Next Article in Journal
Mechanisms Controlling MicroRNA Expression in Tumor
Next Article in Special Issue
Mesenchymal Stem Cell-Derived Extracellular Vesicles for Therapeutic Use and in Bioengineering Applications
Previous Article in Journal
Proactive Immunotherapeutic Approaches against Inflammatory Breast Cancer May Improve Patient Outcomes
Previous Article in Special Issue
Exosomes Derived from Adipose Mesenchymal Stem Cells Promote Diabetic Chronic Wound Healing through SIRT3/SOD2
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Biomaterials and Extracellular Vesicle Delivery: Current Status, Applications and Challenges

1
Department of Oral Biology, College of Dentistry, University of Illinois Chicago, Chicago, IL 60612, USA
2
School of Dentistry, Virginia Commonwealth University, Richmond, VA 23298, USA
*
Author to whom correspondence should be addressed.
Cells 2022, 11(18), 2851; https://doi.org/10.3390/cells11182851
Submission received: 19 August 2022 / Revised: 5 September 2022 / Accepted: 7 September 2022 / Published: 13 September 2022
(This article belongs to the Special Issue Mesenchymal Stem Cell-Derived Extracellular Vesicles)

Abstract

:
In this review, we will discuss the current status of extracellular vesicle (EV) delivery via biopolymeric scaffolds for therapeutic applications and the challenges associated with the development of these functionalized scaffolds. EVs are cell-derived membranous structures and are involved in many physiological processes. Naïve and engineered EVs have much therapeutic potential, but proper delivery systems are required to prevent non-specific and off-target effects. Targeted and site-specific delivery using polymeric scaffolds can address these limitations. EV delivery with scaffolds has shown improvements in tissue remodeling, wound healing, bone healing, immunomodulation, and vascular performance. Thus, EV delivery via biopolymeric scaffolds is becoming an increasingly popular approach to tissue engineering. Although there are many types of natural and synthetic biopolymers, the overarching goal for many tissue engineers is to utilize biopolymers to restore defects and function as well as support host regeneration. Functionalizing biopolymers by incorporating EVs works toward this goal. Throughout this review, we will characterize extracellular vesicles, examine various biopolymers as a vehicle for EV delivery for therapeutic purposes, potential mechanisms by which EVs exert their effects, EV delivery for tissue repair and immunomodulation, and the challenges associated with the use of EVs in scaffolds.

1. Introduction

The use of extracellular vesicles (EVs) in lieu of stems cells in scaffolds has become increasingly popular in recent years. Stem cells such as mesenchymal stem cells (MSCs) have immunomodulatory and differentiation effects, however, they have been found to cause abnormal differentiation and tumor formation [1]. MSCs exert their therapeutic functions via their secretome, including EVs [2]. For tissue engineering, EVs are an attractive alternative to stem cell transplantation as they have emerged as important mediators of cellular communication and can directly affect a number of biological processes in target cells [3]. Thus, regenerative research is shifting from the use of stem cells to the use of EVs.
Scaffolds serve as an approach to restore form and function to diseased, damaged, and lost tissue by acting as the ECM that supports the cells and their fate and function [4]. Various natural and synthetic biopolymers can be used to create such scaffolds. Incorporating EVs into the synthesis of scaffolds provides a system that supports host regeneration through structural and physiological means. In this review, we discuss EV integration into different types of biopolymers for a variety of therapeutic applications. Additionally, we briefly highlight the potential of using EV-functionalized scaffolds for tissue repair in different organs systems as well as the possible roles EVs play in immunomodulation. Finally, we examine the challenges in EV loading, integrity, delivery, and scaling up.

2. Extracellular Vesicles (EVs)

Extracellular vesicles are nanosized cell-derived membranous structures and can be categorized into several subclasses including exosomes (40–160 nm), microvesicles (150–1000 nm), and apoptotic bodies (>1000 nm) [5] (Figure 1). Exosomes are formed by the inward budding of endosomes and results in the generation of intraluminal vesicles within multivesicular bodies (MVBs) [6]. When MVBs fuse with the plasma membrane, the intraluminal vesicles are released into the extracellular space and are then referred to as exosomes [6]. It is unlikely that researchers will be able to capture live images of EV release in order to assign EVs to subclasses [7]. Thus, the authors often refer to EV subclasses based on the physical characteristics (e.g., small EVs (sEVs)), biochemical composition (e.g., Annexin A5-stained EVs), conditions (e.g., hypoxic EVs), or cell of origin (e.g., hMSC EVs) [7]. Ultimately, it is important to continue studying specific and reliable markers of EV subtypes, so that a consensus regarding nomenclature can be established.
EV isolation poses several challenges. For instance, the complete isolation of EVs from other entities, such as proteins and RNA granules, is unlikely [7,8]. Furthermore, there is no consensus on the best method for EV isolation as well as to how pure an EV preparation should be, as this depends on the experimental question and the purpose of the EVs [7,9]. For example, therapeutic applications in which function is most important may not require highly purified EVs, whereas attributing a biomarker to vesicles will likely require highly purified EVs [7]. Nevertheless, researchers are able to isolate EVs from other non-EV components as well as different types of EVs from each other to various degrees using different available techniques such as ultracentrifugation and size-exclusion chromatography [7,10,11,12,13].
A major issue regarding EV characterization is that a relatively high number (17%) of published EV-related articles do not provide EV characterization, and about half of the EV-related articles (55%) only used an antibody-based assay to detect EV proteins [14]. EV characterization is necessary to assess different isolation methods and establish that biomarkers or functions are associated with EVs rather than other co-isolated substances [7]. Examples of EV characterization and quality-control methods include particle tracking, Western blotting, electron microscopy, RNA profiling, and advanced cytometry [15,16]. Thus, it is crucial that the authors include EV characterization in their studies, whether that be by providing EV quantification, protein composition, single vesicle analysis, topology of EV-associated components, or other means [7].
EVs play roles in many physiological and pathological processes [5,17] including suppressing inflammatory responses [18,19], modulating cellular function and regenerating tissue injuries [20,21], and modulating the immune system [22]. Once endocytosed by target cells, EVs trigger a cellular response designated by the parental cell to the target cell [20]. In other words, the roles that EVs fulfill depend on their origin [23], and when taken up by target cells, EVs release their contents to enact changes in gene expression (Figure 1) [24]. The cargo EVs carry include proteins, lipids, and genetic material (e.g., miRNA) [23]. Studies have investigated specific EV cargoes that are related to positive therapeutic outcomes. These studies will be discussed further in the following sections. Ultimately, EV contents allow for the regulation of phenotype, function, and immune cell homing, highlighting the potential benefits of using EVs in bioengineering [25].

3. Scaffolds as a Solution for EV Delivery

Despite the therapeutic potential of EVs, they have short-lived effects when administered alone. It has been shown that systemically administered EVs have a short half-life and are quickly cleared from the body, making it difficult to realize the long-term effects of EV treatment [26,27]. As a result, the therapeutic effect of EVs is minimal. One solution to this issue is the utilization of scaffolds for the localized delivery and controlled release of EVs. Scaffolds can contain EVs and deliver them to sites of interest over time, thus optimizing the EV therapeutic potential. Furthermore, scaffolds provide a proper environment for tissue regeneration by providing space for host cell migration, mechanical support and integrity, and even cell signaling via bioactive molecules (e.g., EVs). Physiologically active EVs interact with surrounding ECM molecules [28]. Thus, it is important that scaffolds mimic the ECM complex to support active-EV delivery. By applying EV-laden scaffolds to injury sites, the EVs in the scaffold are protected and released from the scaffold in a sustained manner [29]. Released EVs can communicate with endogenous cells and extracellular components, which allows them to play a role in therapeutic effects such as the remodeling process [29]. Overall, scaffolds bridge the therapeutic benefits of extracellular vesicles and clinical application by creating an appropriate environment for EV delivery.

4. Biopolymers as Delivery Systems

There are two main types of biopolymers used in bioengineering: natural and synthetic biopolymers (Figure 1). Natural biomacromolecules that have been studied include silk fibroin, collagen, gelatin, chitosan, and hyaluronic acid. Widely studied synthetic biopolymers for scaffolds include polyethylene glycol (PEG), polycaprolactone (PCL), poly(lactic-co-glycolic acid) (PLGA), and poly(L-lactide) (PLLA). Each type of biomaterial comes with benefits and challenges. For example, natural biomaterials are inherently variable depending on the biological source, may have unwanted biological residues, and there may be issues concerning mechanical stability [30]. Meanwhile, synthetic biomaterials lack native tissue topography and structure that is more typical of natural biomaterials and may contribute to a toxic environment due to chemical crosslinking [31]. In general, immunogenicity, limited mechanical properties, and the lack of consistency are always areas of concern when it comes to engineering scaffolds [32]. Additionally, issues that may arise with the use of cell-based technology, such as EVs, include regulatory hurdles in terms of safety and clinical translation (e.g., mutated cell DNA and off-the-shelf-storage, respectively) [33]. Nevertheless, biopolymer-based scaffolds remain one of the most widely used methods for the delivery of biological signals, especially in the field of tissue engineering. In the following sections, we discuss the natural and synthetic biopolymers frequently used for scaffold fabrication and the therapeutic effects of incorporating EVs into these scaffolds.

5. Natural Biopolymer Scaffolds for Therapeutic EV Delivery

Natural biopolymers such as alginate, silk fibroin, collagen, gelatin, and chitosan have numerous advantages as scaffold materials. They all mimic the natural ECM and are also biocompatible, biodegradable, and cause fewer immunogenic reactions [34,35,36]. Furthermore, these natural biopolymers can promote cell adhesion, migration, and differentiation function, as well as produce non-cytotoxic degradation products that can be removed via metabolic pathways in vivo [34]. Thus, natural biopolymers offer select advantages for engineering scaffolds for therapeutic applications. Enhancing the natural biopolymer scaffolds with EVs has shown positive therapeutic effects regarding a myriad of tissues and diseases (Table 1). Below, we highlight examples of factors that have been delivered with commonly studied natural biopolymers as well as several studies that utilize EV-laden natural biopolymer scaffolds for various therapies.

5.1. Sodium Alginate

Sodium alginate (SA) is a linear polysaccharide derived from brown seaweed [55]. More specifically, SA is a derivative of alginic acid and is composed of α-1-guluornic (G) and 1,4-linked-β-D-mannuronic (M) monomers [55]. Alginate has been used to deliver various classes of drugs including NSAIDs [56], chemotherapeutics [57,58], and anesthetics [59]. Hormones such as insulin [60] and salmon calcitonin [61] have also been delivered via alginate. Furthermore, neuropeptides [62], genetic material [63], and probiotics [64] have been delivered using alginate.
EV-laden alginate-based hydrogels have been studied for a wide range of therapeutic applications including diabetic wound healing [37], peripheral nerve regeneration [38], and myocardial infarction (MI) [26]. Lv et al. delivered bone marrow mesenchymal stem cell (MSC)-derived small EVs (sEVs) to the heart using a natural sodium alginate hydrogel as a therapy for myocardial infarction. The hydrogels were embedded with sEVs by simply mixing the sEV solution with the sodium alginate solution prior to hydrogel formation with calcium chloride solution [26]. Additionally, the authors found that hydrogels formed with 0.5% or 1% calcium chloride solutions resulted in nearly all of the sEVs being released by day 10 with a quick burst of sEV release during the first couple of days compared to a 2% calcium chloride solution [26].
Lv et al. reasoned that hydrogels with a quicker sEV release profile would better suit a myocardial infarction model [26]. The authors labeled sEVs and found through ex vivo imaging that sEVs embedded in the hydrogel (sEV-gel) were retained in the heart compared to freely injected sEVs [26]. Lv et al. examined the expression levels of miRNAs (miR) related to anti-apoptosis and pro-angiogenesis and performed TUNEL staining; they found that treatment using the sEV-gel showed decreased cardiac cell apoptosis [26]. Additionally, Lv et al. looked at the number of CD68+ macrophages as well as the ratio of CD206+ to CD68+ macrophages and found that the sEV-gel treatment promoted M1 to M2 macrophage polarization only a couple days after myocardial infarction [26]. Furthermore, Lv et al. used CD31+ staining, α-SMA staining, and Western blotting to show that sEV-gel treatment promoted angiogenesis [26]. Using echocardiography and histology, Lv et al. saw that the sEV-gel treatment resulted in improved cardiac function and enhanced scar thickness compared to only using sEVs [26].
Overall, the authors demonstrated that the sEV-gel treatment promoted angiogenesis, reduced cardiac apoptosis and fibrosis, and improved cardiac function after MI [26]. This study highlights the importance of using a scaffold to allow for an appropriate, efficient, and locally concentrated delivery of EVs, for without some type of retention system, administered EVs will be quickly cleared by the body and will have a minimal therapeutic effect.

5.2. Silk Fibroin

Silk fibroin (SF) is a hydrophobic protein from the Bombyx mori silkworm that self-assembles into strong and resilient materials [65,66]. Silk fibroin on its own is biocompatible, has controllable biodegradability, and has tunable mechanical properties [67]. It also causes minimal inflammation of host tissue, is low-cost, and easy to use [67]. Anti-proliferative [68], anti-inflammatory [69,70], anabolic [70], and anti-retroviral drugs [71] have been delivered using silk fibroin. Additional drugs include anti-inflammatory compounds such as curcumin [72,73] and chemotherapeutics [74,75]. Aside from drug delivery, SF has been utilized for the delivery of antibodies [76], proteins [76,77], hormones (e.g., insulin) [78], genetic material [79], and cells (e.g., mesenchymal stem cells) [80].
Cunnane et al. examined the effect of human adipose-derived mesenchymal stem cell EVs (hADMSC EVs) on vascular cells in vitro [33]. They found that the application of these EVs on smooth muscle cells and endothelial cells increased proliferation as well as migration in a dose-dependent manner [33]. Cunnane et al. then vacuum-seeded EVs into porous silk-based tubular scaffolds by turning mounted scaffolds within a vacuum chamber and infusing the scaffolds with an EV isolate. Using a micro bicinchoninic acid protein assay and fluorescent imaging, they found that this method of seeding retained a greater amount of protein and increased EV coverage, respectively, within the scaffold compared to the soak-loading method [33].
Cunnane et al. implanted the silk-based scaffolds into rat aortas to study the remodeling capacity of the EV-doped scaffolds. After 8 weeks, the explants were stained for cell, collagen, and elastin distribution [33]. Additionally, elastin and collagen content assays were performed to quantify protein deposition within each explant [33]. Their in vivo findings showed that the inclusion of EVs in the scaffold wall improved patency and matrix deposition, including more elastin and collagen production, which is crucial for neo-tissue formation [33]. This study demonstrates that EVs play an effective bioinstructive role when incorporated into and delivered by SF-based vascular grafts.

5.3. Chitosan

Chitosan is a cationic polysaccharide derived from chitin and is made up of diglucose amine and N-acetyl glucose amine groups [81,82,83]. Antibiotics [81,83], antivirals [84], and immunosuppressants [85] have been delivered using chitosan. Additionally, chitosan has been utilized to deliver insulin [82] and genetic material [86,87,88].
Chitosan-based scaffolds have been used to deliver EVs to improve bone defect repair [41], corneal diseases [42], skin wound healing [32], and articular cartilage injuries [43]. Wu et al. developed chitosan-based thermosensitive hydrogels laden with bone mesenchymal stem cell (BMSC)-derived sEVs to accelerate osteogenesis and angiogenesis. After isolating the sEVs, Wu et al. characterized the sEVs through analyzing the size distribution and morphology of the sEVs as well as through Western blotting to detect sEV-specific surface markers [41]. The authors also ensured that the sEVs could be internalized by BMSCs and HUVECs [41]. In vitro experimentation included examining the ALP activity of BMSCs when exposed to sEVs. The results indicated the early-stage osteogenic differentiation of BMSCs when exposed to sEVs, shown by elevated ALP activity in the sEV groups [41]. Furthermore, Wu et al. found through examining the mRNA (OCN, OPN, and Runx2) and protein (OCN, OPN, and RUNX2) levels that BMSC-sEVs can upregulate osteogenic gene expression [41]. The authors also studied the migratory capability of HUVECs exposed to sEVs and found that the proliferation of HUVECs exposed to sEVs increased compared to the control group [41]. Furthermore, mRNA and protein levels relevant to angiogenesis increased in cells exposed to sEVs [41]. Wu et al. added β-glycerphosphate to chitosan to formulate a thermosensitive injectable hydrogel, and they found that sEVs embedded in this hydrogel showed a good slow-release performance of 80% sEV release on day 8, with a slowed release rate thereafter [41].
The in vivo micro-CT results of a calvarial defect model showed that hydrogels embedded with sEVs resulted in a greater area of newly formed bone compared to other groups [41]. Histological staining showed that there was newly formed bone in the sEV-hydrogel and hydrogel-only groups compared to the control group, which resulted in a defect mainly filled with fibrotic connective tissue [41]. Immunohistochemical staining of bone defect sections indicated more CD31+ in the sEV-hydrogel group compared to the hydrogel-only group, which indicated new vessel formation within the bone defect [41]. Overall, the in vivo experimentation demonstrated that sEVs promote calvarial defect repair and enhanced osteogenesis and angiogenesis [41].
Wu et al. examined the potential cause of their observed results by studying the relationship between miR-21 and SPRY2. After performing the reporter assays and rescue experiments, they found that the angiogenic protein levels in cells transfected with miR-21 mimics were higher than the control cells [41]. These experiments indicated that exosomal miR-21 may promote HUVEC migration and angiogenesis by targeting SPRY2 [41].
Overall, Wu et al. successfully developed a thermosensitive injectable chitosan-based hydrogel laden with BMSC-derived sEVs. The hydrogel promoted bone healing and served as a scaffold for sEVs [41]. The sEV-loaded hydrogel promoted bone healing in vivo by enhancing angiogenesis, which may be mediated by miR-21 expression upregulation in sEVs and the regulation of SPRY2 by miR-21 [41]. This study highlights the potential of delivering EVs via scaffolds to promote bone regeneration and the importance of understanding the mechanism behind positive results to continue improving the therapeutic outcomes.

5.4. Collagen

Collagen is the most abundant protein in mammals [89] and is composed of three intertwined α-chains [90]. Some of the functions of collagen include cell adhesion and migration, tissue repair, and scaffolding [91]. Collagen has proven to be useful in medical applications as a delivery tool. Similar to the natural biopolymers discussed thus far, collagen has been utilized to deliver drugs [92,93], cells [94,95], and bioactive substances with antioxidant properties [96]. It is also possible to deliver growth factors using collagen [97,98].
Collagen-based scaffolds have been utilized for therapeutic purposes including bone regeneration [39] and endometrium regeneration [40]. Xin et al. designed a collagen scaffold containing umbilical cord-derived mesenchymal stem cell (UC-MSC)-derived exosomes for endometrial regeneration in a rat endometrium-damage model [40]. The authors confirmed successful exosome extraction through TEM, NTA, and Western blot [40]. Xin et al. added exosome suspension dropwise to the collagen scaffold, and they obtained a sustained release profile with a majority of exosomes being released within 14 days [40].
Xin et al. examined short-term and long-term outcomes from a rat endometrium-damage model involving exosome-collagen, collagen-only, exosome-only, and no treatment (control) groups [40]. Through H&E staining, Xin et al. found that transplantation of the exosome-collagen scaffold promoted endometrium regeneration and glandular reconstruction, which was related to rapid cell proliferation and re-epithelialization [40]. Additionally, Xin et al. performed immunostaining and Masson’s trichrome staining and found that transplantation of the exosome-collagen resulted in excellent neovascularization, reduced fibrosis formation, and promoted collagen remodeling [40]. Through immunohistochemical staining and subsequent software analysis, Xin et al. found that a high number of anti-estrogen receptor α (ERα) positive cells and anti-progesterone receptor (PR) positive cells were present 30 days after exosome-collagen treatment, which suggests the rapid functional recovery of the regenerated endometrium [40]. Furthermore, Xin et al. showed through Evans blue staining that the implantation of the exosome-collagen scaffold resulted in the structural and functional reconstruction of endometrium that could support implantation and the development of embryos in vivo [40].
Xin et al. investigated the potential mechanisms behind the promising outcomes observed with exosome-collagen treatment. The authors found that exosome-collagen treatment promoted the macrophage infiltration within 7 days, with high numbers of M1 macrophages in the exosome-collagen and collagen-only groups within 3 days, likely due to the immunogenicity of the implanted collagen scaffold [40]. Additionally, the exosome-collagen group showed the highest numbers of M2 macrophages within 7 days, indicating that exosome incorporation may have induced the polarization of macrophages to the M2 phenotype [40]. Furthermore, Xin et al. examined the M1 and M2 macrophage-related cytokines. When looking at the M1-related cytokines, they found that the inclusion of exosomes in the scaffold eased inflammation due to the foreign body reaction to collagen within 7 days [40]. Furthermore, there was an enhanced expression of M2-related cytokines with exosome-collagen transplantation. Altogether, transplantation of the exosome-collagen scaffold promoted M1 macrophage infiltration during the early stages of wound healing and induced macrophage transition from an M1 to a M2 phenotype during a later stage of healing [40].
Through analyzing their RNA-Seq data against a publicly-available database, Xin et al. identified a top candidate related to macrophage immunomodulation (miR-223-3p), which has been reported to promote macrophage polarization to an M2 phenotype, as the key cargo within exosomes [40]. Xin et al. believe that miR-223-3p may target Stmn1 within macrophages to confer functional benefits [40]. Overall, Xin et al. developed a local exosome chitosan-based delivery system that could promote endometrium regeneration and fertility restoration [40]. They found that this system involved a mechanism-of-action related to M2 macrophage polarization with miRNA-223-3p serving as a top key component within exosomes [40].

