Next Article in Journal
Two-Step Chemo-Microbial Degradation of Post-Consumer Polyethylene Terephthalate (PET) Plastic Enabled by a Biomass-Waste Catalyst
Previous Article in Journal
Optimizing Spinal Fusion Cage Design to Improve Bone Substitute Filling on Varying Disc Heights: A 3D Printing Study
Previous Article in Special Issue
Direct Integration of 3D Printing and Cryogel Scaffolds for Bone Tissue Engineering
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Delivery of Growth Factors to Enhance Bone Repair

Department of Orthopaedic Surgery, University of Southern California Keck School of Medicine, 1500 San Pablo St., Los Angeles, CA 90033, USA
*
Author to whom correspondence should be addressed.
Bioengineering 2023, 10(11), 1252; https://doi.org/10.3390/bioengineering10111252
Submission received: 15 September 2023 / Revised: 20 October 2023 / Accepted: 25 October 2023 / Published: 26 October 2023
(This article belongs to the Special Issue Advances in Enabling Technologies for Bone Tissue Engineering)

Abstract

:
The management of critical-sized bone defects caused by nonunion, trauma, infection, malignancy, pseudoarthrosis, and osteolysis poses complex reconstruction challenges for orthopedic surgeons. Current treatment modalities, including autograft, allograft, and distraction osteogenesis, are insufficient for the diverse range of pathology encountered in clinical practice, with significant complications associated with each. Therefore, there is significant interest in the development of delivery vehicles for growth factors to aid in bone repair in these settings. This article reviews innovative strategies for the management of critical-sized bone loss, including novel scaffolds designed for controlled release of rhBMP, bioengineered extracellular vesicles for delivery of intracellular signaling molecules, and advances in regional gene therapy for sustained signaling strategies. Improvement in the delivery of growth factors to areas of significant bone loss has the potential to revolutionize current treatment for this complex clinical challenge.

Graphical Abstract

1. Introduction

1.1. Bone Loss Challenge

Critical-sized bone defects (Figure 1) caused by nonunion, trauma, infection, malignancy, spine pseudoarthrosis, and osteolysis pose complex reconstructive challenges for orthopedic surgeons. Failure to heal bone defects is multifactorial, with potential complications occurring at each stage of healing [1]. To achieve successful bone regeneration, there are four essential elements: (1) release of growth factors to recruit osteoprogenitor cells, (2) responding cells that mobilize to the defect, (3) osteoconductive matrix to support bone formation, and (4) a sufficient vascular supply [2]. Multiple strategies have been developed to address impairments in bone healing, including the use of autograft, allograft, growth factors, and distraction osteogenesis for larger defects [3,4,5,6,7]. Over 500,000 bone grafting procedures occur annually in the United States, with bone graft substitutes costing upwards of USD 3000 per application, resulting in USD 5 billion in annual healthcare expenditures [8,9,10,11]. Despite advances in technology and surgical techniques, significant challenges remain, including donor site morbidity for autograft harvest, insufficient graft availability, pseudoarthrosis or persistent nonunion, and prolonged immobilization in distraction osteogenesis.

1.2. Growth Factors for Treatment

Currently, there is significant interest in the use of growth factors to address these complex reconstructive challenges. Dating back to the 19th century, demineralized bone matrix (DBM) has been used as both xenograft and allograft for the treatment of large bone defects with mixed success [8,12,13,14]. It would take almost a century before the landmark study by Urist in 1965, where he inferred that “autoinduction” of host cell populations was responsible for the ectopic bone formation seen with implantation of allogeneic DBM [15]. This theorized inductive substance was termed bone morphogenetic protein (BMP), although it was not isolated at the time [16]. Reddi and Anderson further elucidated this pathway of DBM function, surmising there must exist some factor within the “soluble extract” allowing for the observed ectopic ossification [17]. In 1988, Muthukumaran and Reddi isolated an “osteogenin-enriched fraction” from DMB samples and demonstrated in a dose-dependent fashion that the combination of the extract with inactive collagenous bone matrix resulted in ectopic bone formation [18]. Although the exact composition of this extract was not yet elucidated, this would lead to the characterization of the now well-described family of growth factors, Bone Morphogenetic Proteins [12]. Wozney et al. further characterized BMPs by successfully preparing human complementary DNA (cDNA) from protein extracts, which allowed for the study of recombinant BMPs’ individual biologic activity as well as the development of gene therapy applications [19,20].

1.3. FDA-Approved Growth Factors for Bone Repair

Secondary bone repair is an intricate and dynamic cascade of molecular and cellular processes within the bone tissue microenvironment that occurs in four phases: (1) hematoma formation, (2) fibrocartilage callous development, (3) hard callus formation, and (4) remodeling [21]. These stages are highly regulated by inflammatory cytokines that temporally mediate growth factor release [22]. Tissue engineering (TE) strategies take advantage of this pathway by selecting a variety of potential growth factors that can be targeted to promote healing. Recombinant human BMP-2 (rhBMP) and platelet-derived growth factor (PDGF) are currently the only FDA-approved therapies for bone healing utilizing growth factors [23]. However, their indications remain narrow, with rhBMP-2 being FDA-approved for acute open tibial shaft fracture, alveolar ridge defects, and anterior lumbar spine surgery and PDGF for ankle and hindfoot fusions [24]. Although PGDF has shown promise in multiple clinical trials for hindfoot fusion in the setting of end-stage osteoarthritis, poor osteoinductive potential limits its application for the treatment of critical-sized bone defects [25,26,27,28]. Several other growth factors can influence the bone repair process, including vascular endothelial growth factor (VEGF) [29,30,31], insulin-like growth factor 1 (IGF-1) [32,33], fibroblast growth factor (FGF) [34,35,36], and transforming growth factor beta (TGF-β) [37]. Although these factors have established roles in bone regeneration, BMPs have consistently demonstrated their centrality to the process through in vitro assays, in vivo preclinical models, human disease, and the development of clinically important therapeutic agents [38,39]. Furthermore, BMPs alone are sufficiently osteoinductive for bone repair, which makes them ideal candidates for tissue engineering applications [40].
To promote optimal bone formation, delivery of growth factors needs to occur in sufficient quantities at the site of interest and at the appropriate time with responding cells available. Multiple preclinical TE strategies have been developed to allow for the osteoinductive signals of prospective growth factors to be paired with osteoconductive scaffolds and cells that can respond to these molecular cascades. In this review, we will highlight delivery options for growth factors, including novel scaffolds for controlled release of rhBMP, bioengineered vesicles for delivery of intracellular signaling molecules, and advances in regional gene therapy for sustained signaling strategies, as shown in Table 1.

2. Carriers of Bone Morphogenetic Protein and Other Growth Factors

BMPs are a family of ligands with at least 30 different members to date. Structurally, BMP homodimers include seven cysteine amino acids assisting in the formation of disulfide bonds as well as with dimerization into its biologically active form [41,42,43]. The canonical pathway of BMP signaling involves binding to cell surface receptors, leading to a complex formation composed of two dimers of type I and II serine/threonine kinase receptors, eventually resulting in activation of the Smad pathway [44,45]. Historically, BMP-2 and BMP-7 have been well described for their roles in osteoinduction. BMP-2, existing during skeletal embryonic development, has been shown as an essential component for bone fracture healing. Similarly, BMP-7 also serves a role in the fetal stage, additionally serving as an anti-inflammatory growth factor that elevates alkaline phosphatase (ALP) activity [44].
Cloned rhBMPs are used commercially as an alternative to allograft. The FDA has approved rhBMP-2 for the treatment of open tibial shaft fractures, anterior lumbar interbody fusion, alveolar ridge augmentation, and sinus lift, as shown in Table 2 [24]. Current FDA-approved rhBMP-2 is delivered on an absorbable collagen sponge (ACS) that can be applied directly to the targeted region [46]. Alternative growth factors have also been investigated in preclinical models using an ACS for delivery, as discussed in Section 2.1. Additionally, rhBMP-7 applied in a putty composed of bovine collagen and carboxymethylcellulose for long bone nonunion and posterolateral lumbar fusion had previously received a Humanitarian Device Exemption but has since been removed from the United States market by its supplier, as it was no longer considered “medically necessary” [24]. Despite these approvals, there is concern regarding the large doses of rhBMP-2 currently used for treatment to enhance its biological activity. Not only expensive, high doses of rhBMP-2 have led to a variety of complications, including heterotopic ossification, clinically significant dysphasia associated with soft tissue edema, and inflammatory reactions [47,48,49,50,51,52,53]. Furthermore, it has been hypothesized that the rapid release of the BMP from the collagen sponge limits the body’s response [54,55,56]. Although the collagen sponge remains the only FDA-approved carrier for rhBMP-2, there is much room for improvement in next-generation carriers (Figure 2). The most common new drug delivery modalities being developed are scaffolds, broadly defined as structures that can support cell growth; these scaffold designs include porous 3D matrices, hydrogels, spheres, and fiber meshes. There are four primary categories of biomaterials used for scaffolds, including natural polymers, inorganic materials, synthetic polymers, and composites [57,58]. Despite the abundance of carriers proposed in the literature, there are significant barriers to bringing new devices to market. Clinical application of next-generation scaffolds will require significant investment and large animal models demonstrating safety and efficacy greater than current treatment modalities. Although significant challenges remain, advancement in scaffold design and composition offers promise in reducing supraphysiologic dosing requirements for growth factors in clinical practice.

2.1. Collagen Polymers

The prevalent connective tissue protein collagen has been well described in wound repair. The efficacy of collagen applied through absorbable sponges fabricated by freeze-drying has been extensively studied in large clinical trials. The BESTT clinical trial performed by Govender et al. demonstrated in a prospective randomized controlled trial of 450 patients with open tibia fractures that rhBMP-2 loaded ACS had significantly fewer hardware failures, infections, and faster wound healing than the control group of intramedullary nail fixation alone [59]. INFUSE® (Medtronic, Minneapolis, MN, USA), a rhBMP-2 loaded ACS, is currently approved by the FDA for human use. However, despite such results, collagen sponges have demonstrated rapid collagen fiber degradation by collagenase enzymes, lack of mechanical strength, and absence of sustained release with less than 5% of the protein noted at two weeks post-implantation [58,60].
Burkus et al. performed a multi-center prospective randomized trial of 279 patients with degenerative disc disease who underwent anterior lumbar interbody fusion (ALIF) with or without the addition of rhBMP on an ACS [61]. At two-year follow-up, the experimental group had a significantly higher fusion rate (94.5%) than the control (88.7%), with similar patient-reported outcomes [61]. Additionally, radiographic healing was assessed at 6, 12, and 24 months in 42 patients, demonstrating osteogenesis in the disc space at 6 months postoperatively in 82% of patients, with most growth occurring between 6 and 12 months post-surgery [62].
Several applications using collagen sponges as carriers for growth factors other than rhBMP-2 have also been described. Kigami et al. treated a 5 mm rat calvarial defect model with an FGF-2 loaded ACS, demonstrating increased blood vessel and bone growth when compared to the ACS-only group [63]. The higher dose group of 0.3% FGF-2 showed significantly higher values in both outcomes when compared to the 0.1% FGF-2 cohort [63]. Additionally, Ueda et al. utilized TGF-β loaded collagen sponges to repair 6 mm skull defects in rabbits and demonstrated significantly more bone formation after six weeks when compared to ACS alone and free TGF-β [64]. However, it must be noted that a calvarial bone defect model is not as biologically stringent as a critical-sized long bone defect.
Several studies have been conducted to characterize the pharmacokinetics of growth factor release from collagen sponges. The BMP-2 ACS complex specifically is susceptible to significant proteolysis early postoperatively, leading to rapid elimination. Enhancements have been made to the classic collagen structure through cross-linking, carbodiimide additives, gamma radiation, and the addition of synthetic or inorganic compounds to increase stability [65]. Specifically, composites with collagen/poly(DL-lactic acid) have demonstrated improvements to the GF delivery system in preclinical models [58,66]. In a 2 cm canine ulna critical-sized defect model, Itoh et al. demonstrated significant bone formation in the experimental groups of 140 ug and 640 ug of rhBMP-2 placed on composite scaffolds [67]. Furthermore, both high and low-dose cohorts enhanced bony union with higher bone mineral content when compared to the PLGA/gelatin sponge alone [68]. Collagen additives such as heparin and fibronectin have also been described to enhance BMP binding and, therefore, lower the therapeutic dose by extending release. Lee et al. described the addition of BMP-2 on an absorbable collagen sponge with heparin sulfate nanofibers in a 5 mm rat femoral critical-sized defect model, finding that lower doses of BMP-2 generated more new bone on histological evaluation as compared to the conventional collagen sponge [69]. Interestingly, BMP contains heparin-binding domains that interact noncovalently with the heparin sulfate nanofibers, therefore prolonging release. Although these advances are promising, to our knowledge, these types of collagen sponges have not yet been FDA-approved to be used as carriers for rhBMP-2.
Gelatin, a form of type I collagen, can be formulated into hydrogels, which serve as an alternative carrier [58]. Other natural polymers that can be synthesized into hydrogels include hyaluronic acid, fibrin, or synthetic polymers such as polyethylene glycol (PEG). The mechanism of action includes entrapment using linkage molecules or with chemical makeup selective to the drug itself. Gel components can be modified to alter release through modifications of hydrogel interactions, fluid dynamics, and target tissue physiologic conditions. Hyaluronic acid is a commonly used material to formulate hydrogels. Electrostatic interactions between BMP-2 and hyaluronan are believed to mediate sustained-release kinetics, therefore prolonging the osteogenic signal [70]. In a 2 × 4 cm minipig cranial defect model, Docherty-Skogh et al. demonstrated a 100% increase in bone volume with the addition of 1.25 mg BMP-2 to an injectable hyaluronan-based hydrogel when compared to hydrogel alone [70]. Hydrogel formulations using other growth factors exist as well. In a model using FGF loaded on a p(HEMA-co-VP) hydrogel polymer scaffold, Mabilleau et al. used 4 × 6 mm cylindrical femoral condyle defects in rabbits to demonstrate increased woven bone and trabecular thickness [71]. However, this difference in bone generation became negligent between the FGF-hydrogel and the hydrogel-alone groups after three months [71].

2.2. Ceramics, Composites, and Applications of 3D Printing

Calcium phosphates formulated as hydroxyapatite (HA), coralline hydroxyapatite, and tricalcium phosphate (TCP) are additional materials used in scaffold development. They offer biocompatibility and the ability to withstand stress and tolerate high mechanical loads; however, drawbacks include brittleness and difficulty in generating highly porous structures needed for bony ingrowth and angiogenesis [58]. Additionally, while osteoconductive, ceramics such as HA lack osteoinductive properties. Calcium phosphates can interact with growth factors through hydrogen bonding, allowing manipulation of release kinetics through structural modifications of the carrier [72].
HA scaffolds used to deliver rhBMP-2 have demonstrated significant bone formation in a variety of settings, including alveolar ridge reconstruction, maxillary sinus augmentation, and segmental bone defects [57]. Utilizing a 3 cm sheep tibia critical-sized defect model, Boer and colleagues demonstrated 2–3 times increased torsional strength of BMP-2 loaded on HA as compared to HA alone [73]. The scaffold was created from bovine cancellous bone processed with a sintering method. Furthermore, using a 6–8 cm fibular critical-sized defect model in primates, Seeherman and colleagues showed treatment with percutaneous injection of rhBMP-2 integrated into a calcium phosphate paste resulted in accelerated healing of 40% compared with untreated sites, with torsional stiffness and maximum torque equal to an intact fibula after 14 weeks [74]. In a rat posterolateral fusion model, hydroxyapatite in the form of a fibrous mesh loaded with rhBMP-2 demonstrated a 60% greater fusion rate when compared to HA alone [75].
To enhance the osteoconductive properties of the scaffold, ceramics have historically been incorporated into composites with natural and synthetic polymers, lending it rigidity and hardness [67,76]. FDA-approved rhPDGF has successfully utilized a TCP-collagen compositive throughout clinical trials to enhance hindfoot arthrodesis [26,27,28]. Furthermore, a combination of collagen and HA has been described to create a material that simulates real bone morphology. Itoh et al. described an HA–collagen composite as a carrier for rhBMP-2 that, after 12 weeks, displayed increased bone mineral density in canine radius, ulna, and tibia defect models [54,68,77]. In another composite formulation, Chen and colleagues demonstrated that gelatin, chitosan, and HA scaffold manufactured through electrospinning and loaded with BMP-2 and VEGF led to accelerated osteogenic and angiogenic growth in a 15 mm rat calvarial defect [78].
Using an FGF-2/calcium phosphate composite layer on HA-ceramic buttons, Tsurushima et al. demonstrated efficacy in bone repair in a 5 mm rat cranial defect [79]. The HA-ceramic buttons were generated by sieving particles below 75 μm, therefore forming disks, which was followed by sintering. Also using FGF-2, Komaki et al. synthesized a β-tricalcium phosphate/collagen scaffold complex to repair a 5 mm rabbit tibial defect, ultimately demonstrating both mechanical and radiological healing at 12-week follow-up, with significantly greater bone formation when compared to the scaffold alone [80].
Three-dimensional printing has also been used to synthesize scaffolds using a variety of materials, most notably ceramics and composites. The printing method selected is determined by the scaffold material, pore size, and geometry [81]. Kolan et al. created a 3D-printed bioactive glass scaffold loaded with BMP-2, which was placed in a 4.6 mm rat calvarial critical-sized defect model [82]. The scaffold was 3D printed in both a traditional cubic formation and a complex biomimetic diamond structure. Interestingly, the diamond structure demonstrated more rapid bone formation as compared to the more traditional architecture. Three-dimensional printing also allows for composite materials to be formed, which may have enhanced osteoconductivity. Teotia et al. created a 3D-printed blend of poly(trimethylene carbonate) and ceramics (TCP or HA) loaded with rhBMP-2, which was placed in an 8 mm rabbit calvarial critical-sized defect [83]. The hydrogel blend used to generate the 3D scaffolds was formulated by dissolving gelatin in deionized water and then adding ceramic polymer resin. The composite scaffold demonstrated improved bony ingrowth compared to poly(trimethylene carbonate) alone, even prior to the addition of rhBMP-2. Through ongoing refinement of materials and architecture, 3D printing offers significant promise to develop highly osteoconductive scaffolds for growth factor delivery in bone repair applications.

