka, R. Smart/Stimuli-Responsive Hydrogels: Cutting-Edge Platforms for Tissue Engineering and Other Biomedical Applications. Mater. Today Bio 2022, 13, 100186. [Google Scholar] [CrossRef]
  72. Chai, Q.; Jiao, Y.; Yu, X. Hydrogels for Biomedical Applications: Their Characteristics and the Mechanisms behind Them. Gels 2017, 3, 6. [Google Scholar] [CrossRef]
  73. Cheng, W.; Wang, X.; Zou, S.; Ni, M.; Lu, Z.; Dai, L.; Su, J.; Yang, K.; Su, X. Fabrication of Black Silicon Microneedle Arrays for High Drug Loading. JFB 2023, 14, 245. [Google Scholar] [CrossRef]
  74. Hartmann, X.H.M.; van der Linde, P.; Homburg, E.F.G.A.; van Breemen, L.C.A.; de Jong, A.M.; Luttge, R. Insertion Process of Ceramic Nanoporous Microneedles by Means of a Novel Mechanical Applicator Design. Pharmaceutics 2015, 7, 503–522. [Google Scholar] [CrossRef]
  75. Lin, Z.; Li, Y.; Meng, G.; Hu, X.; Zeng, Z.; Zhao, B.; Lin, N.; Liu, X.Y. Reinforcement of Silk Microneedle Patches for Accurate Transdermal Delivery. Biomacromolecules 2021, 22, 5319–5326. [Google Scholar] [CrossRef] [PubMed]
  76. Shriky, B.; Babenko, M.; Whiteside, B.R. Dissolving and Swelling Hydrogel-Based Microneedles: An Overview of Their Materials, Fabrication, Characterization Methods, and Challenges. Gels 2023, 9, 806. [Google Scholar] [CrossRef] [PubMed]
  77. Aldawood, F.K.; Andar, A.; Desai, S. A Comprehensive Review of Microneedles: Types, Materials, Processes, Characterizations and Applications. Polymers 2021, 13, 2815. [Google Scholar] [CrossRef] [PubMed]
  78. Ahmed Saeed AL-Japairai, K.; Mahmood, S.; Hamed Almurisi, S.; Reddy Venugopal, J.; Rebhi Hilles, A.; Azmana, M.; Raman, S. Current Trends in Polymer Microneedle for Transdermal Drug Delivery. Int. J. Pharm. 2020, 587, 119673. [Google Scholar] [CrossRef] [PubMed]
  79. Luo, Z.; Sun, W.; Fang, J.; Lee, K.; Li, S.; Gu, Z.; Dokmeci, M.R.; Khademhosseini, A. Biodegradable Gelatin Methacryloyl Microneedles for Transdermal Drug Delivery. Adv. Healthc. Mater. 2019, 8, 1801054. [Google Scholar] [CrossRef] [PubMed]
  80. Rouphael, N.G.; Paine, M.; Mosley, R.; Henry, S.; McAllister, D.V.; Kalluri, H.; Pewin, W.; Frew, P.M.; Yu, T.; Thornburg, N.J.; et al. The Safety, Immunogenicity, and Acceptability of Inactivated Influenza Vaccine Delivered by Microneedle Patch (TIV-MNP 2015): A Randomised, Partly Blinded, Placebo-Controlled, Phase 1 Trial. Lancet 2017, 390, 649–658. [Google Scholar] [CrossRef]
  81. Ito, Y.; Hagiwara, E.; Saeki, A.; Sugioka, N.; Takada, K. Feasibility of Microneedles for Percutaneous Absorption of Insulin. Eur. J. Pharm. Sci. 2006, 29, 82–88. [Google Scholar] [CrossRef]
  82. Hong, X.; Wei, L.; Wu, F.; Wu, Z.; Chen, L.; Liu, Z.; Yuan, W. Dissolving and Biodegradable Microneedle Technologies for Transdermal Sustained Delivery of Drug and Vaccine. Drug Des. Dev. Ther. 2013, 945, 945–952. [Google Scholar]
  83. Ono, A.; Azukizawa, H.; Ito, S.; Nakamura, Y.; Asada, H.; Quan, Y.-S.; Kamiyama, F.; Katayama, I.; Hirobe, S.; Okada, N. Development of Novel Double-Decker Microneedle Patches for Transcutaneous Vaccine Delivery. Int. J. Pharm. 2017, 532, 374–383. [Google Scholar] [CrossRef]
  84. Younas, A.; Dong, Z.; Hou, Z.; Asad, M.; Li, M.; Zhang, N. A Chitosan/Fucoidan Nanoparticle-Loaded Pullulan Microneedle Patch for Differential Drug Release to Promote Wound Healing. Carbohydr. Polym. 2023, 306, 120593. [Google Scholar] [CrossRef]
  85. Chen, M.-C.; Lai, K.-Y.; Ling, M.-H.; Lin, C.-W. Enhancing Immunogenicity of Antigens through Sustained Intradermal Delivery Using Chitosan Microneedles with a Patch-Dissolvable Design. Acta Biomater. 2018, 65, 66–75. [Google Scholar] [CrossRef]
  86. González-Vázquez, P.; Larrañeta, E.; McCrudden, M.T.C.; Jarrahian, C.; Rein-Weston, A.; Quintanar-Solares, M.; Zehrung, D.; McCarthy, H.; Courtenay, A.J.; Donnelly, R.F. Transdermal Delivery of Gentamicin Using Dissolving Microneedle Arrays for Potential Treatment of Neonatal Sepsis. J. Control Release 2017, 265, 30–40. [Google Scholar] [CrossRef]
  87. Zhang, X.; Hasani-Sadrabadi, M.M.; Zarubova, J.; Dashtimighadam, E.; Haghniaz, R.; Khademhosseini, A.; Butte, M.J.; Moshaverinia, A.; Aghaloo, T.; Li, S. Immunomodulatory Microneedle Patch for Periodontal Tissue Regeneration. Matter 2022, 5, 666–682. [Google Scholar] [CrossRef]
  88. Bui, V.D.; Son, S.; Xavier, W.; Jung, J.M.; Lee, J.; Shin, S.; Um, W.; An, J.Y.; Kim, C.H.; Song, Y.; et al. Dissolving Microneedles for Long-Term Storage and Transdermal Delivery of Extracellular Vesicles. Biomaterials 2022, 287, 121644. [Google Scholar] [CrossRef]