5.5. Hyaluronic Acid

Hyaluronic acid (HA) is a linear polysaccharide with repeating units of D-glucuronic acid and N-acetyl-D-glucosamine [90]. HA is characterized by a strong water binding ability and is found in the extracellular matrix [90]. HA has been used to deliver anti-bacterial drugs [99], immunosuppressants [100], anabolic and anti-inflammatory compounds [70,101,102,103,104]. The delivery of chemotherapeutics using hyaluronic acid have been well-studied [105,106,107,108]. Examples of these delivered therapeutics include doxorubicin [109,110,111,112], cisplatin [113], and cantharidin [114]. Additionally, anti-oxidants [115], flavonols [116], anti-photoaging agents [117], and peptides [118] have been delivered using HA. Furthermore, HA has been used to deliver cells (including cell secretome) [119,120], genetic material [112], growth factors [102,121], and hormones [122].
Hyaluronic acid-based scaffolds have been used to deliver EVs as a therapy for tendon repair [45] and osteoarthritis cartilage injuries [46]. K. Song et al. isolated exosomes from tendon derived stem cells (TDSC-Exos), loaded a hyaluronic acid scaffold with the exosomes, and studied the therapeutic effects of this system for tendon repair. The authors examined exosome size distribution, morphology, and the presence of exosome-related markers (CD9, CD63, CD81, and TSG101) using Western blot [45]. They also used the protein concentration of exosomes as a representation of exosome concentration throughout the study [45]. The authors then studied the effects of TDSC-Exos on the proliferation and function of tenocytes in vitro and found that high concentrations of exosomes (100 μg/mL) could protect tenocytes from oxidative stress and serum deprivation [45]. Additionally, the treatment of tenocytes with TDSC-Exos increased type 1 collagen production and elevated tendon-specific marker (Scx, Col1a1, and Dcn) expression [45]. The authors then produced a scaffold by irradiation, containing a uniform exosome distribution that could serve as a sustained exosome-release system (50% of exosomes retained after 14 days) [45].
Song et al. then created a rat tendon defect model and used the hyaluronic scaffold (pHA) or the hyaluronic scaffold containing exosomes (pHA-TDSC-Exos) to fill the gap in the patellar tendons [45]. Not only did they see decreased wound visibility with the pHA-TDSC-Exos group, but the authors found through H&E staining that the wound healing outcomes of this group were significantly better than the pHA and control groups and could enhance tendon repair in this model [45]. Masson trichrome staining showed better collagen fiber arrangement with the pHA-TDSC-Exos group, and immunohistochemistry was used to show that this group promoted the early repair of the injured tendon with earlier type III collagen presence and reduction during the tendon healing process compared to the other groups [45]. Furthermore, pHA-TDSC-Exos facilitated the restoration of the biomechanical properties of the injured tendon [45].
With increasing evidence of the important roles miRNAs play during tissue repair, Song et al. performed RNA sequencing to find miRNAs that were expressed significantly higher in TDSC-Exos compared to tenocytes and chose to focus on miR-144-3p [45]. Song et al. showed that miR-144-3p enhanced cell proliferation and migration. They found that miR-144-3p from TDSC-Exos played an important role in tendon repair by targeting ARID1A in tenocytes [45]. ARID1A is a key component of the switch/sucrose non-fermentable ATP-dependent chromatin-remodeling complex, which plays a critical role in cell cycle modulation [45]. MiR-144-3p is only one potential mechanism by which TDSC-Exos may exert therapeutic effects [45]. Overall, Song et al. showed that TDSC-derived exosomes could promote tendon repair and that miR-144-3p transferred from these exosomes enhanced tenocyte proliferation and migration by targeting ARID1A.

5.6. Gelatin

Gelatin is a widely used natural biopolymer in regenerative medicine and tissue engineering [123]. Similar to the natural biopolymers discussed above, gelatin has been utilized to deliver various types of drugs including anti-fungal/anti-yeast drugs [124], anti-inflammatory agents [125,126,127], antibiotics [128,129,130], and chemotherapeutics [131,132,133,134]. Other factors such as probiotics [135], vitamins [136], peptides [137], and ions [138,139] have been delivered using gelatin. Furthermore, researchers have used gelatin to deliver cells [140,141,142], signaling molecules [143,144], and genetic material [145,146].
Gelatin-based scaffolds have been utilized for bone regenerative purposes [44]. Man et al. epigenetically enhanced osteoblast-derived EVs with the histone deacetylase inhibitor Trichostatin A (TSA) to promote EV osteoinductive potency [44]. In this study, Man et al. used gelatin methacryloyl (GelMA) functionalized with synthetic nanoclay laponite (LAP) (GelMA-LAP), which binds, stabilizes, and improves biofactor retention [44]. They characterized these enhanced EVs (TSA-EVs) using TEM imaging, nano-flow cytometry, single-particle phenotyping, measuring EV protein content, and EV release kinetics from the GelMA-LAP hydrogel [44]. The authors also found that the TSA-EVs released from the GelMA-LAP hydrogel were internalized by hBMSCs, and these EVs promoted hBMSC proliferation and migration [44]. Furthermore, the authors found that these released TSA-EVs enhanced histone acetylation and mineralization of hBMSCs [44]. Man et al. went further to evaluate the effects of TSA-EVs on hBMSC extracellular matrix mineralization within the GelMA-LAP hydrogels by assessing the ALP activity, collagen production, and calcium deposition [44]. There was enhanced ALP activity in the hBMSCs within the TSA-EV hydrogels compared to the untreated osteoblast-EVs (MO-EV) and EV-free groups [44]. hBMSC collagen production within the hydrogel was evaluated through picrosirius red staining, and TSA-EV treatment resulted in the greatest collagen content with a high dosage of TSA-EVs (50 µg/mL) (TSA-EV-50), resulting in the greatest collagen production [44]. Furthermore, TSA-EV-50 gels resulted in greater calcium deposition compared to other groups, as found through alizarin red staining [44].
Man et al. believe that the osteoinductive effect of TSA-EVs within the GelMA-LAP hydrogel is due to the 3D matrix (rather than a 2D matrix). More specifically, the 3D matrix elicits an altered cellular response to chemical and physical stimulation, so hBMSCs within the GelMA-LAP hydrogels may be more receptive to osteoinductive stimulation, which is induced by TSA-EVs compared to hBMSCs in 2D culture [44]. Additionally, the 3D microenvironment of the GelMA-LAP hydrogel may have altered the epigenetic landscape of encapsulated hBMSCs, which may have primed the cells with enhanced differentiation capacity compared to the 2D cultured cells [44]. Finally, the 3D microenvironment of the hydrogel and ECM produced by the hBMSCs likely influenced the sequestering of bioactive factors (e.g., EVs) within the secretome, which would further facilitate mineralization within the GelMA-LAP hydrogel [44]. Altogether, these findings demonstrate the improved therapeutic potential of epigenetically enhanced EVs delivered via a gelatin-based scaffold for bone regeneration.

5.7. Natural Biopolymer Composite Scaffolds

To help overcome issues regarding the mechanical properties of scaffolds, researchers have combined biopolymers to create composite scaffolds. Collagen plays a structural support role in wound healing and also controls cellular functions such as cell shape and differentiation, migration, and some protein synthesis [32]. However, collagen typically has poor mechanical performance, in that it has low strength, and is degraded within days at body temperature [32,147]. Abolgheit et al. used a chitosan-collagen scaffold seeded with bone marrow-derived mesenchymal stem cells (BM-MSCs) or the EVs secreted by BM-MSCs and examined skin wound healing. By cross-linking chitosan into the scaffold, Abolgheit et al. were able to obtain a scaffold with mechanical properties, such as pore size and distribution, similar to that of soft tissues [32]. Furthermore, chitosan has bactericidal properties, is biocompatible, cytocompatible, and is cost-effective [32].
Abolgheit et al. seeded BM-MSC EVs into collagen-chitosan scaffolds, however, the exact method of EV loading was not described, and EV loading efficiency was not examined. Furthermore, they did not examine EV functionality in vitro in this study. Nevertheless, they demonstrated that their scaffolds laden with MSCs or EVs showed enhanced macrophage count [32]. Furthermore, greater amounts of collagen deposition with better alignment was observed with scaffolds containing EVs compared to scaffolds with MSCs, demonstrating the advantages of using EVs rather than stem cells [32]. Abolgheit et al. found that it was the scaffold itself that was responsible for accelerated wound healing while the addition of EVs to the scaffold positively impacted the quality of wound healing. This study emphasizes the great effect that EVs may have on remodeling and wound healing, especially when incorporated into a natural biopolymer-based scaffold.
In this section, we highlighted the therapeutic advantages of incorporating EVs into natural biopolymer-based scaffolds. Some of the demonstrated advantages of EV use include improved outcomes after myocardial infarction, bone healing, and wound healing. Challenges associated with natural biopolymers such as a lack of mechanical support and reproducibility may be addressed through the production of composite scaffolds. It is clear that natural scaffolds embedded with EVs are a promising strategy for a multitude of therapeutic applications.

6. Synthetic Biopolymer Scaffolds for Therapeutic EV Delivery

The use of synthetic biomaterials addresses concerns that are associated with natural biomaterials such as inconsistent starting material and sterility. However, synthetic biopolymer-based scaffolds come with their own disadvantages. The mechanical properties of synthetic biopolymers may differ from natural biopolymers or tissues in terms of stiffness and elasticity, and they may also lack biocompatibility and biochemical cues (e.g., protein motifs and release of bioactive peptides) that are characteristic of natural biopolymers [148]. Despite these disadvantages, synthetic biopolymers can also be modified to reach various therapeutic goals. More specifically, the use of synthetic biopolymers allows for greater control over characteristics of the material including the chemistry, charge, and stiffness [149]. Researchers have studied a wide array of synthetic biopolymers including polyethylene glycol (PEG), polycaprolactone (PCL), poly(lactic-co-glycolic acid) (PLGA), poly(L-lactide) (PLLA), and poly(lactic acid) (PLA). Some of the benefits of utilizing synthetic biopolymers is increased batch-to-batch consistency and greater control over the physical and chemical properties of the material. Various synthetic biopolymer-based scaffolds have been utilized for EV delivery to improve healing and disease states (Table 1). Here, we discuss these therapeutic applications of EV-laden synthetic scaffolds in addition to factors that have been delivered using common synthetic biopolymers.

6.1. Polyethylene Glycol (PEG)

PEG is an FDA-approved, hydrophilic, and flexible polymer that has been proven to be safe for use in biomedical applications [150]. PEG has been used to deliver chemotherapeutics [150,151,152,153,154,155,156,157,158,159], anti-inflammatory drugs [160], antibiotics [161,162], and bisphosphonates [163]. Additionally, vitamins [164], phenols [165], and hormones [166] have been delivered using PEG. Furthermore, PEG has been utilized for cell [167], signaling molecule [143], and genetic material delivery [168,169,170].
Exosomes have been delivered via PEG hydrogels for cutaneous wound healing [47,171]. Additionally, macrophages adopt polarization states in response to the local microenvironment [47]. Based on Kwak et al.’s miRNA-sequencing data, exosomes derived from M1 macrophages (M1-Exos) and M2 macrophages (M2-Exos) contain proteins and miRNAs that are capable of shifting macrophage polarity [47]. Thus, Kwak et al. utilized M2-Exos to induce the reprogramming of nearby proinflammatory M1 macrophages toward an anti-inflammatory M2 phenotype [47]. They then encapsulated the M2-Exos in hydrolytically degradable PEG hydrogels (M2-Exogel) and found that the degradation time was adjustable from 6 to 27 days through controlling the crosslinking density and tightness [47].
Kwak et al. used a full-thickness excisional wound model to assess the therapeutic effects of the M2-Exogel in vivo. They treated the wounds with saline, hydrogel alone, free exosomes, or M2-Exogel. Using immunohistochemistry and cytokine expression analyses, Kwak et al. showed the successful local transition of M1 macrophages to M2 macrophages within the lesion for more than 6 days [47]. The M2-Exogel served as a long-term supply of the critical concentration of exosomes needed to initiate and sustain the reprogramming of M1 to M2 macrophages, ultimately contributing to improved wound healing [47]. Kwak et al. found that localizing M2-Exos in the hydrogel led to rapid wound closure and increased healing quality compared to other groups [47]. More specifically, the wound size significantly decreased after day 8 in the M2-Exogel group compared to other groups [47]. Furthermore, they stained closed wound tissues with Masson’s trichrome and saw that the M2-Exogels produced superior results in the stable closure of full-thickness skin wounds as well as enhanced the dermal adipogenesis and hair follicle regeneration compared to freely injected exosomes [47]. Altogether, the PEG hydrogel-based exosome delivery system serves as a method to locally regulate the polarization state of macrophages, which is critical for tissue homeostasis and proper wound repair [47].

6.2. Polycaprolactone (PCL)

PCL is a linear, hydrophobic, aliphatic polyester with high mechanical strength and is biocompatible as well as biodegradable [172,173]. Chemotherapeutics [154,157,159,174,175,176,177,178,179], antimicrobials [162,180,181], and anti-inflammatory drugs [160,182] have been delivered using PCL. Furthermore, PCL has been used for the delivery of hypotensive agents [183], protease inhibitors [184], anticonvulsants [172], and sulfonylureas [185]. In addition to drug delivery, PCL has been used to deliver polyphenols [186], antiretrovirals, and hormones [187]. Similar to the previously discussed biopolymers, the delivery of genetic material [169,188,189], growth factors [190], and cells [191] using PCL has been studied.
Synthetic biopolymer-based scaffolds containing EVs have been used to improve vascular performance and functionality (Table 1). Wei et al. were interested in using heparin-functionalized vascular PCL grafts to enhance anti-thrombogenicity. The authors fabricated the tubular PCL grafts using electrospinning, modified the grafts with heparin, and loaded MSC-derived small EVs (MSC-sEVs) by soaking the scaffolds in sEV solution [49]. The authors observed a uniform distribution of sEVs on the grafts using a confocal laser scanning microscope [49].
Wei et al. examined the in vivo stability of the grafts by in vivo imaging using labeled sEVs and found that the bioluminescence intensities were higher in the heparinized scaffold group [49]. The authors then studied the performance of the PCL vascular grafts modified with heparin and loaded with sEVs in a hyperlipidemia rat model [49]. sEVs play many roles in this model. Wei et al. found that heparin enhanced anti-thrombogenicity while the addition of immunomodulatory sEVs inhibited thrombosis and calcification, which therefore improved the patency of the graft [49]. The patency rate was measured using color doppler ultrasound, and H&E and von Kossa staining was used for calcification detection [49]. Additionally, bioactive molecules (e.g., VEGF and miRNA 126) from the MSC-sEVs enhanced endothelium and vascular smooth muscle regeneration, shown through H&E staining and immunofluorescence staining with the CD31 antibody, α-SMA antibody, and myosin heavy chain [49]. Furthermore, flow cytometric analysis revealed that sEVs induced polarization from pro-inflammatory and atherogenic M1 macrophages to anti-inflammatory and anti-osteogenic M2c macrophages [49]. This study further highlights how synthetic biomaterials can be enhanced using EVs to create translational scaffolds for regenerative medicine.

6.3. Poly(Lactic-Co-Glycolic Acid) (PLGA)

PLGA is a copolymer that is similar to PCL in that it is a biocompatible, biodegradable, and flexible biopolymer [192,193]. The delivery of antibiotics [194,195,196,197,198] and chemotherapeutics [199,200,201,202,203,204] using PLGA has been well-studied. Dopamine agonists [205], anticonvulsants [206], statins [207], and immunosuppressants [208] have also been delivered using PLGA. Furthermore, growth factors [209], various proteins [210,211,212], hormones [213,214], genetic material [215], and vaccines [216] have been delivered using PLGA.
PLGA scaffolds containing EVs have been studied to improve bone defects [52] and chronic kidney disease [51]. Ko et al. designed a PLGA-based scaffold to deliver stem cell-derived EVs for kidney regeneration. The composite scaffold was composed of PLGA, magnesium hydroxide, and decellularized porcine kidney extracellular matrix, and polydeoxyribonucleotide (PDRN) and was fabricated using ice particle leaching [51]. The scaffold was enhanced with EVs that were derived from TNF-α/IFN-γ-primed UC-MSCs (TI-EVs) [51]. Ko et al. characterized the EVs by shape and size. They then studied the effector molecules within the EVs; more specifically, Ko et al. focused on proteins within TI-EVs and unprimed EVs (UC-EVs). The results indicated that treating UC-MSCs with TNF-α/IFN-γ enhanced the cellular uptake capabilities of secreted EVs and induced changes in protein cargo, which is indicative of kidney tissue regeneration [51].
In a partial nephrectomy mouse model, the scaffold containing PDRN and EVs induced glomerular regeneration and the restoration of kidney function [51]. More specifically, the kidney developmental factors (Pax2, Wt1, and Emx2) increased in expression with PME/PDRN/TI-EV scaffold treatment [51]. Additionally, there was an increase in the population of Pax2-expressing host cells, which indicates that the scaffold can facilitate host renal stem/progenitor cell infiltration [51]. Furthermore, there was an increased expression of pro-angiogenic growth factors (FGF2, HGF, and VEGF) in the PME/PDRN/TI-EV scaffold group [51]. Ko et al. determined the total number of functional glomeruli using H&E staining, with the PME/PDRN/TI-EV group showing the best results [51]. They then examined renal function recovery by evaluating the serum creatinine and blood urea nitrogen (BUN) levels. They found significantly better metabolic function in the PME/PDRN/TI-EV group [51]. Furthermore, the glomerular filtration rate (GFR) was restored in the PME/PDRN/TI-EV group (227.2 μL/min) to a level similar to that of native mice (232.5 μL/min) [51]. These results show structural and functional kidney tissue regeneration with the use of the PME/PDRN/TI-EV scaffold. Overall, the biochemical cues from TI-EVs and PDRN as well as the biophysical cues from the PLGA scaffold serve as potential tissue engineering platforms for kidney tissue regeneration [51].

6.4. Poly(L-Lactide) (PLLA)

PLLA degrades by nonenzymatic hydrolysis and its by-products are eliminated via normal cell metabolism [217]. Thus, it is biodegradable and biocompatible. The delivery of antibiotics [218,219,220,221] and chemotherapeutics [222,223,224,225,226] using PLLA has been frequently studied as well as anti-inflammatory drugs [227], anti-psychotics [228], acetylcholinesterase inhibitors [229], and ocular disease therapeutics [230,231]. In addition to drug delivery, cells [232,233], growth factors [234,235,236,237], and genetic material [238] have been delivered using PLLA. Furthermore, PLLA has been utilized to deliver hormones and fertilizer [239].
Swanson et al. engineered a biodegradable PLLA-based delivery platform to control the release of exosomes from microspheres to promote craniofacial bone healing. More specifically, they used PLGA and PEG triblock copolymer microspheres to encapsulate and control the timed release of human dental pulp stem cell (hDPSC)-derived exosomes [54]. This delivery platform was integrated with a 3D tissue engineered PLLA scaffold. They found that microspheres containing exosomes demonstrated a linear and consistent release profile over a longer period of time when attached to a PLLA scaffold compared to freely suspended microspheres [54]. They also found through NTA and TEM that the exosomes maintained their characteristic diameter and morphology throughout their encapsulation and release [54]. Furthermore, Swanson et al. confirmed the in vitro bioactivity of the exosome-containing microspheres by culturing mouse BMSCs on nanofibrous scaffolds functionalized with the microspheres. They used the colorimetric calcium assay to examine hydroxyapatite mineralization and energy dispersive X-ray spectroscopy (EDX) to determine the spatial distribution of elements in the construct [54]. It was found that microsphere-functionalized constructs and groups treated with exogenous exosomes showed an increased calcium phosphate content and a decreased proportion of organic components as minerals was deposited [54].
Furthermore, the authors demonstrated exosome functionality in vivo by implanting constructs in mice and subsequent staining (Masson’s Trichrome and von Kossa staining). This revealed that constructs with exosome-containing microspheres increased hECM deposition and promoted early mineralization compared to blank microsphere constructs and blank scaffolds, which attributes the function to the exosomes [54]. Furthermore, the constructs were used in a calvarial bone defect model. After 8 weeks, the functionalized constructs were laden with cells, collagen-rich matrix, marrow-containing bone tissue, and were integrated with the host at 8 weeks [54]. Additionally, the µCT of the skulls showed that localized delivery of the exosomes via microspheres in the scaffold resulted in the best regenerative outcome among the treatment groups [54].
Overall, the functionalized scaffold system was able to recruit endogenous cells and stimulate bone tissue neogenesis in vivo [54]. The exosomes used in this study provided pro-mineralization cues that guided local progenitor cells toward osteogenic differentiation both in vitro and in vivo [54]. By incorporating exosomes into a synthetic biopolymer-based scaffold, researchers can work toward overcoming the inherent lack of biochemical cues in synthetic biomaterials to achieve therapeutic effects such as bone healing.

6.5. Poly(Lactic Acid) (PLA)

PLA is biocompatible and biodegradable via hydrolysis and enzymatic activity and is also highly hydrophobic [240]. PLA has a wide range of mechanical and physical properties. Similar to the other discussed synthetic biopolymers, the delivery of antibiotics [241,242,243] and chemotherapeutics [240,244,245,246,247,248,249,250,251] using PLA has been well-studied. Additionally, therapeutics for fibrosis [252], arthritis [253], and osteoporosis [254] have been delivered using PLA. Furthermore, PLA has been used to deliver cells [255], growth factors [256], genetic material [257,258], and hormones [259]. Finally, polysaccharides [260], peptides [261], and plant-derived compounds [262,263] have been delivered via PLA.
The physical properties of PLA can be tailored, and PLA has already been used in other materials such as sutures, stents, and in oral surgery [48]. Calcium silicates (CaSi) have been shown to stimulate new bone formation in bone defects [48]. When doped with CaSi, PLA scaffolds have shown high values of bulk porosity, adequate thermal-mechanical properties, and can release Ca2+, OH, and nucleate apatite [48]. Gandolfi et al. aimed to develop a mineral-doped PLA-based scaffold functionalized with EVs to improve osteogenic commitment of human adipose-derived MSCs. Gandolfi et al. saw that mineral-doped PLA scaffolds adsorbed red-labelled human adipose mesenchymal stem cell (hAD-MSC)-derived exosomes [48]. The exosomes were then released and internalized by the cultured hAD-MSCs, and the osteogenic commitment and properties of the hAD-MSCs were confirmed by examining the gene expression of hAD-MSCs; the examined genes included Collagen type 1, Osteocalcin, and Runx [48]. Overall, mineral and exosome-doped PLA-based porous scaffolds provided a suitable bone-forming microenvironment, triggered the osteogenic commitment of the cells, and improved the osteogenic properties of the cells [48]. As a result of the minerals and exosomes providing osteogenic cues, this scaffold system has potential in regenerative bone healing. Future work for Gandolfi et al. includes implanting this functionalized scaffold in vivo to study the potential pro-osteogenic effects on MSCs present at the wound site [48]
In this section, we examined the therapeutic advantages using synthetic biopolymer-based scaffolds endowed with EVs. Some of the demonstrated benefits of EV incorporation are enhanced wound healing, bone tissue neogenesis, kidney regeneration, and improved vascular performance and functionality.

