OBM Transplantation

(ISSN 2577-5820)

OBM Transplantation (ISSN 2577-5820) is an international peer-reviewed Open Access journal published quarterly online by LIDSEN Publishing Inc., which covers all evidence-based scientific studies related to transplantation, including: transplantation procedures and the maintenance of transplanted tissues or organs; assimilation of grafted tissue and the reconstitution of removed organs or parts of organs; transplantation of heart, lung, kidney, liver, pancreatic islets and bone marrow, etc. Areas related to clinical and experimental transplantation are also of interest.

OBM Transplantation is committed to rapid review and publication, and we aim at serving the international transplant community with high accessibility as well as relevant and high quality content.

The journal publishes all types of articles in English. There is no restriction on the length of the papers. We encourage authors to be concise but present their results in as much detail as necessary, as reviewers are expected to emphasize scientific rigor and reproducibility.

 
 

Publication Speed (median values for papers published in 2024): Submission to First Decision: 6.7 weeks; Submission to Acceptance: 14.4 weeks; Acceptance to Publication: 4 days (1-2 days of FREE language polishing included)

 
 
Open Access Review

Pharmacological Strategies to Enhance Bone Graft Integration: Emerging Agents and Molecular Pathways

Garzain Bint e Attar , Mohd. Ashif Khan *

  1. Department of Translational and Clinical Research, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India

Correspondence: Mohd. Ashif Khan

Academic Editor: Eric M. Bluman

Special Issue: Bone Grafting in Trauma and Elective Orthopaedics

Received: May 15, 2025 | Accepted: December 18, 2025 | Published: December 31, 2025

OBM Transplantation 2025, Volume 9, Issue 4, doi:10.21926/obm.transplant.2504262

Recommended citation: Bint e Attar G, Khan MA. Pharmacological Strategies to Enhance Bone Graft Integration: Emerging Agents and Molecular Pathways. OBM Transplantation 2025; 9(4): 262; doi:10.21926/obm.transplant.2504262.

© 2025 by the authors. This is an open access article distributed under the conditions of the Creative Commons by Attribution License, which permits unrestricted use, distribution, and reproduction in any medium or format, provided the original work is correctly cited.

Abstract

Bone grafting remains a cornerstone technique in orthopedic and reconstructive surgery, yet achieving successful graft integration continues to pose significant challenges, particularly in conditions such as osteoporosis, diabetes mellitus, and large bone defects. Traditional graft materials such as autografts, allografts, xenografts, and synthetics, often encounter limitations including immune rejection, poor vascularization, and insufficient osteogenic support. Emerging pharmacological strategies have shown promise in enhancing graft integration by modulating bone-healing pathways, promoting angiogenesis, and regulating inflammatory responses. This review comprehensively explores the biological mechanisms underlying bone repair, including the roles of key molecular pathways such as Wnt/β-catenin, BMP signaling, VEGF-mediated angiogenesis, and the RANK/RANKL/OPG axis. It further examines the therapeutic application of osteoinductive agents (e.g., BMPs, PTH analogs), anti-resorptive drugs (e.g., bisphosphonates, Denosumab), angiogenic modulators (e.g., VEGF, PDGF), and biologics targeting inflammatory cytokines (e.g., TNF-α, IL-6, IL-1β). Innovative approaches such as nanotechnology-based drug delivery, scaffold-based release systems, and gene therapy are also discussed for their potential to achieve localized, controlled, and sustained enhancement of graft performance. While several FDA-approved agents, such as rhBMP-2 and PDGF-BB, have advanced clinical practice, persistent challenges including variability in patient healing, delivery limitations, adverse effects, and regulatory hurdles, highlight the need for continued research. Future directions emphasize the development of multifunctional, personalized therapeutics that actively guide bone regeneration, supported by rigorous translational studies to ensure clinical efficacy and safety.

Graphical abstract

Click to view original image

Keywords

Bone graft integration; pharmacological strategies; osteoinductive agents; angiogenesis; inflammatory cytokines; nanotechnology-based drug delivery

1. Introduction

Bone Grafting is a surgical technique used to replace missing bone tissue through donor-to-recipient transfers of bone cells, employing autogenous, allogeneic, or synthetic materials to both support structure and facilitate bone healing [1]. Orthopedic and reconstructive surgery use this essential procedure to repair skeletal defects that arise from different causes, such as trauma, congenital anomalies, infection, or tumor resection [2]. The main purpose of bone grafting is to promote new bone growth, enabling patients to regain functional bone structure in affected areas. The selection of graft material is a fundamental factor for achieving positive clinical results [3].

Medical practice utilizes various bone grafts, which differ in their biological responses and mechanical properties [4]. Autografts are the preferred choice because they originate from patient tissue, which possesses remarkable osteogenic, osteoinductive, and osteoconductive properties. However, the quantity of available graft tissue, along with donor-site complications, limits the use of autografts [5]. The use of human donor allografts offers practical surgical advantages, although these materials pose risks of immunological reactions and disease transmission [6]. Xenografts that use non-human tissue and synthetic grafts made from ceramics and polymers provide scalable alternatives, though they fail to achieve the same biological activity as autografts [7]. This diversity in graft options highlights a persistent clinical dilemma: graft choice alone cannot consistently ensure reliable integration across all patient populations.

The range of available grafting materials does not resolve the clinical challenge of achieving proper graft integration. The main obstacle to graft success stems from rejection events driven by incompatible immune responses, particularly in allografts and xenografts [8,9]. Additionally, poor vascularization at the graft site impairs nutrient delivery and cellular infiltration, thereby compromising bone regeneration [10]. Tissue damage becomes worse when an immune response becomes too active because it blocks the process of new bone formation [11]. These limitations highlight the need for therapeutic strategies that do not merely replace the defect but actively modulate the biological environment to support integration.

The clinical treatment of bone defects becomes much more challenging when treating patients whose physiological state makes bone healing and repair very difficult [12]. Osteoporosis affects bone density and the balance of bone remodeling, reducing the support required for graft stability and integration [13]. Such bones with limited osteogenic potential tend to heal poorly, delaying bone repair. Diabetes mellitus causes additional complications because elevated blood glucose levels destroy bone-forming cells (osteoblasts) while blocking the development of new blood vessels needed for graft nourishment [14]. The healing process is delayed, and the risk of graft failure increases [15]. Large bone defects exceed the natural healing abilities of human tissues, resulting in an insufficient ability to close gaps. The healing process becomes challenging since these defects need complete cellular activity together with scaffold support and vascularization, yet these elements are often absent.

Given these limitations, there is increasing interest in using pharmacological agents to enhance bone graft success. The agents demonstrate benefit by maintaining cell viability while enhancing bone tissue integration and accelerating tissue repair at implant sites. Growth factors such as BMPs and VEGF drive osteogenic and vascular pathways, while anti-inflammatory biologics may prevent immune-mediated graft damage [16]. Angiogenic modulators improve vascular supply, and osteoinductive compounds accelerate matrix deposition. Importantly, these drugs are increasingly viewed not as standalone solutions but as synergistic adjuncts that can tailor the biological environment to meet the demands of challenging clinical scenarios such as osteoporosis, diabetes, and large bone defects [10].

Thus, this review examines emerging pharmacological agents and molecular pathways that have the potential to enhance bone graft integration, with particular emphasis on their mechanisms of action, clinical evidence, and translational relevance.

2. Mechanisms of Bone Healing and Graft Integration

Bone healing is a coordinated biological process that restores the structural and mechanical integrity of bone following injury or surgical intervention [12]. Successful graft integration similarly depends on the timely orchestration of cellular, molecular, and vascular events that parallel native bone repair [17]. The healing process progresses through overlapping inflammatory, repair, and remodeling phases [18], each governed by specific signaling pathways that regulate cell recruitment, differentiation, and matrix turnover. Understanding these mechanisms is essential because current pharmacological strategies target defined checkpoints within these pathways to optimize graft incorporation.

2.1 Phases of Bone Healing

2.1.1 Inflammatory Phase

The inflammatory phase initiates immediately after injury and establishes the microenvironment required for subsequent repair [19]. Hematoma formation at the fracture site recruits neutrophils, macrophages, and lymphocytes [20], which secrete cytokines such as IL-1, IL-6, TNF-α, and TGF-β, triggering downstream healing cascades [19,21]. These mediators not only clear necrotic tissue but also activate mesenchymal stem cells (MSCs) to commit toward osteogenic lineages [22]. Because early inflammatory signaling influences MSC recruitment and vascular ingrowth, disturbances in this phase can directly compromise graft acceptance.

2.1.2 Repair Phase

Soft callus formation occurs first during the repair phase before the development of a hard callus. The process of mesenchymal stem cell differentiation results in the formation of chondrocytes and osteoblasts, leading to the development of cartilaginous and bony matrices, respectively [23]. Osteoblast activation is the main feature of this phase because it drives bone formation [24]. Concurrently, angiogenesis is stimulated when vascular endothelial growth factor (VEGF) is released into the tissue space, as both processes enable the damaged area to be revascularized and deliver the oxygen and nutrients required for regeneration [25]. The cartilaginous matrix is replaced by mineralized bone through endochondral ossification as the tissue progresses through its structural recovery [26]. The effectiveness of this phase is particularly relevant for graft integration, as insufficient vascularization is a major contributor to graft failure.

2.1.3 Remodeling Phase

The remodeling phase converts immature woven bone into mechanically resilient lamellar bone [27]. This process depends on coordinated osteoclast-mediated resorption and osteoblast-mediated deposition, influenced by mechanical loading and molecular cues [28]. Remodeling can continue for months and is essential for restoring biomechanical strength and long-term graft stability [29]. Modulation of this phase through antiresorptive or osteoanabolic therapies is therefore a key consideration in enhancing graft outcomes.

2.2 Key Molecular Pathways in Bone Regeneration

Several signaling pathways regulate bone healing, and many emerging pharmacological agents modulate these pathways to improve graft incorporation (Figure 1).

Click to view original image

Figure 1 Overview of major molecular pathways involved in bone graft integration and the pharmacological agents targeting them. The flowchart illustrates BMP/SMAD signaling (targeted by rhBMP-2 and rhBMP-7), Wnt/β-catenin activation (enhancing osteoblast differentiation), the RANK/RANKL/OPG axis (targeted by Denosumab), and VEGF-mediated angiogenesis (modulated by PDGF-BB and pro-angiogenic agents).

2.2.1 Wnt/β-Catenin Signaling Pathway

The Wnt/β-catenin pathway plays a central role in osteoblast lineage commitment and bone matrix formation [30,31]. Activation of Wnt ligands stabilizes β-catenin, allowing its nuclear translocation and the subsequent induction of osteogenic genes such as RUNX2 and osterix [32]. Impairment of Wnt signaling disrupts osteoblast maturation and compromises fracture healing and graft integration [33]. Interactions between Wnt and BMP pathways further illustrate the pathway’s importance in coordinated bone regeneration.

2.2.2 Bone Morphogenetic Protein (BMP) Signaling Pathway

BMPs, particularly BMP-2, BMP-4 and BMP-7, form part of the TGF-β superfamily to regulate bone formation as fundamental molecular factors [34]. Cell-surface receptors (BMPRs) bind BMPs, activating signaling that leads to SMAD protein activation and osteogenic gene transcription in the nucleus. The osteoblastic differentiation of mesenchymal stem cells, together with matrix mineralization, results from BMP signaling [35]. Recombinant BMPs have been used clinically to enhance bone healing and graft incorporation [36,37]. However, limitations such as cost and dose-related adverse effects have prompted interest in more targeted osteoinductive therapies.

