You are on page 1of 17

REVIEW

For reprint orders, please contact: reprints@futuremedicine.com

Administration of growth factors for bone regeneration


Growth factors (GFs) such as BMPs, FGFs, VEGFs and IGFs have signicant impacts on osteoblast behavior, and thus have been widely utilized for bone tissue regeneration. Recently, securing biological stability for a sustainable and controllable release to the target tissue has been a challenge to practical applications. This challenge has been addressed to some degree with the development of appropriate carrier materials and delivery systems. This review highlights the importance and roles of those GFs, as well as their proper administration for targeting bone regeneration. Additionally, the invitro and invivo performance of those GFs with or without the use of carrier systems in the repair and regeneration of bone tissue is systematically addressed. Moreover, some recent advances in the utility of the GFs, such as using fusion technology, are also reviewed.
KEYWORDS: BMP n bone regeneration n carrier material n delivery system n FGF nfusion protein n growth factor n IGF n VEGF n wound healing

Growth factors (GFs) have been found in all tissues and are known to stimulate cellular growth, proliferation, migration and differentiation. Many GFs have been proposed for bone tissue engineering applications. GFs are usually stored in the extracellular matrix (ECM); however, they are released from the ECM after injury and then contribute to the bone repair process. During bone repair, cells of the bone environment, such as bone marrow stromal cells, endothelial cells, broblasts, inammatory cells and osteoblasts, produce GFs [1] . Among others, periosteum, a dense irregular connective tissue lining the outer surface of bone, is considered important as this contains a population of progenitor cells that directly differentiate into osteoblasts. Many GFs, including BMPs, VEGFs, FGFs, IGFs and PDGFs, have been engaged in directing and stimulating periosteal progenitors to osteoblastic differentiation during the bone-healing process[210] . The active roles of GFs with respect to the cells mentioned above have been comprehensively identied both invitro and invivo. GFs may act in an autocrine fashion and inuence cells of the same class or in a paracrine fashion and affect different cells. Accordingly, a variety of GFs have been widely used for bone tissue engineering. BMPs are stored within the ECM of mature bone [11] and their expression is also upregulated in the early phases of fracture repair [12] . Through previous studies, BMP-2, -4, -6 and -7 (also called OP-1) are widely known as osteoinductive factors [13,14] . Expression levels of the different BMPs
10.2217/RME.12.1 2012 Future Medicine Ltd

vary throughout the healing process, which causes up- or down-regulation of other BMP family members [14] . BMPs can inuence the expression of other GFs [13] . BMP-2 is involved in the expression of osteogenic markers via induction of the MAPK pathway [15] and osteogenic nuclear transcription factor Cbfa-1/Runx2 [13] . Thus, BMPs have been widely used in preclinical and clinical trials [16,17] . Among the FGFs, FGF-2 plays a role in the bone-remodeling process by maintaining the ne balance between boneforming osteoblasts and bone-resorbing osteoclasts [18] and supporting neovascularization, similar to VEGFs [13] . In addition, FGF-2 promotes osteogenic differentiation via downstream activation of Cbfa-1/Runx2 [19] . VEGFs themselves are powerful angiogenic factors that are responsible for mediating vascular responses in tissues [20,21] . They are particularly important for wound healing and are expressed in fracture sites where they promote new neovascularization and angiogenesis via recruitment of endothelial cells [13,22] . IGFs are also thought to have an important role in bone healing. IGF-I and -II are both expressed by cells of the musculoskeletal system and are highly expressed in bone fracture sites, although IGF-I has the most potent effect on bone metabolic activity [23] . IGFs induce expression and deposition of the typeI collagen matrix and also maintain the anabolic, bone-forming state by preventing expression of collagenase by osteoblasts [13,23] . Although the effects of GFs are pleiotropic, their short biological half-life, lack of long-term
Regen. Med. (2012) 7(3), 369385

Ye-Rang Yun1, JunHyeogJang2 EunyiJeon2, WonmoKang2, SujinLee2, Jong-Eun Won1,3, HaeWon Kim*1,3,4 &Ivan Wall3,5
Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 330-714, Korea 2 Department of Biochemistry, Inha University School of Medicine, Incheon 400-712, Korea 3 Department of Nanobiomedical Science & WCU Research Center, Dankook University Graduate School, Cheonan 330-714, Korea 4 Department of Biomaterials Science, School of Dentistry, Dankook University, Cheonan 330-714, Korea 5 Department of Biochemical Engineering, University College London, Torrington Place, London WC1E 7JE, UK *Author for correspondence: Tel.: +82 41 550 3081 Fax: +82 41 550 3085 kimhw@dku.edu Authors contributed equally
1

part of

ISSN 1746-0751

369

REVIEW

Yun, Jang, Jeon etal.

stability and tissue selectivity, and possible toxicity and risk of tumor-promoting activity limit their practical therapeutic application and thus demand controllable and sustainable delivery [24] . Selection of an appropriate delivery system for GFs is essential to induce a specic biological effect. For example, locally administered solutions of FGF-2 did not promote bone regeneration in rabbit skull defects, whereas FGF-2 incorporated into gelatin hydrogels did [25] . Moreover, BMP in an insoluble bone matrix failed to induce bone growth around titanium implants because of unsuitable carrier properties [26] . Similarly, IGF-I failed to induce bone repair when delivered directly into the osteotomy via an osmotic pump [27,28] , but was successful when embedded in biodegradable microspheres to heal segmental long bone defects [29] . In the following sections, we will briey overview the requirements of biomaterial scaffolds that might be employed to deliver GFs and also explain the importance and roles of various GFs that have been used for bone regeneration based on in vitro and in vivo experimental results.

Considerations for carrier materials When designing appropriate delivery systems, the carrier materials need to satisfy some essential criteria. Specically, the materials should be biocompatible, noncytotoxic and nonimmuno genic to prevent any adverse effects on residing and recruited cells and neighboring tissues. Moreover, the chemistry of materials should be tailored to support cell adhesion and proliferation. Ultimately, the implanted carriers should be degraded by enzymes and/or circulating biological uid. Commonly used materials include natural polymers such as collagen, gelatin, brin, alginate, silk, hyaluronic acid and chitosan [30] , as well as synthetic polymers, including the a-hydroxy esters such as poly(lactic acid), poly(glycolic acid), poly(anhydride), poly(phosphazene), poly(propylene fumarate) and poly(ethylene glycol) [3145] . Although most GF delivery carriers have been produced from polymeric materials, inorganic materials are also used to promote bone formation during specic pharmacological intervention for musculoskeletal problems. Specically, calcium phosphates and silicate glasses have been processed to deliver GFs [4656] . BOX1 presents the carrier materials and matrix types used for the delivery of various GFs for bone regeneration. Those carrier compositions have been processed in the form of micro-/nano-particles, porous scaffolds, bers and hydrogels for specic
370
Regen. Med. (2012) 7 (3)

applications. The GFs have either been incorporated within the structure of carriers or bound onto the surface. In either case, when the GFs were introduced through chemical bonds (covalently, ionically or afnity bindings), a more sustainable release could be achieved, possibly leading to potentiating the efcacy of GFs. When the carriers incorporate the GFs inside the structure, the chemical interaction between GFs and carriers and the degradation rate of the carriers will dominate the release prole of GFs, and the delivery kinetics is more tunable/controllable, allowing prolonged release. On the other hand, when introduced on the surface of carriers, the effects of GFs may occur more rapidly in a manner of direct interaction with the cells in the surroundings. Most of all, timed delivery of GFs to the site over certain periods at appropriate doses is the most critical issue that needs deep consideration in the design of delivery systems. In fact, the repair and regeneration process of bone is a highly complex and harmonized event involving various GFs. Therefore, many recent studies have focused on the delivery of dual/multiple GFs. These studies have been conducted in response to the nding that treatments with GF combinations exhibited additive or even synergistic effects on bone formation. Occasionally, administration of multiple GFs has been found to inhibit bone formation. For example, in a rat tibia fracture model, the combined application of IGF-I and TGF-b1 showed a synergistic effect in fracture healing [57] , whereas the combined administration of BMP-2 and FGF-1 with collagen sponge resulted in decreased bone formation in the same model [58] . These studies demonstrate that the combination of GFs should be carefully considered, and that adequate release kinetics might be critical for successful bone regeneration. For example, should dual GFs be released in combination or sequentially? Dual or multiple GF delivery regimes complicate the design of controlled-release devices, potentially requiring separated compartments to provide timely and sequential release.

BMP Among GFs, BMPs are known to play a critical role in bone regeneration. BMPs are multi functional GFs that belong to the TGF- b super family due to their striking similarities in structure and sequence [11] . Currently, there are 20 known BMP family members, which share some degree of similarity [59] . They are involved in a variety of functions in bone. Predominantly,
future science group

Administration of growth factors for bone regeneration

REVIEW

Box 1. Carrier materials for bone tissue regeneration.


Carrier materials Natural polymers: collagen, brin, alginate, silk, hyaluronic acid and chitosan Synthetic polymers: poly(lactic acid), poly(glycolic acid), poly(lactic-co-glycolic acid), poly(caprolactone), poly(anhydride), poly(phospazene), poly(propylene fumarate) and poly(ethylene glycol) Inorganic materials: hydroxyapatite, tricalcium phosphate, calcium phosphate cement and bioactive glasses Matrix types: microparticles, nanoparticles, porous scaffolds, microbers, nanobers and hydrogels Growth factors: BMP-2, BMP-4, BMP-7, FGF-2, TGF- b1, TGF- b2, TGF- b3, VEGF, IGF-I, IGF-II, combinations of two growth factors and peptides

they promote bone formation throughout development and after injury in adults [6062] . BMP signaling is mediated via BMP receptors (BMPRs). Activation of BMPRs leads to intracellular signaling transmitted by Smad proteins, thereby inducing the transcription of specic genes. Their function is primarily modulated by extracellular BMP-binding proteins such as noggin and chordin, which inhibit BMP signaling by blocking their interaction with BMPRs. Active BMPs are disulde-linked dimers and can be either heterodimers (two different family members) or homodimers (two identical family members). The composition of the dimer profoundly inuences bone cell function. Thus, heterodimers of BMP-2/7 or BMP-4/7 produce a greater morphogenic effect than homodimers of any of these factors [63] . Initially, BMPs were isolated from various cell types and then puried before usage; however, since the 1980s, BMPs have been produced using recombinant DNA technology and batch production methods. In early studies, highly puried BMPs were used invitro. For instance, BMP-2 and BMP-3 retain bioactivity in chondrocytes and osteoblastic cells [64] . The osteogenic activity of BMPs invitro has only been identied since the development of recombinant DNA technology, and there have been many reports of its activity [65,66] . Moreover, 14 different BMPs have been investigated in various cell types in recent studies. The results of these studies suggested an osteogenic hierarchical model in which BMP-2, -6 and -9 might play an important role in induction of osteoblast differentiation of mesenchymal stem cells, whereas most other BMPs stimulate osteogenesis in mature osteoblasts. Overall, the BMPs with greatest osteogenic capacity invitro are BMP-2, -4, -6, -7 and -9 [67] . The benecial effects of BMPs for repairing bone defects such as bone fracture, spinal fusion and osteoporosis have been demonstrated invivo in a variety of animal models [6883] . Of
future science group

the BMPs, the therapeutic potential of recombinant human BMP-2 and -7 (rhBMP-2 and rhBMP7) has been identied in bone wound healing models, as well as in craniomaxillo facial, periodontal and dental diseases [60] . Various studies have reported the multiple functions of BMPs at different dosages for specic cell types such as osteoblasts, chondroblasts and stem cells [82] . For instance, elevated concentrations of BMPs accelerate bone growth [83] , whereas tooth ankylosis can be induced by BMP-2 in a dosedependent fashion following application for periodontal regeneration [84] . Contrary to other BMPs, BMP-3 inhibits BMP-2-induced osteogenic differentiation. Furthermore, BMP3knockout mice exhibited increased bone density, which indicates that BMP-3 is both an antagonist and inhibitor of BMP-induced osteogenesis invivo[85] . Gene transfer technologies have received a great deal of attention due to their potential to aid healing by permitting targeted delivery and sustained expression of BMPs within osseous lesions [86] . This and other examples of regenerative medicine approaches in numerous exvivo studies have demonstrated that BMPs show great promise for bone tissue regeneration strategies [81,87104] . A summary of the application of BMPs for bone tissue regeneration invitro, invivo and exvivo is presented in TAbLE1. For many years, the clinical use of BMPs was severely limited due to several problems. First, potential health risks, such as the transmission of slow viruses, which cause disease in a slow, progressive manner over a time-course spanning months to years and consequently remain undetected for long periods, or induction of malignancies have prevented straightforward translation to the clinic [105] . Second, the use of high doses not only raises safety concerns due to its mitogenic potential, but also greatly increases costs. For example, to induce bone regeneration, BMPs need to be administered at high doses that are greater than the levels at which they occur
www.futuremedicine.com

371

REVIEW

Yun, Jang, Jeon etal.