2.3. Synthetic Polymers

Synthetic polymers have garnered attention within recent decades as effective delivery systems as the ability to synthesize specific properties allows for control over the drug release pathway. In addition to being easily reproduced, synthetic polymers such as polylactide (PLA), polyglycolide (PLG) and copolymer poly(d,l-lactide-co-glycolide) (PLGA) and PLLA are biodegradable and avoid the immunogenicity risk associated with animal-based biomaterials [54]. Although much research has been directed towards 3D porous scaffolds, other formulations of synthetic polymers have been pursued, such as microspheres, fibers, and sheets [54]. Using a 7.9 mm rabbit calvarial defect model treated with BMP-2 incorporated in PLGA microspheres suspended in carboxymethylcellulose (CMC), Woo et al. created immediate and sustained-release preparations of growth factor by altering the physical properties of the microspheres [54]. PLGA microspheres were synthesized through a double emulsion technique. Sustained-release microspheres had detectable BMP-2 release at 21 days post-implantation compared to 7 days in the immediate-release implants. Additionally, the prolonged-release implant exhibited 75–79% calvarial defect healing compared to the 45% in the immediate-release cohort at 6 weeks post-implantation [54]. In a rat calvarial defect model, Weber et al. demonstrated that rhBMP-2 PLGA microspheres increased bone thickness by almost 100% compared to a rhBMP-2 gelatin-hydrogel cohort [77].
As discussed previously, altering a scaffold’s biochemical makeup can allow for tighter control over its release kinetics. There has been significant interest in creating scaffolds that respond to stimuli such as pH change, temperature, or local enzymatic degradation. In regions of local acidosis, substrates with pH sensitivity, such as poly(acrylic acid), allow for the facilitation of drug delivery. Various polymers combine this pH sensitivity with temperature dependency as well, allowing for phase transitions at physiologic temperatures [84,85,86,87,88,89,90,91]. One such polymer (PCLA-PEG-PCLA) was altered by Kim and colleagues to include thermo- and pH-sensitive components and injected into the dorsum of mice [92]. The thermos- and pH-sensitive polymer demonstrated a phase transition from suspension to gel In vivo, with high encapsulation efficiencies of rhBMP-2 and human mesenchymal stem cells with bone tissue formation after 7 weeks [92]. Additionally, multiple studies have demonstrated that the incorporation of molecules with proteolytic sensitivity, such as susceptibility to matrix metalloprotease (MMP) and/or plasmin, can be developed as novel controlled-release scaffolds [90,93,94,95,96].
Temporal associations between different growth factors have also been assessed. Utilizing a composite scaffold of PPF/gelatin + PLGA microparticles, Kempen et al. designed a temporal system of delivering both VEGF and BMP-2 [97]. PLGA microspheres, generated through double emulsion, were formulated into a composite with PPF through photo-crosslinking. Interestingly, there was significantly greater ectopic bone formation in the combined growth factor treatment group than in the scaffold and BMP-2 alone, with the authors deducing the benefit of sequential angiogenic and osteogenic signals in bone formation [97]. This dual release and synergistic effect of VEGF and BMP-2 in early bone generation was further studied using a porous poly(propylene fumarate) scaffold to repair 5 mm rat femoral defects, resulting at four weeks in significantly higher ectopic bone formation than BMP only, VEGF only, or empty scaffolds [97]. Lastly, Sharmin et al. developed a polymer-coated allograft that sequentially releases VEGF followed by BMP-2 in an attempt to replicate physiologic growth factor cascades [98]. Differential release kinetics were hypothesized to be a function of molecular weight differences between VEGF and BMP-2, resulting in prolonged entrapment of VEGF in surface pores. In a 6 mm rat femoral critical-sized defect model, the VEGF/BMP-2 loaded polymer-coated allograft exhibited significant bone growth from four to eight weeks compared to BMP-2 alone polymer-coated allograft, again suggesting that temporal release of growth factors may enhance osteogenesis [99].

3. Extracellular Vesicles

Extracellular vesicles (EVs) are double phospholipid bilayer nanoparticles that are released by most cell types to mediate cell-to-cell communication [100]. Historically, they have been categorized by size and biogenesis: apoptotic bodies secreted by dying cells and exosomes secreted by viable cells through outward budding of the plasma membrane [101]. They are naturally occurring biological transporters that carry lipids, proteins, nucleic acids, and other bioactive molecules [102]. Additionally, EVs can serve as carriers of growth factors and downstream signaling molecules that have been shown to activate cellular cascades that promote bone remodeling through increased osteoblast proliferation, angiogenesis, and immunoregulation [103]. Their phospholipid bilayer facilitates these functions by acting as a barrier against the rapid clearance of cargo, protecting against enzymatic degradation, and facilitating the crossing of biological membranes [104,105]. For these reasons, there has been an extensive preclinical evaluation of EVs as delivery vehicles of growth factors and downstream signaling molecules as a novel strategy to repair critical-sized defects [100].

3.1. Role of Extracellular Vesicles in Growth Factor Delivery and Bone Repair

The cargo within EVs largely reflects the parent cell from which they originate [106]. The most clinically relevant cell types for derived EVs in bone tissue engineering include osteoblasts [107], osteocytes [108], and mesenchymal stem cells [109,110,111,112]. There has been mounting evidence demonstrating that EVs serve as important mediators of MSC paracrine effects through intercellular communication and transfer of bioactive molecules [100,113]. EVs present an opportunity to improve upon traditional stem cell therapy through enhanced biocompatibility and targeting of cells [113,114,115]. These therapeutic effects have been observed in preclinical models of bone regeneration, and to our knowledge, there is one registered clinical trial utilizing MSC-derived EVs to address bone defects [116]. In addition to highlighting the role of EVs and their respective cargo in bone regeneration (Table 3), we will discuss strategies for engineering EVs to optimize cargo yield and delivery of growth factors (Figure 3).

In Vivo Extracellular Vesicle Mediated Delivery of Growth Factors

The effects of EVs in bone regeneration are mediated by intercellular communication relayed by their cargo [118,120]. Critical-sized defect models have demonstrated the pro-osteogenic and angiogenic effects of EV cargo delivery through the transcriptional upregulation of osteocalcin (OCN), type I collagen (COL I), alkaline phosphatase (ALP), osteopontin (OPN), vascular endothelial factor (VEGF), angiopoietin 1 (ANG1) and angiopoietin 2 (ANG2) [111,126,127]. In a rat femoral nonunion model, animals treated with 1010 particles of bone marrow mesenchymal stem cell-derived EVs (BMMSC-EVs) had significantly increased fracture callus observed on radiographs, improved bone volume on microCT, and histologic evidence of healing on postoperative week 8 compared to non-treated controls [117]. Furthermore, PCR and Western blotting demonstrated increased gene expression of VEGF and hypoxia-inducible factor 1-alpha (HIF-1α) at the nonunion site in the experimental cohort. Additionally, Zhang et al. found an upregulation of proteins involved in the BMP-2/Smad1/RUNX pathway, a signaling cascade found to increase osteogenic differentiation and essential for fracture healing [118,120]. The upregulation of osteogenic proteins and activation of growth factor-mediated signaling cascades implies the presence of crosstalk between EV cargo and pro-osteogenic pathways important for tissue engineering applications [117].

3.2. Engineering Extracellular Vesicles to Optimize Growth Factor Delivery

Although naturally occurring EVs play a role in growth factor delivery, TE strategies have been developed to enhance their therapeutic impact on bone repair. Endogenous and exogenous strategies have demonstrated effectiveness in selectively loading EVs, increasing payload, and improving homing efficiency to bone defects [128,129,130,131]. Endogenous engineering is the enhancement of parent cells to produce desired EV phenotypes, while exogenous engineering is a direct manipulation of EVs. Engineering techniques to optimize EV delivery of growth factors include gene transduction, electroporation, sonication, preconditioning, surface modification, and mechanical manipulation [132,133,134]. Despite ongoing improvements in selective cargo loading, significant variability in preclinical dosing paradigms hinders clinical translation [135]. EV protein weight and particle dosing protocols inadequately assess growth factor quantity and may not be appropriate for clinical application. Although significant challenges remain, ongoing advances in EV cargo analysis offer promise in standardizing doses for the treatment of large bone defects.

3.2.1. Endogenous Engineering of Parent Cells

Endogenous EV cargo loading through engineering parent cells is an attractive strategy to increase growth factor yield and delivery. Preconditioning of parent cells by environmental manipulation is one technique that has been investigated [136]. A common strategy is hypoxic preconditioning of MSCs, as this has been shown to increase the angiogenic effects of secreted EVs as HIF-1α is upregulated and stabilized in hypoxic conditions [137]. In an in vivo study utilizing this method, 100 μL of umbilical mesenchymal stem cell (uMSC) derived EVs stimulated bone healing in a rat femoral fracture model by enhancing the proliferation of human umbilical vein endothelial cells (HUVECs), which in turn, produced angiogenic growth factors [120]. Physical methods of preconditioning have also been investigated. Osteocytes are mechanosensitive cells that alter gene expression when under mechanical stress. Eichholz et al. studied the release of EVs from osteocytes under different loading environments and found that 1 μg of mechanically activated osteocyte-derived EVs (MA-Evs) upregulate histone H4, which promotes osteoblastogenesis through the inhibition of RANKL-RANK [124].
Viral/nonviral transduction (see Section 4.2 and Section 4.3) of parent cells to constitutively express osteogenic and angiogenic-related proteins is another attractive endogenous engineered alternative [122,138]. Transduced parent cells can dramatically increase the production of osteogenic- and angiogenic-related growth factors, which maximizes the therapeutic effects of their released EVs [139]. Li et al. evaluated BMMSCs transfected with an adenovirus to overexpress HIF-1α for the treatment of steroid-induced osteonecrosis of the femoral head in a rabbit model [119]. The EVs released from these cells were isolated and injected into the femoral head and compared to non-transfected control EVs. There was greater micro-vessel density and trabecular bone formation within the transfected EV group secondary to the endogenous cargo loading of growth factor [119].

3.2.2. Exogenous Engineering of Extracellular Vesicles

Exogenous engineering of EVs allows for more precise cargo loading. Two strategies under investigation are electroporation and sonoporation, which cause transient permeability of the plasma membrane to facilitate loading. In an in vivo study using a rat radial defect model, a plasmid carrying VEGF was introduced into EVs derived from a chondrogenic progenitor cell line via electroporation, and 10 μg of the engineered EVs were injected into the defect site, resulting in greater bone formation as shown on microCT compared to unmodified EVs [140]. Alternative methods to enhance the homing efficiency of engineered EVs are also in development. EVs express specific surface proteins that aid in cell targeting and promote communication with their surroundings [141]. Surface modification of EVs has been shown to improve cell targeting and delivery of cargo, therefore enhancing activation of pro-osteogenic signaling cascades [142]. Luo et al. utilized an osteoporotic mouse model with a BMMSC-targeting aptamer that was conjugated to bone marrow stromal cell-derived extracellular vesicles (ST-EVs) to demonstrate enhanced ST-EV homing efficiency [143]. The animals treated with 10 μg of an intravenous injection of the modified EVs demonstrated improvement in bone mineral density compared to the nonmodified EV cohort [143]. The aptamer-conjugated ST-EVs had better accumulation in bone with increased trabecular formation when compared to unconjugated ST-EVs.

3.2.3. Optimizing Extracellular Vesicles for Growth Factor Delivery

A primary concern in utilizing EVs in bone defects is their rapid enzymatic clearance. The addition of scaffolds, as discussed in Section 2.1, Section 2.2 and Section 2.3, can help maintain a local EV concentration by prolonging release [114]. The pairing of scaffolds and engineered EVs has been an area of significant interest to optimize the regenerative effects of EV cargo further. Li et al. found that human ADSC-derived EVs loaded on a PLGA/polydopamine(pDA) scaffold promoted the recruitment of endogenous MSCs to increase bone formation in a mouse 5 mm critical-sized calvarial defect when compared to the scaffold alone [100]. PLGA/pDA loaded with EVs demonstrated sustained release of EVs over eight days compared to four in unmodified PGLA, demonstrating significant potential for optimization of release kinetics [95]. As described earlier, modifications to natural polymers, synthetic polymers, hydrogels, ceramics, and composites have all been described and have been successfully implemented in sustained-release strategies for EVs to enhance the repair of bone defects [144,145,146,147].

4. Role of Regional Gene Therapy in Growth Factor Delivery

Delivery options for growth factors discussed in earlier sections of this review rely upon the in-situ biology of the fracture environment to provide osteogenic cells to aid in bone healing. Despite the highly regulated repair process that exists [148], critical-sized defects disrupt the environment beyond the endogenous repair capabilities, resulting in nonunion. Advances in tissue engineering over the last few decades have yielded technology that helps to address these challenges. Local delivery of nucleic acids or cells transduced with genetic sequences that express growth factors has shown promise in overcoming the obstacles posed by disrupted healing environments. The strength of regional gene therapy is that osteoinductive growth factors can be directed to the defects along with essential cell populations placed on an osteoconductive scaffold, therefore overcoming the disrupted healing process [149].

4.1. In Vivo Versus Ex Vivo Regional Gene Therapy

In vivo and ex vivo regional gene therapy techniques have been developed with successful application in bone regeneration. In vivo therapy involves either viral or nonviral vectors carrying genes of interest being directly administered either locally or systemically to the patient. The vector is then free to transfer genetic material to cellular populations either through specific or nonspecific binding. The strength of in vivo gene therapy is that it is performed as a single-stage procedure without the need for prior cell harvest. This has significant potential advantages over ex vivo therapy from a clinical feasibility and cost reduction perspective, but there are certain limitations associated with this strategy. Challenges with in vivo regional gene therapy include nonspecific selection of target cells, inability to place an osteoconductive scaffold, and inefficient transduction. Despite these limitations, there continues to be significant interest in the development of in vivo regional gene therapy with promising preclinical results in bone regeneration [149].
Ex vivo gene therapy requires harvesting autologous or allogeneic cells of interest, culturing and expanding cell populations, and finally, transducing genetic material prior to implantation at the site of interest. One of the advantages of ex vivo regional gene therapy is that a large quantity of healthy transduced cells can be prepared, which is important for healing large defects. There has been a significant effort to identify cells that can be abundantly harvested, easily expanded, consistently transduced, and highly expressed growth factors [2]. The osteogenic potential of human adipose-derived stem cells and human bone marrow-derived stem cells (BMMSC) has been evaluated in bone-healing applications [150,151]. Interestingly, lentivirus BMP-2 transduced human adipose-derived stem cells resulted in significantly higher levels of BMP-2 production in vitro and also superior healing in a rat critical-sized femoral defect model (Figure 4) [150,151]. Furthermore, adipose tissue is readily harvested through liposuction procedures rather than more invasive bone marrow harvests from the iliac crest, which is an important consideration for clinical use.
The drawback to ex vivo therapy is that autologous cells often need to be collected in staged procedures to allow time for preparation. To overcome this obstacle for clinical development, same-day techniques have been developed and proven effective [150,152]. Additionally, allogeneic cells may be feasible protein carriers, which would allow the preparation of transduced cells in advance of definitive treatments. The development of an “off-the-shelf” product would significantly improve the utility of ex vivo therapy in clinical applications.