7. EV Delivery via Scaffolds for Tissue Repair

Given the complex nature of the various systems and tissues that make up a living organism, it is crucial that tissue engineers design scaffolds that can restore, maintain, or improve specific tissues or whole organ function. In other words, scaffold-based delivery systems must be tailored to achieve a specific function with consideration of the natural tissue and environment that is being mimicked. As a result of the wide range of available biopolymers and the necessity for specificity in tissue engineering, there are many different engineered delivery systems for EVs. The use of biopolymers functionalized with EVs provides a novel method to address and aid tissue repair in various systems or organs. Below, we highlight the diversity of tissues that may benefit from EV delivery via biopolymer scaffolds to improve tissue repair. Furthermore, we briefly examine examples or considerations for EV delivery via scaffolds within each discussed tissue type.

7.1. Nerve Repair

The central nervous system (CNS) is notorious for limited regenerative capacity. Additionally, due to the high prevalence of CNS conditions (e.g., stroke), it is crucial to develop therapies to restore function to pre-injury levels [264]. It is important that the developed scaffolds mimic the CNS microenvironments in terms of viscoelasticity to facilitate the migration and differentiation of endogenous stem cells within the scaffold [264]. An example of a promising delivery vehicle is PLLA scaffolds coated with collagen IV, which have been shown to serve as excellent matrices for astrocytes [265]. This scaffold may prove useful for EV delivery in the future. Another consideration is incorporating EVs into hydrogel scaffolds to maximize the therapeutic potential of EVs in intracerebral administration [264]. Furthermore, exosomes have been delivered from a fibrin gel to accelerate recovery from spinal cord injury [266]. VGF (nerve growth factor inducible) was abundant in the exosomes used in this study and played a key role in increased oligodendrogenesis in vitro and in vivo, thus aiding in functional recovery. It is expected to see a greater development of EV delivery via scaffolds for neural repair in the future.

7.2. Bone Repair

Bone tissue is dynamic and has a regenerative capacity, as seen through bone remodeling [267]. However, the bone regenerative process may become impaired due to infections, genetic disorder, trauma, etc. and require bone grafting [267]. Conventional autologous bone grafts are relatively easy to obtain and include osteogenic, osteoinductive, and osteoconductive properties [268]. Additionally, autologous bone grafts do not raise an immune response nor do they transmit infectious diseases as the graft is the host’s own tissue [268]. However, autologous bone grafts require an additional operative time, can be difficult to mold to the receiving site, and are associated with donor site morbidity [269]. Furthermore, the volume of bone that can be harvested from a site is limited, and transplanted bone may resorb [270]. With increasing evidence of EV-laden scaffolds aiding in bone healing, it may be possible to design a biomaterial that could replace autologous bone grafts [48]. This would avoid donor site morbidity, provide needed bone volume, and likely reduce costs.
Biopolymers for bone regenerative purposes that have been well-studied include ceramic, polymer, and composite materials [270]. Typically, scaffolds for bone regeneration utilize organic and inorganic biomaterials due to bone tissue naturally containing inorganic and organic phases [270]. EVs can be tailored or engineered for purposes such as bone regeneration. For example, osteoblast-derived EVs have been shown to promote bone marrow-derived mesenchymal stem cell differentiation into osteoblasts [270]. Some other designs include specifically engineering EVs to enhance osteogenic differentiation [271] and creating irregular scaffolds to better reflect the microstructure of the cortical/cancellous bone unit [272]. Ultimately, there are numerous approaches and methods to improve bone regeneration, and integrating EVs with scaffolds is one of the most promising avenues with excellent translational potential.

7.3. Cardiovascular Tissue Repair

When it comes to the cardiovascular system, heart failure is one of the worldwide leading causes of death [273]. Heart transplants are the standard curative therapy for end-stage heart failure, however, donor shortages and the requirement of lifelong immunosuppression after organ transplantation drives the field of cardiovascular tissue engineering [273]. One of the main goals of cardiovascular tissue engineering is developing regenerative grafts/scaffolds to restore lost cardiac tissue. An example of this is using tubular scaffolds to bypass or replace defective arterial segments [33]. Another example incorporates MSC-derived EVs into a cardiac scaffold for local and systemic immunomodulation following myocardial infarction [274]. Furthermore, small EVs may be incorporated into alginate scaffolds for delivery and retention in the heart, thus improving the therapeutic effects of the small EVs [26]. EV incorporation into such scaffolds works toward restoring lost cardiac tissue.

7.4. Wound Healing

The regenerative effects of EVs in terms of wound healing have been explored. The prevalence of combat, burn, and diabetic wounds demonstrate the need for therapies that facilitate wound healing. Exosomes can promote the proliferation and migration of fibroblasts as well as regulate Type I and III collagen and fibronectin expression [275]. Furthermore, chitosan-based hydrogels enriched with exosomes have shown positive effects on wound healing by promoting angiogenesis and tissue granulation formation [35]. Additionally, Zhao et al. found that gelatin methacryloyl (GelMA) hydrogels embedded with exosomes showed accelerated re-epithelialization, collagen maturity promotion, and improved angiogenesis in vivo [276]. Wound healing approaches that deliver EVs via scaffolds have been heavily studied and have a lot of translational potential [32,37].

8. EV Delivery via Scaffolds for Immunomodulation

The immune cells and mediators play important roles in mediating tissue homeostasis and reparative processes through a variety of orchestrated events and by being involved in inflammation, angiogenesis, and stem/progenitor cell activities such as proliferation and differentiation [277,278,279]. Similar to MSCs that have immunomodulatory functions, the immunomodulatory function associated with MSC-exosomes/EVs involves promoting the M2-like macrophage phenotype, Treg population, and TH2 immune responses [280,281]. Both immunomodulatory and proangiogenic factors were detected in MSC exosomes through proteomic profiling [282]. It was reported that most of the cells absorbing MSC-derived exosomes from the scaffolds were immune cells [282]. This may imply the importance of immune responses in tissue reactions to scaffolds with and without EVs and the necessity to understand and use this mechanism to guide the biological communication network between different cell types. In addition, the cellular communication network between the immunomodulation and regeneration processes in exosome-functionalized scaffolds still needs to be elucidated in detail.
Inflammation is indispensable and irreplaceable in the process of tissue repair and both positively and negatively regulates the tissue repair process. The tissue repair cascade starts from a pro-inflammatory reaction and must be regulated in a timely manner to ellicit appropriate tissue repair/regeneration. Along this line, MSC exosome–functionalized scaffolds were shown to induce innate and adaptive immunomodulatory responses toward tissue repair by proactively recruiting immune cells and modulating orchestrated M2/TH2/Treg responses locally and on the systemic level [282]. In addition, three-dimensional-printed scaffolds consisting of bioceramic-induced macrophage exosomes were demonstrated to regulate immunomodulation, osteogenesis, and angiogenesis [283].

9. Challenges in EV Loading, Integrity, Delivery, and Scaling Up

Despite the positive therapeutic effects of EVs, challenges regarding the uniform EV loading of scaffolds remain. More specifically, EV or secretome solutions may not reach the innermost regions of scaffolds depending on the EV loading method [284]. For example, Bari et al. showed that the lyosecretome solution did not reach the innermost regions of their 3D printed PCL scaffold when an adsorption loading method was used [284]. Additionally, this simple adsorption method was accompanied with high batch-to-batch loading variability [284]. However, Bari et al. utilized a second loading method involving co-printing PCL and lyosecretome-containing alginate, which demonstrated a slower release profile as well as the homogeneous loading of proteins/lipids [284]. It remains to be elucidated whether this second method reduces the batch-to-batch loading variability. Each of these methods are useful for specific goals (e.g., quick or prolonged EV release), however, adsorption loading methods may result in EV loading variability.
In another study, Xing et al. modified a silk fibroin/PCL scaffold with polydopamine (pDA). They found through laser scanning confocal microscopy that pDA modification of the scaffold led to significantly more efficient EV loading compared to conventional superficial EV adsorption onto the silk fibroin/PCL scaffold [285]. Xing et al. also found that the EV adsorption technique led to a burst release effect with EV depletion within nine days [285]. Meanwhile, in the pDA-modified scaffold, EVs demonstrated a slow-release profile with about 10% of EVs retained within the scaffold after nine days [285]. These studies highlighted overcoming challenges regarding EV loading efficiency and sustained EV release from the scaffolds. Researchers may want to consider scaffold loading alternatives to simple EV adsorption such as unique 3D printing scaffold design or biopolymer modification to overcome the discussed challenges.
An important concern regarding EV delivery is the maintenance of EV lipid membrane integrity. The lipid membrane plays an important role in protecting the nucleic acid cargo of EVs, which is responsible for the physiological effects of EVs [54]. Thus, it is crucial that the lipid membrane of EVs remains intact throughout scaffold loading and delivery. EV membrane integrity may be evidenced through electron microscopy (although this method is not high-throughput) or through assays that examine proteins that are tethered to the EV membrane (e.g., CD81) [286]. Furthermore, EV integrity may be studied by examining the EV characteristics prior to scaffold loading and after release from the scaffolds. For instance, Huang et al. found that engineered EVs maintained their integrity before scaffold loading and after delivery [20]. This was demonstrated through nanoparticle tracking analysis (NTA) and polydispersity index (PDI) in which EVs released from RGD peptide-containing hydrogels maintained a similar size distribution and PDI compared to the pre-scaffold encapsulation control group [20]. Following this confirmation, they found that the EVs maintained their desired functionality in vitro and in vivo after hydrogel encapsulation [20].
Nevertheless, there is a need for more widespread confirmation of maintained EV membrane integrity and bioactivity. For instance, Su et al. loaded and tethered exosomes using polyethylenimine (PEI) molecules to a PCL-based scaffold [282]. They found that the exosomes released from this scaffold did not lose their function regarding modulating macrophages to M2-like phenotypes in vitro compared to freely suspended exosome vesicles [282]. Even though Su et al. characterized the diameter and zeta potential of exosomes before scaffold loading, they did not report the examination of these same parameters following exosome release from the scaffold. Many studies such as [287,288] characterized EVs prior to scaffold loading and went on to examine the in vitro and in vivo bioactivity of the loaded scaffolds. These studies failed to confirm the physical integrity of the EVs when released from the scaffolds. Overall, many current approaches have demonstrated positive bioactive effects of EVs delivered from the scaffolds, however, few studies have confirmed EV membrane integrity after delivery. It is important that researchers take this extra confirmation step so that they can attribute the observed functionality to EVs, thus strengthening and supporting their findings.
Furthermore, there are issues concerning the optimal scaffold pore size and pore size distribution of the scaffolds. Pore parameters play a role in controlling the release of EVs, and the pore size contributes significantly to the effects the microenvironment has on cellular functions (e.g., cell adhesion and polarization) [289,290]. Additionally, the average pore size affects the rheological phenotype and diffusivity of the scaffolds [289]. Furthermore, the distribution of pore sizes in a scaffold affects the analysis of the mechanical properties of the scaffold, diffusive processes, and cell migration through the scaffold [289]. There is no agreement on the optimal porosity value or pore size, however, as long as mechanical properties of a scaffold are satisfied, porosity values over 90% are recommended, along with a pore size range from 10 to at least 200 μm [48].
Other challenges concerning scaffolds include the consistency of produced scaffolds and scaling up production. When it comes to scaling up efforts, engineered scaffolds may work well at a small scale, however, it may be difficult to scale up these materials for larger trials, unless the production of each type of unique scaffold is standardized. Furthermore, EV isolation may also need to be standardized to allow for consistency and reproducibility, especially for scaling up measures.

10. Conclusions

In this review, we discussed the important roles EVs play and their therapeutic potential. We also explored the different aspects of natural and synthetic biopolymers and how EVs may be incorporated into these scaffolds. Delivering EVs bypasses the concerns regarding the use of stem cells in tissue engineering. As evidenced by the discussed studies, scaffolds functionalized with EVs have a wide range of applicability including tissue remodeling, wound healing, and bone healing. Even though natural and synthetic biopolymer-based scaffolds each have their own advantages and disadvantages, their therapeutic potential can be enhanced by incorporating EVs. Throughout this review, we highlighted the potential mechanisms by which EVs impart therapeutic effects and their role in immunomodulation. Overall, there are many types of biopolymers that may be used to deliver EVs, and research concerning how EVs exert therapeutic effects is ongoing.

Author Contributions

Conceptualization, S.R. and K.S.L.; Methodology, K.S.L.; Software, K.S.L.; Validation, K.S.L., S.S., L.F.C. and S.R.; Formal Analysis, K.S.L.; Investigation, K.S.L. and S.S.; Resources, S.R. and K.S.L.; Data Curation, K.S.L. and S.S.; Writing—Original Draft Preparation, K.S.L.; Writing—Review & Editing, L.F.C., S.R., S.S. and K.S.L.; Visualization, K.S.L.; Supervision, S.R.; Project Administration, K.S.L. and S.R.; Funding Acquisition, S.R. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by National Institutes of Health R01 DE027404 (SR) and R01 DE030495 (SR).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Acknowledgments