Table 1. Application of BMPs for bone tissue regeneration.


Subfamily
Invitro BMP-2 BMP-2, -3 14 BMPs BMP-2

Model
ROB-C26, ROB-C20 cells Chondrocyte, MC3T3-E1 cells C2C12, C3H10T1/2, TE-85 cells Mandible defects in rats Femoral defects in rabbits Pelvic defects in sheep Tibial defects in sheep Femoral defects in rats Femoral fracture in rats Osteotomy/ostectomy in horses Spine fusion in rats Mandible defects in rats Ulnar osteotomy in rabbits Cranial defects in mice Cranial defects in pigs Mandible defects in rats Femoral defects in rats

Biological effect
Stimulate osteoblastic maturation Promote osteoblastic differentiation BMP-2, -6 and -9 induce osteoblastic differentiation Promote mandibular distraction osteogenesis Enhance fracture healing Retard bone formation Enhance fracture healing Enhance healing of segmental bone defects Promote bone formation in fracture region Accelerate bone formation and osteotomy healing Enhance osteoblastic differentiation Repair bony defects in the craniofacial region Accelerate healing Repair healing bone defects Accelerate bone regeneration Accelerate bone regeneration Enhance healing of segmental femoral defects Induce heterotopic and orthotopic bone formation Repair segmental femoral defects Repair femoral defects Repair experimentally induced osteonecrosis Repair bone defects Induce bone formation Enhance spine fusion Induce bone formation Promote healing of osteoporotic bone fracture Accelerate bone healing Enhance cranial bone regeneration Repair segmental bone defects Enhance bone healing Enhance spine fusion

Ref.
[66] [64] [67] [68] [69,70] [71] [72] [7375] [76] [77] [78] [80] [81] [87] [88] [81] [89] [90] [91] [92] [93] [94,95] [96] [97] [98] [99] [100] [101] [102] [103] [104]

Invivo

BMP-2, -4 BMP-2, -6 BMP-2, -7 BMP-2, -9 BMP-6 Exvivo BMP-2

BMP-2, -7 BMP-4 BMP-7

Femoral defects in goats Tibial defects in goats Spine fusion in mice Spine fusion in rats Spine fusion in rabbits Osteoporotic rat model Osteotomy in horses Cranial defects in mice Femoral defects in rats Cranial defects in rabbits Spine fusion in rats

naturally in bone. Another concern was the lack of appropriate delivery systems required for the clinical use of BMPs. Recombinant BMPs have been shown to have a bone-inducing capacity, but require special carriers to maintain their activity at low doses [106] . The function of the carrier matrix is to immobilize the bone-inducing protein at a particular site for a sufcient amount of time to allow bone induction to occur. Studies using various carrier materials have demonstrated that combinations of carrier materials and BMPs enhance osteogenic activities. Various materials and fabricated forms are used as carriers. Natural polymers, including collagen, gelatin, brin, alginate, chitosan and silk, are widely employed as carriers, and the use of these compounds has resulted in signicant induction of osteogenesis [107113] . BMP-2-derived oligopeptide covalently coupled to alginate hydrogel was found to induce ectopic bone formation in rats [114] . Chitosan was also reported to be an effective carrier [115117] . Synthetic polymers, which primarily include a-hydroxy esters, such as poly(lactic acid),
372
Regen. Med. (2012) 7 (3)

poly(glycolic acid) and their copolymers, have also been used as BMP carriers. The use of such compounds has been found to accelerate bone formation both invitro and invivo [45,109,118] . Because metallic implants are commonly used in bone, BMPs have been coated on the surface of metals. The rhBMP-2 coating on titanium scaffolds has been studied in various species and types of defects, including minipigs with tibia implants, aged sheep implanted below the tibial plateau, nonaged sheep subjected to alveolar segmental osteotomy, rabbits with femural and tibial implants and rats with subcutaneous implantations [119124] . In these studies, BMPcoated titanium scaffolds successfully facilitated bone regeneration, which generally occurred in a concentration-dependent manner. Moreover, the combined use of BMP with a second GF can have synergistic effects on bone bonding. For example, adult male dogs treated with titanium implants containing both rhTGFb2 and rhBMP-2 were found to have a greater implant xation strength than groups that received either type of GF alone [125] . In terms of the carrier,
future science group

Administration of growth factors for bone regeneration

REVIEW

combining different materials can improve the delivery potential [48,126,127] . For example, a novel chitosan/g-PGA composite scaffold for the controlled release of rhBMP-2 has been established and can release larger amounts of the GF in a sustained and controlled manner. Some BMPs have been investigated in both animal studies and human clinical trials [128132] . This has led to several commercially available BMP-based products that will be discussed later in this review.

FGF Originally detected as substances in the brain and pituitary gland, FGFs isolated from these areas were found to promote the growth of broblasts [301] . FGFs are representative GFs that have potential effects on the repair and regeneration of tissues [133] . The FGF family comprises at least 22 different monomeric proteins with molecular weights of 1618 kDa. These compounds bear 55% amino acid sequence homology and are encoded by distinct genes [134] . The 22 members of the FGF family mediate their cellular responses by binding to and activating different isoforms of their receptors encoded by the four receptor tyrosine kinases (i.e., FGFR1, FGFR2, FGFR3 and FGFR4) [135] . FGFs mediate their effects via activation of signaling pathways, including R AS/MAPK, PI3K/AKT and PLC g, among which the RAS/MAPK pathway is known to be predominant [136] . FGFs have various biological functions that occur both invivo and invitro, including roles in mitogenesis, cellular migration, differentiation, angiogenesis and wound healing [133] . Application of FGFs for regenerative medicine may be suitable for promoting regeneration in a wide range of tissues, including skin, blood vessels, muscle, adipose, tendons/ligaments, cartilage, teeth, nerves and bone. Indeed, FGFs are key regulators of bone development [137] , and bone regeneration can be achieved by using bone replacement material that comprises one or more polypeptides having the biological action of FGFs in a porous matrix [132] . FGF pathways can mediate the effects of FGF signaling on osteoblast gene regulation. These pathways can stimulate proliferation of osteogenic cells and chondrocytes and promote cell differentiation and angiogenesis [138] . A brief summary of the roles of different FGF family members in bone growth is presented in FIGURE1. Among the 22 members of the FGF ligand family, FGF-2, -9 and -18 in particular play important roles during bone development. Knockout
future science group

models have revealed that FGF-2-/- adult mice have decreased skeletal bone mass. Moreover, FGF-9-/- embryonic mice exhibit delayed initiation of chondrogenesis. Finally, FGF-18 -/embryonic mice with reportedly smaller cranial vaults and widened cranial sutures have been described [137] . Similar to BMPs, the release effect of FGFs from various carrier materials has been investigated to identify appropriate delivery strategies that regulate the sustained and targeted release of FGFs. FGF-1 and -2 have primarily been used for such studies. For example, in calvarial defects in rats, FGF-1 administered via a coated agarose gel implant was found to promote osteogenesis for cranioplasty [139] . FGF-2 delivered in a hyaluronan or gelatin hydrogel has also been reported to accelerate bone healing invivo [140] . In addition, the osteoinductive effect of hydroxyapatite implants coated with FGF-2 was greatly enhanced compared with uncoated control implants in a rabbit model [141] . Other FGF-binding scaffolds for bone regeneration have also been reported. Collagens with perlecan domain I improved binding of FGF-2, and perlecan domain I-coated electrospun collagen bers were more effective than heparin bovine serum albumin collagen bers at binding FGF-2. Because FGF-2 regulates cell growth, differentiation, migration and survival, the
Preosteoblast Proliferation FGF-2, FGF-4, FGF-9, FGF-18

Osteoblast

Osteoid Matrix

Differentiation FGF-2, FGF-4, FGF-8, FGF-9, FGF-18 Mineralization FGF-2, FGF-9, FGF-18 Osteocyte

Regen. Med. Future Science Group (2012)

Protects against cell apoptosis FGF-2

Figure 1. FGF functions for bone growth.

www.futuremedicine.com

373

REVIEW

Yun, Jang, Jeon etal.

ability to effectively bind FGF-2 to the matrix surface is a key improvement in the fabrication of scaffolds for successful tissue engineering [142] . When titanium nonwoven fabrics with FGF-2 delivery systems of gelatinhydroxyapatite hydrogel microspheres were implanted in rabbit skull defects, bone regeneration was signicantly enhanced compared wth cases without the FGF-2 delivery system or simply with combined systems of FGF-2 in titaniumhydroxyapatite, suggesting the importance of a proper delivery system [143,144] . The negatively charged gelatin protein structure may help the ionic bonding with oppositely charged FGF-2, and consequently sustained release from the system invivo condition.