4.2. Viral Vectors

Viral vectors have been successfully employed in regional gene therapy in preclinical applications for bone repair [20]. Viral vectors are replication incompetent viruses that have been genetically modified to carry transgenes that can be delivered to cellular populations, as shown in Figure 5. Historically, these vectors have been more efficient than nonviral vectors in transducing cells to express genes, although safety concerns include off-target transduction, severe immune response, and potential tumorigenesis [149]. Despite these challenges, viral vectors have been employed for the treatment of Wiskott–Aldrich syndrome, leukodystrophies, thalassemias, familial lipoprotein lipase deficiency, congenital amaurosis, and other genetic conditions [153,154,155,156]. Additionally, in preclinical models, viral vectors carrying genetic sequences for growth factors have been used to heal fractures and critical-sized defects as well as enhance spinal fusions. Selection of viral vectors is critical to this technique, given differences in immunogenicity, tropism, duration of gene expression, and packaging capabilities.
The first viral vectors utilized in bone tissue engineering were derivatives of adenovirus due to their low pathogenicity, high tropism, ability to carry large DNA sequences, and familiarity with oncogenic applications [157,158,159,160]. The major challenge posed by AV vectors is that they do not integrate into the host genome, which limits the duration of gene expression [161,162]. Additionally, AV vectors have significant immunogenicity, often requiring the use of immune-incompetent preclinical models [163,164,165].
Adeno-associated virus (AAV) has also been investigated as a potential vector for the delivery of growth factor in regional gene therapy for bone repair, given its reduced immunogenicity, transduction of dividing and nondividing cells, and prolonged gene expression [166]. Although AAV has proven effective in gene therapy applications, complex packaging restricts transgene size to less than 5.0 kb, which severely limits its efficiency in bone repair [167]. Additionally, in vitro studies have demonstrated poor transduction potential with significantly reduced BMP-2 production without osteogenic differentiation in multiple human stem cell lines compared to a lentiviral vector (LV) [168].
Lentivirus is a single-stranded RNA virus derived from HIV-1, which integrates into the host genome, allowing for prolonged transgene expression [169]. Furthermore, LV can infect nondividing cells and integrate into host-transcribed gene regions rather than regulatory sites, which reduces its oncogenic potential [153]. Safety modifications have been implemented for third-generation LV vectors, including the deletion of non-essential viral proteins and the separation of genomes to multiple plasmids, therefore reducing the probability of reconstitution [170,171]. LV has been successfully applied in multiple preclinical models of bone healing and spinal fusions due to its robust transduction efficacy, sustained expression of growth factor transgenes, and low immunogenicity profile [161,163,172,173].

Viral Vector Delivery of Growth Factor in Bone Repair

Viral vectors have shown significant promise in regional gene therapy for the delivery of growth factors in bone repair applications, as demonstrated by our lab. They have predominantly been used to transduce the BMP-2 gene, as shown in Table 4 and Table 5, but there are preclinical studies investigating their use with other growth factors as well. An early study by Lieberman et al. used an AV vector carrying a BMP-2 cDNA (Ad-BMP-2) to transfect autologous rat bone marrow cells (RBMC) implanted onto a DBM carrier [20]. The loaded carrier was placed in an 8 mm critical defect in a rat hind leg, which was successfully healed at 12 weeks postoperatively. Additionally, biomechanical properties were restored in the operated limb when compared to contralateral controls. AV vectors have also been successfully employed in spinal fusion models utilizing multiple growth factors, including BMP-2, BMP-6, BMP-7, and BMP-9 [20,174,175,176,177,178]. In these studies, AV-BMP was administered through in vivo or in vitro techniques, with successful fusion seen on radiographs and microCT demonstrating continuous contact with posterior spinal elements. Challenges with using AV to deliver growth factors have also become apparent in preclinical models. Significant inflammatory responses have been observed with the use of Ad-BMP-2, which can inhibit the production of ectopic bone; however, administration of immunosuppressive agents such as tacrolimus alongside Ad-BMP-2 may reduce the immune response and improve bone regeneration [164,165,179].
Given the favorable tissue engineering profile for LV vectors, as discussed previously, these have been extensively employed in the delivery of growth factors in bone-healing applications. Viral integration of LV can provide sustained release of transduced BMP-2 for longer than 3 months, which has been shown to improve healing rates on XR, increase bone volume to tissue volume on microCT, and increase energy to failure on biomechanical testing when compared to AV-BMP-2 in a critical-sized rat radius defect model [161,162]. Additionally, preclinical studies have demonstrated success with transducing human-harvested cells to treat critical-sized defects in animal models. Vakhshori et al. obtained adipose tissue from elective liposuction procedures of female donors and isolated human adipose-derived stem cells (hASC), which were transduced using an LV-BMP-2 vector (Figure 6) [151]. The transduced hASCs were then loaded on a tricalcium phosphate/hydroxyapatite carrier and placed in an athymic rat 6 mm critical-sized femoral defect model. An athymic rat model was used to avoid an immune response to the hASCs. At 12 weeks postoperatively, 13 of 14 animals with LV-BMP-2 hASCs successfully healed the femoral defect, which was equivalent to treatment with rhBMP-2. Furthermore, the LV-BMP-2 hASC cohort demonstrated significantly increased bone volume on microCT, greater circumferential bone formation on histology, and improved biomechanics compared to non-transduced cohorts. The ongoing investigation of LV-transduced human-derived stem cells provides an exciting opportunity for the bench-to-bedside application of this technology.
In addition to the delivery of BMP, there are a few studies demonstrating the feasibility of regional gene therapy in the delivery of alternative growth factors in bone repair. Peng et al. prepared a retroviral vector with VEGF and BMP-4 cDNAs, which were used in combination to transduce muscle-derived stem cells loaded on a gelatin sponge to heal a mouse 6 mm calvarial defect [186]. Interestingly, combined VEGF/BMP-4 regional gene therapy enhanced bone formation but only at ratios of 1:5 and 1:1, with inhibitory effects seen at 5:1. Ito et al. demonstrated in a rat fracture model that 4 mm structural allograft coated with AAV-VEGF and AAV-RANKL could revitalize the tissue resulting in remodeling of the graft with radiographic evidence of healing [187]. Lastly, in a rat alveolar defect model, Ad-PDGF placed on a type I collagen sponge demonstrated significantly improved trabecular bone thickness, defect filling, and biomechanical properties compared to a sponge with Ad-Luciferase control [188].

4.3. Nonviral Vector Delivery of Growth Factor in Bone Repair

Given concerns of immunogenicity, pathogenicity, and possible tumorigenesis with the use of viral vectors, there have been significant efforts underway to develop nonviral transduction techniques. Traditionally, these techniques have resulted in low levels of transgene expression in target cells, which has limited their application in bone regeneration. Despite these challenges, preclinical applications have utilized liposomes, polymers, plasmids, sonoporation, and electroporation to deliver growth factor genes into target cell populations with varying success [189,190,191,192].
Although less frequently applied than viral vector-mediated regional gene therapy, multiple groups have demonstrated bone regeneration using nonviral methods. Sonoporation has been investigated as a potential physical delivery mechanism, given its noninvasive nature and technical reproducibility [190,191]. Bez et al. created a miniature pig 1 cm critical-sized tibial defect model and placed a collagen scaffold within the wound [190]. By postoperative day 14, the experimental group received an injection of BMP-6 plasmid mixed with microbubbles at the fracture site, followed by ultrasonic pulses until bubble oscillations were no longer visualized. At 5 days post-injection, only 40% of the cells at the defect were found to be transduced; however, there was 120-fold higher BMP-6 production compared to empty plasmid controls. At 8 weeks postoperatively, there was 75% restoration of the defect, which was similar to autograft controls with biomechanical equivalence. Feichtinger et al. also used sonoporation to transduce a plasmid containing both BMP-2 and BMP-7 genes into cells to heal a rat 4 mm femoral defect model [192]. Interestingly, passive gene transfer was found to have a 61% transduction success compared to 100% of the animals with ultrasonic pulses. Unfortunately, only 2/6 (33%) experimental animals had successful bony unions, as seen on microCT, compared to 1/6 (16.7%) with passive transduction alone.
Alternative nonviral techniques have made use of polymers to effectively deliver growth factors in preclinical models. Curtin et al. compared the effectiveness of polyethyleneimine (PEI) and nano-hydroxyapatite (nHA) vectors loaded on collagen-nHA scaffolds to heal a mouse 7 mm cranial defect by creating PEI-BMP-2/PEI-VEGF and nHA-BMP-2/nHA-VEGF [193]. Interestingly, PEI-BMP-2 and PEI-VEGF resulted in the highest mesenchymal stem cell (MSC) growth factor production; however, in vitro studies demonstrated greater calcium deposition with nHA-BMP-2 and similar endothelial proliferation with both VEGF vectors. Furthermore, nHA-BMP-2/nHA-VEGF dual therapy loaded on a hydroxyapatite scaffold resulted in the greatest healing in a mouse cranial defect model with PEI-BMP-2/PEI-VEGF dual therapy, not resulting in improvement compared to scaffold alone.

4.4. Role of Scaffolds in Gene Therapy

In addition to providing an osteoinductive surface for bony ingrowth, scaffold design greatly impacts the controlled delivery of growth factors in regional gene therapy. As discussed previously, scaffolds can be composed of various biomaterials and can transport not only recombinant protein but also plasmid DNA (pDNA), chemically modified RNA, and genetically modified MSCs. One area of interest is the utilization of 3D-printed scaffolds, which have the added benefit of being customized to fit any bone defect that may be encountered and clearly have clinical potential. Alluri et al. obtained preoperative microCT scans of a 24-week-old rat’s femur, which was templated for a 3D printed β-tricalcium phosphate scaffold [194]. The 3D printed scaffold was then loaded with LV-BMP-2 transduced RBMCs prior to implantation in a rat 6 mm femoral critical-sized defect. Interestingly, all rats with transduced RBMCs loaded onto the scaffold demonstrated healing at 12 weeks postoperatively. The ongoing development of 3D-printed scaffolds in regional gene therapy presents an opportunity to treat clinically complex defects that off-the-shelf products would inadequately address.
Additionally, scaffolds can be used in a nonviral context, forming what are traditionally termed gene-activated matrices (GAMs). Challenges with GAMs include low transduction efficacy in a limited population of available osteoprogenitor cells. Despite these obstacles, GAMs have been used in clinical applications of bone regeneration. Bozo et al. employed a collagen-hydroxyapatite scaffold and pDNA-VEGF complex to repair a mandibular nonunion in a 37-year-old patient with a resected fibrous dysplasia who failed multiple bone grafting procedures [195]. At 12 months postoperatively, Bozo et al. reported that there was significant integration of the GAM, healing of the defect, and remodeling of the nonunion site without observed adverse events [195]. Given this early success, a nonrandomized trial was conducted with 20 patients with maxillofacial bone defects who were treated with a GAM composed of ostacalcium-carrying pDNA-VEGF [196]. At 6 months postoperatively, all patients demonstrated significantly increased bone density and bone healing with the grafting area composed of newly formed tissue and GAM resorption.

5. Conclusions and Future Directions

Delivery of growth factor for repair of large bone defects is a promising solution to this complex clinical challenge. As discussed, local delivery of growth factors, extracellular vesicles, and regional gene therapy continue to be viable options for further clinical development. Significant challenges remain, including the development of osteoinductive scaffolds with sustained growth factor release, selective loading of extracellular vesicles, and optimization of gene therapy for reduced immunogenicity. A major advancement in the development of all these technologies will likely be achieved through improved scaffold design. Although natural polymers and ceramics are most prevalent in clinical practice today, improvements in synthetic polymers and signal-responsive scaffolds can dramatically improve the delivery of growth factors. Additionally, an increased understanding of extracellular vesicles and their cargo has the potential to change how stem cells are utilized in bone regeneration applications. Engineered EVs with intracellular osteoinductive cargo may allow for modulation of molecular pathways not currently achievable with local delivery of growth factor. Furthermore, an improved understanding of biologically critical pathways for bone regeneration, such as SMAD, WNT, Notch, and Hedge Hodge, may provide alternative therapeutic targets [197]. Emerging technology such as CRISPR and the use of miRNA may greatly improve our ability to target cellular pathways through modification of parent cells [198,199,200,201]. Finally, regional gene therapy has significant clinical potential. The combination of locally produced osteoinductive signaling generated through selected cellular populations placed on an osteoconductive scaffold has the potential to revolutionize how these bone defects are treated in clinical settings [198,199,200,201]. Ongoing development of ex vivo allogeneic regional gene therapy provides an exciting opportunity to develop an “off-the-shelf” therapeutic to treat large bone defects [202,203].

