Abstract
Fracture nonunion remains an unresolved complication after extremity fracture, with notable costs to patient quality of life and health systems. Nonunion is defined by the inability of fracture ends to unite without evidence of progressive healing over time. Approximately 2 to 10% of all fractures go onto nonunion, with increased rates observed in specific fracture locations and patient populations. Despite advances in fixation techniques and bone grafting, current treatments remain limited and frequently fail to restore durable bone healing. In this review, the current state of emerging biologic and bioengineering therapies for nonunion will be summarized, with a focus on how these advances may shift treatment from palliative reconstruction toward durable healing. Biological therapies such as growth factors, stem cells, and gene-modified constructs show promise but face challenges of short half-life, inconsistent efficacy, and safety concerns. Emerging approaches, including controlled-release scaffolds, immunomodulatory materials, stem cell-derived exosomes, and gene therapy platforms, offer opportunities to more precisely restore the osteogenic, angiogenic, and immunologic environment required for union.
1. Introduction
Extremity fractures represent a common and morbid physical injury frequently seen after higher energy trauma. Globally, over 178 million new fractures were estimated in 2019 by the Global Burden of Diseases, Injuries, and Risk Factors Study group [,]. While the majority of fractures go on to heal, a notable subset may not. The incidence of nonunion widely varies, with reported rates ranging from 2% to 50%, depending on the fracture type and presence of concomitant risk factors [,,,]. The true incidence is likely between 2% and 5% for the majority of cases, though specific bones, such as the scaphoid, clavicle, and femur have a risk exceeding 10% [,,,,,]. Further, there are subsets of fractures and injury patterns which may portend a nonunion rate of well over 30% [,]. Fracture-related factors that influence nonunion risk include location, presence of concomitant fractures, open fractures, and poor residual fracture alignment after initial reduction attempts [,,]. To that end, studies have reported close to a 20% nonunion rate in open tibial fractures, with the rate increasing if there is vascular compromise [,,,]. Due to the significant prevalence of nonunited fractures, understanding their causes and management strategies is of high importance.
Nonunion is defined by the United States Federal Drug Administration as a fracture which has not united by nine months after injury or one in which there is failure of progression towards union over three months []. While this definition can be useful to define a broad framework for understanding nonunion, patient and fracture specific factors must also be considered to more precisely capture the clinical picture. To more pragmatically characterize nonunion, others have defined it as a fracture that will not unite without further intervention []. Radiographic measures of nonunion have also been proposed. These include the Radiographic Union Scale in Tibia fractures (RUST) and the Radiographic Union Scale in Humeral fractures (RUSHU) scales, which score the probability of successful healing []. In addition to radiographic findings, laboratory measures are also key in defining the exact etiology of nonunion. These measures include the evaluation of inflammatory markers, such as C-reactive protein (CRP) to rule out infectious causes, as well as metabolically active endocrine factors, like vitamin D and parathyroid hormone []. Importantly, none of these measures should be used in isolation to define or predict the risk of nonunion, thus highlighting the patient-specific framework necessary to accurately diagnose this pathology.
Nonunion has been classified into several distinct categories, including septic and aseptic forms. Aseptic nonunion is further subclassified into hypertrophic, oligotrophic, and atrophic forms which references both the radiographic appearance of the fracture, as well as the presumed local microenvironment driving the observed radiographic findings []. A hypertrophic nonunion is presumed to have poor fixation, thus leading to excessive motion about the fracture ends, however, with good local biological healing potential []. This combination results in excessive callus formation, but without discrete resolution of fracture lines nor remodeling of the cartilaginous scaffold. Conversely, atrophic nonunion represents poor local biology which is radiographically characterized by lack of callus about the fracture site and resorption of the fracture ends []. While these two classifications are important conceptually, the majority of nonunions are best classified as oligotrophic nonunions which have a combination of poor fixation and poor local biology, and commonly appear with minimal callus []. These classifications help physicians define nonunion but its impact is better understood by examining the risk factors that lead to poor outcomes and the consequences on patients’ lives.
2. Clinical Context
2.1. Clinical Significance
Nonunited fractures have a significant impact on patients’ quality of life, often causing a notable loss of mobility and severe pain [,,,]. Several studies report substandard quality of life for patients with nonunion, comparable to multiple sclerosis and certain cancers [,,,]. Additionally, there is a significant impact on mental health. Patients with nonunion experience increased levels of anxiety and depression, related to their declining health, mobility issues, multiple procedures, uncertainty surrounding the healing process, and social isolation related to their persistent health challenges and prolonged treatment course [,,,]. Nonunited fractures also impose significant socioeconomic burdens on both the patient and the healthcare system as a whole. Direct costs, such as office visits, increased number of surgical procedures, and prescriptions increase expenses for the patient [,,]. Additionally, patients require increased physical therapy sessions, home health services and prolonged hospital stays, further exacerbating their physical and financial burden [,,]. Moreover, indirect costs, primarily related to lost productivity and wages from the inability to work, represent the largest financial cost of nonunions [,,,,,]. Given the extensive economic impact of nonunion through both direct and indirect costs to the patient, the healthcare system, and to society, the development of effective treatment solutions is vital.
2.2. Risk Factors
There are many risk factors for nonunion. Injury-related factors are particularly consequential, compromising the mechanical stability and local biological environment for healing. These are commonly observed in higher energy injury mechanisms and include soft tissue damage and greater bone loss leading to difficulties in maintaining fracture stability [,,,]. These factors are often associated with injury to the local vascular supply, which decreases the availability of oxygen and nutrients, thereby delaying healing and impairing an effective immune response. Given the risk of wound contamination during the injury, surgery, and postoperatively, the inability to mount an effective immune response may result in a fracture related infection, which further heightens nonunion risk [,,]. Furthermore, several comorbid health conditions have been associated with nonunion, including diabetes, rheumatoid arthritis, and primary osteoarthritis which have been shown to increase the risk of nonunion by up to 40% [,,]. Additionally, modifiable patient factors such as obesity, alcoholism, and smoking amplify this risk [,,]. Smoking has been shown to impair fracture healing by reducing blood flow, in part through suppression of vascular endothelial growth factor (VEGF [VEGFA]) and von Willebrand factor (vWF [VFW]), which are essential for angiogenesis. Smoking also decreases the expression of genes critical for osteogenesis, including alkaline phosphatase (ALP [ALPL]) and bone morphogenetic proteins (BMPs) []. Similarly, alcohol disrupts the Wnt pathway, a molecular pathway important for bone healing and the genesis of osteoblasts []. Many of these risk factors are thought to influence bone healing by creating a pro-inflammatory environment that promotes osteoclasts and decreases osteoblastic activity, leading to increased bone resorption [,,,,,]. In particular, diseases such as osteoarthritis, diabetes and rheumatoid arthritis lead to chronic, systemic inflammation which negatively influences bone remodeling [,].
While comorbidity related risk factors for non-union have been established, genetic predisposition has also emerged as an area of investigation. In a study comparing peripheral venous blood samples from patients with nonunion to successfully healed fractures, polymorphisms within the platelet derived growth factor (PDGF) gene emerged as a potential genetic risk factor []. Additionally, a gain-of-function polymorphism within a matrix metalloproteinase, an enzyme critical to extracellular matrix remodeling, was associated with accelerated fracture healing [,]. Freedman et al. investigated single nucleotide polymorphisms (SNPs) in BMPs, reporting an inverse relationship with bone mineral density (BMD). Similarly, a genome-wide association study further identified ADAMTS18 and TGFBR3 as genes contributing to inter-ethnic variation in BMD []. Overall, these polymorphisms have several implications in the potential for early identification of patients at risk and targeted therapeutic strategies.
3. Pathophysiology
3.1. Bone Healing
Bone healing can occur through primary (direct) or secondary (indirect) healing (Figure 1).
Figure 1.
Schematic of primary and secondary bone healing. Primary bone healing (top) occurs under conditions of absolute stability, where osteoclasts create cutting cones that traverse the fracture line, followed by osteoblasts depositing lamellar bone in the reversal and closing zones, resulting in direct remodeling without callus formation []. Secondary bone healing (bottom) occurs under conditions of relative stability and follows a staged progression beginning with (1) hematoma formation within 24 h, providing a preliminary scaffold for inflammatory cell infiltration, (2) an inflammatory phase marked by neutrophil and macrophage recruitment and release of cytokines, (3) stabilization of a cartilaginous soft callus with early vascular invasion and progenitor cell differentiation over the first week, and (4) formation of a mineralized hard callus by one month, ultimately leading to bridging and consolidation of the fracture []. Created in BioRender. Shelby, H. (2025) https://BioRender.com/l9yx33f.
Primary healing occurs when the fragments are reduced and stabilized under compression with minimal motion at the fracture site. Perren’s strain theory lays the foundation for this understanding. Strain is the measure of the relative deformation of a material when a given force is applied and it ultimately determines the type of tissue that can form in the fracture gap. For instance, in the condition of absolute stability with inter-fragmentary strain less than 2%, lamellar bone formation will occur without significant callus formation (primary healing). Conversely, a range of 2–10% will encourage secondary bone healing with callus formation. Strain outside this range disrupts osteogenesis, promotes fibrocartilaginous differentiation of mesenchymal stem cells (MSCs), and ultimately increases the risk of delayed union or nonunion []. In high-strain environments, woven bone cannot tolerate the deformation, thus interrupting the hard callus phase, preventing stable bridging.
Expanding on primary bone healing, in the context of absolute stability, bones are able to heal via remodeling of the lamellar bone and Haversian canals with minimal callus formation. This process involves osteoclasts, specialized cells responsible for bone resorption, forming “cutting cones” at the end of the Haversian canals near the fracture site. The cutting cones create tunnels across the fracture lines, followed by osteoblasts generating bone across the plane []. Paradoxically, the process of direct bone healing typically takes longer compared to indirect healing. It is worth noting that if rigid fixation is imperfect, potentially through micro-motion from poor compression or implant loosening, the strain may exceed tolerance for direct bone formation, and the callus will not form []. This will ultimately lead to the development of a nonunion. At the cellular level, when mechanical stability is achieved and interfragmentary strain remains low, MSCs experience a stiff microenvironment that enhances cytoskeletal tension, activating integrin–FAK–Src complexes and downstream mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) cascades. These pathways converge on Wnt/β-catenin signaling, promoting nuclear translocation of RUNX2 and Osterix (SP7) to drive osteogenic differentiation [,]. Conversely, excessive micromotion or unstable fixation alters these mechanical inputs, leading to diversion toward chondrogenic or fibroblastic phenotypes with impaired bone formation [].
Secondary bone healing is more common and involves both intramembranous and endochondral ossification. Following injury, disruption of blood vessels supplying the bone leads to the formation of a hematoma, which serves as a scaffold for immune and progenitor cell infiltration (Figure 2).
Figure 2.
Cellular and molecular events during the early inflammatory phase of secondary bone healing. Following fracture, hypoxia, acidosis, and tissue necrosis trigger the release of debris and inflammatory mediators. Neutrophils are rapidly recruited and secrete pro-inflammatory cytokines (interleukins [IL-1, IL-6], tumor necrosis factor- α [TNF-α]), while monocytes (M0) differentiate into M1 macrophages under the influence of pathogen-associated molecular patterns (PAMPs), damage-associated molecular patterns (DAMPs), and inflammatory cytokines (interferon-γ [INF-γ], TNF-α, IL-1) [,]. M1 macrophages promote osteoclast activity for necrotic tissue clearance and sustain a pro-inflammatory environment []. Polarization of M1 macrophages to M2 macrophages is driven by IL-4, IL-10, and IL-13, marking the resolution of early inflammation to initiate tissue repair via secretion of IL-10 and transforming growth factor-β (TGF-β) []. M2 macrophages stimulate reparative granuloma formation, serving as a template for callus. Concurrently, stromal cell-derived factor 1 (SDF1 [CXCL12]) recruits MSCs, which differentiate into osteoblasts (via BMP-2 signaling) and support angiogenesis (via VEGF signaling) []. Created in BioRender. Wier, J. (2025) https://BioRender.com/7ezf2ps.
This acute inflammatory phase represents the first stage of secondary bone healing and generally peaks within 24 h and ends after 7 days. It is characterized by wide platelet degradation that results in the subsequent release of pro-inflammatory cytokines, including interleukins (IL-1, IL-6) and TNF- α [,]. These cytokines result in the recruitment and proliferation of macrophages and neutrophils. First, neutrophils are recruited and then monocytes/macrophages infiltrate into the fracture site. PAMPs and DAMPs, as well as pro-inflammatory cytokines (e.g., INF-γ, TNF-α, IL-1) promote the M1 (pro-inflammatory) phenotype of macrophages. M1 macrophages amplify the inflammatory reaction, recruit additional immune cells, and phagocytose microorganisms, necrotic tissue, and the provisional fibrin matrix. After neutrophils and macrophages clear the area of cellular debris, the process transitions to the resolution of inflammation and formation of a reparative granuloma that forms the template for the subsequent callus. During the resolution of acute inflammation, macrophages are polarized from an M1 phenotype to an M2 (pro-healing) phenotype by anti-inflammatory cytokines IL-4, IL-10, and IL-13. (Figure 2) Bone marrow derived stem cells (BMSC) are attracted locally by cytokines such as TNF-α and SDF1 [,]. Recruited inflammatory cells and BMSCs participate in critical inter-cellular communication or crosstalk via pro-inflammatory cytokines and anti-inflammatory cytokines. Moreover, the release of multiple local proteins, including BMPs and additional growth factors (e.g., VEGF, PDGF, fibroblast growth factor-2 [FGF-2], TGF-β) further potentiates the initiation of osteogenesis and angiogenesis []. The following phase is characterized by the formation of a callus, which involves both intramembranous ossification and endochondral ossification. At the periosteum, intramembranous ossification occurs and a hard callus forms directly, whereby periosteal MSCs differentiate into osteoblasts that directly form woven bone [,].
BMPs, along with other regulatory factors, play an integral role in this process of callus formation (Figure 3).
Figure 3.
Cellular mechanisms underlying osteogenesis. Bone morphogenetic protein (BMP) signaling activates SMAD1/5/8, which associate with SMAD4 and translocates to the nucleus, while canonical Wnt/β-catenin signaling, mediated through LRP5/6 and Dishevelled, stabilizes β-catenin to promote transcriptional activity [,]. In parallel, mechanotransduction activate integrins and Ca2+ channels, initiating intracellular signaling cascades through Src, FAK, and MAPK pathways []. These converging pathways regulate key transcription factors, including RUNX2, Osterix, DLX5, MSX2, and TCF/LEF, which drive the expression of osteogenic genes such as ALP, collagen type I (COL1A1), osteocalcin (OCN [BGLAP]), and osteopontin (OPN [SPP1]) [,]. Together, these mechanisms coordinate matrix mineralization, angiogenesis, and osteoblast differentiation. The lower panel illustrates the stepwise progression from MSCs to pre-osteoblasts, mature osteoblasts, and ultimately mineralized osteoid, highlighting the integrated molecular and cellular processes essential for bone formation. Created in BioRender. Bergren, S. (2025) https://BioRender.com/qt1yy6z.
BMP initiates the signal transduction cascade by binding to cell surface receptors and forming a heterotetrameric complex composed of two dimers of type I and type II serine/threonine kinase receptors. Activated BMP receptors phosphorylate Smad proteins (specifically Smad 1, Smad 5, and Smad 8) and these bind to a common mediator protein, Smad 4, to form a complex []. This complex translocates to the nucleus where it functions as a transcription factor. Downstream, the complex cooperates with additional transcription factors (e.g., RUNX2, Osterix, DLX5, MSX2) to upregulate key genes promoting osteogenesis. These include ALP, involved in matrix mineralization, COL1A1, integral to type I collagen synthesis, OCN, a late osteoblast marker, OPN, involved in bone matrix adhesion, and VEGF, which promotes angiogenesis for callus vascularization []. Smad-independent pathways also contribute to bone healing. These include MAPK cascades and small GTPases, which promote osteoblast differentiation and early proliferation of osteoprogenitor cells. RUNX2, known as the “master regulator of osteoblast differentiation,” also inhibits chondrocytes from acquiring the phenotypes of permanent cartilage chondrocytes, allowing this cartilage to be replaced by bone, making RUNX2 a key player in endochondral ossification []. Finally, the last stage of bone remodeling can persist for several months. The process involves the coordinated interaction of signaling pathways, featuring BMP, FGF, and parathyroid hormone-related protein (PTHrP). Continued migration of osteoblast and osteoclasts are contributors to “coupled remodeling”, the dynamic equilibrium between bone resorption and bone formation [].
3.2. Impaired Healing
Despite this coordinated and tightly regulated process, bone healing does not always occur successfully. Among areas of investigation, the acute inflammatory phase of bone healing is one component that seems to influence the progression of nonunion. Imbalance in the early macrophage polarization signals and other paracrine factors which promote the orchestrated transition through each phase of healing can lead to delays and potential cessation in osteogenic progression. Persistence of an M1-dominant milieu has been linked to nonunion, as well as impaired callus formation [,,]. Conversely, the coordinated induction of M2 macrophages promotes vascular invasion and osteoblast recruitment [,]. Additional murine models have been used to demonstrate that unregulated acute inflammation impairs fracture healing [,,]. In a clinically relevant model, Recknagel et al. used a blast wave generator to induce chest trauma following an osteotomy, demonstrating delayed healing with increased neutrophil infiltration and elevated pro-inflammatory cytokines at the fracture site [,]. Moreover, unstable fractures which are commonly associated with delayed healing, have been shown to exhibit significantly more cytotoxic T cells and leukocytes in the hematoma neighboring the injury when compared to those with stabilized fractures [,].
The local biology, including the role of adequate blood supply, is another key factor implicated in non-union. It was long theorized that atrophic non-unions were avascular and biologically inert. However, this framework has been largely disproven, with evidence that these tissues retain vascularity even in the setting of atrophic nonunion. To that end, Reed et al. reported no significant difference in vessel density between atrophic non-unions, hypertrophic nonunions and healing fractures []. Nevertheless, adequate blood supply remains a key component in promoting healing and reducing the risk of fracture related infections. Local protein factors, such as BMPs have also been implicated in the dysregulation of bone repair. Kloen et al. reported evidence of ongoing BMP signaling in non-union tissue, while other studies have revealed an imbalance between BMP’s expression and that of their inhibitors in this setting [,,]. Similarly, matrix metalloproteinases (MMPs) have been highlighted in non-union pathophysiology as these proteins bind and degrade BMP-2 []. The molecular and biochemical pathways underlying impaired fracture repair remain an area of investigation, with ongoing research working to identify therapeutic targets to restore osteogenesis.
4. Current Solutions to Address Nonunion
Several important factors are necessary for bony union, which include the presence of osteogenic cells, osteoinductive stimulus, osteoconductive scaffold, and a robust vascular supply [,]. Current interventions are designed to reintroduce these key elements of osteogenesis that are deficient in nonunited fractures. Techniques such as bone grafting, local cellular and molecular therapies, or a manufactured scaffold, aim to mimic the bone growth environment and compensate for any deficient elements needed for bone formation [,,] (Table 1). These interventions strive to augment missing structural or biologic elements needed for durable fracture healing and mimic the original pathways necessary for bone development and repair.
Table 1.
Summary of recent key clinical and non-clinical studies related to fracture healing.
4.1. Nonsurgical
Non-invasive methods are more commonly considered in cases of delayed union or in patients where surgical intervention is precluded. These include prolonged immobilization and external modes of bone growth stimulation (BGS). Initial management of delayed union can consist of an extended period of bracing or casting of the affected limb []. However, these methods commonly do not result in complete fracture healing and carry a much higher risk of malunion which may further exacerbate the functional deficits caused by the fracture. BGS is an adjunct to immobilization, with the goal of reducing the rate of nonunion and facilitating healing if delayed. Examples of external BGS include low intensity pulsed ultrasound (LIPUS), electrical stimulation (ESTIM)/pulsed electromagnetic fields (PEMF), and extracorporeal shockwave therapy (ESWT). The clinical interest in LIPUS has grown over time, while ESTIM has fallen out of favor, with several reviews demonstrating unfavorable healing rates ranging from 55 to 61% [,]. Conversely, the use of LIPUS in the setting of nonunion has been shown to have an 82% healing rate, with even greater healing rates for fragility fracture patients with non-unions [,]. Despite this encouraging data, several randomized controlled trials evaluating the effect of LIPUS on nonunion healing demonstrated no superiority compared to a control group [,]. As such, there is limited adoption of these external BGS methods in modern orthopaedics.
4.2. Surgical Solutions
4.2.1. Traditional Surgical Approaches
Nonunited fractures are commonly treated with internal fixation using plates or intramedullary nails (IMNs), typically combined with a bone graft [,,]. These require either increasing or decreasing the strain the fracture sees depending on the type of fixation primarily used and the character of the resulting nonunion. In the setting of hypertrophic nonunions, which commonly have excessive bone formation and impede appropriate bony opposition, partial decortication is often employed to reduce the osseous burden and allow for optimal fixation []. Different surgical constructs are used to elicit primary or secondary healing. Implants like IMNs, which promote secondary healing, allow for the mechanical continuity of bone to be restored so that daily movements can physically stimulate healing through mechanotransduction []. In certain cases, compression plating, which evokes primary healing, is used, in which a metal plate is fixed to the fracture segments and screws are used to provide a compressive force between the bony ends (Figure 4).
Figure 4.
Ulnar nonunion with compression plating. 35-year-old female that presented 2 months after sustaining an ulnar fracture and was found to have nonunion of her fracture. The patient was treated with an open reduction and internal fixation with a compression plate. (A) Radiograph from when the patient was first diagnosed with nonunion (orange arrow). (B) Radiograph 3 months after fixation.
Additional techniques in the setting of large bone defects include the Masquelet technique and distraction osteogenesis. The Masquelet technique involves implanting a polymethyl methacrylate cement spacer in the bone defect that acts as a foreign body to induce a vascularized membrane []. The cement spacer induces inflammation and edema at the site of nonunion causing a membrane to form. This membrane will release growth factors and cytokines to induce angiogenesis, forming a vascular supply for the healing bone [,]. This process can take several months, after which the spacer is removed and the empty void is filled with autologous bone graft (Figure 5).
Figure 5.