VEGF VEGFs are important GFs that stimulate the growth of new blood vessels. With molecular weights ranging from 34 to 42 kDa, VEGFs are secreted by various human and rodent tumor cell lines. The VEGF family contains VEGF-A, VEGF-B, PlGF, VEGF-C and VEGF-D [145] . Different isoforms of VEGF can be generated through alternative splicing, including VEGF121, VEGF145, VEGF165, VEGF183, VEGF189 and VEGF206. Among these, the major forms (i.e.,VEGF121, VEGF165 and VEGF189) are secreted by most cell types [146] . VEGFs have various functions. The primary function of VEGFs is related to both vasculogenesis and angiogenesis. Accordingly, VEGFs are known as powerful angiogenic factors that stimulate proliferation and migration of endothelial cells, resulting in the formation of tubular blood vessels. Moreover, VEGFs play a crucial role in bone formation and healing through recruitment, survival and activity of bone forming cells [147] . The process of angiogenesis involves new vessel formation from a pre-existing vascular network. These newly formed vessels are essential for transporting nutrients and oxygen, migration of cells and preservation of the appropriate metabolic microenvironment during bone regeneration and bone repair [148] . Therefore, the activation of angiogenic factors using VEGFs leads to coordinated angiogenesis and bone regeneration [149] . Additionally, VEGFs were found to enhance the migration of osteoprogenitor cells [150] and stimulate migration and differentiation of primary human osteoblasts [151,152] . Furthermore, the expression of VEGF165 by osteoblasts was actively upregulated during osteogenesis [153] . These results suggest that VEGFs can enhance
374
Regen. Med. (2012) 7 (3)

bone formation. Interestingly, VEGF in combination with BMP-2 enhanced BMP-2-induced bone formation, further indicating that additive or synergistic effects can be achieved using combinations of GFs for bone regeneration. VEGF can also act as a mediator of various other osteoinductive factors, including TGFb1, IGF and FGF-2, which in turn control the expression of VEGF [154] . Therefore, the combined application of VEGFs with other GFs could synergistically enhance bone formation and fracture healing. VEGFs not only enhance the formation of bone directly, but also indirectly via the induction of angiogenesis at the fracture site, enabling the delivery of additional circulating progenitor cells [154] . TAbLE2 shows a summary of the application of VEGFs for bone tissue regeneration invitro [146,149,151,152,155158] and invivo[150,157,159161] . Due to the rapid diffusion of GFs away from the injury site after injection, delivery of VEGF was performed using poly(lactic-co-glycolic acid) (PLGA) biopolymer carriers [162] . In a rat calvarial defect model, signicant improvements in neovasculatrization, bone coverage and bone mineral density were observed after implantation of VEGF scaffolds compared with PLGA control scaffolds [162] . In another study, VEGF coated onto the surface of bioactive glass resulted in additive bone healing effects within the rat critical- sized defect [163] . Moreover, PLGA scaffolds seeded with human bone marrow stromal cells containing combinations of condensed plasmid DNA encoding BMP-4 and VEGF promoted new bone formation [164] .

IGF IGFs are polypeptides with high sequence similarity to insulin. There are two IGF ligands (IGF-I and IGF-II) and two cell-surface receptors (IGF1R and IGF2R). IGF-I has been used for the treatment of injuries in various tissues. IGF-I generally functions by stimulating cell proliferation and survival and inhibiting apoptosis [165] . IGF-I is the most abundant GF present in skeletal tissues [166] . It is an important regulator of skeletal growth and development [167] , as well as a regulator of osteoblastic function [166] . In addition, IGF-I stimulates the proliferation and differentiation of osteoblastic precursor cells [168] . This osteoblastic function is mediated through the PI3K pathway by stabilizing b -catenin, thereby enhancing Wnt-dependent activity [169] . In a monolayer model of wound healing, IGF-I secreted from osteoblasts directed
future science group

Administration of growth factors for bone regeneration

REVIEW

Table 2. Application of VEGFs for bone tissue regeneration.


Cell/animal type
Invitro Endothelial cells

Biological effect

Ref.
[146] [149] [155] [156] [157] [146] [158] [157] [159] [150,160,161]

Proliferation, sprouting, migration and tube formation Osteoblasts Migration, proliferation and survival Chondrocytes Conversion of cartilage into bone and osteoblasts Muscle-derived stem cells Differentiation and proliferation (angiogenesis) Mouse osteoprogenitor cells Ossication and angiogenesis Enhancement of bone formation and healing Bone formation Mouse Rabbit Resorption of cartilage Blood vessel formation, ossication and new bone (callus) maturation Bone healing and increased bone density

Invivo

the migration of osteoblasts, promoted cell spreading and regulated cell polarization [170] . Similarly, in developing fracture calluses, IGF-I and its receptors are widely expressed in periosteal cells, mesenchymal cells, proliferating chondrocytes and osteoblasts, suggesting that it plays a coordinated role among several cell types during fracture healing [171] . There is invivo evidence that serum IGF-I concentrations are positively correlated with skeletal mass. Increases in serum IGF-I levels during growth lead to enhancement of all bone phenotypic traits and may play a protective role later on during aging. Such increases also emphasize the contribution of serum IGF-I to both cortical size and bone density [172] . By contrast, locally produced skeletal IGF-I plays a greater role in trabecular bone integrity. This is demonstrated in transgenic mice expressing IGF-I in osteoblasts and in conditional IGF-I receptor-null mice, which display decreased osteoblast numbers and function, causing reduced bone formation and trabecular bone volume [173] . Global deletion of IGF-I in mice resulted in postnatal growth retardation [174] ; similarly, in IGF-I heterozygous mice, reduction of serum IGF-I levels resulted in a change of body weight and reduced cortical bone mineral density [175] . Other studies in mice with reduced serum IGF-I levels also showed signicant decreases in transverse bone growth. Reductions in serum IGF-I during postnatal growth are extremely important markers of a potential compromise in bone robustness and increased fracture risk during adulthood and aging. IGF-I also induces VEGF expression in skeletal cells, and so may enhance the direct and indirect positive effects of VEGF on endochondral bone formation and osteoblastic differentiation and function [176] . Only a few invivo studies investigated the use of intra-articular injections of IGF-I in mice,
future science group

dogs and rabbits. These studies all demonstrated little therapeutic effect of the periodic use of IGF-I alone for cartilage and subchondral bone repair in osteoarthritic joints [176] . The effect of IGF release from carrier materials has also been investigated. When IGF-I was coated onto a dextran-co-gelatin, periodontal tissue regeneration was enhanced [177] . In another study, IGF-I entrapped in PLGA micro particles also enhanced new bone formation [178] . Moreover, the combined delivery of TGF-b and IGF-I within gelatin hydrogels to promote bone formation was evaluated, and the combined implantation of TGF-b and IGF-I was shown to have greater bone healing potential than IGF-I treatment alone [179] . The long-term effects of combined TGF-b and IGF-I on fracture healing have also been determined, and while enhanced bone regeneration was evident in a rat closed tibial fracture model for up to 6weeks, by 12weeks, no difference in bone regeneration status remained in comparison with control fractures [180] . However, when IGF-I and TGF-b were coated onto titanium with poly(l-lactide acid), they failed to induce ectopic bone formation in ovine muscle [181] . Nevertheless, IGF-I and IGF-II delivered within collagen gels were found to enhance the repair of facial osseous defects[182] .

Other GFs HGF HGF has been shown to stimulate both osteoblast proliferation and osteoclast chemotactic migration [183] . In bone tissue, HGF is produced by osteoclasts and induces responses in both osteoblasts and osteoclasts. In osteoblasts, HGF induces DNA synthesis, whereas in osteoclasts, in addition to DNA synthesis, HGF also stimulates chemotactic-oriented migration, as previously observed in epithelial and endothelial cells [184] . Cell migration is important in the
www.futuremedicine.com

375

REVIEW

Yun, Jang, Jeon etal.

early steps of bone resorption, when osteoclast precursors are chemotacticly attracted to the bone matrix. In these cells, HGF also stimulates DNA synthesis, thereby controlling both migration and proliferation of osteoclasts at the sites of bone resorption [185] . HGF reportedly exerts benecial effects on bone formation at the boneimplant interface of orthopedic prostheses [184] . However, HGF has occasionally been found to inhibit BMP-2-induced bone formation through activation of ERK1/2 when the cells are treated with both HGF and BMP-2 simultaneously [186] . Furthermore, HGF was shown to inhibit the BMP-2-induced expression of Runx2 [187] , which, along with osterix, is an essential transcription factor for osteogenic differentiation [188] . Treatment with HGF prior to BMP-2 was shown to promote cell proliferation without disturbing BMP-2-induced osteoblast differentiation. Thus, HGF treatment can potentially be benecial for increasing cell numbers, but the timing of HGF treatment is considered to be critical and should be carefully determined for successful bone formation induced by BMPs [186] . HGF readily adsorbs to the bone mineral surface, maintains integrity (i.e., does not degrade) over time and enhances osteoblast differentiation. These characteristics make HGF a promising potential candidate for GF coating of bone implant/scaffold surfaces [183] . EGF EGF plays an important role in osteoblast growth and development [189] . EGF alone promotes osteoblast proliferation, and the combination of EGF with BMP-2 and -7 signicantly upregulates this proliferation [190] . The effect of EGF on osteoblast growth may occur through specic receptors that are expressed by the osteoblasts. EGF delivered either alone or in combination with BMP-2 and -7 also induced expression of ALP and OC proteins, but in spite of this, there is an overall net negative effect on bone mineralization due to inhibition of nodule formation [190] . Moreover, EGF combined with calcitriol was able to stimulate osteoblast differentiation, characterized by increased synthesis of both ALP and OC [189] . EGFR signaling is also involved in bone resorption. EGF has the ability to strongly stimulate bone resorption in cultured fetal rat long bones, newborn mouse calvarial cultures and long-term human marrow cultures, suggesting that this GF regulates osteoclastogenesis and bone resorption [191] .
376
Regen. Med. (2012) 7 (3)

MGF Many splice variants of the IGF-I gene can be produced that have different functions [192] , and one splice alternative variant is MGF [193] . MGF acts locally as a tissue repair factor that responds to changes in either physiological conditions or environmental stimuli [193] . The MGF splice variant of IGF-I is produced when cells experience significant physical trauma such as mechanical overload, severe temperature uctuations or hypoxia, among others [192] . The presence of a unique E domain (a 48-bp insert between exons 5 and 6 introduced during splicing) in the C-terminal region of MGF (called MGF-Ct24E) functionally distinguishes MGF from other IGF-I isoforms [194] . Recently, MGF and its E peptide have attracted much attention because of their reported woundhealing role [195] . MGF and MGF-Ct24E were found to improve osteoblast proliferation [192] . Particularly, MGF-Ct24E increased proliferation activity compared with IGF-I. To date, the study of MGF using carrier materials has not been investigated, although MGF is a candidate for targeting bone tissue regeneration. Therefore, further study on MGF is needed. PDGF PDGF is produced by skeletal cells. These cells express three pdgf genes (i.e., pdgfa, pdgfb and pdgfc), indicating that PDGF may act as an autocrine regulator of skeletal cell function. PDGF is present in bone matrix and is secreted by platelets at the site of fracture during early tissue repair [196] . It is a potent mitogen that facilitates wound healing and stimulates bone repair by expanding osteoblastic precursor cells [197,198] . However, additional signals are required to induce the differentiation of these cells towards mature osteoblasts [166] . The angiogenic effects of PDGF, which are similar to the effects of VEGF, may also be favorable to osseous wound repair. PDGFs have a similar structure to VEGF, and PDGF-BB enhances FGF-2-stimulated VEGF release [199] . These additional factors can accelerate and enhance the bone-induction effects of PDGF, promoting greater bone volume, and thus enabling earlier implant placement and loading [197] . Collagen gels loaded with PDGF-BB were found to enhance their bone-regenerative effect on tibial osteotomies in rabbits [200] . Furthermore, PDGF-BB within a chitosan/ tricalcium phosphate sponge carrier was also shown to have bone-regenerative effects [201] . Taken together, these studies suggest that
future science group

Administration of growth factors for bone regeneration

REVIEW

delivery of PDGF within carriers can enhance bone regeneration strategies.