Author Contributions

Conceptualization, J.R.B., C.K.M. and J.R.L.; Methodology, J.R.B., T.S. and F.H.; Software, F.H.; Validation, J.R.B., C.K.M. and J.R.L.; Investigation, J.R.L., T.S. and F.H.; Resources, J.R.L.; Data Curation, J.R.B., C.K.M. and J.R.L.; Writing—Original Draft Preparation, J.R.B., T.S. and F.H.; Writing—Review and Editing, J.R.B., C.K.M. and J.R.L.; Supervision, J.R.B. and J.R.L.; Project Administration, J.R.B. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data was reported in this review article. Cited studies can be accessed from the references already included.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Brinker, M.R.; O’Connor, D.P. The biological basis for nonunions. JBJS Rev. 2016, 4, e3. [Google Scholar] [CrossRef] [PubMed]
  2. Carofino, B.C.; Lieberman, J.R. Gene Therapy Applications for Fracture-Healing. JBJS 2008, 90, 99. [Google Scholar] [CrossRef] [PubMed]
  3. Nauth, A.; Lee, M.; Gardner, M.J.; Brinker, M.R.; Warner, S.J.; Tornetta, P.; Leucht, P. Principles of Nonunion Management: State of the Art. J. Orthop. Trauma 2018, 32, S52–S57. [Google Scholar] [CrossRef] [PubMed]
  4. Sen, M.K.; Miclau, T. Autologous iliac crest bone graft: Should it still be the gold standard for treating nonunions? Injury 2021, 52, S18–S22. [Google Scholar] [CrossRef] [PubMed]
  5. Garrison, K.; Shemilt, I.; Donell, S.; Ryder, J.; Mugford, M.; Harvey, I.; Song, F. Bone morphogenetic protein (BMP) for fracture healing in adults. Cochrane Database Syst. Rev. 2008, 2010, CD006950. [Google Scholar] [CrossRef]
  6. Gulabi, D.; Erdem, M.; Cecen, G.S.; Avci, C.C.; Saglam, N.; Saglam, F. Ilizarov Fixator Combined With an Intramedullary Nail for Tibial Nonunions With Bone Loss: Is It Effective? Clin. Orthop. Relat. Res. 2014, 472, 3892–3901. [Google Scholar] [CrossRef]
  7. Giannoudis, P.V.; Faour, O.; Goff, T.; Kanakaris, N.; Dimitriou, R. Masquelet technique for the treatment of bone defects: Tips-tricks and future directions. Injury 2011, 42, 591–598. [Google Scholar] [CrossRef]
  8. Baldwin, P.; Li, D.J.; Auston, D.A.; Mir, H.S.; Yoon, R.S.; Koval, K.J. Autograft, Allograft, and Bone Graft Substitutes: Clinical Evidence and Indications for Use in the Setting of Orthopaedic Trauma Surgery. J. Orthop. Trauma 2019, 33, 203–213. [Google Scholar] [CrossRef] [PubMed]
  9. Bostrom, M.P.G.; Seigerman, D.A. The Clinical Use of Allografts, Demineralized Bone Matrices, Synthetic Bone Graft Substitutes and Osteoinductive Growth Factors: A Survey Study. HSS J. 2005, 1, 9–18. [Google Scholar] [CrossRef]
  10. Perez, J.R.; Kouroupis, D.; Li, D.J.; Best, T.M.; Kaplan, L.; Correa, D. Tissue Engineering and Cell-Based Therapies for Fractures and Bone Defects. Front. Bioeng. Biotechnol. 2018, 6, 340516. [Google Scholar] [CrossRef]
  11. O’Keefe, R.J.; Mao, J. Bone tissue engineering and regeneration: From discovery to the clinic—An overview. Tissue Eng. Part B Rev. 2011, 17, 389–392. [Google Scholar] [CrossRef]
  12. Gruskin, E.; Doll, B.A.; Futrell, F.W.; Schmitz, J.P.; Hollinger, J.O. Demineralized bone matrix in bone repair: History and use. Adv. Drug Deliv. Rev. 2012, 64, 1063–1077. [Google Scholar] [CrossRef]
  13. Watson, T.J. Distraction Osteogenesis. JAAOS J. Am. Acad. Orthop. Surg. 2006, 14, S168–S174. [Google Scholar] [CrossRef]
  14. Dimitriou, R.; Mataliotakis, G.I.; Angoules, A.G.; Kanakaris, N.K.; Giannoudis, P.V. Complications following autologous bone graft harvesting from the iliac crest and using the RIA: A systematic review. Injury 2011, 42, S3–S15. [Google Scholar] [CrossRef]
  15. Urist, M.R. Bone: Formation by autoinduction. Science 1965, 150, 893–899. [Google Scholar] [CrossRef] [PubMed]
  16. Urist, M.R.; Strates, B.S. Bone Morphogenetic Protein. J. Dent. Res. 1971, 50, 1392–1406. [Google Scholar] [CrossRef] [PubMed]
  17. Reddi, A.H.; Anderson, W.A. Collagenous bone matrix-induced endochondral ossification hemopoiesis. J. Cell Biol. 1976, 69, 557–572. [Google Scholar] [CrossRef]
  18. Muthukumaran, N.; Ma, S.; Reddi, A.H. Dose-dependence of and threshold for optimal bone induction by collagenous bone matrix and osteogenin-enriched fraction. Coll. Relat. Res. 1988, 8, 433–441. [Google Scholar] [CrossRef] [PubMed]
  19. Wozney, J.M.; Rosen, V.; Celeste, A.J.; Mitsock, L.M.; Whitters, M.J.; Kriz, R.W.; Hewick, R.M.; Wang, E.A. Novel regulators of bone formation: Molecular clones and activities. Science 1988, 242, 1528–1534. [Google Scholar] [CrossRef]
  20. Lieberman, J.R.; Daluiski, A.; Stevenson, S.; Jolla, L.; Wu, L.; McAllister, P.; Lee, Y.P.; Kabo, J.M.; Finerman, G.A.; Berk, A.J.; et al. The Effect of Regional Gene Therapy with Bone Morphogenetic Protein-2-Producing Bone-Marrow Cells on the Repair of Segmental Femoral Defects in Rats. JBJS 1999, 81, 905–917. [Google Scholar] [CrossRef]
  21. ElHawary, H.; Baradaran, A.; Abi-Rafeh, J.; Vorstenbosch, J.; Xu, L.; Efanov, J.I. Bone Healing and Inflammation: Principles of Fracture and Repair. Semin. Plast. Surg. 2021, 35, 198–203. [Google Scholar] [CrossRef] [PubMed]
  22. Dimitriou, R.; Tsiridis, E.; Giannoudis, P.V. Current concepts of molecular aspects of bone healing. Injury 2005, 36, 1392–1404. [Google Scholar] [CrossRef] [PubMed]
  23. Gillman, C.E.; Jayasuriya, A.C. FDA-approved bone grafts and bone graft substitute devices in bone regeneration. Mater. Sci. Eng. C 2021, 130, 112466. [Google Scholar] [CrossRef]
  24. FDA. Recombinant Human Bone Morphogenetic Protein; FDA: Silver Spring, MD, USA, 2023. [Google Scholar]
  25. DiGiovanni, C.W.; Petricek, J.M. The evolution of rhPDGF-BB in musculoskeletal repair and its role in foot and ankle fusion surgery. Foot Ankle Clin. 2010, 15, 621–640. [Google Scholar] [CrossRef]
  26. Daniels, T.R.; Younger, A.S.E.; Penner, M.J.; Wing, K.J.; Le, I.L.D.; Russell, I.S.; Lalonde, K.-A.; Evangelista, P.T.; Quiton, J.D.; Glazebrook, M.; et al. Prospective Randomized Controlled Trial of Hindfoot and Ankle Fusions Treated With rhPDGF-BB in Combination With a β-TCP-Collagen Matrix. Foot Ankle Int. 2015, 36, 739–748. [Google Scholar] [CrossRef] [PubMed]
  27. Daniels, T.R.; Anderson, J.; Swords, M.P.; Maislin, G.; Donahue, R.; Pinsker, E.; Quiton, J.D. Recombinant Human Platelet-Derived Growth Factor BB in Combination With a Beta-Tricalcium Phosphate (rhPDGF-BB/β-TCP)-Collagen Matrix as an Alternative to Autograft. Foot Ankle Int. 2019, 40, 1068–1078. [Google Scholar] [CrossRef]
  28. Digiovanni, C.W.; Baumhauer, J.; Lin, S.S.; Berberian, W.S.; Flemister, A.S.; Enna, M.J.; Evangelista, P.; Newman, J. Prospective, randomized, multi-center feasibility trial of rhPDGF-BB versus autologous bone graft in a foot and ankle fusion model. Foot Ankle Int. 2011, 32, 344–354. [Google Scholar] [CrossRef]
  29. Street, J.; Bao, M.; DeGuzman, L.; Bunting, S.; Peale, F.V.; Ferrara, N.; Steinmetz, H.; Hoeffel, J.; Cleland, J.L.; Daugherty, A.; et al. Vascular endothelial growth factor stimulates bone repair by promoting angiogenesis and bone turnover. Proc. Natl. Acad. Sci. USA 2002, 99, 9656–9661. [Google Scholar] [CrossRef]
  30. Keramaris, N.C.; Calori, G.M.; Nikolaou, V.S.; Schemitsch, E.H.; Giannoudis, P.V. Fracture vascularity and bone healing: A systematic review of the role of VEGF. Injury 2008, 39, S45–S57. [Google Scholar] [CrossRef]
  31. Eckardt, H.; Ding, M.; Lind, M.; Hansen, E.S.; Christensen, K.S.; Hvid, I. Recombinant human vascular endothelial growth factor enhaces bone healing in an experimental nonunion model. J. Bone Jt. Surg. Ser. B 2005, 87, 1434–1438. [Google Scholar] [CrossRef]
  32. Kawai, M.; Rosen, C.J. The Insulin-Like Growth Factor System in Bone: Basic and Clinical Implications. Endocrinol. Metab. Clin. N. Am. 2012, 41, 323–333. [Google Scholar] [CrossRef]
  33. Reible, B.; Schmidmaier, G.; Moghaddam, A.; Westhauser, F. Insulin-Like Growth Factor-1 as a Possible Alternative to Bone Morphogenetic Protein-7 to Induce Osteogenic Differentiation of Human Mesenchymal Stem Cells In Vitro. Int. J. Mol. Sci. 2018, 19, 1674. [Google Scholar] [CrossRef]
  34. Fei, Q.; Li, J.; Lin, J.; Li, D.; Wang, B.; Meng, H.; Wang, Q.; Su, N.; Yang, Y. Risk Factors for Surgical Site Infection After Spinal Surgery: A Meta-Analysis. World Neurosurg. 2016, 95, 507–515. [Google Scholar] [CrossRef]
  35. Radomsky, M.L.; Aufdemorte, T.B.; Swain, L.D.; Fox, W.C.; Spiro, R.C.; Poser, J.W. Novel formulation of fibroblast growth factor-2 in a hyaluronan gel accelerates fracture healing in nonhuman primates. J. Orthop. Res. 1999, 17, 607–614. [Google Scholar] [CrossRef] [PubMed]
  36. Charoenlarp, P.; Rajendran, A.K.; Iseki, S. Role of fibroblast growth factors in bone regeneration. Inflamm. Regen. 2017, 37, 10. [Google Scholar] [CrossRef] [PubMed]
  37. Rosier, R.N.; O’Keefe, R.J.; Hicks, D.G. The Potential Role of Transforming Growth Factor Beta in Fracture Healing. Clin. Orthop. Relat. Res. (1976–2007) 1998, 355, S294–S300. [Google Scholar] [CrossRef] [PubMed]
  38. Nauth, A.; Ristevski, B.; Li, R.; Schemitsch, E.H. Growth factors and bone regeneration: How much bone can we expect? Injury 2011, 42, 574–579. [Google Scholar] [CrossRef] [PubMed]
  39. Cao, X.; Chen, D. The BMP signaling and in vivo bone formation. Gene 2005, 357, 1–8. [Google Scholar] [CrossRef]
  40. Salazar, V.S.; Gamer, L.W.; Rosen, V. BMP signalling in skeletal development, disease and repair. Nat. Rev. Endocrinol. 2016, 12, 203–221. [Google Scholar] [CrossRef]
  41. Wang, R.N.; Green, J.; Wang, Z.; Deng, Y.; Qiao, M.; Peabody, M.; Zhang, Q.; Ye, J.; Yan, Z.; Denduluri, S.; et al. Bone Morphogenetic Protein (BMP) signaling in development and human diseases. Genes Dis. 2014, 1, 87–105. [Google Scholar] [CrossRef]
  42. Zhu, L.; Liu, Y.; Wang, A.; Zhu, Z.; Li, Y.; Zhu, C.; Che, Z.; Liu, T.; Liu, H.; Huang, L. Application of BMP in Bone Tissue Engineering. Front. Bioeng. Biotechnol. 2022, 10, 810880. [Google Scholar] [CrossRef]
  43. Gipson, G.R.; Goebel, E.J.; Hart, K.N.; Kappes, E.C.; Kattamuri, C.; McCoy, J.C.; Thompson, T.B. Structural perspective of BMP ligands and signaling. Bone 2020, 140, 115549. [Google Scholar] [CrossRef]
  44. El Bialy, I.; Jiskoot, W.; Reza Nejadnik, M. Formulation, Delivery and Stability of Bone Morphogenetic Proteins for Effective Bone Regeneration. Pharm. Res. 2017, 34, 1152–1170. [Google Scholar] [CrossRef] [PubMed]
  45. Bessa, P.C.; Casal, M.; Reis, R.L. Bone morphogenetic proteins in tissue engineering: The road from laboratory to clinic, part II (BMP delivery). J. Tissue Eng. Regen. Med. 2008, 2, 81–96. [Google Scholar] [CrossRef]
  46. Sampath, T.K.; Reddi, A.H. Dissociative extraction and reconstitution of extracellular matrix components involved in local bone differentiation. Proc. Natl. Acad. Sci. USA 1981, 78, 7599–7603. [Google Scholar] [CrossRef] [PubMed]
  47. Cahill, K.S.; Chi, J.H.; Day, A.; Claus, E.B. Prevalence, complications, and hospital charges associated with use of bone-morphogenetic proteins in spinal fusion procedures. JAMA 2009, 302, 58–66. [Google Scholar] [CrossRef]
  48. Kelly, M.P.; Savage, J.W.; Bentzen, S.M.; Hsu, W.K.; Ellison, S.A.; Anderson, P.A. Cancer risk from bone morphogenetic protein exposure in spinal arthrodesis. J. Bone Jt. Surg. Am. 2014, 96, 1417–1422. [Google Scholar] [CrossRef] [PubMed]
  49. Cooper, G.S.; Kou, T.D. Risk of Cancer Following Lumbar Fusion Surgery With Recombinant Human Bone Morphogenic Protein-2 (rhBMP-2): An Analysis Using a Commercially Insured Patient Population. Int. J. Spine Surg. 2018, 12, 260–268. [Google Scholar] [CrossRef]
  50. Chrastil, J.; Low, J.B.; Whang, P.G.; Patel, A.A. Complications associated with the use of the recombinant human bone morphogenetic proteins for posterior interbody fusions of the lumbar spine. Spine 2013, 38, E1020–E1027. [Google Scholar] [CrossRef] [PubMed]
  51. Mesfin, A.; Buchowski, J.M.; Zebala, L.P.; Bakhsh, W.R.; Aronson, A.B.; Fogelson, J.L.; Hershman, S.; Kim, H.J.; Ahmad, A.; Bridwell, K.H. High-dose rhBMP-2 for adults: Major and minor complications: A study of 502 spine cases. J. Bone Jt. Surg. Am. 2013, 95, 1546–1553. [Google Scholar] [CrossRef]
  52. Smucker, J.D.; Rhee, J.M.; Singh, K.; Yoon, S.T.; Heller, J.G. Increased swelling complications associated with off-label usage of rhBMP-2 in the anterior cervical spine. Spine 2006, 31, 2813–2819. [Google Scholar] [CrossRef] [PubMed]
  53. Wen, Y.-D.; Jiang, W.-M.; Yang, H.-L.; Shi, J.-H. Exploratory meta-analysis on dose-related efficacy and complications of rhBMP-2 in anterior cervical discectomy and fusion: 1,539,021 cases from 2003 to 2017 studies. J. Orthop. Transl. 2020, 24, 166–174. [Google Scholar] [CrossRef] [PubMed]
  54. Woo, B.H.; Fink, B.F.; Page, R.; Schrier, J.A.; Jo, Y.W.; Jiang, G.; DeLuca, M.; Vasconez, H.C.; DeLuca, P.P. Enhancement of bone growth by sustained delivery of recombinant human bone morphogenetic protein-2 in a polymeric matrix. Pharm. Res. 2001, 18, 1747–1753. [Google Scholar] [CrossRef] [PubMed]
  55. McKay, W.F.; Peckham, S.M.; Badura, J.M. A comprehensive clinical review of recombinant human bone morphogenetic protein-2 (INFUSE Bone Graft). Int. Orthop. 2007, 31, 729–734. [Google Scholar] [CrossRef]
  56. Visser, R.; Arrabal, P.M.; Becerra, J.; Rinas, U.; Cifuentes, M. The effect of an rhBMP-2 absorbable collagen sponge-targeted system on bone formation in vivo. Biomaterials 2009, 30, 2032–2037. [Google Scholar] [CrossRef]
  57. Vo, T.N.; Kasper, F.K.; Mikos, A.G. Strategies for controlled delivery of growth factors and cells for bone regeneration. Adv. Drug Deliv. Rev. 2012, 64, 1292–1309. [Google Scholar] [CrossRef]
  58. Arias-Betancur, A.; Badilla-Wenzel, N.; Astete-Sanhueza, Á.; Farfán-Beltrán, N.; Dias, F.J. Carrier systems for bone morphogenetic proteins: An overview of biomaterials used for dentoalveolar and maxillofacial bone regeneration. Jpn. Dent. Sci. Rev. 2022, 58, 316–327. [Google Scholar] [CrossRef] [PubMed]