Nonunion of pilon fracture with induced membrane and local biologic therapy. 27-year-old male who was in a motor vehicle accident resulting in a pilon fracture. One year after his injury, the patient was diagnosed with septic tibial nonunion and underwent repair with an induced membrane, iliac autograft and rhBMP-2. (A) Radiograph of initial injury demonstrating comminuted distal tibial fracture with intra-articular extension and ipsilateral fibula fracture. (B) Radiograph of primary hardware at over 6 months after initial fixation when the patient was diagnosed with infected nonunion. Orange arrow pointing at persistent fracture lines. (C) Radiograph of antibiotic spacer (purple oval) resulting in an induced membrane (not visible) with external fixator in place to hold alignment. (D) Radiograph 3 years following repair with evidence of fracture consolidation (purple arrow) and new plate fixation.
Gaillard et al. demonstrated that this technique can be successful in humeral fracture nonunions, with 15 out of 15 nonunion achieving union []. Success has also been shown with nonunions of the tibia, radius, ulna and the femur, with rates of union between approximately 85–95% [,,,,,].
Distraction osteogenesis has shown promise in promoting healing in nonunited fractures with large defects []. In this technique, an osteotomy is performed, and the bone is progressively distracted as new bone fills in the gap (Figure 6).
Figure 6.
Distraction osteogenesis of the tibia following nonunion. 57-year-old male with an open tibial and fibular septic nonunion following a motorcycle collision. (A) Radiograph of initial injury demonstrating a displaced segmental distal third tibia fracture with ipsilateral fibula fracture. (B) Initial fracture fixation using intramedullary nailing to allow for secondary bone healing. (C) Radiograph after debridement and saucerization of regions of tibial osteomyelitis with dynamic external fixator in place. Region of regenerate highlighted with orange arrow. (D) Definitive fixation with intramedullary nail after removal of dynamic external fixator at 4 months after initial external fixator placement. (E) Most recent radiograph at 6 weeks after intramedullary nail placement with evidence of early callus about region of regenerate (purple arrow).
Additional strategies include an externally controlled dynamic intramedullary nail that uses a magnetic mechanism to provide compression or distraction at the fracture site. In a study of five tibial and nine femur nonunions, an intramedullary compression nail was able to achieve union in 13/14 cases []. While these surgical techniques can be successful, the biological or mechanical environments may not always be fully restored. Moreover, these techniques may require multiple surgical procedures which increase the risk of complications, including infection and devascularization of the local environment, which further prolongs healing and increases nonunion risk. In this setting of multiple surgeries and potential complications, an immense psychosocial burden is placed on patients and patients are commonly confronted with having to choose amputation despite multiple prolonged treatment courses []. As such, opportunities to reduce the nonunion risk during the primary surgery or limit the interventions to a single repeat surgery are of high importance.
4.2.2. Surgical Solutions with Biologic Therapy
In addition to targeting the biomechanical environment, surgical exposure allows for the direct application of biologic therapies. Bone grafting is a commonly used option to bridge small bone defects and can be sourced autologously or as an acellular allograft [,]. Autologous bone graft has several advantages over allograft, which only serves as an osteoconductive matrix, as it contains a patient’s own osteogenic cells and osteoinductive proteins, as well as serving as a scaffold for osteoconduction. Common regions used to obtain autograft include the iliac crest, the tibia, and the femoral intramedullary canal []. However, local graft can be obtained from nearly any site. The major limitation of autologous bone graft is availability and donor site morbidity, as it is typically harvested from a discrete site outside the zone of injury thus requiring additional surgical exposure. Vascularized bone graft is an attractive option for large bone defects as it not only provides the benefits of autologous bone, but also a direct vascular conduit to the implanted tissue. However, the risk of morbidity and additional surgical procedures is particularly relevant for vascularized bone graft, such as free fibula transfers, which require significant surgical expertise and are associated with clinically relevant rates of wound complications and chronic pain [].
While autograft is limited to ~20–40 cm3 of available tissue, bone allograft is widely available at large quantities, however, at a significant monetary cost []. Allogeneic bone grafts are harvested from one individual, living or cadaveric, and implanted into another. These grafts are predominantly osteoconductive in nature, serving as a scaffold for new bone formation but may maintain some osteoinductive properties []. Critically, once prepared and sterilized, these grafts no longer contain osteogenic cells or key growth factors, serving primarily as a scaffold for bony ingrowth []. The use of allografts has limited utility in nonunion treatment because of their poor biological activity with evidence of higher risk of re-fracture and nonunion when used in large bone defects []. Despite the widespread use of these bone grafts, the aforementioned limitations highlight the need to investigate adjunctive therapies.
Bone marrow aspirate concentrate (BMAC) can be used as the sole graft material or as a potential adjunct to grafts for bone regeneration (Figure 7).
Figure 7.
Femur nonunion with BMAC and nail dynamization. 27-year-old male with a nonunion following intramedullary nailing of a displaced comminuted femur fracture. Six months after his initial surgery the patient was diagnosed with nonunion of the fracture. (A,B) Radiographic findings of femur nonunion with residual fracture line through region of hypertrophic callus (orange arrow). (C) Distal locking screws (orange circle) in initial intramedullary nail construct. (D,E) Radiographic evidence of union at one year after nail dynamization and BMAC placement, with resolution of multicortical fracture lines (purple arrow). (F) Evidence of removed distal locking screws (purple circle) to dynamize intramedullary nail for compression of proximal fracture segment.
BMAC is obtained through aspiration of bone marrow, usually from the iliac crest. The aspirate is then processed to concentrate MSCs and associated growth factors such as VEGF, PDGF, and BMPs [,,]. BMAC has shown to lead to union in 75% to 100% of cases, particularly with septic nonunions [,,]. Further, the addition of BMAC to the bone grafts was shown to accelerate healing times in nonunion compared to patients treated with standard surgical fixation and bone graft alone []. Despite its promising clinical applications, one of the main limitations is the variable amount of stem cells and the heterogeneity of its contents across different individuals. In a study of 8 patients, the percentage of MSCs within BMAC generated from each patient’s bone marrow ranged from 0.0001% to 0.003% []. Another study highlighted how the age of a patient and the origin of the bone marrow influenced the amount of mononuclear cells, which includes stem cells, within BMAC, with younger patients having a higher proportion of monocular cells and the iliac crest producing more cells than the tibia []. Furthermore, another disadvantage of this method is the iatrogenic pain and morbidity associated with bone marrow retrieval. Overall, BMAC is a promising adjunct for nonunion treatment, but improved isolation techniques and more high-quality research are needed to better understand its efficacy in this context.
Platelet rich plasma (PRP) is another adjunct to bone grafts that can be used in nonunion cases. PRP is typically sourced by isolating platelets and their associated growth factors from autologous blood. Through this process, PRP is proposed to deliver a concentrated milieu of critical growth factors, such as PDGF, TGF-β, and VEGF, as well as immunomodulatory protein such as interleukins which promote bone healing [,]. Wang et al. demonstrated that in a retrospective study of 66 patients with femoral nonunions, PRP significantly increased healing rates by 20% and decreased healing time by almost 4 months []. Additionally, similar to BMAC, the use of PRP has demonstrated enhanced healing rates relative to surgical fixation and bone grafts []. Although PRP has the potential to be effective in improving healing in nonunion, the evidence remains mixed. A recent meta-analysis showed that PRP may lead to a faster time to union but results were inconclusive on whether it positively influenced healing rates []. Specifically, concerns remain about the reproducibility of PRP, with evidence that the molecular composition varies widely between individuals and collection methods [,,]. Given these inconsistencies, more data is required to precisely define its effect on nonunion and its optimal use.
5. Emerging Strategies for Nonunion Repair
Emerging strategies leverage local biology to enhance bone formation through the use of growth factors, immunomodulation, scaffolds, local drug delivery, stem cells and gene therapy. Each of these components targets a key factor in bone stimulation (Table 2).
Table 2.
Summary of advantages and limitations of clinical and preclinical modalities used to promote fracture healing.
5.1. Exogenous Growth Factors
Growth factors target the upregulation of osteogenic differentiation and proliferation, with potential targets including not only osteoinductive factors, but also those related to angiogenesis and immunomodulation [,]. Multiple families of growth factors are presently being investigated as potential therapeutic targets, such as VEGF, PDGF, TNF-ɑ, and the TGF-β superfamily including BMPs [] (Table 3). However, their utility is limited by rapid diffusion and short local retention times, which hinder sustained biological activity at the target site []. As a result, localized delivery systems are being developed to enhance the spatial and temporal availability of these factors, thereby promoting more effective therapeutic responses.
Table 3.
Summary of key non-BMP growth factors implicated in fracture healing.
5.1.1. BMP
Although several strategies leveraging osteoinductive growth factors have also shown promise, BMP-2 has emerged as the most popular to enhance bone regeneration. Recombinant BMP-2 (rhBMP-2) is an analog designed to stimulate differentiation of MSCs into osteoblasts which has achieved FDA approval as an adjunctive treatment modality for acute open tibial fractures []. A series of preclinical studies have demonstrated that rhBMP-2 enhances bone healing when combined with various delivery systems and scaffold materials. It has been shown that rhBMP-2 administered with human periosteum-derived cells recapitulate physiological fracture healing in mouse tibia, while subsequent studies in rat models confirm rhBMP-2′s role in promoting hypertrophic chondrocyte formation and mineralized bone bridging [,]. When used with allografts or synthetic scaffolds, rhBMP-2 significantly improved integration and mechanical strength, often outperforming autograft controls. To that end, several studies demonstrated that rhBMP-2-loaded constructs induce rapid lamellar bone formation and bridging in rat and canine models [,,]. These combined approaches highlight emerging strategies to augment rhBMP-2 functionality by potentiating its delivery and enhancing bone stability which could ultimately be applied to nonunion scenarios. Despite some evidence of clinical efficacy, concerns related to local adverse tissue reactions due to the high initial dose required to achieve prolonged effects have hampered its widespread use. Specifically, its short half-life necessitates a high local dose to sustain therapeutic effects []. The delivery system is a central limitation to its use, with the only FDA approved carrier for rhBMP being an absorbable clinical sponge [,]. While the sponge allows localized delivery, it exhibits poor control over release kinetics and can lead to rapid burst delivery. This necessitates using a supraphysiologic dose to maintain therapeutic levels. The high burst release has been associated with excessive soft-tissue swelling and formation of seromas [,]. Additionally, the high-dose use of rhBMP-2 has been associated with heterotopic bone formation, soft tissue swelling, osteoclast-driven bone resorption, and oncologic risks in doses exceeding 40 milligrams []. These adverse effects underscore a need for an improved delivery system with controlled-released scaffolds. Multiple approaches have been proposed to achieve this controlled release, including the use of scaffold encapsulation or more specific affinity-based methods targeting key interactions between BMP-2 and functionalized surface substrates. A highly cited example is heparin-mediated BMP-2 delivery using heparin microparticles blended into carriers [,]. In a rat critically sized femoral defect, this approach increased in vivo BMP-2 retention, tightened spatial localization of new bone to the defect, and reduced heterotopic ossification []. A complementary line of studies engineered heparan-sulfate variants (HS3) embedded in collagen/mineral composites to triple BMP-2 retention and extend release to ~4 weeks, yielding significantly greater osteogenic differentiation in vitro []. Similarly, heparin-dopamine conjugates demonstrated significantly improved bone formation than heparin alone on a biologic scaffold []. Collagen-anchoring bridge proteins with dual affinity for BMP-2 and collagen have also produced strong, site-restricted osteogenesis in murine spinal fusion models at reduced doses by tethering BMP-2 directly to the matrix []. These data show that specific molecular interactions are able to modulate spatiotemporal release profiles to enhance defect repair while curbing off-target bone formation.
Beyond optimizing release profiles, research involving BMP has shifted toward more clinically relevant chronic nonunion models. These models use delayed treatment following a period of nonunion development, better simulating clinical scenarios. In this context, Bosemark et al. identified a combination of BMP-7, and systemic bisphosphonates as most effective in nonunited rat femurs [,]. Similar findings were found by DeBaun et al. and Kaipel et al., who used variations of BMP-2 delivery via polycaprolactone scaffolds and fibrin matrices to achieve complete union in chronic defects [,]. However, limitations to BMP-2 therapy have also been noted in this setting, such as dose-dependent heterotopic ossification, and inferior mechanical properties in BMP-induced bone formation [,]. Other adverse effects noted by these studies included ectopic bone formation, rapid BMP clearance, adipogenesis, and bone cysts. These findings underscore the therapeutic potential of BMPs in nonunion repair while highlighting the need for careful dose modulation and safety evaluation prior to clinical translation.
5.1.2. Alternative Molecular Mediators of Bone Healing
In addition to BMP, several alternative growth factors have been explored for the treatment of nonunion, with varying degrees of success []. VEGF enhances angiogenesis and has shown promise in bone regeneration when delivered via gene-activated matrices, although its efficacy falls short when compared to BMP []. Murine models have demonstrated that supra-physiologic concentrations of VEGF can accelerate healing of long bone fractures; however, evidence of direct benefit in human fracture healing remains limited [,]. Additionally, PDGF has been considered a “starter” signal for the wound-healing cascade in tissue regeneration. However, while studies on supplemental rhPDGF have shown improvements in bone healing, it generally fails to achieve complete union alone []. Combinations of rhPDGF with β-TCP and collagen matrices have shown encouraging results in stimulating union; however, additional data on its efficacy are presently needed [,,,,]. Other investigations of molecular targets include sclerostin inhibitors such as romosozumab, which have demonstrated significantly improved bone volume in ulnar defects of cynomolgus monkeys []. Another endogenously derived growth factor which has shown promise in promoting bony union is parathyroid hormone (PTH). By upregulating RUNX2 and OSX, PTH promotes osteoblast differentiation, an important osteoinductive process in bone repair []. Pulsed administration of rhPTH, specifically teriparatide, has been shown in preclinical animal models to enhance bone healing by stimulating callus formation, improving mechanical strength, accelerating endochondral ossification, and increasing union rates [,,,]. From a clinical perspective, several studies demonstrate a modest benefit in rhPTH administration in fracture healing. A meta-analysis of eight randomized trials involving 524 patients found that rhPTH treatment significantly reduced healing time, improved pain relief and functional outcomes, albeit without affecting overall union rates []. Data in younger, non-osteoporotic patients is even less clear, with only a single trial of 13 patients examining the effect of rhPTH on union []. Although these mediators of bone healing have potential, there is collectively limited data in their utility and efficacy in treating fracture nonunion.
5.2. Immunomodulation
Another evolving strategy in the treatment of nonunion is targeting the acute inflammatory phase of bone healing, optimizing early progression toward bony union. Prostaglandin E2 (PGE2) is a proposed target in this inflammatory pathway that has shown promise in improving bone formation. Selective agonists of the PGE2 and PGE4 receptors have resulted in enhanced bone formation and biomechanical strength [,,]. Furthermore, dual administration of a selective PGE4 receptor agonist and rhBMP-2 has demonstrated significantly improved bone healing in murine models []. By targeting a similar biological pathway, Rundle et al. demonstrated accelerated fracture healing in a rat model by overexpressing COX-2 using a retroviral vector system []. Biologic scaffolds are another proposed solution to facilitate the coordinated transition between these inflammatory phases and have shown promise in promoting regeneration. These are manufactured to ensure an optimal surface topography and an exact internal porous architecture to facilitate osteogenesis with maintained structural properties. Notably, these scaffolds have also been shown to modulate the immune microenvironment by enhancing immune cell adhesion, migration, and macrophage polarization []. By leveraging varied scaffold materials and properties, several studies have demonstrated increased M2 polarization and promotion of the restorative inflammatory phase [,,,,]. Xuan et al. demonstrated that a 3D-printed ordered bredigite scaffold promoted M2 polarization by enhancing IL-10 expression while suppressing TNF-α, while also promoting BMSC proliferation, thereby accelerating defect healing in a rat critical-sized femoral model []. Incorporating both inflammatory and osteoinductive mediators has also demonstrated success in facilitating improved osteogenesis. Similarly, Xiong et al. reported that digital light processing-fabricated hydroxyapatite (HA) scaffolds with hierarchical porous structures regulated the immune microenvironment by increasing CD206+ M2 macrophages and reducing iNOS+ M1 macrophages, resulting in elevated BMP-2 secretion, VEGF release, and significantly improved bone formation in their rabbit skull defect model []. Others have engineered functionalized scaffolds using metal ions, bioactive molecules, or piezoelectric materials to provide specific osteoinductive cues, while also modulating macrophage phenotypes []. Another therapeutic approach involves the incorporation of BMP-2 and osteoimmunomodulatory agents, which have demonstrated significantly improved BMP-2-induced in vivo osteogenesis, suggesting that inflammatory inhibition is crucial for maximizing BMP-2 efficacy [,]. Given the essential role of the acute inflammatory phase in modifying the risk of nonunion, identifying how to optimally leverage growth factors and maximize scaffold design to promote bone regeneration will remain a key avenue of future research.
5.3. Scaffolds
Numerous in vitro and in vivo studies have evaluated the therapeutic performance of a broad range of scaffold compositions and architectures, particularly focusing on the capacity of these constructs to support vascularization and osteogenesis. These scaffolds widely range in terms of their material composition and therapeutic effect, with high degrees of modifiability through the use of bioengineering and 3D printing modalities (Table 4).
Table 4.
Overview of material properties of scaffold-based therapies.
5.3.1. Bioceramics
Synthetic bone substitutes can be used to support local osteoconduction with potential osteoinductive properties, as modifications to their surface topography can promote osteoblastic differentiation of MSCs by mimicking the mechanotransductive characteristics of the native extracellular matrix []. Bioceramics are a class of inorganic materials that have been widely explored for the treatment of long bone nonunion and segmental defects due to their innate bioactivity, structural rigidity, and ability to emulate the mineral phase of natural bone. They are composed primarily of calcium-phosphate ceramics such as HA and tricalcium phosphate (TCP), as well as bioactive glass (BAG). Additionally, bioceramics are able to be coated with synthetic biodegradable polymers such as polylactic acid (PLA), polyglycolic acid (PGA), poly-ε-caprolactone (PCL), or polylactic acid co-glycolic acid (PLGA), or natural compounds such as collagen and gelatin to improve structural strength [,]. For example, collagen-wrapped HA scaffolds implanted in rabbit femoral defects resulted in bone formation with mechanical strength equivalent to autologous graft and 3D printed PLA scaffolds delivering rhBMP-2 to sheep metatarsal defects significantly higher bone formation than controls [,]. Importantly, these materials can also be manufactured to fabricate anatomically customized scaffolds with specific porosity and mechanical properties [,]. Engineered microporous structures of bioceramics can facilitate angiogenesis and mineral deposition, with porous TCP scaffolds demonstrating greater bone volume and increased blood vessels in critical sized defect models [,]. Lu et al. reported that diamond pore TCP scaffolds induced osteogenic differentiation and significantly improved bone healing through the Ras pathway, implicating the link between mechanotransductory mechanisms and cell fate []. Several other studies have demonstrated success with piezoelectrically engineered barium titanate scaffolds to accelerate bone repair [,,]. Conceptually these leverage load-driven piezoelectric currents to simulate the natural bioelectricity formed during native bone regeneration. Despite their promise, the majority of bioceramics remain brittle and mechanically inferior to certain synthetic polymers, and their regenerative efficacy may be more influenced by scaffold architecture or fabrication method than the ceramic composition itself [,]. Nevertheless, their integration with other polymers continues to expand their clinical potential in nonunion repair, with hybrid systems enabling controlled therapeutic delivery.
5.3.2. Natural Polymers
Natural polymers have garnered substantial interest due to their intrinsic biocompatibility given their resemblance to native extracellular matrix components []. Materials currently under active investigation include gelatin, collagen, alginate, chitosan, silk fibroin, and hyaluronic acid, each exhibiting distinct degradation kinetics and fabrication compatibility. However, the biological origin of these materials also introduces variability in mechanical integrity and immune compatibility. Some formulations have been associated with increased inflammatory responses which may impair bone healing or elicit adverse host reactions [,]. Additionally, the mechanical strength of most natural polymers is inferior to synthetic alternatives, limiting their application in load-bearing environments unless reinforced or modified. To overcome these limitations, hybrid approaches incorporating natural polymers with ceramics, synthetic polymers, or growth factors have been explored to optimize scaffold mechanical integrity, degradation profile, and osteoinductive capacity in the treatment of bone nonunion [].
Among natural polymers, gelatin has been widely utilized due to its derivation from collagen, excellent biocompatibility, and ease of fabrication into porous scaffolds. Gelatin scaffolds have been used as carriers for endothelial progenitor cells to enhance angiogenesis and bone regeneration in critically sized rat femur defects, demonstrating improved healing when compared to PRP []. Collagen, another prominent scaffold material, represents the primary organic component of bone, and its low immunogenicity and cell-adhesive properties make it ideal for tissue engineering applications [,]. When combined with calcium phosphates or demineralized bone matrix, collagen scaffolds can mimic the composite structure and mineral content of native bone. Schwarz et al. demonstrated significant bone regeneration in rat femur defect models using collagen sponges loaded with BMP-2, though the osteogenic response was largely attributed to the growth factor rather than the collagen matrix alone [].
Other natural polymers under active investigation include alginate and silk fibroin. Alginate can be readily cross-linked under mild conditions to form hydrogels with adaptable porosity and mechanical stiffness. Although alginate is non-toxic and cost-effective, it lacks natural degradability in mammals and often requires irradiation or chemical oxidation to optimize its degradation kinetics [,]. Modified alginate scaffolds have been shown to sustain BMP-2 release and promote bone formation more effectively than unmodified versions or even collagen sponges in rat critical-sized defects [,]. Silk fibroin, derived from silkworm cocoons, exhibits favorable mechanical properties, slow biodegradation, and biocompatibility. These characteristics make it suitable for fabricating scaffolds with customized porosity and topography to support osteoconduction and vascular infiltration [,]. In a rat femoral defect model, Deininger et al. created anisotropically porous silk fibroin scaffolds coated with apatite and loaded with low-dose rhBMP-2 (2.5 µg), achieving substantial bone regeneration while reducing required growth factor concentration []. While no single natural polymer is universally optimal, strategic combinations of materials with bioactive agents can yield scaffolds that effectively support bone healing and nonunion resolution across a range of anatomical and mechanical contexts. However, these scaffolds will need to be tested in clinically relevant animal models to assess their efficacy in more biologically equivalent environments.