Fusion proteins & peptides Fusion proteins are formed by combining sequences from two or more genes that originally coded for those individual proteins. The single protein product contains functional characteristics of each individual protein. This is made possible by the fact that a particular protein function is often specically attributable to a single domain of that protein. Therefore, the whole protein does not have to be fused, but only the sequences containing the functional domains are of interest. Fusion proteins have been widely used for identifying and purifying proteins, (e.g., using hexa-His peptide [6Histag], fusing a GST protein or FLAG peptide). In addition, fusion proteins can have toxins or antibodies attached to them, offering the opportunity to target specic cell types, such as tumor cells. Because of their wide range of uses, fusion proteins are being widely investigated. FGFs are effective GFs for fusion with a variety of proteins. For example, FGF-1 conjugated with heparan sulfate proteoglycan was constructed to protect FGF-1 from proteolytic degradation [202] . In addition, fusion of FGF-2 and OC has been investigated [144] . The FGF-2mOC product has advantages for bone repair and regeneration owing to the synergistic effects of FGF and OC. The hydroxyapatite (HA)-binding capacity of FGF-2mOC protein is signicantly greater than that of native FGF-2, due to the high afnity of OC to HA. Furthermore, fusion of FGF-2 and mOC in a HA surface-immobilized state results in a signicant increase in cell proliferation and differentiation of MC3T3-E1 osteoblastic cells. These properties, which are not exhibited by native FGF-2, may offer a novel strategy to potentiate the therapeutic effect of FGF-2 in bone repair and regeneration. In particular, collagen-binding domain (CBD)-based GF fusion proteins have been found to be useful for combining GFs with collagen carriers to injured tissues in some studies. For instance, fusion protein delivery of VEGF has been investigated when Ishikawa etal. established the bronectin CBD (FNCBD)VEGF121 [203] . FNCBDVEGF121 stably maintained an optimally high and local concentration of VEGF within the collagen matrix, stimulating both endothelial cells and endothelial progenitor cells insitu, thereby supplying a vascular regeneration niche. The same group also established a collagen-binding GF
future science group

consisting of EGF and FNCBD, designated FNCBDEGF [204] . FNCBDEGF is a biologically active fusion protein that was able to stably bind to collagen materials and exert its GF activity even after collagen binding. Thus, in an injured artery, infused FNCBDEGF remained bound to collagen exposed in the injured tissues, even after blood circulation was restored. Injection of the fusion protein into the rat hindlimbs was found to be effective for direct administration to muscular tissue. The effects of a HGF fusion protein exhibiting collagen-binding activity (CBDHGF) were studied to determine the re-endothelialization and neointimal formation in a balloon-injured rat carotid artery [205] . CBDHGF accelerated re-endothelialization and neointimal formation invivo compared with both the negative control and HGF-treated groups. TGF-b1 was also engineered to generate a fusion protein bearing a CBD to selectively target typeI collagen and ensure the slow and localized release of the GF [206] . Taken together, CBD fusion protein is a useful vehicle to deliver vascular GFs to injured arteries. Genetically engineered fusion proteins with specic binding domains for the ECM are becoming particularly attractive for bone targeting.

Regulator-approved products: towards the clinic There are still relatively few recombinant GF products on the market for bone regeneration purposes. Of fundamental importance for the delivery of biologics, such as recombinant GFs, is that they are safe to use and do not compromise the wellbeing of the patient. The second main requirement is that they actually demonstrate efcacy in the context of accelerated tissue regeneration responses, yielding substantial improvement in regeneration capacity. The US FDA has approved two BMP therapeutic products for clinical application. Both BMP-2 and -7 are available commercially for clinical use as the active components of the products Infuse (Medtronic Sofamor Danek; MN, USA) and OP-1 (Stryker Biotechnology; MA, USA). These BMPs have been rigorously investigated in both animal studies and human trials [128132] , with many successful results. Specifically, BMP-2 (Infuse) has been approved for the treatment of fusion of the lumbar spine in patients with degenerative disc disease. It has also been approved for certain oral and maxillofacial uses and has been demon strated to be highly effective in the treatment of
www.futuremedicine.com

377

REVIEW

Yun, Jang, Jeon etal.

tibial fracture [130] . BMP-7 has been approved as an alternative to autografting in recalcitrant long bone nonunions [207] and in compromised patients requiring revision posterolateral lumbar spinal fusion. However, recent evidence suggests that the safety prole of BMP treatment is not as high as was originally thought, and patients treated with BMP-2 for spinal fusion have an increased risk of cancer [208] . Therefore these
Executive summary

treatments should only be used with signicant caution. PDGF has been used to stimulate bone regeneration in dental procedures. Being incorporated into a calcium phosphate matrix, it has been demon strated to provide safe and effective stimulation of bone regeneration in patients with severe periodontal bone loss [209] . It is available under the trade name GEM21S (Osteohealth Company,

Background Growth factors promote cell responses and are important mediators of wound healing after injury. They hold promise for use in tissue engineering and for exogenous stimulation of intrinsic regenerative responses if delivered to the injury site using appropriate carrier materials. Considerations for carrier materials Materials that will be implanted should be biocompatible, nontoxic and nonimmunogenic, and ideally will degrade over time by the action of enzymes or circulating body uid. Polymer materials that are generally used are either natural (i.e., collagen) or synthetic (i.e., poly[lactic acid] or poly[glycolic acid]), andin the context of bone regeneration, inorganic materials such as calcium phosphate, which supports bone cell responses, are oftenused. BMP Most BMPs stimulate bone regeneration (BMP-3 being a notable exception), and delivery of BMPs using synthetic polymers has proven successful in accelerating bone regeneration in animal models. FGF FGF is a key regulator of bone development and has broad effects on repair and regeneration responses, including cell proliferation, migration, differentiation and angiogenesis. VEGF VEGF is an important growth factor that mediates angiogenic and vasculogenic responses, which are helpful for production of bone tissue as it requires a vascular supply. VEGF in combination with BMP2 enhances the osteogenic effects of BMP2. IGF IGF is an important regulator of skeletal growth and development and of osteoblast function, particularly proliferation and differentiation. Delivery using a variety of carrier materials in animal models of osteoid defects has yielded favorable regenerative repsonses. Other factors HGF HGF stimulates osteoblast proliferation, osteoclast proliferation and chemotaxis to regulate bone turnover, but inhibits BMP-2-induced bone formation. EGF EGF stimulates osteoblast proliferation and has a synergistic effect in combination with BMP-2 or -7. MGF MGF is an IGF splice variant that stimulates osteoblast proliferation and is a general positive regulator of wound healing. PDGF PDGF is a potent inducer of proliferation, but in itself is not capable of directing osteoblastic precursors towards a mature phenotype. Fusion proteins Fusion proteins that are engineered to contain functionally active components of two or more individual proteins have proven effective at enhancing bone regeneration. Notably, fusion proteins of FGF-2 combined with OC have been successfully employed to enhance cell responses on hydroxyapatite-coated scaffolds due to the dual impact of mitogenic FGF-2 and maturation-inducing OC. Regulator-approved products: towards the clinic Several recombinant protein products are currently available on the market for bone-regeneration therapy. Specically, products containing BMP-2, BMP-7 or PDGF as the active ingredient were successful in several clinical trials. However, in light of recent evidence, regulator concern still exists regarding the safety of recombinant protein products that have mitogenic capacity. Therefore, it is necessary to proceed with caution in the development of recombinant growth factors and delivery methods for bone regeneration.

378

Regen. Med. (2012) 7 (3)

future science group

Administration of growth factors for bone regeneration

REVIEW

NY, USA) and meets the International Standard for biocompatibility of medical devices [401] . To date, no severe adverse effects have been reported, and it is therefore very promising as a treatment strategy. Broadly speaking, orthopedic and oral surgery have beneted greatly from commercially available GFs in the last few years. Positive patient responses have been reported in many clinical trials. However, recently reported safety concerns highlight the continued requirement to carry out more research and understand the global system effects of GFs on the patient. It also highlights the need to understand precisely the dosages that will elicit positive effects without compromising safety and the need to use biomaterials that will carefully control and regulate GF release in a precise and localized manner.

bone regeneration. Future strategies to restore functions in difcult-to-heal bone fractures will depend on carefully controlled, targeted delivery of osteogenic GFs to re-establish the healthy tissue architecture. In addition, administration of multiple GFs in combination is expected to be required to maximize the potential of GFs. Many strategies are being explored for producing carrier systems that will ensure targeted delivery in a sustained and controlled manner, including both synthetic and natural degradable biomaterials. A biotechnological approach using fusion proteins that preserve the active domains of the GFs to stimulate regenerative cells, as well as containing matrix-specic binding motifs to be effectively loaded onto delivery systems, offers an exciting addition to the current arsenal of GF-based therapy in bone regeneration.
Financial & competing interests disclosure
This work was supported by the Priority Research Centers Program (grant no.: 2009-0093829) and World Class University program (grant no.: R31-10069) through the National Research Foundation funded by the Ministry of Education, Science and Technology. The authors have no other relevant afliations or nancial involvement with any organization or entity with a nancial interest in or nancial conict with the subject matter or materials discussed in the manuscript apart from those disclosed. No writing assistance was utilized in the production of this manuscript.
endothelial growth factor receptors. Mol. Biol. Rep. 38(2), 14431450 (2011). chondrogenesis invitro. Osteoarth. Cartil. 11(1), 5564 (2003).
11

Future perspective GFs are generally stored in the ECM of bone tissue. GFs, including BMPs, FGFs, VEGFs and IGFs, are released after injury and contribute to bone repair, and have consequently been proposed for use in bone regeneration applications. There is considerable potential to use these biofactors for regenerative medicine approaches that not only manage fracture healing, but also actively enhance the regenerative process. Indeed, BMPs have already been tested clinically and recombinant proteins are available on the market to enhance
References
Papers of special note have been highlighted as: n of interest nn of considerable interest
1 6

Kneser U, Schaefer DJ, Polykandriotis E, Horch RE. Tissue engineering of bone: the reconstructive surgeons point of view. J.Cell Mol. Med . 10(1), 719 (2006). Yu YY, Lieu S, Lu C, Colnot C. Bone morphogenetic protein 2 stimulates endochondral ossication by regulating periosteal cell fate during bone repair. Bone 47(1), 6573 (2010). Wang Q, Huang C, Xue M etal. Expression of endogenous BMP-2 in periosteal progenitor cells is essential for bone healing. Bone 48(3), 524532 (2011). Hanada K, Solchaga LA, Caplan AI etal. BMP-2 induction and TGF-beta 1 modulation of rat periosteal cell chondrogenesis. J. Cell. Biochem. 81(2), 284294 (2001). Hah YS, Jun JS, Lee SG etal. Vascular endothelial growth factor stimulates osteoblastic differentiation of cultured human periosteal-derived cells expressing vascular
7

Samee M, Kasugai S, Kondo H etal. Bone morphogenetic protein-2 (BMP-2) and vascular endothelial growth factor (VEGF) transfection to human periosteal cells enhances osteoblast differentiation and bone formation. J. Pharmacol. Sci. 108(1), 1831 (2008). Casap N, Venezia NB, Wilensky A etal. VEGF facilitates periosteal distractioninduced osteogenesis in rabbits: a microcomputerized tomography study. Tissue Eng. Part A 14(2), 247253 (2008). Gruber R, Karreth F, Frommlet F etal. Platelets are mitogenic for periosteumderived cells. J. Orthop. Res. 21(5), 941948 (2003). Stevens MM, Marini RP, Martin I etal. FGF-2 enhances TGF-beta 1-induced periosteal chondrogenesis. J. Orthop. Res. 22(5), 11141119 (2008). Fukumoto T, Sperling JW, Sanyal A etal. Combined effects of insulin-like growth factor-1 and transforming growth factor-beta1 on periosteal mesenchymal cells during

Malafaya PB, Silva GA, Reis RL. Drug delivery therapies I. General trends and its importance on bone tissue engineering applications. Curr. Opin. Solid State Mater. Sci. 6, 283295 (2002). formation. Eur. Spine J. 10(Suppl.2), S102S109 (2001).