  89. Sartawi, Z.; Blackshields, C.; Faisal, W. Dissolving Microneedles: Applications and Growing Therapeutic Potential. J. Control. Release 2022, 348, 186–205. [Google Scholar] [CrossRef]
  90. Liu, Y.; Zhang, H.; Zhang, J.; Zheng, Y. Transient Swelling of Polymeric Hydrogels: A New Finite Element Solution Framework. Int. J. Solids Struct. 2016, 80, 246–260. [Google Scholar] [CrossRef]
  91. Mozhdehi, D.; Ayala, S.; Cromwell, O.R.; Guan, Z. Self-healing multiphase polymers via dynamic metal-ligand interactions. J. Am. Chem. Soc. 2014, 136, 16128–16131. [Google Scholar] [CrossRef] [PubMed]
  92. Liao, H.; Guo, X.; Wan, P.; Yu, G. Conductive MXene Nanocomposite Organohydrogel for Flexible, Healable, Low-Temperature Tolerant Strain Sensors. Adv. Funct. Mater. 2019, 29, 1904507. [Google Scholar] [CrossRef]
  93. Liu, X.; Liu, J.; Lin, S.; Zhao, X. Hydrogel Machines. Mater. Today 2020, 36, 102–124. [Google Scholar] [CrossRef]
  94. Sun, W.; Duan, T.; Cao, Y.; Li, H. An Injectable Self-Healing Protein Hydrogel with Multiple Dissipation Modes and Tunable Dynamic Response. Biomacromolecules 2019, 20, 4199–4207. [Google Scholar] [CrossRef] [PubMed]
  95. Shen, H.; Lin, Q.; Tang, H.; Tian, Y.; Zhang, X. Fabrication of Temperature- and Alcohol-Responsive Photonic Crystal Hydrogel and Its Application for Sustained Drug Release. Langmuir 2022, 38, 3785–3794. [Google Scholar] [CrossRef] [PubMed]
  96. Su, X.; Feng, Y.; Shi, H.; Wang, F.; Wang, Z.; Hou, S.; Song, X.; Yang, J.; Liu, L. A Hydrogel Dressing with Tunable Critical Temperature and Photothermal Modulating Melittin Release for Multiply Antibacterial Treatment. Int. J. Biol. Macromol. 2023, 239, 124272. [Google Scholar] [CrossRef]
  97. Chen, G.; Zhou, Y.; Dai, J.; Yan, S.; Miao, W.; Ren, L. Calcium Alginate/PNIPAAm Hydrogel with Body Temperature Response and Great Biocompatibility: Application as Burn Wound Dressing. Int. J. Biol. Macromol. 2022, 216, 686–697. [Google Scholar] [CrossRef] [PubMed]
  98. Jiang, B.; Liu, X.; Yang, C.; Yang, Z.; Luo, J.; Kou, S.; Liu, K.; Sun, F. Injectable, Photoresponsive Hydrogels for Delivering Neuroprotective Proteins Enabled by Metal-Directed Protein Assembly. Sci. Adv. 2020, 6, eabc4824. [Google Scholar] [CrossRef]
  99. Odinotski, S.; Dhingra, K.; GhavamiNejad, A.; Zheng, H.; GhavamiNejad, P.; Gaouda, H.; Mohammadrezaei, D.; Poudineh, M. A Conductive Hydrogel-Based Microneedle Platform for Real-Time pH Measurement in Live Animals. Small 2022, 18, e2200201. [Google Scholar] [CrossRef]
  100. Edirisinghe, D.I.U.; D’Souza, A.; Ramezani, M.; Carroll, R.J.; Chicón, Q.; Muenzel, C.L.; Soule, J.; Monroe, M.B.B.; Patteson, A.E.; Makhlynets, O.V. Antibacterial and Cytocompatible pH-Responsive Peptide Hydrogel. Molecules 2023, 28, 4390. [Google Scholar] [CrossRef]
  101. Xu, Z.; Liu, G.; Liu, P.; Hu, Y.; Chen, Y.; Fang, Y.; Sun, G.; Huang, H.; Wu, J. Hyaluronic Acid-Based Glucose-Responsive Antioxidant Hydrogel Platform for Enhanced Diabetic Wound Repair. Acta Biomater. 2022, 147, 147–157. [Google Scholar] [CrossRef]
  102. Yu, H.; Huang, C.; Kong, X.; Ma, J.; Ren, P.; Chen, J.; Zhang, X.; Luo, H.; Chen, G. Nanoarchitectonics of Cartilage-Targeting Hydrogel Microspheres with Reactive Oxygen Species Responsiveness for the Repair of Osteoarthritis. ACS Appl. Mater. Interfaces 2022, 14, 40711–40723. [Google Scholar] [CrossRef]
  103. Su, M.; Ruan, L.; Dong, X.; Tian, S.; Lang, W.; Wu, M.; Chen, Y.; Lv, Q.; Lei, L. Current State of Knowledge on Intelligent-Response Biological and Other Macromolecular Hydrogels in Biomedical Engineering: A Review. Int. J. Biol. Macromol. 2023, 227, 472–492. [Google Scholar] [CrossRef]
  104. Liang, Y.; He, J.; Guo, B. Functional Hydrogels as Wound Dressing to Enhance Wound Healing. ACS Nano 2021, 15, 12687–12722. [Google Scholar] [CrossRef]
  105. Guo, Z.; Liu, H.; Shi, Z.; Lin, L.; Li, Y.; Wang, M.; Pan, G.; Lei, Y.; Xue, L. Responsive Hydrogel-Based Microneedle Dressing for Diabetic Wound Healing. J. Mater. Chem. B 2022, 10, 3501–3511. [Google Scholar] [CrossRef]
  106. Razzaghi, M.; Seyfoori, A.; Pagan, E.; Askari, E.; Hassani Najafabadi, A.; Akbari, M. 3D Printed Hydrogel Microneedle Arrays for Interstitial Fluid Biomarker Extraction and Colorimetric Detection. Polymers 2023, 15, 1389. [Google Scholar] [CrossRef]