5.3.3. Hydrogels
Hydrogels are garnering increased attention in the field of orthopaedics and bone regeneration. Due to the unique architecture of hydrogels, physiologic cues within the local environment (e.g., pH, temperature, etc.) allow them to degrade in response to stimuli, thus more effectively controlling the release of a therapeutic substance [,,]. They can be made with a variety of organic substances such as chitosan, collagen, alginate, and gelatin to mimic the natural extracellular matrix []. Moreover, hydrogel mechanical properties have also shown to directly influence MSC fate, with biomimetic compositions promoting cell adhesion, osteogenic differentiation, and bone repair [,,]. As such, hydrogels can also provide an environment for both osteogenesis and angiogenesis, further facilitating their effectiveness in promoting bone union []. Broadly, hydrogels have shown success as a medium for local drug delivery, with several in vitro and in vivo studies demonstrating improved healing and increased osteoinductivity when applying hydrogels together with local therapy [,,,,]. To that end, hydrogels containing BMSCs or encapsulated rhBMP-2 implanted into bone defect models resulted in improved osteogenesis and successful union [,]. Furthermore, Yu et al. reported successful use of gelatin methacryloyl hydrogel scaffolds in conjunction with preosteoblast-derived matrices in a rabbit model of radial bone defects, highlighting their application in bone regeneration strategies requiring both bioactivity and structural support [,,,]. Even with their current successful applications in nonunion, hydrogels lack mechanical strength and are subject to rapid breakdown, particularly limiting their use in large bone defect scenarios [,,]. Collectively, these studies underscore the versatility of hydrogels as biomimetic scaffolds that not only enable controlled therapeutic delivery but also provide a supportive microenvironment for osteogenesis and angiogenesis, thereby positioning them as promising adjuncts in bone regeneration and union; however, more studies are needed to discover mechanisms to limit the degradation and improve the mechanical strength of hydrogels without compromising their biocompatibility.
5.4. Stem Cell-Based Therapy
Stem cells represent a promising therapeutic avenue for bone union because of their capacity to differentiate directly into osteoblasts [,,,]. Among these, MSCs have garnered particular attention, given their ability to differentiate into osteoblasts, chondrocytes, and adipocytes while also exerting immunomodulatory and pro-angiogenic effects []. BMSCs remain the most extensively studied, although their harvest can be invasive and associated with donor-site pain []. Alternative sources include adipose-derived MSCs (ADSCs) and those harvested from synovial fluid, dental pulp, and umbilical cord blood, which have demonstrated comparable differentiation capacity to BMSCs [,,,]. Prior investigations using direct autologous MSC injections into fracture or nonunion sites have reported radiographic bridging and improved biomechanical properties within months of treatment [,,]. In a recent meta-analysis of 21 studies including 866 patients, Cui et al. evaluated MSC therapy in the setting of nonunion, reporting improved early healing and lower rates of complication than with standard bone graft treatments []. Importantly, the regenerative efficacy of MSC therapy appears to depend on the differentiation status of transplanted cells. In a rat femoral nonunion model with periosteal cauterization and marrow ablation, percutaneous delivery of osteogenically predifferentiated MSCs (OCPs) significantly enhanced bone healing compared to undifferentiated MSCs and untreated controls []. ADSCs are increasingly studied as these cells can be readily obtained from procedures such as liposuction []. In a large animal model, Schubert et al. evaluated osteoblast-differentiated ADSCs in pigs with CT-confirmed fibrotic nonunion and treatment resulted in complete bone fusion, further supporting the therapeutic potential of stem cells in chronic nonunion repair []. However, overall union rates at final follow-up were equivalent to untreated cohorts. Taken together, MSC-based therapy holds substantial promise for accelerating fracture healing and nonunion repair. Ultimately, despite the regenerative potential of MSC therapy, the transplanted cells often show limited survival and a rapid decline in osteogenic activity within the defect site [].
Scaffold-assisted MSC delivery using ceramic or polymeric constructs further enhances osteoconductivity and bone regeneration, with injectable hydrogels and decellularized matrices mimicking the biomechanical and biochemical milieu of native bone [,,,]. Studies by Maiti et al. and Ninu et al. demonstrated that scaffolds seeded with autologous, allogeneic, or xenogeneic MSCs accelerated healing of critically sized rabbit radial defects, with BMSCs enhancing bone formation particularly when combined with osteoconductive bioceramics such as silica-coated HA [,]. Similarly, nanohydroxyapatite/gelatin scaffolds pre-seeded with BMSCs and loaded with BMP-6 significantly enhanced in vitro osteogenesis and in vivo calvarial defect healing in rats []. Moreover, Kim et al. explored the osteogenic potential of canine ADSCs delivered via an osteogenic cell sheet seeded on a composite PCL/β-TCP scaffold in a canine radial segmental defect model which significantly outperformed undifferentiated ADSCs or scaffold-only controls in terms of new bone volume, highlighting the value of stem cells and scaffold pairing in large animal models of nonunion [].
Exosomes, a subtype of extracellular vesicles (EVs), are thought to play a large role in the regenerative properties of stem cells and can be composed of lipids, proteins, mRNA or miRNA [,]. They are able to reach target cells either through diffusion to surrounding tissue or in circulation, using specific cell-mediated interactions through ligands or endocytosis, to release their contents into cells []. Several studies have investigated extracellular vesicle-based therapies, particularly the application of exosomes, in promoting fracture healing. Qin et al. demonstrated that BMSC-derived EVs suspended in a hydrogel enhanced osteogenic gene expression and osteogenic differentiation, enabling bone regeneration in a critical-sized calvarial defect rat model []. Further, Narayanan et al. cultured human marrow-derived stromal cells (HMSCs) with pro-osteogenic exosomes and observed that these exosomes were endocytosed by HMSCs and created an upregulation of key osteogenic growth factors such as TGF-β-1 and BMP-9 []. Narayanan et al. also performed an in vivo study using a murine model that showed collagen scaffolds containing HMSCs and osteogenic miRNA-based exosomes had increased vascularization and osteogenic differentiation compared to controls []. Moreover, exosomes offer a variety of advantages: the lipid bilayer protects its contents from degradation, they are thought to retain the immune privilege nature of their parent cells and they can be stored for long periods of time without loss of potency []. These studies demonstrate how stem cell derived exosomes can induce fracture healing while avoiding challenges of other therapies.
However, despite promising preclinical data, clinical application of stem cells still raises concerns. While MSCs are typically characterized by low immunogenicity, immune rejection remains a possibility with allogeneic use []. In a systematic review of 555 patients, approximately 15% of the patients treated with allogeneic MSCs developed associated antibodies []. However, these antibodies were found to not be correlated with treatment safety or cause significant side effects in patients. More critically, the long-term persistence and differentiation behavior of MSCs remain poorly understood, with theoretical risks of uncontrolled growth or tumorigenesis. Corcoran et al. showed MSCs can facilitate the spread of cancer cells into bone marrow, increasing the metastasis of cancer []. Though MSCs are not intrinsically oncogenic, they can facilitate tumor progression under certain conditions, making rigorous safety evaluations essential prior to widespread clinical adoption [].
5.5. Integration of Gene Therapy
Gene therapy approaches involve the modification of a cell’s genetic material to induce differential expression of effector genes. In vivo and ex vivo viral vector or nonviral mediated systems have been used with varied efficacy, with ex vivo viral vector delivery serving as the gold standard at present [,,]. Local MSC-based gene therapy presents an evolving paradigm for enhancing fracture healing, particularly in complex defects or dysvascular bone environments. By genetically modifying MSCs to overexpress specific therapeutic proteins, sustained and localized bioactivity can be achieved without the need for repeated protein administration. Viral vector-mediated gene delivery has further accelerated the translational potential of MSC-based therapies for bone healing. Adenoviral (AD), lentiviral (LV), and adeno-associated viral (AAV) vectors have all been used to transduce growth factor genes such as BMP-2, BMP-6, BMP-7, and VEGF, enabling both in vitro and in vivo application to fracture and fusion models [,,,]. This strategy has been successfully performed in animal models using several cell sources, including BMSCs and ADSCs [,,,]. In an early study, Lieberman et al. showed that BMSCs transduced with BMP-2 via AD vectors healed over 90% of critical sized femoral defects in a murine model with restored mechanical properties []. Further, Liao et al. showed that the co-expression of BMP-6 and VEGF in nude rates resulted in increased bone and blood vessel growth []. Kumar et al. also demonstrated that murine MSCs transduced to express with both BMP2 and VEGF led to greater bone formation and vascularity in a segmental bone defect model []. More recently, single-cell RNA sequencing of rat femoral defects treated with ex vivo regional gene therapy versus rhBMP-2 revealed that gene therapy not only induced a more robust chondrogenic response but also created an anti-inflammatory microenvironment with reduced fibroblast-driven profibrotic signaling, offering mechanistic insight into its advantages over recombinant protein delivery []. While ex vivo transduction is popular, it can be time consuming and requires isolation of patient compatible cells []. Given this, Baltzer et al. explored the efficacy of in vivo transduction by adding AD vectors encoding BMP-2 directly to rabbit femoral defects, resulting in significantly increased radiographic healing and stronger biomechanical properties of new bone []. Additionally, to shorten turnaround time, Evans et al. demonstrated a same-day gene therapy approach for bone defect repair by transducing fat and muscle tissue containing MSCs with AD-BMP-2 intraoperatively, then reimplanting it into critical sized rat femoral defects. This approach achieved bone bridging within 10 days and full mechanical restoration by 8 weeks, importantly demonstrating the effectiveness of rapid transduction []. Virk et al. took a similar approach harvesting femoral and tibial BMSC from Lewis rats, transducing the cells ex vivo with LV-TSTA-BMP-2, then reimplanting the transduced cells into a critical-sized femoral defect all within the same day, resulting in earlier radiographic healing and higher bone volume compared to the traditional two-step transduction approach []. Bougioukli et al. took this approach further, using a “next day approach” where human- derived BMSCs were isolated and transduced overnight to be implanted into critical-sized rat femoral defects the next day; however this approach did not lead to superior healing compared to the two-step approach []. While BMP is most often used, other gene constructs, including VEGF/BMP-4 and PDGF, have also proven effective in specific bone defect models, although efficacy may be ratio- and site-dependent [,,]. Specifically, ratios of 0.2:1 and 1:1 VEGF transduced cells to BMP-4 transduced cells showed increased bone area in murine fracture models, with significantly increased bony bridging and union rates compared to a ratio of 5:1 or VEGF alone []. While many studies have looked at gene therapy for bone regeneration in rat models, Pelled et al. was one of the first to extend its application to a large animal model. Using a minipig, they demonstrated that fibrin gel cellularized with BMSCs expressing BMP-6 led to greater vertebral bone repair on microCT compared to their fibrin only control []. These investigators also evaluated various ways of delivering target genes into these stem cells without the use of viral vectors, such as with ultrasound and with nucleofection [,,]. These studies underscore the therapeutic potential of viral vector–modified MSCs in orthopedic applications as well as the complexities associated with identifying optimal delivery modes.
An important limitation of these models is the pro-inflammatory response to viral vectors, as the immune system recognizes these vectors as infectious viruses, prompting inflammation and tissue infiltration of immune cells [,,,]. For example, stem cells transduced with an AD vector to express BMP-2 can lead to an inflammatory response that hinders bone formation []. Further, immune suppression with FK506 has been shown to lead to superior bony bridging in rat femoral critical-sized defects treated with AD-BMP-2 transduced MSCs []. Despite the immunogenicity of viral vectors, ex vivo gene transfer has been used to significantly limit the in vivo exposure to viral antigens, thereby reducing the immune response to these vectors [,,]. Vakhshori et al. implanted human ADSCs transduced with LV-BMP-2 into a critical-sized defect of athymic rats, showing a majority of defects healed at 12 weeks, matching the performance of rhBMP-2 while demonstrating superior histology and biomechanics over control groups []. Moreover, there is evidence of limited biodistribution and toxicity related to LV gene therapy. For instance, Bell et al. demonstrated minimal viral copies in internal organs 84 days after implantation of human ADSCs transduced with a lentivirus to express BMP-2 into a rat femoral defect model []. In contrast to AD or AAV systems, LVs produce a more limited innate immune response. LVs have been found to express fewer viral proteins post-transduction and support effective transgene expression with less inflammatory signaling []. Comparing bone repair studies, LV-BMP2 featured more robust bone formation with months-long expression, whereas AD methods exhibited shorter expression periods with stronger host response [,,]. To that end, Feeley et al. found that in two murine bone-formation models, LV delivery sustained transgene expression for nearly 3 months versus approximately 21 days with AD systems []. Although further research is needed to understand the safety and optimal delivery of gene therapy in bone regeneration, the integration of gene therapy with stem cell therapy demonstrates great potential for the treatment of nonunions.
6. Future Directions
With the development of novel technologies in the space of bioengineering and molecular therapies, emerging techniques and approaches show early promise in furthering bone regeneration and nonunion repair. These include the use of nanotechnology and stimulus sensitive biomaterials for delivery of therapeutic compounds in a spatiotemporally controlled manner. Nanotechnologies represent a broad category of biotechnology which has been used to enhance cellular differentiation and promote bone formation both in vitro and in vivo []. Both organic and synthetic nanoparticles represent avenues for targeted drug delivery while also providing biomechanical signals for osteogenic differentiation []. These have been combined with bioorthogonal delivery systems to allow for coordinated release of therapies in a localized and controlled manner []. Demonstrating two in vivo use cases, Lee et al. and Sun et al. controlled the sequential release of rhBMP-2 through specialized HA-collagen or mesoporous BAG scaffolds in the setting of critical-sized bone defects to enhance bone healing in rat and canine models []. The continued evolution of molecular tools to directly target genes and their regulatory systems are another evolving field which may improve these interventions. These include the use of CRISPR-Cas9, CRISPR interference, and non-coding RNA systems to modulate gene therapy-based systems [,]. Finally, the integration of the aforementioned technologies to produce an optimal delivery device which enables the release of an exact milieu of key regulatory proteins remains a major goal. Park et al. leveraged several of these emerging concepts to improve bone tissue regeneration. They used CRISPR/Cas9 to overexpress BMP2 and VEGF in MSCs which were delivered in a multifunctional vitamin-D-incorporated magnesium hydroxide PLGA scaffold designed to optimally induce macrophage polarization and coordinated angio-osteogenesis with evidence of a robust healing response in vivo []. Despite early successes, given the complexity of bone healing in both the acute and chronic setting, integrated systems such as those employed by Park et al. are likely needed to produce optimal results. Further optimization of these therapies may be discovered with the use of machine learning (ML) algorithms; however, the utility of these advances have yet to be validated []. ML has also been used to identify patients at risk of nonunion using radiographic parameters, however in their current form, are unable to achieve the high levels of sensitivity or specificity required for widespread clinical adoption [,,]. Moreover, while genetic, transcriptomic, and molecular signatures related to nonunion risk have been identified, their integration into a clinically useful tool has yet to be established, thus ML models will be a key approach for their optimization [,]. Finally, further large animal research is needed for translation of the pre-clinical data discussed herein, particularly given that rodents lack true Haversian cortical bone remodeling thus are not ideal models of bone repair []. In this context, regional gene therapy and scaffold-based delivery show some promise given their efficacy in large animal models in inducing enhanced bone repair; however, further efficacy and safety data are needed prior to clinical trials [,,,].
7. Concluding Remarks
Nonunion remains a complex orthopedic challenge with no gold standard treatment. Nonunion is highly prevalent and places a significant burden on patients and the healthcare system. Surgical intervention is the primary approach to treatment; however, these commonly require multiple surgeries and continue to have non-insignificant failure rates. Emerging treatments aim to replicate the ideal environment for bone healing with growing emphasis on restoring the functional and structural integrity of bone. Stem-cell-based therapies combined with gene therapy hold a promising future as they can be integrated with scaffolds and key growth factors for bone regeneration. Nonetheless, further research is needed to develop the optimal delivery system, cell type and gene targets to find an effective solution for nonunion.
Author Contributions
Conceptualization, J.W. and J.R.L.; resources, J.T.P.; writing—original draft preparation, J.W., H.S. and S.B.; writing—review and editing, J.W. and J.R.L.; visualization, J.W., H.S. and S.B.; supervision, J.T.P. and J.R.L.; project administration, J.W. and J.R.L. 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
Not applicable.
Conflicts of Interest
The authors J.W., H.S., and S.B. declare no conflicts of interest and declare that the review was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest. J.T.P. reports royalties from BodyCad and Globus Medical, consulting fees from Stryker, Depuy Synthes, BodyCad, and Globus Medical, research support from Patient Centered Outcomes Research Institute and Orthopaedic Trauma Association, board of committee membership with AO North America and Orthopaedic Trauma Association but all have no role of interest in the work which could be construed as a potential conflict of interest, J.R.L. reports royalties from DePuy Inc and Hip Innovation Technology, and board or committee membership with Western Orthopaedic Association and The Hip Society but all have no role of interest in the work which could be construed as a potential conflict of interest.