12 Lind M, B nger C. Factors stimulating bone

13 Jadlowiec JA, Celil AB, Hollinger JO.

Bonetissue engineering: recent advances andpromising therapeutic agents. ExpertOpin. Biol. Ther. 3(3), 409423 (2003).
14

Yoon ST, Boden SD. Osteoinductive molecules in orthopaedics: basic science and preclinical studies. Clin. Orthop. Relat. Res. 395, 3343 (2002). Gallea S, Lallemand F, At A etal. Activation of mitogen-activated protein kinase cascades is involved in regulation ofbone morphogenetic protein-2-induced osteoblast differentiation in pluripotent C2C12 cells. Bone 28(5), 491498 (2001).

15

10

future science group

www.futuremedicine.com

379

REVIEW
16

Yun, Jang, Jeon etal.

Bruder SP, Fox BS. Tissue engineering of bone. Cell based strategies. Clin. Orthop. Relat. Res. 367(Suppl.), S68S83 (1999). Cook SD. Preclinical and clinical evaluation of osteogenic protein-1 (BMP-7) in bony sites. Orthopedics 22(7), 669671 (1999). Mackay AM, Beck SC, Murphy JM etal. Chondrogenic differentiation of cultured human mesenchymal stem cells from marrow. Tissue Eng. 4(4), 415428 (1998). Franceschi RT, Xiao G. Regulation of the osteoblast-specic transcription factor, Runx2: responsiveness to multiple signal transduction pathways. J. Cell. Biochem. 88(3), 446454 (2003). Molecular pathways regulating mobilization of marrow-derived stem cells for tissue revascularization. Trends Mol. Med . 9(3), 109117 (2003).

29 Luginbuehl V, Meinel L, Merkle HP, Gander

41 Lakshmi S, Katti DS, Laurencin CT.

17

B. Localized delivery of growth factors for bone repair. Eur. J. Pharm. Biopharm. 58(2), 197208 (2004).
30 Seeherman H, Wozney JM. Delivery of bone

Biodegradable polyphosphazenes for drug delivery applications. Adv. Drug Deliv. Rev. 55(4), 467482 (2003).
42 Lutolf MP, Weber FE, Schmoekel HG etal.

18

morphogenetic proteins for orthopedic tissue regeneration. Cytokine Growth Factor Rev. 16(3), 329345 (2005).
31

19

Anseth KS, Shastri VR, Langer R. Photopolymerizable degradable polyanhydrides with osteocompatibility. Nat. Biotechnol . 17(2), 156159 (1999). Herbst H, Pohle T. Fibrin glue, healing of gastric mucosal injury, and expression of growth factors: results from a human. Invivo study. Gastrointest. Endosc. 61(4), 560567 (2005).

Repair of bone defects using synthetic mimetics of collagenous extracellular matrices. Nat. Biotechnol . 21(5), 513518 (2003).
n

Describes the use of synthetic materials that are amenable to routine manufacture to replicate collagen extracellular matrices. Biodegradable poly-d,l-lactic acidpolyethylene glycol block copolymers as a BMP delivery system for inducing bone. J.Bone Joint Surg. Am . 83-A(Suppl. 1, Pt 2), S92S98 (2001).

32 Becker JC, Beckbauer M, Domschke W,

43 Saito N, Okada T, Horiuchi H etal.

20 Rabbany SY, Heissig B, Hattori K, Rafoo S.

33 Behravesh E, Yasko AW, Engel PS, Mikos

21

Uchida S, Sakai A, Kudo H etal. Vascular endothelial growth factor is expressed along with its receptors during the healing process of bone and bone marrow after drill-hole injury in rats. Bone 32(5), 491501 (2003). Vascular endothelial growth factor principally acts as the main angiogenic factor in the early stage of human osteoblastogenesis. J. Biochem. 133(5), 633639 (2003).

AG. Synthetic biodegradable polymers for orthopaedic applications. Clin. Orthop. Relat. Res. 367(Suppl.), S118S129 (1999).
34 Boyan BD, Lohmann CH, Somers A etal.

44 Saito N, Okada T, Horiuchi H etal. Local

bone formation by injection of recombinant human bone morphogenetic protein-2 contained in polymer carriers. Bone 32(4), 381386 (2003).
45

22 Furumatsu T, Shen ZN, Kawai A etal.

Potential of porous poly-d,l-lactide-coglycolide particles as a carrier for recombinant human bone morphogenetic protein-2 during osteoinduction invivo. J. Biomed. Mater. Res. 46(1), 5159 (1999).
35

Yoneda M, Terai H, Imai Y etal. Repair of an intercalated long bone defect with a synthetic biodegradable bone-inducing implant. Biomaterials 26(25), 51455152 (2005). medicine: from ceramics to calcium phosphate cements. Injury 31(Suppl. 4), 3747 (2000).

23 Canalis E, Agnusdei D. Insulin-like growth

factors and their role in osteoporosis. Calcif. Tissue Int. 58(3), 133134 (1996).
24 Putney SD, Burke PA. Improving protein

Cowan CM, Aalami OO, Shi YY etal. Bone morphogenetic protein 2 and retinoic acid accelerate invivo bone formation, osteoclast recruitment, and bone turnover. Tissue Eng. 11(34), 645658 (2005). etal. Effect of varied release kinetics of the osteogenic thrombin peptide TP508 from biodegradable, polymeric scaffolds on bone formation invivo. J. Biomed. Mater. Res. A 72(4), 343353 (2005).

46 Bohner M. Calcium orthophosphates in

36 Hedberg EL, Kroese-Deutman HC, Shih CK

47 Bouxsein ML, Turek TJ, Blake CA etal.

therapeutics with sustained-release formulations. Nat. Biotechnol . 16(2), 153157 (1998).


n

Recombinant human bone morphogenetic protein-2 accelerates healing in a rabbit ulnar osteotomy model. J. Bone Joint Surg. Am. 83-A(8), 12191230 (2001).
48 Polak SJ, Levengood SK, Sheeler MB etal.

Describes the importance of regulating the rate of release of active protein components from a formulated product. regeneration by basic broblast growth factor complexed with biodegradable hydrogels. Biomaterials 19(79), 807815 (1998).

37 Hollister SJ, Lin CY, Saito E etal.

25 Tabata Y, Yamada K, Miyamoto S etal. Bone

Engineering craniofacial scaffolds. Orthod.Craniofac. Res. 8(3), 162173 (2005).


38 Huang W, Carlsen B, Wulur I etal. BMP-2

Analysis of the roles of microporosity and BMP-2 on multiple measures of bone regeneration and healing in calcium phosphate scaffolds. Acta Biomater. 7(4), 17601771 (2011).
49 Jingushi S, Urabe K, Okazaki K etal.

26 Stenport VF, Roos-Jansaker AM, Renvert S

etal. Failure to induce supracrestal bone growth between and around partially inserted titanium implants using bone morphogenetic protein (BMP): an experimental study in dogs. Clin. Oral Implants Res. 14(2), 219225 (2003).
27 Aspenberg A, Albrektsson T, Thormgren KG.

exerts differential effects on differentiation of rabbit bone marrow stromal cells grown in two-dimensional and three-dimensional systems and is required for invitro bone formation in a PLGA scaffold. Exp. Cell. Res. 299(2), 325334 (2004).
39 Jones AA, Buser D, Schenk R, Wozney J,

Intramuscular bone induction by human recombinant bone morphogenetic protein-2 with beta-tricalcium phosphate as a carrier: invivo bone banking for muscle-pedicle autograft. J. Orthop. Sci. 7(4), 490494 (2002).
50 Laffargue P, Hildebrand HF, Rtaimate M

Local application of growth-factor IGF-1 to healing bone. Experiments with a titanium chamber in rabbits. Acta Orthop. Scand . 60(5), 607610 (1989).
28 Kirkeby OJ, Ekeland A. No effects of local

Cochran DL. The effect of rhBMP-2 around endosseous implants with and without membranes in the canine model. J.Periodontol . 77(7), 11841193 (2006).
40 Kim SE, Park JH, Cho YW etal. Porous

etal. Evaluation of human recombinant bone morphogenetic protein-2-loaded tricalcium phosphate implants in rabbits bone defects. Bone 25(Suppl. 2), S55S58 (1999).
51

somatomedin C on bone repair. Continuous infusion in rats. Acta Orthop. Scand . 63(4), 447450 (1992).

chitosan scaffold containing microspheres loaded with transforming growth factorbeta1: implications for cartilage tissue engineering. Acta J. Control. Release 91(3), 365374 (2003).

Ripamonti U, Ma SS, van den Heever B, Reddi AH. Osteogenin, a bone morphogenetic protein, adsorbed on porous hydroxyapatite substrata, induces rapid bone differentiation in calvarial defects of adult primates. Plast. Reconstr. Surg. 90(3), 382393 (1992).

380

Regen. Med. (2012) 7 (3)

future science group

Administration of growth factors for bone regeneration

REVIEW

52

Saito N, Okada T, Horiuchi H etal. Abiodegradable polymer as a cytokine delivery system for inducing bone formation. Nat. Biotechnol. 19(4), 332335 (2001).
65

differentiated phenotype of rabbit chondrocytes and osteoblastic MC3T3-E1 cells invitro. J. Bone Miner. Res. 6(12), 13731385 (1991). Cheng SL, Lou J, Wright NM etal. Invitro and invivo induction of bone formation using a recombinant adenoviral vector carrying the human BMP-2 gene. Calcif. Tissue Int. 68(2), 8794 (2001). Recombinant human bone morphogenetic protein-2 stimulates osteoblastic maturation and inhibits myogenic differentiation invitro. J. Cell Biol . 113(3), 681687 (1991).
n

induced by direct retroviral-based gene therapy with bone morphogenetic protein-4. Bone 32(6), 591601 (2003).
77 Ishihara A, Shields KM, Litsky AS etal.

53 Seeherman H, Li R, Bouxsein M etal.

rhBMP-2/calcium phosphate matrix accelerates osteotomy-site healing in a nonhuman primate model at multiple treatment times and concentrations. J. Bone Joint Surg. Am. 88(1), 144160 (2006).
54 Seeherman HJ, Bouxsein M, Kim H etal.

Osteogenic gene regulation and relative acceleration of healing by adenoviral mediated transfer of human BMP-2 or -6 in equine osteotomy and ostectomy models. J. Orthop. Res. 26(6), 764771 (2008).
78 Zhu W, Rawlins BA, Boachie-Adjei O etal.

66 Yamaguchi A, Katagiri T, Ikeda T etal.

Recombinant human bone morphogenetic protein-2 delivered in an injectable calcium phosphate paste accelerates osteotomy-site healing in a nonhuman primate model. J.Bone Joint Surg. Am. 86-A(9), 19611972 (2004).
55

Suzuki A, Terai H, Toyoda H etal. Abiodegradable delivery system for antibiotics and recombinant human bone morphogenetic protein-2: a potential treatment for infected bone defects. J. Orthop. Res. 24(3), 327332 (2006). Ectopic bone formation in titanium mesh loaded with bone morphogenetic protein and coated with calcium phosphate. Plast. Reconstr. Surg. 108(2), 434443 (2001).