2.2.3 RANK/RANKL/OPG Axis

Bone resorption, together with osteoclastogenesis, is controlled through the RANK/RANKL/OPG system [38]. Osteoblasts and stromal cells create RANKL to activate RANK on osteoclast precursors, which drives their differentiation and activation process [39]. Osteoprotegerin (OPG) functions as a decoy receptor made by osteoblasts to block RANKL binding to RANK, which prevents osteoclast development and function [40]. Maintaining an appropriate balance between resorption and formation is essential for graft remodeling. This axis also underlies antiresorptive therapies used to stabilize grafted bone.

2.2.4 Vascular Endothelial Growth Factor-Mediated Angiogenesis

The process of angiogenesis serves as a vital support mechanism during bone repair, particularly when treating large defects or inadequate blood supply conditions of grafts [41]. Vascular Endothelial Growth Factor (VEGF) is the main regulatory factor that stimulates endothelial cells to multiply and migrate directionally to form new blood vessels [42]. The newly developed blood vessels provide vital delivery of oxygen, nutrients, and signaling molecules, which support the regeneration of bone tissue. Furthermore, the VEGF signaling pathway operates in concert with Bone Morphogenetic Proteins (BMPs) to promote improved blood vessel development and bone tissue formation [43]. Additionally, under low-oxygen conditions, which are common at the injury site, hypoxia-inducible factors (HIFs) are activated to enhance VEGF expression, thereby strengthening the connection between the bone healing processes of angiogenesis and osteogenesis [44,45].

3. Pharmacological Strategies for Enhancing Bone Graft Integration

Pharmacological agents that enhance bone graft integration modulate osteogenesis, resorption, angiogenesis, and inflammation (Table 1). These strategies target defined molecular pathways involved in graft incorporation, thereby complementing surgical techniques and improving clinical outcomes:

Table 1 Pharmacological Agents Enhancing Bone Graft Integration – Mechanisms and Therapeutic Roles.

3.1 Osteoinductive and Anabolic Agents

Osteoinductive and anabolic agents enhance graft integration by stimulating osteoblast differentiation and matrix deposition. Their efficacy stems from activating specific signaling pathways that replicate or amplify physiological bone regeneration.

3.1.1 Bone Morphogenetic Proteins (BMPs)

BMPs operate as transformative regulators of bone formation and healing within the members of the transforming growth factor-beta (TGF-β) superfamily. The osteoinductive properties of BMP-2 and BMP-7 enable these proteins to trigger the transformation of mesenchymal stem cells into osteoblasts in a highly effective manner [65,66]. This process involves activation of the Smad signaling cascade, and when Smad signaling is activated, it stimulates the expression of osteogenic genes while the cell produces bone matrix components [67]. The clinical use of recombinant human BMPs (rhBMPs) exists for spinal fusion surgeries as well as non-union fracture repair and multiple orthopedic procedures, all of which aim to improve graft incorporation [67]. Nevertheless, the use of mesenchymal stem cell therapy requires careful evaluation because it has been associated with reported complications, which include ectopic bone formation and local inflammatory responses [68,69].

3.1.2 Parathyroid Hormone and Analogs

The administration of parathyroid hormone (PTH), along with its analog teriparatide (PTH 1-34), gives periodic short-term benefits to bone structure through enhanced osteoblast action and expanded cellular survival [70,71]. Through PTH1 receptor activation, cAMP and PKA signaling pathways become activated to mediate these effects. The United States Food and Drug Administration approves PTH analogs for osteoporosis treatment, while ongoing research assesses their potential to accelerate fracture healing and improve bone graft attachment to host tissue [72]. Research indicates that these agents enhance bone quality while accelerating graft-host union, especially in patients with reduced healing capacity [73,74,75,76].

3.2 Anti-Resorptive Agents

Bone graft integration requires a balanced coupling of resorption and formation. Excessive early resorption can destabilize grafts, making anti-resorptive therapies a valuable adjunct in selected scenarios.

3.2.1 Bisphosphonates

The synthetic compounds known as bisphosphonates show a strong attraction to hydroxyapatite in bone, which results in their targeted accumulation at active bone remodeling sites [77] (Figure 2). Osteoclasts absorb bisphosphonates (BPs) during bone resorption, and the drugs disrupt the mevalonate pathway, leading to prenylation defects in small GTPase proteins and, eventually, osteoclast cell death, followed by reduced bone breakdown [78]. Research demonstrates that BPs effectively improve graft survival rates by preventing excessive bone resorption [79,80]. The local application of BPs at low to moderate doses has been shown to be effective in enhancing both graft bone formation and mechanical strength according to research [81]. However, the incorporation of grafts into bone tissue can be compromised when BPs are administered systemically or when high local concentrations are used [80,81]. The therapeutic window for BP use in grafting is narrow; thus, the correct application of BP during grafting requires specialized drug-delivery systems because the effective dosage range is very narrow. Implant coatings that contain BP deliver the drug specifically to target areas to achieve high local impact without causing substantial systemic drug absorption [49].

Click to view original image

Figure 2 Classification of Bisphosphonates for Bone Graft Integration.

3.2.2 Denosumab

Denosumab is a fully human monoclonal antibody that binds to RANKL, preventing its interaction with the RANK receptor on osteoclast precursors [51]. Osteoclast differentiation stops, and their function, along with survival, is inhibited, leading to reduced bone resorption. The suppressive action of Denosumab on osteoclasts makes it an effective bone grafting support, especially for procedures at risk of substantial bone loss [82]. Research findings show that denosumab treatment improves both callus development and fracture gap healing in complex therapeutic situations [52,53]. Moreover, injectable bone graft composites containing this agent demonstrate promising results for controlling bone remodeling balance according to research [83].

The benefits of denosumab treatment for bone resorption control require close monitoring because stopping the medication leads to increased bone turnover [84]. Additionally, potential adverse effects such as hypocalcemia and osteonecrosis of the jaw require vigilance [85].

3.3 Angiogenic and Vascular-Enhancing Agents

Early vascularization is crucial for graft survival, nutrient delivery, and osteogenesis. Agents that promote angiogenesis directly influence graft success.

3.3.1 Vascular Endothelial Growth Factor (VEGF) and Platelet-Derived Growth Factor (PDGF) - Based Therapies

The angiogenic factor VEGF strongly drives endothelial cell growth and migration, leading to the formation of new blood vessels [86]. Bone graft revascularization occurs via VEGF, which enables the transport of essential nutrients and oxygen to the graft site [41]. Scientific investigations have proven that external VEGF treatments improve bone development and blood vessel network growth in laboratory animals with bone injuries [87,88]. During hypoxic conditions, which frequently occur in graft sites, VEGF expression increases to stimulate angiogenesis [89]. Studies have shown that VEGF interacts with the Notch and Noggin signaling pathways, demonstrating its complex role in linking angiogenesis to osteogenesis [90,91].

PDGF functions with VEGF to recruit pericytes and smooth muscle cells to stabilize newly developed blood vessels [55]. The graft site attracts mesenchymal stem cells (MSCs) through PDGF action, which later transform into osteoblasts for bone formation [92]. Research indicates that simultaneous administration of PDGF and VEGF produces concurrent benefits for vascularization and osteogenesis, supporting their joint use as a promising approach to improve bone graft integration [10,93].

3.3.2 Hypoxia-Inducible Factors (HIFs)

Hypoxia-Inducible Factors (HIFs), including HIF-1α, act as transcription factors that detect low oxygen levels to activate genes that control both angiogenesis and metabolic processes [56]. The hypoxic response mechanism in bone tissue depends heavily on HIF-1α functions to adapt to conditions that occur after graft implantation. The hypoxic environment causes HIF-1α to stabilize while it moves to the nucleus to activate VEGF and other target genes [94]. Visualizing the revascularization of the graft site becomes possible through VEGF upregulation because it enhances angiogenesis levels. The protein HIF-1α controls both the differentiation process and activity levels of osteoblasts, which in turn stimulates bone tissue regeneration [95,96]. Research has investigated deferoxamine (DFO) as well as other prolyl hydroxylase inhibitors for their ability to stabilize HIF-1α to promote bone healing [96,97,98]. Experimental research has demonstrated that DFO administration enhances bone regeneration through increased tissue repair because of new blood vessel formation in animal subjects [57]. This pathway represents a promising target for grafts placed in poorly vascularized environments.

3.4 Biologics Targeting Inflammatory Cytokines

Local inflammatory conditions strongly affect bone graft integration because tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6) and interleukin-1 beta (IL-1β) function as vital cytokines in this process. Graft failure becomes more likely when pro-inflammatory cytokines, which promote healing, exceed surpass normal levels, as they then inhibit osteogenesis [99,100,101].

Osteoclastogenesis is enhanced by TNF-α stimulates its development, leading to increased bone resorption [102]. Research shows that increased TNF-α levels create obstacles to both bone repair and implant material incorporation into the bone [103]. The biologic agents etanercept and infliximab show promise in managing TNF-α activity through their TNF-α inhibitory properties to enhance bone regeneration according to research [102,104,105]. Research findings show that inhibitor drugs decrease inflammation and enhance osteoblast differentiation, thereby improving graft integration [104,106,107].

The inflammatory response cytokine IL-6 plays dual functions within bone metabolism [108]. IL-6 supports osteoblast function under certain conditions, but it also drives osteoclast differentiation, leading to bone resorption [109]. Clinical studies have examined the use of the monoclonal antibody tocilizumab to target IL-6 because it aims to control bone resorption while maintaining bone formation [110,111]. Clinical studies show that blocking IL-6 enhances both bone density and graft stability [112,113].

IL-1β functions as a powerful inflammatory mediator that promotes osteoclast cell activity and suppresses osteoblast cell function to lead to bone loss [114]. The inflammatory effects of IL-1 receptor antagonist Anakinra have been shown to reduce these cellular responses [60,115]. The scientific evidence shows that inhibiting IL-1β creates beneficial conditions for bone maintenance and supports essential processes needed for graft integration [116].

The strategic use of biologic agents to control tissue inflammation appears to hold great potential for promoting bone graft integration. These therapies reduce harmful cytokine effects, which help establish a better environment for bone regeneration and graft incorporation.

3.5 Other Potential Pharmacological Strategies

Sclerostin inhibitors such as romosozumab enhance Wnt signaling and stimulate robust osteoblast activity, thereby increasing bone mass and strength [61,117]. These medicines enhance both osteoblast activity and bone formation by blocking sclerostin, resulting in stronger, denser bone. The pharmaceutical company is studying the use of romosozumab locally for bone grafting procedures despite its current systemic administration for osteoporosis treatment [62]. Delivering Wnt signaling stimulants at the implant site would improve graft integration while minimizing systemic drug effects, delivering superior healing results.

The integration of bone grafts requires the successful prevention of infections as a vital component. Preventing infection risks during bone graft integration relies on the widespread use of local antibiotic delivery systems, including polymethyl methacrylate (PMMA) beads and calcium sulfate pellets [63,64]. The traditional use of PMMA beads exists, but they lack resorbable properties, and patients may need surgical removal. The antibiotic release profile of calcium sulfate beads allows for extended drug delivery at therapeutic levels to the graft site as they are biodegradable [118,119]. Studies show that antibiotics-loaded calcium sulfate beads, including vancomycin and tobramycin, can stop bacterial growth and biofilm development that leads to graft failure [120]. Moreover, the resorption of calcium sulfate eliminates the need for further procedures to remove delivery systems, thereby reducing patient morbidity [121].