  107. Ma, W.; Zhang, X.; Liu, Y.; Fan, L.; Gan, J.; Liu, W.; Zhao, Y.; Sun, L. Polydopamine Decorated Microneedles with Fe-MSC-Derived Nanovesicles Encapsulation for Wound Healing. Adv. Sci. 2022, 9, 2103317. [Google Scholar] [CrossRef]
  108. Chi, J.; Zhang, X.; Chen, C.; Shao, C.; Zhao, Y.; Wang, Y. Antibacterial and Angiogenic Chitosan Microneedle Array Patch for Promoting Wound Healing. Bioact. Mater. 2020, 5, 253–259. [Google Scholar] [CrossRef] [PubMed]
  109. Li, B.; Lu, G.; Liu, W.; Liao, L.; Ban, J.; Lu, Z. Formulation and Evaluation of PLGA Nanoparticulate-Based Microneedle System for Potential Treatment of Neurological Diseases. Int. J. Nanomed. 2023, 18, 3745–3760. [Google Scholar] [CrossRef] [PubMed]
  110. Yin, M.; Wu, J.; Deng, M.; Wang, P.; Ji, G.; Wang, M.; Zhou, C.; Blum, N.T.; Zhang, W.; Shi, H.; et al. Multifunctional Magnesium Organic Framework-Based Microneedle Patch for Accelerating Diabetic Wound Healing. ACS Nano 2021, 15, 17842–17853. [Google Scholar] [CrossRef]
  111. Wu, T.; Hou, X.; Li, J.; Ruan, H.; Pei, L.; Guo, T.; Wang, Z.; Ci, T.; Ruan, S.; He, Y.; et al. Microneedle-Mediated Biomimetic Cyclodextrin Metal Organic Frameworks for Active Targeting and Treatment of Hypertrophic Scars. ACS Nano 2021, 15, 20087–20104. [Google Scholar] [CrossRef] [PubMed]
  112. Choi, S.Y.; Kwon, H.J.; Ahn, G.R.; Ko, E.J.; Yoo, K.H.; Kim, B.J.; Lee, C.; Kim, D. Hyaluronic Acid Microneedle Patch for the Improvement of Crow’s Feet Wrinkles. Dermatol. Ther. 2017, 30, e12546. [Google Scholar] [CrossRef] [PubMed]
  113. Xing, M.; Liu, H.; Meng, F.; Ma, Y.; Zhang, S.; Gao, Y. Design and Evaluation of Complex Polypeptide-Loaded Dissolving Microneedles for Improving Facial Wrinkles in Different Areas. Polymers 2022, 14, 4475. [Google Scholar] [CrossRef]
  114. He, M.; Yang, G.; Zhao, X.; Zhang, S.; Gao, Y. Intradermal Implantable PLGA Microneedles for Etonogestrel Sustained Release. J. Pharm. Sci. 2020, 109, 1958–1966. [Google Scholar] [CrossRef]
  115. Jin, X.; Zhu, D.D.; Chen, B.Z.; Ashfaq, M.; Guo, X.D. Insulin Delivery Systems Combined with Microneedle Technology. Adv. Drug Deliv. Rev. 2018, 127, 119–137. [Google Scholar] [CrossRef]
  116. Jamaledin, R.; Yiu, C.K.Y.; Zare, E.N.; Niu, L.-N.; Vecchione, R.; Chen, G.; Gu, Z.; Tay, F.R.; Makvandi, P. Advances in Antimicrobial Microneedle Patches for Combating Infections. Adv. Mater. 2020, 32, 2002129. [Google Scholar] [CrossRef]
  117. Wolf, P. The Nature and Significance of Platelet Products in Human Plasma. Br. J. Haematol. 1967, 13, 269–288. [Google Scholar] [CrossRef]
  118. Schrier, S.L.; Godin, D.; Gould, R.G.; Swyryd, B.; Junga, I.; Seeger, M. Characterization of Microvesicles Produced by Shearing of Human Erythrocyte Membranes. Biochim. Biophys. Acta 1971, 233, 26–36. [Google Scholar] [CrossRef]
  119. Anderson, H.C. Electron Microscopic Studies of Induced Cartilage Development and Calcification. J. Cell Biol. 1967, 35, 81–101. [Google Scholar] [CrossRef]
  120. Azoidis, I.; Cox, S.C.; Davies, O.G. The Role of Extracellular Vesicles in Biomineralisation: Current Perspective and Application in Regenerative Medicine. J. Tissue Eng. 2018, 9, 2041731418810130. [Google Scholar] [CrossRef]
  121. Tan, T.T.; Toh, W.S.; Lai, R.C.; Lim, S.K. Practical Considerations in Transforming MSC Therapy for Neurological Diseases from Cell to EV. Exp. Neurol. 2022, 349, 113953. [Google Scholar] [CrossRef]
  122. Gill, S.; Catchpole, R.; Forterre, P. Extracellular Membrane Vesicles in the Three Domains of Life and Beyond. FEMS Microbiol. Rev. 2019, 43, 273–303. [Google Scholar] [CrossRef]
  123. Ratajczak, M.Z.; Ratajczak, J. Extracellular Microvesicles/Exosomes: Discovery, Disbelief, Acceptance, and the Future? Leukemia 2020, 34, 3126–3135. [Google Scholar] [CrossRef]
  124. Fyfe, J.; Casari, I.; Manfredi, M.; Falasca, M. Role of Lipid Signalling in Extracellular Vesicles-Mediated Cell-to-Cell Communication. Cytokine Growth Factor. Rev. 2023, 73, 20–26. [Google Scholar] [CrossRef] [PubMed]
  125. Jadli, A.S.; Ballasy, N.; Edalat, P.; Patel, V.B. Inside(Sight) of Tiny Communicator: Exosome Biogenesis, Secretion, and Uptake. Mol. Cell Biochem. 2020, 467, 77–94. [Google Scholar] [CrossRef] [PubMed]
  126. Sheta, M.; Taha, E.A.; Lu, Y.; Eguchi, T. Extracellular Vesicles: New Classification and Tumor Immunosuppression. Biology 2023, 12, 110. [Google Scholar] [CrossRef] [PubMed]