Provides one of the rst reports on the potent effect of BMP-2 on ostoegenic induction. Osteogenic activity of the fourteen types of human bone morphogenetic proteins (BMPs). J. Bone Joint Surg. Am. 85-A(8), 15441552 (2003).

Combined bone morphogenetic protein-2 and-7 gene transfer enhances osteoblastic differentiation and spine fusion in a rodent model. J. Bone Miner. Res. 19(12), 20212032 (2004).
79 Alden TD, Beres EJ, Laurent JS etal. The use

67 Cheng H, Jiang W, Phillips FM etal.

of bone morphogenetic protein gene therapy in craniofacial bone repair. J. Craniofac. Surg. 11(1), 2430 (2000).
80 Bertone AL, Pittman DD, Bouxsein ML etal.

56 Vehof JW, Mahmood J, Takita H etal.

68 Ashinoff RL, Cetrulo CL, Galiano RD etal.

Bone morphogenic protein-2 gene therapy for mandibular distraction osteogenesis. Ann. Plast. Surg. 52(6), 585591 (2004).
69 Baltzer AW, Lattermann C, Whalen JD etal.

Adenoviral-mediated transfer of human BMP-6 gene accelerates healing in a rabbit ulnar osteotomy model. J. Orthop. Res. 22(6), 12611270 (2004).
81

57 Schmidmaier G, Wildemann B, Gbelein T

etal. Synergistic effect of IGF-I and TGF-beta1 on fracture healing in rats: single versus combined application of IGF-I and TGF-beta 1. Acta Orthop. Scand . 74(5), 604610 (2003).
58 Vonau RL, Bostrom MP, Aspenberg P, Sams

Genetic enhancement of fracture repair: healing of an experimental segmental defect by adenoviral transfer of the BMP-2 gene. Gene Ther. 7(9), 734739 (2000).
70 Southwood LL, Frisbie DD, Kawcak CE etal.

Park J, Ries J, Gelse K etal. Bone regeneration in critical size defects by cell-mediated BMP-2 gene transfer: a comparison of adenoviral vectors and liposomes. Gene Ther. 10(13), 10891098 (2003). Retroviral-based gene therapy with cyclooxygenase-2 promotes the union of bony callus tissues and accelerates fracture healing in the rat. J. Gene Med . 10(3), 229241 (2008).

82 Rundle CH, Strong DD, Chen ST etal.

AE. Combination of growth factors inhibits bone ingrowth in the bone harvest chamber. Clin. Orthop. Relat. Res. 386, 243251 (2001).
59 Ducy P, Karsenty G. The family of bone

Evaluation of Ad-BMP-2 for enhancing fracture healing in an infected defect fracture rabbit model. J. Orthop. Res. 22(1), 6672 (2004).
71 Egermann M, Lill CA, Griesbeck K etal.

83 Sykaras N, Opperman LA. Bone

morphogenetic proteins. Kidney Int. 57(6), 22072214 (2000).


60 Kirker-Head CA. Potential applications and

Effect of BMP-2 gene transfer on bone healing in sheep. Gene Ther. 13(17), 12901299 (2006).
72 Egermann M, Baltzer AW, Adamaszek S etal.

morphogenetic proteins (BMPs): how do they function and what can they offer the clinician? J. Oral Sci. 45(2), 5773 (2003).
84 Sigurdsson TJ, Nygaard L, Tatakis DN etal.

delivery strategies for bone morphogenetic proteins. Adv. Drug Deliv. Rev. 43(1), 6592 (2000).
61

Direct adenoviral transfer of bone morphogenetic protein-2 cDNA enhances fracture healing in osteoporotic sheep. Hum. Gene Ther. 17(5), 507517 (2006).
73 Betz OB, Betz VM, Nazarian A etal. Direct

Periodontal repair in dogs: evaluation of rhBMP-2 carriers. Int. J. Periodontics Restorative Dent. 16(6), 524537 (1996).
85 Rosen V. Harnessing the parathyroid

Reddi AH. Regulation of cartilage and bone differentiation by bone morphogenetic proteins. Curr. Opin. Cell Biol . 4(5), 850855 (1992). regeneration: potential role of bone morphogenetic proteins. J. Periodont. Res. 29(4), 225235 (1994).

percutaneous gene delivery to enhance healing of segmental bone defects. J. Bone Joint Surg. Am. 88(2), 355365 (2006).
74

hormone, Wnt, and bone morphogenetic protein signaling cascades for successful bone tissue engineering. Tissue Eng. Part B Rev. 17(6), 475479 (2011).
86 Wang JC, Kanim LE, Yoo S etal. Effect of

62 Ripamonti U, Reddi AH. Periodontal

63 Kawabata M, Imamura T, Miwazono K.

Betz OB, Betz VM, Nazarian A etal. Delayed administration of adenoviral BMP-2 vector improves the formation of bone in osseous defects. Gene Ther. 14(13), 10391044 (2007). segmental bone defects by direct percutaneous gene delivery: effect of vector dose. Hum. Gene Ther. 18(10), 907915 (2007).

regional gene therapy with bone morphogenetic protein-2-producing bone marrow cells on spinal fusion in rats. J. Bone Joint Surg. Am. 85-A(5), 905911 (2003).
87 Lee JY, Musqrave D, Pelinkovic D etal.

Signal transduction by bone morphogenetic proteins. Cytokine Growth Factor Rev. 99(1), 4961 (1998).
64 Hiraki Y, Inoue H, Shiqeno C etal. Bone

75 Betz VM, Betz OB, Glatt V etal. Healing of

Effect of bone morphogenetic protein-2expressing muscle-derived cells on healing of critical-sized bone defects in mice. J. Bone Joint Surg. Am. 83-A(7), 10321039 (2001).
88 Chang SC, Lin TM, Chung HY etal.

morphogenetic proteins (BMP-2 and BMP-3) promote growth and expression of the

76 Rundle CH, Miyakoshi N, Kasukawa Y etal.

Invivo bone formation in fracture repair

Large-scale bicortical skull bone regeneration using exvivo replication-defective adenoviral-

future science group

www.futuremedicine.com

381

REVIEW

Yun, Jang, Jeon etal.

mediated bone morphogenetic protein-2 gene-transferred bone marrow stromal cells and composite biomaterials. Neurosurgery 65(Suppl. 6), 7583 (2009).
89 Hsu WK, Suqiyama O, Park SH etal.

BMP2 therapy to accelerate bone healing in an equine osteotomy model. J. Orthop. Res. 28(3), 403411 (2010).
101 Koh JT, Zhao Z, Wang Z etal.

collagen gel. Laryngoscope 109(10), 15801584 (1999).


113 Welch RD, Jones AL, Bucholz RW etal.

Lentiviral-mediated BMP-2 gene transfer enhances healing of segmental femoral defects in rats. Bone 40(4), 931938 (2007).
90 Lieberman JR, Le LQ, Wu L etal. Regional

Combinatorial gene therapy with BMP2/7 enhances cranial bone regeneration. J. Dent. Res. 87(9), 845849 (2008).
102 Shen HC, Peng H, Usas A etal. Structural

Effect of recombinant human bone morphogenetic protein-2 on fracture healing in a goat tibial fracture model. J. Bone Miner. Res. 13(9), 14831490 (1998).
114 Suzuki Y, Tanihara M, Suzuki K etal.

gene therapy with a BMP-2-producing murine stromal cell line induces heterotopic and orthotopic bone formation in rodents. J.Orthop. Res. 16(3), 330339 (1998).
91

and functional healing of critical-size segmental bone defects by transduced muscle-derived cells expressing BMP4. J.Gene Med . 6(9), 984991 (2004).
103 Breitbart AS, Grande DA, Mason JM etal.

Alginate hydrogel linked with synthetic oligopeptide derived from BMP-2 allows ectopic osteoinduction invivo. J. Biomed. Mater. Res. 50(3), 405409 (2000).
115 Jiang T, Abdel-Fattah WI, Laurencin CT.

Lieberman JR, Daluiski A, Stevenson S etal. The effect of regional gene therapy with bone morphogenetic protein-2-producing bone-marrow cells on the repair of segmental femoral defects in rats. J. Bone Joint Surg. Am. 81(7), 905917 (1999). of critically sized femoral defects, using genetically modied mesenchymal stem cells from human adipose tissue. Tissue Eng. 11(12), 120129 (2005).

Gene-enhanced tissue engineering: applications for bone healing using cultured periosteal cells transduced retrovirally with the BMP-7 gene. Ann. Plast. Surg. 42(5), 488495 (1999).
104 Hidaka C, Goshi K, Rawlins B, Boachie-

Invitro evaluation of chitosan/poly(lactic acid-glycolic acid) sintered microsphere scaffolds for bone tissue engineering. Biomaterials 27(28), 48944903 (2006).
116 Kim LS, Park JW, Kwon IC, Baik BS, Cho

92 Peterson B, Zhang J, Iqlesias R etal. Healing

Adjei O, Crystal RG. Enhancement of spine fusion using combined gene therapy and tissue engineering BMP-7-expressing bone marrow cells and allograft bone. Spine 28(18), 20492057 (2003).
105 Wang EA, Rosen V, Cordes P etal.

BC. Role of BMP, betaig-h3, and chitosan in early bony consolidation in distraction osteogenesis in a dog model. Plast. Reconstr. Surg. 109(6), 19661977 (2002).
117 Park YJ, Lee YM, Park SN etal. Platelet

93 Tang TT, Lu B, Yue B etal. Treatment of

osteonecrosis of the femoral head with hBMP-2 -gene-modied tissue-engineered bone in goats. J.Bone Joint Surg. Br. 89(1), 127129 (2007).
94 Dai KR, Xu XL, Tang TT etal. Repairing of

Purication and characterization of other distinct bone-inducing factors. Proc. Natl Acad. Sci. USA 85(24), 94849488 (1988).
106 Wang EA, Rosen V, DAlessandro JS etal.

derived growth factor releasing chitosan sponge for periodontal bone regeneration. Biomaterials 21(2), 153159 (2000).
118 Woo BH, Fink BF, Paqe R etal. Enhancement

goat tibial bone defects with BMP-2 gene-modied tissue engineered bone. Calcif. Tissue Int. 77(1), 5561 (2005).
95 Xu XL, Tang T, Dai K etal. Immune

Recombinant human morphogenic protein induces bone formation. Proc. Natl Acad. Sci. USA 87(6), 22202224 (1990).
107 Arnander C, Westermark A, Veltheim R etal.

of bone growth by sustained delivery of recombinant human bone morphogenetic protein-2 in a polymeric matrix. Pharm. Res. 18(12), 17471753 (2001).
119 Becker J, Kirsch A, Schwarz F etal. Bone

response and effect of adenovirus-mediated human BMP-2 gene transfer on the repair of segmental tibial bone defects in goats. Acta Orthop. 76(5), 637646 (2005).
96 Hasharoni A, Zilberman Y, Turgeman G etal.