4. Comparative Insights Across Drug Classes and Key Translational Bottlenecks

Pharmacological agents used to enhance bone graft integration vary widely in their mechanisms, dosing requirements, and translational readiness. Osteoinductive factors such as BMP-2 and BMP-7 provide strong anabolic signals but require high doses and controlled delivery to avoid adverse effects, such as ectopic ossification [122]. Systemic anabolic agents, including intermittent PTH, offer more balanced remodeling support, although their benefits depend heavily on precise dosing schedules [70]. Anti-resorptive therapies (bisphosphonates and Denosumab) help stabilize grafts by reducing excessive osteoclastic activity, yet excessive suppression of turnover can delay long-term graft maturation [123]. Angiogenic agents such as VEGF and PDGF enhance early vascularization but face challenges due to short half-lives and difficulty achieving sustained, physiologic release at the graft site [124]. Immunomodulatory biologics targeting pro-inflammatory cytokines show promise in reducing graft resorption; however, concerns about systemic immunosuppression and optimal timing limit broader application [125]. Across all classes, key translational barriers persist, including narrow and poorly defined dosing windows, uncertainty surrounding the optimal timing of administration relative to graft placement, and ongoing challenges with spatiotemporally controlled delivery. Additionally, long-term safety concerns ranging from aberrant bone formation to immune dysregulation continue to impede routine clinical use. Addressing these limitations is essential to advance pharmacologic strategies that reliably support graft healing in clinical practice.

5. Mechanistic Pathways: Distinguishing Causal Evidence from Correlational Findings

Recent pharmacological approaches to improve bone graft integration act through diverse biological pathways; however, the strength of mechanistic evidence varies considerably. Pathways with clear causal roles are those demonstrated through genetic models, targeted inhibition, or controlled temporal manipulation. BMP-mediated Smad1/5/8 signaling has been directly linked to osteoblast differentiation and new bone formation, with knockout models showing failure of graft incorporation [65]. Similarly, intermittent PTH activation of the Wnt/β-catenin pathway has been shown to have a causal effect on osteoprogenitor expansion and improved graft remodeling [126]. VEGF-driven angiogenesis also has strong causal support, as vascular blockade consistently impairs graft survival in both preclinical and clinical settings [86].

In contrast, several pathways frequently described in the literature remain associative rather than mechanistically proven. Anti-inflammatory cytokine modulation (e.g., TNF-α or IL-6 inhibition) correlates with reduced graft resorption, yet causal links are less definitive because immunosuppression affects multiple downstream processes [127]. Similarly, many nanoparticle- or peptide-based therapies show improvements in bone volume or histology, but their specific intracellular targets remain incompletely elucidated, making the mechanistic relationship more speculative [128].

Overall, differentiating causally validated pathways from correlational findings is essential for translating pharmacologic strategies into predictable clinical interventions. Strengthening mechanistic clarity will also guide drug selection, dosing windows, and combination therapy design in future clinical applications.

6. Emerging Approaches for Targeted Drug Delivery

6.1 Nanotechnology-Based Drug Delivery

Nanotechnology has emerged as a key strategy for enhancing bone graft integration by enabling site-specific, controlled delivery of therapeutic agents while minimizing systemic exposure [129,130]. The nanoscale dimensions and elevated surface area of nanoparticles (NPs) enable engineers to develop therapeutic delivery systems targeting bone graft sites for improved osteointegration while reducing systemic toxicity [129]. NPs designed for drug delivery systems can transport different bioactive molecules, such as growth factors, antibiotics, and genetic material, to create favorable conditions for bone regeneration [131].

Mesoporous silica nanoparticles (MSNs) have been used to deliver combined bone morphogenetic proteins (BMPs) and antibiotics a single system [132]. Core–shell mesoporous silica nanoparticles containing BMP-2 at the core and gentamicin at the shell have shown dual capabilities for both osteogenic regeneration and antibacterial protection according to research [133]. Additionally, chitosan-based nanohybrid hydrogels with dual functionalities as bone regenerative scaffolds and drug delivery platforms for extended drug release capabilities at the treatment area have been developed [134].

Furthermore, titanium dioxide (TiO2) nanostructures have been explored for drug delivery applications because they exhibit superior biocompatibility and effective bone integration [135]. The drug delivery properties of TiO2 nanotubes have been developed to incorporate both antibiotics and growth factors, leading to effective bone healing and infection prevention [129,136]. Similarly, superparamagnetic iron oxide nanoparticles (SPIONs) offer the advantage of magnetic targeting, allowing for the directed delivery of drugs to specific bone sites under an external magnetic field, thereby enhancing the precision of treatment [137,138].

Overall, nanotechnology provides spatiotemporally controlled drug delivery, a feature critical for synchronized bone healing and the reduction of common postoperative complications. Recent AI-driven strategies, including the accelerated discovery of self-assembling peptides, are further expanding the design space for next-generation nanocarriers and enabling more precise control over therapeutic release profiles [139].

6.2 Scaffold-Based Drug Release Systems

Scaffold-based systems integrate mechanical support with localized therapeutic delivery, offering a dual advantage in bone graft procedures [140]. Advances in 3D printing enable fabrication of defect-specific scaffolds with controlled porosity and architecture, improving both tissue integration and drug-loading efficiency [141,142]. Drug molecules can be integrated into scaffolds to enable controlled drug delivery at the implantation site. The integration of scaffolds containing vascular endothelial growth factor (VEGF) with cephalexin antibiotics enables both proangiogenic and antibacterial properties, thereby enhancing bone regeneration while preventing infections [143].

Research has shown extensive use of biodegradable polymers including poly (lactic-co-glycolic acid) (PLGA) and gelatin because they enable the controlled delivery of growth factors [144]. PLGA nanoparticles containing bone morphogenetic protein-2 (BMP-2) have been shown to drive osteogenic stem cell differentiation thus demonstrating utility in bone tissue engineering [145]. Similarly, the release of growth factors through gelatin microparticles enables controlled biochemical delivery systems that enhance calcium deposition, a key indicator of bone healing [146].

These scaffold-based systems both give support to structures while preparing environments that promote bone healing by delivering therapeutic agents to specific treatment locations [147]. Recent advances in musculoskeletal organoid systems provide more physiologically relevant platforms for evaluating scaffold–tissue interactions and may help reduce dependence on animal models in preclinical bone regeneration research [148]. Innovations in hydrogel-based organoid culture further contribute to understanding how advanced biomaterials influence cellular organization and may guide the refinement of scaffold-mediated drug delivery strategies [149]. Such advanced drug delivery systems integrated into bone graft procedures offer the opportunity to enhance clinical outcomes and eliminate systemic drug use while reducing side effects.

6.3 Gene Therapy Approaches

Gene therapy offers a fundamentally different mechanism for promoting bone regeneration by enabling sustained, endogenous production of osteoinductive proteins at the graft site [150,151]. The approaches for delivering genes in bone regeneration can be divided into two main types: in vivo and ex vivo delivery [152]. The direct application of genetic material within the patient's body constitutes in vivo gene therapy because it targets cells at the graft site [153]. Ex vivo gene therapy requires medical professionals to extract patient cells followed by laboratory modification to produce osteoinductive factors before returning the cells to the graft site [153]. Both methods have shown potential in promoting bone healing and integration.

The clinical translation of gene- and biologic-based therapeutics is limited not only by their biological potency but also by delivery feasibility. Viral vectors, particularly adenoviruses, are the primary choice used in gene therapy because of their high transduction efficiency [154]. However, researchers now work on non-viral delivery systems because viral vectors present risks, including immune responses and mutagenicity [155]. Consequently, attention has shifted to non-viral carriers, including liposomes and nanoparticles, which offer safer profiles but lower transfection efficiency [156]. Liposomal delivery of BMP-2 genes has been shown to enhance bone regeneration in animal models [157,158].

Despite the potential benefits, several challenges hinder the clinical translation of gene therapy in bone regeneration [159]. The safety and efficiency of gene delivery vectors, along with potential immune responses and uncontrolled gene expression that can lead to adverse events, represent significant concerns for gene therapy. Moreover, the main challenge in tissue engineering is achieving controlled gene expression at the graft site without harming adjacent tissues. Addressing these challenges requires the development of advanced delivery systems and thorough preclinical studies to ensure safety and efficacy. The increasing integration of AI tools into scientific research is also accelerating biomaterial design, predictive modelling, and literature synthesis, supporting the development of next-generation drug delivery systems and regenerative platforms [160].

7. FDA-Approved Pharmacological Agents for Enhancing Bone Graft Integration

The U.S. Food and Drug Administration (FDA) has approved several pharmacological agents for boosting bone graft integration in orthopedic and dental procedures as listed in Table 2. These include osteoinductive growth factors such as BMPs, angiogenic mediators like PDGF-BB, and synthetic peptides that enhance cell–matrix interactions.

Table 2 FDA-approved agents for bone graft integration.

The FDA approved Recombinant human Bone Morphogenetic Protein-2 (rhBMP-2) as the most popular therapeutic agent for anterior lumbar interbody fusion in 2002, making it the first of its kind [161]. The FDA approved rhBMP-2 for tibial nonunion treatment in 2004 under the commercial name Infuse (Medtronic) and for specific oral and maxillofacial procedures in 2007 [165]. As an osteoinductive agent, rhBMP-2 drives mesenchymal stem cells to differentiate into osteoblasts and accelerates bone formation [166].

The FDA approved recombinant human Bone Morphogenetic Protein-7 (rhBMP-7), known commercially as OP-1 (Stryker), through a Humanitarian Device Exemption (HDE) to treat long bone nonunions in 2001 [46]. The product delivered promising outcomes, but the market withdrew it in 2014 due to both commercial aspects and regulatory modifications [164].

The FDA approved Platelet-Derived Growth Factor-BB (PDGF-BB) because it functions as an essential component for bone repair through its mechanisms of angiogenesis and cellular recruitment [167]. The product received FDA approval in 2005 for the treatment of periodontal defects before gaining clearance in 2015 for use in ankle and hindfoot fusion procedures under the names GEM 21S and Augment [162,163]. PDGF-BB stimulates complete tissue regeneration through dual effects on cell multiplication and matrix development [168].

Furthermore, the P-15 synthetic peptide under the brand name i-Factor (Cerapedics) received FDA approval in 2015 for cervical spine fusion procedures at the C3-C7 levels [164]. While BMP-based therapies remain the most widely studied osteoinductive agents, P-15 represents a different class of bone graft substitute. P-15 is a collagen-mimetic peptide that promotes osteoblast attachment and matrix mineralization through integrin-mediated signaling, thereby improving the early cell–matrix interface essential for graft incorporation [169]. This synthetic peptide mimics the cell-binding domain of type I collagen to promote cellular adhesion and initiate essential osteogenic signaling pathways [169]. Clinical studies demonstrate fusion rates comparable to autograft bone [170,171], although the evidence base remains smaller than for BMP-2 or PDGF-BB. Nevertheless, P-15 offers a promising alternative for spinal and periodontal applications, particularly where osteoconductive support is prioritized.

8. Natural Bioactive Agents in Bone Graft Integration

Natural bioactive compounds have emerged as promising adjuncts for enhancing bone graft integration due to their osteogenic, anti-inflammatory, and pro-angiogenic properties. Compounds such as curcumin, resveratrol, icariin, quercetin, berberine, and osthole promote osteoblast differentiation by activating key pathways, including Wnt/β-catenin, BMP/Smad, and MAPK, while simultaneously inhibiting osteoclastogenesis by downregulating RANKL-mediated signalling [172,173]. Their anti-inflammatory activity, particularly suppression of TNF-α, IL-1β, and IL-6, creates a more favorable microenvironment for graft incorporation [174,175]. Recent research highlights advancements in delivery strategies designed to overcome poor bioavailability and rapid degradation [176]. Curcumin-loaded nanoparticles, icariin-functionalized bioactive scaffolds, and resveratrol-encapsulated hydrogels have demonstrated enhanced bone regeneration, improved vascularization, and superior graft-host integration in preclinical models [177]. Moreover, hybrid systems combining natural compounds with growth factors or ceramic scaffolds show synergistic effects on osteogenesis and mineralization [178]. Despite these promising outcomes, the translational potential of natural agents remains limited by the absence of standardized dosing, variability in compound purity, and insufficient long-term clinical studies. Continued research focusing on optimized delivery systems, pharmacokinetics, and controlled clinical evaluations is necessary to advance these agents toward clinical application in bone grafting.