  127. Gomez, I.; Ward, B.; Souilhol, C.; Recarti, C.; Ariaans, M.; Johnston, J.; Burnett, A.; Mahmoud, M.; Luong, L.A.; West, L.; et al. Neutrophil Microvesicles Drive Atherosclerosis by Delivering miR-155 to Atheroprone Endothelium. Nat. Commun. 2020, 11, 214. [Google Scholar] [CrossRef] [PubMed]
  128. Thakur, A.; Ke, X.; Chen, Y.-W.; Motallebnejad, P.; Zhang, K.; Lian, Q.; Chen, H.J. The Mini Player with Diverse Functions: Extracellular Vesicles in Cell Biology, Disease, and Therapeutics. Protein Cell 2022, 13, 631–654. [Google Scholar] [CrossRef] [PubMed]
  129. Janouskova, O.; Herma, R.; Semeradtova, A.; Poustka, D.; Liegertova, M.; Malinska, H.A.; Maly, J. Conventional and Nonconventional Sources of Exosomes–Isolation Methods and Influence on Their Downstream Biomedical Application. Front. Mol. Biosci. 2022, 9, 846650. [Google Scholar] [CrossRef]
  130. Gámez-Valero, A.; Monguió-Tortajada, M.; Carreras-Planella, L.; la Franquesa, M.; Beyer, K.; Borràs, F.E. Size-Exclusion Chromatography-Based Isolation Minimally Alters Extracellular Vesicles’ Characteristics Compared to Precipitating Agents. Sci. Rep. 2016, 6, 33641. [Google Scholar] [CrossRef] [PubMed]
  131. Gupta, D.; Zickler, A.M.; El Andaloussi, S. Dosing Extracellular Vesicles. Adv. Drug Deliv. Rev. 2021, 178, 11396. [Google Scholar] [CrossRef]
  132. Théry, C.; Amigorena, S.; Raposo, G.; Clayton, A. Isolation and Characterization of Exosomes from Cell Culture Supernatants and Biological Fluids. Curr. Protoc. Cell Biol. 2006, 3, 3.22.1–3.22.29. [Google Scholar] [CrossRef]
  133. Greening, D.W.; Xu, R.; Ji, H.; Tauro, B.J.; Simpson, R.J. A Protocol for Exosome Isolation and Characterization: Evaluation of Ultracentrifugation, Density-Gradient Separation, and Immunoaffinity Capture Methods. Methods Mol. Biol. 2015, 1295, 179–209. [Google Scholar]
  134. Johnson, J.; Law, S.Q.K.; Shojaee, M.; Hall, A.S.; Bhuiyan, S.; Lim, M.B.L.; Silva, A.; Kong, K.J.W.; Schoppet, M.; Blyth, C.; et al. First-in-human Clinical Trial of Allogeneic, Platelet-derived Extracellular Vesicles as a Potential Therapeutic for Delayed Wound Healing. J. Extracell. Vesicle 2023, 12, 12332. [Google Scholar] [CrossRef] [PubMed]
  135. Zhang, M.; Jin, K.; Gao, L.; Zhang, Z.; Li, F.; Zhou, F.; Zhang, L. Methods and technologies for exosome isolation and characterization. Small Methods 2018, 2, 1800021. [Google Scholar] [CrossRef]
  136. Nwokwu, C.D.; Ishraq Bari, S.M.; Hutson, K.H.; Brausell, C.; Nestorova, G.G. ExoPRIME: Solid-Phase Immunoisolation and OMICS Analysis of Surface-Marker-Specific Exosomal Subpopulations. Talanta 2022, 236, 122870. [Google Scholar] [CrossRef]
  137. Doyle, L.M.; Wang, M.Z. Overview of Extracellular Vesicles, Their Origin, Composition, Purpose, and Methods for Exosome Isolation and Analysis. Cells 2019, 8, 727. [Google Scholar] [CrossRef]
  138. Batrakova, E.V.; Kim, M.S. Using Exosomes, Naturally-Equipped Nanocarriers, for Drug Delivery. J. Control. Release 2015, 219, 396–405. [Google Scholar] [CrossRef] [PubMed]
  139. Zaborowski, M.P.; Balaj, L.; Breakefield, X.O.; Lai, C.P. Extracellular Vesicles: Composition, Biological Relevance, and Methods of Study. Bioscience 2015, 65, 783–797. [Google Scholar] [CrossRef]
  140. Zeringer, E.; Barta, T.; Li, M.; Vlassov, A.V. Strategies for Isolation of Exosomes. Cold Spring Harb. Protoc. 2015, 2015, 319–323. [Google Scholar] [CrossRef]
  141. Shin, H.; Han, C.; Labuz, J.M.; Kim, J.; Kim, J.; Cho, S.; Gho, Y.S.; Takayama, S.; Park, J. High-yield isolation of extracellular vesicles using aqueous two-phase system. Sci. Rep. 2015, 5, 13103. [Google Scholar] [CrossRef]
  142. Zarovni, N.; Corrado, A.; Guazzi, P.; Zocco, D.; Lari, E.; Radano, G.; Muhhina, J.; Fondelli, C.; Gavrilova, J.; Chiesi, A. Integrated Isolation and Quantitative Analysis of Exosome Shuttled Proteins and Nucleic Acids Using Immunocapture Approaches. Methods 2015, 87, 46–58. [Google Scholar] [CrossRef]
  143. Li, P.; Kaslan, M.; Lee, S.H.; Yao, J.; Gao, Z. Progress in Exosome Isolation Techniques. Theranostics 2017, 7, 789–804. [Google Scholar] [CrossRef]
  144. Lee, K.; Shao, H.; Weissleder, R.; Lee, H. Acoustic Purification of Extracellular Microvesicles. ACS Nano 2015, 9, 2321–2327. [Google Scholar] [CrossRef]
  145. Zhao, Z.; Yang, Y.; Zeng, Y.; He, M. A Microfluidic ExoSearch Chip for Multiplexed Exosome Detection towards Blood-Based Ovarian Cancer Diagnosis. Lab. Chip 2016, 16, 489–496. [Google Scholar] [CrossRef]