Figure 1 was created with BioRender.com (https://biorender.com/).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Liu, H.; Li, R.; Liu, T.; Yang, L.; Yin, G.; Xie, Q. Immunomodulatory Effects of Mesenchymal Stem Cells and Mesenchymal Stem Cell-Derived Extracellular Vesicles in Rheumatoid Arthritis. Front. Immunol. 2020, 11, 1912. [Google Scholar] [CrossRef] [PubMed]
  2. Zhao, M.; Liu, S.; Wang, C.; Wang, Y.; Wan, M.; Liu, F.; Gong, M.; Yuan, Y.; Chen, Y.; Cheng, J.; et al. Mesenchymal Stem Cell-Derived Extracellular Vesicles Attenuate Mitochondrial Damage and Inflammation by Stabilizing Mitochondrial DNA. ACS Nano 2021, 15, 1519–1538. [Google Scholar] [CrossRef] [PubMed]
  3. Branscome, H.; Paul, S.; Khatkar, P.; Kim, Y.; Barclay, R.A.; Pinto, D.O.; Yin, D.; Zhou, W.; Liotta, L.A.; El-Hage, N.; et al. Stem Cell Extracellular Vesicles and Their Potential to Contribute to the Repair of Damaged CNS Cells. J. Neuroimmune Pharmacol. 2020, 15, 520–537. [Google Scholar] [CrossRef] [PubMed]
  4. Webber, M.J.; Khan, O.F.; Sydlik, S.A.; Tang, B.C.; Langer, R. A Perspective on the Clinical Translation of Scaffolds for Tissue Engineering. Ann. Biomed. Eng. 2015, 43, 641–656. [Google Scholar] [CrossRef] [PubMed]
  5. Shahin, H.I.; Radnaa, E.; Tantengco, O.A.G.; Kechichian, T.; Kammala, A.K.; Sheller-Miller, S.; Taylor, B.D.; Menon, R. Microvesicles and Exosomes Released by Amnion Epithelial Cells under Oxidative Stress Cause Inflammatory Changes in Uterine Cells. Biol. Reprod. 2021, 105, 464–480. [Google Scholar] [CrossRef]
  6. Fonseka, P.; Marzan, A.L.; Mathivanan, S. Introduction to the Community of Extracellular Vesicles. In New Frontiers: Extracellular Vesicles; Mathivanan, S., Fonseka, P., Nedeva, C., Atukorala, I., Eds.; Subcellular Biochemistry; Springer International Publishing: Cham, Switzerland, 2021; pp. 3–18. [Google Scholar] [CrossRef]
  7. Théry, C.; Witwer, K.W.; Aikawa, E.; Alcaraz, M.J.; Anderson, J.D.; Andriantsitohaina, R.; Antoniou, A.; Arab, T.; Archer, F.; Atkin-Smith, G.K.; et al. Minimal Information for Studies of Extracellular Vesicles 2018 (MISEV2018): A Position Statement of the International Society for Extracellular Vesicles and Update of the MISEV2014 Guidelines. J. Extracell. Vesicles 2018, 7, 1535750. [Google Scholar] [CrossRef]
  8. Rai, A.; Fang, H.; Fatmous, M.; Claridge, B.; Poh, Q.H.; Simpson, R.J.; Greening, D.W. A Protocol for Isolation, Purification, Characterization, and Functional Dissection of Exosomes. Methods Mol. Biol. 2021, 2261, 105–149. [Google Scholar] [CrossRef]
  9. Ter-Ovanesyan, D.; Norman, M.; Lazarovits, R.; Trieu, W.; Lee, J.-H.; Church, G.M.; Walt, D.R. Framework for Rapid Comparison of Extracellular Vesicle Isolation Methods. eLife 2021, 10, e70725. [Google Scholar] [CrossRef]
  10. Brennan, K.; Martin, K.; FitzGerald, S.P.; O’Sullivan, J.; Wu, Y.; Blanco, A.; Richardson, C.; Mc Gee, M.M. A Comparison of Methods for the Isolation and Separation of Extracellular Vesicles from Protein and Lipid Particles in Human Serum. Sci. Rep. 2020, 10, 1039. [Google Scholar] [CrossRef] [Green Version]
  11. Torres Crigna, A.; Fricke, F.; Nitschke, K.; Worst, T.; Erb, U.; Karremann, M.; Buschmann, D.; Elvers-Hornung, S.; Tucher, C.; Schiller, M.; et al. Inter-Laboratory Comparison of Extracellular Vesicle Isolation Based on Ultracentrifugation. Transfus. Med. Hemother. 2021, 48, 48–59. [Google Scholar] [CrossRef]
  12. Sharma, S.; LeClaire, M.; Wohlschlegel, J.; Gimzewski, J. Impact of Isolation Methods on the Biophysical Heterogeneity of Single Extracellular Vesicles. Sci. Rep. 2020, 10, 13327. [Google Scholar] [CrossRef] [PubMed]
  13. Monguió-Tortajada, M.; Morón-Font, M.; Gámez-Valero, A.; Carreras-Planella, L.; Borràs, F.E.; Franquesa, M. Extracellular-Vesicle Isolation from Different Biological Fluids by Size-Exclusion Chromatography. Curr. Protoc. Stem Cell Biol. 2019, 49, e82. [Google Scholar] [CrossRef] [PubMed]
  14. Van Deun, J.; Mestdagh, P.; Agostinis, P.; Akay, Ö.; Anand, S.; Anckaert, J.; Martinez, Z.A.; Baetens, T.; Beghein, E.; Bertier, L.; et al. EV-TRACK: Transparent Reporting and Centralizing Knowledge in Extracellular Vesicle Research. Nat. Methods 2017, 14, 228–232. [Google Scholar] [CrossRef] [PubMed]
  15. Priglinger, E.; Strasser, J.; Buchroithner, B.; Weber, F.; Wolbank, S.; Auer, D.; Grasmann, E.; Arzt, C.; Sivun, D.; Grillari, J.; et al. Label-Free Characterization of an Extracellular Vesicle-Based Therapeutic. J. Extracell. Vesicles 2021, 10, e12156. [Google Scholar] [CrossRef] [PubMed]
  16. Crescitelli, R.; Lässer, C.; Lötvall, J. Isolation and Characterization of Extracellular Vesicle Subpopulations from Tissues. Nat. Protoc. 2021, 16, 1548–1580. [Google Scholar] [CrossRef]
  17. Shirazi, S.; Huang, C.-C.; Kang, M.; Lu, Y.; Ravindran, S.; Cooper, L.F. The Importance of Cellular and Exosomal MiRNAs in Mesenchymal Stem Cell Osteoblastic Differentiation. Sci. Rep. 2021, 11, 5953. [Google Scholar] [CrossRef]
  18. Kim, S.-D.; Kang, S.A.; Kim, Y.-W.; Yu, H.S.; Cho, K.-S.; Roh, H.-J. Screening and Functional Pathway Analysis of Pulmonary Genes Associated with Suppression of Allergic Airway Inflammation by Adipose Stem Cell-Derived Extracellular Vesicles. Stem Cells Int. 2020, 2020, 5684250. [Google Scholar] [CrossRef]
  19. Liu, W.; Yu, M.; Xie, D.; Wang, L.; Ye, C.; Zhu, Q.; Liu, F.; Yang, L. Melatonin-Stimulated MSC-Derived Exosomes Improve Diabetic Wound Healing through Regulating Macrophage M1 and M2 Polarization by Targeting the PTEN/AKT Pathway. Stem Cell Res. Ther. 2020, 11, 259. [Google Scholar] [CrossRef]
  20. Huang, C.-C.; Kang, M.; Shirazi, S.; Lu, Y.; Cooper, L.F.; Gajendrareddy, P.; Ravindran, S. 3D Encapsulation and Tethering of Functionally Engineered Extracellular Vesicles to Hydrogels. Acta Biomater. 2021, 126, 199–210. [Google Scholar] [CrossRef]
  21. Kang, M.; Huang, C.-C.; Lu, Y.; Shirazi, S.; Gajendrareddy, P.; Ravindran, S.; Cooper, L.F. Bone Regeneration Is Mediated by Macrophage Extracellular Vesicles. Bone 2020, 141, 115627. [Google Scholar] [CrossRef]
  22. Cho, K.-S.; Kang, S.A.; Kim, S.-D.; Mun, S.-J.; Yu, H.S.; Roh, H.-J. Dendritic Cells and M2 Macrophage Play an Important Role in Suppression of Th2-Mediated Inflammation by Adipose Stem Cells-Derived Extracellular Vesicles. Stem Cell Res. 2019, 39, 101500. [Google Scholar] [CrossRef] [PubMed]
  23. Lee, K.S.; Lee, J.; Kim, H.K.; Yeom, S.H.; Woo, C.H.; Jung, Y.J.; Yun, Y.E.; Park, S.Y.; Han, J.; Kim, E.; et al. Extracellular Vesicles from Adipose Tissue-derived Stem Cells Alleviate Osteoporosis through Osteoprotegerin and MiR-21-5p. J. Extracell. Vesicles 2021, 10, e12152. [Google Scholar] [CrossRef] [PubMed]
  24. Garcia-Martin, R.; Wang, G.; Brandão, B.B.; Zanotto, T.M.; Shah, S.; Kumar Patel, S.; Schilling, B.; Kahn, C.R. MicroRNA Sequence Codes for Small Extracellular Vesicle Release and Cellular Retention. Nature 2022, 601, 446–451. [Google Scholar] [CrossRef]
  25. Harrell, C.R.; Jovicic, N.; Djonov, V.; Arsenijevic, N.; Volarevic, V. Mesenchymal Stem Cell-Derived Exosomes and Other Extracellular Vesicles as New Remedies in the Therapy of Inflammatory Diseases. Cells 2019, 8, 1605. [Google Scholar] [CrossRef]
  26. Lv, K.; Li, Q.; Zhang, L.; Wang, Y.; Zhong, Z.; Zhao, J.; Lin, X.; Wang, J.; Zhu, K.; Xiao, C.; et al. Incorporation of Small Extracellular Vesicles in Sodium Alginate Hydrogel as a Novel Therapeutic Strategy for Myocardial Infarction. Theranostics 2019, 9, 7403–7416. [Google Scholar] [CrossRef] [PubMed]
  27. Xing, H.; Zhang, Z.; Mao, Q.; Wang, C.; Zhou, Y.; Zhou, X.; Ying, L.; Xu, H.; Hu, S.; Zhang, N. Injectable Exosome-Functionalized Extracellular Matrix Hydrogel for Metabolism Balance and Pyroptosis Regulation in Intervertebral Disc Degeneration. J. Nanobiotechnol. 2021, 19, 264. [Google Scholar] [CrossRef] [PubMed]
  28. Hao, D.; Swindell, H.S.; Ramasubramanian, L.; Liu, R.; Lam, K.S.; Farmer, D.L.; Wang, A. Extracellular Matrix Mimicking Nanofibrous Scaffolds Modified With Mesenchymal Stem Cell-Derived Extracellular Vesicles for Improved Vascularization. Front. Bioeng. Biotechnol. 2020, 8, 633. [Google Scholar] [CrossRef]
  29. Lazar, S.; Mor, S.; Chen, J.; Hao, D.; Wang, A. Bioengineered Extracellular Vesicle-Loaded Bioscaffolds for Therapeutic Applications in Regenerative Medicine. Extracell. Vesicles Circ. Nucl. Acids 2021, 2, 175–178. [Google Scholar] [CrossRef]
  30. Mallick, S.P.; Singh, B.N.; Rastogi, A.; Srivastava, P. Design and Evaluation of Chitosan/Poly(l-Lactide)/Pectin Based Composite Scaffolds for Cartilage Tissue Regeneration. Int. J. Biol. Macromol. 2018, 112, 909–920. [Google Scholar] [CrossRef]
  31. Akiyama, N.; Yamamoto-Fukuda, T.; Takahashi, H.; Koji, T. In Situ Tissue Engineering with Synthetic Self-Assembling Peptide Nanofiber Scaffolds, PuraMatrix, for Mucosal Regeneration in the Rat Middle-Ear. Int. J. Nanomed. 2013, 8, 2629–2640. [Google Scholar] [CrossRef]
  32. Abolgheit, S.; Abdelkader, S.; Aboushelib, M.; Omar, E.; Mehanna, R. Bone Marrow-Derived Mesenchymal Stem Cells and Extracellular Vesicles Enriched Collagen Chitosan Scaffold in Skin Wound Healing (a Rat Model). J. Biomater. Appl. 2021, 36, 128–139. [Google Scholar] [CrossRef] [PubMed]
  33. Cunnane, E.M.; Lorentz, K.L.; Ramaswamy, A.K.; Gupta, P.; Mandal, B.B.; O’Brien, F.J.; Weinbaum, J.S.; Vorp, D.A. Extracellular Vesicles Enhance the Remodeling of Cell-Free Silk Vascular Scaffolds in Rat Aortae. ACS Appl. Mater. Interfaces 2020, 12, 26955–26965. [Google Scholar] [CrossRef] [PubMed]
  34. Asadpour, S.; Kargozar, S.; Moradi, L.; Ai, A.; Nosrati, H.; Ai, J. Natural Biomacromolecule Based Composite Scaffolds from Silk Fibroin, Gelatin and Chitosan toward Tissue Engineering Applications. Int. J. Biol. Macromol. 2020, 154, 1285–1294. [Google Scholar] [CrossRef]
  35. Nooshabadi, V.T.; Khanmohamadi, M.; Valipour, E.; Mahdipour, S.; Salati, A.; Malekshahi, Z.V.; Shafei, S.; Amini, E.; Farzamfar, S.; Ai, J. Impact of Exosome-Loaded Chitosan Hydrogel in Wound Repair and Layered Dermal Reconstitution in Mice Animal Model. J. Biomed. Mater. Res. Part A 2020, 108, 2138–2149. [Google Scholar] [CrossRef]
  36. Shafei, S.; Khanmohammadi, M.; Heidari, R.; Ghanbari, H.; Taghdiri Nooshabadi, V.; Farzamfar, S.; Akbariqomi, M.; Sanikhani, N.S.; Absalan, M.; Tavoosidana, G. Exosome Loaded Alginate Hydrogel Promotes Tissue Regeneration in Full-Thickness Skin Wounds: An in Vivo Study. J. Biomed. Mater. Res. A 2020, 108, 545–556. [Google Scholar] [CrossRef]
  37. Zhang, Y.; Zhang, P.; Gao, X.; Chang, L.; Chen, Z.; Mei, X. Preparation of Exosomes Encapsulated Nanohydrogel for Accelerating Wound Healing of Diabetic Rats by Promoting Angiogenesis. Mater. Sci. Eng. C 2021, 120, 111671. [Google Scholar] [CrossRef]
  38. Yang, Z.; Yang, Y.; Xu, Y.; Jiang, W.; Shao, Y.; Xing, J.; Chen, Y.; Han, Y. Biomimetic Nerve Guidance Conduit Containing Engineered Exosomes of Adipose-Derived Stem Cells Promotes Peripheral Nerve Regeneration. Stem Cell Res. Ther. 2021, 12, 442. [Google Scholar] [CrossRef] [PubMed]
  39. Imanishi, Y.; Hata, M.; Matsukawa, R.; Aoyagi, A.; Omi, M.; Mizutani, M.; Naruse, K.; Ozawa, S.; Honda, M.; Matsubara, T.; et al. Efficacy of Extracellular Vesicles from Dental Pulp Stem Cells for Bone Regeneration in Rat Calvarial Bone Defects. Inflamm Regen 2021, 41, 12. [Google Scholar] [CrossRef]
  40. Xin, L.; Lin, X.; Zhou, F.; Li, C.; Wang, X.; Yu, H.; Pan, Y.; Fei, H.; Ma, L.; Zhang, S. A Scaffold Laden with Mesenchymal Stem Cell-Derived Exosomes for Promoting Endometrium Regeneration and Fertility Restoration through Macrophage Immunomodulation. Acta Biomater. 2020, 113, 252–266. [Google Scholar] [CrossRef]
  41. Wu, D.; Qin, H.; Wang, Z.; Yu, M.; Liu, Z.; Peng, H.; Liang, L.; Zhang, C.; Wei, X. Bone Mesenchymal Stem Cell-Derived SEV-Encapsulated Thermosensitive Hydrogels Accelerate Osteogenesis and Angiogenesis by Release of Exosomal MiR-21. Front. Bioeng. Biotechnol. 2022, 9, 829136. [Google Scholar] [CrossRef]
  42. Tang, Q.; Lu, B.; He, J.; Chen, X.; Fu, Q.; Han, H.; Luo, C.; Yin, H.; Qin, Z.; Lyu, D.; et al. Exosomes-Loaded Thermosensitive Hydrogels for Corneal Epithelium and Stroma Regeneration. Biomaterials 2022, 280, 121320. [Google Scholar] [CrossRef] [PubMed]
  43. Nikhil, A.; Kumar, A. Evaluating Potential of Tissue-Engineered Cryogels and Chondrocyte Derived Exosomes in Articular Cartilage Repair. Biotechnol. Bioeng. 2022, 119, 605–625. [Google Scholar] [CrossRef] [PubMed]
  44. Man, K.; Barroso, I.A.; Brunet, M.Y.; Peacock, B.; Federici, A.S.; Hoey, D.A.; Cox, S.C. Controlled Release of Epigenetically-Enhanced Extracellular Vesicles from a GelMA/Nanoclay Composite Hydrogel to Promote Bone Repair. Int. J. Mol. Sci. 2022, 23, 832. [Google Scholar] [CrossRef] [PubMed]
  45. Song, K.; Jiang, T.; Pan, P.; Yao, Y.; Jiang, Q. Exosomes from Tendon Derived Stem Cells Promote Tendon Repair through MiR-144-3p-Regulated Tenocyte Proliferation and Migration. Stem Cell Res. Ther. 2022, 13, 80. [Google Scholar] [CrossRef]
  46. Heirani-Tabasi, A.; Hosseinzadeh, S.; Rabbani, S.; Tafti, S.H.A.; Jamshidi, K.; Soufizomorrod, M.; Soleimani, M. Cartilage Tissue Engineering by Co-Transplantation of Chondrocyte Extracellular Vesicles and Mesenchymal Stem Cells, Entrapped in Chitosan–Hyaluronic Acid Hydrogel. Biomed. Mater. 2021, 16, 055003. [Google Scholar] [CrossRef]
  47. Kwak, G.; Cheng, J.; Kim, H.; Song, S.; Lee, S.J.; Yang, Y.; Jeong, J.H.; Lee, J.E.; Messersmith, P.B.; Kim, S.H. Sustained Exosome-Guided Macrophage Polarization Using Hydrolytically Degradable PEG Hydrogels for Cutaneous Wound Healing: Identification of Key Proteins and MiRNAs, and Sustained Release Formulation. Small 2022, 18, 2200060. [Google Scholar] [CrossRef]
  48. Gandolfi, M.G.; Gardin, C.; Zamparini, F.; Ferroni, L.; Esposti, M.D.; Parchi, G.; Ercan, B.; Manzoli, L.; Fava, F.; Fabbri, P.; et al. Mineral-Doped Poly(L-Lactide) Acid Scaffolds Enriched with Exosomes Improve Osteogenic Commitment of Human Adipose-Derived Mesenchymal Stem Cells. Nanomaterials 2020, 10, 432. [Google Scholar] [CrossRef]
  49. Wei, Y.; Wu, Y.; Zhao, R.; Zhang, K.; Midgley, A.C.; Kong, D.; Li, Z.; Zhao, Q. MSC-Derived SEVs Enhance Patency and Inhibit Calcification of Synthetic Vascular Grafts by Immunomodulation in a Rat Model of Hyperlipidemia. Biomaterials 2019, 204, 13–24. [Google Scholar] [CrossRef]
  50. Wang, X.; Ao, J.; Lu, H.; Zhao, Q.; Ma, Y.; Zhang, J.; Ren, H.; Zhang, Y. Osteoimmune Modulation and Guided Osteogenesis Promoted by Barrier Membranes Incorporated with S-Nitrosoglutathione (GSNO) and Mesenchymal Stem Cell-Derived Exosomes. Int. J. Nanomed. 2020, 15, 3483–3496. [Google Scholar] [CrossRef]
  51. Ko, K.-W.; Park, S.-Y.; Lee, E.H.; Yoo, Y.-I.; Kim, D.-S.; Kim, J.Y.; Kwon, T.G.; Han, D.K. Integrated Bioactive Scaffold with Polydeoxyribonucleotide and Stem-Cell-Derived Extracellular Vesicles for Kidney Regeneration. ACS Nano 2021, 15, 7575–7585. [Google Scholar] [CrossRef]
  52. Li, W.; Liu, Y.; Zhang, P.; Tang, Y.; Zhou, M.; Jiang, W.; Zhang, X.; Wu, G.; Zhou, Y. Tissue-Engineered Bone Immobilized with Human Adipose Stem Cells-Derived Exosomes Promotes Bone Regeneration. ACS Appl. Mater. Interfaces 2018, 10, 5240–5254. [Google Scholar] [CrossRef] [PubMed]
  53. Yang, Y.; Zhu, Z.; Gao, R.; Yuan, J.; Zhang, J.; Li, H.; Xie, Z.; Wang, Y. Controlled Release of MSC-Derived Small Extracellular Vesicles by an Injectable Diels-Alder Crosslinked Hyaluronic Acid/PEG Hydrogel for Osteoarthritis Improvement. Acta Biomater. 2021, 128, 163–174. [Google Scholar] [CrossRef] [PubMed]
  54. Swanson, W.B.; Zhang, Z.; Xiu, K.; Gong, T.; Eberle, M.; Wang, Z.; Ma, P.X. Scaffolds with Controlled Release of Pro-Mineralization Exosomes to Promote Craniofacial Bone Healing without Cell Transplantation. Acta Biomater. 2020, 118, 215–232. [Google Scholar] [CrossRef] [PubMed]
  55. Homem, N.C.; Tavares, T.D.; Miranda, C.S.; Antunes, J.C.; Amorim, M.T.P.; Felgueiras, H.P. Functionalization of Crosslinked Sodium Alginate/Gelatin Wet-Spun Porous Fibers with Nisin Z for the Inhibition of Staphylococcus aureus-Induced Infections. Int. J. Mol. Sci. 2021, 22, 1930. [Google Scholar] [CrossRef] [PubMed]
  56. Athamneh, T.; Amin, A.; Benke, E.; Ambrus, R.; Gurikov, P.; Smirnova, I.; Leopold, C.S. Pulmonary Drug Delivery with Aerogels: Engineering of Alginate and Alginate-Hyaluronic Acid Microspheres. Pharm. Dev. Technol. 2021, 26, 509–521. [Google Scholar] [CrossRef]
  57. Elbialy, N.S.; Mohamed, N. Alginate-Coated Caseinate Nanoparticles for Doxorubicin Delivery: Preparation, Characterisation, and in Vivo Assessment. Int. J. Biol. Macromol. 2020, 154, 114–122. [Google Scholar] [CrossRef]
  58. Sheng, Y.; Gao, J.; Yin, Z.-Z.; Kang, J.; Kong, Y. Dual-Drug Delivery System Based on the Hydrogels of Alginate and Sodium Carboxymethyl Cellulose for Colorectal Cancer Treatment. Carbohydr. Polym. 2021, 269, 118325. [Google Scholar] [CrossRef]
  59. Sarheed, O.; Dibi, M.; Ramesh, K.V.R.N.S.; Drechsler, M. Fabrication of Alginate-Based O/W Nanoemulsions for Transdermal Drug Delivery of Lidocaine: Influence of the Oil Phase and Surfactant. Molecules 2021, 26, 2556. [Google Scholar] [CrossRef]
  60. Rajalekshmy, G.; Rekha, M. Synthesis and Evaluation of an Alginate-Methacrylate Xerogel for Insulin Delivery towards Wound Healing Applications. Ther. Deliv. 2021, 12, 215–234. [Google Scholar] [CrossRef]
  61. Chen, Z.; Yu, P.; Miao, Z.; Zhang, H.; Xiao, H.; Xie, J.; Ding, C.; Li, J. Sulfated Alginate Based Complex for Sustained Calcitonin Delivery and Enhanced Osteogenesis. Biomed. Mater. 2021, 16, 035022. [Google Scholar] [CrossRef]
  62. Kumar, A.; Belhaj, M.; DiPette, D.J.; Potts, J.D. A Novel Alginate-Based Delivery System for the Prevention and Treatment of Pressure-Overload Induced Heart Failure. Front. Pharmacol. 2020, 11, 602952. [Google Scholar] [CrossRef] [PubMed]
  63. Alallam, B.; Altahhan, S.; Taher, M.; Mohd Nasir, M.H.; Doolaanea, A.A. Electrosprayed Alginate Nanoparticles as CRISPR Plasmid DNA Delivery Carrier: Preparation, Optimization, and Characterization. Pharmaceuticals 2020, 13, 158. [Google Scholar] [CrossRef] [PubMed]
  64. Han, C.; Xiao, Y.; Liu, E.; Su, Z.; Meng, X.; Liu, B. Preparation of Ca-Alginate-Whey Protein Isolate Microcapsules for Protection and Delivery of L. Bulgaricus and L. Paracasei. Int. J. Biol. Macromol. 2020, 163, 1361–1368. [Google Scholar] [CrossRef] [PubMed]
  65. Bossi, A.M.; Bucciarelli, A.; Maniglio, D. Molecularly Imprinted Silk Fibroin Nanoparticles. ACS Appl. Mater. Interfaces 2021, 13, 31431–31439. [Google Scholar] [CrossRef]
  66. Rockwood, D.N.; Preda, R.C.; Yücel, T.; Wang, X.; Lovett, M.L.; Kaplan, D.L. Materials Fabrication from Bombyx Mori Silk Fibroin. Nat. Protoc. 2011, 6, 1612–1631. [Google Scholar] [CrossRef]
  67. Guan, Y.; You, H.; Cai, J.; Zhang, Q.; Yan, S.; You, R. Physically Crosslinked Silk Fibroin/Hyaluronic Acid Scaffolds. Carbohydr. Polym. 2020, 239, 116232. [Google Scholar] [CrossRef]
  68. Lee, J.; Jang, E.H.; Kim, J.H.; Park, S.; Kang, Y.; Park, S.; Lee, K.; Kim, J.-H.; Youn, Y.-N.; Ryu, W. Highly Flexible and Porous Silk Fibroin Microneedle Wraps for Perivascular Drug Delivery. J. Control. Release 2021, 340, 125–135. [Google Scholar] [CrossRef]
  69. Opálková Šišková, A.; Kozma, E.; Opálek, A.; Kroneková, Z.; Kleinová, A.; Nagy, Š.; Kronek, J.; Rydz, J.; Eckstein Andicsová, A. Diclofenac Embedded in Silk Fibroin Fibers as a Drug Delivery System. Materials 2020, 13, 3580. [Google Scholar] [CrossRef]
  70. Ziadlou, R.; Rotman, S.; Teuschl, A.; Salzer, E.; Barbero, A.; Martin, I.; Alini, M.; Eglin, D.; Grad, S. Optimization of Hyaluronic Acid-Tyramine/Silk-Fibroin Composite Hydrogels for Cartilage Tissue Engineering and Delivery of Anti-Inflammatory and Anabolic Drugs. Mater. Sci. Eng. C 2021, 120, 111701. [Google Scholar] [CrossRef]
  71. Crakes, K.R.; Herrera, C.; Morgan, J.L.; Olstad, K.; Hessell, A.J.; Ziprin, P.; LiWang, P.J.; Dandekar, S. Efficacy of Silk Fibroin Biomaterial Vehicle for in Vivo Mucosal Delivery of Griffithsin and Protection against HIV and SHIV Infection Ex Vivo. J. Int. AIDS Soc. 2020, 23, e25628. [Google Scholar] [CrossRef]
  72. Crivelli, B.; Bari, E.; Perteghella, S.; Catenacci, L.; Sorrenti, M.; Mocchi, M.; Faragò, S.; Tripodo, G.; Prina-Mello, A.; Torre, M.L. Silk Fibroin Nanoparticles for Celecoxib and Curcumin Delivery: ROS-Scavenging and Anti-Inflammatory Activities in an in Vitro Model of Osteoarthritis. Eur. J. Pharm. Biopharm. 2019, 137, 37–45. [Google Scholar] [CrossRef] [PubMed]
  73. Laomeephol, C.; Ferreira, H.; Kanokpanont, S.; Neves, N.M.; Kobayashi, H.; Damrongsakkul, S. Dual-Functional Liposomes for Curcumin Delivery and Accelerating Silk Fibroin Hydrogel Formation. Int. J. Pharm. 2020, 589, 119844. [Google Scholar] [CrossRef] [PubMed]