9. Clinical Applications and Outcomes of Pharmacological Agents in Bone Regeneration and Graft Integration

Table 3 summarizes clinical evidence on the application of rhBMP-2, rhPDGF-BB, and P-15 in bone regeneration and periodontal therapy. Across multiple case reports, rhBMP-2 has demonstrated predictable enhancement of alveolar ridge height, peri-implant bone formation, and maxillofacial defect reconstruction. Reported outcomes include vertical bone gains of 5–5.5 mm [179], improved peri-implant parameters in elderly patients [180], stable supracrestal augmentation [181], and effective reconstruction of atrophic maxillae [182]. Its utility also extends to orthopedic indications, with complete long-bone union in nonunion cases [183] and successful healing of mandibular and humeral defects [184,185,186,187].

Table 3 Summary of Clinical Applications and Outcomes of rhBMP-2, rhPDGF-BB, and P-15 in Bone Regeneration and Periodontal Treatments.

rhPDGF-BB has shown consistent benefit in periodontal and peri-implant regenerative procedures, particularly in defects requiring enhanced angiogenesis. It has enabled successful reconstruction of peri-implantitis-associated ridge defects [188] and complete Grade II furcation closure when combined with FDBA [189].

The biomimetic peptide P-15 supports periodontal regeneration by promoting cell adhesion and matrix formation, with clinical evidence demonstrating regeneration of cementum, bone, and periodontal ligament without adverse effects such as ankylosis or root resorption [190].

10. Challenges and Limitations

Despite major advances in bone grafting and adjunct pharmacological therapies, several biological and translational barriers continue to limit consistent clinical success. Patient-specific variability remains a central challenge; comorbidities such as aging, osteoporosis, diabetes, smoking, and nutritional deficiencies significantly impair osteogenesis and angiogenesis, thereby compromising graft incorporation [191]. Osteoporosis reduces bone mass and remodeling capacity, while diabetes disrupts osteoblast activity and microvascular stability, collectively increasing the risk of delayed union and graft failure [192,193]. Immune-mediated responses further hinder integration, particularly with allografts and xenografts, where inflammatory cytokine release accelerates graft degradation [194]. Although anti-inflammatory therapies may mitigate these effects, systemic administration carries risks including immunosuppression, infection, and delayed wound repair [195]. Different patient populations may respond uniquely to pharmacologic adjuncts due to variations in bone biology and systemic health. Individuals with osteoporosis often exhibit reduced osteogenic potential. They may benefit more from anabolic or anti-resorptive agents, whereas patients with diabetes typically experience impaired angiogenesis and heightened inflammatory responses, requiring therapies that target both vascular and immune pathways. These differences highlight the importance of patient-specific stratification when selecting adjunctive pharmacologic therapies for graft integration.

Insufficient vascularization represents another major limitation, particularly in large defects or regions with compromised perfusion [4]. While angiogenic agents such as VEGF and PDGF show strong experimental efficacy, their clinical translation is constrained by rapid degradation, difficulty in achieving controlled release, and potential toxicity [196]. Drug delivery itself remains problematic: systemic administration often results in low target-site concentrations and off-target effects, whereas local delivery systems face issues of burst release, uneven distribution, and material instability.

Emerging nanotechnology and scaffold-based delivery systems also present notable risks. Nanoparticles may accumulate in off-target tissues, induce oxidative stress, or trigger unintended immune responses due to unpredictable degradation and protein corona formation. Scaffold systems can fail through inconsistent degradation, insufficient mechanical strength, or inflammatory reactions to synthetic polymers. These risks, combined with challenges in reproducibility and regulatory validation, currently limit the clinical readiness of these advanced delivery platforms.

In addition to biological barriers, emerging delivery systems also face significant technical limitations. Nanoparticles may suffer from low loading efficiency, unpredictable in vivo degradation, and rapid clearance by the mononuclear phagocyte system. Scaffold-based systems often struggle with inconsistent pore interconnectivity, inadequate mechanical strength in load-bearing regions, and difficulty achieving uniform drug distribution throughout the matrix. Gene delivery platforms further present risks of uncontrolled expression, genomic integration, and variable transfection efficiency, which can compromise both safety and therapeutic reliability. These technical constraints collectively limit the reproducibility and clinical scalability of advanced delivery technologies.

A major translational limitation across pharmacologic classes is the lack of standardized dosing windows and timing protocols relative to graft placement. Many osteoinductive, angiogenic, and anti-resorptive agents display narrow therapeutic ranges, with early or excessive dosing impairing essential inflammatory and remodeling processes. Furthermore, long-term safety profiles remain incompletely characterized, particularly regarding ectopic ossification with BMPs, rebound bone turnover with Denosumab, immunomodulation-related infection risk with cytokine inhibitors, and delayed adverse events associated with gene or nanoparticle-based therapies.

Beyond biological barriers, regulatory and translational constraints limit the progression of advanced therapies. Gene therapy, nanoparticle-based systems, and bioactive scaffolds require extensive safety testing, long-term data, and substantial financial investment before clinical approval [197,198]. The absence of standardized evaluation metrics for graft performance further complicates comparison across studies. High costs also restrict access, particularly in low-resource healthcare settings.

In addition, several approved pharmacological agents carry clinically significant adverse effects. Bisphosphonates and Denosumab may impair normal bone remodeling and have been associated with osteonecrosis of the jaw and atypical fractures [199,200,201]. Similarly, BMP-based therapies may cause ectopic bone formation or inflammatory reactions when dosing is not well controlled [202]. Finally, the field lacks robust long-term clinical evidence. Most available data originate from preclinical studies or short-duration trials, limiting understanding of graft durability, mechanical stability, and long-time functional outcomes. Much of the available clinical evidence presented in this review consists only of small case series or isolated case reports, which sit at the lowest tier of the evidence hierarchy and cannot establish causality or predict long-term clinical performance. As a result, current conclusions must be interpreted cautiously until supported by larger, rigorously designed clinical trials. This evidence gap continues to delay the development of standardized clinical guidelines and the broad adoption of emerging therapies.

Regulatory experience with bone-related biologics shows that successful translation depends on clear safety profiles and tightly controlled dosing. Agents like BMP-2 and PDGF received restricted clinical indications due to concerns about dose-related adverse events. In contrast, newer options such as gene-based constructs and combination biologic platforms remain in early-phase trials due to complex regulatory pathways and unresolved long-term safety issues. These outcomes help identify which emerging therapies are realistically positioned for future clinical use.

11. Future Directions and Conclusion

Despite advancements in bone grafting techniques, challenges such as immune rejection, poor vascularization, and impaired healing in comorbid conditions persist [203]. Future research should focus on refining osteoinductive agents such as BMPs and PTH analogs, to minimize complications like ectopic bone formation, while ensuring that anti-resorptives such as bisphosphonates and Denosumab are administered with precise control to maintain balanced remodeling.

The clinical application of angiogenic therapies using VEGF, PDGF, and HIF stabilizers demands improved delivery and dosage methods before reaching clinical implementation. The development of immunomodulatory biologics that focus on TNF-α, IL-1β, and IL-6 needs additional research to establish a microenvironment that promotes regeneration instead of inflammatory responses.

Emerging technological developments featuring nanocarriers with bioengineered scaffolds and gene therapy deliver localized medications sustainably for better integration between tissues and reduced systemic effects. These platforms hold a promising clinical future when integrated with practice-based evidence from strong preclinical and clinical studies.

However, several knowledge gaps still limit the predictability of clinical translation. Optimal dosing windows, long-term safety profiles, and comparative effectiveness data remain insufficient across most drug classes. Many agents also present inherent trade-offs between efficacy and risk, particularly when used at higher or prolonged doses. Emerging combination strategies that pair osteoinductive, angiogenic, and immunomodulatory mechanisms may offer a more balanced therapeutic benefit. Still, their advantages over single-agent regimens require systematic evaluation in well-designed clinical studies. Addressing these gaps will be essential for guiding next-generation, evidence-driven bone graft therapeutics.

In conclusion, the future of bone grafting lies in developing multifunctional, personalized, and biologically active systems that not only replace bone but also actively stimulate and guide regeneration. Continued interdisciplinary collaboration and rigorous translational research will be critical for optimizing long-term outcomes and advancing the next generation of bone repair therapies.

Author Contributions

Garzain Bint e Attar conceptualized the review, performed the comprehensive literature search, curated and analyzed the data, and drafted the original manuscript. Dr. Mohd. Ashif Khan contributed to the study design, provided critical intellectual input, supervised the work, and revised the manuscript for important scientific content. Both authors reviewed, edited, and approved the final version of the manuscript.

Competing Interests

The authors have declared that no competing interests exist.