  146. Davies, R.T.; Kim, J.; Jang, S.C.; Choi, E.-J.; Gho, Y.S.; Park, J. Microfluidic Filtration System to Isolate Extracellular Vesicles from Blood. Lab. Chip 2012, 12, 5202–5210. [Google Scholar] [CrossRef]
  147. Bang, O.Y.; Kim, E.H.; Cho, Y.H.; Oh, M.J.; Chung, J.-W.; Chang, W.H.; Kim, Y.-H.; Yang, S.W.; Chopp, M. Circulating Extracellular Vesicles in Stroke Patients Treated With Mesenchymal Stem Cells: A Biomarker Analysis of a Randomized Trial. Stroke 2022, 53, 2276–2286. [Google Scholar] [CrossRef] [PubMed]
  148. Shuen, T.W.H.; Alunni-Fabbroni, M.; Öcal, E.; Malfertheiner, P.; Wildgruber, M.; Schinner, R.; Pech, M.; Benckert, J.; Sangro, B.; Kuhl, C.; et al. Extracellular Vesicles May Predict Response to Radioembolization and Sorafenib Treatment in Advanced Hepatocellular Carcinoma: An Exploratory Analysis from the SORAMIC Trial. Clin. Cancer Res. 2022, 28, 3890–3901. [Google Scholar] [CrossRef] [PubMed]
  149. Agrahari, V.; Agrahari, V.; Burnouf, P.-A.; Chew, C.H.; Burnouf, T. Extracellular Microvesicles as New Industrial Therapeutic Frontiers. Trends Biotechnol. 2019, 37, 707–729. [Google Scholar] [CrossRef] [PubMed]
  150. Yuan, T.; Meijia, L.; Xinyao, C.; Xinyue, C.; Lijun, H. Exosome Derived from Human Adipose-derived Stem Cell Improve Wound Healing Quality: A Systematic Review and Meta-analysis of Preclinical Animal Studies. Int. Wound J. 2023, 20, 2424–2439. [Google Scholar] [CrossRef] [PubMed]
  151. Kalluri, R.; LeBleu, V.S. The Biology, Function, and Biomedical Applications of Exosomes. Science 2020, 367, eaau6977. [Google Scholar] [CrossRef] [PubMed]
  152. Kolonics, F.; Szeifert, V.; Timár, C.I.; Ligeti, E.; Lőrincz, Á.M. The Functional Heterogeneity of Neutrophil-Derived Extracellular Vesicles Reflects the Status of the Parent Cell. Cells 2020, 9, 2718. [Google Scholar] [CrossRef]
  153. Zhang, B.; Yeo, R.W.Y.; Lai, R.C.; Sim, E.W.K.; Chin, K.C.; Lim, S.K. Mesenchymal Stromal Cell Exosome-Enhanced Regulatory T-Cell Production through an Antigen-Presenting Cell-Mediated Pathway. Cytotherapy 2018, 20, 687–696. [Google Scholar] [CrossRef]
  154. Brambilla, D.; Luciani, P.; Leroux, J.-C. Breakthrough Discoveries in Drug Delivery Technologies: The next 30 Years. J. Control. Release 2014, 190, 9–14. [Google Scholar] [CrossRef]
  155. Huang, C.-C.; Kang, M.; Lu, Y.; Shirazi, S.; Diaz, J.I.; Cooper, L.F.; Gajendrareddy, P.; Ravindran, S. Functionally Engineered Extracellular Vesicles Improve Bone Regeneration. Acta Biomater. 2020, 109, 182–194. [Google Scholar] [CrossRef]
  156. Pan, Z.; Sun, W.; Chen, Y.; Tang, H.; Lin, W.; Chen, J.; Chen, C. Extracellular Vesicles in Tissue Engineering: Biology and Engineered Strategy. Adv. Heal. Mater. 2022, 11, e2201384. [Google Scholar] [CrossRef]
  157. Henrich, S.E.; McMahon, K.M.; Plebanek, M.P.; Calvert, A.E.; Feliciano, T.J.; Parrish, S.; Tavora, F.; Mega, A.; De Souza, A.; Carneiro, B.A.; et al. Prostate Cancer Extracellular Vesicles Mediate Intercellular Communication with Bone Marrow Cells and Promote Metastasis in a Cholesterol-Dependent Manner. J. Extracell. Vesicles 2020, 10, e12042. [Google Scholar] [CrossRef]
  158. Lobb, R.J.; Lima, L.G.; Möller, A. Exosomes: Key Mediators of Metastasis and Pre-Metastatic Niche Formation. Semin. Cell Dev. Biol. 2017, 67, 3–10. [Google Scholar] [CrossRef]
  159. Zhang, L.; Yu, D. Exosomes in Cancer Development, Metastasis, and Immunity. Biochim. Biophys. Acta (BBA)—Rev. Cancer 2019, 1871, 455–468. [Google Scholar] [CrossRef] [PubMed]
  160. Berumen Sánchez, G.; Bunn, K.E.; Pua, H.H.; Rafat, M. Extracellular Vesicles: Mediators of Intercellular Communication in Tissue Injury and Disease. Cell Commun. Signal. 2021, 19, 104. [Google Scholar] [CrossRef]
  161. Rahmati, S.; Khazaei, M.; Nadi, A.; Alizadeh, M.; Rezakhani, L. Exosome-loaded scaffolds for regenerative medicine in hard tissues. Tissue Cell 2023, 82, 102102. [Google Scholar] [CrossRef]
  162. Lin, Y.-N.; Mesquita, T.; Sanchez, L.; Chen, Y.-H.; Liu, W.; Li, C.; Rogers, R.; Wang, Y.; Li, X.; Wu, D.; et al. Extracellular Vesicles from Immortalized Cardiosphere-Derived Cells Attenuate Arrhythmogenic Cardiomyopathy in Desmoglein-2 Mutant Mice. Eur. Heart J. 2021, 42, 3558–3571. [Google Scholar] [CrossRef]
  163. Jiang, D.; Xiong, G.; Feng, H.; Zhang, Z.; Chen, P.; Yan, B.; Chen, L.; Gandhervin, K.; Ma, C.; Li, C.; et al. Donation of Mitochondria by iPSC-Derived Mesenchymal Stem Cells Protects Retinal Ganglion Cells against Mitochondrial Complex I Defect-Induced Degeneration. Theranostics 2019, 9, 2395–2410. [Google Scholar] [CrossRef] [PubMed]