  74. Pérez Quiñones, J.; Roschger, C.; Zierer, A.; Peniche-Covas, C.; Brüggemann, O. Self-Assembled Silk Fibroin-Based Aggregates for Delivery of Camptothecin. Polymers 2021, 13, 3804. [Google Scholar] [CrossRef]
  75. Rahmani, H.; Fattahi, A.; Sadrjavadi, K.; Khaledian, S.; Shokoohinia, Y. Preparation and Characterization of Silk Fibroin Nanoparticles as a Potential Drug Delivery System for 5-Fluorouracil. Adv. Pharm. Bull. 2019, 9, 601–608. [Google Scholar] [CrossRef]
  76. Yavuz, B.; Morgan, J.L.; Herrera, C.; Harrington, K.; Perez-Ramirez, B.; LiWang, P.J.; Kaplan, D.L. Sustained Release Silk Fibroin Discs: Antibody and Protein Delivery for HIV Prevention. J. Control. Release 2019, 301, 1–12. [Google Scholar] [CrossRef] [PubMed]
  77. Teuschl, A.H.; Zipperle, J.; Huber-Gries, C.; Kaplan, D.L. Silk Fibroin Based Carrier System for Delivery of Fibrinogen and Thrombin as Coagulant Supplements. J. Biomed. Mater. Res. A 2017, 105, 687–696. [Google Scholar] [CrossRef]
  78. Zhu, M.; Liu, Y.; Jiang, F.; Cao, J.; Kundu, S.C.; Lu, S. Combined Silk Fibroin Microneedles for Insulin Delivery. ACS Biomater. Sci. Eng. 2020, 6, 3422–3429. [Google Scholar] [CrossRef]
  79. Zhang, H.; Lai, L.; Wang, Y.; Ye, B.; Deng, S.; Ding, A.; Teng, L.; Qiu, L.; Chen, J. Silk Fibroin for CpG Oligodeoxynucleotide Delivery. ACS Biomater. Sci. Eng. 2019, 5, 6082–6088. [Google Scholar] [CrossRef]
  80. Li, T.; Song, X.; Weng, C.; Wang, X.; Wu, J.; Sun, L.; Gong, X.; Zeng, W.-N.; Yang, L.; Chen, C. Enzymatically Crosslinked and Mechanically Tunable Silk Fibroin/Pullulan Hydrogels for Mesenchymal Stem Cells Delivery. Int. J. Biol. Macromol. 2018, 115, 300–307. [Google Scholar] [CrossRef]
  81. Amiri, N.; Ajami, S.; Shahroodi, A.; Jannatabadi, N.; Amiri Darban, S.; Fazly Bazzaz, B.S.; Pishavar, E.; Kalalinia, F.; Movaffagh, J. Teicoplanin-Loaded Chitosan-PEO Nanofibers for Local Antibiotic Delivery and Wound Healing. Int. J. Biol. Macromol. 2020, 162, 645–656. [Google Scholar] [CrossRef]
  82. Bahmanpour, A.; Ghaffari, M.; Milan, P.B.; Moztarzadeh, F.; Mozafari, M. Synthesis and Characterization of Thermosensitive Hydrogel Based on Quaternized Chitosan for Intranasal Delivery of Insulin. Biotechnol. Appl. Biochem. 2020, 68, 247–256. [Google Scholar] [CrossRef] [PubMed]
  83. Esfandiari, F.; Motazedian, M.H.; Asgari, Q.; Morowvat, M.H.; Molaei, M.; Heli, H. Paromomycin-Loaded Mannosylated Chitosan Nanoparticles: Synthesis, Characterization and Targeted Drug Delivery against Leishmaniasis. Acta Trop. 2019, 197, 105072. [Google Scholar] [CrossRef] [PubMed]
  84. Deshkar, S.; Sikchi, S.; Thakre, A.; Kale, R. Poloxamer Modified Chitosan Nanoparticles for Vaginal Delivery of Acyclovir. Pharm. Nanotechnol. 2021, 9, 141–156. [Google Scholar] [CrossRef]
  85. Zaman, M.; Iqbal, A.; Haider Rizvi, S.F.; Hussain, M.A.; Jamshaid, T.; Jamshaid, M. Chitosan Based Controlled Release Drug Delivery of Mycophenolate Mofetil Loaded in Nanocarriers System: Synthesis and in-Vitro Evaluation. Drug Dev. Ind. Pharm. 2021, 47, 477–483. [Google Scholar] [CrossRef] [PubMed]
  86. Panão Costa, J.; Carvalho, S.; Jesus, S.; Soares, E.; Marques, A.P.; Borges, O. Optimization of Chitosan-α-Casein Nanoparticles for Improved Gene Delivery: Characterization, Stability, and Transfection Efficiency. AAPS PharmSciTech 2019, 20, 132. [Google Scholar] [CrossRef] [PubMed]
  87. Soliman, O.Y.; Alameh, M.G.; De Cresenzo, G.; Buschmann, M.D.; Lavertu, M. Efficiency of Chitosan/Hyaluronan-Based MRNA Delivery Systems In Vitro: Influence of Composition and Structure. J. Pharm. Sci. 2020, 109, 1581–1593. [Google Scholar] [CrossRef] [PubMed]
  88. Babii, O.; Wang, Z.; Liu, G.; Martinez, E.C.; van Drunen Littel-van den Hurk, S.; Chen, L. Low Molecular Weight Chitosan Nanoparticles for CpG Oligodeoxynucleotides Delivery: Impact of Molecular Weight, Degree of Deacetylation, and Mannosylation on Intracellular Uptake and Cytokine Induction. Int. J. Biol. Macromol. 2020, 159, 46–56. [Google Scholar] [CrossRef]
  89. Wang, X.; Ronsin, O.; Gravez, B.; Farman, N.; Baumberger, T.; Jaisser, F.; Coradin, T.; Hélary, C. Nanostructured Dense Collagen-Polyester Composite Hydrogels as Amphiphilic Platforms for Drug Delivery. Adv. Sci. 2021, 8, 2004213. [Google Scholar] [CrossRef]
  90. Kupper, S.; Kłosowska-Chomiczewska, I.; Szumała, P. Collagen and Hyaluronic Acid Hydrogel in Water-in-Oil Microemulsion Delivery Systems. Carbohydr. Polym. 2017, 175, 347–354. [Google Scholar] [CrossRef]
  91. Domene, C.; Jorgensen, C.; Abbasi, S.W. A Perspective on Structural and Computational Work on Collagen. Phys. Chem. Chem. Phys. 2016, 18, 24802–24811. [Google Scholar] [CrossRef]
  92. Cheng, O.T.; Stein, A.P.; Babajanian, E.; Hoppe, K.R.; Li, S.; Jung, H.; Abrol, A.; Akkus, A.; Younesi, M.; Altawallbeh, G.; et al. Heparin-Mediated Antibiotic Delivery from an Electrochemically-Aligned Collagen Sheet. Biomed. Mater. Eng. 2021, 32, 159–170. [Google Scholar] [CrossRef] [PubMed]
  93. Zeng, Y.; Zhou, M.; Mou, S.; Yang, J.; Yuan, Q.; Guo, L.; Zhong, A.; Wang, J.; Sun, J.; Wang, Z. Sustained Delivery of Alendronate by Engineered Collagen Scaffold for the Repair of Osteoporotic Bone Defects and Resistance to Bone Loss. J. Biomed. Mater. Res. A 2020, 108, 2460–2472. [Google Scholar] [CrossRef] [PubMed]
  94. Zhang, S.; Huang, D.; Lin, H.; Xiao, Y.; Zhang, X. Cellulose Nanocrystal Reinforced Collagen-Based Nanocomposite Hydrogel with Self-Healing and Stress-Relaxation Properties for Cell Delivery. Biomacromolecules 2020, 21, 2400–2408. [Google Scholar] [CrossRef] [PubMed]
  95. Zhou, X.; Wang, J.; Fang, W.; Tao, Y.; Zhao, T.; Xia, K.; Liang, C.; Hua, J.; Li, F.; Chen, Q. Genipin Cross-Linked Type II Collagen/Chondroitin Sulfate Composite Hydrogel-like Cell Delivery System Induces Differentiation of Adipose-Derived Stem Cells and Regenerates Degenerated Nucleus Pulposus. Acta Biomater. 2018, 71, 496–509. [Google Scholar] [CrossRef]
  96. Anghel, N.; Dinu, M.V.; Zaltariov, M.; Pamfil, D.; Spiridon, I. New Cellulose-Collagen-Alginate Materials Incorporated with Quercetin, Anthocyanins and Lipoic Acid. Int. J. Biol. Macromol. 2021, 181, 30–40. [Google Scholar] [CrossRef]
  97. Qu, Y.; Cao, C.; Wu, Q.; Huang, A.; Song, Y.; Li, H.; Zuo, Y.; Chu, C.; Li, J.; Man, Y. The Dual Delivery of KGF and BFGF by Collagen Membrane to Promote Skin Wound Healing. J. Tissue Eng. Regen. Med. 2018, 12, 1508–1518. [Google Scholar] [CrossRef]
  98. Ruehle, M.A.; Li, M.-T.A.; Cheng, A.; Krishnan, L.; Willett, N.J.; Guldberg, R.E. Decorin-Supplemented Collagen Hydrogels for the Co-Delivery of Bone Morphogenetic Protein-2 and Microvascular Fragments to a Composite Bone-Muscle Injury Model with Impaired Vascularization. Acta Biomater. 2019, 93, 210–221. [Google Scholar] [CrossRef]
  99. Gerton, M.L.; Mann, B.K. Mucoadhesive Hyaluronic Acid-Based Films for Vaginal Delivery of Metronidazole. J. Biomed. Mater. Res. B Appl. Biomater. 2021, 109, 1706–1712. [Google Scholar] [CrossRef]
  100. Du, H.; Liu, P.; Zhu, J.; Lan, J.; Li, Y.; Zhang, L.; Zhu, J.; Tao, J. Hyaluronic Acid-Based Dissolving Microneedle Patch Loaded with Methotrexate for Improved Treatment of Psoriasis. ACS Appl. Mater. Interfaces 2019, 11, 43588–43598. [Google Scholar] [CrossRef]
  101. Camara, C.I.; Bertocchi, L.; Ricci, C.; Bassi, R.; Bianchera, A.; Cantu’, L.; Bettini, R.; Del Favero, E. Hyaluronic Acid-Dexamethasone Nanoparticles for Local Adjunct Therapy of Lung Inflammation. Int. J. Mol. Sci. 2021, 22, 10480. [Google Scholar] [CrossRef]
  102. Cho, J.-A.; Kim, B.-J.; Hwang, Y.-J.; Woo, S.-W.; Noh, T.-S.; Suh, M.-W. Effect and Biocompatibility of a Cross-Linked Hyaluronic Acid and Polylactide-Co-Glycolide Microcapsule Vehicle in Intratympanic Drug Delivery for Treating Acute Acoustic Trauma. Int. J. Mol. Sci. 2021, 22, 5720. [Google Scholar] [CrossRef] [PubMed]
  103. Lai, H.; Ding, X.; Ye, J.; Deng, J.; Cui, S. PH-Responsive Hyaluronic Acid-Based Nanoparticles for Targeted Curcumin Delivery and Enhanced Cancer Therapy. Colloids Surf. B Biointerfaces 2021, 198, 111455. [Google Scholar] [CrossRef] [PubMed]
  104. Zhang, Y.; Zhang, K.; Wang, Z.; Hu, H.; Jing, Q.; Li, Y.; Guo, T.; Feng, N. Transcutol® P/Cremophor® EL/Ethyl Oleate-Formulated Microemulsion Loaded into Hyaluronic Acid-Based Hydrogel for Improved Transdermal Delivery and Biosafety of Ibuprofen. AAPS PharmSciTech 2019, 21, 22. [Google Scholar] [CrossRef]
  105. Liu, D.; Zhang, Q.; Wang, J.; Guan, S.; Cai, D.; Liu, J. Inhibition of Growth and Metastasis of Breast Cancer by Targeted Delivery of 17-Hydroxy-Jolkinolide B via Hyaluronic Acid-Coated Liposomes. Carbohydr. Polym. 2021, 257, 117572. [Google Scholar] [CrossRef] [PubMed]
  106. Rezaei, S.; Kashanian, S.; Bahrami, Y.; Cruz, L.J.; Motiei, M. Redox-Sensitive and Hyaluronic Acid-Functionalized Nanoparticles for Improving Breast Cancer Treatment by Cytoplasmic 17α-Methyltestosterone Delivery. Molecules 2020, 25, 1181. [Google Scholar] [CrossRef]
  107. Sagbas Suner, S.; Ari, B.; Onder, F.C.; Ozpolat, B.; Ay, M.; Sahiner, N. Hyaluronic Acid and Hyaluronic Acid: Sucrose Nanogels for Hydrophobic Cancer Drug Delivery. Int. J. Biol. Macromol. 2019, 126, 1150–1157. [Google Scholar] [CrossRef]
  108. Zhang, Q.; Wang, J.; Liu, D.; Zhu, W.; Guan, S.; Fan, L.; Cai, D. Targeted Delivery of Honokiol by Zein/Hyaluronic Acid Core-Shell Nanoparticles to Suppress Breast Cancer Growth and Metastasis. Carbohydr. Polym. 2020, 240, 116325. [Google Scholar] [CrossRef]
  109. Lei, M.; Chen, G.; Zhang, M.; Lei, J.; Li, T.; Li, D.; Zheng, H. A PH-Sensitive Drug Delivery System Based on Hyaluronic Acid Co-Deliver Doxorubicin and Aminoferrocene for the Combined Application of Chemotherapy and Chemodynamic Therapy. Colloids Surf. B Biointerfaces 2021, 203, 111750. [Google Scholar] [CrossRef]
  110. Li, M.; Li, J.-P.; Wang, Y.-S.; He, Q. [Construction and Anti-tumor Effect Evaluation of a Dual-Responsive Hyaluronic Acid Carbon Quantum Dot-Gelatin Nano-Drug Delivery System]. Sichuan Da Xue Xue Bao Yi Xue Ban 2021, 52, 577–584. [Google Scholar] [CrossRef]
  111. Lu, B.; Xiao, F.; Wang, Z.; Wang, B.; Pan, Z.; Zhao, W.; Zhu, Z.; Zhang, J. Redox-Sensitive Hyaluronic Acid Polymer Prodrug Nanoparticles for Enhancing Intracellular Drug Self-Delivery and Targeted Cancer Therapy. ACS Biomater. Sci. Eng. 2020, 6, 4106–4115. [Google Scholar] [CrossRef]
  112. Zhang, X.; Pan, J.; Yao, M.; Palmerston Mendes, L.; Sarisozen, C.; Mao, S.; Torchilin, V.P. Charge Reversible Hyaluronic Acid-Modified Dendrimer-Based Nanoparticles for SiMDR-1 and Doxorubicin Co-Delivery. Eur. J. Pharm. Biopharm. 2020, 154, 43–49. [Google Scholar] [CrossRef] [PubMed]
  113. Serini, S.; Cassano, R.; Bruni, M.; Servidio, C.; Calviello, G.; Trombino, S. Characterization of a Hyaluronic Acid and Folic Acid-Based Hydrogel for Cisplatin Delivery: Antineoplastic Effect in Human Ovarian Cancer Cells in Vitro. Int. J. Pharm. 2021, 606, 120899. [Google Scholar] [CrossRef] [PubMed]
  114. Sun, S.; Shang, E.; Ju, A.; Li, Y.; Wu, Q.; Li, Q.; Yang, Y.; Guo, Y.; Yang, D.; Lv, S. Tumor-Targeted Hyaluronic Acid-MPEG Modified Nanostructured Lipid Carriers for Cantharidin Delivery: An in Vivo and in Vitro Study. Fitoterapia 2021, 155, 105033. [Google Scholar] [CrossRef]
  115. Kim, Y.; Bhattaccharjee, S.A.; Beck-Broichsitter, M.; Banga, A.K. Fabrication and Characterization of Hyaluronic Acid Microneedlesto Enhance Delivery of Magnesium Ascorbyl Phosphate into Skin. Biomed Microdevices 2019, 21, 104. [Google Scholar] [CrossRef] [PubMed]
  116. Chen, S.; Han, Y.; Wang, Y.; Yang, X.; Sun, C.; Mao, L.; Gao, Y. Zein-Hyaluronic Acid Binary Complex as a Delivery Vehicle of Quercetagetin: Fabrication, Structural Characterization, Physicochemical Stability and in Vitro Release Property. Food Chem. 2019, 276, 322–332. [Google Scholar] [CrossRef] [PubMed]
  117. De Oliveira, M.M.; Nakamura, C.V.; Auzély-Velty, R. Boronate-Ester Crosslinked Hyaluronic Acid Hydrogels for Dihydrocaffeic Acid Delivery and Fibroblasts Protection against UVB Irradiation. Carbohydr. Polym. 2020, 247, 116845. [Google Scholar] [CrossRef]
  118. Liu, Y.; Wang, Y.; Yang, J.; Zhang, H.; Gan, L. Cationized Hyaluronic Acid Coated Spanlastics for Cyclosporine A Ocular Delivery: Prolonged Ocular Retention, Enhanced Corneal Permeation and Improved Tear Production. Int. J. Pharm. 2019, 565, 133–142. [Google Scholar] [CrossRef]
  119. Hamilton, M.; Harrington, S.; Dhar, P.; Stehno-Bittel, L. Hyaluronic Acid Hydrogel Microspheres for Slow Release Stem Cell Delivery. ACS Biomater. Sci. Eng. 2021, 7, 3754–3763. [Google Scholar] [CrossRef]
  120. Liu, F.; Hu, S.; Yang, H.; Li, Z.; Huang, K.; Su, T.; Wang, S.; Cheng, K. Hyaluronic Acid Hydrogel Integrated with Mesenchymal Stem Cell-Secretome to Treat Endometrial Injury in a Rat Model of Asherman’s Syndrome. Adv. Healthc. Mater. 2019, 8, e1900411. [Google Scholar] [CrossRef]
  121. Jenjob, R.; Nguyen, H.-P.; Kim, M.-K.; Jiang, Y.; Kim, J.J.; Yang, S.-G. Bisphosphonate-Conjugated Photo-Crosslinking Polyanionic Hyaluronic Acid Microbeads for Controlled BMP2 Delivery and Enhanced Bone Formation Efficacy. Biomacromolecules 2021, 22, 4138–4145. [Google Scholar] [CrossRef]
  122. Chen, Z.; Han, S.; Yang, X.; Xu, L.; Qi, H.; Hao, G.; Cao, J.; Liang, Y.; Ma, Q.; Zhang, G.; et al. Overcoming Multiple Absorption Barrier for Insulin Oral Delivery Using Multifunctional Nanoparticles Based on Chitosan Derivatives and Hyaluronic Acid. Int. J. Nanomed. 2020, 15, 4877–4898. [Google Scholar] [CrossRef] [PubMed]
  123. Derkach, S.R.; Kolotova, D.S.; Voron’ko, N.G.; Obluchinskaya, E.D.; Malkin, A.Y. Rheological Properties of Fish Gelatin Modified with Sodium Alginate. Polymers 2021, 13, 743. [Google Scholar] [CrossRef]
  124. Dolci, L.S.; Albertini, B.; Di Filippo, M.F.; Bonvicini, F.; Passerini, N.; Panzavolta, S. Development and in Vitro Evaluation of Mucoadhesive Gelatin Films for the Vaginal Delivery of Econazole. Int. J. Pharm. 2020, 591, 119979. [Google Scholar] [CrossRef]
  125. Oliveira, M.B.; da Silva, J.B.; Montanha, M.C.; Kimura, E.; Diniz, A.; Bruschi, M.L. Design and Characterization of Mucoadhesive Gelatin-Ethylcellulose Microparticles for the Delivery of Curcumin to the Bladder. Curr. Drug Deliv. 2018, 15, 1112–1122. [Google Scholar] [CrossRef]
  126. Patel, S.; Srivastava, S.; Singh, M.R.; Singh, D. Preparation and Optimization of Chitosan-Gelatin Films for Sustained Delivery of Lupeol for Wound Healing. Int. J. Biol. Macromol. 2018, 107, 1888–1897. [Google Scholar] [CrossRef] [PubMed]
  127. Zhang, S.; Kang, L.; Hu, S.; Hu, J.; Fu, Y.; Hu, Y.; Yang, X. Carboxymethyl Chitosan Microspheres Loaded Hyaluronic Acid/Gelatin Hydrogels for Controlled Drug Delivery and the Treatment of Inflammatory Bowel Disease. Int. J. Biol. Macromol. 2021, 167, 1598–1612. [Google Scholar] [CrossRef] [PubMed]
  128. Cao, S.; Li, L.; Du, Y.; Gan, J.; Wang, J.; Wang, T.; Liu, Y.; Liu, W.; Zhou, Y.; Gao, X.; et al. Porous Gelatin Microspheres for Controlled Drug Delivery with High Hemostatic Efficacy. Colloids Surf. B Biointerfaces 2021, 207, 112013. [Google Scholar] [CrossRef] [PubMed]
  129. Fan, Z.; Cheng, P.; Yin, G.; Wang, Z.; Han, J. In Situ Forming Oxidized Salecan/Gelatin Injectable Hydrogels for Vancomycin Delivery and 3D Cell Culture. J. Biomater. Sci. Polym. Ed. 2020, 31, 762–780. [Google Scholar] [CrossRef]
  130. Nouri-Felekori, M.; Khakbiz, M.; Nezafati, N.; Mohammadi, J.; Eslaminejad, M.B. Comparative Analysis and Properties Evaluation of Gelatin Microspheres Crosslinked with Glutaraldehyde and 3-Glycidoxypropyltrimethoxysilane as Drug Delivery Systems for the Antibiotic Vancomycin. Int. J. Pharm. 2019, 557, 208–220. [Google Scholar] [CrossRef]
  131. Chen, X.; Fan, M.; Tan, H.; Ren, B.; Yuan, G.; Jia, Y.; Li, J.; Xiong, D.; Xing, X.; Niu, X.; et al. Magnetic and Self-Healing Chitosan-Alginate Hydrogel Encapsulated Gelatin Microspheres via Covalent Cross-Linking for Drug Delivery. Mater. Sci. Eng. C Mater. Biol. Appl. 2019, 101, 619–629. [Google Scholar] [CrossRef]
  132. Jahanban-Esfahlan, R.; Derakhshankhah, H.; Haghshenas, B.; Massoumi, B.; Abbasian, M.; Jaymand, M. A Bio-Inspired Magnetic Natural Hydrogel Containing Gelatin and Alginate as a Drug Delivery System for Cancer Chemotherapy. Int. J. Biol. Macromol. 2020, 156, 438–445. [Google Scholar] [CrossRef] [PubMed]
  133. Vaghasiya, K.; Ray, E.; Singh, R.; Jadhav, K.; Sharma, A.; Khan, R.; Katare, O.P.; Verma, R.K. Efficient, Enzyme Responsive and Tumor Receptor Targeting Gelatin Nanoparticles Decorated with Concanavalin-A for Site-Specific and Controlled Drug Delivery for Cancer Therapy. Mater. Sci. Eng. C Mater. Biol. Appl. 2021, 123, 112027. [Google Scholar] [CrossRef] [PubMed]
  134. Zhou, K.; Zhu, Y.; Chen, X.; Li, L.; Xu, W. Redox- and MMP-2-Sensitive Drug Delivery Nanoparticles Based on Gelatin and Albumin for Tumor Targeted Delivery of Paclitaxel. Mater. Sci. Eng. C Mater. Biol. Appl. 2020, 114, 111006. [Google Scholar] [CrossRef] [PubMed]
  135. Albadran, H.A.; Monteagudo-Mera, A.; Khutoryanskiy, V.V.; Charalampopoulos, D. Development of Chitosan-Coated Agar-Gelatin Particles for Probiotic Delivery and Targeted Release in the Gastrointestinal Tract. Appl. Microbiol. Biotechnol. 2020, 104, 5749–5757. [Google Scholar] [CrossRef] [PubMed]
  136. Demir, G.C.; Erdemli, Ö.; Keskin, D.; Tezcaner, A. Xanthan-Gelatin and Xanthan-Gelatin-Keratin Wound Dressings for Local Delivery of Vitamin C. Int. J. Pharm. 2022, 614, 121436. [Google Scholar] [CrossRef]
  137. Amit, C.; Muralikumar, S.; Janaki, S.; Lakshmipathy, M.; Therese, K.L.; Umashankar, V.; Padmanabhan, P.; Narayanan, J. Designing and Enhancing the Antifungal Activity of Corneal Specific Cell Penetrating Peptide Using Gelatin Hydrogel Delivery System. Int. J. Nanomed. 2019, 14, 605–622. [Google Scholar] [CrossRef]
  138. Galdopórpora, J.M.; Morcillo, M.F.; Ibar, A.; Perez, C.J.; Tuttolomondo, M.V.; Desimone, M.F. Development of Silver Nanoparticles/Gelatin Thermoresponsive Nanocomposites: Characterization and Antimicrobial Activity. Curr. Pharm. Des. 2019, 25, 4121–4129. [Google Scholar] [CrossRef]
  139. Luo, R.; Huang, Y.; Yuan, X.; Yuan, Z.; Zhang, L.; Han, J.; Zhao, Y.; Cai, Q. Controlled Co-Delivery System of Magnesium and Lanthanum Ions for Vascularized Bone Regeneration. Biomed. Mater. 2021, 16, 065024. [Google Scholar] [CrossRef]
  140. Fan, C.; Zhan, S.-H.; Dong, Z.-X.; Yang, W.; Deng, W.-S.; Liu, X.; Wang, D.-A.; Sun, P. Cross-Linked Gelatin Microsphere-Based Scaffolds as a Delivery Vehicle of MC3T3-E1 Cells: In Vitro and in Vivo Evaluation. Mater. Sci. Eng. C Mater. Biol. Appl. 2020, 108, 110399. [Google Scholar] [CrossRef]
  141. Tang, Y.; Tong, X.; Conrad, B.; Yang, F. Injectable and in Situ Crosslinkable Gelatin Microribbon Hydrogels for Stem Cell Delivery and Bone Regeneration in Vivo. Theranostics 2020, 10, 6035–6047. [Google Scholar] [CrossRef]
  142. Yuan, Z.; Yuan, X.; Zhao, Y.; Cai, Q.; Wang, Y.; Luo, R.; Yu, S.; Wang, Y.; Han, J.; Ge, L.; et al. Injectable GelMA Cryogel Microspheres for Modularized Cell Delivery and Potential Vascularized Bone Regeneration. Small 2021, 17, e2006596. [Google Scholar] [CrossRef] [PubMed]
  143. Bai, Y.; Moeinzadeh, S.; Kim, S.; Park, Y.; Lui, E.; Tan, H.; Zhao, W.; Zhou, X.; Yang, Y.P. Development of PLGA-PEG-COOH and Gelatin-Based Microparticles Dual Delivery System and E-Beam Sterilization Effects for Controlled Release of BMP-2 and IGF-1. Part. Part. Syst. Charact. 2020, 37, 2000180. [Google Scholar] [CrossRef] [PubMed]
  144. Yu, J.R.; Janssen, M.; Liang, B.J.; Huang, H.-C.; Fisher, J.P. A Liposome/Gelatin Methacrylate Nanocomposite Hydrogel System for Delivery of Stromal Cell-Derived Factor-1α and Stimulation of Cell Migration. Acta Biomater. 2020, 108, 67–76. [Google Scholar] [CrossRef] [PubMed]
  145. Attarwala, H.Z.; Suri, K.; Amiji, M.M. Pharmacokinetics and Biodistribution Analysis of Small Interference RNA for Silencing Tissue Transglutaminase-2 in Celiac Disease After Oral Administration in Mice Using Gelatin-Based Multicompartmental Delivery Systems. Bioelectricity 2020, 2, 167–174. [Google Scholar] [CrossRef]
  146. Takanche, J.S.; Kim, J.-E.; Kim, J.-S.; Yi, H.-K. Guided Bone Regeneration with a Gelatin Layer and Adenoviral Delivery of C-Myb Enhances Bone Healing in Rat Tibia. Regen. Med. 2020, 15, 1877–1890. [Google Scholar] [CrossRef]
  147. Negishi, J.; Funamoto, S.; Hashimoto, Y.; Yanagisawa, K. PLA-Collagen Composite Scaffold Fabrication by Vacuum Pressure Impregnation. Tissue Eng. Part C Methods 2019, 25, 742–747. [Google Scholar] [CrossRef]