References
- Wang, X.; Huang, J.; Hu, J.; Ma, X.; Huang, Z.; Zhu, J.; Zhu, K.; Zhang, C. Global Burden, Trends and Forecast Analysis of Extremity Fractures Based on GBD 2021. Int. J. Surg. 2025, 111, 4884–4897. [Google Scholar] [CrossRef] [PubMed]
- Feng, J.; Zhang, C.; Li, B.; Zhan, S.; Wang, S.; Song, C. Global Burden of Hip Fracture: The Global Burden of Disease Study. Osteoporos. Int. 2024, 35, 41–52. [Google Scholar] [CrossRef]
- Mills, L.A.; Aitken, S.A.; Simpson, A.H.R.W. The Risk of Non-Union per Fracture: Current Myths and Revised Figures from a Population of over 4 Million Adults. Acta Orthop. 2017, 88, 434–439. [Google Scholar] [CrossRef]
- Zura, R.; Xiong, Z.; Einhorn, T.; Watson, J.T.; Ostrum, R.F.; Prayson, M.J.; Della Rocca, G.J.; Mehta, S.; McKinley, T.; Wang, Z.; et al. Epidemiology of Fracture Nonunion in 18 Human Bones. JAMA Surg. 2016, 151, e162775. [Google Scholar] [CrossRef]
- Calori, G.M.; Mazza, E.; Colombo, M.; Ripamonti, C.; Tagliabue, L. Treatment of Long Bone Non-Unions with Polytherapy: Indications and Clinical Results. Injury 2011, 42, 587–590. [Google Scholar] [CrossRef]
- Roberts, L.; Ahmed, M.R.; Agrawal, D.K. Current Strategies in the Prevention and Management of Infection in Open Fractures. J. Orthop. Sports Med. 2025, 7, 218–229. [Google Scholar] [CrossRef]
- Kawamura, K.; Chung, K.C. Treatment of Scaphoid Fractures and Nonunions. J. Hand Surg. Am. 2008, 33, 988–997. [Google Scholar] [CrossRef]
- Jones, G.L.; McCluskey, G.M.; Curd, D.T. Nonunion of the Fractured Clavicle: Evaluation, Etiology, and Treatment. J. South. Orthop. Assoc. 2000, 9, 43–54. [Google Scholar]
- Mundi, R.; Pincus, D.; Schemitsch, E.; Ekhtiari, S.; Paterson, J.M.; Chaudhry, H.; Leis, J.A.; Redelmeier, D.A.; Ravi, B. Association Between Periprosthetic Joint Infection and Mortality Following Primary Total Hip Arthroplasty. J. Bone Jt. Surg. 2024, 106, 1546–1552. [Google Scholar] [CrossRef]
- Tian, R.; Zheng, F.; Zhao, W.; Zhang, Y.; Yuan, J.; Zhang, B.; Li, L. Prevalence and Influencing Factors of Nonunion in Patients with Tibial Fracture: Systematic Review and Meta-Analysis. J. Orthop. Surg. Res. 2020, 15, 377. [Google Scholar] [CrossRef] [PubMed]
- Jensen, S.S.; Jensen, N.M.; Gundtoft, P.H.; Kold, S.; Zura, R.; Viberg, B. Risk Factors for Nonunion Following Surgically Managed, Traumatic, Diaphyseal Fractures: A Systematic Review and Meta-Analysis. EFORT Open. Rev. 2022, 7, 516–525. [Google Scholar] [CrossRef] [PubMed]
- Dickson, K.M.D.; Katzman, S.M.D.; Delgado, E.M.D. Delayed Unions and Nonunions of Open Tibial Fractures: Correlation with Arteriography Results. Clin. Orthop. Relat. Res. 1994, 302, 189–193. [Google Scholar] [CrossRef]
- Schnetz, M.; Wengert, A.; Ruckes, C.; Jakobi, T.; Klug, A.; Gramlich, Y. Open Fractures of the Lower Leg: Outcome and Risk-Factor Analysis for Fracture-Related Infection and Nonunion in a Single Center Analysis of 187 Fractures. Injury 2025, 56, 112303. [Google Scholar] [CrossRef]
- Westgeest, J.; Weber, D.; Dulai, S.K.; Bergman, J.W.; Buckley, R.; Beaupre, L.A. Factors Associated with Development of Nonunion or Delayed Healing After an Open Long Bone Fracture: A Prospective Cohort Study of 736 Subjects. J. Orthop. Trauma. 2016, 30, 149. [Google Scholar] [CrossRef]
- Castillo, I.A.; Heiner, J.A.; Meremikwu, R.I.; Kellam, J.; Warner, S.J. Where Are We in 2022? A Summary of 11,000 Open Tibia Fractures Over 4 Decades. J. Orthop. Trauma 2023, 37, e326–e334. [Google Scholar] [CrossRef]
- Thomas, J.D.; Kehoe, J.L. Bone Nonunion. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2025. [Google Scholar]
- Ghanem, W.; Ezzeddine, H.; Saad, R.; Kiwan, E.; Dahdouh, R.; Fakih, O.; Sakhat, G.; Alam, E.; Najjar, J.; Assaf, F.; et al. State of the Nonunion: A Review of the Latest Literature. Orthop. Rev. 2025, 17, 129085. [Google Scholar] [CrossRef]
- Leow, J.M.; Clement, N.D.; Tawonsawatruk, T.; Simpson, C.J.; Simpson, A.H.R.W. The Radiographic Union Scale in Tibial (RUST) Fractures. Bone Jt. Res. 2016, 5, 116–121. [Google Scholar] [CrossRef]
- Mouliou, D.S. C-Reactive Protein: Pathophysiology, Diagnosis, False Test Results and a Novel Diagnostic Algorithm for Clinicians. Diseases 2023, 11, 132. [Google Scholar] [CrossRef]
- Solomin, L.N.; Semenistyy, A.A.; Komarov, A.V.; Khominets, V.V.; Sheridan, G.A.; Rozbruch, S.R. Universal Long Bone Nonunion Classification. Strateg. Trauma. Limb Reconstr. 2023, 18, 169–173. [Google Scholar] [CrossRef]
- Gómez-Barrena, E.; Rosset, P.; Lozano, D.; Stanovici, J.; Ermthaller, C.; Gerbhard, F. Bone Fracture Healing: Cell Therapy in Delayed Unions and Nonunions. Bone 2015, 70, 93–101. [Google Scholar] [CrossRef]
- Nicholson, J.; Makaram, N.; Simpson, A.; Keating, J. Fracture Nonunion in Long Bones: A Literature Review of Risk Factors and Surgical Management. Injury 2021, 52, S3–S11. [Google Scholar] [CrossRef] [PubMed]
- Vincken, L.; Van Der Broeck, L.; Geurts, J.; Qiu Shao, S.S.; Poeze, M.; Blokhuis, T.J. The Effect of Post-Traumatic Long Bone Non-Unions on Health-Related Quality of Life. Injury 2023, 54, 110929. [Google Scholar] [CrossRef] [PubMed]
- Schottel, P.C.; O’Connor, D.P.; Brinker, M.R. Time Trade-Off as a Measure of Health-Related Quality of Life: Long Bone Nonunions Have a Devastating Impact. J. Bone Jt. Surg. Am. 2015, 97, 1406–1410. [Google Scholar] [CrossRef]
- Wichlas, F.; Tsitsilonis, S.; Disch, A.C.; Haas, N.P.; Hartmann, C.; Graef, F.; Schwabe, P. Long-Term Functional Outcome and Quality of Life after Successful Surgical Treatment of Tibial Nonunions. Int. Orthop. 2015, 39, 521–525. [Google Scholar] [CrossRef]
- Maisenbacher, T.C.; Rollmann, M.F.; Menger, M.M.; Braun, N.R.; Braun, B.J.; Herath, S.C.; Stuby, F.; Nuessler, A.K.; Histing, T.; Reumann, M.K. Direct and Indirect Costs of Long Bone Fracture Nonunions of the Lower Limb. Bone Jt. Res. 2025, 14, 341–350. [Google Scholar] [CrossRef]
- Hak, D.J.; Fitzpatrick, D.; Bishop, J.A.; Marsh, J.L.; Tilp, S.; Schnettler, R.; Simpson, H.; Alt, V. Delayed Union and Nonunions: Epidemiology, Clinical Issues, and Financial Aspects. Injury 2014, 45, S3–S7. [Google Scholar] [CrossRef]
- Noferesti, A.; Movahedi Aliabadi, M.; Bagheri, F. Anxiety in Trauma Patients with Nonunion Diaphyseal Bone Fractures. Int. Orthop. SICOT 2025, 49, 1821–1827. [Google Scholar] [CrossRef]
- Johnson, L.; Igoe, E.; Kleftouris, G.; Papachristos, I.V.; Papakostidis, C.; Giannoudis, P.V. Physical Health and Psychological Outcomes in Adult Patients with Long-Bone Fracture Non-Unions: Evidence Today. J. Clin. Med. 2019, 8, 1998. [Google Scholar] [CrossRef]
- Brinker, M.R.; Hanus, B.D.; Sen, M.; O’Connor, D.P. The Devastating Effects of Tibial Nonunion on Health-Related Quality of Life. J. Bone Jt. Surg. Am. 2013, 95, 2170–2176. [Google Scholar] [CrossRef]
- Alt, V.; Donell, S.T.; Chhabra, A.; Bentley, A.; Eicher, A.; Schnettler, R. A Health Economic Analysis of the Use of rhBMP-2 in Gustilo–Anderson Grade III Open Tibial Fractures for the UK, Germany, and France. Injury 2009, 40, 1269–1275. [Google Scholar] [CrossRef]
- Flores, M.J.; Brown, K.E.; O’Marr, J.M.; Adejuyigbe, B.; Rodarte, P.; Gomez-Alvarado, F.; Nwachuku, K.; Urva, M.; Shearer, D. The Economic Impact of Infection and/or Nonunion on Long-Bone Shaft Fractures: A Systematic Review. OTA Int. 2024, 7, e337. [Google Scholar] [CrossRef]
- Antonova, E.; Le, T.K.; Burge, R.; Mershon, J. Tibia Shaft Fractures: Costly Burden of Nonunions. BMC Musculoskelet. Disord. 2013, 14, 42. [Google Scholar] [CrossRef]
- Iliaens, J.; Onsea, J.; Hoekstra, H.; Nijs, S.; Peetermans, W.E.; Metsemakers, W.-J. Fracture-Related Infection in Long Bone Fractures: A Comprehensive Analysis of the Economic Impact and Influence on Quality of Life. Injury 2021, 52, 3344–3349. [Google Scholar] [CrossRef]
- Sprague, S.; Bhandari, M. An Economic Evaluation of Early versus Delayed Operative Treatment in Patients with Closed Tibial Shaft Fractures. Arch. Orthop. Trauma. Surg. 2002, 122, 315–323. [Google Scholar] [CrossRef]
- Busse, J.W.; Bhandari, M.; Sprague, S.; Johnson-Masotti, A.P.; Gafni, A. An Economic Analysis of Management Strategies for Closed and Open Grade I Tibial Shaft Fractures. Acta Orthop. 2005, 76, 705–712. [Google Scholar] [CrossRef]
- Chang, C.-J.; Jou, I.-M.; Wu, T.-T.; Su, F.-C.; Tai, T.-W. Cigarette Smoke Inhalation Impairs Angiogenesis in Early Bone Healing Processes and Delays Fracture Union. Bone Jt. Res. 2020, 9, 99–107. [Google Scholar] [CrossRef]
- Roper, P.M.; Abbasnia, P.; Vuchkovska, A.; Natoli, R.M.; Callaci, J.J. Alcohol-Related Deficient Fracture Healing Is Associated With Activation of FoxO Transcription Factors in Mice. J. Orthop. Res. 2016, 34, 2106–2115. [Google Scholar] [CrossRef]
- Claes, L.; Recknagel, S.; Ignatius, A. Fracture Healing under Healthy and Inflammatory Conditions. Nat. Rev. Rheumatol. 2012, 8, 133–144. [Google Scholar] [CrossRef]
- Eby, J.M.; Sharieh, F.; Callaci, J.J. Impact of Alcohol on Bone Health, Homeostasis and Fracture Repair. Curr. Pathobiol. Rep. 2020, 8, 75–86. [Google Scholar] [CrossRef]
- Huston, P. Why Osteoarthritis of the Knee Is Called “a Wound That Does Not Heal” and Why Tai Chi Is an Effective Treatment. Front. Med. 2023, 10, 1208326. [Google Scholar] [CrossRef]
- Jiao, H.; Xiao, E.; Graves, D.T. Diabetes and Its Effect on Bone and Fracture Healing. Curr. Osteoporos. Rep. 2015, 13, 327–335. [Google Scholar] [CrossRef] [PubMed]
- Tarantino, U.; Cariati, I.; Greggi, C.; Gasbarra, E.; Belluati, A.; Ciolli, L.; Maccauro, G.; Momoli, A.; Ripanti, S.; Falez, F.; et al. Skeletal System Biology and Smoke Damage: From Basic Science to Medical Clinic. Int. J. Mol. Sci. 2021, 22, 6629. [Google Scholar] [CrossRef] [PubMed]
- Zeckey, C.; Mommsen, P.; Andruszkow, H.; Macke, C.; Frink, M.; Stübig, T.; Hüfner, T.; Krettek, C.; Hildebrand, F. The Aseptic Femoral and Tibial Shaft Non-Union in Healthy Patients–An Analysis of the Health-Related Quality of Life and the Socioeconomic Outcome. Open Orthop. J. 2011, 5, 193–197. [Google Scholar] [CrossRef]
- Knäuper, V.; López-Otin, C.; Smith, B.; Knight, G.; Murphy, G. Biochemical Characterization of Human Collagenase-3. J. Biol. Chem. 1996, 271, 1544–1550. [Google Scholar] [CrossRef] [PubMed]
- Kosaki, N.; Takaishi, H.; Kamekura, S.; Kimura, T.; Okada, Y.; Minqi, L.; Amizuka, N.; Chung, U.-I.; Nakamura, K.; Kawaguchi, H.; et al. Impaired Bone Fracture Healing in Matrix Metalloproteinase-13 Deficient Mice. Biochem. Biophys. Res. Commun. 2007, 354, 846–851. [Google Scholar] [CrossRef]
- Xiong, D.-H.; Liu, X.-G.; Guo, Y.-F.; Tan, L.-J.; Wang, L.; Sha, B.-Y.; Tang, Z.-H.; Pan, F.; Yang, T.-L.; Chen, X.-D.; et al. Genome-Wide Association and Follow-Up Replication Studies Identified ADAMTS18 and TGFBR3 as Bone Mass Candidate Genes in Different Ethnic Groups. Am. J. Hum. Genet. 2009, 84, 388–398. [Google Scholar] [CrossRef]
- Bolamperti, S.; Villa, I.; Rubinacci, A. Bone Remodeling: An Operational Process Ensuring Survival and Bone Mechanical Competence. Bone Res. 2022, 10, 48. [Google Scholar] [CrossRef]
- Sheen, J.R.; Mabrouk, A.; Garla, V.V. Fracture Healing Overview. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2025. [Google Scholar]
- Epari, D.R.; Taylor, W.R.; Heller, M.O.; Duda, G.N. Mechanical Conditions in the Initial Phase of Bone Healing. Clin. Biomech. 2006, 21, 646–655. [Google Scholar] [CrossRef]
- Wazen, R.M.; Currey, J.A.; Guo, H.; Brunski, J.B.; Helms, J.A.; Nanci, A. Micromotion-Induced Strain Fields Influence Early Stages of Repair at Bone-Implant Interfaces. Acta Biomater. 2013, 9, 6663–6674. [Google Scholar] [CrossRef]
- Liu, Z.; Wang, Q.; Zhang, J.; Qi, S.; Duan, Y.; Li, C. The Mechanotransduction Signaling Pathways in the Regulation of Osteogenesis. Int. J. Mol. Sci. 2023, 24, 14326. [Google Scholar] [CrossRef]
- Yavropoulou, M.P.; Yovos, J.G. The Molecular Basis of Bone Mechanotransduction. J. Musculoskelet. Neuronal Interact. 2016, 16, 221–236. [Google Scholar]
- Li, R.; Liang, L.; Dou, Y.; Huang, Z.; Mo, H.; Wang, Y.; Yu, B. Mechanical Strain Regulates Osteogenic and Adipogenic Differentiation of Bone Marrow Mesenchymal Stem Cells. Biomed. Res. Int. 2015, 2015, 873251. [Google Scholar] [CrossRef]
- Lafuente-Gracia, L.; Borgiani, E.; Nasello, G.; Geris, L. Towards in Silico Models of the Inflammatory Response in Bone Fracture Healing. Front. Bioeng. Biotechnol. 2021, 9, 703725. [Google Scholar] [CrossRef]
- Maruyama, M.; Rhee, C.; Utsunomiya, T.; Zhang, N.; Ueno, M.; Yao, Z.; Goodman, S.B. Modulation of the Inflammatory Response and Bone Healing. Front. Endocrinol. 2020, 11, 386. [Google Scholar] [CrossRef]
- Hu, K.; Shang, Z.; Yang, X.; Zhang, Y.; Cao, L. Macrophage Polarization and the Regulation of Bone Immunity in Bone Homeostasis. J. Inflamm. Res. 2023, 16, 3563–3580. [Google Scholar] [CrossRef]
- Chen, K.; Jiao, Y.; Liu, L.; Huang, M.; He, C.; He, W.; Hou, J.; Yang, M.; Luo, X.; Li, C. Communications Between Bone Marrow Macrophages and Bone Cells in Bone Remodeling. Front. Cell Dev. Biol. 2020, 8, 598263. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Loi, F.; Córdova, L.A.; Pajarinen, J.; Lin, T.; Yao, Z.; Goodman, S.B. Inflammation, Fracture and Bone Repair. Bone 2016, 86, 119–130. [Google Scholar] [CrossRef]
- Maruyama, M.; Moeinzadeh, S.; Guzman, R.A.; Zhang, N.; Storaci, H.W.; Utsunomiya, T.; Lui, E.; Huang, E.E.; Rhee, C.; Gao, Q.; et al. The Efficacy of Lapine Preconditioned or Genetically Modified IL4 Over-Expressing Bone Marrow-Derived Mesenchymal Stromal Cells in Corticosteroid-Associated Osteonecrosis of the Femoral Head in Rabbits. Biomaterials 2021, 275, 120972. [Google Scholar] [CrossRef] [PubMed]
- Burgan, J.; Rahmati, M.; Lee, M.; Saiz, A.M. Innate Immune Response to Bone Fracture Healing. Bone 2025, 190, 117327. [Google Scholar] [CrossRef]
- Houschyar, K.S.; Tapking, C.; Borrelli, M.R.; Popp, D.; Duscher, D.; Maan, Z.N.; Chelliah, M.P.; Li, J.; Harati, K.; Wallner, C.; et al. Wnt Pathway in Bone Repair and Regeneration–What Do We Know So Far. Front. Cell Dev. Biol. 2018, 6, 170. [Google Scholar] [CrossRef] [PubMed]
- Wu, M.; Wu, S.; Chen, W.; Li, Y.-P. The Roles and Regulatory Mechanisms of TGF-β and BMP Signaling in Bone and Cartilage Development, Homeostasis and Disease. Cell Res. 2024, 34, 101–123. [Google Scholar] [CrossRef] [PubMed]
- Zhang, X.; Guo, J.; Zhou, Y.; Wu, G. The Roles of Bone Morphogenetic Proteins and Their Signaling in the Osteogenesis of Adipose-Derived Stem Cells. Tissue Eng. Part B Rev. 2014, 20, 84–92. [Google Scholar] [CrossRef] [PubMed]
- Sanchez-Duffhues, G.; Williams, E.; Goumans, M.-J.; Heldin, C.-H.; ten Dijke, P. Bone Morphogenetic Protein Receptors: Structure, Function and Targeting by Selective Small Molecule Kinase Inhibitors. Bone 2020, 138, 115472. [Google Scholar] [CrossRef]
- Huang, W.; Yang, S.; Shao, J.; Li, Y.-P. Signaling and Transcriptional Regulation in Osteoblast Commitment and Differentiation. Front. Biosci. 2007, 12, 3068–3092. [Google Scholar] [CrossRef]
- Baniwal, S.K.; Shah, P.K.; Shi, Y.; Haduong, J.H.; DeClerck, Y.A.; Gabet, Y.; Frenkel, B. Runx2 Promotes Both Osteoblastogenesis and Novel Osteoclastogenic Signals in ST2 Mesenchymal Progenitor Cells. Osteoporos. Int. 2012, 23, 1399–1413. [Google Scholar] [CrossRef]
- Kushchayeva, Y.; Pestun, I.; Kushchayev, S.; Radzikhovska, N.; Lewiecki, E.M. Advancement in the Treatment of Osteoporosis and the Effects on Bone Healing. J. Clin. Med. 2022, 11, 7477. [Google Scholar] [CrossRef]
- Avin, K.G.; Dominguez, J.M.; Chen, N.X.; Hato, T.; Myslinski, J.J.; Gao, H.; Liu, Y.; McKinley, T.O.; Brown, K.M.; Moe, S.M.; et al. Single-Cell RNAseq Provides Insight into Altered Immune Cell Populations in Human Fracture Nonunions. J. Orthop. Res. 2023, 41, 1060–1069. [Google Scholar] [CrossRef]
- De Seny, D.; Cobraiville, G.; Leprince, P.; Fillet, M.; Collin, C.; Mathieu, M.; Hauzeur, J.-P.; Gangji, V.; Malaise, M.G. Biomarkers of Inflammation and Innate Immunity in Atrophic Nonunion Fracture. J. Transl. Med. 2016, 14, 258. [Google Scholar] [CrossRef]
- Vi, L.; Baht, G.S.; Whetstone, H.; Ng, A.; Wei, Q.; Poon, R.; Mylvaganam, S.; Grynpas, M.; Alman, B.A. Macrophages Promote Osteoblastic Differentiation In-Vivo: Implications in Fracture Repair and Bone Homeostasis. J. Bone Min. Res. 2015, 30, 1090–1102. [Google Scholar] [CrossRef]
- Schlundt, C.; El Khassawna, T.; Serra, A.; Dienelt, A.; Wendler, S.; Schell, H.; van Rooijen, N.; Radbruch, A.; Lucius, R.; Hartmann, S.; et al. Macrophages in Bone Fracture Healing: Their Essential Role in Endochondral Ossification. Bone 2018, 106, 78–89. [Google Scholar] [CrossRef]
- Spiller, K.L.; Nassiri, S.; Witherel, C.E.; Anfang, R.R.; Ng, J.; Nakazawa, K.R.; Yu, T.; Vunjak-Novakovic, G. Sequential Delivery of Immunomodulatory Cytokines to Facilitate the M1-to-M2 Transition of Macrophages and Enhance Vascularization of Bone Scaffolds. Biomaterials 2015, 37, 194–207. [Google Scholar] [CrossRef] [PubMed]
- Flierl, M.A.; Perl, M.; Rittirsch, D.; Bartl, C.; Schreiber, H.; Fleig, V.; Schlaf, G.; Liener, U.; Brueckner, U.B.; Gebhard, F.; et al. The Role of C5a in the Innate Immune Response after Experimental Blunt Chest Trauma. Shock 2008, 29, 25–31. [Google Scholar] [CrossRef] [PubMed]