  164. Damania, A.; Jaiman, D.; Teotia, A.K.; Kumar, A. Mesenchymal Stromal Cell-Derived Exosome-Rich Fractionated Secretome Confers a Hepatoprotective Effect in Liver Injury. Stem Cell Res. Ther. 2018, 9, 31. [Google Scholar] [CrossRef] [PubMed]
  165. Gonçalves, R.D.J.; Vasques, J.; Da Silva-Junior, A.; Gubert, F.; Mendez-Otero, R. Mesenchymal Stem Cell- and Extracellular Vesicle-Based Therapies for Alzheimer′s Disease: Progress, Advantages, and Challenges. Neural Regen. Res. 2023, 18, 1645–1651. [Google Scholar] [PubMed]
  166. Xu, X.; Liang, Y.; Li, X.; Ouyang, K.; Wang, M.; Cao, T.; Li, W.; Liu, J.; Xiong, J.; Li, B.; et al. Exosome-Mediated Delivery of Kartogenin for Chondrogenesis of Synovial Fluid-Derived Mesenchymal Stem Cells and Cartilage Regeneration. Biomaterials 2021, 269, 120539. [Google Scholar] [CrossRef] [PubMed]
  167. Keshtkar, S.; Azarpira, N.; Ghahremani, M.H. Mesenchymal Stem Cell-Derived Extracellular Vesicles: Novel Frontiers in Regenerative Medicine. Stem Cell Res. Ther. 2018, 9, 63. [Google Scholar] [CrossRef]
  168. Bian, D.; Wu, Y.; Song, G.; Azizi, R.; Zamani, A. The Application of Mesenchymal Stromal Cells (MSCs) and Their Derivative Exosome in Skin Wound Healing: A Comprehensive Review. Stem Cell Res. Ther. 2022, 13, 24. [Google Scholar] [CrossRef]
  169. Zhang, X.; Gan, J.; Fan, L.; Luo, Z.; Zhao, Y. Bioinspired Adaptable Indwelling Microneedles for Treatment of Diabetic Ulcers. Adv. Mater. 2023, 35, 2210903. [Google Scholar] [CrossRef]
  170. Ti, D.; Hao, H.; Tong, C.; Liu, J.; Dong, L.; Zheng, J.; Zhao, Y.; Liu, H.; Fu, X.; Han, W. LPS-Preconditioned Mesenchymal Stromal Cells Modify Macrophage Polarization for Resolution of Chronic Inflammation via Exosome-Shuttled Let-7b. J. Transl. Med. 2015, 13, 308. [Google Scholar] [CrossRef]
  171. Lee, Y.; Ni, J.; Beretov, J.; Wasinger, V.C.; Graham, P.; Li, Y. Recent Advances of Small Extracellular Vesicle Biomarkers in Breast Cancer Diagnosis and Prognosis. Mol. Cancer 2023, 22, 33. [Google Scholar] [CrossRef]
  172. Zhang, Z.; Liu, X.; Yang, X.; Jiang, Y.; Li, A.; Cong, J.; Li, Y.; Xie, Q.; Xu, C.; Liu, D. Identification of Faecal Extracellular Vesicles as Novel Biomarkers for the Non-Invasive Diagnosis and Prognosis of Colorectal Cancer. J. Extracell. Vesicles 2023, 12, e12300. [Google Scholar] [CrossRef]
  173. Bunduc, S.; Gede, N.; Váncsa, S.; Lillik, V.; Kiss, S.; Juhász, M.F.; Erőss, B.; Szakács, Z.; Gheorghe, C.; Mikó, A.; et al. Exosomes as Prognostic Biomarkers in Pancreatic Ductal Adenocarcinoma—A Systematic Review and Meta-Analysis. Transl. Res. 2022, 244, 126–136. [Google Scholar] [CrossRef]
  174. Nanou, A.; Mol, L.; Coumans, F.A.W.; Koopman, M.; Punt, C.J.A.; Terstappen, L.W.M.M. Endothelium-Derived Extracellular Vesicles Associate with Poor Prognosis in Metastatic Colorectal Cancer. Cells 2020, 9, 2688. [Google Scholar] [CrossRef]
  175. Kharaziha, P.; Chioureas, D.; Rutishauser, D.; Baltatzis, G.; Lennartsson, L.; Fonseca, P.; Azimi, A.; Hultenby, K.; Zubarev, R.; Ullén, A.; et al. Molecular Profiling of Prostate Cancer Derived Exosomes May Reveal a Predictive Signature for Response to Docetaxel. Oncotarget 2015, 6, 21740–21754. [Google Scholar] [CrossRef]
  176. Murali, V.P.; Holmes, C.A. Biomaterial-Based Extracellular Vesicle Delivery for Therapeutic Applications. Acta Biomater. 2021, 124, 88–107. [Google Scholar] [CrossRef]
  177. Dinh, P.-U.C.; Paudel, D.; Brochu, H.; Popowski, K.D.; Gracieux, M.C.; Cores, J.; Huang, K.; Hensley, M.T.; Harrell, E.; Vandergriff, A.C.; et al. Inhalation of Lung Spheroid Cell Secretome and Exosomes Promotes Lung Repair in Pulmonary Fibrosis. Nat. Commun. 2020, 11, 1064. [Google Scholar] [CrossRef]
  178. Hu, L.I.; Wang, J.; Zhou, X.; Xiong, Z.; Zhao, J.; Yu, R.; Huang, F.; Zhang, H.; Chen, L. Exosomes Derived from Human Adipose Mensenchymal Stem Cells Accelerates Cutaneous Wound Healing via Optimizing the Characteristics of Fibroblasts. Sci. Rep. 2016, 6, 32993. [Google Scholar] [CrossRef]
  179. Fang, A.; Wang, Y.; Guan, N.; Zuo, Y.; Lin, L.; Guo, B.; Mo, A.; Wu, Y.; Lin, X.; Cai, W.; et al. Porous Microneedle Patch with Sustained Delivery of Extracellular Vesicles Mitigates Severe Spinal Cord Injury. Nat. Commun. 2023, 14, 4011. [Google Scholar] [CrossRef]