  59. Govender, S.; Csimma, C.; Genant, H.K.; Valentin-Opran, A. Recombinant Human Bone Morphogenetic Protein-2 for Treatment of Open Tibial Fractures. J. Bone Jt. Surg.-Am. Vol. 2002, 84, 2123–2134. [Google Scholar] [CrossRef]
  60. Xu, Q.; Torres, J.E.; Hakim, M.; Babiak, P.M.; Pal, P.; Battistoni, C.M.; Nguyen, M.; Panitch, A.; Solorio, L.; Liu, J.C. Collagen- and hyaluronic acid-based hydrogels and their biomedical applications. Mater. Sci. Eng. R Rep. Rev. J. 2021, 146, 100641. [Google Scholar] [CrossRef] [PubMed]
  61. Burkus, J.K.; Gornet, M.F.; Dickman, C.A.; Zdeblick, T.A. Anterior lumbar interbody fusion using rhBMP-2 with tapered interbody cages. J. Spinal Disord. Tech. 2002, 15, 337–349. [Google Scholar] [CrossRef]
  62. Burkus, J.K.; Haid, R.W.; Traynelis, V.C.; Mummaneni, P.V. Long-term clinical and radiographic outcomes of cervical disc replacement with the Prestige disc: Results from a prospective randomized controlled clinical trial. J. Neurosurg. Spine 2010, 13, 308–318. [Google Scholar] [CrossRef] [PubMed]
  63. Kigami, R.; Sato, S.; Tsuchiya, N.; Yoshimakai, T.; Arai, Y.; Ito, K. FGF-2 angiogenesis in bone regeneration within critical-sized bone defects in rat calvaria. Implant Dent. 2013, 22, 422–427. [Google Scholar] [CrossRef] [PubMed]
  64. Ueda, H.; Hong, L.; Yamamoto, M.; Shigeno, K.; Inoue, M.; Toba, T.; Yoshitani, M.; Nakamura, T.; Tabata, Y.; Shimizu, Y. Use of collagen sponge incorporating transforming growth factor-beta1 to promote bone repair in skull defects in rabbits. Biomaterials 2002, 23, 1003–1010. [Google Scholar] [CrossRef]
  65. Lee, J.-S.; Lee, S.-K.; Kim, B.-S.; Im, G.-I.; Cho, K.-S.; Kim, C.-S. Controlled release of BMP-2 using a heparin-conjugated carrier system reduces in vivo adipose tissue formation. J. Biomed. Mater. Res. A 2015, 103, 545–554. [Google Scholar] [CrossRef]
  66. García-García, P.; Reyes, R.; Pérez-Herrero, E.; Arnau, M.R.; Évora, C.; Delgado, A. Alginate-hydrogel versus alginate-solid system. Efficacy in bone regeneration in osteoporosis. Mater. Sci. Eng. C 2020, 115, 111009. [Google Scholar] [CrossRef]
  67. Itoh, T.; Mochizuki, M.; Nishimura, R.; Matsunaga, S.; Kadosawa, T.; Kokubo, S.; Yokota, S.; Sasaki, N. Repair of Ulnar Segmental Defect by Recombinant Human Bone Morphogenetic Protein-2 in Dogs. J. Vet. Med. Sci. 1998, 60, 451–458. [Google Scholar] [CrossRef] [PubMed]
  68. Itoh, S.; Kikuchi, M.; Takakuda, K.; Nagaoka, K.; Koyama, Y.; Tanaka, J.; Shinomiya, K. Implantation study of a novel hydroxyapatite/collagen (HAp/col) composite into weight-bearing sites of dogs. J. Biomed. Mater. Res. 2002, 63, 507–515. [Google Scholar] [CrossRef]
  69. Lee, S.S.; Huang, B.J.; Kaltz, S.R.; Sur, S.; Newcomb, C.J.; Stock, S.R.; Shah, R.N.; Stupp, S.I. Bone regeneration with low dose BMP-2 amplified by biomimetic supramolecular nanofibers within collagen scaffolds. Biomaterials 2013, 34, 452–459. [Google Scholar] [CrossRef]
  70. Docherty-Skogh, A.-C.; Bergman, K.; Waern, M.J.; Ekman, S.; Hultenby, K.; Ossipov, D.; Hilborn, J.; Bowden, T.; Engstrand, T. Bone Morphogenetic Protein-2 Delivered by Hyaluronan-Based Hydrogel Induces Massive Bone Formation and Healing of Cranial Defects in Minipigs. Plast. Reconstr. Surg. 2010, 125, 1383–1392. [Google Scholar] [CrossRef]
  71. Mabilleau, G.; Aguado, E.; Stancu, I.C.; Cincu, C.; Baslé, M.F.; Chappard, D. Effects of FGF-2 release from a hydrogel polymer on bone mass and microarchitecture. Biomaterials 2008, 29, 1593–1600. [Google Scholar] [CrossRef]
  72. Levingstone, T.J.; Herbaj, S.; Dunne, N.J. Calcium Phosphate Nanoparticles for Therapeutic Applications in Bone Regeneration. Nanomaterials 2019, 9, 1570. [Google Scholar] [CrossRef] [PubMed]
  73. den Boer, F.C.; Wippermann, B.W.; Blokhuis, T.J.; Patka, P.; Bakker, F.C.; Haarman, H.J.T.M. Healing of segmental bone defects with granular porous hydroxyapatite augmented with recombinant human osteogenic protein-I or autologous bone marrow. J. Orthop. Res. 2003, 21, 521–528. [Google Scholar] [CrossRef] [PubMed]
  74. Seeherman, H.J.; Bouxsein, M.; Kim, H.; Li, R.; Li, X.J.; Aiolova, M.; Wozney, J.M. Recombinant human bone morphogenetic protein-2 delivered in an injectable calcium phosphate paste accelerates osteotomy-site healing in a nonhuman primate model. J. Bone Jt. Surg. Am. 2004, 86, 1961–1972. [Google Scholar] [CrossRef]
  75. Moore, W.R.; Graves, S.E.; Bain, G.I. Synthetic bone graft substitutes. ANZ J. Surg. 2001, 71, 354–361. [Google Scholar] [CrossRef] [PubMed]
  76. Mohd Roslan, M.R.; Mohd Kamal, N.L.; Abdul Khalid, M.F.; Mohd Nasir, N.F.; Cheng, E.M.; Beh, C.Y.; Tan, J.S.; Mohamed, M.S. The State of Starch/Hydroxyapatite Composite Scaffold in Bone Tissue Engineering with Consideration for Dielectric Measurement as an Alternative Characterization Technique. Materials 2021, 14, 1960. [Google Scholar] [CrossRef]
  77. Weber, F.E.; Eyrich, G.; Grätz, K.W.; Maly, F.E.; Sailer, H.F. Slow and continuous application of human recombinant bone morphogenetic protein via biodegradable poly(lactide-co-glycolide) foamspheres. Int. J. Oral Maxillofac. Surg. 2002, 31, 60–65. [Google Scholar] [CrossRef]
  78. Chen, S.; Shi, Y.; Zhang, X.; Ma, J. Evaluation of BMP-2 and VEGF loaded 3D printed hydroxyapatite composite scaffolds with enhanced osteogenic capacity in vitro and in vivo. Mater. Sci. Eng. C Mater. Biol. Appl. 2020, 112, 110893. [Google Scholar] [CrossRef]
  79. Tsurushima, H.; Marushima, A.; Suzuki, K.; Oyane, A.; Sogo, Y.; Nakamura, K.; Matsumura, A.; Ito, A. Enhanced bone formation using hydroxyapatite ceramic coated with fibroblast growth factor-2. Acta Biomater. 2010, 6, 2751–2759. [Google Scholar] [CrossRef]
  80. Komaki, H.; Tanaka, T.; Chazono, M.; Kikuchi, T. Repair of segmental bone defects in rabbit tibiae using a complex of beta-tricalcium phosphate, type I collagen, and fibroblast growth factor-2. Biomaterials 2006, 27, 5118–5126. [Google Scholar] [CrossRef]
  81. Mayfield, C.K.; Ayad, M.; Lechtholz-Zey, E.; Chen, Y.; Lieberman, J.R. 3D-Printing for Critical Sized Bone Defects: Current Concepts and Future Directions. Bioengineering 2022, 9, 680. [Google Scholar] [CrossRef]
  82. Kolan, K.C.R.; Huang, Y.-W.; Semon, J.A.; Leu, M.C. 3D-printed Biomimetic Bioactive Glass Scaffolds for Bone Regeneration in Rat Calvarial Defects. Int. J. Bioprint. 2020, 6, 274. [Google Scholar] [CrossRef]
  83. Teotia, A.K.; Dienel, K.; Qayoom, I.; van Bochove, B.; Gupta, S.; Partanen, J.; Seppälä, J.; Kumar, A. Improved Bone Regeneration in Rabbit Bone Defects Using 3D Printed Composite Scaffolds Functionalized with Osteoinductive Factors. ACS Appl. Mater. Interfaces 2020, 12, 48340–48356. [Google Scholar] [CrossRef]
  84. Lutolf, M.P.; Weber, F.E.; Schmoekel, H.G.; Schense, J.C.; Kohler, T.; Müller, R.; Hubbell, J.A. Repair of bone defects using synthetic mimetics of collagenous extracellular matrices. Nat. Biotechnol. 2003, 21, 513–518. [Google Scholar] [CrossRef]
  85. Rizzi, S.C.; Ehrbar, M.; Halstenberg, S.; Raeber, G.P.; Schmoekel, H.G.; Hagenmüller, H.; Müller, R.; Weber, F.E.; Hubbell, J.A. Recombinant protein-co-PEG networks as cell-adhesive and proteolytically degradable hydrogel matrixes. Part II: Biofunctional characteristics. Biomacromolecules 2006, 7, 3019–3029. [Google Scholar] [CrossRef] [PubMed]
  86. Pratt, A.B.; Weber, F.E.; Schmoekel, H.G.; Müller, R.; Hubbell, J.A. Synthetic extracellular matrices for in situ tissue engineering. Biotechnol. Bioeng. 2004, 86, 27–36. [Google Scholar] [CrossRef]
  87. Liu, M.; Sun, Y.; Zhang, Q. Emerging Role of Extracellular Vesicles in Bone Remodeling. J. Dent. Res. 2018, 97, 859–868. [Google Scholar] [CrossRef]
  88. Patois, E.; Osorio-da Cruz, S.; Tille, J.-C.; Walpoth, B.; Gurny, R.; Jordan, O. Novel thermosensitive chitosan hydrogels: In vivo evaluation. J. Biomed. Mater. Res. A 2009, 91, 324–330. [Google Scholar] [CrossRef]
  89. Lutolf, M.P.; Lauer-Fields, J.L.; Schmoekel, H.G.; Metters, A.T.; Weber, F.E.; Fields, G.B.; Hubbell, J.A. Synthetic matrix metalloproteinase-sensitive hydrogels for the conduction of tissue regeneration: Engineering cell-invasion characteristics. Proc. Natl. Acad. Sci. USA 2003, 100, 5413–5418. [Google Scholar] [CrossRef] [PubMed]
  90. Na, K.; Kim, S.W.; Sun, B.K.; Woo, D.G.; Yang, H.N.; Chung, H.M.; Park, K.H. Osteogenic differentiation of rabbit mesenchymal stem cells in thermo-reversible hydrogel constructs containing hydroxyapatite and bone morphogenic protein-2 (BMP-2). Biomaterials 2007, 28, 2631–2637. [Google Scholar] [CrossRef]
  91. Garbern, J.C.; Hoffman, A.S.; Stayton, P.S. Injectable pH- and temperature-responsive poly(N-isopropylacrylamide-co-propylacrylic acid) copolymers for delivery of angiogenic growth factors. Biomacromolecules 2010, 11, 1833–1839. [Google Scholar] [CrossRef]
  92. Kim, H.K.; Shim, W.S.; Kim, S.E.; Lee, K.-H.; Kang, E.; Kim, J.-H.; Kim, K.; Kwon, I.C.; Lee, D.S. Injectable in situ-forming pH/thermo-sensitive hydrogel for bone tissue engineering. Tissue Eng. Part A 2009, 15, 923–933. [Google Scholar] [CrossRef]
  93. Puddu, P.; Puddu, G.M.; Cravero, E.; Muscari, S.; Muscari, A. The involvement of circulating microparticles in inflammation, coagulation and cardiovascular diseases. Can. J. Cardiol. 2010, 26, e140–e145. [Google Scholar] [CrossRef]
  94. Davies, O.G. Extracellular vesicles: From bone development to regenerative orthopedics. Mol. Ther. J. Am. Soc. Gene Ther. 2023, 31, 1251–1274. [Google Scholar] [CrossRef]
  95. Ren, J.; He, W.; Zheng, L.; Duan, H. From structures to functions: Insights into exosomes as promising drug delivery vehicles. Biomater. Sci. 2016, 4, 910–921. [Google Scholar] [CrossRef] [PubMed]
  96. Shahabipour, F.; Barati, N.; Johnston, T.P.; Derosa, G.; Maffioli, P.; Sahebkar, A. Exosomes: Nanoparticulate tools for RNA interference and drug delivery. J. Cell. Physiol. 2017, 232, 1660–1668. [Google Scholar] [CrossRef] [PubMed]
  97. Kempen, D.H.R.; Lu, L.; Heijink, A.; Hefferan, T.E.; Creemers, L.B.; Maran, A.; Yaszemski, M.J.; Dhert, W.J.A. Effect of local sequential VEGF and BMP-2 delivery on ectopic and orthotopic bone regeneration. Biomaterials 2009, 30, 2816–2825. [Google Scholar] [CrossRef]
  98. Sharmin, F.; Adams, D.; Pensak, M.; Dukas, A.; Lieberman, J.; Khan, Y. Biofunctionalizing devitalized bone allografts through polymer-mediated short and long term growth factor delivery: Biofunctionalizing Devitalized Bone Allografts. J. Biomed. Mater. Res. A 2015, 103, 2847–2854. [Google Scholar] [CrossRef] [PubMed]
  99. Sharmin, F.; O’Sullivan, M.; Malinowski, S.; Lieberman, J.R.; Khan, Y. Large scale segmental bone defect healing through the combined delivery of VEGF and BMP-2 from biofunctionalized cortical allografts. J. Biomed. Mater. Res. B Appl. Biomater. 2019, 107, 1002–1010. [Google Scholar] [CrossRef] [PubMed]
  100. 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]
  101. González-González, A.; García-Sánchez, D.; Dotta, M.; Rodríguez-Rey, J.; Pérez-Campo, F. Mesenchymal stem cells secretome: The cornerstone of cell-free regenerative medicine. World J. Stem Cells 2020, 12, 1529–1552. [Google Scholar] [CrossRef]
  102. Liang, X.; Ding, Y.; Zhang, Y.; Tse, H.-F.; Lian, Q. Paracrine Mechanisms of Mesenchymal Stem Cell-Based Therapy: Current Status and Perspectives. Cell Transplant. 2014, 23, 1045–1059. [Google Scholar] [CrossRef] [PubMed]
  103. Hade, M.D.; Suire, C.N.; Suo, Z. Mesenchymal Stem Cell-Derived Exosomes: Applications in Regenerative Medicine. Cells 2021, 10, 1959. [Google Scholar] [CrossRef] [PubMed]
  104. Malekpour, K.; Hazrati, A.; Zahar, M.; Markov, A.; Zekiy, A.O.; Navashenaq, J.G.; Roshangar, L.; Ahmadi, M. The Potential Use of Mesenchymal Stem Cells and Their Derived Exosomes for Orthopedic Diseases Treatment. Stem Cell Rev. Rep. 2022, 18, 933–951. [Google Scholar] [CrossRef]
  105. Qin, Y.; Sun, R.; Wu, C.; Wang, L.; Zhang, C. Exosome: A Novel Approach to Stimulate Bone Regeneration through Regulation of Osteogenesis and Angiogenesis. Int. J. Mol. Sci. 2016, 17, 712. [Google Scholar] [CrossRef] [PubMed]
  106. van der Koog, L.; Gandek, T.B.; Nagelkerke, A. Liposomes and Extracellular Vesicles as Drug Delivery Systems: A Comparison of Composition, Pharmacokinetics, and Functionalization. Adv. Healthc. Mater. 2022, 11, 2100639. [Google Scholar] [CrossRef] [PubMed]
  107. Niedermair, T.; Lukas, C.; Li, S.; Stöckl, S.; Craiovan, B.; Brochhausen, C.; Federlin, M.; Herrmann, M.; Grässel, S. Influence of Extracellular Vesicles Isolated From Osteoblasts of Patients with Cox-Arthrosis and/or Osteoporosis on Metabolism and Osteogenic Differentiation of BMSCs. Front. Bioeng. Biotechnol. 2020, 8, 615520. [Google Scholar] [CrossRef]
  108. Ren, J.; Yu, R.; Xue, J.; Tang, Y.; Su, S.; Liao, C.; Guo, Q.; Guo, W.; Zheng, J. How Do Extracellular Vesicles Play a Key Role in the Maintenance of Bone Homeostasis and Regeneration? A Comprehensive Review of Literature. Int. J. Nanomed. 2022, 17, 5375–5389. [Google Scholar] [CrossRef]
  109. Tsiapalis, D.; O’Driscoll, L. Mesenchymal Stem Cell Derived Extracellular Vesicles for Tissue Engineering and Regenerative Medicine Applications. Cells 2020, 9, 991. [Google Scholar] [CrossRef]
  110. Osugi, M.; Katagiri, W.; Yoshimi, R.; Inukai, T.; Hibi, H.; Ueda, M. Conditioned media from mesenchymal stem cells enhanced bone regeneration in rat calvarial bone defects. Tissue Eng. Part A 2012, 18, 1479–1489. [Google Scholar] [CrossRef]
  111. Takeuchi, R.; Katagiri, W.; Endo, S.; Kobayashi, T. Exosomes from conditioned media of bone marrow-derived mesenchymal stem cells promote bone regeneration by enhancing angiogenesis. PLoS ONE 2019, 14, e0225472. [Google Scholar] [CrossRef]
  112. Furuta, T.; Miyaki, S.; Ishitobi, H.; Ogura, T.; Kato, Y.; Kamei, N.; Miyado, K.; Higashi, Y.; Ochi, M. Mesenchymal Stem Cell-Derived Exosomes Promote Fracture Healing in a Mouse Model. Stem Cells Transl. Med. 2016, 5, 1620–1630. [Google Scholar] [CrossRef]