- Recknagel, S.; Bindl, R.; Kurz, J.; Wehner, T.; Ehrnthaller, C.; Knöferl, M.W.; Gebhard, F.; Huber-Lang, M.; Claes, L.; Ignatius, A. Experimental Blunt Chest Trauma Impairs Fracture Healing in Rats. J. Orthop. Res. 2011, 29, 734–739. [Google Scholar] [CrossRef] [PubMed]
- Recknagel, S.; Bindl, R.; Brochhausen, C.; Göckelmann, M.; Wehner, T.; Schoengraf, P.; Huber-Lang, M.; Claes, L.; Ignatius, A. Systemic Inflammation Induced by a Thoracic Trauma Alters the Cellular Composition of the Early Fracture Callus. J. Trauma. Acute Care Surg. 2013, 74, 531–537. [Google Scholar] [CrossRef]
- Schell, H.; Thompson, M.S.; Bail, H.J.; Hoffmann, J.-E.; Schill, A.; Duda, G.N.; Lienau, J. Mechanical Induction of Critically Delayed Bone Healing in Sheep: Radiological and Biomechanical Results. J. Biomech. 2008, 41, 3066–3072. [Google Scholar] [CrossRef]
- Schmidt-Bleek, K.; Schell, H.; Schulz, N.; Hoff, P.; Perka, C.; Buttgereit, F.; Volk, H.-D.; Lienau, J.; Duda, G.N. Inflammatory Phase of Bone Healing Initiates the Regenerative Healing Cascade. Cell Tissue Res. 2012, 347, 567–573. [Google Scholar] [CrossRef]
- Reed, A.A.C.; Joyner, C.J.; Brownlow, H.C.; Simpson, A.H.R.W. Human Atrophic Fracture Non-unions Are Not Avascular. J. Orthop. Res. 2002, 20, 593–599. [Google Scholar] [CrossRef]
- Fajardo, M.; Liu, C.-J.; Egol, K. Levels of Expression for BMP-7 and Several BMP Antagonists May Play an Integral Role in a Fracture Nonunion: A Pilot Study. Clin. Orthop. Relat. Res. 2009, 467, 3071–3078. [Google Scholar] [CrossRef]
- Kloen, P.; Loots, G.G.; Hamdy, R.C.; Smit, T.H. Bridging the Gap: Compressing Non-Unions for Proper Cellular Signaling. Med. Hypotheses 2022, 160, 110794. [Google Scholar] [CrossRef]
- Kwong, F.N.K.; Hoyland, J.A.; Freemont, A.J.; Evans, C.H. Altered Relative Expression of BMPs and BMP Inhibitors in Cartilaginous Areas of Human Fractures Progressing towards Nonunion. J. Orthop. Res. 2009, 27, 752–757. [Google Scholar] [CrossRef] [PubMed]
- Fajardo, M.; Liu, C.-J.; Ilalov, K.; Egol, K.A. Matrix Metalloproteinases That Associate with and Cleave Bone Morphogenetic Protein-2 In Vitro Are Elevated in Hypertrophic Fracture Nonunion Tissue. J. Orthop. Trauma 2010, 24, 557–563. [Google Scholar] [CrossRef] [PubMed]
- Evans, C.H. Advances in Regenerative Orthopedics. Mayo Clin. Proc. 2013, 88, 1323–1339. [Google Scholar] [CrossRef] [PubMed]
- Khan, W.S.; Rayan, F.; Dhinsa, B.S.; Marsh, D. An Osteoconductive, Osteoinductive, and Osteogenic Tissue-Engineered Product for Trauma and Orthopaedic Surgery: How Far Are We? Stem Cells Int. 2012, 2012, 236231. [Google Scholar] [CrossRef]
- 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]
- Patterson, J.; Lieberman, J. Segmental defect repair and nonunion. In Orthobiologics: Scientific and Clinical Solutions for Orthopaedic Surgeons; Lippincott Williams & Wilkins: Philadelphia, PA, USA, 2024. [Google Scholar]
- Mazzotta, A.; Stagni, C.; Rocchi, M.; Rani, N.; Del Piccolo, N.; Filardo, G.; Dallari, D. Bone Marrow Aspirate Concentrate/Platelet-Rich Fibrin Augmentation Accelerates Healing of Aseptic Upper Limb Nonunions. J. Orthop. Traumatol. 2021, 22, 21. [Google Scholar] [CrossRef]
- Laubach, M.; Suresh, S.; Herath, B.; Wille, M.-L.; Delbrück, H.; Alabdulrahman, H.; Hutmacher, D.W.; Hildebrand, F. Clinical Translation of a Patient-Specific Scaffold-Guided Bone Regeneration Concept in Four Cases with Large Long Bone Defects. J. Orthop. Transl. 2022, 34, 73–84. [Google Scholar] [CrossRef]
- Xie, C.; Wang, C.; Huang, Y.; Li, Q.; Tian, X.; Huang, W.; Yin, D. Therapeutic Effect of Autologous Bone Grafting with Adjuvant Bone Morphogenetic Protein on Long Bone Nonunion: A Systematic Review and Meta-Analysis. J. Orthop. Surg. Res. 2022, 17, 298. [Google Scholar] [CrossRef]
- Choi, W.; Kim, B.-S.; Cho, W.-T.; Lim, E.J.; Choi, J.S.; Ryu, Y.K.; Cho, J.-W.; Sakong, S.; Oh, J.-K. Efficacy and Safety of Recombinant Human Bone Morphogenetic Protein-2 (rhBMP-2) Combined with Autologous Bone for the Treatment of Long Bone Nonunion: A Report of a Prospective Case Series. Injury 2024, 55, 111711. [Google Scholar] [CrossRef]
- Moyal, A.J.; Li, A.W.; Adelstein, J.M.; Moon, T.J.; Napora, J.K. Bone Marrow Aspirate and Bone Marrow Aspirate Concentrate: Does the Literature Support Use in Long-Bone Nonunion and Provide New Insights into Mechanism of Action? Eur. J. Orthop. Surg. Traumatol. 2024, 34, 2871–2880. [Google Scholar] [CrossRef]
- Zhu, J.; Han, D.; Sun, Y.; Zhao, C. Platelet-Rich Plasma in the Treatment of Delayed Union and Nonunion Fractures: An Umbrella Meta-Analysis. J. Orthop. Sci. 2024, 30, 894–904. [Google Scholar] [CrossRef]
- Tanavalee, C.; Ngarmukos, S.; Amarase, C.; Tantavisut, S.; Jaruthien, N.; Tanavalee, A. A Randomized Controlled Trial of Teriparatide for Accelerating Bone Union and Improving Clinical Outcomes in Patients with Pertrochanteric Fracture Fixation. Sci. Rep. 2025, 15, 19465. [Google Scholar] [CrossRef] [PubMed]
- Pelled, G.; Sheyn, D.; Tawackoli, W.; Jun, D.S.; Koh, Y.; Su, S.; Cohn Yakubovich, D.; Kallai, I.; Antebi, B.; Da, X.; et al. BMP6-Engineered MSCs Induce Vertebral Bone Repair in a Pig Model: A Pilot Study. Stem Cells Int. 2016, 2016, 6530624. [Google Scholar] [CrossRef] [PubMed]
- Brunello, G.; Panda, S.; Schiavon, L.; Sivolella, S.; Biasetto, L.; Del Fabbro, M. The Impact of Bioceramic Scaffolds on Bone Regeneration in Preclinical In Vivo Studies: A Systematic Review. Materials 2020, 13, 1500. [Google Scholar] [CrossRef] [PubMed]
- Liu, T.; Fang, W.; Wu, G.; Li, Y.; Pathak, J.L.; Liu, Y. Low Dose BMP2-Doped Calcium Phosphate Graft Promotes Bone Defect Healing in a Large Animal Model. Front. Cell Dev. Biol. 2020, 8, 613891. [Google Scholar] [CrossRef]
- DeBaun, M.R.; Salazar, B.P.; Bai, Y.; Gardner, M.J.; Yang, Y.P.; Pan, C.C.; Stahl, A.M.; Moeinzadeh, S.; Kim, S.; Lui, E.; et al. A Bioactive Synthetic Membrane Improves Bone Healing in a Preclinical Nonunion Model. Injury 2022, 53, 1368–1374. [Google Scholar] [CrossRef]
- De La Vega, R.E.; van Griensven, M.; Zhang, W.; Coenen, M.J.; Nagelli, C.V.; Panos, J.A.; Peniche Silva, C.J.; Geiger, J.; Plank, C.; Evans, C.H.; et al. Efficient Healing of Large Osseous Segmental Defects Using Optimized Chemically Modified Messenger RNA Encoding BMP-2. Sci. Adv. 2022, 8, eabl6242. [Google Scholar] [CrossRef]
- da Rocha, L.R.; Dias, R.B.; Fernandes, M.B.C.; Prinz, R.; Eirado, T.P.; de Souza Costa, I.; Monteiro, M.J.; da Silva, C.E.R.; Dos Santos, C.T.; Fogagnolo, F. A New Option for Bone Regeneration: A Rapid Methodology for Cellularization of Allograft with Human Bone Marrow Stromal Cells with in Vivo Bone-Forming Potential. Injury 2023, 54 (Suppl. 6), 110777. [Google Scholar] [CrossRef]
- Garot, C.; Schoffit, S.; Monfoulet, C.; Machillot, P.; Deroy, C.; Roques, S.; Vial, J.; Vollaire, J.; Renard, M.; Ghanem, H.; et al. 3D-Printed Osteoinductive Polymeric Scaffolds with Optimized Architecture to Repair a Sheep Metatarsal Critical-Size Bone Defect. Adv. Healthc. Mater. 2023, 12, e2301692. [Google Scholar] [CrossRef]
- Sun, L.; Niu, H.; Wu, Y.; Dong, S.; Li, X.; Kim, B.Y.S.; Liu, C.; Ma, Y.; Jiang, W.; Yuan, Y. Bio-Integrated Scaffold Facilitates Large Bone Regeneration Dominated by Endochondral Ossification. Bioact. Mater. 2024, 35, 208–227. [Google Scholar] [CrossRef]
- Li, X.; Asuncion, F.; Ominsky, M.; Niu, Q.-T.; Akesson, K.E.; Wang, J.; Lieberman, J.; Ke, H.Z. High-Dose Romosozumab Promoted Bone Regeneration of Critical-Size Ulnar Defect Filled with Demineralized Bone Matrix in Nonhuman Primates. J. Orthop. Transl. 2025, 54, 1–7. [Google Scholar] [CrossRef]
- Aleem, I.S.; Aleem, I.; Evaniew, N.; Busse, J.W.; Yaszemski, M.; Agarwal, A.; Einhorn, T.; Bhandari, M. Efficacy of Electrical Stimulators for Bone Healing: A Meta-Analysis of Randomized Sham-Controlled Trials. Sci. Rep. 2016, 6, 31724. [Google Scholar] [CrossRef] [PubMed]
- Griffin, X.L.; Costa, M.L.; Parsons, N.; Smith, N. Electromagnetic Field Stimulation for Treating Delayed Union or Non-Union of Long Bone Fractures in Adults. Cochrane Database Syst. Rev. 2011, 4, CD008471. [Google Scholar] [CrossRef] [PubMed]
- Leighton, R.; Watson, J.T.; Giannoudis, P.; Papakostidis, C.; Harrison, A.; Steen, R.G. Healing of Fracture Nonunions Treated with Low-Intensity Pulsed Ultrasound (LIPUS): A Systematic Review and Meta-Analysis. Injury 2017, 48, 1339–1347. [Google Scholar] [CrossRef] [PubMed]
- Leighton, R.; Phillips, M.; Bhandari, M.; Zura, R. Low Intensity Pulsed Ultrasound (LIPUS) Use for the Management of Instrumented, Infected, and Fragility Non-Unions: A Systematic Review and Meta-Analysis of Healing Proportions. BMC Musculoskelet Disord 2021, 22, 532. [Google Scholar] [CrossRef]
- TRUST Investigators Writing Group; Busse, J.W.; Bhandari, M.; Einhorn, T.A.; Schemitsch, E.; Heckman, J.D.; Tornetta, P.; Leung, K.-S.; Heels-Ansdell, D.; Makosso-Kallyth, S.; et al. Re-Evaluation of Low Intensity Pulsed Ultrasound in Treatment of Tibial Fractures (TRUST): Randomized Clinical Trial. BMJ 2016, 355, i5351. [Google Scholar] [CrossRef]
- White, N.J.; Patterson, E.D.; Dhaliwal, G.S.; Hildebrand, K.A. WECAN Low-Intensity Pulsed Ultrasound Versus Sham in the Treatment of Operatively Managed Scaphoid Nonunions: The SNAPU Randomized Controlled Trial. J. Bone Jt. Surg. 2024, 106, 1573–1582. [Google Scholar] [CrossRef]
- Mittal, K.K.; Gupta, H.; Kaushik, N. Reunion of Post Nail Aseptic Non-Union of Diaphyseal Femoral Fractures by Augmentation Plating, Decortication and Bone Grafting–Replacement for Exchange Nailing. Injury 2021, 52, 1529–1533. [Google Scholar] [CrossRef]
- Bégué, T.; Mouchantaf, M.; Aurégan, J.-C. Aseptic Humeral Shaft Nonunion. Orthop. Traumatol. Surg. Res. 2023, 109, 103462. [Google Scholar] [CrossRef]
- Ayalon, O.; Rettig, S.A.; Tedesco, L.J. Bone Graft and Fixation Options in the Surgical Management of Scaphoid Nonunion. J. Am. Acad. Orthop. Surg. 2024, 33, e989–e999. [Google Scholar] [CrossRef]
- Alford, A.I.; Nicolaou, D.; Hake, M.; McBride-Gagyi, S. Masquelet’s Induced Membrane Technique: Review of Current Concepts and Future Directions. J. Orthop. Res. 2021, 39, 707–718. [Google Scholar] [CrossRef] [PubMed]
- Hoit, G.; Kain, M.S.; Sparkman, J.W.; Norris, B.L.; Conway, J.D.; Watson, J.T.; Tornetta, P.; Nauth, A. The Induced Membrane Technique for Bone Defects: Basic Science, Clinical Evidence, and Technical Tips. OTA Int. 2021, 4, e106(1–5). [Google Scholar] [CrossRef] [PubMed]
- Wu, J.-H.; Bao, Q.-W.; Wang, S.-K.; Zhou, P.-Y.; Xu, S.-G. Mechanisms of the Masquelet Technique to Promote Bone Defect Repair and Its Influencing Factors. Chin. J. Traumatol. 2025, 28, 157–163. [Google Scholar] [CrossRef] [PubMed]
- Gaillard, J.; Masquelet, A.-C.; Boutroux, P.; Cambon-Binder, A. Induced-Membrane Treatment of Refractory Humeral Non-Union with or without Bone Defect. Orthop. Traumatol. Surg. Res. 2020, 106, 803–811. [Google Scholar] [CrossRef]
- Chloros, G.D.; Kanakaris, N.K.; Harwood, P.J.; Giannoudis, P.V. Induced Membrane Technique for Acute Bone Loss and Nonunion Management of the Tibia. OTA Int. 2022, 5, e170. [Google Scholar] [CrossRef]
- Griffin, J.T.; Landy, D.C.; Sneed, C.R.; Liu, B.; Kavolus, M.; Pectol, R.W.; Gitajn, I.L.; Oh, J.-K.; Aneja, A. Masquelet Technique for the Tibia: A Systematic Review and Meta-Analysis of Contemporary Outcomes. J. Orthop. Trauma 2023, 37, e36–e44. [Google Scholar] [CrossRef]
- Ma, X.-Y.; Liu, B.; Yu, H.-L.; Zhang, X.; Xiang, L.-B.; Zhou, D.-P. Induced Membrane Technique for the Treatment of Infected Forearm Nonunion: A Retrospective Study. J. Hand Surg. 2022, 47, 583.e1–583.e9. [Google Scholar] [CrossRef]
- Moreillon, P.; Que, Y.A.; Bayer, A.S. Pathogenesis of Streptococcal and Staphylococcal Endocarditis. Infect. Dis. Clin. North. Am. 2002, 16, 297–318. [Google Scholar] [CrossRef]
- Singh, S.; Toci, G.R.; Kapadia, K.; Colon, A.; Greenberg, P.; Iyer, H.; Katt, B.; Shah, A. Vascularized Bone Grafting Versus the 2-Stage Masquelet Technique for Upper-Extremity Bone Reconstruction: A Meta-Analysis. J. Hand Surg. Am. 2023, 48, 984–992. [Google Scholar] [CrossRef]
- Rouse, B.J.; Sheridan, G.A.; Page, B.J.; Fragomen, A.T.; Rozbruch, S.R. Hypertrophic Nonunion Management with Distraction Osteogenesis: A Scoping Review of the Literature. OTA Int. 2024, 7, e342. [Google Scholar] [CrossRef]
- Fragomen, A.T.; Wellman, D.; Rozbruch, S.R. The PRECICE Magnetic IM Compression Nail for Long Bone Nonunions: A Preliminary Report. Arch. Orthop. Trauma. Surg. 2019, 139, 1551–1560. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Gómez-Barrena, E.; Ehrnthaller, C. Long Bone Uninfected Non-Union: Grafting Techniques. EFORT Open Rev. 2024, 9, 329–338. [Google Scholar] [CrossRef] [PubMed]
- Migliorini, F.; La Padula, G.; Torsiello, E.; Spiezia, F.; Oliva, F.; Maffulli, N. Strategies for Large Bone Defect Reconstruction after Trauma, Infections or Tumour Excision: A Comprehensive Review of the Literature. Eur. J. Med. Res. 2021, 26, 118. [Google Scholar] [CrossRef]
- 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]
- Bray, C.C.; Walker, C.M.; Spence, D.D. Orthobiologics in Pediatric Sports Medicine. Orthop. Clin. North. Am. 2017, 48, 333–342. [Google Scholar] [CrossRef]
- Chahla, J.; Mannava, S.; Cinque, M.E.; Geeslin, A.G.; Codina, D.; LaPrade, R.F. Bone Marrow Aspirate Concentrate Harvesting and Processing Technique. Arthrosc. Tech. 2017, 6, e441–e445. [Google Scholar] [CrossRef]
- Holton, J.; Imam, M.; Ward, J.; Snow, M. The Basic Science of Bone Marrow Aspirate Concentrate in Chondral Injuries. Orthop. Rev. 2016, 8, 6659. [Google Scholar] [CrossRef]
- Kim, G.B.; Seo, M.-S.; Park, W.T.; Lee, G.W. Bone Marrow Aspirate Concentrate: Its Uses in Osteoarthritis. Int. J. Mol. Sci. 2020, 21, 3224. [Google Scholar] [CrossRef]
- Lin, K.; VandenBerg, J.; Putnam, S.M.; Parks, C.D.; Spraggs-Hughes, A.; McAndrew, C.M.; Ricci, W.M.; Gardner, M.J. Bone Marrow Aspirate Concentrate with Cancellous Allograft versus Iliac Crest Bone Graft in the Treatment of Long Bone Nonunions. OTA Int. 2019, 2, e012. [Google Scholar] [CrossRef]
- Van Vugt, T.A.G.; Geurts, J.A.P.; Blokhuis, T.J. Treatment of Infected Tibial Non-Unions Using a BMAC and S53P4 BAG Combination for Reconstruction of Segmental Bone Defects: A Clinical Case Series. Injury 2021, 52, S67–S71. [Google Scholar] [CrossRef] [PubMed]
- Brozovich, A.; Sinicrope, B.J.; Bauza, G.; Niclot, F.B.; Lintner, D.; Taraballi, F.; McCulloch, P.C. High Variability of Mesenchymal Stem Cells Obtained via Bone Marrow Aspirate Concentrate Compared with Traditional Bone Marrow Aspiration Technique. Orthop. J. Sports Med. 2021, 9, 23259671211058459. [Google Scholar] [CrossRef] [PubMed]
- Cavallo, C.; Boffa, A.; de Girolamo, L.; Merli, G.; Kon, E.; Cattini, L.; Santo, E.; Grigolo, B.; Filardo, G. Bone Marrow Aspirate Concentrate Quality Is Affected by Age and Harvest Site. Knee Surg. Sports Traumatol. Arthrosc. 2023, 31, 2140–2151. [Google Scholar] [CrossRef] [PubMed]
- Rodriguez, I.A.; Growney Kalaf, E.A.; Bowlin, G.L.; Sell, S.A. Platelet-Rich Plasma in Bone Regeneration: Engineering the Delivery for Improved Clinical Efficacy. Biomed. Res. Int. 2014, 2014, 392398. [Google Scholar] [CrossRef]
- Wang, S.; Chen, J.; Wu, J.; Peng, Z.; Wang, K.; Zhang, X.; Wu, F.; Jie, K. Based on the Diamond Concept, Application of Platelet-Rich Plasma in the Treatment of Aseptic Femoral Shaft Nonunion: A Retrospective Controlled Study on 66 Patients. Injury 2025, 56, 112325. [Google Scholar] [CrossRef]
- Oh, J.H.; Kim, W.; Park, K.U.; Roh, Y.H. Comparison of the Cellular Composition and Cytokine-Release Kinetics of Various Platelet-Rich Plasma Preparations. Am. J. Sports Med. 2015, 43, 3062–3070. [Google Scholar] [CrossRef]
- Sugaya, H.; Yoshioka, T.; Kato, T.; Taniguchi, Y.; Kumagai, H.; Hyodo, K.; Ohneda, O.; Yamazaki, M.; Mishima, H. Comparative Analysis of Cellular and Growth Factor Composition in Bone Marrow Aspirate Concentrate and Platelet-Rich Plasma. Bone Marrow Res. 2018, 2018, 1549826. [Google Scholar] [CrossRef]
- Sundman, E.A.; Cole, B.J.; Fortier, L.A. Growth Factor and Catabolic Cytokine Concentrations Are Influenced by the Cellular Composition of Platelet-Rich Plasma. Am. J. Sports Med. 2011, 39, 2135–2140. [Google Scholar] [CrossRef]
- Jing, N.; Hou, Y.-C.; Zhang, J.-C.; Xu, G.; Lei, M.; Tang, X.; Chen, W.; Ni, H.; Zhang, F. Cracking the Code: Understanding ESWT’s Role in Bone Fracture Healing. J. Orthop. Transl. 2025, 50, 403–412. [Google Scholar] [CrossRef]
- Phillips, M.R.; Harrison, A.; Mehta, S.; Nolte, P.A.; Bhandari, M.; Zura, R. A Scoping Review of Operative and Non-Invasive Management in the Treatment of Non-Unions. Injury 2022, 53, 3872–3878. [Google Scholar] [CrossRef]
- Gillman, C.E.; Jayasuriya, A.C. FDA-Approved Bone Grafts and Bone Graft Substitute Devices in Bone Regeneration. Mater. Sci. Eng. C Mater. Biol. Appl. 2021, 130, 112466. [Google Scholar] [CrossRef] [PubMed]
- Gómez-Barrena, E.; Rosset, P.; Gebhard, F.; Hernigou, P.; Baldini, N.; Rouard, H.; Sensebé, L.; Gonzalo-Daganzo, R.M.; Giordano, R.; Padilla-Eguiluz, N.; et al. Feasibility and Safety of Treating Non-Unions in Tibia, Femur and Humerus with Autologous, Expanded, Bone Marrow-Derived Mesenchymal Stromal Cells Associated with Biphasic Calcium Phosphate Biomaterials in a Multicentric, Non-Comparative Trial. Biomaterials 2019, 196, 100–108. [Google Scholar] [CrossRef] [PubMed]