References

  1. Miron RJ. Optimized bone grafting. Periodontol 2000. 2024; 94: 143-160. [CrossRef] [Google scholar]
  2. Nashi N, Kagda FH. Current concepts of bone grafting in trauma surgery. J Clin Orthop Trauma. 2023; 43: 102231. [CrossRef] [Google scholar]
  3. Wang W, Yeung KW. Bone grafts and biomaterials substitutes for bone defect repair: A review. Bioact Mater. 2017; 2: 224-247. [CrossRef] [Google scholar]
  4. Oryan A, Alidadi S, Moshiri A, Maffulli N. Bone regenerative medicine: Classic options, novel strategies, and future directions. J Orthop Surg Res. 2014; 9: 18. [CrossRef] [Google scholar]
  5. Wiecheć E, Hybiak J, Kieda C. Introduction to transplantology. In: Stem cells and biomaterials for regenerative medicine. Academic Press; 2019. pp. 73-83. [CrossRef] [Google scholar]
  6. Land WG. Innate immunity and allograft rejection. Reference Module in Biomedical Sciences. Amsterdam, Netherlands: Elsevier; 2014. doi: 10.1016/B978-0-12-801238-3.00128-8. [CrossRef] [Google scholar]
  7. Meenu G, George VT, Manohar R, Thomas NG. Applications of xenografts in periodontal regeneration. IP Int J Periodontol Implantol. 2021; 6: 184-191. [CrossRef] [Google scholar]
  8. Vadori M, Cozzi E. Immunological challenges and therapies in xenotransplantation. Cold Spring Harb Perspect Med. 2014; 4: a015578. [CrossRef] [Google scholar]
  9. Platt JL. Graft acceptance and rejection. In: Encyclopedia of cancer. Berlin, Heidelberg: Springer; 2008. p. 1293. [CrossRef] [Google scholar]
  10. Jang HJ, Yoon JK. The role of vasculature and angiogenic strategies in bone regeneration. Biomimetics. 2024; 9: 75. [CrossRef] [Google scholar]
  11. Yang N, Liu Y. The role of the immune microenvironment in bone regeneration. Int J Med Sci. 2021; 18: 3697-3707. [CrossRef] [Google scholar]
  12. Sheen JR, Mabrouk A, Garla VV. Fracture healing overview. StatPearls [Internet]. Treasure Island, FL: StatPearls Publishing; 2023. Available from:
    https://www.ncbi.nlm.nih.gov/books/NBK551678/.
  13. Liang B, Burley G, Lin S, Shi YC. Osteoporosis pathogenesis and treatment: Existing and emerging avenues. Cell Mol Biol Lett. 2022; 27: 72. [CrossRef] [Google scholar]
  14. Pavya G, Babu NA. Effect of diabetes in osseointegration of dental implant-A review. Biomed Pharmacol J. 2015; 8: 353-358. [CrossRef] [Google scholar]
  15. Stahl A, Yang YP. Regenerative approaches for the treatment of large bone defects. Tissue Eng Part B Rev. 2021; 27: 539-547. [CrossRef] [Google scholar]
  16. Schönmeyr BH, Soares M, Avraham T, Clavin NW, Gewalli F, Mehrara BJ. Vascular endothelial growth factor inhibits bone morphogenetic protein 2 expression in rat mesenchymal stem cells. Tissue Eng Part A. 2010; 16: 653-662. [CrossRef] [Google scholar]
  17. Dimitriou R, Tsiridis E, Giannoudis PV. Current concepts of molecular aspects of bone healing. Injury. 2005; 36: 1392-1404. [CrossRef] [Google scholar]
  18. Kalish BT, Kieran MW, Puder M, Panigrahy D. The growing role of eicosanoids in tissue regeneration, repair, and wound healing. Prostaglandins Other Lipid Mediat. 2013; 104-105: 130-138. [CrossRef] [Google scholar]
  19. Molitoris KH, Huang M, Baht GS. Osteoimmunology of fracture healing. Curr Osteoporos Rep. 2024; 22: 330-339. [CrossRef] [Google scholar]
  20. Marsell R, Einhorn TA. The biology of fracture healing. Injury. 2011; 42: 551-555. [CrossRef] [Google scholar]
  21. Ozaki A, Tsunoda M, Kinoshita S, Saura R. Role of fracture hematoma and periosteum during fracture healing in rats: Interaction of fracture hematoma and the periosteum in the initial step of the healing process. J Orthop Sci. 2000; 5: 64-70. [CrossRef] [Google scholar]
  22. Gu F, Zhang K, Li J, Xie X, Wen Q, Sui Z, et al. Changes of migration, immunoregulation and osteogenic differentiation of mesenchymal stem cells in different stages of inflammation. Int J Med Sci. 2022; 19: 25-33. [CrossRef] [Google scholar]
  23. Knight MN, Hankenson KD. Mesenchymal stem cells in bone regeneration. Adv Wound Care. 2013; 2: 306-316. [CrossRef] [Google scholar]
  24. Ottewell PD. The role of osteoblasts in bone metastasis. J Bone Oncol. 2016; 5: 124-127. [CrossRef] [Google scholar]
  25. Lv N, Zhou Z, Hou M, Hong L, Li H, Qian Z, et al. Research progress of vascularization strategies of tissue-engineered bone. Front Bioeng Biotechnol. 2024; 11: 1291969. [CrossRef] [Google scholar]
  26. Hernández-García F, Fernández-Iglesias Á, Rodríguez Suárez J, Gil Peña H, López JM, Pérez RF. The crosstalk between cartilage and bone in skeletal growth. Biomedicines. 2024; 12: 2662. [CrossRef] [Google scholar]
  27. Steppe L, Megafu M, Tschaffon-Müller ME, Ignatius A, Haffner-Luntzer M. Fracture healing research: Recent insights. Bone Rep. 2023; 19: 101686. [CrossRef] [Google scholar]
  28. Šromová V, Sobola D, Kaspar P. A brief review of bone cell function and importance. Cells. 2023; 12: 2576. [CrossRef] [Google scholar]
  29. Feng X, McDonald JM. Disorders of bone remodeling. Annu Rev Pathol. 2011; 6: 121-145. [CrossRef] [Google scholar]
  30. Wang X, Qu Z, Zhao S, Luo L, Yan L. Wnt/β-catenin signaling pathway: Proteins' roles in osteoporosis and cancer diseases and the regulatory effects of natural compounds on osteoporosis. Mol Med. 2024; 30: 193. [CrossRef] [Google scholar]
  31. Heo JS, Lee SY, Lee JC. Wnt/β-catenin signaling enhances osteoblastogenic differentiation from human periodontal ligament fibroblasts. Mol Cells. 2010; 30: 449-454. [CrossRef] [Google scholar]
  32. Liu J, Xiao Q, Xiao J, Niu C, Li Y, Zhang X, et al. Wnt/β-catenin signalling: Function, biological mechanisms, and therapeutic opportunities. Signal Transduct Target Ther. 2022; 7: 3. [CrossRef] [Google scholar]
  33. Avery D, Morandini L, Sheakley LS, Shah AH, Bui L, Abaricia JO, et al. Canonical Wnt signaling enhances pro-inflammatory response to titanium by macrophages. Biomaterials. 2022; 289: 121797. [CrossRef] [Google scholar]
  34. Ducy P, Karsenty G. The family of bone morphogenetic proteins. Kidney Int. 2000; 57: 2207-2214. [CrossRef] [Google scholar]
  35. Yamaguchi A, Sakamoto K, Minamizato T, Katsube K, Nakanishi S. Regulation of osteoblast differentiation mediated by BMP, Notch, and CCN3/NOV. Jpn Dent Sci Rev. 2008; 44: 48-56. [CrossRef] [Google scholar]
  36. Argintar E, Edwards S, Delahay J. Bone morphogenetic proteins in orthopaedic trauma surgery. Injury. 2011; 42: 730-734. [CrossRef] [Google scholar]
  37. Liu Q, Peng X, Liu X, Mou X, Guo Y, Yang L, et al. Advances in the application of bone morphogenetic proteins and their derived peptides in bone defect repair. Compos B Eng. 2023; 262: 110805. [CrossRef] [Google scholar]
  38. Wada T, Nakashima T, Hiroshi N, Penninger JM. RANKL-RANK signaling in osteoclastogenesis and bone disease. Trends Mol Med. 2006; 12: 17-25. [CrossRef] [Google scholar]
  39. Tobeiha M, Moghadasian MH, Amin N, Jafarnejad S. RANKL/RANK/OPG pathway: A mechanism involved in exercise-induced bone remodeling. Biomed Res Int. 2020; 2020: 6910312. [CrossRef] [Google scholar]
  40. Reid P, Holen I. Pathophysiological roles of osteoprotegerin (OPG). Eur J Cell Biol. 2009; 88: 1-17. [CrossRef] [Google scholar]
  41. Hankenson KD, Dishowitz M, Gray C, Schenker M. Angiogenesis in bone regeneration. Injury. 2011; 42: 556-561. [CrossRef] [Google scholar]
  42. Ferrara N. Role of vascular endothelial growth factor in the regulation of angiogenesis. Kidney Int. 1999; 56: 794-814. [CrossRef] [Google scholar]
  43. Li B, Wang H, Qiu G, Su X, Wu Z. Synergistic effects of vascular endothelial growth factor on bone morphogenetic proteins induced bone formation in vivo: Influencing factors and future research directions. Biomed Res Int. 2016; 2016: 2869572. [CrossRef] [Google scholar]
  44. Tao J, Miao R, Liu G, Qiu X, Yang B, Tan X, et al. Spatiotemporal correlation between HIF-1α and bone regeneration. FASEB J. 2022; 36: e22520. [CrossRef] [Google scholar]
  45. Schipani E, Maes C, Carmeliet G, Semenza GL. Regulation of osteogenesis‐angiogenesis coupling by HIFs and VEGF. J Bone Miner Res. 2009; 24: 1347-1353. [CrossRef] [Google scholar]
  46. 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. [CrossRef] [Google scholar]
  47. Wu M, Chen G, Li YP. TGF-β and BMP signaling in osteoblast, skeletal development, and bone formation, homeostasis and disease. Bone Res. 2016; 4: 16009. [CrossRef] [Google scholar]
  48. Takahata M, Schwarz EM, Chen T, O'Keefe RJ, Awad HA. Delayed short‐course treatment with teriparatide (PTH 1-34) improves femoral allograft healing by enhancing intramembranous bone formation at the graft-host junction. J Bone Miner Res. 2012; 27: 26-37. [CrossRef] [Google scholar]
  49. Zhang J, Bai H, Bai M, Wang X, Li Z, Xue H, et al. Bisphosphonate-incorporated coatings for orthopedic implants functionalization. Mater Today Bio. 2023; 22: 100737. [CrossRef] [Google scholar]
  50. Ozturk AM, Tabak AY, Aktekin CN, Altay M, Erdemli E, Karahuseyinoglu S, et al. Alendronate enhances antibiotic-impregnated bone grafts in the treatment of osteomyelitis. Int Orthop. 2008; 32: 821-827. [CrossRef] [Google scholar]
  51. Kostenuik PJ, Nguyen HQ, McCabe J, Warmington KS, Kurahara C, Sun N, et al. Denosumab, a fully human monoclonal antibody to RANKL, inhibits bone resorption and increases BMD in knock‐in mice that express chimeric (murine/human) RANKL. J Bone Miner Res. 2009; 24: 182-195. [CrossRef] [Google scholar]
  52. Agarwala S, Vijayvargiya M. Repurposing denosumab for recalcitrant bone healing. BMJ Case Rep. 2021; 14: e238460. [CrossRef] [Google scholar]
  53. Aldhilan MM, Almahamed MH, Abdel-Wanis ME. Denosumab and metatarsal fracture healing: Potential benefits with delayed remodeling: A case report. Ann Med Surg. 2024; 86: 3786-3790. [CrossRef] [Google scholar]
  54. Hu K, Olsen BR. Vascular endothelial growth factor control mechanisms in skeletal growth and repair. Dev Dyn. 2017; 246: 227-234. [CrossRef] [Google scholar]
  55. Raica M, Cimpean AM. Platelet-derived growth factor (PDGF)/PDGF receptors (PDGFR) axis as target for antitumor and antiangiogenic therapy. Pharmaceuticals. 2010; 3: 572-599. [CrossRef] [Google scholar]
  56. Zhang J, Yao M, Xia S, Zeng F, Liu Q. Systematic and comprehensive insights into HIF-1 stabilization under normoxic conditions: Implications for cellular adaptation and therapeutic strategies in cancer. Cell Mol Biol Lett. 2025; 30: 2. [CrossRef] [Google scholar]
  57. Donneys A, Farberg AS, Tchanque-Fossuo CN, Deshpande SS, Buchman SR. Deferoxamine enhances the vascular response of bone regeneration in mandibular distraction osteogenesis. Plast Reconstr Surg. 2012; 129: 850-856. [CrossRef] [Google scholar]