  180. Shi, M.; Yang, Q.; Monsel, A.; Yan, J.; Dai, C.; Zhao, J.; Shi, G.; Zhou, M.; Zhu, X.; Li, S.; et al. Preclinical Efficacy and Clinical Safety of Clinical-Grade Nebulized Allogenic Adipose Mesenchymal Stromal Cells-Derived Extracellular Vesicles. J. Extracell. Vesicles 2021, 10, e12134. [Google Scholar] [CrossRef]
  181. Sun, T.; Li, M.; Liu, Q.; Yu, A.; Cheng, K.; Ma, J.; Murphy, S.; McNutt, P.M.; Zhang, Y. Insights into Optimizing Exosome Therapies for Acute Skin Wound Healing and Other Tissue Repair. Front. Med. 2024. [Google Scholar] [CrossRef]
  182. Zhang, S.; Yang, L.; Liu, J.; Li, H.; Hong, S.; Hong, L. Microneedle Systems: Cell, Exosome, and Nucleic Acid Based Strategies. Biomater. Sci. 2023, 11, 7018–7033. [Google Scholar] [CrossRef] [PubMed]
  183. Yuan, J.; Yang, H.; Liu, C.; Shao, L.; Zhang, H.; Lu, K.; Wang, J.; Wang, Y.; Yu, Q.; Zhang, Y.; et al. Microneedle Patch Loaded with Exosomes Containing MicroRNA-29b Prevents Cardiac Fibrosis after Myocardial Infarction. Adv. Healthc. Mater. 2023, 12, 2202959. [Google Scholar] [CrossRef]
  184. Wolf, M.; Poupardin, R.W.; Ebner-Peking, P.; Andrade, A.C.; Blöchl, C.; Obermayer, A.; Gomes, F.G.; Vari, B.; Maeding, N.; Eminger, E.; et al. A Functional Corona around Extracellular Vesicles Enhances Angiogenesis, Skin Regeneration and Immunomodulation. J. Extracell. Vesicles 2022, 11, e12207. [Google Scholar] [CrossRef]
  185. Gan, J.; Zhang, X.; Ma, W.; Zhao, Y.; Sun, L. Antibacterial, Adhesive, and MSC Exosomes Encapsulated Microneedles with Spatio-Temporal Variation Functions for Diabetic Wound Healing. Nano Today 2022, 47, 101630. [Google Scholar] [CrossRef]
  186. Zeng, J.; Sun, Z.; Zeng, F.; Gu, C.; Chen, X. M2 Macrophage-Derived Exosome-Encapsulated Microneedles with Mild Photothermal Therapy for Accelerated Diabetic Wound Healing. Mater. Today Bio 2023, 20, 100649. [Google Scholar] [CrossRef]
  187. Mbituyimana, B.; Adhikari, M.; Qi, F.; Shi, Z.; Fu, L.; Yang, G. Microneedle-Based Cell Delivery and Cell Sampling for Biomedical Applications. J. Control Release 2023, 362, 692–714. [Google Scholar] [CrossRef]
  188. Li, L.; Zhang, Y.; Mu, J.; Chen, J.; Zhang, C.; Cao, H.; Gao, J. Transplantation of Human Mesenchymal Stem-Cell-Derived Exosomes Immobilized in an Adhesive Hydrogel for Effective Treatment of Spinal Cord Injury. Nano Lett. 2020, 20, 4298–4305. [Google Scholar] [CrossRef]
  189. Liu, A.; Wang, Q.; Zhao, Z.; Wu, R.; Wang, M.; Li, J.; Sun, K.; Sun, Z.; Lv, Z.; Xu, J.; et al. Nitric Oxide Nanomotor Driving Exosomes-Loaded Microneedles for Achilles Tendinopathy Healing. ACS Nano 2021, 15, 13339–13350. [Google Scholar] [CrossRef]
  190. Gaurav, I.; Thakur, A.; Iyaswamy, A.; Wang, X.; Chen, X.; Yang, Z. Factors Affecting Extracellular Vesicles Based Drug Delivery Systems. Molecules 2021, 26, 1544. [Google Scholar] [CrossRef]
  191. Yerneni, S.S.; Yalcintas, E.P.; Smith, J.D.; Averick, S.; Campbell, P.G.; Ozdoganlar, O.B. Skin-Targeted Delivery of Extracellular Vesicle-Encapsulated Curcumin Using Dissolvable Microneedle Arrays. Acta Biomater. 2022, 149, 198–212. [Google Scholar] [CrossRef]
  192. Song, K.; Hao, Y.; Tan, X.; Huang, H.; Wang, L.; Zheng, W. Microneedle-mediated delivery of Ziconotide-loaded liposomes fused with exosomes for analgesia. J. Control. Release 2023, 356, 448–462. [Google Scholar] [CrossRef]
  193. 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]
  194. Nazerian, Y.; Nazerian, A.; Mohamadi-Jahani, F.; Sodeifi, P.; Jafarian, M.; Javadi, S.A. Hydrogel-Encapsulated Extracellular Vesicles for the Regeneration of Spinal Cord Injury. Front. Neurosci. 2023, 17, 1309172. [Google Scholar] [CrossRef]
  195. Gondaliya, P.; Sayyed, A.A.; Bhat, P.; Mali, M.; Arya, N.; Khairnar, A.; Kalia, K. Mesenchymal Stem Cell-Derived Exosomes Loaded with miR-155 Inhibitor Ameliorate Diabetic Wound Healing. Mol. Pharm. 2022, 19, 1294–1308. [Google Scholar] [CrossRef]
  196. Yan, L.; Liu, G.; Wu, X. The Umbilical Cord Mesenchymal Stem Cell-Derived Exosomal lncRNA H19 Improves Osteochondral Activity through miR-29b-3p/FoxO3 Axis. Clin. Transl. Med. 2021, 11, e255. [Google Scholar] [CrossRef]
  197. Zhang, Y.; Pan, Y.; Liu, Y.; Li, X.; Tang, L.; Duan, M.; Li, J.; Zhang, G. Exosomes Derived from Human Umbilical Cord Blood Mesenchymal Stem Cells Stimulate Regenerative Wound Healing via Transforming Growth Factor-β Receptor Inhibition. Stem Cell Res. Ther. 2021, 12, 434. [Google Scholar] [CrossRef]