- Nashi, N.; Kagda, F.H. Current Concepts of Bone Grafting in Trauma Surgery. J. Clin. Orthop. Trauma. 2023, 43, 102231. [Google Scholar] [CrossRef] [PubMed]
- Dave, U.; Rubin, J.; Shah, H.; Gerhold, C.; McCormick, J.R.; Bi, A.S.; Yuh, C.; Rossi, L.A.; Chahla, J. Bone Marrow Aspirate Concentrate (BMAC) Harvested in the Axial and Appendicular Skeleton Does Not Differ in Progenitor Cell Count: A Systematic Review and Meta-Analysis. J. Orthop. 2025, 63, 216–223. [Google Scholar] [CrossRef]
- Impieri, L.; Pezzi, A.; Hadad, H.; Peretti, G.M.; Mangiavini, L.; Rossi, N. Orthobiologics in Delayed Union and Non-Union of Adult Long Bones Fractures: A Systematic Review. Bone Rep. 2024, 21, 101760. [Google Scholar] [CrossRef]
- Lieberman, J.R.; Daluiski, A.; Stevenson, S.; Jolla, L.; Wu, L.; McALLISTER, P.; Lee, Y.P.; Kabo, J.M.; Finerman, G.A.M.; 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. [Google Scholar] [CrossRef]
- Paulini, M.; Camal Ruggieri, I.N.; Ramallo, M.; Alonso, M.; Rodriguez-Cabello, J.C.; Esbrit, P.; Mardegan Issa, J.P.; Feldman, S. Recombinant Proteins-Based Strategies in Bone Tissue Engineering. Biomolecules 2021, 12, 3. [Google Scholar] [CrossRef]
- Miri, Z.; Haugen, H.J.; Loca, D.; Rossi, F.; Perale, G.; Moghanian, A.; Ma, Q. Review on the Strategies to Improve the Mechanical Strength of Highly Porous Bone Bioceramic Scaffolds. J. Eur. Ceram. Soc. 2024, 44, 23–42. [Google Scholar] [CrossRef]
- Percival, K.M.; Paul, V.; Husseini, G.A. Recent Advancements in Bone Tissue Engineering: Integrating Smart Scaffold Technologies and Bio-Responsive Systems for Enhanced Regeneration. Int. J. Mol. Sci. 2024, 25, 6012. [Google Scholar] [CrossRef]
- Rodriguez-Merchan, C.; Gomez-Castresana, F. Internal Fixation of Nonunions. Clin. Orthop. Relat. Res. 2004, 419, 13–20. [Google Scholar] [CrossRef]
- Schulze, F.; Lang, A.; Schoon, J.; Wassilew, G.I.; Reichert, J. Scaffold Guided Bone Regeneration for the Treatment of Large Segmental Defects in Long Bones. Biomedicines 2023, 11, 325. [Google Scholar] [CrossRef] [PubMed]
- Farjaminejad, S.; Farjaminejad, R.; Hasani, M.; Garcia-Godoy, F.; Abdouss, M.; Marya, A.; Harsoputranto, A.; Jamilian, A. Advances and Challenges in Polymer-Based Scaffolds for Bone Tissue Engineering: A Path Towards Personalized Regenerative Medicine. Polymers 2024, 16, 3303. [Google Scholar] [CrossRef] [PubMed]
- Saurav, S.; Sharma, P.; Kumar, A.; Tabassum, Z.; Girdhar, M.; Mamidi, N.; Mohan, A. Harnessing Natural Polymers for Nano-Scaffolds in Bone Tissue Engineering: A Comprehensive Overview of Bone Disease Treatment. CIMB 2024, 46, 585–611. [Google Scholar] [CrossRef] [PubMed]
- Wang, X.; Zeng, J.; Gan, D.; Ling, K.; He, M.; Li, J.; Lu, Y. Recent Strategies and Advances in Hydrogel-Based Delivery Platforms for Bone Regeneration. Nano-Micro Lett. 2024, 17, 73. [Google Scholar] [CrossRef]
- Cui, C.; Lin, F.; Xia, L.; Zhang, X. Mesenchymal Stem Cells Therapy for the Treatment of Non-Union Fractures: A Systematic Review and Meta-Analysis. BMC Musculoskelet Disord 2025, 26, 245. [Google Scholar] [CrossRef]
- Lalu, M.M.; McIntyre, L.; Pugliese, C.; Fergusson, D.; Winston, B.W.; Marshall, J.C.; Granton, J.; Stewart, D.J. Canadian Critical Care Trials Group Safety of Cell Therapy with Mesenchymal Stromal Cells (SafeCell): A Systematic Review and Meta-Analysis of Clinical Trials. PLoS ONE 2012, 7, e47559. [Google Scholar] [CrossRef]
- Lee, H.-Y.; Hong, I.-S. Double-Edged Sword of Mesenchymal Stem Cells: Cancer-Promoting versus Therapeutic Potential. Cancer Sci. 2017, 108, 1939–1946. [Google Scholar] [CrossRef]
- Thurairajah, K.; Briggs, G.D.; Balogh, Z.J. Stem Cell Therapy for Fracture Non-Union: The Current Evidence from Human Studies. J. Orthop. Surg. 2021, 29, 23094990211036545. [Google Scholar] [CrossRef]
- Arjomandnejad, M.; Dasgupta, I.; Flotte, T.R.; Keeler, A.M. Immunogenicity of Recombinant Adeno-Associated Virus (AAV) Vectors for Gene Transfer. BioDrugs 2023, 37, 311–329. [Google Scholar] [CrossRef]
- Chu, X.; Xiong, Y.; Lu, L.; Wang, Y.; Wang, J.; Zeng, R.; Hu, L.; Yan, C.; Zhao, Z.; Lin, S.; et al. Research Progress of Gene Therapy Combined with Tissue Engineering to Promote Bone Regeneration. APL Bioeng. 2024, 8, 031502. [Google Scholar] [CrossRef]
- Cornetta, K.; Lin, T.-Y.; Pellin, D.; Kohn, D.B. Meeting FDA Guidance Recommendations for Replication-Competent Virus and Insertional Oncogenesis Testing. Mol. Ther. Methods Clin. Dev. 2023, 28, 28–39. [Google Scholar] [CrossRef]
- Gallo, M.C.; Elias, A.; Reynolds, J.; Ball, J.R.; Lieberman, J.R. Regional Gene Therapy for Bone Tissue Engineering: A Current Concepts Review. Bioengineering 2025, 12, 120. [Google Scholar] [CrossRef]
- De Pace, R.; Molinari, S.; Mazzoni, E.; Perale, G. Bone Regeneration: A Review of Current Treatment Strategies. JCM 2025, 14, 1838. [Google Scholar] [CrossRef]
- Kirankumar, S.; Gurusamy, N.; Rajasingh, S.; Sigamani, V.; Vasanthan, J.; Perales, S.G.; Rajasingh, J. Modern Approaches on Stem Cells and Scaffolding Technology for Osteogenic Differentiation and Regeneration. Exp. Biol. Med. 2022, 247, 433–445. [Google Scholar] [CrossRef]
- Wang, Z.; Ren, L.; Li, Z.; Qiu, Q.; Wang, H.; Huang, X.; Ma, D. Impact of Different Cell Types on the Osteogenic Differentiation Process of Mesenchymal Stem Cells. Stem Cells Int. 2025, 2025, 5551222. [Google Scholar] [CrossRef]
- Zhao, T.; Chu, Z.; Ma, J.; Ouyang, L. Immunomodulation Effect of Biomaterials on Bone Formation. J. Funct. Biomater. 2022, 13, 103. [Google Scholar] [CrossRef] [PubMed]
- Wang, P.; Berry, D.; Moran, A.; He, F.; Tam, T.; Chen, L.; Chen, S. Controlled Growth Factor Release in 3D-Printed Hydrogels. Adv. Heal Mater. 2020, 9, e1900977. [Google Scholar] [CrossRef]
- Shibuya, M. Vascular Endothelial Growth Factor (VEGF) and Its Receptor (VEGFR) Signaling in Angiogenesis. Genes. Cancer 2011, 2, 1097–1105. [Google Scholar] [CrossRef] [PubMed]
- Andrae, J.; Gallini, R.; Betsholtz, C. Role of Platelet-Derived Growth Factors in Physiology and Medicine. Genes. Dev. 2008, 22, 1276–1312. [Google Scholar] [CrossRef] [PubMed]
- Cohn-Schwartz, D.; Schary, Y.; Yalon, E.; Krut, Z.; Da, X.; Schwarz, E.M.; Gazit, D.; Pelled, G.; Gazit, Z. PTH-Induced Bone Regeneration and Vascular Modulation Are Both Dependent on Endothelial Signaling. Cells 2022, 11, 897. [Google Scholar] [CrossRef]
- Menger, M.M.; Tobias, A.L.; Bauer, D.; Bleimehl, M.; Scheuer, C.; Menger, M.D.; Histing, T.; Laschke, M.W. Parathyroid Hormone Stimulates Bone Regeneration in an Atrophic Non-Union Model in Aged Mice. J. Transl. Med. 2023, 21, 844. [Google Scholar] [CrossRef]
- Shin, J.-O.; Lee, J.-B.; Lee, S.; Kim, J.-W. Enhancing Bone Regeneration and Osseointegration Using rhPTH(1-34) and Dimeric R25CPTH(1-34) in an Osteoporotic Beagle Model. Elife 2024, 13, RP93830. [Google Scholar] [CrossRef]
- Zhang, S.; Ding, L.; Chen, G.; Zhang, J.; Ge, W.; Qu, Y. Enhanced Bone Regeneration via Local Low-Dose Delivery of PTH1-34 in a Composite Hydrogel. Front. Bioeng. Biotechnol. 2023, 11, 1209752. [Google Scholar] [CrossRef] [PubMed]
- Zou, Z.; Wang, L.; Zhou, Z.; Sun, Q.; Liu, D.; Chen, Y.; Hu, H.; Cai, Y.; Lin, S.; Yu, Z.; et al. Simultaneous Incorporation of PTH(1-34) and Nano-Hydroxyapatite into Chitosan/Alginate Hydrogels for Efficient Bone Regeneration. Bioact. Mater. 2021, 6, 1839–1851. [Google Scholar] [CrossRef] [PubMed]
- Ishack, S.; Mediero, A.; Wilder, T.; Ricci, J.L.; Cronstein, B.N. Bone Regeneration in Critical Bone Defects Using Three-Dimensionally Printed β-Tricalcium Phosphate/Hydroxyapatite Scaffolds Is Enhanced by Coating Scaffolds with Either Dipyridamole or BMP-2. J. Biomed. Mater. Res. B Appl. Biomater. 2017, 105, 366–375. [Google Scholar] [CrossRef] [PubMed]
- Mediero, A.; Wilder, T.; Perez-Aso, M.; Cronstein, B.N. Direct or Indirect Stimulation of Adenosine A2A Receptors Enhances Bone Regeneration as Well as Bone Morphogenetic Protein-2. FASEB J. 2015, 29, 1577–1590. [Google Scholar] [CrossRef]
- Witek, L.; Alifarag, A.M.; Tovar, N.; Lopez, C.D.; Cronstein, B.N.; Rodriguez, E.D.; Coelho, P.G. Repair of Critical-Sized Long Bone Defects Using Dipyridamole-Augmented 3D-Printed Bioactive Ceramic Scaffolds. J. Orthop. Res. 2019, 37, 2499–2507. [Google Scholar] [CrossRef]
- Marie, P.J.; Miraoui, H.; Sévère, N. FGF/FGFR Signaling in Bone Formation: Progress and Perspectives. Growth Factors 2012, 30, 117–123. [Google Scholar] [CrossRef]
- Karnes, J.M.; Daffner, S.D.; Watkins, C.M. Multiple Roles of Tumor Necrosis Factor-Alpha in Fracture Healing. Bone 2015, 78, 87–93. [Google Scholar] [CrossRef]
- Bolander, J.; Ji, W.; Leijten, J.; Teixeira, L.M.; Bloemen, V.; Lambrechts, D.; Chaklader, M.; Luyten, F.P. Healing of a Large Long-Bone Defect through Serum-Free In Vitro Priming of Human Periosteum-Derived Cells. Stem Cell Rep. 2017, 8, 758–772. [Google Scholar] [CrossRef]
- Hettiaratchi, M.H.; Krishnan, L.; Rouse, T.; Chou, C.; McDevitt, T.C.; Guldberg, R.E. Heparin-Mediated Delivery of Bone Morphogenetic Protein-2 Improves Spatial Localization of Bone Regeneration. Sci. Adv. 2020, 6, eaay1240. [Google Scholar] [CrossRef]
- Krishnan, L.; Priddy, L.B.; Esancy, C.; Li, M.-T.A.; Stevens, H.Y.; Jiang, X.; Tran, L.; Rowe, D.W.; Guldberg, R.E. Hydrogel-Based Delivery of rhBMP-2 Improves Healing of Large Bone Defects Compared with Autograft. Clin. Orthop. Relat. Res. 2015, 473, 2885–2897. [Google Scholar] [CrossRef] [PubMed]
- Minier, K.; Touré, A.; Fusellier, M.; Fellah, B.; Bouvy, B.; Weiss, P.; Gauthier, O. BMP-2 Delivered from a Self-Crosslinkable CaP/Hydrogel Construct Promotes Bone Regeneration in a Critical-Size Segmental Defect Model of Non-Union in Dogs. Vet. Comp. Orthop. Traumatol. 2014, 27, 411–421. [Google Scholar] [CrossRef]
- Yasuda, H.; Yano, K.; Wakitani, S.; Matsumoto, T.; Nakamura, H.; Takaoka, K. Repair of Critical Long Bone Defects Using Frozen Bone Allografts Coated with an rhBMP-2-Retaining Paste. J. Orthop. Sci. 2012, 17, 299–307. [Google Scholar] [CrossRef] [PubMed]
- 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]
- 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]
- Zara, J.N.; Siu, R.K.; Zhang, X.; Shen, J.; Ngo, R.; Lee, M.; Li, W.; Chiang, M.; Chung, J.; Kwak, J.; et al. High Doses of Bone Morphogenetic Protein 2 Induce Structurally Abnormal Bone and Inflammation in Vivo. Tissue Eng. Part A 2011, 17, 1389–1399. [Google Scholar] [CrossRef]
- 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]
- 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]
- Carragee, E.J.; Chu, G.; Rohatgi, R.; Hurwitz, E.L.; Weiner, B.K.; Yoon, S.T.; Comer, G.; Kopjar, B. Cancer Risk after Use of Recombinant Bone Morphogenetic Protein-2 for Spinal Arthrodesis. J. Bone Jt. Surg. Am. 2013, 95, 1537–1545. [Google Scholar] [CrossRef]
- Kim, S.E.; Song, S.-H.; Yun, Y.P.; Choi, B.-J.; Kwon, I.K.; Bae, M.S.; Moon, H.-J.; Kwon, Y.-D. The Effect of Immobilization of Heparin and Bone Morphogenic Protein-2 (BMP-2) to Titanium Surfaces on Inflammation and Osteoblast Function. Biomaterials 2011, 32, 366–373. [Google Scholar] [CrossRef] [PubMed]
- Quang Le, B.; Chun Tan, T.; Lee, S.-B.; Woong Jang, J.; Sik Kim, Y.; Soo Lee, J.; Won Choi, J.; Sathiyanathan, P.; Nurcombe, V.; Cool, S.M. A Biomimetic Collagen-Bone Granule-Heparan Sulfate Combination Scaffold for BMP2 Delivery. Gene 2021, 769, 145217. [Google Scholar] [CrossRef] [PubMed]
- Kim, T.-H.; Yun, Y.-P.; Park, Y.-E.; Lee, S.-H.; Yong, W.; Kundu, J.; Jung, J.W.; Shim, J.-H.; Cho, D.-W.; Kim, S.E.; et al. In Vitro and in Vivo Evaluation of Bone Formation Using Solid Freeform Fabrication-Based Bone Morphogenic Protein-2 Releasing PCL/PLGA Scaffolds. Biomed. Mater. 2014, 9, 025008. [Google Scholar] [CrossRef] [PubMed]
- Briquez, P.S.; Tsai, H.-M.; Watkins, E.A.; Hubbell, J.A. Engineered Bridge Protein with Dual Affinity for Bone Morphogenetic Protein-2 and Collagen Enhances Bone Regeneration for Spinal Fusion. Sci. Adv. 2021, 7, eabh4302. [Google Scholar] [CrossRef]
- Bosemark, P.; Isaksson, H.; McDonald, M.M.; Little, D.G.; Tägil, M. Augmentation of Autologous Bone Graft by a Combination of Bone Morphogenic Protein and Bisphosphonate Increased Both Callus Volume and Strength. Acta Orthop. 2013, 84, 106–111. [Google Scholar] [CrossRef]
- Bosemark, P.; Isaksson, H.; Tägil, M. Influence of Systemic Bisphosphonate Treatment on Mechanical Properties of BMP-Induced Calluses in a Rat Fracture Model: Comparison of Three-Point Bending and Twisting Test. J. Orthop. Res. 2014, 32, 721–726. [Google Scholar] [CrossRef]
- Kaipel, M.; Schützenberger, S.; Schultz, A.; Ferguson, J.; Slezak, P.; Morton, T.J.; Van Griensven, M.; Redl, H. BMP-2 but Not VEGF or PDGF in Fibrin Matrix Supports Bone Healing in a Delayed-union Rat Model. J. Orthop. Res. 2012, 30, 1563–1569. [Google Scholar] [CrossRef]
- Ihn, H.; Kang, H.; Iglesias, B. Regional Gene Therapy with Transduced Human Cells: The Influence of “Cell Dose” on Bone Repair. Tissue Eng. Part A 2021, 27, 1422–1433. [Google Scholar] [CrossRef]
- Panos, J.A.; Coenen, M.J.; Nagelli, C.V.; McGlinch, E.B.; Atasoy-Zeybek, A.; De Padilla, C.L.; Coghlan, R.F.; Johnstone, B.; Ferreira, E.; Porter, R.M.; et al. IL-1Ra Gene Transfer Potentiates BMP2-Mediated Bone Healing by Redirecting Osteogenesis toward Endochondral Ossification. Mol. Ther. 2023, 31, 420–434. [Google Scholar] [CrossRef]
- Hu, K.; Olsen, B.R. The Roles of Vascular Endothelial Growth Factor in Bone Repair and Regeneration. Bone 2016, 91, 30–38. [Google Scholar] [CrossRef]
- He, S.; Fang, J.; Zhong, C.; Ren, F.; Wang, M. Controlled pVEGF Delivery via a Gene-Activated Matrix Comprised of a Peptide-Modified Non-Viral Vector and a Nanofibrous Scaffold for Skin Wound Healing. Acta Biomater. 2022, 140, 149–162. [Google Scholar] [CrossRef]
- Orth, M.; Fritz, T.; Stutz, J.; Scheuer, C.; Ganse, B.; Bullinger, Y.; Lee, J.S.; Murphy, W.L.; Laschke, M.W.; Menger, M.D.; et al. Local Application of Mineral-Coated Microparticles Loaded with VEGF and BMP-2 Induces the Healing of Murine Atrophic Non-Unions. Front. Bioeng. Biotechnol. 2022, 9, 809397. [Google Scholar] [CrossRef]
- Novak, S.; Madunic, J.; Shum, L.; Vucetic, M.; Wang, X.; Tanigawa, H.; Ghosh, M.; Sanjay, A.; Kalajzic, I. PDGF Inhibits BMP2-Induced Bone Healing. NPJ Regen. Med. 2023, 8, 3. [Google Scholar] [CrossRef]
- 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] [PubMed]
- DiGiovanni, C.W.; Lin, S.S.; Baumhauer, J.F.; Daniels, T.; Younger, A.; Glazebrook, M.; Anderson, J.; Anderson, R.; Evangelista, P.; Lynch, S.E.; et al. Recombinant Human Platelet-Derived Growth Factor-BB and Beta-Tricalcium Phosphate (rhPDGF-BB/β-TCP): An Alternative to Autogenous Bone Graft. J. Bone Jt. Surg. Am. 2013, 95, 1184–1192. [Google Scholar] [CrossRef] [PubMed]
- Hollinger, J.O.; Onikepe, A.O.; MacKrell, J.; Einhorn, T.; Bradica, G.; Lynch, S.; Hart, C.E. Accelerated Fracture Healing in the Geriatric, Osteoporotic Rat with Recombinant Human Platelet-derived Growth Factor-bb and an Injectable Beta-tricalcium Phosphate/Collagen Matrix. J. Orthop. Res. 2008, 26, 83–90. [Google Scholar] [CrossRef] [PubMed]
- Nayak, V.V.; Costello, J.P.; Ehlen, Q.T.; Slavin, B.V.; Mirsky, N.A.; Kelly, S.; Suarez, C.; Daunert, S.; Witek, L.; Coelho, P.G. A rhPDGF-BB/Bovine Type I Collagen/β-TCP Mixture for the Treatment of Critically Sized Non-union Tibial Defects: An in Vivo Study in Rabbits. J. Orthop. Res. 2024, 42, 1998–2006. [Google Scholar] [CrossRef]
- Kitcharanant, N.; Chattipakorn, N.; Chattipakorn, S.C. The Effect of Intermittent Parathyroid Hormone on Bone Lengthening: Current Evidence to Inform Future Effective Interventions. Osteoporos. Int. 2023, 34, 1657–1675. [Google Scholar] [CrossRef]
- Andreassen, T.T.; Ejersted, C.; Oxlund, H. Intermittent Parathyroid Hormone (1-34) Treatment Increases Callus Formation and Mechanical Strength of Healing Rat Fractures. J. Bone Min. Res. 1999, 14, 960–968. [Google Scholar] [CrossRef]
- Cipriano, C.A.; Issack, P.S.; Shindle, L.; Werner, C.M.L.; Helfet, D.L.; Lane, J.M. Recent Advances Toward the Clinical Application of PTH (1-34) in Fracture Healing. HSS J. 2009, 5, 149–153. [Google Scholar] [CrossRef]
- Holzer, G.; Majeska, R.J.; Lundy, M.W.; Hartke, J.R.; Einhorn, T.A. Parathyroid Hormone Enhances Fracture Healing. A Preliminary Report. Clin. Orthop. Relat. Res. 1999, 366, 258–263. [Google Scholar] [CrossRef] [PubMed]
- Kumabe, Y.; Lee, S.Y.; Waki, T.; Iwakura, T.; Takahara, S.; Arakura, M.; Kuroiwa, Y.; Fukui, T.; Matsumoto, T.; Matsushita, T.; et al. Triweekly Administration of Parathyroid Hormone (1–34) Accelerates Bone Healing in a Rat Refractory Fracture Model. BMC Musculoskelet. Disord. 2017, 18, 545. [Google Scholar] [CrossRef] [PubMed]
- Hong, H.; Song, T.; Liu, Y.; Li, J.; Jiang, Q.; Song, Q.; Deng, Z. The Effectiveness and Safety of Parathyroid Hormone in Fracture Healing: A Meta-Analysis. Clinics 2019, 74, e800. [Google Scholar] [CrossRef] [PubMed]
- Eastman, K.; Gerlach, M.; Piec, I.; Greeves, J.; Fraser, W. Effectiveness of Parathyroid Hormone (PTH) Analogues on Fracture Healing: A Meta-Analysis. Osteoporos. Int. 2021, 32, 1531–1546. [Google Scholar] [CrossRef]
- Cameron, K.O.; Lefker, B.A.; Ke, H.Z.; Li, M.; Zawistoski, M.P.; Tjoa, C.M.; Wright, A.S.; DeNinno, S.L.; Paralkar, V.M.; Owen, T.A.; et al. Discovery of CP-533536: An EP2 Receptor Selective Prostaglandin E2 (PGE2) Agonist That Induces Local Bone Formation. Bioorganic Med. Chem. Lett. 2009, 19, 2075–2078. [Google Scholar] [CrossRef]