  148. Phelan, M.A.; Kruczek, K.; Wilson, J.H.; Brooks, M.J.; Drinnan, C.T.; Regent, F.; Gerstenhaber, J.A.; Swaroop, A.; Lelkes, P.I.; Li, T. Soy Protein Nanofiber Scaffolds for Uniform Maturation of Human Induced Pluripotent Stem Cell-Derived Retinal Pigment Epithelium. Tissue Eng. Part C Methods 2020, 26, 433–446. [Google Scholar] [CrossRef]
  149. Galler, K.M.; Brandl, F.P.; Kirchhof, S.; Widbiller, M.; Eidt, A.; Buchalla, W.; Göpferich, A.; Schmalz, G. Suitability of Different Natural and Synthetic Biomaterials for Dental Pulp Tissue Engineering. Tissue Eng. Part A 2018, 24, 234–244. [Google Scholar] [CrossRef]
  150. Sharma, P.K.; Singh, Y. Glyoxylic Hydrazone Linkage-Based PEG Hydrogels for Covalent Entrapment and Controlled Delivery of Doxorubicin. Biomacromolecules 2019, 20, 2174–2184. [Google Scholar] [CrossRef]
  151. Chai, D.; Hao, B.; Hu, R.; Zhang, F.; Yan, J.; Sun, Y.; Huang, X.; Zhang, Q.; Jiang, H. Delivery of Oridonin and Methotrexate via PEGylated Graphene Oxide. ACS Appl. Mater. Interfaces 2019, 11, 22915–22924. [Google Scholar] [CrossRef]
  152. Emami, J.; Maghzi, P.; Hasanzadeh, F.; Sadeghi, H.; Mirian, M.; Rostami, M. PLGA-PEG-RA-Based Polymeric Micelles for Tumor Targeted Delivery of Irinotecan. Pharm. Dev. Technol. 2018, 23, 41–54. [Google Scholar] [CrossRef] [PubMed]
  153. Gong, Y.-H.; Shu, M.; Xie, J.-H.; Zhang, C.; Cao, Z.; Jiang, Z.-Z.; Liu, J. Enzymatic Synthesis of PEG-Poly(Amine-Co-Thioether Esters) as Highly Efficient PH and ROS Dual-Responsive Nanocarriers for Anticancer Drug Delivery. J. Mater. Chem. B 2019, 7, 651–664. [Google Scholar] [CrossRef] [PubMed]
  154. Manjili, H.K.; Malvandi, H.; Mousavi, M.S.; Attari, E.; Danafar, H. In Vitro and in Vivo Delivery of Artemisinin Loaded PCL-PEG-PCL Micelles and Its Pharmacokinetic Study. Artif. Cells Nanomed. Biotechnol. 2018, 46, 926–936. [Google Scholar] [CrossRef] [PubMed]
  155. Nasab, S.H.; Amani, A.; Ebrahimi, H.A.; Hamidi, A.A. Design and Preparation of a New Multi-Targeted Drug Delivery System Using Multifunctional Nanoparticles for Co-Delivery of SiRNA and Paclitaxel. J. Pharm. Anal. 2021, 11, 163–173. [Google Scholar] [CrossRef]
  156. Nguyen, P.T.H.; Le, B.T.; Ninh, H.D.; La, D.D. Ultrasonic-Assisted Synthesis of Fe–BTC–PEG Metal–Organic Complex: An Effective and Safety Nanocarrier for Anticancer Drug Delivery. ACS Omega 2021, 6, 33419–33427. [Google Scholar] [CrossRef] [PubMed]
  157. Ni, R.; Duan, D.; Li, B.; Li, Z.; Li, L.; Ming, Y.; Wang, X.; Chen, J. Dual-Modified PCL-PEG Nanoparticles for Improved Targeting and Therapeutic Efficacy of Docetaxel against Colorectal Cancer. Pharm. Dev. Technol. 2021, 26, 910–921. [Google Scholar] [CrossRef]
  158. Wu, J.; Wang, X.; Zhu, B.; He, Q.; Ren, F.; Tong, F.; Jiang, W.; He, X. PH-Sensitive Magnetic Drug Delivery System via Layer-by-Layer Self-Assembly of CS/PEG and Its Controlled Release of DOX. J. Biomater. Sci. Polym. Ed. 2020, 31, 1057–1070. [Google Scholar] [CrossRef]
  159. Yang, J.-G.; Zhang, J.; Chen, X.-J.; Zhou, G. Stable Loading and Delivery of Icaritin Using PEG-PCL Micelles for Effective Treatment of Oral Squamous Cell Carcinoma. Curr. Drug. Deliv. 2021, 18, 975–983. [Google Scholar] [CrossRef]
  160. Alami-Milani, M.; Zakeri-Milani, P.; Valizadeh, H.; Salehi, R.; Jelvehgari, M. Preparation and Evaluation of PCL-PEG-PCL Micelles as Potential Nanocarriers for Ocular Delivery of Dexamethasone. Iran. J. Basic Med. Sci. 2018, 21, 153–164. [Google Scholar] [CrossRef]
  161. Sharma, P.K.; Halder, M.; Srivastava, U.; Singh, Y. Antibacterial PEG-Chitosan Hydrogels for Controlled Antibiotic/Protein Delivery. ACS Appl. Bio Mater. 2019, 2, 5313–5322. [Google Scholar] [CrossRef]
  162. Singh, S.; Alrobaian, M.M.; Molugulu, N.; Agrawal, N.; Numan, A.; Kesharwani, P. Pyramid-Shaped PEG-PCL-PEG Polymeric-Based Model Systems for Site-Specific Drug Delivery of Vancomycin with Enhance Antibacterial Efficacy. ACS Omega 2020, 5, 11935–11945. [Google Scholar] [CrossRef] [PubMed]
  163. Yamashita, S.; Katsumi, H.; Sakane, T.; Yamamoto, A. Bone-Targeting Dendrimer for the Delivery of Methotrexate and Treatment of Bone Metastasis. J. Drug Target. 2018, 26, 818–828. [Google Scholar] [CrossRef] [PubMed]
  164. Zhang, Y.; Lane, M.E.; Moore, D.J. An Investigation of the Influence of PEG 400 and PEG-6-Caprylic/Capric Glycerides on Dermal Delivery of Niacinamide. Polymers 2020, 12, 2907. [Google Scholar] [CrossRef] [PubMed]
  165. Zhang, M.; Xu, C.; Liu, D.; Han, M.K.; Wang, L.; Merlin, D. Oral Delivery of Nanoparticles Loaded With Ginger Active Compound, 6-Shogaol, Attenuates Ulcerative Colitis and Promotes Wound Healing in a Murine Model of Ulcerative Colitis. J. Crohn’s Colitis 2018, 12, 217–229. [Google Scholar] [CrossRef]
  166. Tan, X.; Yin, N.; Liu, Z.; Sun, R.; Gou, J.; Yin, T.; Zhang, Y.; He, H.; Tang, X. Hydrophilic and Electroneutral Nanoparticles to Overcome Mucus Trapping and Enhance Oral Delivery of Insulin. Mol. Pharm. 2020, 17, 3177–3191. [Google Scholar] [CrossRef]
  167. Ghuman, H.; Matta, R.; Tompkins, A.; Nitzsche, F.; Badylak, S.F.; Gonzalez, A.L.; Modo, M. ECM Hydrogel Improves the Delivery of PEG Microsphere-Encapsulated Neural Stem Cells and Endothelial Cells into Tissue Cavities Caused by Stroke. Brain Res. Bull. 2021, 168, 120–137. [Google Scholar] [CrossRef]
  168. Amani, A.; Kabiri, T.; Shafiee, S.; Hamidi, A. Preparation and Characterization of PLA-PEG-PLA/PEI/DNA Nanoparticles for Improvement of Transfection Efficiency and Controlled Release of DNA in Gene Delivery Systems. Iran. J. Pharm. Res. 2019, 18, 125–141. [Google Scholar]
  169. Poudel, S.; Napit, P.R.; Briski, K.P.; Mattheolabakis, G. Oral Delivery of Nucleic Acids with Passive and Active Targeting to the Intestinal Tissue Using Polymer-Based Nanocarriers. Pharmaceutics 2021, 13, 1075. [Google Scholar] [CrossRef]
  170. Yu, M.; Wang, K.; Zhang, H.; Liu, Q.; Wang, J.; Cao, L.; Li, W.; Wang, K.; Hong, Z. DOTAP-Incorporated PEG-PLGA Nanoparticles for Efficient In Vitro and In Vivo Gene Delivery. J. Biomed. Nanotechnol. 2018, 14, 281–293. [Google Scholar] [CrossRef]
  171. Jiang, T.; Liu, S.; Wu, Z.; Li, Q.; Ren, S.; Chen, J.; Xu, X.; Wang, C.; Lu, C.; Yang, X.; et al. ADSC-Exo@MMP-PEG Smart Hydrogel Promotes Diabetic Wound Healing by Optimizing Cellular Functions and Relieving Oxidative Stress. Mater. Today Bio 2022, 16, 100365. [Google Scholar] [CrossRef]
  172. Mohamady Hussein, M.A.; Guler, E.; Rayaman, E.; Cam, M.E.; Sahin, A.; Grinholc, M.; Sezgin Mansuroglu, D.; Sahin, Y.M.; Gunduz, O.; Muhammed, M.; et al. Dual-Drug Delivery of Ag-Chitosan Nanoparticles and Phenytoin via Core-Shell PVA/PCL Electrospun Nanofibers. Carbohydr. Polym. 2021, 270, 118373. [Google Scholar] [CrossRef] [PubMed]
  173. Zavan, B.; Gardin, C.; Guarino, V.; Rocca, T.; Cruz Maya, I.; Zanotti, F.; Ferroni, L.; Brunello, G.; Chachques, J.-C.; Ambrosio, L.; et al. Electrospun PCL-Based Vascular Grafts: In Vitro Tests. Nanomaterials 2021, 11, 751. [Google Scholar] [CrossRef] [PubMed]
  174. Brandt, J.V.; Piazza, R.D.; Dos Santos, C.C.; Vega-Chacón, J.; Amantéa, B.E.; Pinto, G.C.; Magnani, M.; Piva, H.L.; Tedesco, A.C.; Primo, F.L.; et al. Synthesis and Colloidal Characterization of Folic Acid-Modified PEG-b-PCL Micelles for Methotrexate Delivery. Colloids Surf. B Biointerfaces 2019, 177, 228–234. [Google Scholar] [CrossRef]
  175. De Lima, J.M.; Castellano, L.R.C.; Bonan, P.R.F.; de Medeiros, E.S.; Hier, M.; Bijian, K.; Alaoui-Jamali, M.A.; da Cruz Perez, D.E.; da Silva, S.D. Chitosan/PCL Nanoparticles Can Improve Anti-Neoplastic Activity of 5-Fluorouracil in Head and Neck Cancer through Autophagy Activation. Int. J. Biochem. Cell Biol. 2021, 134, 105964. [Google Scholar] [CrossRef] [PubMed]
  176. Hassankhani Rad, A.; Asiaee, F.; Jafari, S.; Shayanfar, A.; Lavasanifar, A.; Molavi, O. Poly(Ethylene Glycol)-Poly(ε-Caprolactone)-Based Micelles for Solubilization and Tumor-Targeted Delivery of Silibinin. Bioimpacts 2020, 10, 87–95. [Google Scholar] [CrossRef]
  177. Jalilzadeh, N.; Samadi, N.; Salehi, R.; Dehghan, G.; Iranshahi, M.; Dadpour, M.R.; Hamishehkar, H. Novel Nano-Vehicle for Delivery and Efficiency of Anticancer Auraptene against Colon Cancer Cells. Sci. Rep. 2020, 10, 1606. [Google Scholar] [CrossRef]
  178. Rezvani, M.; Mohammadnejad, J.; Narmani, A.; Bidaki, K. Synthesis and in Vitro Study of Modified Chitosan-Polycaprolactam Nanocomplex as Delivery System. Int. J. Biol. Macromol. 2018, 113, 1287–1293. [Google Scholar] [CrossRef]
  179. Yu, T.; Wu, C.; Zhu, C.; He, Y.; Yang, D.; Cheng, Y.; Gao, X. Oral Administration of Liposome-Apatinib and Locally Delivery of Docetaxel/MPEG-PCL by Fibrin Glue Synergistically Improve Therapeutic Effect in Colorectal Cancer. J. Biomed. Nanotechnol. 2018, 14, 2077–2091. [Google Scholar] [CrossRef]
  180. Mirzaeei, S.; Mansurian, M.; Asare-Addo, K.; Nokhodchi, A. Metronidazole- and Amoxicillin-Loaded PLGA and PCL Nanofibers as Potential Drug Delivery Systems for the Treatment of Periodontitis: In Vitro and In Vivo Evaluations. Biomedicines 2021, 9, 975. [Google Scholar] [CrossRef]
  181. Ramazani, A.; Keramati, M.; Malvandi, H.; Danafar, H.; Kheiri Manjili, H. Preparation and in Vivo Evaluation of Anti-Plasmodial Properties of Artemisinin-Loaded PCL-PEG-PCL Nanoparticles. Pharm. Dev. Technol. 2018, 23, 911–920. [Google Scholar] [CrossRef]
  182. Al-Lawati, H.; Vakili, M.R.; Lavasanifar, A.; Ahmed, S.; Jamali, F. Delivery and Biodistribution of Traceable Polymeric Micellar Diclofenac in the Rat. J. Pharm. Sci. 2019, 108, 2698–2707. [Google Scholar] [CrossRef] [PubMed]
  183. Samy, K.E.; Cao, Y.; Kim, J.; Konichi da Silva, N.R.; Phone, A.; Bloomer, M.M.; Bhisitkul, R.B.; Desai, T.A. Co-Delivery of Timolol and Brimonidine with a Polymer Thin-Film Intraocular Device. J. Ocul. Pharmacol. Ther. 2019, 35, 124–131. [Google Scholar] [CrossRef] [PubMed]
  184. Kurd, M.; Sadegh Malvajerd, S.; Rezaee, S.; Hamidi, M.; Derakhshandeh, K. Oral Delivery of Indinavir Using MPEG-PCL Nanoparticles: Preparation, Optimization, Cellular Uptake, Transport and Pharmacokinetic Evaluation. Artif. Cells Nanomed. Biotechnol. 2019, 47, 2123–2133. [Google Scholar] [CrossRef]
  185. Danafar, H.; Jaberizadeh, H.; Andalib, S. In Vitro and in Vivo Delivery of Gliclazide Loaded MPEG-PCL Micelles and Its Kinetic Release and Solubility Study. Artif. Cells Nanomed. Biotechnol. 2018, 46, 1625–1636. [Google Scholar] [CrossRef] [PubMed]
  186. Dziadek, M.; Dziadek, K.; Checinska, K.; Zagrajczuk, B.; Golda-Cepa, M.; Brzychczy-Wloch, M.; Menaszek, E.; Kopec, A.; Cholewa-Kowalska, K. PCL and PCL/Bioactive Glass Biomaterials as Carriers for Biologically Active Polyphenolic Compounds: Comprehensive Physicochemical and Biological Evaluation. Bioact. Mater. 2020, 6, 1811–1826. [Google Scholar] [CrossRef] [PubMed]
  187. Li, L.; Gatto, G.J.; Brand, R.M.; Krovi, S.A.; Cottrell, M.L.; Norton, C.; van der Straten, A.; Johnson, L.M. Long-Acting Biodegradable Implant for Sustained Delivery of Antiretrovirals (ARVs) and Hormones. J. Control. Release 2021, 340, 188–199. [Google Scholar] [CrossRef] [PubMed]
  188. Khodaei, M.; Rostamizadeh, K.; Taromchi, A.H.; Monirinasab, H.; Fathi, M. DDAB Cationic Lipid-MPEG, PCL Copolymer Hybrid Nano-Carrier Synthesis and Application for Delivery of SiRNA Targeting IGF-1R into Breast Cancer Cells. Clin. Transl. Oncol. 2021, 23, 1167–1178. [Google Scholar] [CrossRef]
  189. Zhang, H.-T.; Yu, M.; Niu, Y.-J.; Liu, W.-Z.; Pang, W.-H.; Ding, J.; Wang, J.-C. Polyarginine-Mediated SiRNA Delivery: A Mechanistic Study of Intracellular Trafficking of PCL-R15/SiRNA Nanoplexes. Mol. Pharm. 2020, 17, 1685–1696. [Google Scholar] [CrossRef]
  190. Kong, D.; Shi, Y.; Gao, Y.; Fu, M.; Kong, S.; Lin, G. Preparation of BMP-2 Loaded MPEG-PCL Microspheres and Evaluation of Their Bone Repair Properties. Biomed. Pharmacother. 2020, 130, 110516. [Google Scholar] [CrossRef]
  191. Chen, M.; Feng, Z.; Guo, W.; Yang, D.; Gao, S.; Li, Y.; Shen, S.; Yuan, Z.; Huang, B.; Zhang, Y.; et al. PCL-MECM-Based Hydrogel Hybrid Scaffolds and Meniscal Fibrochondrocytes Promote Whole Meniscus Regeneration in a Rabbit Meniscectomy Model. ACS Appl. Mater. Interfaces 2019, 11, 41626–41639. [Google Scholar] [CrossRef]
  192. Qian, Y.; Zhou, X.; Zhang, F.; Diekwisch, T.G.H.; Luan, X.; Yang, J. Triple PLGA/PCL Scaffold Modification Including Silver-Impregnation, Collagen-Coating, and Electrospinning Significantly Improve Biocompatibility, Antimicrobial, and Osteogenic Properties for Oro-Facial Tissue Regeneration. ACS Appl. Mater. Interfaces 2019, 11, 37381–37396. [Google Scholar] [CrossRef] [PubMed]
  193. Stromberg, Z.R.; Lisa Phipps, M.; Magurudeniya, H.D.; Pedersen, C.A.; Rajale, T.; Sheehan, C.J.; Courtney, S.J.; Bradfute, S.B.; Hraber, P.; Rush, M.N.; et al. Formulation of Stabilizer-Free, Nontoxic PLGA and Elastin-PLGA Nanoparticle Delivery Systems. Int. J. Pharm. 2021, 597, 120340. [Google Scholar] [CrossRef] [PubMed]
  194. Anversa Dimer, F.; de Souza Carvalho-Wodarz, C.; Goes, A.; Cirnski, K.; Herrmann, J.; Schmitt, V.; Pätzold, L.; Abed, N.; De Rossi, C.; Bischoff, M.; et al. PLGA Nanocapsules Improve the Delivery of Clarithromycin to Kill Intracellular Staphylococcus Aureus and Mycobacterium Abscessus. Nanomedicine 2020, 24, 102125. [Google Scholar] [CrossRef] [PubMed]
  195. Gebreel, R.M.; Edris, N.A.; Elmofty, H.M.; Tadros, M.I.; El-Nabarawi, M.A.; Hassan, D.H. Development and Characterization of PLGA Nanoparticle-Laden Hydrogels for Sustained Ocular Delivery of Norfloxacin in the Treatment of Pseudomonas Keratitis: An Experimental Study. Drug Des. Dev. Ther. 2021, 15, 399–418. [Google Scholar] [CrossRef]
  196. Govoni, M.; Lamparelli, E.P.; Ciardulli, M.C.; Santoro, A.; Oliviero, A.; Palazzo, I.; Reverchon, E.; Vivarelli, L.; Maso, A.; Storni, E.; et al. Demineralized Bone Matrix Paste Formulated with Biomimetic PLGA Microcarriers for the Vancomycin Hydrochloride Controlled Delivery: Release Profile, Citotoxicity and Efficacy against S. Aureus. Int. J. Pharm. 2020, 582, 119322. [Google Scholar] [CrossRef]
  197. Jadidi, A.; Salahinejad, E.; Sharifi, E.; Tayebi, L. Drug-Delivery Ca-Mg Silicate Scaffolds Encapsulated in PLGA. Int. J. Pharm. 2020, 589, 119855. [Google Scholar] [CrossRef]
  198. Sun, M.; Zhu, C.; Long, J.; Lu, C.; Pan, X.; Wu, C. PLGA Microsphere-Based Composite Hydrogel for Dual Delivery of Ciprofloxacin and Ginsenoside Rh2 to Treat Staphylococcus Aureus-Induced Skin Infections. Drug Deliv. 2020, 27, 632–641. [Google Scholar] [CrossRef]
  199. Allavena, P.; Palmioli, A.; Avigni, R.; Sironi, M.; La Ferla, B.; Maeda, A. PLGA Based Nanoparticles for the Monocyte-Mediated Anti-Tumor Drug Delivery System. J. Biomed. Nanotechnol. 2020, 16, 212–223. [Google Scholar] [CrossRef]
  200. Chung, K.; Ullah, I.; Kim, N.; Lim, J.; Shin, J.; Lee, S.C.; Jeon, S.; Kim, S.H.; Kumar, P.; Lee, S.-K. Intranasal Delivery of Cancer-Targeting Doxorubicin-Loaded PLGA Nanoparticles Arrests Glioblastoma Growth. J. Drug Target. 2020, 28, 617–626. [Google Scholar] [CrossRef]
  201. Nguyen, H.X.; Banga, A.K. Delivery of Methotrexate and Characterization of Skin Treated by Fabricated PLGA Microneedles and Fractional Ablative Laser. Pharm. Res. 2018, 35, 68. [Google Scholar] [CrossRef]
  202. Trujillo-Nolasco, R.M.; Morales-Avila, E.; Ocampo-García, B.E.; Ferro-Flores, G.; Gibbens-Bandala, B.V.; Escudero-Castellanos, A.; Isaac-Olive, K. Preparation and in Vitro Evaluation of Radiolabeled HA-PLGA Nanoparticles as Novel MTX Delivery System for Local Treatment of Rheumatoid Arthritis. Mater. Sci. Eng. C Mater. Biol. Appl. 2019, 103, 109766. [Google Scholar] [CrossRef] [PubMed]
  203. Yao, W.; Yao, J.; Qian, F.; Que, Z.; Yu, P.; Luo, T.; Zheng, D.; Zhang, Z.; Tian, J. Paclitaxel-Loaded and Folic Acid-Modified PLGA Nanomedicine with Glutathione Response for the Treatment of Lung Cancer. Acta Biochim. Biophys. Sin. 2021, 53, 1027–1036. [Google Scholar] [CrossRef] [PubMed]
  204. Yin, Y.; Wang, J.; Yang, M.; Du, R.; Pontrelli, G.; McGinty, S.; Wang, G.; Yin, T.; Wang, Y. Penetration of the Blood-Brain Barrier and the Anti-Tumour Effect of a Novel PLGA-LysoGM1/DOX Micelle Drug Delivery System. Nanoscale 2020, 12, 2946–2960. [Google Scholar] [CrossRef]
  205. Chatzitaki, A.-T.; Jesus, S.; Karavasili, C.; Andreadis, D.; Fatouros, D.G.; Borges, O. Chitosan-Coated PLGA Nanoparticles for the Nasal Delivery of Ropinirole Hydrochloride: In Vitro and Ex Vivo Evaluation of Efficacy and Safety. Int. J. Pharm. 2020, 589, 119776. [Google Scholar] [CrossRef] [PubMed]
  206. Nigam, K.; Kaur, A.; Tyagi, A.; Nematullah, M.; Khan, F.; Gabrani, R.; Dang, S. Nose-to-Brain Delivery of Lamotrigine-Loaded PLGA Nanoparticles. Drug Deliv. Transl. Res. 2019, 9, 879–890. [Google Scholar] [CrossRef]
  207. Sun, Y.; Long, D. Preparation, Characterization and in Vitro/in Vivo Evaluation of Lovastatin-Loaded PLGA Microspheres by Local Administration for Femoral Head Necrosis. Drug Des. Dev. Ther. 2021, 15, 601–610. [Google Scholar] [CrossRef]
  208. Saffari, T.M.; Chan, K.; Saffari, S.; Zuo, K.J.; McGovern, R.M.; Reid, J.M.; Borschel, G.H.; Shin, A.Y. Combined Local Delivery of Tacrolimus and Stem Cells in Hydrogel for Enhancing Peripheral Nerve Regeneration. Biotechnol. Bioeng. 2021, 118, 2804–2814. [Google Scholar] [CrossRef]
  209. Pan, S.; Qi, Z.; Li, Q.; Ma, Y.; Fu, C.; Zheng, S.; Kong, W.; Liu, Q.; Yang, X. Graphene Oxide-PLGA Hybrid Nanofibres for the Local Delivery of IGF-1 and BDNF in Spinal Cord Repair. Artif. Cells Nanomed. Biotechnol. 2019, 47, 651–664. [Google Scholar] [CrossRef]
  210. Attias Cohen, S.; Kingma, P.S.; Whitsett, J.A.; Goldbart, R.; Traitel, T.; Kost, J. SP-D Loaded PLGA Nanoparticles as Drug Delivery System for Prevention and Treatment of Premature Infant’s Lung Diseases. Int. J. Pharm. 2020, 585, 119387. [Google Scholar] [CrossRef]
  211. Mahmoud, M.Y.; Sapare, S.; Curry, K.C.; Demuth, D.R.; Steinbach-Rankins, J.M. Rapid Release Polymeric Fibers for Inhibition of Porphyromonas Gingivalis Adherence to Streptococcus Gordonii. Front. Chem. 2019, 7, 926. [Google Scholar] [CrossRef]
  212. Song, Y.; Shi, Y.; Zhang, L.; Hu, H.; Zhang, C.; Yin, M.; Chu, L.; Yan, X.; Zhao, M.; Zhang, X.; et al. Synthesis of CSK-DEX-PLGA Nanoparticles for the Oral Delivery of Exenatide to Improve Its Mucus Penetration and Intestinal Absorption. Mol. Pharm. 2019, 16, 518–532. [Google Scholar] [CrossRef] [PubMed]
  213. Mohammadpour, F.; Hadizadeh, F.; Tafaghodi, M.; Sadri, K.; Mohammadpour, A.H.; Kalani, M.R.; Gholami, L.; Mahmoudi, A.; Chamani, J. Preparation, in Vitro and in Vivo Evaluation of PLGA/Chitosan Based Nano-Complex as a Novel Insulin Delivery Formulation. Int. J. Pharm. 2019, 572, 118710. [Google Scholar] [CrossRef] [PubMed]
  214. Wang, W.; Yu, C.; Zhang, F.; Li, Y.; Zhang, B.; Huang, J.; Zhang, Z.; Jin, L. Improved Oral Delivery of Insulin by PLGA Nanoparticles Coated with 5β-Cholanic Acid Conjugated Glycol Chitosan. Biomed. Mater. 2021, 16, 064103. [Google Scholar] [CrossRef] [PubMed]
  215. Jo, A.; Ringel-Scaia, V.M.; McDaniel, D.K.; Thomas, C.A.; Zhang, R.; Riffle, J.S.; Allen, I.C.; Davis, R.M. Fabrication and Characterization of PLGA Nanoparticles Encapsulating Large CRISPR-Cas9 Plasmid. J. Nanobiotechnol. 2020, 18, 16. [Google Scholar] [CrossRef] [PubMed]
  216. Zhu, J.; Qin, F.; Ji, Z.; Fei, W.; Tan, Z.; Hu, Y.; Zheng, C. Mannose-Modified PLGA Nanoparticles for Sustained and Targeted Delivery in Hepatitis B Virus Immunoprophylaxis. AAPS PharmSciTech 2019, 21, 13. [Google Scholar] [CrossRef]
  217. Ramesh, B.; Cherian, K.M.; Fakoya, A.O.J. Fabrication and Electrospinning of 3D Biodegradable Poly-l-Lactic Acid (PLLA) Nanofibers for Clinical Application. In Stem Cell Nanotechnology: Methods and Protocols; Turksen, K., Ed.; Methods in Molecular Biology; Springer: New York, NY, USA, 2020; pp. 119–128. [Google Scholar] [CrossRef]
  218. Ghaeini-Hesaroeiye, S.; Boddohi, S.; Vasheghani-Farahani, E. Dual Responsive Chondroitin Sulfate Based Nanogel for Antimicrobial Peptide Delivery. Int. J. Biol. Macromol. 2020, 143, 297–304. [Google Scholar] [CrossRef]
  219. He, J.; Hu, X.; Cao, J.; Zhang, Y.; Xiao, J.; Peng, L.; Chen, D.; Xiong, C.; Zhang, L. Chitosan-Coated Hydroxyapatite and Drug-Loaded Polytrimethylene Carbonate/Polylactic Acid Scaffold for Enhancing Bone Regeneration. Carbohydr. Polym. 2021, 253, 117198. [Google Scholar] [CrossRef]
  220. Kiani, M.H.; Ali, S.; Qadry, A.; Arshad, R.; Aslam, A.; Shahnaz, G. Polyethylene Imine Conjugated Supramolecular Stereocomplexed Nanomicelles for Intracellular Delivery of Rifampicin against Mycobacterium Bovis. Colloids Surf. B Biointerfaces 2021, 206, 111976. [Google Scholar] [CrossRef]
  221. Lim, D.-J.; McCormick, J.; Skinner, D.; Zhang, S.; Elder, J.B.; McLemore, J.G.; Allen, M.; West, J.M.; Grayson, J.W.; Rowe, S.M.; et al. Controlled Delivery of Ciprofloxacin and Ivacaftor via Sinus Stent in a Preclinical Model of Pseudomonas Sinusitis. Int. Forum Allergy Rhinol. 2020, 10, 481–488. [Google Scholar] [CrossRef]