  113. Zhang, Y.; Xie, Y.; Hao, Z.; Zhou, P.; Wang, P.; Fang, S.; Li, L.; Xu, S.; Xia, Y. Umbilical Mesenchymal Stem Cell-Derived Exosome-Encapsulated Hydrogels Accelerate Bone Repair by Enhancing Angiogenesis. ACS Appl. Mater. Interfaces 2021, 13, 18472–18487. [Google Scholar] [CrossRef] [PubMed]
  114. Wang, X.; Thomsen, P. Mesenchymal stem cell–derived small extracellular vesicles and bone regeneration. Basic Clin. Pharmacol. Toxicol. 2021, 128, 18–36. [Google Scholar] [CrossRef] [PubMed]
  115. Zhou, M.; Xi, J.; Cheng, Y.; Sun, D.; Shu, P.; Chi, S.; Tian, S.; Ye, S. Reprogrammed mesenchymal stem cells derived from iPSCs promote bone repair in steroid-associated osteonecrosis of the femoral head. Stem Cell Res. Ther. 2021, 12, 175. [Google Scholar] [CrossRef] [PubMed]
  116. ClinicalTrials.gov. Treatment of Patients with Bone Tissue Defects Using Mesenchymal Stem Cells Enriched by Extracellular Vesicles. Available online: https://classic.clinicaltrials.gov/ct2/show/NCT05520125 (accessed on 10 August 2023).
  117. Zhang, L.; Jiao, G.; Ren, S.; Zhang, X.; Li, C.; Wu, W.; Wang, H.; Liu, H.; Zhou, H.; Chen, Y. Exosomes from bone marrow mesenchymal stem cells enhance fracture healing through the promotion of osteogenesis and angiogenesis in a rat model of nonunion. Stem Cell Res. Ther. 2020, 11, 38. [Google Scholar] [CrossRef]
  118. Zhang, Y.; Cao, X.; Li, P.; Fan, Y.; Zhang, L.; Ma, X.; Sun, R.; Liu, Y.; Li, W. microRNA-935-modified bone marrow mesenchymal stem cells-derived exosomes enhance osteoblast proliferation and differentiation in osteoporotic rats. Life Sci. 2021, 272, 119204. [Google Scholar] [CrossRef]
  119. Li, H.; Liu, D.; Li, C.; Zhou, S.; Tian, D.; Xiao, D.; Zhang, H.; Gao, F.; Huang, J. Exosomes secreted from mutant-HIF-1α-modified bone-marrow-derived mesenchymal stem cells attenuate early steroid-induced avascular necrosis of femoral head in rabbit. Cell Biol. Int. 2017, 41, 1379–1390. [Google Scholar] [CrossRef]
  120. Zhang, Y.; Hao, Z.; Wang, P.; Xia, Y.; Wu, J.; Xia, D.; Fang, S.; Xu, S. Exosomes from human umbilical cord mesenchymal stem cells enhance fracture healing through HIF-1α-mediated promotion of angiogenesis in a rat model of stabilized fracture. Cell Prolif. 2019, 52, e12570. [Google Scholar] [CrossRef]
  121. Qi, X.; Zhang, J.; Yuan, H.; Xu, Z.; Li, Q.; Niu, X.; Hu, B.; Wang, Y.; Li, X. Exosomes Secreted by Human-Induced Pluripotent Stem Cell-Derived Mesenchymal Stem Cells Repair Critical-Sized Bone Defects through Enhanced Angiogenesis and Osteogenesis in Osteoporotic Rats. Int. J. Biol. Sci. 2016, 12, 836–849. [Google Scholar] [CrossRef]
  122. Cui, Y.; Luan, J.; Li, H.; Zhou, X.; Han, J. Exosomes derived from mineralizing osteoblasts promote ST2 cell osteogenic differentiation by alteration of microRNA expression. FEBS Lett. 2016, 590, 185–192. [Google Scholar] [CrossRef]
  123. Uenaka, M.; Yamashita, E.; Kikuta, J.; Morimoto, A.; Ao, T.; Mizuno, H.; Furuya, M.; Hasegawa, T.; Tsukazaki, H.; Sudo, T.; et al. Osteoblast-derived vesicles induce a switch from bone-formation to bone-resorption in vivo. Nat. Commun. 2022, 13, 1066. [Google Scholar] [CrossRef]
  124. Eichholz, K.F.; Woods, I.; Riffault, M.; Johnson, G.P.; Corrigan, M.; Lowry, M.C.; Shen, N.; Labour, M.-N.; Wynne, K.; O’Driscoll, L.; et al. Human bone marrow stem/stromal cell osteogenesis is regulated via mechanically activated osteocyte-derived extracellular vesicles. Stem Cells Transl. Med. 2020, 9, 1431–1447. [Google Scholar] [CrossRef]
  125. Lv, P.-Y.; Gao, P.-F.; Tian, G.-J.; Yang, Y.-Y.; Mo, F.-F.; Wang, Z.-H.; Sun, L.; Kuang, M.-J.; Wang, Y.-L. Osteocyte-derived exosomes induced by mechanical strain promote human periodontal ligament stem cell proliferation and osteogenic differentiation via the miR-181b-5p/PTEN/AKT signaling pathway. Stem Cell Res. Ther. 2020, 11, 295. [Google Scholar] [CrossRef]
  126. Wang, C.-L.; Xiao, F.; Wang, C.-D.; Zhu, J.-F.; Shen, C.; Zuo, B.; Wang, H.; Li, D.; Wang, X.-Y.; Feng, W.-J.; et al. Gremlin2 Suppression Increases the BMP-2-Induced Osteogenesis of Human Bone Marrow-Derived Mesenchymal Stem Cells Via the BMP-2/Smad/Runx2 Signaling Pathway. J. Cell. Biochem. 2017, 118, 286–297. [Google Scholar] [CrossRef] [PubMed]
  127. Bunnell, B.A. Adipose Tissue-Derived Mesenchymal Stem Cells. Cells 2021, 10, 3433. [Google Scholar] [CrossRef] [PubMed]
  128. Macías, I.; Alcorta-Sevillano, N.; Infante, A.; Rodríguez, C.I. Cutting Edge Endogenous Promoting and Exogenous Driven Strategies for Bone Regeneration. Int. J. Mol. Sci. 2021, 22, 7724. [Google Scholar] [CrossRef]
  129. Gholami, L.; Nooshabadi, V.T.; Shahabi, S.; Jazayeri, M.; Tarzemany, R.; Afsartala, Z.; Khorsandi, K. Extracellular vesicles in bone and periodontal regeneration: Current and potential therapeutic applications. Cell Biosci. 2021, 11, 16. [Google Scholar] [CrossRef]
  130. Han, Z.; Liu, S.; Pei, Y.; Ding, Z.; Li, Y.; Wang, X.; Zhan, D.; Xia, S.; Driedonks, T.; Witwer, K.W.; et al. Highly efficient magnetic labelling allows MRI tracking of the homing of stem cell-derived extracellular vesicles following systemic delivery. J. Extracell. Vesicles 2021, 10, e12054. [Google Scholar] [CrossRef] [PubMed]
  131. Tan, S.S.H.; Tjio, C.K.E.; Wong, J.R.Y.; Wong, K.L.; Chew, J.R.J.; Hui, J.H.P.; Toh, W.S. Mesenchymal Stem Cell Exosomes for Cartilage Regeneration: A Systematic Review of Preclinical In Vivo Studies. Tissue Eng. Part B Rev. 2021, 27, 1–13. [Google Scholar] [CrossRef]
  132. Ma, S.; Zhang, Y.; Li, S.; Li, A.; Li, Y.; Pei, D. Engineering exosomes for bone defect repair. Front. Bioeng. Biotechnol. 2022, 10, 1091360. [Google Scholar] [CrossRef]
  133. Bei, H.P.; Hung, P.M.; Yeung, H.L.; Wang, S.; Zhao, X. Bone-a-Petite: Engineering Exosomes towards Bone, Osteochondral, and Cartilage Repair. Small 2021, 17, 2101741. [Google Scholar] [CrossRef]
  134. Xing, Y.; Yerneni, S.S.; Wang, W.; Taylor, R.E.; Campbell, P.G.; Ren, X. Engineering pro-angiogenic biomaterials via chemoselective extracellular vesicle immobilization. Biomaterials 2022, 281, 121357. [Google Scholar] [CrossRef]
  135. Gupta, D.; Zickler, A.M.; El Andaloussi, S. Dosing extracellular vesicles. Adv. Drug Deliv. Rev. 2021, 178, 113961. [Google Scholar] [CrossRef]
  136. Hertel, F.C.; da Silva, A.S.; de Paula Sabino, A.; Valente, F.L.; Reis, E.C.C. Preconditioning Methods to Improve Mesenchymal Stromal Cell-Derived Extracellular Vesicles in Bone Regeneration—A Systematic Review. Biology 2022, 11, 733. [Google Scholar] [CrossRef]
  137. Hnatiuk, A.P.; Ong, S.-G.; Olea, F.D.; Locatelli, P.; Riegler, J.; Lee, W.H.; Jen, C.H.; De Lorenzi, A.; Giménez, C.S.; Laguens, R.; et al. Allogeneic Mesenchymal Stromal Cells Overexpressing Mutant Human Hypoxia-Inducible Factor 1-α (HIF1-α) in an Ovine Model of Acute Myocardial Infarction. J. Am. Heart Assoc. 2016, 5, e003714. [Google Scholar] [CrossRef] [PubMed]
  138. Valenta, T.; Hausmann, G.; Basler, K. The many faces and functions of β-catenin. EMBO J. 2012, 31, 2714–2736. [Google Scholar] [CrossRef]
  139. Man, K.; Eisenstein, N.M.; Hoey, D.A.; Cox, S.C. Bioengineering extracellular vesicles: Smart nanomaterials for bone regeneration. J. Nanobiotechnol. 2023, 21, 137. [Google Scholar] [CrossRef]
  140. Zha, Y.; Li, Y.; Lin, T.; Chen, J.; Zhang, S.; Wang, J. Progenitor cell-derived exosomes endowed with VEGF plasmids enhance osteogenic induction and vascular remodeling in large segmental bone defects. Theranostics 2021, 11, 397–409. [Google Scholar] [CrossRef] [PubMed]
  141. Xu, J.; Wang, Y.; Hsu, C.-Y.; Gao, Y.; Meyers, C.A.; Chang, L.; Zhang, L.; Broderick, K.; Ding, C.; Peault, B.; et al. Human perivascular stem cell-derived extracellular vesicles mediate bone repair. eLife 2019, 8, e48191. [Google Scholar] [CrossRef] [PubMed]
  142. Xie, H.; Wang, Z.; Zhang, L.; Lei, Q.; Zhao, A.; Wang, H.; Li, Q.; Cao, Y.; Jie Zhang, W.; Chen, Z. Extracellular Vesicle-functionalized Decalcified Bone Matrix Scaffolds with Enhanced Pro-angiogenic and Pro-bone Regeneration Activities. Sci. Rep. 2017, 7, 45622. [Google Scholar] [CrossRef]
  143. Luo, Z.-W.; Li, F.-X.-Z.; Liu, Y.-W.; Rao, S.-S.; Yin, H.; Huang, J.; Chen, C.-Y.; Hu, Y.; Zhang, Y.; Tan, Y.-J.; et al. Aptamer-functionalized exosomes from bone marrow stromal cells target bone to promote bone regeneration. Nanoscale 2019, 11, 20884–20892. [Google Scholar] [CrossRef]
  144. Yang, S.; Zhu, B.; Yin, P.; Zhao, L.; Wang, Y.; Fu, Z.; Dang, R.; Xu, J.; Zhang, J.; Wen, N. Integration of Human Umbilical Cord Mesenchymal Stem Cells-Derived Exosomes with Hydroxyapatite-Embedded Hyaluronic Acid-Alginate Hydrogel for Bone Regeneration. ACS Biomater. Sci. Eng. 2020, 6, 1590–1602. [Google Scholar] [CrossRef]
  145. 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]
  146. Yan, H.-C.; Yu, T.-T.; Li, J.; Qiao, Y.-Q.; Wang, L.-C.; Zhang, T.; Li, Q.; Zhou, Y.-H.; Liu, D.-W. The Delivery of Extracellular Vesicles Loaded in Biomaterial Scaffolds for Bone Regeneration. Front. Bioeng. Biotechnol. 2020, 8, 1015. [Google Scholar] [CrossRef] [PubMed]
  147. Kang, M.; Lee, C.; Lee, M. Bioactive Scaffolds Integrated with Liposomal or Extracellular Vesicles for Bone Regeneration. Bioengineering 2021, 8, 137. [Google Scholar] [CrossRef]
  148. Bahney, C.S.; Zondervan, R.L.; Allison, P.; Theologis, A.; Ashley, J.W.; Ahn, J.; Miclau, T.; Marcucio, R.S.; Hankenson, K.D. Cellular biology of fracture healing; Cellular biology of fracture healing. J. Orthop. Res. 2019, 37, 35–50. [Google Scholar] [CrossRef]
  149. Collon, K.; Gallo, M.C.; Lieberman, J.R. Musculoskeletal tissue engineering: Regional gene therapy for bone repair. Biomaterials 2021, 275, 120901. [Google Scholar] [CrossRef] [PubMed]
  150. Bougioukli, S.; Alluri, R.; Pannell, W.; Sugiyama, O.; Vega, A.; Tang, A.; Skorka, T.; Park, S.H.; Oakes, D.; Lieberman, J.R. Ex vivo gene therapy using human bone marrow cells overexpressing BMP-2: “Next-day” gene therapy versus standard “two-step” approach. Bone 2019, 128, 115032. [Google Scholar] [CrossRef] [PubMed]
  151. Vakhshori, V.; Bougioukli, S.; Sugiyama, O.; Kang, H.P.; Tang, A.H.; Park, S.-H.; Lieberman, J.R. Ex vivo regional gene therapy with human adipose-derived stem cells for bone repair. Bone 2020, 138, 115524. [Google Scholar] [CrossRef]
  152. Virk, M.S.; Sugiyama, O.; Park, S.H.; Gambhir, S.S.; Adams, D.J.; Drissi, H.; Lieberman, J.R. Same Day Ex-vivo regional gene therapy: A novel strategy to enhance bone repair. Mol. Ther. 2011, 19, 960–968. [Google Scholar] [CrossRef] [PubMed]
  153. Cavazzana-Calvo, M.; Payen, E.; Negre, O.; Wang, G.; Hehir, K.; Fusil, F.; Down, J.; Denaro, M.; Brady, T.; Westerman, K.; et al. Transfusion independence and HMGA2 activation after gene therapy of human β-thalassaemia. Nature 2010, 467, 318–322. [Google Scholar] [CrossRef]
  154. Aiuti, A.; Biasco, L.; Scaramuzza, S.; Ferrua, F.; Cicalese, M.P.; Baricordi, C.; Dionisio, F.; Calabria, A.; Giannelli, S.; Castiello, M.C.; et al. Lentiviral hematopoietic stem cell gene therapy in patients with wiskott-aldrich syndrome. Science 2013, 341, 1233151. [Google Scholar] [CrossRef]
  155. Sessa, M.; Lorioli, L.; Fumagalli, F.; Acquati, S.; Redaelli, D.; Baldoli, C.; Canale, S.; Lopez, I.D.; Morena, F.; Calabria, A.; et al. Lentiviral haemopoietic stem-cell gene therapy in early-onset metachromatic leukodystrophy: An ad-hoc analysis of a non-randomised, open-label, phase 1/2 trial. Lancet 2016, 388, 476–487. [Google Scholar] [CrossRef]
  156. Keeler, A.M.; Flotte, T.R. Recombinant Adeno-Associated Virus Gene Therapy in Light of Luxturna (and Zolgensma and Glybera): Where Are We, and How Did We Get Here? Annu. Rev. Virol. 2019, 6, 601–621. [Google Scholar] [CrossRef]
  157. Vorburger, S.A.; Hunt, K.K. Adenoviral Gene Therapy. Oncologist 2002, 7, 46–59. [Google Scholar] [CrossRef]
  158. Mcconnell, M.J.; Imperiale, M.J. Biology of Adenovirus and Its Use as a Vector for Gene Therapy. Hum. Gene Ther. 2004, 15, 1022–1033. [Google Scholar] [CrossRef]
  159. Turgeman, G.; Pittman, D.D.; Müller, R.; Gowda Kurkalli, B.; Zhou, S.; Pelled, G.; Peyser, A.; Zilberman, Y.; Moutsatsos, I.K.; Gazit, D. Engineered human mesenchymal stem cells: A novel platform for skeletal cell mediated gene therapy. J. Gene Med. 2001, 3, 240–251. [Google Scholar] [CrossRef]
  160. Lieberman, J.R.; Le, L.Q.; Wu, L.; Finerman, G.A.M.; Berk, A.; Witte, O.N.; Stevenson, S. Regional gene therapy with a BMP-2-producing murine stromal cell line induces heterotopic and orthotopic bone formation in rodents. J. Orthop. Res. 1998, 16, 330–339. [Google Scholar] [CrossRef]
  161. Feeley, B.T.; Conduah, A.H.; Sugiyama, O.; Krenek, L.; Chen, I.S.Y.; Lieberman, J.R. In vivo molecular imaging of adenoviral versus lentiviral gene therapy in two bone formation models. J. Orthop. Res. 2006, 24, 1709–1721. [Google Scholar] [CrossRef]
  162. Virk, M.S.; Conduah, A.; Park, S.H.; Liu, N.; Sugiyama, O.; Cuomo, A.; Kang, C.; Lieberman, J.R. Influence of short-term adenoviral vector and prolonged lentiviral vector mediated bone morphogenetic protein-2 expression on the quality of bone repair in a rat femoral defect model. Bone 2008, 42, 921–931. [Google Scholar] [CrossRef]
  163. Musgrave, D.S.; Bosch, P.; Ghivizzani, S.; Robbins, P.D.; Evans, C.H.; Huard, J. Adenovirus-mediated direct gene therapy with bone morphogenetic protein-2 produces bone. Bone 1999, 24, 541–547. [Google Scholar] [CrossRef]
  164. Kaihara, S.; Bessho, K.; Okubo, Y.; Sonobe, J.; Kawai, M.; Iizuka, T. Simple and effective osteoinductive gene therapy by local injection of a bone morphogenetic protein-2-expressing recombinant adenoviral vector and FK506 mixture in rats. Gene Ther. 2004, 11, 439–447. [Google Scholar] [CrossRef]