- Graham, S.; Gamie, Z.; Polyzois, I.; Narvani, A.A.; Tzafetta, K.; Tsiridis, E.; Helioti, M.; Mantalaris, A.; Tsiridis, E. Prostaglandin EP2 and EP4 Receptor Agonists in Bone Formation and Bone Healing: In Vivo and in Vitro Evidence. Expert. Opin. Investig. Drugs 2009, 18, 746–766. [Google Scholar] [CrossRef]
- Li, M.; Thompson, D.D.; Paralkar, V.M. Prostaglandin E2 Receptors in Bone Formation. Int. Orthop. 2007, 31, 767–772. [Google Scholar] [CrossRef]
- Kamolratanakul, P.; Hayata, T.; Ezura, Y.; Kawamata, A.; Hayashi, C.; Yamamoto, Y.; Hemmi, H.; Nagao, M.; Hanyu, R.; Notomi, T.; et al. Nanogel-Based Scaffold Delivery of Prostaglandin E(2) Receptor-Specific Agonist in Combination with a Low Dose of Growth Factor Heals Critical-Size Bone Defects in Mice. Arthritis Rheum. 2011, 63, 1021–1033. [Google Scholar] [CrossRef]
- Rundle, C.H.; Strong, D.D.; Chen, S.-T.; Linkhart, T.A.; Sheng, M.H.-C.; Wergedal, J.E.; Lau, K.-H.W.; Baylink, D.J. Retroviral-Based Gene Therapy with Cyclooxygenase-2 Promotes the Union of Bony Callus Tissues and Accelerates Fracture Healing in the Rat. J. Gene Med. 2008, 10, 229–241. [Google Scholar] [CrossRef]
- Abedi, N.; Sadeghian, A.; Kouhi, M.; Haugen, H.J.; Savabi, O.; Nejatidanesh, F. Immunomodulation in Bone Tissue Engineering: Recent Advancements in Scaffold Design and Biological Modifications for Enhanced Regeneration. ACS Biomater. Sci. Eng. 2025, 11, 1269–1290. [Google Scholar] [CrossRef]
- Xuan, Y.; Li, L.; Zhang, C.; Zhang, M.; Cao, J.; Zhang, Z. The 3D-Printed Ordered Bredigite Scaffold Promotes Pro-Healing of Critical-Sized Bone Defects by Regulating Macrophage Polarization. Int. J. Nanomed. 2023, 18, 917–932. [Google Scholar] [CrossRef]
- Yunna, C.; Mengru, H.; Lei, W.; Weidong, C. Macrophage M1/M2 Polarization. Eur. J. Pharmacol. 2020, 877, 173090. [Google Scholar] [CrossRef] [PubMed]
- Mahon, O.R.; Browe, D.C.; Gonzalez-Fernandez, T.; Pitacco, P.; Whelan, I.T.; Von Euw, S.; Hobbs, C.; Nicolosi, V.; Cunningham, K.T.; Mills, K.H.G.; et al. Nano-Particle Mediated M2 Macrophage Polarization Enhances Bone Formation and MSC Osteogenesis in an IL-10 Dependent Manner. Biomaterials 2020, 239, 119833. [Google Scholar] [CrossRef] [PubMed]
- Xiong, S.; Zhang, Y.; Zeng, J.; Zhou, J.; Liu, S.; Wei, P.; Liu, H.; Yi, F.; Wan, Z.; Xiong, L.; et al. DLP Fabrication of HA Scaffold with Customized Porous Structures to Regulate Immune Microenvironment and Macrophage Polarization for Enhancing Bone Regeneration. Mater. Today Bio 2024, 24, 100929. [Google Scholar] [CrossRef] [PubMed]
- Li, L.; Li, J.; Guo, J.; Zhang, H.; Zhang, X.; Yin, C.; Wang, L.; Zhu, Y.; Yao, Q. 3D Molecularly Functionalized Cell-Free Biomimetic Scaffolds for Osteochondral Regeneration. Adv. Funct. Mater. 2019, 29, 1807356. [Google Scholar] [CrossRef]
- Bougioukli, S.; Jain, A.; Sugiyama, O.; Tinsley, B.A.; Tang, A.H.; Tan, M.H.; Adams, D.J.; Kostenuik, P.J.; Lieberman, J.R. Combination Therapy with BMP-2 and a Systemic RANKL Inhibitor Enhances Bone Healing in a Mouse Critical-Sized Femoral Defect. Bone 2016, 84, 93–103. [Google Scholar] [CrossRef]
- Ratanavaraporn, J.; Furuya, H.; Tabata, Y. Local Suppression of Pro-Inflammatory Cytokines and the Effects in BMP-2-Induced Bone Regeneration. Biomaterials 2012, 33, 304–316. [Google Scholar] [CrossRef]
- Hannink, G.; Arts, J.J.C. Bioresorbability, Porosity and Mechanical Strength of Bone Substitutes: What Is Optimal for Bone Regeneration? Injury 2011, 42 (Suppl. 2), S22–S25. [Google Scholar] [CrossRef]
- Bose, S.; Roy, M.; Bandyopadhyay, A. Recent Advances in Bone Tissue Engineering Scaffolds. Trends Biotechnol. 2012, 30, 546–554. [Google Scholar] [CrossRef]
- Eliaz, N.; Metoki, N. Calcium Phosphate Bioceramics: A Review of Their History, Structure, Properties, Coating Technologies and Biomedical Applications. Materials 2017, 10, 334. [Google Scholar] [CrossRef]
- Chinnasami, H.; Dey, M.K.; Devireddy, R. Three-Dimensional Scaffolds for Bone Tissue Engineering. Bioengineering 2023, 10, 759. [Google Scholar] [CrossRef]
- Farazin, A.; Darghiasi, S.F. Advanced Polymeric Scaffolds for Bone Tissue Regeneration. Explor. BioMat-X 2025, 2, 101340. [Google Scholar] [CrossRef]
- Filippi, M.; Born, G.; Chaaban, M.; Scherberich, A. Natural Polymeric Scaffolds in Bone Regeneration. Front. Bioeng. Biotechnol. 2020, 8, 474. [Google Scholar] [CrossRef]
- Da Cunha, M.R.; Maia, F.L.M.; Iatecola, A.; Massimino, L.C.; Plepis, A.M.d.G.; Martins, V.d.C.A.; Da Rocha, D.N.; Mariano, E.D.; Hirata, M.C.; Ferreira, J.R.M.; et al. In Vivo Evaluation of Collagen and Chitosan Scaffold, Associated or Not with Stem Cells, in Bone Repair. J. Funct. Biomater. 2023, 14, 357. [Google Scholar] [CrossRef]
- Fu, F.; Zhu, X.; Qin, Z.; Wang, J.-J.; Xu, C.; Wang, L.-N.; Tu, Y.; Zhang, S.; Li, R.-X.; Li, X.-H.; et al. Differential Degradation Rate and Underlying Mechanism of a Collagen/Chitosan Complex in Subcutis, Spinal Cord and Brain Tissues of Rat. J. Mater. Sci: Mater. Med. 2018, 29, 35. [Google Scholar] [CrossRef] [PubMed]
- Azaman, F.A.; Brennan Fournet, M.E.; Sheikh Ab Hamid, S.; Zawawi, M.S.F.; Da Silva Junior, V.A.; Devine, D.M. Enhancement of Scaffold In Vivo Biodegradability for Bone Regeneration Using P28 Peptide Formulations. Pharmaceuticals 2023, 16, 876. [Google Scholar] [CrossRef] [PubMed]
- Tan, Y.; Huang, H.; Ayers, D.C.; Song, J. Modulating Viscoelasticity, Stiffness, and Degradation of Synthetic Cellular Niches via Stoichiometric Tuning of Covalent versus Dynamic Noncovalent Cross-Linking. ACS Cent. Sci. 2018, 4, 971–981. [Google Scholar] [CrossRef]
- Foudazi, R.; Zowada, R.; Manas-Zloczower, I.; Feke, D.L. Porous Hydrogels: Present Challenges and Future Opportunities. Langmuir 2023, 39, 2092–2111. [Google Scholar] [CrossRef] [PubMed]
- Robinson, M.; Douglas, S.; Michelle Willerth, S. Mechanically Stable Fibrin Scaffolds Promote Viability and Induce Neurite Outgrowth in Neural Aggregates Derived from Human Induced Pluripotent Stem Cells. Sci. Rep. 2017, 7, 6250. [Google Scholar] [CrossRef]
- Sun, S.; Cui, Y.; Yuan, B.; Dou, M.; Wang, G.; Xu, H.; Wang, J.; Yin, W.; Wu, D.; Peng, C. Drug Delivery Systems Based on Polyethylene Glycol Hydrogels for Enhanced Bone Regeneration. Front. Bioeng. Biotechnol. 2023, 11, 1117647. [Google Scholar] [CrossRef]
- Zhang, X.; Li, Y.; Ma, Z.; He, D.; Li, H. Modulating Degradation of Sodium Alginate/Bioglass Hydrogel for Improving Tissue Infiltration and Promoting Wound Healing. Bioact. Mater. 2021, 6, 3692–3704. [Google Scholar] [CrossRef] [PubMed]
- Amini, A.R.; Adams, D.J.; Laurencin, C.T.; Nukavarapu, S.P. Optimally Porous and Biomechanically Compatible Scaffolds for Large-Area Bone Regeneration. Tissue Eng. Part A 2012, 18, 1376–1388. [Google Scholar] [CrossRef] [PubMed]
- Guda, T.; Walker, J.A.; Singleton, B.M.; Hernandez, J.W.; Son, J.-S.; Kim, S.-G.; Oh, D.S.; Appleford, M.R.; Ong, J.L.; Wenke, J.C. Guided Bone Regeneration in Long-Bone Defects with a Structural Hydroxyapatite Graft and Collagen Membrane. Tissue Eng. Part A 2013, 19, 1879–1888. [Google Scholar] [CrossRef] [PubMed]
- Valtanen, R.S.; Yang, Y.P.; Gurtner, G.C.; Maloney, W.J.; Lowenberg, D.W. Synthetic and Bone Tissue Engineering Graft Substitutes: What Is the Future? Injury 2021, 52, S72–S77. [Google Scholar] [CrossRef]
- Cabrera Pereira, A.; Tovar, N.; Nayak, V.V.; Mijares, D.Q.; Smay, J.E.; Torroni, A.; Flores, R.L.; Witek, L. Direct Inkjet Writing Type 1 Bovine Collagen/Β-tricalcium Phosphate Scaffolds for Bone Regeneration. J. Biomed. Mater. Res. 2024, 112, e35347. [Google Scholar] [CrossRef]
- Tovar, N.; Witek, L.; Atria, P.; Sobieraj, M.; Bowers, M.; Lopez, C.D.; Cronstein, B.N.; Coelho, P.G. Form and Functional Repair of Long Bone Using 3D-printed Bioactive Scaffolds. J. Tissue Eng. Regen. Med. 2018, 12, 1986–1999. [Google Scholar] [CrossRef]
- Zhang, T.; Huang, L.; Zhang, X.; Guo, J.; Shen, Y.; Luo, Y. 3D Printed Bioceramic Scaffolds with Fully Interconnected Channel Networks for Enhanced Vascularized Bone Regeneration. Biomater. Sci. 2025, 13, 4830–4845. [Google Scholar] [CrossRef]
- Wang, X.; Nie, Z.; Chang, J.; Lu, M.L.; Kang, Y. Multiple Channels with Interconnected Pores in a Bioceramic Scaffold Promote Bone Tissue Formation. Sci. Rep. 2021, 11, 20447. [Google Scholar] [CrossRef]
- Lu, Q.; Diao, J.; Wang, Y.; Feng, J.; Zeng, F.; Yang, Y.; Kuang, Y.; Zhao, N.; Wang, Y. 3D Printed Pore Morphology Mediates Bone Marrow Stem Cell Behaviors via RhoA/ROCK2 Signaling Pathway for Accelerating Bone Regeneration. Bioact. Mater. 2023, 26, 413–424. [Google Scholar] [CrossRef]
- Joo, S.; Gwon, Y.; Kim, S.; Park, S.; Kim, J.; Hong, S. Piezoelectrically and Topographically Engineered Scaffolds for Accelerating Bone Regeneration. ACS Appl. Mater. Interfaces 2024, 16, 1999–2011. [Google Scholar] [CrossRef]
- Dai, X.; Yao, X.; Zhang, W.; Cui, H.; Ren, Y.; Deng, J.; Zhang, X. The Osteogenic Role of Barium Titanate/Polylactic Acid Piezoelectric Composite Membranes as Guiding Membranes for Bone Tissue Regeneration. Int. J. Nanomed. 2022, 17, 4339–4353. [Google Scholar] [CrossRef] [PubMed]
- Tian, L.; Yong, S.; Jiao, Z.; Zhang, W.; Xi, Y.; Huang, D.; Bie, X.; Li, C.; Shi, G.; Zhao, Y.; et al. Piezoelectric Composites with BaTiO3 NPs as Guiding Membranes: Reconstructing the Bioelectric Microenvironment for Enhanced Bone Regeneration. J. Mater. Chem. C 2025, 13, 11850–11868. [Google Scholar] [CrossRef]
- Fernandez De Grado, G.; Keller, L.; Idoux-Gillet, Y.; Wagner, Q.; Musset, A.-M.; Benkirane-Jessel, N.; Bornert, F.; Offner, D. Bone Substitutes: A Review of Their Characteristics, Clinical Use, and Perspectives for Large Bone Defects Management. J. Tissue Eng. 2018, 9, 2041731418776819. [Google Scholar] [CrossRef] [PubMed]
- Kim, M.S.; Kim, G. Three-Dimensional Electrospun Polycaprolactone (PCL)/Alginate Hybrid Composite Scaffolds. Carbohydr. Polym. 2014, 114, 213–221. [Google Scholar] [CrossRef]
- Leng, Y.; Yang, F.; Wang, Q.; Li, Z.; Yuan, B.; Peng, C.; Ren, G.; Wang, Z.; Cui, Y.; Wang, Y.; et al. Material-Based Therapy for Bone Nonunion. Mater. Des. 2019, 183, 108161. [Google Scholar] [CrossRef]
- Sapudom, J.; Kongsema, M.; Methachittipan, A.; Damrongsakkul, S.; Kanokpanont, S.; Teo, J.C.M.; Khongkow, M.; Tonsomboon, K.; Thongnuek, P. Degradation Products of Crosslinked Silk Fibroin Scaffolds Modulate the Immune Response but Not Cell Toxicity. J. Mater. Chem. B 2023, 11, 3607–3616. [Google Scholar] [CrossRef]
- Godbout, C.; Ryan, G.; Ramnaraign, D.J.; Hegner, C.; Desjardins, S.; Gagnon, S.; Bates, B.D.; Whatley, I.; Schemitsch, E.H.; Nauth, A. Optimal Delivery of Endothelial Progenitor Cells in a Rat Model of Critical-size Bone Defects. J. Orthop. Res. 2024, 42, 193–201. [Google Scholar] [CrossRef]
- Liao, H.; Zhong, Z.; Liu, Z.; Li, L.; Ling, Z.; Zou, X. Bone Mesenchymal Stem Cells Co-Expressing VEGF and BMP-6 Genes to Combat Avascular Necrosis of the Femoral Head. Exp. Ther. Med. 2018, 15, 954–962. [Google Scholar] [CrossRef]
- Pek, Y.S.; Gao, S.; Arshad, M.S.M.; Leck, K.-J.; Ying, J.Y. Porous Collagen-Apatite Nanocomposite Foams as Bone Regeneration Scaffolds. Biomaterials 2008, 29, 4300–4305. [Google Scholar] [CrossRef]
- Schwartz, L.; Ganta, A.; Konda, S.; Leucht, P.; Rivero, S.; Egol, K.A. Tibial Plateau Fracture Surgical Care Using Standardized Protocols Over Time: A Single Center’s Longitudinal View. J. Orthop. Trauma. 2023, 37, 627–632. [Google Scholar] [CrossRef]
- Bouhadir, K.H.; Lee, K.Y.; Alsberg, E.; Damm, K.L.; Anderson, K.W.; Mooney, D.J. Degradation of Partially Oxidized Alginate and Its Potential Application for Tissue Engineering. Biotechnol. Prog. 2001, 17, 945–950. [Google Scholar] [CrossRef]
- Ivanovska, J.; Zehnder, T.; Lennert, P.; Sarker, B.; Boccaccini, A.R.; Hartmann, A.; Schneider-Stock, R.; Detsch, R. Biofabrication of 3D Alginate-Based Hydrogel for Cancer Research: Comparison of Cell Spreading, Viability, and Adhesion Characteristics of Colorectal HCT116 Tumor Cells. Tissue Eng. Part C Methods 2016, 22, 708–715. [Google Scholar] [CrossRef] [PubMed]
- Boerckel, J.D.; Kolambkar, Y.M.; Dupont, K.M.; Uhrig, B.A.; Phelps, E.A.; Stevens, H.Y.; García, A.J.; Guldberg, R.E. Effects of Protein Dose and Delivery System on BMP-Mediated Bone Regeneration. Biomaterials 2011, 32, 5241–5251. [Google Scholar] [CrossRef] [PubMed]
- Priddy, L.B.; Chaudhuri, O.; Stevens, H.Y.; Krishnan, L.; Uhrig, B.A.; Willett, N.J.; Guldberg, R.E. Oxidized Alginate Hydrogels for Bone Morphogenetic Protein-2 Delivery in Long Bone Defects. Acta Biomater. 2014, 10, 4390–4399. [Google Scholar] [CrossRef] [PubMed]
- Meinel, L.; Kaplan, D.L. Silk Constructs for Delivery of Musculoskeletal Therapeutics. Adv. Drug Deliv. Rev. 2012, 64, 1111–1122. [Google Scholar] [CrossRef]
- Stuckensen, K.; Schwab, A.; Knauer, M.; Muiños-López, E.; Ehlicke, F.; Reboredo, J.; Granero-Moltó, F.; Gbureck, U.; Prósper, F.; Walles, H.; et al. Tissue Mimicry in Morphology and Composition Promotes Hierarchical Matrix Remodeling of Invading Stem Cells in Osteochondral and Meniscus Scaffolds. Adv. Mater. 2018, 30, 1706754. [Google Scholar] [CrossRef]
- Deininger, C.; Wagner, A.; Heimel, P.; Salzer, E.; Vila, X.M.; Weißenbacher, N.; Grillari, J.; Redl, H.; Wichlas, F.; Freude, T.; et al. Enhanced BMP-2-Mediated Bone Repair Using an Anisotropic Silk Fibroin Scaffold Coated with Bone-like Apatite. Int. J. Mol. Sci. 2021, 23, 283. [Google Scholar] [CrossRef]
- Feng, Y.; Guo, W.; Hu, L.; Yi, X.; Tang, F. Application of Hydrogels as Sustained-Release Drug Carriers in Bone Defect Repair. Polymers 2022, 14, 4906. [Google Scholar] [CrossRef]
- Huang, H.; Qi, X.; Chen, Y.; Wu, Z. Thermo-Sensitive Hydrogels for Delivering Biotherapeutic Molecules: A Review. Saudi Pharm. J. 2019, 27, 990–999. [Google Scholar] [CrossRef]
- Li, R.; Liang, J.; He, Y.; Qin, J.; He, H.; Lee, S.; Pang, Z.; Wang, J. Sustained Release of Immunosuppressant by Nanoparticle-Anchoring Hydrogel Scaffold Improved the Survival of Transplanted Stem Cells and Tissue Regeneration. Theranostics 2018, 8, 878–893. [Google Scholar] [CrossRef]
- Fan, Z.; Cheng, P.; Wang, D.; Zhao, Y.; Wang, Z.; Han, J. Design and Investigation of Salecan/Chitosan Hydrogel Formulations with Improved Antibacterial Performance and 3D Cell Culture Function. J. Biomater. Sci. Polym. Ed. 2020, 31, 2268–2284. [Google Scholar] [CrossRef]
- Whitehead, J.; Griffin, K.H.; Gionet-Gonzales, M.; Vorwald, C.E.; Cinque, S.E.; Leach, J.K. Hydrogel Mechanics Are a Key Driver of Bone Formation by Mesenchymal Stromal Cell Spheroids. Biomaterials 2021, 269, 120607. [Google Scholar] [CrossRef] [PubMed]
- Wu, D.T.; Jeffreys, N.; Diba, M.; Mooney, D.J. Viscoelastic Biomaterials for Tissue Regeneration. Tissue Eng. Part C Methods 2022, 28, 289–300. [Google Scholar] [CrossRef] [PubMed]
- Li, J.; Wang, W.; Li, M.; Song, P.; Lei, H.; Gui, X.; Zhou, C.; Liu, L. Biomimetic Methacrylated Gelatin Hydrogel Loaded With Bone Marrow Mesenchymal Stem Cells for Bone Tissue Regeneration. Front. Bioeng. Biotechnol. 2021, 9, 770049. [Google Scholar] [CrossRef] [PubMed]
- Wagner, J.M.; Conze, N.; Lewik, G.; Wallner, C.; Brune, J.C.; Dittfeld, S.; Jaurich, H.; Becerikli, M.; Dadras, M.; Harati, K.; et al. Bone Allografts Combined with Adipose-Derived Stem Cells in an Optimized Cell/Volume Ratio Showed Enhanced Osteogenesis and Angiogenesis in a Murine Femur Defect Model. J. Mol. Med. 2019, 97, 1439–1450. [Google Scholar] [CrossRef]
- Chatterjee, K.; Lin-Gibson, S.; Wallace, W.E.; Parekh, S.H.; Lee, Y.J.; Cicerone, M.T.; Young, M.F.; Simon, C.G. The Effect of 3D Hydrogel Scaffold Modulus on Osteoblast Differentiation and Mineralization Revealed by Combinatorial Screening. Biomaterials 2010, 31, 5051–5062. [Google Scholar] [CrossRef]
- McBeth, C.; Lauer, J.; Ottersbach, M.; Campbell, J.; Sharon, A.; Sauer-Budge, A.F. 3D Bioprinting of GelMA Scaffolds Triggers Mineral Deposition by Primary Human Osteoblasts. Biofabrication 2017, 9, 015009. [Google Scholar] [CrossRef]
- Yu, F.T.H.; Armstrong, J.K.; Tripette, J.; Meiselman, H.J.; Cloutier, G. A Local Increase in Red Blood Cell Aggregation Can Trigger Deep Vein Thrombosis: Evidence Based on Quantitative Cellular Ultrasound Imaging. J. Thromb. Haemost. 2011, 9, 481–488. [Google Scholar] [CrossRef]
- Yue, K.; Trujillo-de Santiago, G.; Alvarez, M.M.; Tamayol, A.; Annabi, N.; Khademhosseini, A. Synthesis, Properties, and Biomedical Applications of Gelatin Methacryloyl (GelMA) Hydrogels. Biomaterials 2015, 73, 254–271. [Google Scholar] [CrossRef]
- Zhang, X.; Pan, Y.; Li, S.; Xing, L.; Du, S.; Yuan, G.; Li, J.; Zhou, T.; Xiong, D.; Tan, H.; et al. Doubly Crosslinked Biodegradable Hydrogels Based on Gellan Gum and Chitosan for Drug Delivery and Wound Dressing. Int. J. Biol. Macromol. 2020, 164, 2204–2214. [Google Scholar] [CrossRef]
- Zhang, Y.; Chen, H.; Zhang, T.; Zan, Y.; Ni, T.; Cao, Y.; Wang, J.; Liu, M.; Pei, R. Injectable Hydrogels from Enzyme-Catalyzed Crosslinking as BMSCs-Laden Scaffold for Bone Repair and Regeneration. Mater. Sci. Eng. C 2019, 96, 841–849. [Google Scholar] [CrossRef]
- Liu, X.; Sun, S.; Wang, N.; Kang, R.; Xie, L.; Liu, X. Therapeutic Application of Hydrogels for Bone-Related Diseases. Front. Bioeng. Biotechnol. 2022, 10, 998988. [Google Scholar] [CrossRef]