  222. Cardoso, M.M.; Peca, I.N.; Lopes, T.; Gardner, R.; Bicho, A. Double-Walled Poly-(D,L-Lactide-Co-Glycolide) (PLGA) and Poly(L-Lactide) (PLLA) Nanoparticles for the Sustained Release of Doxorubicin. Polymers 2021, 13, 3230. [Google Scholar] [CrossRef]
  223. Tan, W.; Gao, C.; Feng, P.; Liu, Q.; Liu, C.; Wang, Z.; Deng, Y.; Shuai, C. Dual-Functional Scaffolds of Poly(L-Lactic Acid)/Nanohydroxyapatite Encapsulated with Metformin: Simultaneous Enhancement of Bone Repair and Bone Tumor Inhibition. Mater. Sci. Eng. C Mater. Biol. Appl. 2021, 120, 111592. [Google Scholar] [CrossRef] [PubMed]
  224. Tu, H.; Dai, F.; Cheng, G.; Yuan, M.; Zhou, X.; Wang, Y.; Zhang, R.; Zheng, Y.; Cheng, Y.; Deng, H. Incorporation of Layered Rectorite into Biocompatible Core-Sheath Nanofibrous Mats for Sustained Drug Delivery. ACS Biomater. Sci. Eng. 2021, 7, 4509–4520. [Google Scholar] [CrossRef] [PubMed]
  225. Wang, Y.; Zhu, P.; Li, G.; Zhu, S.; Liu, K.; Liu, Y.; He, J.; Lei, J. Amphiphilic Carboxylated Cellulose-g-Poly(l-Lactide) Copolymer Nanoparticles for Oleanolic Acid Delivery. Carbohydr. Polym. 2019, 214, 100–109. [Google Scholar] [CrossRef] [PubMed]
  226. Wang, Y.; Cui, S.; Wu, B.; Zhang, Q.; Jiang, W. PLA-Based Core-Shell Structure Stereocomplexed Nanoparticles with Enhanced Loading and Release Profile of Paclitaxel. Front. Biosci. (Landmark Ed.) 2021, 26, 517–532. [Google Scholar] [CrossRef]
  227. Birhanu, G.; Tanha, S.; Akbari Javar, H.; Seyedjafari, E.; Zandi-Karimi, A.; Kiani Dehkordi, B. Dexamethasone Loaded Multi-Layer Poly-l-Lactic Acid/Pluronic P123 Composite Electrospun Nanofiber Scaffolds for Bone Tissue Engineering and Drug Delivery. Pharm. Dev. Technol. 2019, 24, 338–347. [Google Scholar] [CrossRef]
  228. Lugasi, L.; Grinberg, I.; Rudnick-Glick, S.; Okun, E.; Einat, H.; Margel, S. Designed Proteinoid Polymers and Nanoparticles Encapsulating Risperidone for Enhanced Antipsychotic Activity. J. Nanobiotechnol. 2020, 18, 149. [Google Scholar] [CrossRef]
  229. Nanaki, S.G.; Spyrou, K.; Bekiari, C.; Veneti, P.; Baroud, T.N.; Karouta, N.; Grivas, I.; Papadopoulos, G.C.; Gournis, D.; Bikiaris, D.N. Hierarchical Porous Carbon-PLLA and PLGA Hybrid Nanoparticles for Intranasal Delivery of Galantamine for Alzheimer’s Disease Therapy. Pharmaceutics 2020, 12, 227. [Google Scholar] [CrossRef]
  230. Liu, W.; Lee, B.-S.; Mieler, W.F.; Kang-Mieler, J.J. Biodegradable Microsphere-Hydrogel Ocular Drug Delivery System for Controlled and Extended Release of Bioactive Aflibercept In Vitro. Curr. Eye Res. 2019, 44, 264–274. [Google Scholar] [CrossRef]
  231. Rudeen, K.M.; Liu, W.; Mieler, W.F.; Kang-Mieler, J.J. Simultaneous Release of Aflibercept and Dexamethasone from an Ocular Drug Delivery System. Curr. Eye Res. 2022, 47, 1034–1042. [Google Scholar] [CrossRef]
  232. Chung, H.-J.; Kim, J.-T.; Kim, H.-J.; Kyung, H.-W.; Katila, P.; Lee, J.-H.; Yang, T.-H.; Yang, Y.-I.; Lee, S.-J. Epicardial Delivery of VEGF and Cardiac Stem Cells Guided by 3-Dimensional PLLA Mat Enhancing Cardiac Regeneration and Angiogenesis in Acute Myocardial Infarction. J. Control. Release 2015, 205, 218–230. [Google Scholar] [CrossRef]
  233. Kong, Y.; Zhao, Y.; Li, D.; Shen, H.; Yan, M. Dual Delivery of Encapsulated BM-MSCs and BMP-2 Improves Osteogenic Differentiation and New Bone Formation. J. Biomed. Mater. Res. A 2019, 107, 2282–2295. [Google Scholar] [CrossRef] [PubMed]
  234. Heidariyan, Z.; Ghanian, M.H.; Ashjari, M.; Farzaneh, Z.; Najarasl, M.; Rezaei Larijani, M.; Piryaei, A.; Vosough, M.; Baharvand, H. Efficient and Cost-Effective Generation of Hepatocyte-like Cells through Microparticle-Mediated Delivery of Growth Factors in a 3D Culture of Human Pluripotent Stem Cells. Biomaterials 2018, 159, 174–188. [Google Scholar] [CrossRef] [PubMed]
  235. Kang, I.-G.; Kim, J.; Park, S.; Kim, H.-E.; Han, C.-M. PLLA Membrane with Embedded Hydroxyapatite Patterns for Improved Bioactivity and Efficient Delivery of Growth Factor. Macromol. Biosci. 2020, 20, e2000136. [Google Scholar] [CrossRef] [PubMed]
  236. Wu, L.; Gu, Y.; Liu, L.; Tang, J.; Mao, J.; Xi, K.; Jiang, Z.; Zhou, Y.; Xu, Y.; Deng, L.; et al. Hierarchical Micro/Nanofibrous Membranes of Sustained Releasing VEGF for Periosteal Regeneration. Biomaterials 2020, 227, 119555. [Google Scholar] [CrossRef] [PubMed]
  237. Xia, B.; Lv, Y. Dual-Delivery of VEGF and NGF by Emulsion Electrospun Nanofibrous Scaffold for Peripheral Nerve Regeneration. Mater. Sci. Eng. C Mater. Biol. Appl. 2018, 82, 253–264. [Google Scholar] [CrossRef]
  238. Muniyandi, P.; Palaninathan, V.; Mizuki, T.; Mohamed, M.S.; Hanajiri, T.; Maekawa, T. Scaffold Mediated Delivery of Dual MiRNAs to Transdifferentiate Cardiac Fibroblasts. Mater. Sci. Eng. C Mater. Biol. Appl. 2021, 128, 112323. [Google Scholar] [CrossRef]
  239. Javazmi, L.; Young, A.; Ash, G.J.; Low, T. Kinetics of Slow Release of Nitrogen Fertiliser from Multi-Layered Nanofibrous Structures. Sci. Rep. 2021, 11, 4871. [Google Scholar] [CrossRef]
  240. Jelonek, K.; Zajdel, A.; Wilczok, A.; Kaczmarczyk, B.; Musiał-Kulik, M.; Hercog, A.; Foryś, A.; Pastusiak, M.; Kasperczyk, J. Comparison of PLA-Based Micelles and Microspheres as Carriers of Epothilone B and Rapamycin. The Effect of Delivery System and Polymer Composition on Drug Release and Cytotoxicity against MDA-MB-231 Breast Cancer Cells. Pharmaceutics 2021, 13, 1881. [Google Scholar] [CrossRef]
  241. Da Costa, D.; Exbrayat-Héritier, C.; Rambaud, B.; Megy, S.; Terreux, R.; Verrier, B.; Primard, C. Surface Charge Modulation of Rifampicin-Loaded PLA Nanoparticles to Improve Antibiotic Delivery in Staphylococcus Aureus Biofilms. J. Nanobiotechnol. 2021, 19, 12. [Google Scholar] [CrossRef]
  242. Salahuddin, N.; Abdelwahab, M.; Gaber, M.; Elneanaey, S. Synthesis and Design of Norfloxacin Drug Delivery System Based on PLA/TiO2 Nanocomposites: Antibacterial and Antitumor Activities. Mater. Sci. Eng. C Mater. Biol. Appl. 2020, 108, 110337. [Google Scholar] [CrossRef]
  243. Salahuddin, N.; Gaber, M.; Elneanaey, S.; Snowdon, M.R.; Abdelwahab, M.A. Co-Delivery of Norfloxacin and Tenoxicam in Ag-TiO2/Poly(Lactic Acid) Nanohybrid. Int. J. Biol. Macromol. 2021, 180, 771–781. [Google Scholar] [CrossRef] [PubMed]
  244. Afsharzadeh, M.; Hashemi, M.; Babaei, M.; Abnous, K.; Ramezani, M. PEG-PLA Nanoparticles Decorated with Small-Molecule PSMA Ligand for Targeted Delivery of Galbanic Acid and Docetaxel to Prostate Cancer Cells. J. Cell. Physiol. 2020, 235, 4618–4630. [Google Scholar] [CrossRef] [PubMed]
  245. Du, X.; Yin, S.; Xu, L.; Ma, J.; Yu, H.; Wang, G.; Li, J. Polylysine and Cysteine Functionalized Chitosan Nanoparticle as an Efficient Platform for Oral Delivery of Paclitaxel. Carbohydr. Polym. 2020, 229, 115484. [Google Scholar] [CrossRef]
  246. Kong, N.; Deng, M.; Sun, X.-N.; Chen, Y.-D.; Sui, X.-B. Polydopamine-Functionalized CA-(PCL-Ran-PLA) Nanoparticles for Target Delivery of Docetaxel and Chemo-Photothermal Therapy of Breast Cancer. Front. Pharmacol. 2018, 9, 125. [Google Scholar] [CrossRef] [PubMed]
  247. Taghavi, S.; Abnous, K.; Babaei, M.; Taghdisi, S.M.; Ramezani, M.; Alibolandi, M. Synthesis of Chimeric Polymersomes Based on PLA-b-PHPMA and PCL-b-PHPMA for Nucleoline Guided Delivery of SN38. Nanomedicine 2020, 28, 102227. [Google Scholar] [CrossRef] [PubMed]
  248. Wang, N.; Shi, J.; Wu, C.; Chu, W.; Tao, W.; Li, W.; Yuan, X. Design of DOX-GNRs-PNIPAM@PEG-PLA Micelle With Temperature and Light Dual-Function for Potent Melanoma Therapy. Front. Chem. 2020, 8, 599740. [Google Scholar] [CrossRef]
  249. Wang, Y.-R.; Yang, S.-Y.; Chen, G.-X.; Wei, P. Barbaloin Loaded Polydopamine-Polylactide-TPGS (PLA-TPGS) Nanoparticles against Gastric Cancer as a Targeted Drug Delivery System: Studies in Vitro and in Vivo. Biochem. Biophys. Res. Commun. 2018, 499, 8–16. [Google Scholar] [CrossRef]
  250. Yao, H.; Song, S.; Miao, X.; Liu, X.; Zhao, J.; Wang, Z.; Shao, X.; Zhang, Y.; Han, G. MPEG-PLA Micelle for Delivery of Effective Parts of Andrographis Paniculata. Curr. Drug Deliv. 2018, 15, 532–540. [Google Scholar] [CrossRef]
  251. Zheng, W.; Li, M.; Lin, Y.; Zhan, X. Encapsulation of Verapamil and Doxorubicin by MPEG-PLA to Reverse Drug Resistance in Ovarian Cancer. Biomed. Pharmacother. 2018, 108, 565–573. [Google Scholar] [CrossRef]
  252. Fayzullin, A.; Churbanov, S.; Ignatieva, N.; Zakharkina, O.; Tokarev, M.; Mudryak, D.; Khristidis, Y.; Balyasin, M.; Kurkov, A.; Golubeva, E.N.; et al. Local Delivery of Pirfenidone by PLA Implants Modifies Foreign Body Reaction and Prevents Fibrosis. Biomedicines 2021, 9, 853. [Google Scholar] [CrossRef]
  253. Abou-ElNour, M.; Ishak, R.A.H.; Tiboni, M.; Bonacucina, G.; Cespi, M.; Casettari, L.; Soliman, M.E.; Geneidi, A.S. Triamcinolone Acetonide-Loaded PLA/PEG-PDL Microparticles for Effective Intra-Articular Delivery: Synthesis, Optimization, in Vitro and in Vivo Evaluation. J. Control. Release 2019, 309, 125–144. [Google Scholar] [CrossRef] [PubMed]
  254. Ferrández-Montero, A.; Eguiluz, A.; Vazquez, E.; Guerrero, J.D.; Gonzalez, Z.; Sanchez-Herencia, A.J.; Ferrari, B. Controlled SrR Delivery by the Incorporation of Mg Particles on Biodegradable PLA-Based Composites. Polymers 2021, 13, 1061. [Google Scholar] [CrossRef] [PubMed]
  255. Han, S.H.; Cha, M.; Jin, Y.-Z.; Lee, K.-M.; Lee, J.H. BMP-2 and HMSC Dual Delivery onto 3D Printed PLA-Biogel Scaffold for Critical-Size Bone Defect Regeneration in Rabbit Tibia. Biomed. Mater. 2020, 16, 015019. [Google Scholar] [CrossRef] [PubMed]
  256. Palazzo, I.; Lamparelli, E.P.; Ciardulli, M.C.; Scala, P.; Reverchon, E.; Forsyth, N.; Maffulli, N.; Santoro, A.; Della Porta, G. Supercritical Emulsion Extraction Fabricated PLA/PLGA Micro/Nano Carriers for Growth Factor Delivery: Release Profiles and Cytotoxicity. Int. J. Pharm. 2021, 592, 120108. [Google Scholar] [CrossRef]
  257. Amani, A.; Dustparast, M.; Noruzpour, M.; Zakaria, R.A.; Ebrahimi, H.A. Design and Invitro Characterization of Green Synthesized Magnetic Nanoparticles Conjugated with Multitargeted Poly Lactic Acid Copolymers for Co-Delivery of SiRNA and Paclitaxel. Eur. J. Pharm. Sci. 2021, 167, 106007. [Google Scholar] [CrossRef]
  258. Senapati, S.; Upadhyaya, A.; Dhruw, S.; Giri, D.; Maiti, P. Controlled DNA Delivery Using Poly(Lactide) Nanoparticles and Understanding the Binding Interactions. J. Phys. Chem. B 2021, 125, 10009–10017. [Google Scholar] [CrossRef]
  259. Ghasemi, R.; Abdollahi, M.; Emamgholi Zadeh, E.; Khodabakhshi, K.; Badeli, A.; Bagheri, H.; Hosseinkhani, S. MPEG-PLA and PLA-PEG-PLA Nanoparticles as New Carriers for Delivery of Recombinant Human Growth Hormone (RhGH). Sci. Rep. 2018, 8, 9854. [Google Scholar] [CrossRef]
  260. Meena, J.; Kumar, R.; Singh, M.; Ahmed, A.; Panda, A.K. Modulation of Immune Response and Enhanced Clearance of Salmonella Typhi by Delivery of Vi Polysaccharide Conjugate Using PLA Nanoparticles. Eur. J. Pharm. Biopharm. 2020, 152, 270–281. [Google Scholar] [CrossRef]
  261. Cai, Y.; Xu, Z.; Shuai, Q.; Zhu, F.; Xu, J.; Gao, X.; Sun, X. Tumor-Targeting Peptide Functionalized PEG-PLA Micelles for Efficient Drug Delivery. Biomater. Sci. 2020, 8, 2274–2282. [Google Scholar] [CrossRef]
  262. Fan, T.; Daniels, R. Preparation and Characterization of Electrospun Polylactic Acid (PLA) Fiber Loaded with Birch Bark Triterpene Extract for Wound Dressing. AAPS PharmSciTech 2021, 22, 205. [Google Scholar] [CrossRef]
  263. Sun, S.; Du, X.; Fu, M.; Khan, A.R.; Ji, J.; Liu, W.; Zhai, G. Galactosamine-Modified PEG-PLA/TPGS Micelles for the Oral Delivery of Curcumin. Int. J. Pharm. 2021, 595, 120227. [Google Scholar] [CrossRef] [PubMed]
  264. Tsintou, M.; Dalamagkas, K.; Moore, T.L.; Rathi, Y.; Kubicki, M.; Rosene, D.L.; Makris, N. The Use of Hydrogel-Delivered Extracellular Vesicles in Recovery of Motor Function in Stroke: A Testable Experimental Hypothesis for Clinical Translation Including Behavioral and Neuroimaging Assessment Approaches. Neural. Regen. Res. 2020, 16, 605–613. [Google Scholar] [CrossRef]
  265. Pavia, F.C.; Di Bella, M.A.; Brucato, V.; Blanda, V.; Zummo, F.; Vitrano, I.; Di Liegro, C.M.; Ghersi, G.; Di Liegro, I.; Schiera, G. A 3D-Scaffold of PLLA Induces the Morphological Differentiation and Migration of Primary Astrocytes and Promotes the Production of Extracellular Vesicles. Mol. Med. Rep. 2019, 20, 1288–1296. [Google Scholar] [CrossRef]
  266. He, X.; Yang, L.; Dong, K.; Zhang, F.; Liu, Y.; Ma, B.; Chen, Y.; Hai, J.; Zhu, R.; Cheng, L. Biocompatible Exosome-Modified Fibrin Gel Accelerates the Recovery of Spinal Cord Injury by VGF-Mediated Oligodendrogenesis. J. Nanobiotechnol. 2022, 20, 360. [Google Scholar] [CrossRef] [PubMed]
  267. Bari, E.; Roato, I.; Perale, G.; Rossi, F.; Genova, T.; Mussano, F.; Ferracini, R.; Sorlini, M.; Torre, M.L.; Perteghella, S. Biohybrid Bovine Bone Matrix for Controlled Release of Mesenchymal Stem/Stromal Cell Lyosecretome: A Device for Bone Regeneration. Int. J. Mol. Sci. 2021, 22, 4064. [Google Scholar] [CrossRef]
  268. Azi, M.L.; Aprato, A.; Santi, I.; Junior, M.K.; Masse, A.; Joeris, A. Autologous Bone Graft in the Treatment of Post-Traumatic Bone Defects: A Systematic Review and Meta-Analysis. BMC Musculoskelet. Disord. 2016, 17, 465. [Google Scholar] [CrossRef]
  269. Rogers, G.F.; Greene, A.K. Autogenous Bone Graft: Basic Science and Clinical Implications. J. Craniofacial Surg. 2012, 23, 323–327. [Google Scholar] [CrossRef]
  270. Emami, A.; Talaei-Khozani, T.; Tavanafar, S.; Zareifard, N.; Azarpira, N.; Vojdani, Z. Synergic Effects of Decellularized Bone Matrix, Hydroxyapatite, and Extracellular Vesicles on Repairing of the Rabbit Mandibular Bone Defect Model. J. Transl. Med. 2020, 18, 361. [Google Scholar] [CrossRef]
  271. 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]
  272. Chen, H.; Liu, Y.; Wang, C.; Zhang, A.; Chen, B.; Han, Q.; Wang, J. Design and Properties of Biomimetic Irregular Scaffolds for Bone Tissue Engineering. Comput. Biol. Med. 2021, 130, 104241. [Google Scholar] [CrossRef]
  273. Yeung, E.; Fukunishi, T.; Bai, Y.; Bedja, D.; Pitaktong, I.; Mattson, G.; Jeyaram, A.; Lui, C.; Ong, C.S.; Inoue, T.; et al. Cardiac Regeneration Using Human IPSC-Derived Biomaterial-Free 3D Bioprinted Cardiac Patch in Vivo. J. Tissue Eng. Regen. Med. 2019, 13, 2031–2039. [Google Scholar] [CrossRef] [PubMed]
  274. Monguió-Tortajada, M.; Prat-Vidal, C.; Martínez-Falguera, D.; Teis, A.; Soler-Botija, C.; Courageux, Y.; Munizaga-Larroudé, M.; Moron-Font, M.; Bayes-Genis, A.; Borràs, F.E.; et al. Acellular Cardiac Scaffolds Enriched with MSC-Derived Extracellular Vesicles Limit Ventricular Remodelling and Exert Local and Systemic Immunomodulation in a Myocardial Infarction Porcine Model. Theranostics 2022, 12, 4656–4670. [Google Scholar] [CrossRef] [PubMed]
  275. Wang, C.; Liang, C.; Wang, R.; Yao, X.; Guo, P.; Yuan, W.; Liu, Y.; Song, Y.; Li, Z.; Xie, X. The Fabrication of a Highly Efficient Self-Healing Hydrogel from Natural Biopolymers Loaded with Exosomes for the Synergistic Promotion of Severe Wound Healing. Biomater. Sci. 2019, 8, 313–324. [Google Scholar] [CrossRef] [PubMed]
  276. Zhao, D.; Yu, Z.; Li, Y.; Wang, Y.; Li, Q.; Han, D. GelMA Combined with Sustained Release of HUVECs Derived Exosomes for Promoting Cutaneous Wound Healing and Facilitating Skin Regeneration. J. Mol. Histol. 2020, 51, 251–263. [Google Scholar] [CrossRef] [PubMed]
  277. Chung, L.; Maestas, D.R.; Housseau, F.; Elisseeff, J.H. Key Players in the Immune Response to Biomaterial Scaffolds for Regenerative Medicine. Adv. Drug Deliv. Rev. 2017, 114, 184–192. [Google Scholar] [CrossRef]
  278. Karin, M.; Clevers, H. Reparative Inflammation Takes Charge of Tissue Regeneration. Nature 2016, 529, 307–315. [Google Scholar] [CrossRef]
  279. Mescher, A.L.; Neff, A.W.; King, M.W. Inflammation and Immunity in Organ Regeneration. Dev. Comp. Immunol. 2017, 66, 98–110. [Google Scholar] [CrossRef]
  280. Chen, W.; Huang, Y.; Han, J.; Yu, L.; Li, Y.; Lu, Z.; Li, H.; Liu, Z.; Shi, C.; Duan, F.; et al. Immunomodulatory Effects of Mesenchymal Stromal Cells-Derived Exosome. Immunol. Res. 2016, 64, 831–840. [Google Scholar] [CrossRef]
  281. Kang, M.; Huang, C.-C.; Gajendrareddy, P.; Lu, Y.; Shirazi, S.; Ravindran, S.; Cooper, L.F. Extracellular Vesicles From TNFα Preconditioned MSCs: Effects on Immunomodulation and Bone Regeneration. Front. Immunol. 2022, 13, 878194. [Google Scholar] [CrossRef]
  282. Su, N.; Hao, Y.; Wang, F.; Hou, W.; Chen, H.; Luo, Y. Mesenchymal Stromal Exosome–Functionalized Scaffolds Induce Innate and Adaptive Immunomodulatory Responses toward Tissue Repair. Sci. Adv. 2021, 7, eabf7207. [Google Scholar] [CrossRef]
  283. Sun, Y.; Zhang, B.; Zhai, D.; Wu, C. Three-Dimensional Printing of Bioceramic-Induced Macrophage Exosomes: Immunomodulation and Osteogenesis/Angiogenesis. NPG Asia Mater. 2021, 13, 72. [Google Scholar] [CrossRef]
  284. Bari, E.; Scocozza, F.; Perteghella, S.; Sorlini, M.; Auricchio, F.; Torre, M.L.; Conti, M. 3D Bioprinted Scaffolds Containing Mesenchymal Stem/Stromal Lyosecretome: Next Generation Controlled Release Device for Bone Regenerative Medicine. Pharmaceutics 2021, 13, 515. [Google Scholar] [CrossRef] [PubMed]
  285. Xing, X.; Han, S.; Ni, Y.; Cheng, G.; Cheng, Y.; Ni, X.; Deng, Y.; Li, Z.; Li, Z. Mussel-Inspired Functionalization of Electrospun Scaffolds with Polydopamine-Assisted Immobilization of Mesenchymal Stem Cells-Derived Small Extracellular Vesicles for Enhanced Bone Regeneration. Int. J. Pharm. 2021, 609, 121136. [Google Scholar] [CrossRef] [PubMed]
  286. Witwer, K.W.; Van Balkom, B.W.M.; Bruno, S.; Choo, A.; Dominici, M.; Gimona, M.; Hill, A.F.; De Kleijn, D.; Koh, M.; Lai, R.C.; et al. Defining Mesenchymal Stromal Cell (MSC)-Derived Small Extracellular Vesicles for Therapeutic Applications. J. Extracell. Vesicles 2019, 8, 1609206. [Google Scholar] [CrossRef]
  287. Xiao, S.; Xiao, C.; Miao, Y.; Wang, J.; Chen, R.; Fan, Z.; Hu, Z. Human Acellular Amniotic Membrane Incorporating Exosomes from Adipose-Derived Mesenchymal Stem Cells Promotes Diabetic Wound Healing. Stem. Cell Res. Ther. 2021, 12, 255. [Google Scholar] [CrossRef] [PubMed]
  288. Jiang, S.; Tian, G.; Yang, Z.; Gao, X.; Wang, F.; Li, J.; Tian, Z.; Huang, B.; Wei, F.; Sang, X.; et al. Enhancement of Acellular Cartilage Matrix Scaffold by Wharton’s Jelly Mesenchymal Stem Cell-Derived Exosomes to Promote Osteochondral Regeneration. Bioact. Mater. 2021, 6, 2711–2728. [Google Scholar] [CrossRef]
  289. Fischer, T.; Hayn, A.; Mierke, C.T. Fast and Reliable Advanced Two-Step Pore-Size Analysis of Biomimetic 3D Extracellular Matrix Scaffolds. Sci. Rep. 2019, 9, 8352. [Google Scholar] [CrossRef]
  290. Hady, T.F.; Hwang, B.; Pusic, A.D.; Waworuntu, R.L.; Mulligan, M.; Ratner, B.; Bryers, J.D. Uniform 40-Μm-Pore Diameter Precision Templated Scaffolds Promote a pro-Healing Host Response by Extracellular Vesicle Immune Communication. J. Tissue Eng. Regen. Med. 2021, 15, 24–36. [Google Scholar] [CrossRef]
Figure 1. An overview of extracellular vesicle (EV) delivery via scaffolds. The contents and categories of the extracellular vesicles as well as what the extracellular vesicles may regulate are described. Common natural and synthetic biomaterials for scaffold fabrication are highlighted.
Figure 1. An overview of extracellular vesicle (EV) delivery via scaffolds. The contents and categories of the extracellular vesicles as well as what the extracellular vesicles may regulate are described. Common natural and synthetic biomaterials for scaffold fabrication are highlighted.
Cells 11 02851 g001
Table 1. The therapeutic applications of natural and synthetic biopolymer-based scaffolds that utilize extracellular vesicles. The type of EV and biopolymer used in each study are indicated as well as the EV incorporation method into the scaffold.
Table 1. The therapeutic applications of natural and synthetic biopolymer-based scaffolds that utilize extracellular vesicles. The type of EV and biopolymer used in each study are indicated as well as the EV incorporation method into the scaffold.
StudyEV SourceScaffold BiomaterialModifications and/or EV Incorporation MethodDisease or TissueAnimal ModelsTherapeutic Outcomes
[33]Human adipose-derived mesenchymal stem cell (hADMSC) EVsSilk-based tubular scaffoldVacuum-seeded EVsCardiovascular diseaseRat abdominal aortic interposition graft modelImproved patency and matrix deposition, including increased elastin and collagen production
[26]Bone marrow-derived mesenchymal stem cell (BMSC) sEVsSodium AlginatesEVs mixed with sodium alginate solutionMyocardial infarction (MI)MI induction in male ratsDecreased cardiac cell apoptosis