  165. Alden, T.D.; Pittman, D.D.; Hankins, G.R.; Beres, E.J.; Engh, J.A.; Das, S.; Hudson, S.B.; Kerns, K.M.; Kallmes, D.F.; Helm, G.A. In Vivo Endochondral Bone Formation Using a Bone Morphogenetic Protein 2 Adenoviral Vector. Hum. Gene Ther. 2004, 10, 2245–2253. [Google Scholar] [CrossRef]
  166. Li, C.; Samulski, R.J. Engineering adeno-associated virus vectors for gene therapy. Nat. Rev. Genet. 2020, 21, 255–272. [Google Scholar] [CrossRef]
  167. Wang, D.; Tai, P.W.L.; Gao, G. Adeno-associated virus vector as a platform for gene therapy delivery. Nat. Rev. Drug Discov. 2019, 18, 358. [Google Scholar] [CrossRef]
  168. Bougioukli, S.; Chateau, M.; Morales, H.; Vakhshori, V.; Sugiyama, O.; Oakes, D.; Longjohn, D.; Cannon, P. Limited potential of AAV-mediated gene therapy in transducing human mesenchymal stem cells for bone repair applications. Gene Ther. 2021, 28, 729–739. [Google Scholar] [CrossRef]
  169. Buchschacher, G.L.; Wong-Staal, F. Development of lentiviral vectors for gene therapy for human diseases. Blood 2000, 95, 2499–2504. [Google Scholar] [CrossRef]
  170. Milone, M.C.; O’Doherty, U. Clinical use of lentiviral vectors. Leukemia 2018, 32, 1529–1541. [Google Scholar] [CrossRef]
  171. Rothe, M.; Modlich, U.; Schambach, A. Biosafety challenges for use of lentiviral vectors in gene therapy. Curr. Gene Ther. 2013, 13, 453–468. [Google Scholar] [CrossRef]
  172. Miyazaki, M.; Sugiyama, O.; Zou, J.; Yoon, S.H.; Wei, F.; Morishita, Y.; Sintuu, C.; Virk, M.S.; Lieberman, J.R.; Wang, J.C. Comparison of lentiviral and adenoviral gene therapy for spinal fusion in rats. Spine 2008, 33, 1410–1417. [Google Scholar] [CrossRef]
  173. Miyazaki, M.; Sugiyama, O.; Tow, B.; Zou, J.; Morishita, Y.; Wei, F.; Napoli, A.; Sintuu, C.; Lieberman, J.R.; Wang, J.C. The effects of lentiviral gene therapy with bone morphogenetic protein-2-producing bone marrow cells on spinal fusion in rats. J. Spinal Disord. Tech. 2008, 21, 372–379. [Google Scholar] [CrossRef] [PubMed]
  174. Alden, T.D.; Pittman, D.D.; Beres, E.J.; Hankins, G.R.; Kallmes, D.F.; Wisotsky, B.M.; Kerns, K.M.; Helm, G.A. Percutaneous spinal fusion using bone morphogenetic protein-2 gene therapy. J. Neurosurg. Spine 1999, 90, 109–114. [Google Scholar] [CrossRef]
  175. Hidaka, C.; Goshi, K.; Rawlins, B.; Boachie-Adjei, O.; Crystal, R.G. Enhancement of spine fusion using combined gene therapy and tissue engineering BMP-7-expressing bone marrow cells and allograft bone. Spine 2003, 28, 2049–2057. [Google Scholar] [CrossRef]
  176. Dumont, R.J.; Dayoub, H.; Li, J.Z.; Dumont, A.S.; Kallmes, D.F.; Hankins, G.R.; Helm, G.A. Ex Vivo Bone Morphogenetic Protein-9 Gene Therapy Using Human Mesenchymal Stem Cells Induces Spinal Fusion in Rodents. Neurosurgery 2002, 51, 1239–1244. [Google Scholar] [CrossRef]
  177. Laurent, J.J.; Webb, K.M.; Beres, E.J.; McGee, K.; Li, J.; Van Rietbergen, B.; Helm, G.A. The use of bone morphogenetic protein—6 gene therapy for percutaneous spinal fusion in rabbits. J. Neurosurg. Spine 2004, 1, 90–94. [Google Scholar] [CrossRef]
  178. Helm, G.A.; Alden, T.D.; Beres, E.J.; Hudson, S.B.; Das, S.; Engh, J.A.; Pittman, D.D.; Kerns, K.M.; Kallmes, D.F. Use of bone morphogenetic protein-9 gene therapy to induce spinal arthrodesis in the rodent. J. Neurosurg. 2000, 92, 191–196. [Google Scholar] [CrossRef]
  179. Wang, J.C.; Kanim, L.E.; Yoo, S.; Campbell, P.A.; Berk, A.J.; Lieberman, J.R. Effect of regional gene therapy with bone morphogenetic protein-2-producing bone marrow cells on spinal fusion in rats. JBJS 2003, 85, 905–911. [Google Scholar] [CrossRef] [PubMed]
  180. Lee, J.Y.; Peng, H.; Usas, A.; Musgrave, D.; Cummins, J.; Pelinkovic, D.; Jankowski, R.; Huard, J.; Lee, J.Y.; Peng, H.; et al. Enhancement of Bone Healing Based on Ex Vivo Gene Therapy Using Human Muscle-Derived Cells Expressing Bone Morphogenetic Protein 2. Hum. Gene Ther. 2004, 13, 1201–1211. [Google Scholar] [CrossRef] [PubMed]
  181. Alluri, R.; Jakus, A.; Bougioukli, S.; Pannell, W.; Sugiyama, O.; Tang, A.; Shah, R.; Lieberman, J.R. 3D printed hyperelastic “bone” scaffolds and regional gene therapy: A novel approach to bone healing. J. Biomed. Mater. Res. A 2018, 106, 1104–1110. [Google Scholar] [CrossRef]
  182. Miyazaki, M.; Zuk, P.A.; Zou, J.; Yoon, S.H.; Wei, F.; Morishita, Y.; Sintuu, C.; Wang, J.C. Comparison of human mesenchymal stem cells derived from adipose tissue and bone marrow for ex vivo gene therapy in rat spinal fusion model. Spine 2008, 33, 863–869. [Google Scholar] [CrossRef]
  183. Kang, H.P.; Ihn, H.; Robertson, D.M.; Chen, X.; Sugiyama, O.; Tang, A.; Hollis, R.; Skorka, T.; Longjohn, D.; Oakes, D.; et al. Regional gene therapy for bone healing using a 3D printed scaffold in a rat femoral defect model. J. Biomed. Mater. Res. A 2021, 109, 2346–2356. [Google Scholar] [CrossRef]
  184. Rundle, C.H.; Miyakoshi, N.; Kasukawa, Y.; Chen, S.T.; Sheng, M.H.C.; Wergedal, J.E.; Lau, K.H.W.; Baylink, D.J. In vivo bone formation in fracture repair induced by direct retroviral-based gene therapy with bone morphogenetic protein-4. Bone 2003, 32, 591–601. [Google Scholar] [CrossRef]
  185. Baltzer, A.W.A.; Lattermann, C.; Whalen, J.D.; Wooley, P.; Weiss, K.; Grimm, M.; Ghivizzani, S.C.; Robbins, P.D.; Evans, C.H. Genetic enhancement of fracture repair: Healing of an experimental segmental defect by adenoviral transfer of the BMP-2 gene. Gene Ther. 2000, 7, 734–739. [Google Scholar] [CrossRef] [PubMed]
  186. Peng, H.; Wright, V.; Usas, A.; Gearhart, B.; Shen, H.C.; Cummins, J.; Huard, J. Synergistic enhancement of bone formation and healing by stem cell-expressed VEGF and bone morphogenetic protein-4. J. Clin. Investig. 2002, 110, 751–759. [Google Scholar] [CrossRef] [PubMed]
  187. Ito, H.; Koefoed, M.; Tiyapatanaputi, P.; Gromov, K.; Goater, J.J.; Carmouche, J.; Zhang, X.; Rubery, P.T.; Rabinowitz, J.; Samulski, R.J.; et al. Remodeling of cortical bone allografts mediated by adherent rAAV-RANKL and VEGF gene therapy. Nat. Med. 2005, 11, 291–297. [Google Scholar] [CrossRef]
  188. Chang, P.C.; Seol, Y.J.; Cirelli, J.A.; Pellegrini, G.; Jin, Q.; Franco, L.M.; Goldstein, S.A.; Chandler, L.A.; Sosnowski, B.; Giannobile, W.V. PDGF-B Gene Therapy Accelerates Bone Engineering and Oral Implant Osseointegration. Gene Ther. 2010, 17, 95. [Google Scholar] [CrossRef] [PubMed]
  189. Al-Dosari, M.S.; Gao, X. Nonviral Gene Delivery: Principle, Limitations, and Recent Progress. AAPS J. 2009, 11, 671–681. [Google Scholar] [CrossRef]
  190. Bez, M.; Sheyn, D.; Tawackoli, W.; Avalos, P.; Shapiro, G.; Giaconi, J.C.; Da, X.; David, S.B.; Gavrity, J.; Awad, H.A.; et al. In situ bone tissue engineering via ultrasound-mediated gene delivery to endogenous progenitor cells in mini-pigs. Sci. Transl. Med. 2017, 9, eaal3128. [Google Scholar] [CrossRef]
  191. Bez, M.; Foiret, J.; Shapiro, G.; Pelled, G.; Ferrara, K.W.; Gazit, D. Nonviral ultrasound-mediated gene delivery in small and large animal models. Nat. Protoc. 2019, 14, 1015–1026. [Google Scholar] [CrossRef] [PubMed]
  192. Feichtinger, G.A.; Hofmann, A.T.; Slezak, P.; Schuetzenberger, S.; Kaipel, M.; Schwartz, E.; Neef, A.; Nomikou, N.; Nau, T.; Van Griensven, M.; et al. Sonoporation increases therapeutic efficacy of inducible and constitutive BMP2/7 in vivo gene delivery. Hum. Gene Ther. Methods 2014, 25, 57–71. [Google Scholar] [CrossRef]
  193. Curtin, C.M.; Tierney, E.G.; McSorley, K.; Cryan, S.-A.; Duffy, G.P.; O’Brien, F.J.; Curtin, C.M.; Tierney, E.G.; McSorley, K.; Duffy, G.P. Combinatorial Gene Therapy Accelerates Bone Regeneration: Non-Viral Dual Delivery of VEGF and BMP2 in a Collagen-Nanohydroxyapatite Scaffold. Adv Healthc Mater. 2014, 4, 223–227. [Google Scholar] [CrossRef]
  194. Alluri, R.; Song, X.; Bougioukli, S.; Pannell, W.; Vakhshori, V.; Sugiyama, O.; Tang, A.; Park, S.H.; Chen, Y.; Lieberman, J.R. Regional gene therapy with 3D printed scaffolds to heal critical sized bone defects in a rat model. J. Biomed. Mater. Res. A 2019, 107, 2174–2182. [Google Scholar] [CrossRef]
  195. Bozo, I.Y.; Deev, R.V.; Drobyshev, A.Y.; Isaev, A.A.; Eremin, I.I. World’s First Clinical Case of Gene-Activated Bone Substitute Application. Case Rep. Dent. 2016, 2016, 8648949. [Google Scholar] [CrossRef]
  196. Bozo, I.Y.; Drobyshev, A.Y.; Redko, N.A.; Komlev, V.S.; Isaev, A.A.; Deev, R.V. Bringing a Gene-Activated Bone Substitute Into Clinical Practice: From Bench to Bedside. Front. Bioeng. Biotechnol. 2021, 9, 599300. [Google Scholar] [CrossRef]
  197. Salhotra, A.; Shah, H.N.; Levi, B.; Longaker, M.T. Mechanisms of bone development and repair. Nat. Rev. Mol. Cell Biol. 2020, 21, 696–711. [Google Scholar] [CrossRef]
  198. Li, C.; Du, Y.; Zhang, T.; Wang, H.; Hou, Z.; Zhang, Y.; Cui, W.; Chen, W. “Genetic scissors” CRISPR/Cas9 genome editing cutting-edge biocarrier technology for bone and cartilage repair. Bioact. Mater. 2023, 22, 254–273. [Google Scholar] [CrossRef]
  199. Ghadakzadeh, S.; Mekhail, M.; Aoude, A.; Hamdy, R.; Tabrizian, M. Small Players Ruling the Hard Game: siRNA in Bone Regeneration. J. Bone Miner. Res. 2016, 31, 475–487. [Google Scholar] [CrossRef]
  200. Leng, Q.; Chen, L.; Lv, Y. RNA-based scaffolds for bone regeneration: Application and mechanisms of mRNA, miRNA and siRNA. Theranostics 2020, 10, 3190. [Google Scholar] [CrossRef]
  201. Rose, L.; Uludaǧ, H. Realizing the potential of gene-based molecular therapies in bone repair. J. Bone Miner. Res. 2013, 28, 2245–2262. [Google Scholar] [CrossRef]
  202. Liu, F.; Ferreira, E.; Porter, R.M.; Glatt, V.; Schinhan, M.; Shen, Z.; Randolph, M.A.; Kirker-Head, C.A.; Wehling, C.; Vrahas, M.S.; et al. Rapid and reliable healing of critical size bone defects with genetically modified sheep muscle. Eur. Cells Mater. 2015, 30, 118–130; discussion 130–131. [Google Scholar] [CrossRef]
  203. De La Vega, R.E.; De Padilla, C.L.; Trujillo, M.; Quirk, N.; Porter, R.M.; Evans, C.H.; Ferreira, E. Contribution of Implanted, Genetically Modified Muscle Progenitor Cells Expressing BMP-2 to New Bone Formation in a Rat Osseous Defect. Mol. Ther. J. Am. Soc. Gene Ther. 2018, 26, 208–218. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Radiograph of a tibial critical-sized bone defect associated with a fracture nonunion complicated by infection. The patient status is post-placement of an external fixator with an intramedullary antibiotic device pending distraction osteogenesis (Radiographs courtesy of Dr Joseph Patterson).
Figure 1. Radiograph of a tibial critical-sized bone defect associated with a fracture nonunion complicated by infection. The patient status is post-placement of an external fixator with an intramedullary antibiotic device pending distraction osteogenesis (Radiographs courtesy of Dr Joseph Patterson).
Bioengineering 10 01252 g001
Figure 2. Features of an optimized delivery system.
Figure 2. Features of an optimized delivery system.
Bioengineering 10 01252 g002
Figure 3. Schematic illustration of a parent cell undergoing ectocytosis of an extracellular vesicle and the engineering strategies to optimize extracellular vesicles (EVs) for bone repair (The figure was created with BioRender.com).
Figure 3. Schematic illustration of a parent cell undergoing ectocytosis of an extracellular vesicle and the engineering strategies to optimize extracellular vesicles (EVs) for bone repair (The figure was created with BioRender.com).
Bioengineering 10 01252 g003
Figure 4. Radiograph of a 6 mm athymic rat critical-sized femoral defect treated with LV-BMP-2 transduced human adipose-derived stem cells loaded on a TCP-HA scaffold at postoperative day 0 (left) with the healed defect at 12 weeks postoperatively (right).
Figure 4. Radiograph of a 6 mm athymic rat critical-sized femoral defect treated with LV-BMP-2 transduced human adipose-derived stem cells loaded on a TCP-HA scaffold at postoperative day 0 (left) with the healed defect at 12 weeks postoperatively (right).
Bioengineering 10 01252 g004
Figure 5. Schematic illustration of two-step transcriptional amplification (TSTA) lentiviral system overexpressing growth factor genes of interest in adipose stem cells. The TSTA system is composed of two separate lentiviral vectors: the GAL4-VP16 (LV-RhMLV-GAL4-VP16) transactivator vector and the transgene vector expressing the growth factor gene (LV-G5-GF gene) (Created with BioRender.com). LV, lentivirus; RhMLV, murine leukemia virus; LTR, long terminal repeat; Ψ, packaging signal; RRE, rev-responsible element; cppt, central polypurine tract; G5 promoter, GAL4 binding site; SIN, self-inactivating; GF, growth factor; ASC, adipose stem cell; BMP-2, bone morphogenetic protein 2.
Figure 5. Schematic illustration of two-step transcriptional amplification (TSTA) lentiviral system overexpressing growth factor genes of interest in adipose stem cells. The TSTA system is composed of two separate lentiviral vectors: the GAL4-VP16 (LV-RhMLV-GAL4-VP16) transactivator vector and the transgene vector expressing the growth factor gene (LV-G5-GF gene) (Created with BioRender.com). LV, lentivirus; RhMLV, murine leukemia virus; LTR, long terminal repeat; Ψ, packaging signal; RRE, rev-responsible element; cppt, central polypurine tract; G5 promoter, GAL4 binding site; SIN, self-inactivating; GF, growth factor; ASC, adipose stem cell; BMP-2, bone morphogenetic protein 2.
Bioengineering 10 01252 g005
Figure 6. Human Adipose-Derived Stem Cells transduced with LV-BMP-2 loaded on a scaffold prior to implantation in a critical-sized bone defect. MSCs, mesenchymal stem cells; LV-BMP-2, lentivirus vector expressing bone morphogenetic protein. (The figure was created with BioRender.com).
Figure 6. Human Adipose-Derived Stem Cells transduced with LV-BMP-2 loaded on a scaffold prior to implantation in a critical-sized bone defect. MSCs, mesenchymal stem cells; LV-BMP-2, lentivirus vector expressing bone morphogenetic protein. (The figure was created with BioRender.com).
Bioengineering 10 01252 g006