Three-dimensional technology and bone morphogenetic protein in frontal bone reconstruction. J. Craniofac. Surg. 17(2), 275279 (2006).
108 Chung C, Mesa J, Miller GJ etal. Effects of

apposition to titanium implants biocoated with recombinant human bone morphogenetic protein-2 (rhBMP-2). A pilot study in dogs. Clin. Oral Investig. 10(3), 217224 (2006).
120 Liu Y, de Groot K, Hunziker EB. BMP-2

Murine spinal fusion induced by engineered mesenchymal stem cells that conditionally express bone morphogenetic protein-2. J.Neurosurg. Spine 3(1), 4752 (2005).
97 Hsu WK, Wang JC, Liu NQ etal. Stem cells

auricular chondrocyte expansion on neocartilage formation in photocrosslinked hyaluronic acid networks. Tissue Eng. 12(9), 26652673 (2006).
109 Kandziora F, Bail H, Schmidmaier G etal.

liberated from biomimetic implant coatings induces and sustains direct ossication in an ectopic rat model. Bone 36(5), 745757 (2005).
121 Rachmiel A, Aizenbud D, Peled M.

from human fat as cellular delivery vehicles in an athymic rat posterolateral spine fusion model. J. Bone Joint Surg. Am. 90(5), 10431052 (2008).
98 Riew KD, Wright NM, Chenq S, Avioli LV,

Bone morphogenetic protein-2 application by a poly(d,l-lactide)-coated interbody cage: invivo results of a new carrier for growth factors. J. Neurosurg. 97(Suppl. 1), 4048 (2002).
110 Karageorgiou V, Meinel L, Hofmann S etal.

Enhancement of bone formation by bone morphogenetic protein-2 during alveolar distraction: an experimental study in sheep. J.Periodontol . 75(11), 15241531 (2004).
122 Sachse A, Wagner A, Keller M etal.

Lou J. Induction of bone formation using a recombinant adenoviral vector carrying the human BMP-2 gene in a rabbit spinal fusion model. Calcif. Tissue Int. 63(4), 357360 (1998).
99 Yi Y, Choi KB, Lim CL etal. Irradiated

Bone morphogenetic protein-2 decorated silk broin lms induce osteogenic differentiation of human bone marrow stromal cells. J.Biomed. Mater. Res. A 71(3), 528537 (2004).
111 Sanchez C, Arribart H, Guille MM.

Osteointegration of hydroxyapatitetitanium implants coated with nonglycosylated recombinant human bonemorphogenetic protein-2 (BMP-2) in aged sheep. Bone 37(5), 699710 (2005).
123 Schierano G, Canuto RA, Navone R etal.

human chondrocytes expressing bone morphogenetic protein 2 promote healing of osteoporotic bone fracture in rats. Tissue Eng. Part A 15(10), 28532863 (2009).
100 Ishihara A, Zekas LJ, Litsky AS, Weisbrode

Biomimetism and bioinspiration as tools for the design of innovative materials and systems. Nat. Mater. 4(4), 277288 (2005).
112 Toung JS, Ogle RC, Morgan RF, Lindsey

Biological factors involved in the osseointegration of oral titanium implants with different surfaces: a pilot study in minipigs. J. Periodontol . 76(10), 17101720 (2005).
124 Stenport VF, Johansson C, Heo SJ, Aspenberg

SE, Bertone AL. Dermal broblast-mediated

WH. Repair of a rodent nasal critical-size osseous defect with osteoblast augmented Regen. Med. (2012) 7 (3)

P, Albrektsson T. Titanium implants and BMP-7 in bone: an experimental model in the

382

future science group

Administration of growth factors for bone regeneration

REVIEW

rabbit. J. Mater. Sci. Mater. Med . 14(3), 247254 (2003).


125 Sumner DR, Turner TM, Urban RM, Virdi

healing potential of frontal and parietal bones. Bone 47(2), 281294 (2010).
137 Lee KM, Santos-Ruiz L, Ferretti P. A

engineering bone. Eur. Cell Mater. 15, 100114 (2008).


151 Mayr-Wohlfart U, Waltenberger J, Hausser H

AS, Inoue N. Additive enhancement of implant xation following combined treatment with rhTGF-beta 2 and rhBMP-2 in a canine model. J. Bone Joint Surg. Am. 88(4), 806817 (2006).
126 Arosarena OA, Collins WL. Defect repair in

single-point mutation in FGFR2 affects cell cycle and Tgfbeta signalling in osteoblasts. Biochim. Biophys. Acta 1802(3), 347355 (2010).
138 Gmez G, Korkiakoski S, Gonzlez MM etal.

etal. Vascular endothelial growth factor stimulates chemotactic migration of primary human osteoblasts. Bone 30(3), 472477 (2002).
152 Orlandini M, Spreaco A, Bardelli M etal.

the rat mandible with bone morphogenic protein 5 and prostaglandin E1. Arch. Otolaryngol. Head Neck Surg. 129(10), 11251130 (2003).
127 Hsieh CY, Hsieh HJ, Liu HC, Wang DM,

Effect of FGF and polylactide scaffolds on calvarial bone healing with growth factor on biodegradable polymer scaffolds. J.Craniofac. Surg. 17(5), 935942 (2006).
139 Cuevas P, de Paz V, Cuevas B etal.

Vascular endothelial growth factor-D activates VEGFR-3 expressed in osteoblasts inducing their differentiation. J. Biol. Chem. 281(26), 1796117967 (2006).
153 Li R, Li CH, Nauth A, McKee MD,

Hou LT. Fabrication and release behavior of a novel freeze-gelled chitosan/g-PGA scaffold as a carrier for rhBMP-2. Dent. Mater. 22(7), 622629 (2006).
128 Boden SD, Zdeblick TA, Sandhu HS, Heim

Osteopromotion for cranioplasty: anexperimental study in rats using acidic broblast growth factor. Surg. Neurol. 47(3), 242246 (1997).
140 Kawaguchi H. Bone fracture and the healing

SE. The use of rhBMP-2 in interbody fusion cages. Denitive evidence of osteoinduction in human: a preliminary report. Spine 25(3), 376381 (2000).
129 Burkus JK, Dorchak JD, Sanders DL.

mechanisms. Fibroblast growth factor-2 and fracture healing. Clin. Calcium 19(5), 653659 (2009).
141 Draenert GF, Draenert K, Tischer T.

Schemitsch EH. Effect of human vascular endothelial growth factor gene transfer on endogenous vascular endothelial growth factor mRNA expression in a rat broblast and osteoblast culture model. J. Orthop. Trauma 24(9), 547551 (2010).
154 Lee SH, Shin H. Matrices and scaffolds for

Radiographic assessment of interbody fusion using recombinant human bone morphogenetic protein type2. Spine 28(4), 372377 (2003).
130 Govender S, Csimma C, Genant HK etal.

Dose-dependent osteoinductive effects of bFGF in rabbits. Growth Factors 27(6), 419424 (2009).
142 Casper CL, Yang W, Farach-Carson MC,

delivery of bioactive molecules in bone and cartilage tissue engineering. Adv. Drug Deliv. Rev. 59(45), 339359 (2007).
155 Devescovi V, Leonardi E, Ciapetti G, Cenni

E. Growth factors in bone repair. Chir. Organi. Mov. 92(3), 161168 (2008).
156 Kronenberg HM. Developmental regulation

Recombinant human bone morphogenetic protein-2 for treatment of open tibial fracture: a prospective, controlled, randomized study of four hundred and fty patients. J. Bone Joint Surg. Am. 84-A(12), 21232134 (2002).
131 Johnson EE, Urist MR, Finermans GA.

Rabolt JF. Coating electropun collagen and gelatin bers with perlecan dimain I for increased growth binding. Biomacromolecules 8(4), 11161123 (2007).
143 Ichinohe N, Kuboki Y, Tabata Y. Bone

of the growth plate. Nature 423(6937), 332336 (2003).


157 Gerber HP, Vu TH, Ryan AM etal. VEGF

regeneration using titanium nonwoven fabrics combined with FGF-2 release from gelatin hydrogel microspheres in rabbit skull defects. Tissue Eng. Part A 14(10), 16631671 (2008).
144 Jeon E, Jang JH. Protein engineering of a

couples hypertrophic cartilage remodeling, ossication and angiogenesis during endochondral bone formation. Nat. Med. 5(6), 623628 (1999).
158 Cui F, Wang X, Liu X etal. VEGF and BMP-6

Repair of segmental defects of the tibia with cancellous bone grafts augmented with human bone morphogenetic protein. A preliminary report. Clin. Orthop. Relat. Res. 236, 249257 (1988).
132 Vaccaro AR, Patel T, Fischgrund J etal.

broblast growth factor-2 protein for targeting to bone mineral hydroxyapatite. Protein Pep. Lett. 16(6), 664667 (2009).
145 Murukesh N, Dive C, Jayson GC. Biomarkers

enhance bone formation mediated by cloned mouse osteoprogenitor cells. Growth Factors 28(5), 306317 (2010).
159 Street J, Bao M, deGuzman L etal. Vascular

Apilot safety and efcacy study of OP-1 putty (rhBMP-7) as an adjunct to iliac crest autograft in posterolateral lumbar fusions. Eur. Spine J. 12(5), 495500 (2003).
133 Yun YR, Won JE, Joen E etal. Fibroblast

of angiogenesis and their role in the development of VEGF inhibitors. Br.J.Cancer 102(1), 818 (2010).
146 Tammela T, Enholm B, Alitalo K, Paavonen

endothelial growth factor stimulates bone repair by promoting angiogenesis and bone turnover. Proc. Natl Acad. Sci. USA 99(15), 96569661 (2002).
160 Phillips AM. Overview of the fracture healing

growth factors: biology, function, and application for tissue regeneration. J.Tissue Eng. 2010, 218142 (2010).
134 Andrades JA, Wu LT, Hall FL, Nimni ME,

K. The biology of vascular endothelial growth factors. Cardiovasc. Res. 65(3), 550563 (2005).
147 Hsiong SX, Mooney DJ. Regeneration of

cascade. Injury 36(Suppl. 3), S5S7 (2005).


161 Eckardt H, Ding M, Lind M etal.

Becerra J. Engineering, expression, and renaturation of a collagen-targeted human bFGF fusion protein. Growth Factors 18(4), 261275 (2001).
135 Eswarakumar VP, Lax I, Schlessinger J.

vascularized bone. Periodontol. 2000 41, 109122 (2006).


148 Kempen DH, Lu L, Heijink A etal. Effect of

Recombinant human vascular endothelial growth factor enhances bone healing in an experimental nonunion model. J. Bone Joint Surg. Br. 87(10), 14341438 (2005).
162 Kaigler D, Wang Z, Horger K, Mooney DJ,

local sequential VEGF and BMP-2 delivery on ectopic and orthotopic bone regeneration. Biomaterials 30(14), 28162825 (2009).
149 Kleinheinz J, Stratmann U, Joos U,

Cellular signaling by broblast growth factor receptors. Cytokine Growth Factor Rev. 16(2), 139149 (2005).
136 Behr B, Panetta NJ, Longaker MT, Quarto N.

Krebsbach PH. VEGF scaffolds enhance angiogenesis and bone regeneration in irradiated osseous defects. J. Bone Miner. Res. 21(5), 735744 (2006).
163 Leach JK, Kaigler D, Wang Z, Krebsbach PH,

Wiesmann HP. VEGF-activated angiogenesis during bone regeneration. J. Oral Maxillofac. Surg. 63(9), 13101316 (2005).
150 Kanczler JM, Oreffo RO. Osteogenesis

Different endogenous threshold levels of broblast growth factor-ligands determine the

andangiogenesis: the potential for

Mooney DJ. Coating of VEGF-releasing scaffolds with bioactive glass for angiogenesis and bone regeneration. Biomaterials 27(17), 32493255 (2006).

future science group

www.futuremedicine.com

383

REVIEW

Yun, Jang, Jeon etal.