Promotion of macrophage polarization

Increased scar thickness and angiogenesis

Improved cardiac function and infarct size
[37] Human umbilical cord mesenchymal stem cell (HUCMSCs) ExosomesPolyvinyl alcohol (PVA)/AlginateExosomes mixed with PVA/alginate solutionDiabetic wound healingFull-thickness wounds on diabetic rat modelProliferation, migration, and angiogenesis of HUVECs

Sped up diabetic wound healing by promoting angiogenesis
[38]ADSC-derived ExosomesSodium AlginateExosomes mixed with alginate solutionPeripheral nerve regenerationRat sciatic nerve defectExosomes containing neurotrophin-3 mRNA (important neurotrophic factor for peripheral nerve regeneration) in scaffold promoted nerve regeneration
[39]Dental pulp stem cell (DPSC)-derived EVsCollagenEVs were injected into the scaffoldBone regenerationRat calvarial bone defectBone formation in center of defects

Broader angiogenesis
[40]HUCMSCsCollagenExosomes added dropwise onto scaffoldEndometrium regeneration and fertility restorationRat endometrium-damage modelInduction of endometrium regeneration, collagen remodeling
Increased expression of estrogen receptor α/progesterone receptor

Restored fertility

Facilitated CD163+ M2 macrophage polarization, reduced inflammation, increased anti-inflammatory responses
[32]BMSC EVsChitosan-Collagen Composite ScaffoldScaffolds seeded with EVsSkin wound healingFull-thickness skin wound on adult male ratsAccelerated skin healing

Enhanced macrophage count

Greater collagen deposition, better collagen alignment and thus, increased mechanical strength
[41]Bone mesenchymal stem cell-derived sEVsChitosansEVs added to hydrogelsBone defect repairCalvarial defect rat modelsEV-loaded hydrogel promoted bone healing by enhancing angiogenesis possibly via upregulation of miR-21 in sEVs
[42]Induced pluripotent stem cell-derived mesenchymal stem cells (iPSC-MSCs) exosomesChitosanExosomes stirred into chitosan solutionCorneal diseasesRat cornea anterior lamellar damage modelPromote repair of damaged corneal epithelium and stromal layer
[43]Chondrocyte exosomesChitosan-Gelatin-Chondroitin Sulfate and Nano-Hydroxyapatite-GelatinExosome suspension added dropwise over scaffoldArticular cartilage injuriesN/AEnhance proliferation and migration of chondrocytes
[44]Osteoblast-derived EVsGelatin Methacryloyl (GelMA)GelMA functionalized with nanoclay laponiteBone regenerationN/AEnhanced proliferation, migration, histone acetylation, mineralization of human bone marrow stromal cells
[45]Tendon derived stem cells (TDSCs) exosomesHyaluronic acidLoaded scaffold–not sufficiently describedTendon repairRat model of tendon defectsPromoted proliferation, migration, collagen type I production, and tendon-specific markers expression in tenocytes

Protected tenocytes from oxidative stress and serum deprivation

Promoted early healing of injured tendons and better fiber arrangement at injury site
[46]Human articular chondrocyte-derived EVsHyaluronic acid/ChitosanLoaded scaffold–not sufficiently describedOsteoarthritis cartilage injuriesRabbit osteochondral defect modelGreater cartilage regeneration

Provide niche for chondrogenic differentiation of MSCs

Hyalin-like cartilage in defect zone
[47]M2 polarized macrophage-derived Exosomes (M2-Exos)PEGDissolved freeze-dried PEG in exosome solutionCutaneous wound healingMouse excisional wound splinting modelLocalization and sustained release of M2-Exos

Effective and prolonged conversion of M1 to M2 macrophages

Enhanced efficiency and quality of wound care
[48]hADMSC-derived exosomesPLAMineral doped PLA scaffolds adsorbed exosomesBone defects including oral bone defectsN/AIncreased osteogenic commitment of MSCs
[49]Mesenchymal stem cell (MSC)-derived sEVsPCLsEVs loaded onto heparin-modified scaffoldCardiovascular diseaseHyperlipidemia rat modelInhibited thrombosis and calcification and thus improved patency of graft

Enhanced endothelium and vascular smooth muscle regeneration

Induced polarization of M1 macrophages to M2c macrophages
[50]MSC-exosomesPCLPCL modified with S-nitrosoglutathione (GSNO); exosomes incubated with scaffoldBone defectsN/ADecreased expression of pro-inflammatory genes in macrophages treated with exosome-loaded scaffold

Accelerated osteogenic differentiation of mesenchymal stem cells
[51]MSC-derived EVsPLGAEVs infused into composite scaffold; polydeoxyribonucleotide (PDRN) addedChronic kidney diseasePartial nephrectomy mouse modelSynergistic interaction of EVs and other added compounds in scaffold alleviates fibrosis and inflammatory response

Cellular proliferation

Angiogenesis

Effective glomerular regeneration

Restoration of kidney function

Develop new blood vessels and induces pro-reparative macrophages
[52]Human adipose-derived stem cell exosomesPLGAScaffolds submerged in exosome solutionBone defectsMouse calvarial defectEnhance bone regeneration partially through osteoinductive effects and promoting mesenchymal stem cell migration and homing in newly formed bone tissue
[53]MSC-sEVsPEG/Hyaluronic AcidsEVs mixed with scaffold solutionOsteoarthritis (OA)Traumatic OA rat modelImproved bioavailability and therapeutic efficacy of MSC-sEVs for OA improvement
[54]Human DPSC-derived exosomesPLLAExosome encapsulation in triblock PLGA-PEG-PLGA microspheres and subsequently incorporated into PLLA scaffoldBone defectsCritical size mouse calvarial bone defectStimulated bone tissue neogenesis

Facilitated bone marrow stromal cell osteogenic differentiation

Guided local progenitor cells towards osteogenic differentiation and bone healing

Accelerated bone healing
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Leung, K.S.; Shirazi, S.; Cooper, L.F.; Ravindran, S. Biomaterials and Extracellular Vesicle Delivery: Current Status, Applications and Challenges. Cells 2022, 11, 2851. https://doi.org/10.3390/cells11182851

AMA Style

Leung KS, Shirazi S, Cooper LF, Ravindran S. Biomaterials and Extracellular Vesicle Delivery: Current Status, Applications and Challenges. Cells. 2022; 11(18):2851. https://doi.org/10.3390/cells11182851

Chicago/Turabian Style

Leung, Kasey S., Sajjad Shirazi, Lyndon F. Cooper, and Sriram Ravindran. 2022. "Biomaterials and Extracellular Vesicle Delivery: Current Status, Applications and Challenges" Cells 11, no. 18: 2851. https://doi.org/10.3390/cells11182851

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