  58. Jimi E, Hirata S, Osawa K, Terashita M, Kitamura C, Fukushima H. The current and future therapies of bone regeneration to repair bone defects. Int J Dent. 2012; 2012: 148261. [CrossRef] [Google scholar]
  59. Zeng M, Xu M, Wang L, Peng P, Yu K. IL-6 inhibitor tocilizumab reduces bone resorption around implants with bacterial infection during osseointegration: A pilot study in rabbits. Int J Oral Maxillofac Implants. 2024; 39: 446-454. [CrossRef] [Google scholar]
  60. Steemers E, Talbi WM, Hogervorst JM, Schoenmaker T, de Vries TJ. IL-1 receptor antagonist anakinra inhibits the effect of il-1β-mediated osteoclast formation by periodontal ligament fibroblasts. Biology. 2025; 14: 250. [CrossRef] [Google scholar]
  61. Lim SY, Bolster MB. Profile of romosozumab and its potential in the management of osteoporosis. Drug Des Devel Ther. 2017; 11: 1221-1231. [CrossRef] [Google scholar]
  62. Uemura T, Yano K, Takamatsu K, Miyashima Y, Yasuda H, Konishi S, et al. Bone healing of distal radius nonunion treated with bridge plating with bone graft substitutes in combination with systemic romosozumab administration: A case report. Jt Dis Relat Surg. 2021; 32: 526-530. [CrossRef] [Google scholar]
  63. Van Vugt TA, Arts JJ, Geurts JA. Antibiotic-loaded polymethylmethacrylate beads and spacers in treatment of orthopedic infections and the role of biofilm formation. Front Microbiol. 2019; 10: 1626. [CrossRef] [Google scholar]
  64. McConoughey SJ, Howlin RP, Wiseman J, Stoodley P, Calhoun JH. Comparing PMMA and calcium sulfate as carriers for the local delivery of antibiotics to infected surgical sites. J Biomed Mater Res B Appl Biomater. 2015; 103: 870-877. [CrossRef] [Google scholar]
  65. Halloran D, Durbano HW, Nohe A. Bone morphogenetic protein-2 in development and bone homeostasis. J Dev Biol. 2020; 8: 19. [CrossRef] [Google scholar]
  66. Wu M, Wu S, Chen W, Li YP. 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. [CrossRef] [Google scholar]
  67. Zou ML, Chen ZH, Teng YY, Liu SY, Jia Y, Zhang KW, et al. The Smad dependent TGF-β and BMP signaling pathway in bone remodeling and therapies. Front Mol Biosci. 2021; 8: 593310. [CrossRef] [Google scholar]
  68. James AW, LaChaud G, Shen J, Asatrian G, Nguyen V, Zhang X, et al. A review of the clinical side effects of bone morphogenetic protein-2. Tissue Eng Part B Rev. 2016; 22: 284-297. [CrossRef] [Google scholar]
  69. Nourbakhsh A, Qureshi R, Heppner C, Shen F, Hassanzadeh H. The use of bone morphogenic protein in cervical spine fusions: A literature review. Semin Spine Surg. 2016; 28: 245-250. [CrossRef] [Google scholar]
  70. Yamashita J, McCauley LK. Effects of intermittent administration of parathyroid hormone and parathyroid hormone related protein on fracture healing: A narrative review of animal and human studies. JBMR Plus. 2019; 3: e10250. [CrossRef] [Google scholar]
  71. Marin F, Ma YL. Teriparatide. In: Osteoporotic fracture and systemic skeletal disorders: Mechanism, assessment, and treatment. Singapore: Springer; 2021. pp. 339-359. [CrossRef] [Google scholar]
  72. Takahata M, Awad HA, O’Keefe RJ, Bukata SV, Schwarz EM. Endogenous tissue engineering: PTH therapy for skeletal repair. Cell Tissue Res. 2012; 347: 545-552. [CrossRef] [Google scholar]
  73. Chan HL, McCauley L. Parathyroid hormone applications in the craniofacial skeleton. J Dent Res. 2013; 92: 18-25. [CrossRef] [Google scholar]
  74. Lou S, Lv H, Li Z, Tang P, Wang Y. Parathyroid hormone analogues for fracture healing: Protocol for a systematic review and meta-analysis of randomised controlled trials. BMJ Open. 2018; 8: e019291. [CrossRef] [Google scholar]
  75. Rocha T, Cavalcanti AS, Leal AC, Dias RB, da Costa RS, de Oliveira Ribeiro G, et al. PTH 1-34 improves devitalized allogenic bone graft healing in a murine femoral critical size defect. Injury. 2021; 52: S3-S12. [CrossRef] [Google scholar]
  76. Leder BZ. Parathyroid hormone and parathyroid hormone-related protein analogs in osteoporosis therapy. Curr Osteoporos Rep. 2017; 15: 110-119. [CrossRef] [Google scholar]
  77. Farrell KB, Karpeisky A, Thamm DH, Zinnen S. Bisphosphonate conjugation for bone specific drug targeting. Bone Rep. 2018; 9: 47-60. [CrossRef] [Google scholar]
  78. Itzstein C, Coxon FP, Rogers MJ. The regulation of osteoclast function and bone resorption by small GTPases. Small GTPases. 2011; 2: 117-130. [CrossRef] [Google scholar]
  79. Jensen PR, Andersen TL, Chavassieux P, Roux JP, Delaisse JM. Bisphosphonates impair the onset of bone formation at remodeling sites. Bone. 2021; 145: 115850. [CrossRef] [Google scholar]
  80. Hong C, Quach A, Lin L, Olson J, Kwon T, Bezouglaia O, et al. Local vs. systemic administration of bisphosphonates in rat cleft bone graft: A comparative study. PLoS One. 2018; 13: e0190901. [CrossRef] [Google scholar]
  81. Maethungkul R, Sangsin A, Chattipakorn N, Chattipakorn SC. Exploring the multifaceted impact of bisphosphonates on bone graft integration: Transitioning from in vivo insights to clinical applications. Arch Toxikol. 2025; 99: 2157-2178. [CrossRef] [Google scholar]
  82. Miyazaki T, Tokimura F, Tanaka S. A review of denosumab for the treatment of osteoporosis. Patient Prefer Adherence. 2014; 8: 463-471. [CrossRef] [Google scholar]
  83. Cheng X, Tian W, Yang J, Wang J, Zhang Y. Engineering approaches to manipulate osteoclast behavior for bone regeneration. Mater Today Bio. 2024; 26: 101043. [CrossRef] [Google scholar]
  84. Kumar S, Wang M, Kim AS, Center JR, McDonald MM, Girgis CM. Denosumab discontinuation in the clinic: Implications of rebound bone turnover and emerging strategies to prevent bone loss and fractures. J Bone Miner Res. 2025; 40: 1017-1034. [CrossRef] [Google scholar]
  85. Costa AG, Lewiecki EM, Bilezikian JP. Denosumab. Primer on the metabolic bone diseases and disorders of mineral metabolism. 9th ed. Hoboken, NJ: John Wiley & Sons, Inc.; 2018. [CrossRef] [Google scholar]
  86. Duffy AM, Bouchier-Hayes DJ, Harmey JH. Vascular endothelial growth factor (VEGF) and its role in non-endothelial cells: Autocrine signalling by VEGF. Madame Curie Bioscience Database [Internet]. Austin, TX: Landes Bioscience; 2013. Available from: https://www.ncbi.nlm.nih.gov/books/NBK6482/.
  87. Street J, Bao M, deGuzman L, Bunting S, Peale Jr FV, Ferrara N, et al. Vascular endothelial growth factor stimulates bone repair by promoting angiogenesis and bone turnover. Proc Natl Acad Sci. 2002; 99: 9656-9661. [CrossRef] [Google scholar]
  88. Peng H, Wright V, Usas A, Gearhart B, Shen HC, Cummins J, et al. Synergistic enhancement of bone formation and healing by stem cell-expressed VEGF and bone morphogenetic protein-4. J Clin Invest. 2002; 110: 751-759. [CrossRef] [Google scholar]
  89. Marti HH. Vascular endothelial growth factor. Madame Curie Bioscience Database [Internet]. Austin, TX: Landes Bioscience; 2000-2013. Available from:
    https://www.ncbi.nlm.nih.gov/books/NBK6149/.
  90. Liu G, Wei J, Xiao W, Xie W, Ru Q, Chen L, et al. Insights into the notch signaling pathway in degenerative musculoskeletal disorders: Mechanisms and perspectives. Biomed Pharmacother. 2023; 169: 115884. [CrossRef] [Google scholar]
  91. Sewduth R, Santoro MM. “Decoding” angiogenesis: New facets controlling endothelial cell behavior. Front Physiol. 2016; 7: 306. [CrossRef] [Google scholar]
  92. Su P, Tian Y, Yang C, Ma X, Wang X, Pei J, et al. Mesenchymal stem cell migration during bone formation and bone diseases therapy. Int J Mol Sci. 2018; 19: 2343. [CrossRef] [Google scholar]
  93. Liu M, Liu Y, Luo F. The role and mechanism of platelet-rich fibrin in alveolar bone regeneration. Biomed Pharmacother. 2023; 168: 115795. [CrossRef] [Google scholar]
  94. Ban HS, Uno M, Nakamura H. Suppression of hypoxia-induced HIF-1α accumulation by VEGFR inhibitors: Different profiles of AAL993 versus SU5416 and KRN633. Cancer Lett. 2010; 296: 17-26. [CrossRef] [Google scholar]
  95. You J, Liu M, Li M, Zhai S, Quni S, Zhang L, et al. The role of HIF-1α in bone regeneration: A new direction and challenge in bone tissue engineering. Int J Mol Sci. 2023; 24: 8029. [CrossRef] [Google scholar]
  96. Chen W, Wu P, Yu F, Luo G, Qing L, Tang J. HIF-1α regulates bone homeostasis and angiogenesis, participating in the occurrence of bone metabolic diseases. Cells. 2022; 11: 3552. [CrossRef] [Google scholar]
  97. Shen H, Ma Y, Qiao Y, Zhang C, Chen J, Zhang R. Application of deferoxamine in tissue regeneration attributed to promoted angiogenesis. Molecules. 2024; 29: 2050. [CrossRef] [Google scholar]
  98. Saber S, Abdelhady R, Elhemely MA, Elmorsy EA, Hamad RS, Abdel-Reheim MA, et al. Nanoscale systems for local activation of hypoxia-inducible factor-1 alpha: A new approach in diabetic wound management. Int J Nanomedicine. 2024; 19: 13735-13762. [CrossRef] [Google scholar]
  99. Azuma Y, Kaji K, Katogi R, Takeshita S, Kudo A. Tumor necrosis factor-α induces differentiation of and bone resorption by osteoclasts. J Biol Chem. 2000; 275: 4858-4864. [CrossRef] [Google scholar]
  100. Steeve KT, Marc P, Sandrine T, Dominique H, Yannick F. IL-6, RANKL, TNF-alpha/IL-1: Interrelations in bone resorption pathophysiology. Cytokine Growth Factor Rev. 2004; 15: 49-60. [CrossRef] [Google scholar]
  101. Gilbert L, He X, Farmer P, Boden S, Kozlowski M, Rubin J, et al. Inhibition of osteoblast differentiation by tumor necrosis factor-α. Endocrinology. 2000; 141: 3956-3964. [CrossRef] [Google scholar]
  102. Vitale RF, Ribeiro FAQ. The role of Tumor Necrosis Factor-Alpha (TNF-α) in bone resorption present in middle ear cholesteatoma. Braz J Otorhinolaryngol. 2007; 73: 117-121. [CrossRef] [Google scholar]
  103. Mountziaris PM, Mikos AG. Modulation of the inflammatory response for enhanced bone tissue regeneration. Tissue Eng Part B Rev. 2008; 14: 179-186. [CrossRef] [Google scholar]
  104. Gerriets V, Goyal A, Khaddour K. Tumor necrosis factor inhibitors. StatPearls [Internet]. Treasure Island, FL: StatPearls Publishing; 2023. Available from: https://www.ncbi.nlm.nih.gov/books/NBK482425/.
  105. Dogra S, Khullar G. Tumor necrosis factor-α antagonists: Side effects and their management. Indian J Dermatol Venereol Leprol. 2013; 79: 35-46. [CrossRef] [Google scholar]
  106. Braun J, Brandt J, Listing J, Zink A, Alten R, Golder W, et al. Treatment of active ankylosing spondylitis with infliximab: A randomised controlled multicentre trial. Lancet. 2002; 359: 1187-1193. [CrossRef] [Google scholar]