- Shi, W.; Jiang, Y.; Wu, T.; Zhang, Y.; Li, T. Advancements in Drug-Loaded Hydrogel Systems for Bone Defect Repair. Regen. Ther. 2023, 25, 174–185. [Google Scholar] [CrossRef]
- Zheng, S.; Zhong, H.; Cheng, H.; Li, X.; Zeng, G.; Chen, T.; Zou, Y.; Liu, W.; Sun, C. Engineering Multifunctional Hydrogel With Osteogenic Capacity for Critical-Size Segmental Bone Defect Repair. Front. Bioeng. Biotechnol. 2022, 10, 899457. [Google Scholar] [CrossRef]
- Rust, P.A.; Kalsi, P.; Briggs, T.W.R.; Cannon, S.R.; Blunn, G.W. Will Mesenchymal Stem Cells Differentiate into Osteoblasts on Allograft? Clin. Orthop. Relat. Res. 2007, 457, 220–226. [Google Scholar] [CrossRef]
- Rasch, A.; Naujokat, H.; Wang, F.; Seekamp, A.; Fuchs, S.; Klüter, T. Evaluation of Bone Allograft Processing Methods: Impact on Decellularization Efficacy, Biocompatibility and Mesenchymal Stem Cell Functionality. PLoS ONE 2019, 14, e0218404. [Google Scholar] [CrossRef] [PubMed]
- Vallés, G.; Bensiamar, F.; Maestro-Paramio, L.; García-Rey, E.; Vilaboa, N.; Saldaña, L. Influence of Inflammatory Conditions Provided by Macrophages on Osteogenic Ability of Mesenchymal Stem Cells. Stem Cell Res. Ther. 2020, 11, 57. [Google Scholar] [CrossRef] [PubMed]
- Poliwoda, S.; Noor, N.; Downs, E.; Schaaf, A.; Cantwell, A.; Ganti, L.; Kaye, A.D.; Mosel, L.I.; Carroll, C.B.; Viswanath, O.; et al. Stem Cells: A Comprehensive Review of Origins and Emerging Clinical Roles in Medical Practice. Orthop. Rev. 2022, 14, 37498. [Google Scholar] [CrossRef] [PubMed]
- de Girolamo, L.; Bertolini, G.; Cervellin, M.; Sozzi, G.; Volpi, P. Treatment of Chondral Defects of the Knee with One Step Matrix-Assisted Technique Enhanced by Autologous Concentrated Bone Marrow: In Vitro Characterisation of Mesenchymal Stem Cells from Iliac Crest and Subchondral Bone. Injury 2010, 41, 1172–1177. [Google Scholar] [CrossRef]
- Horwitz, E.M.; Le Blanc, K.; Dominici, M.; Mueller, I.; Slaper-Cortenbach, I.; Marini, F.C.; Deans, R.J.; Krause, D.S.; Keating, A. Clarification of the Nomenclature for MSC: The International Society for Cellular Therapy Position Statement. Cytotherapy 2005, 7, 393–395. [Google Scholar] [CrossRef]
- Jia, Z.; Wang, S.; Liu, Q. Identification of Differentially Expressed Genes by Single-Cell Transcriptional Profiling of Umbilical Cord and Synovial Fluid Mesenchymal Stem Cells. J. Cell Mol. Med. 2020, 24, 1945–1957. [Google Scholar] [CrossRef] [PubMed]
- Stanko, P.; Kaiserova, K.; Altanerova, V.; Altaner, C. Comparison of Human Mesenchymal Stem Cells Derived from Dental Pulp, Bone Marrow, Adipose Tissue, and Umbilical Cord Tissue by Gene Expression. Biomed. Pap. Med. Fac. Univ. Palacky. Olomouc Czech Repub. 2014, 158, 373–377. [Google Scholar] [CrossRef]
- Benavides-Castellanos, M.P.; Garzón-Orjuela, N.; Linero, I. Effectiveness of Mesenchymal Stem Cell-Conditioned Medium in Bone Regeneration in Animal and Human Models: A Systematic Review and Meta-Analysis. Cell Regen. 2020, 9, 5. [Google Scholar] [CrossRef]
- Ismail, H.D.; Phedy, P.; Kholinne, E.; Djaja, Y.P.; Kusnadi, Y.; Merlina, M.; Yulisa, N.D. Mesenchymal Stem Cell Implantation in Atrophic Nonunion of the Long Bones: A Translational Study. Bone Jt. Res. 2016, 5, 287–293. [Google Scholar] [CrossRef] [PubMed]
- Kim, M.-S.; Chung, H.-J.; Kim, K.-I. Optimal Concentration of Mesenchymal Stem Cells for Fracture Healing in a Rat Model with Long Bone Fracture. World J. Stem Cells 2022, 14, 839–850. [Google Scholar] [CrossRef]
- Peters, A.; Toben, D.; Lienau, J.; Schell, H.; Bail, H.J.; Matziolis, G.; Duda, G.N.; Kaspar, K. Locally Applied Osteogenic Predifferentiated Progenitor Cells Are More Effective Than Undifferentiated Mesenchymal Stem Cells in the Treatment of Delayed Bone Healing. Tissue Eng. Part A 2009, 15, 2947–2954. [Google Scholar] [CrossRef]
- Buchert, J.M.; Lotz, B.; Diederichs, S.; Richter, W. Adipose-Derived Stromal Cells: Isolation, Expansion, and Differentiation. Methods Mol. Biol. 2023, 2598, 75–85. [Google Scholar] [CrossRef]
- Schubert, T.; Lafont, S.; Beaurin, G.; Grisay, G.; Behets, C.; Gianello, P.; Dufrane, D. Critical Size Bone Defect Reconstruction by an Autologous 3D Osteogenic-like Tissue Derived from Differentiated Adipose MSCs. Biomaterials 2013, 34, 4428–4438. [Google Scholar] [CrossRef]
- Granero-Moltó, F.; Weis, J.A.; Miga, M.I.; Landis, B.; Myers, T.J.; O’Rear, L.; Longobardi, L.; Jansen, E.D.; Mortlock, D.P.; Spagnoli, A. Regenerative Effects of Transplanted Mesenchymal Stem Cells in Fracture Healing. Stem Cells 2009, 27, 1887–1898. [Google Scholar] [CrossRef]
- Ji, X.; Yuan, X.; Ma, L.; Bi, B.; Zhu, H.; Lei, Z.; Liu, W.; Pu, H.; Jiang, J.; Jiang, X.; et al. Mesenchymal Stem Cell-Loaded Thermosensitive Hydroxypropyl Chitin Hydrogel Combined with a Three-Dimensional-Printed Poly(ε-Caprolactone)/Nano-Hydroxyapatite Scaffold to Repair Bone Defects via Osteogenesis, Angiogenesis and Immunomodulation. Theranostics 2020, 10, 725–740. [Google Scholar] [CrossRef]
- Lee, D.J.; Miguez, P.; Kwon, J.; Daniel, R.; Padilla, R.; Min, S.; Zalal, R.; Ko, C.-C.; Shin, H.W. Decellularized Pulp Matrix as Scaffold for Mesenchymal Stem Cell Mediated Bone Regeneration. J. Tissue Eng. 2020, 11, 2041731420981672. [Google Scholar] [CrossRef] [PubMed]
- Park, S.; Tofighi, A.; Wang, X.; Strunk, M.; Ricketts, T.; Chang, J.; Kaplan, D.L. Calcium Phosphate Combination Biomaterials as Human Mesenchymal Stem Cell Delivery Vehicles for Bone Repair. J. Biomed. Mater. Res. 2011, 97B, 235–244. [Google Scholar] [CrossRef] [PubMed]
- Šponer, P.; Filip, S.; Kučera, T.; Brtková, J.; Urban, K.; Palička, V.; Kočí, Z.; Syka, M.; Bezrouk, A.; Syková, E. Utilizing Autologous Multipotent Mesenchymal Stromal Cells Andβ-Tricalcium Phosphate Scaffold in Human Bone Defects: A Prospective, Controlled Feasibility Trial. BioMed Res. Int. 2016, 2016, 1–12. [Google Scholar] [CrossRef] [PubMed]
- Maiti, S.K.; Shivakumar, M.U.; Mohan, D.; Kumar, N.; Singh, K.P. Mesenchymal Stem Cells of Different Origin-Seeded Bioceramic Construct in Regeneration of Bone Defect in Rabbit. Tissue Eng. Regen. Med. 2018, 15, 477–492. [Google Scholar] [CrossRef]
- Ninu, A.R.; Maiti, S.K.; Kumar, S.; Kumar, S.; Sangeetha, P.; Kritaniya, D.; Gupta, S.; Saxena, A.; Kumar, N. Isolation, Proliferation, Characterization and in Vivo Osteogenic Potential of Bone-Marrow Derived Mesenchymal Stem Cells (rBMSC) in Rabbit Model. Indian. J. Exp. Biol. 2017, 55, 79–87. [Google Scholar]
- Li, X.; Zhang, R.; Tan, X.; Li, B.; Liu, Y.; Wang, X. Synthesis and Evaluation of BMMSC-Seeded BMP-6/nHAG/GMS Scaffolds for Bone Regeneration. Int. J. Med. Sci. 2019, 16, 1007–1017. [Google Scholar] [CrossRef]
- Kim, Y.; Lee, S.H.; Kang, B.; Kim, W.H.; Yun, H.; Kweon, O. Comparison of Osteogenesis between Adipose-Derived Mesenchymal Stem Cells and Their Sheets on Poly-ε-Caprolactone/β-Tricalcium Phosphate Composite Scaffolds in Canine Bone Defects. Stem Cells Int. 2016, 2016, 8414715. [Google Scholar] [CrossRef]
- Hao, Z.; Lu, J.; Wang, S.; Wu, H.; Zhang, Y.; Xu, S. Stem Cell-derived Exosomes: A Promising Strategy for Fracture Healing. Cell Prolif. 2017, 50, e12359. [Google Scholar] [CrossRef]
- Hu, J.-C.; Zheng, C.-X.; Sui, B.-D.; Liu, W.-J.; Jin, Y. Mesenchymal Stem Cell-Derived Exosomes: A Novel and Potential Remedy for Cutaneous Wound Healing and Regeneration. World J. Stem Cells 2022, 14, 318–329. [Google Scholar] [CrossRef]
- Qin, Y.; Wang, L.; Gao, Z.; Chen, G.; Zhang, C. Bone Marrow Stromal/Stem Cell-Derived Extracellular Vesicles Regulate Osteoblast Activity and Differentiation In Vitro and Promote Bone Regeneration In Vivo. Sci. Rep. 2016, 6, 21961. [Google Scholar] [CrossRef]
- Narayanan, R.; Huang, C.-C.; Ravindran, S. Hijacking the Cellular Mail: Exosome Mediated Differentiation of Mesenchymal Stem Cells. Stem Cells Int. 2016, 2016, 3808674. [Google Scholar] [CrossRef]
- Ankrum, J.A.; Ong, J.F.; Karp, J.M. Mesenchymal stem cells: Immune evasive, not immune privileged. Nat. Biotechnol. 2014, 32, 252–260. [Google Scholar] [CrossRef] [PubMed]
- Sanabria-de la Torre, R.; Quiñones-Vico, M.I.; Fernández-González, A.; Sánchez-Díaz, M.; Montero-Vílchez, T.; Sierra-Sánchez, Á.; Arias-Santiago, S. Alloreactive Immune Response Associated to Human Mesenchymal Stromal Cells Treatment: A Systematic Review. J. Clin. Med. 2021, 10, 2991. [Google Scholar] [CrossRef] [PubMed]
- Corcoran, K.E.; Trzaska, K.A.; Fernandes, H.; Bryan, M.; Taborga, M.; Srinivas, V.; Packman, K.; Patel, P.S.; Rameshwar, P. Mesenchymal Stem Cells in Early Entry of Breast Cancer into Bone Marrow. PLoS ONE 2008, 3, e2563. [Google Scholar] [CrossRef]
- Ridge, S.M.; Sullivan, F.J.; Glynn, S.A. Mesenchymal Stem Cells: Key Players in Cancer Progression. Mol. Cancer 2017, 16, 31. [Google Scholar] [CrossRef] [PubMed]
- Shapiro, G.; Lieber, R.; Gazit, D.; Pelled, G. Recent Advances and Future of Gene Therapy for Bone Regeneration. Curr. Osteoporos. Rep. 2018, 16, 504–511. [Google Scholar] [CrossRef]
- De la Vega, R.E.; Atasoy-Zeybek, A.; Panos, J.A.; VAN Griensven, M.; Evans, C.H.; Balmayor, E.R. Gene Therapy for Bone Healing: Lessons Learned and New Approaches. Transl. Res. 2021, 236, 1–16. [Google Scholar] [CrossRef]
- Watson-Levings, R.S.; Palmer, G.D.; Levings, P.P.; Dacanay, E.A.; Evans, C.H.; Ghivizzani, S.C. Gene Therapy in Orthopaedics: Progress and Challenges in Pre-Clinical Development and Translation. Front. Bioeng. Biotechnol. 2022, 10, 901317. [Google Scholar] [CrossRef]
- Al-Dosari, M.S.; Gao, X. Nonviral Gene Delivery: Principle, Limitations, and Recent Progress. AAPS J. 2009, 11, 671–681. [Google Scholar] [CrossRef]
- 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]
- 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]
- 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] [PubMed]
- Kumar, S.; Wan, C.; Ramaswamy, G.; Clemens, T.L.; Ponnazhagan, S. Mesenchymal Stem Cells Expressing Osteogenic and Angiogenic Factors Synergistically Enhance Bone Formation in a Mouse Model of Segmental Bone Defect. Mol. Ther. 2010, 18, 1026–1034. [Google Scholar] [CrossRef]
- Lin, C.-Y.; Lin, K.-J.; Kao, C.-Y.; Chen, M.-C.; Lo, W.-H.; Yen, T.-C.; Chang, Y.-H.; Hu, Y.-C. The Role of Adipose-Derived Stem Cells Engineered with the Persistently Expressing Hybrid Baculovirus in the Healing of Massive Bone Defects. Biomaterials 2011, 32, 6505–6514. [Google Scholar] [CrossRef] [PubMed]
- Sarkar, A.; Gallo, M.C.; Bell, J.A.; Mayfield, C.K.; Ball, J.R.; Ayad, M.; Lechtholz-Zey, E.; Chang, S.W.; Sugiyama, O.; Evseenko, D.; et al. Ex Vivo Regional Gene Therapy Compared to Recombinant BMP-2 for the Treatment of Critical-Size Bone Defects: An In Vivo Single-Cell RNA-Sequencing Study. Bioengineering 2025, 12, 29. [Google Scholar] [CrossRef]
- Razi Soofiyani, S.; Baradaran, B.; Lotfipour, F.; Kazemi, T.; Mohammadnejad, L. Gene Therapy, Early Promises, Subsequent Problems, and Recent Breakthroughs. Adv. Pharm. Bull. 2013, 3, 249–255. [Google Scholar] [CrossRef]
- 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]
- Evans, C.H.; Liu, F.J.; Glatt, V. Use of Genetically Modified Muscle and Fat Grafts to Repair Defects in Bone and Cartilage. Eur. Cell Mater. 2009, 18, 96–111. [Google Scholar] [CrossRef]
- Virk, M.S.; Sugiyama, O.; Park, S.H. Same Day” Ex-Vivo Regional Gene Therapy: A Novel Strategy to Enhance Bone Repair. Mol. Ther. 2011, 19, 960–968. [Google Scholar] [CrossRef]
- 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]
- 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, 95–104. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed]
- 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]
- 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] [PubMed]
- 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]
- Liu, F.; Ferreira, E.; Porter, R.M. Rapid and Reliable Healing of Critical Size Bone Defects with Genetically Modified Sheep Muscle. Eur. Cell Mater. 2015, 30, 118–131. [Google Scholar] [CrossRef]
- Shirley, J.L.; de Jong, Y.P.; Terhorst, C.; Herzog, R.W. Immune Responses to Viral Gene Therapy Vectors. Mol. Ther. 2020, 28, 709–722. [Google Scholar] [CrossRef]
- 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. 1999, 10, 2245–2253. [Google Scholar] [CrossRef]
- RE, V.; MJ, C.; SA, M. Effects of FK506 on the Healing of Diaphyseal, Critical Size Defects in the Rat Femur. Eur. Cell Mater. 2020, 40, 160–171. [Google Scholar] [CrossRef]
- Bell, J.A.; Collon, K.; Mayfield, C.; Gallo, M.C.; Chang, S.W.; Sugiyama, O.; Tang, A.H.; Hollis, R.; Chopra, S.; Kohn, D.B.; et al. Biodistribution of Lentiviral Transduced Adipose-Derived Stem Cells for “Ex-Vivo” Regional Gene Therapy for Bone Repair. Gene Ther. 2023, 30, 826–834. [Google Scholar] [CrossRef]
- Vakhshori, V.; Bougioukli, S.; Sugiyama, O. Ex Vivo Regional Gene Therapy with Human Adipose-Derived Stem Cells for Bone Repair. Bone 2020, 138, 115524. [Google Scholar] [CrossRef]
- Bougioukli, S.; Saitta, B.; Sugiyama, O.; Tang, A.H.; Elphingstone, J.; Evseenko, D.; Lieberman, J.R. Lentiviral Gene Therapy for Bone Repair Using Human Umbilical Cord Blood-Derived Mesenchymal Stem Cells. Hum. Gene Ther. 2019, 30, 906–917. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed]
- Sonobe, J.; Okubo, Y.; Kaihara, S.; Miyatake, S.-I.; Bessho, K. Osteoinduction by Bone Morphogenetic Protein 2-Expressing Adenoviral Vector: Application of Biomaterial to Mask the Host Immune Response. Hum. Gene Ther. 2004, 15, 659–668. [Google Scholar] [CrossRef] [PubMed]
- Bauso, L.V.; La Fauci, V.; Longo, C.; Calabrese, G. Bone Tissue Engineering and Nanotechnology: A Promising Combination for Bone Regeneration. Biology 2024, 13, 237. [Google Scholar] [CrossRef]
- Siqueira, F.S.; Gomes, A.D.J.; Lunardi, C.N. Integrating Nanotechnology With 3D Printing for Drug Delivery: Bone Regeneration and Beyond. Nano Select 2025, 6, e70030. [Google Scholar] [CrossRef]
- Cui, Y.; Li, N.; Wu, J.; Wang, X.; Zhou, P.; Bai, J.; Liu, Y. Navigating the Landscape of Materials-Based RNA Delivery Systems in Bone Healing. Adv. Funct. Mater. 2025, e09383. [Google Scholar] [CrossRef]
- Iyer, S.S. Epigenetic Regulation of Bone Healing: Implications for Fracture Repair and Clinical Treatment Strategies. Yale J. Biol. Med. 2025, 98, 159–170. [Google Scholar] [CrossRef]
- Park, S.-Y.; Lee, J.-K.; Lee, S.-H.; Kim, D.-S.; Jung, J.-W.; Kim, J.H.; Baek, S.-W.; You, S.; Hwang, D.-Y.; Han, D.K. Multifunctional Vitamin D-Incorporated PLGA Scaffold with BMP/VEGF-Overexpressed Tonsil-Derived MSC via CRISPR/Cas9 for Bone Tissue Regeneration. Mater. Today Bio 2024, 28, 101254. [Google Scholar] [CrossRef]
- Da Silva, V.A.; Sharma, R.; Shteinberg, E.; Patel, V.; Bhardwaj, L.; Garay, T.; Yu, B.; Willerth, S.M. Machine Learning Approaches to 3D Models for Drug Screening. Biomed. Mater. Devices 2024, 2, 695–720. [Google Scholar] [CrossRef]
- Degenhart, C.; Engelhardt, L.; Niemeyer, F.; Erne, F.; Braun, B.; Gebhard, F.; Schütze, K. Computer-Based Mechanobiological Fracture Healing Model Predicts Non-Union of Surgically Treated Diaphyseal Femur Fractures. J. Clin. Med. 2023, 12, 3461. [Google Scholar] [CrossRef] [PubMed]
- Donie, C.; Reumann, M.K.; Hartung, T.; Braun, B.J.; Histing, T.; Endo, S.; Hirche, S. Predictive Model Development to Identify Failed Healing in Patients after Non-Union Fracture Surgery. In Proceedings of the 2024 46th Annual International Conference of the IEEE Engineering in Medicine and Biology Society (EMBC), Orlando, FL, USA, 15–19 July 2024; IEEE: Piscataway, NJ, USA, 2024; pp. 1–4. [Google Scholar] [CrossRef]
- Yu, H.; Mu, Q.; Wang, Z.; Guo, Y.; Zhao, J.; Wang, G.; Wang, Q.; Meng, X.; Dong, X.; Wang, S.; et al. A Study on Early Diagnosis for Fracture Non-Union Prediction Using Deep Learning and Bone Morphometric Parameters. Front. Med. 2025, 12, 1547588. [Google Scholar] [CrossRef] [PubMed]
- Breulmann, F.L.; Hatt, L.P.; Schmitz, B.; Wehrle, E.; Richards, R.G.; Della Bella, E.; Stoddart, M.J. Prognostic and Therapeutic Potential of microRNAs for Fracture Healing Processes and Non-Union Fractures: A Systematic Review. Clin. Transl. Med. 2023, 13, e1161. [Google Scholar] [CrossRef] [PubMed]
- Chitwood, J.R.; Chakraborty, N.; Hammamieh, R.; Moe, S.M.; Chen, N.X.; Kacena, M.A.; Natoli, R.M. Predicting Fracture Healing with Blood Biomarkers: The Potential to Assess Patient Risk of Fracture Nonunion. Biomarkers 2021, 26, 703–717. [Google Scholar] [CrossRef]
- Ribitsch, I.; Baptista, P.M.; Lange-Consiglio, A.; Melotti, L.; Patruno, M.; Jenner, F.; Schnabl-Feichter, E.; Dutton, L.C.; Connolly, D.J.; van Steenbeek, F.G.; et al. Large Animal Models in Regenerative Medicine and Tissue Engineering: To Do or Not to Do. Front. Bioeng. Biotechnol. 2020, 8, 972. [Google Scholar] [CrossRef]
- Dai, K.; Xu, X.; Tang, T.; Zhu, Z.; Yu, C.; Xu, M.; Zhu, L.; Hao, Y.; Lou, J. BMP-2 gene modified tissue-engineered bone repairing segmental tibial bone defects in goats. Zhonghua Yi Xue Za Zhi 2003, 83, 1345–1349. [Google Scholar]
- Arinzeh, T.L.; Peter, S.J.; Archambault, M.P.; van den Bos, C.; Gordon, S.; Kraus, K.; Smith, A.; Kadiyala, S. Allogeneic Mesenchymal Stem Cells Regenerate Bone in a Critical-Sized Canine Segmental Defect. J. Bone Jt. Surg. Am. 2003, 85, 1927–1935. [Google Scholar] [CrossRef]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2025 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).