164 Huang YC, Kaigler D, Rice KG, Krebsbach

PH, Mooney DJ. Combined angiogenic and osteogenic factor delivery enhances bone marrow stromal cell-driven bone regeneration. J. Bone Miner. Res. 20(5), 848857 (2005).
165 Sell C, Baserga R, Rubin R. Insulin-like

PI3K signaling pathway through the hypoxiaincucible factor-2alpha. Endocrinology 143(2), 420425 (2002).
177 Chen FM, Zhao YM, Wu H etal.

primary human osteoblast. J. Cell. Biochem. 86(2), 348356 (2002).


188 Standal T, Abildgaard N, Faqerli UM etal.

growth factor I (IGFI) and the IGF-I receptor prevent etoposide-induced apoptosis. Cancer Res. 55(2), 303306 (1995).
166 Canalis E. Growth factor control of bone

Enhancement of periodontal tissue regeneration by locally controlled delivery of insulin-like growth factor-I from dextran-cogelatin microspheres. J. Control. Release 114(2), 209222 (2006).
178 Meinel L, Zoidis E, Zapf J etal. Localized

HGF inhibits BMP-induced osteoblastogenesis: possible implications for the bone disease of multiple myeloma. Blood 109(7), 30243030 (2010).
189 Yarram SJ, Tasman C, Gidley J etal.

mass. J. Cell. Biochem. 108(4), 769777 (2009).


167 Elis S, Courtland HW, Wu Y etal. Elevated

insulin-like growth factor I delivery to enhance new bone formation. Bone 33(4), 660672 (2003).
179 Srouji S, Rachmiel A, Blimenfeld I, Livne E.

Epidermal growth factor and calcitriol synergistically induce osteoblast maturation. Mol. Cell. Endocrinol . 220(12), 920 (2010).
190 Laamme C, Curt S, Rouabhia M.

serum IGF-1 levels synergize PTH action on the skeleton only when the tissue IGF-1 axis is intact. J. Bone Miner. Res. 25(9), 20512058 (2010).
168 Zoidis E, Ghirlanda-Keller C, Schmid C.

Mandibular defect repair by TGF-beta and IGF-1 released from a biodegradable osteoconductive hydrogel. J. Craniomaxillofac. Surg. 33(2), 7984 (2005).
180 Schmidmaier G, Wildemann B, Ostapowicz

Epidermal growth factor and bone morphogenetic proteins upregulate osteoblast proliferation and osteoblastic markers and inhibit bone nodule formation. Arch. Oral Biol . 55(9), 689701 (2010).
191 Zhang X, Tamasi J, Lu X etal. Epidermal

Stimulation of glucose transport in osteoblastic cells by parathyroid hormone and insulin-like growth factor I. Mol. Cell. Biochem. 348(12), 3342 (2010).
169 Playford MP, Bicknell D, Bodmer WF,

D etal. Long-term effects of local growth factor (IGF-I and TGF-beta 1) treatment on fracture healing. A safety study for using growth factors. J. Orthop. Res. 22(3), 514519 (2004).
181 Wildemann B, Kandziora F, Krummrey G

growth factor receptor plays an anabolic role in bone metabolism invivo. J. Bone Miner. Res. 26(5), 10221034 (2011).
192 Deng M, Zhang B, Wang K etal. Mechano

Macaulay VM. Insulin-like growth factor 1 regulates the location, stability, and transcriptional activity of beta-catenin. Proc. Natl Acad. Sci. USA 97(22), 1210312108 (2000).
170 Eingartner C, Coerper S, Fritz J etal. Growth

factors in distraction osteogenesis. Immunohistological pattern of TGF-beta1 and IGF-1 in human callus induced by distraction osteogenesis. Int. Orthop. 23(5), 253259 (1999).
171 Nakasaki M, Yoshioka K, Miyamoto Y etal.

etal. Local and controlled release of growth factors (combination of IGF-I and TGFbetaI, and BMP-2 alone) from a polylactide coating of titanium implants does not lead to ectopic bone formation in sheep muscle. J.Control. Release 95(2), 249256 (2004).
182 Toung JS, Ogle RC, Morgan RF, Lindsey

growth factor E peptide promotes osteoblasts proliferation and bone-defect healing in rabbits . Int. Orthop. 35(7), 10991106 (2010).
193 Dai Z, Wu F, Yeugn EW, Li Y. IGF-IEc

expression, regulation and biological function in different tissues. Growth Horm. IGF Res. 20(4), 275281 (2010).
194 Milingos DS, Philippou A, Armakolas A etal.

WH. Insulinlike growth factor 1- and 2-augmented collagen gel repair of facial osseous defects. Arch. Otolaryngol. Head Neck Surg. 125(4), 451455 (1999).
183 Hossain M, Irwin R, Baumann MJ, McCabe

IGF-I secreted by osteoblasts acts as a potent chemotactic factor for osteoblasts. Bone 43(5), 869879 (2008).
172 Yakar S, Courtland HW, Clemmons D.

Insulin-like growth factor-1Ec (MGF) expression in eutopic and ectopic endometrium: characterization of the MGF E peptide actions invitro. Mol. Med . 17(12), 2128 (2010).
195 Carpenter V, Matthews K, Devlin G etal.

IGF-1 and bone: new discoveries from mouse models. J. Bone Miner. Res. 25(12), 22672276 (2010).
173 Ahmed SF, Farquharson C. The effect of GH

LR. Hepatocyte growth factor (HGF) adsorption kinetics and enhancement of osteoblast differentiation on hydroxyapatite surfaces. Biomaterial 26(15), 25952602 (2005).
184 Zambonin G, Camerino C, Greco G etal.

Mechano-growth factor reduces loss of cardiac function in acute myocardial infarction. Heart Lung Circ. 17(1), 3339 (2008).
196 Rao MV, Berk J, Almojaly SA etal. Effects of

and IGF1 on linear growth and skeletal development and their modulation by SOCS proteins. J. Endocrinol . 206(3), 249259 (2010).
174 Liu JP, Baker J, Perkins AS, Robertson EJ,

Hydroxyapatite coated with hepatocyte growth factor (HGF) stimulates human osteoblasts invitro. J. Bone Joint Surg. Br. 82(3), 457460 (1998).
185 Grano M, Galimi F, Zambonin G etal.

platelet-derived growth factor, vitamin D and parathyroid hormone on osteoblasts derived from cancer patients on chronic bisphosphonate therapy. Int. J. Mol. Med . 23(3), 407413 (2010).
197 Chang PC, Seol YJ, Cirelli JA etal. PDGF-B

Efstratiadis A. Mice carrying null mutations of the genes encoding insulin-like growth factor I (Igf-1) and type1 IGF receptor (Igf1r). Cell 75(1), 5972 (1993).
175 Kawai M, Rosen CJ. The IGF-I regulatory

Hepatocyte growth factor is a coupling factor for osteoclasts andosteoblasts invitro. Proc. Natl Acad. Sci. USA 93(15), 76447648 (1996).
186 Kawasaki T, Niki Y, Miyamoto T etal.

gene therapy accelerates bone engineering and oral implant osseointegration. Gene Ther. 17(1), 95104 (2008).
198 Young CS, Bradica G, Hart CE et al.

system and its impact on skeletal and energy homeostasis. J. Cell. Biochem. 111(1), 1419 (2010).
176 Akeno N, Robins J, Zhang M, Czyzyk-

Theeffect of timing in the administration of hepatocyte growth factor to modulate BMP-2-induced osteoblast differentiation. Biomaterials 31(6), 11911198 (2009).
187 Viereck V, Siggelkow H, Tauber S etal.

Preclinical toxicology studies of recombinant human platelet-derived growth factor-BB either alone or in combination with beta-tricalcium phosphate and type I collagen. J. Tissue Eng. 2010, 246215 (2010).
199 Zhang J, Cao R, Zhang Y etal. Differential

Krzeska MF, Clemens TL. Induction of vascular endothelial growth factor by IGF-1 in osteoblast-like cells is mediated by the

Differential regulation of Cbfa1/Funx2 and osteocalcin gene expression by vitamin-D3, dexamethasone, and local growth factors in

roles of PDGFR- a and PDGFR-b in angiogenesis and vessel stability. FASEB J. 23(1), 153163 (2009).

384

Regen. Med. (2012) 7 (3)

future science group

Administration of growth factors for bone regeneration

REVIEW

200 Nash TJ, Howlett CR, Martin C etal. Effect

of platelet-derived growth factor on tibial osteotomies in rabbits. Bone 15(2), 203208 (1994).
201 Lee YM, Park YJ, Lee SJ etal. The bone

having collagen-binding activity (CBD HGF) accelerates re-endothelialization and intimal hyperplasia in balloon-injured rat carotid artery. J. Atheroscler. Thromb. 14(4), 185191 (2007).
206 Andrades JA, Han B, Becerra J etal.

209 Nevins M, Giannobile WV, McGuire MK

regenerative effect of platelet-derived growth factor-BB delivered with a chitosan/tricalcium phosphate sponge carrier. J. Periodontol . 71(3), 418424 (2000).
202 Yoneda A, Asada M, Oda Y, Suzuki M,

etal. Platelet-derived growth factor (rhPDGF-BB) stimulates bone ll and rate of attachment level gain. Results of a large, multicenter randomized controlled trial. J.Periodontol . 76(12), 22052215 (2005).
nn

Imamura T. Engineering of an FGFproteoglycan fusion protein with heparin-independent, mitogenic activity. Nat. Biotechnol . 18(6), 641644 (2000).
203 Ishikawa T, Eguchi M, Wada M etal.

Arecombinant human TGF-beta 1 fusion protein with collagen-binding domain promotes migration, growth, and differentiation of bone marrow mesenchymal cells. Exp. Cell Res. 250(2), 485498 (1999).
207 Friedlaender GE, Perry CR, Cole JD etal.

Reports the successful outcome of the delivery of recombinant human PDGF in 180 patients with periodontal bone loss, demonstrating a successful transition from the bench to the bedside.

Establishment of a functionally active collagen-binding vascular endothelial growth factor fusion protein in situ. Arterioscler. Thromb. Vasc. Biol. 26(9), 19982004 (2006).
204 Ishikawa T, Terai H, Yamamoto T, Harada K,

Osteogenic protein-1 (bone morphogenetic protein-7) in the treatment of tibial nonunions. J. Bone Joint Surg. Am. 83A(Suppl.1, Pt2), S151S158 (2001).
208 Carragee EJ, Hurwitz EL, Weiner BK.

Patent
301 Nies B, Dingeldein E, Wahlig H.

Darmstadt, Germany, Inc. US006118043A (2000).

Kitajima T. Delivery of a growth factor fusion protein having collagen-binding activity to wound tissues. Artif. Organs 27(2), 147154 (2003).
205 Ohkawara N, Ueda H, Shinozaki S etal.

Acritical review of recombinant human bone morphogenetic protein-2 trials in spinal surgery: emerging safety concerns and lessons learned. Spine J. 11(6), 471491 (2011).
nn

Website
401 International Standards Organisation.

Hepatocyte growth factor fusion protein

Analyzes current clinical trial and patient follow-up data and raises safety concerns that will aid our understanding of the requirements for the development of targeted growth factor delivery methods.

Biological evaluation of medical devices part 1: evaluation and testing. ISO 10993-1 (1997). www.fda.gov/MedicalDevices/ DeviceRegulationandGuidance/ GuidanceDocuments/ucm080735.htm

future science group

www.futuremedicine.com

385

You might also like