  107. King N. Biologic therapies in ankylosing spondylitis: Efficacy and safety profile. Int J Clin Rheumatol. 2025; 20: 380-382. Available from:
    https://www.openaccessjournals.com/articles/biologic-therapies-in-ankylosing-spondylitis-efficacy-and-safety-profile.pdf.
  108. Dawalibi A, Alosaimi AA, Mohammad KS. Balancing the scales: The dual role of interleukins in bone metastatic microenvironments. Int J Mol Sci. 2024; 25: 8163. [CrossRef] [Google scholar]
  109. McGregor NE, Murat M, Elango J, Poulton IJ, Walker EC, Crimeen-Irwin B, et al. IL-6 exhibits both cis-and trans-signaling in osteocytes and osteoblasts, but only trans-signaling promotes bone formation and osteoclastogenesis. J Biol Chem. 2019; 294: 7850-7863. [CrossRef] [Google scholar]
  110. Lorenzo JA. The role of interleukin-6 in bone. J Endocr Soc. 2020; 4: bvaa112. [CrossRef] [Google scholar]
  111. Briot K, Rouanet S, Schaeverbeke T, Etchepare F, Gaudin P, Perdriger A, et al. The effect of tocilizumab on bone mineral density, serum levels of dickkopf-1 and bone remodeling markers in patients with rheumatoid arthritis. Joint Bone Spine. 2015; 82: 109-115. [CrossRef] [Google scholar]
  112. Hashizume M, Mihara M. The roles of interleukin-6 in the pathogenesis of rheumatoid arthritis. Arthritis. 2011; 2011: 765624. [CrossRef] [Google scholar]
  113. Narazaki M, Kishimoto T. Current status and prospects of IL-6–targeting therapy. Expert Rev Clin Pharmacol. 2022; 15: 575-592. [CrossRef] [Google scholar]
  114. Cheng R, Wu Z, Li M, Shao M, Hu T. Interleukin-1β is a potential therapeutic target for periodontitis: A narrative review. Int J Oral Sci. 2020; 12: 2. [CrossRef] [Google scholar]
  115. Anakinra. In: Meyler's side effects of drugs: The international encyclopedia of adverse drug reactions and interactions. 15th ed. Amsterdam, Netherlands: Elsevier; 2006. pp. 215-216. doi: 10.1016/B0-44-451005-2/00866-4. [CrossRef] [Google scholar]
  116. Abramson SB, Amin A. Blocking the effects of IL-1 in rheumatoid arthritis protects bone and cartilage. Rheumatology. 2002; 41: 972-980. [CrossRef] [Google scholar]
  117. Aditya S, Rattan A. Sclerostin inhibition: A novel target for the treatment of postmenopausal osteoporosis. J Midlife Health. 2021; 12: 267-275. [CrossRef] [Google scholar]
  118. Cursaru A, Cursaru R, Iordache S, Costache MA, Cretu BS, Serban B, et al. The use of dissolvable synthetic calcium impregnated with antibiotic in osteoarticular infection in patients with diabetes. Life. 2024; 14: 1335. [CrossRef] [Google scholar]
  119. Doub JB, Bork JT, Heil E, Stafford K, Banoub M, Karwowski JK, et al. Effectiveness and safety of biodegradable calcium sulfate antibiotic beads as adjuvant therapy in vascular graft infections. Open Forum Infect Dis. 2021; 8: ofaa650. [CrossRef] [Google scholar]
  120. Howlin RP, Brayford MJ, Webb JS, Cooper JJ, Aiken SS, Stoodley P. Antibiotic-loaded synthetic calcium sulfate beads for prevention of bacterial colonization and biofilm formation in periprosthetic infections. Antimicrob Agents Chemother. 2015; 59: 111-120. [CrossRef] [Google scholar]
  121. Andreacchio A, Alberghina F, Paonessa M, Cravino M, De Rosa V, Canavese F. Tobramycin-impregnated calcium sulfate pellets for the treatment of chronic osteomyelitis in children and adolescents. J Pediatr Orthop B. 2019; 28: 189-195. [CrossRef] [Google scholar]
  122. Olthof MG, Lu L, Tryfonidou MA, Loozen LD, Pouran B, Yaszemski MJ, et al. The osteoinductive effect of controlled bone morphogenic protein 2 release is location dependent. Tissue Eng Part A. 2019; 25: 193-202. [CrossRef] [Google scholar]
  123. Misof BM, Fratzl-Zelman N, Paschalis EP, Roschger P, Klaushofer K. Long-term safety of antiresorptive treatment: Bone material, matrix and mineralization aspects. Bonekey Rep. 2015; 4: 634. [CrossRef] [Google scholar]
  124. Chu H, Wang Y. Therapeutic angiogenesis: Controlled delivery of angiogenic factors. Ther Deliv. 2012; 3: 693-714. [CrossRef] [Google scholar]
  125. Sharma Y, Arora M, Bala K. The potential of immunomodulators in shaping the future of healthcare. Discov Med. 2024; 1: 37. [CrossRef] [Google scholar]
  126. Hu L, Chen W, Qian A, Li YP. Wnt/β-catenin signaling components and mechanisms in bone formation, homeostasis, and disease. Bone Res. 2024; 12: 39. [CrossRef] [Google scholar]
  127. Shen H, Goldstein DR. IL-6 and TNF-α synergistically inhibit allograft acceptance. J Am Soc Nephrol. 2009; 20: 1032-1040. [CrossRef] [Google scholar]
  128. Shirazi AN, Vadlapatla R, Koomer A, Nguyen A, Khoury V, Parang K. Peptide-based inorganic nanoparticles as efficient intracellular delivery systems. Pharmaceutics. 2025; 17: 1123. [CrossRef] [Google scholar]
  129. Farjaminejad S, Farjaminejad R, Garcia-Godoy F. Nanoparticles in bone regeneration: A narrative review of current advances and future directions in tissue engineering. J Funct Biomater. 2024; 15: 241. [CrossRef] [Google scholar]
  130. Cheng H, Chawla A, Yang Y, Li Y, Zhang J, Jang HL, et al. Development of nanomaterials for bone-targeted drug delivery. Drug Discov Today. 2017; 22: 1336-1350. [CrossRef] [Google scholar]
  131. Zhang G, Zhen C, Yang J, Wang J, Wang S, Fang Y, et al. Recent advances of nanoparticles on bone tissue engineering and bone cells. Nanoscale Adv. 2024; 6: 1957-1973. [CrossRef] [Google scholar]
  132. Yi M, Nie Y, Zhang C, Shen B. Application of mesoporous silica nanoparticle-chitosan-loaded BMP-2 in the repair of bone defect in chronic osteomyelitis. J Immunol Res. 2022; 2022: 4450196. [CrossRef] [Google scholar]
  133. Xu C, Cao Y, Lei C, Li Z, Kumeria T, Meka AK, et al. Polymer-mesoporous silica nanoparticle core-shell nanofibers as a dual-drug-delivery system for guided tissue regeneration. ACS Appl Nano Mater. 2020; 3: 1457-1467. [CrossRef] [Google scholar]
  134. Shan J, Yu Y, Liu X, Chai Y, Wang X, Wen G. Recent advances of chitosan-based composite hydrogel materials in application of bone tissue engineering. Heliyon. 2024; 10: e37431. [CrossRef] [Google scholar]
  135. Jafari S, Mahyad B, Hashemzadeh H, Janfaza S, Gholikhani T, Tayebi L. Biomedical applications of TiO2 nanostructures: Recent advances. Int J Nanomed. 2020; 15: 3447-3470. [CrossRef] [Google scholar]
  136. Park J, Cimpean A, Tesler AB, Mazare A. Anodic TiO2 nanotubes: Tailoring osteoinduction via drug delivery. Nanomaterials. 2021; 11: 2359. [CrossRef] [Google scholar]
  137. Iyer SR, Xu S, Stains JP, Bennett CH, Lovering RM. Superparamagnetic iron oxide nanoparticles in musculoskeletal biology. Tissue Eng Part B Rev. 2017; 23: 373-385. [CrossRef] [Google scholar]
  138. Suciu M, Ionescu CM, Ciorita A, Tripon SC, Nica D, Al-Salami H, et al. Applications of superparamagnetic iron oxide nanoparticles in drug and therapeutic delivery, and biotechnological advancements. Beilstein J Nanotechnol. 2020; 11: 1092-1109. [CrossRef] [Google scholar]
  139. Shi Y, Hu H. AI accelerated discovery of self-assembling peptides. Biomater Transl. 2023; 4: 291-293. [Google scholar]
  140. Anil S, Al-Sulaimani AF, Beeran AE, Chalisserry EP, Varma HP, Al Amri MD. Drug delivery systems in bone regeneration and implant dentistry. In: Current concepts in dental implantology. London, UK: IntechOpen; 2015. pp. 239-265. [CrossRef] [Google scholar]
  141. Alex Y, Vincent S, Divakaran N, Uthappa UT, Srinivasan P, Mubarak S, et al. Pioneering bone regeneration: A review of cutting-edge scaffolds in tissue engineering. Bioprinting. 2024; 43: e00364. [CrossRef] [Google scholar]
  142. Brachet A, Bełżek A, Furtak D, Geworgjan Z, Tulej D, Kulczycka K, et al. Application of 3D printing in bone grafts. Cells. 2023; 12: 859. [CrossRef] [Google scholar]
  143. Shineh G, Patel K, Mobaraki M, Tayebi L. Functional approaches in promoting vascularization and angiogenesis in bone critical-sized defects via delivery of cells, growth factors, drugs, and particles. J Funct Biomater. 2023; 14: 99. [CrossRef] [Google scholar]
  144. Alsaab HO, Alharbi FD, Alhibs AS, Alanazi NB, Alshehri BY, Saleh MA, et al. PLGA-based nanomedicine: History of advancement and development in clinical applications of multiple diseases. Pharmaceutics. 2022; 14: 2728. [CrossRef] [Google scholar]
  145. Choe G, Lee M, Oh S, Seok JM, Kim J, Im S, et al. Three-dimensional bioprinting of mesenchymal stem cells using an osteoinductive bioink containing alginate and BMP-2-loaded PLGA nanoparticles for bone tissue engineering. Biomater Adv. 2022; 136: 212789. [CrossRef] [Google scholar]
  146. Bektas C, Mao Y. Hydrogel microparticles for bone regeneration. Gels. 2023; 10: 28. [CrossRef] [Google scholar]
  147. Lv Z, Ji Y, Wen G, Liang X, Zhang K, Zhang W. Structure-optimized and microenvironment-inspired nanocomposite biomaterials in bone tissue engineering. Burns Trauma. 2024; 12: tkae036. [CrossRef] [Google scholar]
  148. Liu D, Gao S, Xu J, Xia W, Chen S, Li B. Advancing musculoskeletal organoids research: Overcoming barriers to reduce animal model dependency. Organoid Res. 2025; 1: 025280024. [CrossRef] [Google scholar]
  149. Jin Y, Chen Q, Gong L, Zheng X, Chen L, Li B, et al. Organoids: Applications and challenges of advanced hydrogels in tissue systems. Organoid Res. 2025; 1: 8262. [CrossRef] [Google scholar]
  150. Evans CH. Gene therapy for bone healing. Expert Rev Mol Med. 2010; 12: e18. [CrossRef] [Google scholar]
  151. Park SY, Kim KH, Kim S, Lee YM, Seol YJ. BMP-2 gene delivery-based bone regeneration in dentistry. Pharmaceutics. 2019; 11: 393. [CrossRef] [Google scholar]
  152. De la Vega RE, Atasoy-Zeybek A, Panos JA, Van Griensven M, Evans CH, Balmayor ER. Gene therapy for bone healing: Lessons learned and new approaches. Transl Res. 2021; 236: 1-16. [CrossRef] [Google scholar]
  153. Gallo MC, Elias A, Reynolds J, Ball JR, Lieberman JR. Regional gene therapy for bone tissue engineering: A current concepts review. Bioengineering. 2025; 12: 120. [CrossRef] [Google scholar]
  154. Lee CS, Bishop ES, Zhang R, Yu X, Farina EM, Yan S, et al. Adenovirus-mediated gene delivery: Potential applications for gene and cell-based therapies in the new era of personalized medicine. Genes Dis. 2017; 4: 43-63. [CrossRef] [Google scholar]
  155. Taghdiri M, Mussolino C. Viral and non-viral systems to deliver gene therapeutics to clinical targets. Int J Mol Sci. 2024; 25: 7333. [CrossRef] [Google scholar]
  156. Butt MH, Zaman M, Ahmad A, Khan R, Mallhi TH, Hasan MM, et al. Appraisal for the potential of viral and nonviral vectors in gene therapy: A review. Genes. 2022; 13: 1370. [CrossRef] [Google scholar]