Table 1. Summary of growth factor delivery strategies in bone repair.
Table 1. Summary of growth factor delivery strategies in bone repair.
Delivery MethodAdvantagesChallenges
Scaffolds
-
Local delivery of purified growth factors with precise concentrations
-
Modifiable composition/geometry to improve osteoconductivity
-
Customizable for complex defects
-
Potential for time-dependent release
-
Biocompatibility dependent upon material
-
Sustained release requires significant modifications
-
Complex manufacturing
Extracellular Vesicles
-
Immune privileged
-
Enhanced crossing of biological membranes for intracellular signaling
-
Modifiable for homing efficiency
-
Protection against enzymatic degradation
-
Isolation and collection
-
Nonspecific cargo loading and concentrations
-
Undefined signaling pathways
Regional Gene Therapy
-
Sustained signaling with viral integration
-
Pairs osteoinductive signaling with responding cells and osteoconductive scaffold
-
Potential inflammatory response dependent upon vector
-
Limited concerns for viral reconstitution and oncogenesis
-
Complex manufacturing
Table 2. FDA-approved clinical applications of recombinant BMP-2.
Table 2. FDA-approved clinical applications of recombinant BMP-2.
SystemsIndicationApproval Date
INFUSE® Bone Graft
  • Alternative to autogenous bone graft for sinus augmentations
  • For localized alveolar ridge augmentations in extraction socket defects
March 2007
  • Expanded indication for spinal fusion procedures in skeletally mature patients with degenerative disc disease at 1 level from L4 to S1
  • Expanded for acute, indication open tibial shaft fractures stabilized with nail fixation
October 2009
INFUSE™ Bone Graft/LT-CAGE™ Lumbar Tapered Fusion Device
  • Indicated for spinal fusion procedures in skeletally mature patients with degenerative disc disease at 1 level from L4 to S1
  • Up to grade 1 spondylolisthesis at the involved level
  • Implantation via anterior open or anterior laparoscopic approach
July 2002
  • Extension of device use from L2 to S1
  • May be used with retrolisthesis
July 2004
  • Indicated for acute, open tibial shaft fractures stabilized with nail fixation
  • Alternative to autogenous bone graft for sinus augmentations
  • For localized alveolar ridge augmentations in extraction socket defects
October 2009
INFUSE™ Bone Graft/Medtronic Interbody Fusion Device
  • Indicated for spinal fusion procedures in skeletally mature patients with degenerative disc disease at one level from L2-S1, who may also have up to Grade I spondylolisthesis or Grade I retrolisthesis at the involved level
December 2015
Table 3. Summary of preclinical studies applying EVs in immunoregulation, osteoclastogenesis, osteoblastogenesis, and angiogenesis. + Bone Marrow Mesenchymal Stem Cell-Derived Extracellular Vesicles.
Table 3. Summary of preclinical studies applying EVs in immunoregulation, osteoclastogenesis, osteoblastogenesis, and angiogenesis. + Bone Marrow Mesenchymal Stem Cell-Derived Extracellular Vesicles.
AuthorParent CellEV DoseScaffold CarrierCargo;
Pathway
Growth Factor, Protein, GeneModelOutcome
Takeuchi et al. [111]hBMMSCs 30 μgAtelocollagen sponge miRNA undefined; unspecified ↑ VEGF, ANG1, ANG2, COLI, OCN, Runx2 Wistar rat calvarial defect In vitro: anti-VEGF antibody decreased expression of osteogenic and angiogenic-related genes; EVs promoted hMSC migration
In vivo: anti-VEGF antibody impaired bone formation
Zhang et al. [117]Rat BMMSCs 100 μL (1010 particles)Local injection BMP;
BMP-2/Smad1/Runx2 and HIF1-1α/VEGF pathways
↑ OCN, OPN, OGN, BMP2, Smad1, Runx2 Wistar rat femoral nonunion In vitro: BMMSC-EVs + promoted proliferation and migration of HUVECs and osteoblast precursors
In vivo: BMMSC-EVs + enhanced osteogenesis, angiogenesis, and fracture healing
Zhang et al. [118]hBMMSCs 25 μg/mLLocal injection miR-935; inhibition of STAT1 ↑ Runx2, ATF4 Sprague–Dawley ovariectomized, osteoporotic rats In vitro: increased ALP activity, enhanced levels of Runx2 and ATF4, enhanced osteoblast proliferation and differentiation
In vivo: increased BMD, BV/TV, TbN, Tb.Th
Li et al. [100]hADSCs 25 μg/mLPLGA/pDA miR-218; unspecified ↑ Runx2, ALP, COL1a1 Murine critical-sized calvarial defect In vitro: enhanced expression of osteoblastogenesis-related genes
In vivo: significantly more new bone formation and recruitment of host MSCs
Li et al. [119] hADSCs0.8 mg/mLGNP hydrogelmiR-451a;
unspecified
↑ M2 marker (CD206)
↓ M1 marker (iNOS)
Sprague–Dawley rat calvarial defectIn vitro: miR-451a promotes the polarization of macrophage phenotypes through the inhibition of MIF
In vivo: immunoregulated bone microenvironment, promoted osteogenesis
Zhang et al. [120]hUCMSCs 100 μL/mLHyStem-HP hydrogel HIF-1α, VEGF;
unspecified
↑ HIF-1α, VEGF, OCN, COL1a1 Rat femoral fracture In vitro: Upregulation of osteogenic- and angiogenic-related gene expression levels
In vivo: promoted angiogenesis and fracture healing through the proliferation of HUVECs
Qi et al. [121] hiPSC-MSCs 100 µgβ-TCP unspecified ↑ OPN, COL1, Runx2 Sprague–Dawley ovariectomized rats with calvarial defectIn vitro: increased ALP activity and expression levels of osteoblast-related genes and increased proliferation of rBMSCs
In vivo: enhanced BV/TV and angiogenesis in a dose-dependent manner
Cui et al. [122]MC3T3-E1 100 µg----miR-1192, miR-680, miR-302a;
Wnt pathway
↑ Runx2, ALP, β-catenin
↓ Axin1
Murine bone marrow-derived stromal cell line (ST2)In vitro: increased osteoblast differentiation and matrix mineralization
Uenaka et al. [123]MC3T3-E1,
Mature osteoblasts
1–5 × 109 particlesGelatin-hydrogel sheetmiR-143-3p;
targeting of Cbfb
↑ Rankl
↓ Runx2, Sp7
Murine critical-sized calvarial defectIn vitro: Inhibition of osteoblast differentiation and promotion of osteoclastogenesis through the suppression of osteoblastic gene expression
In vivo: inhibition of bone repair and promotion of bone resorption
Eichholz et al. [124]MLO-Y4 osteocyte-like cells 1 μg----Annexin A5, Histone H4;
inhibition of RANKL-RANK
↑ (CM-F):
Histone H4, COX2, OCN, OPN, Runx2, OSX, ALP
hMSCs In vitro: CM-F treatment groups enhanced osteogenesis, osteoblastogenesis
Lv et al. [125]MLO-Y4 osteocyte-like cells 10 μL----miR181b-5p; PTEN/AKT pathway ↑ ALP, BMP2, Runx2 hPDLSC In vitro: promoted osteogenic proliferation and differentiation in mechanically strained MLO-Y4 cells
↑, increased; ↓, decreased; hBMMSCs, human bone marrow mesenchymal stem cell; miRNA, microRNA; VEGF, vascular endothelial growth factor; ANG1, angiopoietin 1; ANG2, angiopoietin 2; COL1, type 1 collagen; OCN, osteocalcin; Runx2, runt-related transcription factor 2; HUVEC, human umbilical vein endothelial cells; BMP-2, bone morphogenetic protein 2; HIF1-1α, hypoxia-inducible factor 1-alpha; OPN, osteopontin; OGN, osteoglycin; STAT1, signal transducer and activator of transcription 1; ATF4, activating transcription factor 4; BMD, bone mineral density; BV/TV, bone volume/tissue volume; TbN, average number of trabeculae per unit length; Tb.Th, mean thickness of trabeculae; hADSCs, human adipose stem cells; PLGA, poly(lactic-co-glycolic) acid; pDA, polydopamine; ALP, alkaline phosphatase; COL1a1, collagen type 1 alpha 1; GNP, gliadin nanoparticles; M2, macrophage phenotype 2; M1, macrophage phenotype 1; MIF, macrophage migration inhibitory factor; iNOS, inducible nitric oxide synthase; hUCMSCs, human umbilical cord mesenchymal stem cells; hiPSC, human-induced pluripotent stem cells; β-TCP, beta-tricalcium phosphate; Rankl, receptor activator of nuclear factor kappa beta; RANK, receptor activator of nuclear factor kappa beta; COX2, prostaglandin-endoperoxide synthase 2; OSX, osterix; hMSCs, human mesenchymal stem cells; CM-F, media undergone fluid shear; PTEN/AKT, phosphate and tensin homolog/protein kinase B; hPDLSC, human periodeontal ligament stem cell.
Table 4. Ex vivo viral vectors used to deliver BMP to treat critical-sized defects and promote spinal fusion in preclinical animal models.
Table 4. Ex vivo viral vectors used to deliver BMP to treat critical-sized defects and promote spinal fusion in preclinical animal models.
AuthorVectorCellCarrierModelResults
Lieberman et al. [160]Ad-BMP2W-20 (murine stromal)DBMSCID Mouse 8 mm Femoral DefectRadiographic healing at 8 wks. Histologic demonstration of lamellar bone formation
Lieberman et al. [20]Ad-BMP2Rat Bone Marrow CellsDBMLewis Rat 8 mm Femoral DefectRadiographic healing at 8 wks with course trabecular bone with remodeling. Equivalent mechanics between operated and non-operated femurs that healed
Dumont et al. [176]Ad-BMP9Human Mesenchymal Stem Cells----Athymic Nude Rat Lumbar FusionMicroCT evidence of ectopic bone formation with histologic demonstration of fusion with posterior spinal elements
Wang et al. [179] Ad-BMP2Rat Bone Marrow CellsDBM or collagen spongeLewis Rat Lumbar FusionRadiograph, histologic, and mechanical testing demonstrate spinal fusion in both carriers
Hidaka et al. [175]Ad-BMP7Rat Bone Marrow CellsAllogeneic AllograftLewis Rat Lumbar FusionRadiograph, histologic, and mechanical testing demonstrate spinal fusion
Lee et al. [180]Ad-BMP2Human MyocytesCollagen MatrixSCID Mouse 5 mm Calvarial DefectBridging bone appears at 2 wks postoperatively with significant healing and periosteum at 4 wks
Feeley et al. [161]LV-BMP2 or Ad-BMP2Rat Bone Marrow CellsCollagen SpongeSCID Mouse 4 mm Radial DefectLV-BMP2 continued BMP2 production at 12 wks compared to only 4 wks for Ad-BMP2, with radiographic and histologic healing for both vectors
Virk et al. [162]LV-BMP2 or Ad-BMP2Rat Bone Marrow CellsCalcium Phosphate plus Type I CollagenLewis Rat 8 mm Femoral DefectHigher rates of healing on radiographs and microCT with improved mechanical properties for LV-BMP2 vs. Ad-BMP2
Virk et al. [152]LV-BMP2“Same-Day” Rat Bone Marrow Cells vs. TraditionalCalcium Phosphate plus Type I CollagenLewis Rat 8 mm Femoral Defect“Same-Day” ex vivo technique resulting in faster rates of healing with increased bone formation and improved biomechanics compared to traditional methods
Alluri et al. [181]LV-BMP2Rat Bone Marrow Cells3D-printed Tricalcium Phosphate ScaffoldLewis Rat 6 mm Femoral DefectRadiographic healing with histology demonstrating trabecular bone circumferentially
Miyazaki et al. [173]LV-BMP2Rat Bone Marrow CellsCollagen CarrierLewis Rat Lumbar FusionRadiographic, microCT, and histologic evidence of healing with resorption of collagen sponge
Miyazaki et al. [172] LV-BMP2 or Ad-BMP2Rat Bone Marrow CellCollagen CarrierLewis Rat Lumbar FusionImproved spinal fusion with LV-BMP2 seen on radiographs, microCT, and histology vs. Ad-BMP2
Miyazaki et al. [182]Ad-BMP2Human Bone Marrow or Adipose Stem CellsCollagen CarrierAthymic Nude Rat Lumbar FusionEquivalent fusion on radiographics, microCT, histology, and mechanical testing between bone marrow and stem cell groups
Vakhshori et al. [151]LV-BMP2Human Adipose Stem CellTricalcium phosphate/HydroxyapatiteAthymic Nude Rat 6 mm Femoral DefectEquivalent radiograph, microCT, histological, and biomechanical testing compared to rhBMP
Kang et al. [183]LV-BMP2Rat Bone Marrow Cells3D Printed Hyperelastic boneLewis Rat 6 mm Femoral DefectRadiographic and histologic evidence of healing with bony ingrowth on scaffold
Ad-BMP2, adenovirus vector expressing a bone morphogenetic protein-2; DBM, demineralized bone matrix; SCID, severe combined immunodeficiency; LV-BMP2, lentivirus vector expressing a bone morphogenetic protein-2; rhBMP, recombinant human bone morphogenetic protein.
Table 5. In vivo viral vectors used to deliver BMP in preclinical animal models.
Table 5. In vivo viral vectors used to deliver BMP in preclinical animal models.
AuthorVectorModelResults
Rundle et al. [184]MLV-BMP2/BMP4 hybridSprague–Dawley Femur FractureSignificant callous formation early post-injection; however, mostly found to be extra-periosteal. Histology failed to demonstrate significant differences in remodeling compared to controls.
Baltzer et al. [185]AV-BMP2New Zealand White Rabbit Femoral DefectDefects were 75% restored after 7 wks and healed after 12 wks in the experimental group only. Histology demonstrated a bridging callus present at 8 wks post-injection.
Helm et al. [178]AV-BMP9Athymic Nude Rat Lumbar Fusion16 wks post-injection microCT demonstrated fusion mass in direct contact with posterior spinal elements without canal compromise. Histological evidence of lamellar bone with marrow cavities developed.
Alden et al. [174]AV-BMP2Athymic Nude Rat Lumbar Fusion12 wks post-injection microCT demonstrated fusion mass in direct contact with posterior spinal elements. Sharp borders observed on histology but no adverse reaction in surrounding paraspinal musculature.
Laurent et al. [177]AV-BMP6New Zealand White Rabbit Lumbar Fusion14 wks post-injection microCT demonstrated fusion mass in direct contact with posterior spinal elements. Histological evidence of bony bridging between transverse processes.
MLV-BMP2/BMP4, murine leukemia virus retroviral vector expressing bone morphogenetic protein 2 and 4. AV-BMP, adenovirus vector expressing bone morphogenetic protein.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Ball, J.R.; Shelby, T.; Hernandez, F.; Mayfield, C.K.; Lieberman, J.R. Delivery of Growth Factors to Enhance Bone Repair. Bioengineering 2023, 10, 1252. https://doi.org/10.3390/bioengineering10111252

AMA Style

Ball JR, Shelby T, Hernandez F, Mayfield CK, Lieberman JR. Delivery of Growth Factors to Enhance Bone Repair. Bioengineering. 2023; 10(11):1252. https://doi.org/10.3390/bioengineering10111252

Chicago/Turabian Style

Ball, Jacob R., Tara Shelby, Fergui Hernandez, Cory K. Mayfield, and Jay R. Lieberman. 2023. "Delivery of Growth Factors to Enhance Bone Repair" Bioengineering 10, no. 11: 1252. https://doi.org/10.3390/bioengineering10111252

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