  198. Yan, C.; Chen, J.; Wang, C.; Yuan, M.; Kang, Y.; Wu, Z.; Li, W.; Zhang, G.; Machens, H.-G.; Rinkevich, Y.; et al. Milk Exosomes-Mediated miR-31-5p Delivery Accelerates Diabetic Wound Healing through Promoting Angiogenesis. Drug Deliv. 2022, 29, 214–228. [Google Scholar] [CrossRef]
  199. Ma, Y.; Brocchini, S.; Williams, G.R. Extracellular Vesicle-Embedded Materials. J. Control. Release 2023, 361, 280–296. [Google Scholar] [CrossRef]
  200. Ju, Y.; Hu, Y.; Yang, P.; Xie, X.; Fang, B. Extracellular Vesicle-Loaded Hydrogels for Tissue Repair and Regeneration. Mater. Today Bio 2023, 18, 100522. [Google Scholar] [CrossRef]
  201. Bordbar-Khiabani, A.; Gasik, M. Smart Hydrogels for Advanced Drug Delivery Systems. Int. J. Mol. Sci. 2022, 23, 3665. [Google Scholar] [CrossRef] [PubMed]
  202. Sun, J.; Wu, X.; Xiao, J.; Zhang, Y.; Ding, J.; Jiang, J.; Chen, Z.; Liu, X.; Wei, D.; Zhou, L.; et al. Hydrogel-Integrated Multimodal Response as a Wearable and Implantable Bidirectional Interface for Biosensor and Therapeutic Electrostimulation. ACS Appl. Mater. Interfaces 2023, 15, 5897–5909. [Google Scholar] [CrossRef] [PubMed]
  203. Samant, P.P.; Niedzwiecki, M.M.; Raviele, N.; Tran, V.; Mena-Lapaix, J.; Walker, D.I.; Felner, E.I.; Jones, D.P.; Miller, G.W.; Prausnitz, M.R. Sampling Interstitial Fluid from Human Skin Using a Microneedle Patch. Sci. Transl. Med. 2020, 12, eaaw0285. [Google Scholar] [CrossRef]
  204. Smith, S.M.; Kumari, A.; Reis, T.; Kang, Y.-T.; Nagrath, S. Abstract 3297: Deep Reactive Ion Etched Microneedle Array for in-Vivo Melanoma Cancer Monitoring via Cancer Exosome Isolation. Cancer Res. 2023, 83, 3297. [Google Scholar] [CrossRef]
  205. Yao, S.; Chi, J.; Wang, Y.; Zhao, Y.; Luo, Y.; Wang, Y. Zn-MOF Encapsulated Antibacterial and Degradable Microneedles Array for Promoting Wound Healing. Adv. Healthc. Mater. 2021, 10, 2100056. [Google Scholar] [CrossRef]
  206. Himawan, A.; Vora, L.K.; Permana, A.D.; Sudir, S.; Nurdin, A.R.; Nislawati, R.; Hasyim, R.; Scott, C.J.; Donnelly, R.F. Where Microneedle Meets Biomarkers: Futuristic Application for Diagnosing and Monitoring Localized External Organ Diseases. Adv. Heal. Mater. 2023, 12, e2202066. [Google Scholar] [CrossRef]
Figure 1. (a) MNs deliver drugs for transdermal and non-transdermal applications. (b) Drug diffusion diagram of different types of MNs.
Figure 1. (a) MNs deliver drugs for transdermal and non-transdermal applications. (b) Drug diffusion diagram of different types of MNs.
Pharmaceutics 16 00326 g001
Table 1. Comparison of exosomal isolation techniques.
Table 1. Comparison of exosomal isolation techniques.
MechanismTechniquesExpertiseStarting VolumesTime
Efficiency
PurityCostRefs
size and densityUltracentrifugation (UC)little100 s of mLstime-
consuming
lowultracentrifuge
high initial cost
[135,139]
Ultrafiltration (UF)littleN/Afaster than UCN/Aminimal
equipment
[112]
Size-exclusion chromatographyN/AN/Aseveral hourspurer than UCrequirement
high cost
[106]
polymer
precipitation
Polyethylene
glycol
precipitation
little100 μL to several mLsquicklownot expensive instrumentation[135,140]
Aqueous two-phase systemeasy
procedure
N/Anearly 15 min,
yield 10 to 15 times higher than UC
N/AN/A[141,142]
immunoaffinity captureEnzyme-linked
immunosorbent assay
common
laboratory experiment
100 μllowN/Aexperiment
materials
[137]
Magneto-ImmunoprecipitationN/Ano up
limit
faster than UCpurer than UCnot expensive instrumentation[137,143]
microfluidics-
based methods
Acoustic
nano-filter
minimal50 μLquickN/AN/A[144]
Immuno-based
microfluidic
isolation
minimal10 s–100 s of μLrapidefficientthe most
cost
[145,146]
Table abbreviations: N/A denotes not applicable. Refs denote references.
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

Wang, X.; Cheng, W.; Su, J. Research Progress of Extracellular Vesicles-Loaded Microneedle Technology. Pharmaceutics 2024, 16, 326. https://doi.org/10.3390/pharmaceutics16030326

AMA Style

Wang X, Cheng W, Su J. Research Progress of Extracellular Vesicles-Loaded Microneedle Technology. Pharmaceutics. 2024; 16(3):326. https://doi.org/10.3390/pharmaceutics16030326

Chicago/Turabian Style

Wang, Xue, Wei Cheng, and Jiandong Su. 2024. "Research Progress of Extracellular Vesicles-Loaded Microneedle Technology" Pharmaceutics 16, no. 3: 326. https://doi.org/10.3390/pharmaceutics16030326

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