  157. Park J, Lutz R, Felszeghy E, Wiltfang J, Nkenke E, Neukam FW, et al. The effect on bone regeneration of a liposomal vector to deliver BMP-2 gene to bone grafts in peri-implant bone defects. Biomaterials. 2007; 28: 2772-2782. [CrossRef] [Google scholar]
  158. Bez M, Pelled G, Gazit D. BMP gene delivery for skeletal tissue regeneration. Bone. 2020; 137: 115449. [CrossRef] [Google scholar]
  159. Watson-Levings RS, Palmer GD, Levings PP, Dacanay EA, Evans CH, Ghivizzani SC. Gene therapy in orthopaedics: Progress and challenges in pre-clinical development and translation. Front Bioeng Biotechnol. 2022; 10: 901317. [CrossRef] [Google scholar]
  160. Xia Z, Wang Q. The emergence of AI tools in scientific writing and research. Biomater Transl. 2023; 4: 1-2. [Google scholar]
  161. Skovrlj B, Marquez-Lara A, Guzman JZ, Qureshi SA. A review of the current published spinal literature regarding bone morphogenetic protein-2: An insight into potential bias. Curr Rev Musculoskelet Med. 2014; 7: 182-188. [CrossRef] [Google scholar]
  162. Kaigler D, Avila G, Wisner-Lynch L, Nevins ML, Nevins M, Rasperini G, et al. Platelet-derived growth factor applications in periodontal and peri-implant bone regeneration. Expert Opin Biol Ther. 2011; 11: 375-385. [CrossRef] [Google scholar]
  163. Govoni M, Vivarelli L, Mazzotta A, Stagni C, Maso A, Dallari D. Commercial bone grafts claimed as an alternative to autografts: Current trends for clinical applications in orthopaedics. Materials. 2021; 14: 3290. [CrossRef] [Google scholar]
  164. Gillman CE, Jayasuriya AC. FDA-approved bone grafts and bone graft substitute devices in bone regeneration. Mater Sci Eng C. 2021; 130: 112466. [CrossRef] [Google scholar]
  165. McKay WF, Peckham SM, Badura JM. A comprehensive clinical review of recombinant human bone morphogenetic protein-2 (INFUSE® Bone Graft). Int Orthop. 2007; 31: 729-734. [CrossRef] [Google scholar]
  166. Kathami N, Moreno-Vicente C, Martín P, Vergara-Arce JA, Ruiz-Hernández R, Gerovska D, et al. rhBMP-2 induces terminal differentiation of human bone marrow mesenchymal stromal cells only by synergizing with other signals. Stem Cell Res Ther. 2024; 15: 124. [CrossRef] [Google scholar]
  167. Baldini N, Cenni E, Ciapetti G, Granchi D, Savarino L. Bone repair and regeneration. In: Bone repair biomaterials. Cambridge, UK: Woodhead Publishing; 2009. pp. 69-105. [CrossRef] [Google scholar]
  168. Takayama T, Imamura K, Yamano S. Growth factor delivery using a collagen membrane for bone tissue regeneration. Biomolecules. 2023; 13: 809. [CrossRef] [Google scholar]
  169. Cheng CT, Vyas PS, McClain IV EJ, Hoelen TC, Arts JJ, McLaughlin C, et al. The osteogenic peptide P-15 for bone regeneration: A narrative review of the evidence for a mechanism of action. Bioengineering. 2024; 11: 599. [CrossRef] [Google scholar]
  170. Hasan S, Al-Jamal M, Miller A, Higginbotham DO, Cavazos DR, Waheed M, et al. Efficacy and outcome measurement of iFactor/ABM/P-15 in lumbar spine surgery: A systematic review. Global Spine J. 2024; 14: 1422-1433. [CrossRef] [Google scholar]
  171. Arnold PM, Vaccaro AR, Sasso RC, Goulet B, Fehlings MG, Heary RF, et al. Six-year follow-up of a randomized controlled trial of i-FACTOR peptide-enhanced bone graft versus local autograft in single-level anterior cervical discectomy and fusion. Neurosurgery. 2023; 92: 725-733. [CrossRef] [Google scholar]
  172. Ouyang X, Ma Q, Zhou C, Tang J, Li M, Qing J, et al. Natural bioactive products in the regulation of bone metabolism and regeneration. Front Pharmacol. 2025; 16: 1683279. [CrossRef] [Google scholar]
  173. Song C, Zhao C. The role of active constituents of in traditional Chinese medicine for primary osteoporosis: A mechanistic review. Front Endocrinol. 2025; 16: 1647984. [CrossRef] [Google scholar]
  174. Li H, Ma L, Zhu N, Liang X, Liu K, Fu X, et al. Natural-origin bioadhesive hydrogel with dual antioxidative and immunoregulatory properties for enhanced angiogenesis and wound healing. Bioact Mater. 2025; 53: 507-521. [CrossRef] [Google scholar]
  175. Liu L, Chen H, Zhao X, Han Q, Xu Y, Liu Y, et al. Advances in the application and research of biomaterials in promoting bone repair and regeneration through immune modulation. Mater Today Bio. 2025; 30: 101410. [CrossRef] [Google scholar]
  176. Hashmi AR, Sekar M, Zahra F, Molugulu N, Wong LS. Advanced drug delivery strategies to overcome solubility and permeability challenges: Driving biopharmaceutical advancements toward commercial success. ACS Omega. 2025; 10: 40769-40792. [CrossRef] [Google scholar]
  177. Astaneh ME, Noori F, Fereydouni N. Curcumin-loaded scaffolds in bone regeneration. Heliyon. 2024; 10: e32566. [CrossRef] [Google scholar]
  178. Saberi A, Kouhjani M, Mohammadi M, Hosta-Rigau L. Novel scaffold platforms for simultaneous induction osteogenesis and angiogenesis in bone tissue engineering: A cutting-edge approach. J Nanobiotechnol. 2023; 21: 351. [CrossRef] [Google scholar]
  179. Katanec D, Granić M, Gabrić Pandurić D, Majstorović M, Trampuš Z. Use of recombinant human bone morphogenetic protein (rhBMP2) in bilateral alveolar ridge augmentation: Case report. Coll Antropol. 2014; 38: 325-330. [Google scholar]
  180. Rotenberg SA, Tatakis DN. Recombinant human bone morphogenetic protein-2 for peri-implant bone regeneration: A case report. J Periodontol. 2011; 82: 1212-1218. [CrossRef] [Google scholar]
  181. Sclar AG, Best SP. The combined use of rhBMP-2/ACS, autogenous bone graft, a bovine bone mineral biomaterial, platelet-rich plasma, and guided bone regeneration at nonsubmerged implant placement for supracrestal bone augmentation. A case report. Int J Oral Maxillofac Implants. 2013; 28: e272-e276. [CrossRef] [Google scholar]
  182. Zétola AL, Verbicaro T, Littieri S, Larson R, Giovanini AF, Deliberador TM. Recombinant human bone morphogenetic protein type 2 in the reconstruction of atrophic maxilla: Case report with long-term follow-up. J Indian Soc Periodontol. 2014; 18: 781-785. [CrossRef] [Google scholar]
  183. Choi W, Kim BS, Cho WT, Lim EJ, Choi JS, Ryu YK, et al. 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. [CrossRef] [Google scholar]
  184. Kaushik M, Srivastava A, Khattri S. Successful integration of recombinant human bone morphogenetic protein-2 (rhBMP-2)-coated dental implant: A case report. Cureus. 2025; 17: e79281. [CrossRef] [Google scholar]
  185. Cicciu M, Herford AS, Stoffella E, Cervino G, Cicciu D. Protein-signaled guided bone regeneration using titanium mesh and Rh-BMP2 in oral surgery: A case report involving left mandibular reconstruction after tumor resection. Open Dent J. 2012; 6: 51-55. [CrossRef] [Google scholar]
  186. Alhalabi L, Zenati M. Case report: The effect of bone morphogenetic proteins (BMP) in comparison with a xenograft in the management of mandibular bone defects. F1000Res. 2024; 13: 20. [CrossRef] [Google scholar]
  187. Tapio RM, Zhu K, Frolov D, Dolgov V, Schmitz M. Open reduction and internal fixation with bone morphogenetic protein-2 for correction of nonunion humeral shaft fracture with pseudoarthrosis in the geriatric population: A case report. J Orthop Case Rep. 2024; 14: 124-128. [CrossRef] [Google scholar]
  188. Mandelaris GA, DeGroot B. Bone construction surgery: A case report using recombinant human platelet‐derived growth factor‐BB. Clin Adv Periodontics. 2022; 12: 262-269. [CrossRef] [Google scholar]
  189. Almutairi AS. Recombinant human PDGF-BB in combination with mineralized freeze-dried bone allograft in the treatment of Grade II furcation involvement: A case report. Asian J Oral Health Allied Sci. 2021; 11: 4. [CrossRef] [Google scholar]
  190. Yukna RA, Salinas TJ, Carr RF. Periodontal regeneration following use of ABM/P-15: A case report. Int J Periodontics Restorative Dent. 2002; 22: 146-155. [Google scholar]
  191. Kant K, Kshirsagar J, Pasha Z, Deshmukh KS, Kale PP. Advances in bone grafting techniques for dental implants: A comprehensive review. IP Int J Periodontol Implantol. 2023; 8: 195-199. [CrossRef] [Google scholar]
  192. Jiao H, Xiao E, Graves DT. Diabetes and its effect on bone and fracture healing. Curr Osteoporos Rep. 2015; 13: 327-335. [CrossRef] [Google scholar]
  193. Rosen CJ. The epidemiology and pathogenesis of osteoporosis. Endotext [Internet]. South Dartmouth, MA: MDText.com, Inc.; 2000. Available from:
    https://www.ncbi.nlm.nih.gov/books/NBK279134/.
  194. Janeway Jr CA, Travers P, Walport M, Shlomchik MJ. Responses to alloantigens and transplant rejection. Immunobiology: The immune system in health and disease. 5th ed. New York, NY: Garland Science; 2001. [Google scholar]
  195. Henrickson SE, Ruffner MA, Kwan M. Unintended immunological consequences of biologic therapy. Curr Allergy Asthma Rep. 2016; 16: 46. [CrossRef] [Google scholar]
  196. Gacche RN, Meshram RJ. Angiogenic factors as potential drug target: Efficacy and limitations of anti-angiogenic therapy. Biochim Biophys Acta Rev Cancer. 2014; 1846: 161-179. [CrossRef] [Google scholar]
  197. Haleem A, Javaid M, Singh RP, Rab S, Suman R. Applications of nanotechnology in medical field: A brief review. Glob Health J. 2023; 7: 70-77. [CrossRef] [Google scholar]
  198. Ho-Shui-Ling A, Bolander J, Rustom LE, Johnson AW, Luyten FP, Picart C. Bone regeneration strategies: Engineered scaffolds, bioactive molecules and stem cells current stage and future perspectives. Biomaterials. 2018; 180: 143-162. [CrossRef] [Google scholar]
  199. Otto S, Pautke C, Van den Wyngaert T, Niepel D, Schiødt M. Medication-related osteonecrosis of the jaw: Prevention, diagnosis and management in patients with cancer and bone metastases. Cancer Treat Rev. 2018; 69: 177-187. [CrossRef] [Google scholar]
  200. Cadieux B, Coleman R, Jafarinasabian P, Lipton A, Orlowski RZ, Saad F, et al. Experience with denosumab (XGEVA®) for prevention of skeletal-related events in the 10 years after approval. J Bone Oncol. 2022; 33: 100416. [CrossRef] [Google scholar]
  201. Lončar Brzak B, Horvat Aleksijević L, Vindiš E, Kordić I, Granić M, Vidović Juras D, et al. Osteonecrosis of the jaw. Dent J. 2023; 11: 23. [CrossRef] [Google scholar]
  202. Sampath TK, Vukicevic S. Biology of bone morphogenetic protein in bone repair and regeneration: A role for autologous blood coagulum as carrier. Bone. 2020; 141: 115602. [CrossRef] [Google scholar]
  203. Schmidt AH. Autologous bone graft: Is it still the gold standard? Injury. 2021; 52: S18-S22. [CrossRef] [Google scholar]
Journal Metrics
2024
CiteScore SJR SNIP
0.40.1310.065
Newsletter
Download PDF Download Full-Text XML Download Citation
0 0

TOP