You are on page 1of 11

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/266325190

Fracture healing: Mechanisms and interventions

Article  in  Nature Reviews Rheumatology · September 2014


DOI: 10.1038/nrrheum.2014.164 · Source: PubMed

CITATIONS READS

450 15,440

2 authors, including:

Louis C Gerstenfeld
Boston University
271 PUBLICATIONS   17,882 CITATIONS   

SEE PROFILE

Some of the authors of this publication are also working on these related projects:

BET bromodomain proteins: their role in obesity, inflammation and cancer View project

Roles for lysyl oxidase in diabetic complications View project

All content following this page was uploaded by Louis C Gerstenfeld on 10 November 2014.

The user has requested enhancement of the downloaded file.


REVIEWS
Fracture healing: mechanisms and interventions
Thomas A. Einhorn and Louis C. Gerstenfeld
Abstract | Fractures are the most common large-organ, traumatic injuries to humans. The repair of bone
fractures is a postnatal regenerative process that recapitulates many of the ontological events of embryonic
skeletal development. Although fracture repair usually restores the damaged skeletal organ to its pre-injury
cellular composition, structure and biomechanical function, about 10% of fractures will not heal normally.
This article reviews the developmental progression of fracture healing at the tissue, cellular and molecular
levels. Innate and adaptive immune processes are discussed as a component of the injury response, as
are environmental factors, such as the extent of injury to the bone and surrounding tissue, fixation and the
contribution of vascular tissues. We also present strategies for fracture treatment that have been tested
in animal models and in clinical trials or case series. The biophysical and biological basis of the molecular
actions of various therapeutic approaches, including recombinant human bone morphogenetic proteins
and parathyroid hormone therapy, are also discussed.
Einhorn, T. A. & Gerstenfeld, L. C. Nat. Rev. Rheumatol. advance online publication 30 September 2014; doi:10.1038/nrrheum.2014.164

Introduction
Fracture healing and bone repair are postnatal processes differentiation of stem cells that form skeletal and vascu­
that mirror many of the ontological events that take place lar tissues. Immediately adjacent to the fracture line, a
during embryonic development of the skeleton and have cartilaginous callus will form. Peripheral to this central
been extensively reviewed elsewhere.1–5 The recapitula- region, at the edges of the new cartilage tissues, the perio­
tion of these ontological processes is believed to make steum swells and primary bone formation is initiated.7,8
fracture healing one of the few postnatal processes that is Concurrent with cartilage tissue development, cells that
truly regenerative, restoring the damaged skeletal organ will form the nascent blood vessels that supply the new
to its pre-injury cellular composition, structure and bio­ bone are recruited and differentiate in the surrounding
mechanical function. Interestingly, a comparison of the muscle sheath.9,10 The increases in the vascular bed that
transcriptome of mouse callus tissues across a 21‑day surrounds and then grows into the callus are further
period of fracture healing showed that about one-third of reflected by the increased blood flow into the area of tissue
the mouse homologues of the genes expressed by human repair. As chondrocyte differentiation progresses, the
embryonic stem cells are preferentially induced. Many of cartilage extracellular matrix undergoes mineralization
the homeotic genes that control appendicular limb devel- and the anabolic phase of fracture repair terminates with
opment also show increased expression during fracture chondrocyte apoptosis.11,12 The histological and c­ellular
healing.6 Finally, all the primary morpho­genetic pathways progression of these events are shown in Figure 1.
that are active during embryonic skeletal develop­ment are The anabolic phase is followed by a prolonged phase in
expressed in fracture calluses,6 and have therefore become which catabolic activities predominate, and is character­
the focal point of efforts to develop new therapies. In this ized by a reduction in the volume of the callus tissues.
Review, we place these biological processes in the context of During this phase of predominately catabolic activity,
how trauma and the immune system, as a component of the such as cartilage resorption, specific anabolic processes
Orthopaedic Surgery,
Boston University injury response, are related to the developmental aspects of continue to take place; secondary bone formation is initi-
Medical Centre, fracture healing. We then review the relationships between ated as the cartilage is resorbed and primary angiogenesis
Doctor’s Office
Building Suite 808,
ontogeny and the recovery of skeletal function. Finally, we continues as the nascent bone tissues replace the cartilage.
720 Harrison Avenue, focus on speci­fic biophysical, local and systemic therapies Subsequently, when bone remodelling begins, the first
Boston, MA 02118, that have been used to promote fracture healing and on the mineralized matrix produced during primary bone for-
USA (T.A.E.).
Orthopaedic Surgery, various b­iological processes they promote. mation is resorbed by osteoclasts, and then the secondary
Boston University bone laid down during the period of cartilage resorption
School of Medicine,
72 East Concord
Phases of fracture healing is also resorbed. As the bony callus tissue continues to be
Street, E243, Fracture healing and skeletal tissue repair involve an resorbed, this prolonged period is characterized by coupled
Boston, MA 02118, ini­tial anabolic phase characterized by an increase in cycles of osteoblast and osteoclast activity in which the
USA (L.C.G.).
tis­sue volume related to the de novo recruitment and callus tissues are remodelled to the bone’s original cortical
Correspondence to: structure (termed ‘coupled remodelling’ here). During this
T.A.E.
thomas.einhorn@ Competing interests period, the marrow space is re-established and the origi-
bmc.org The authors declare no competing interests. nal marrow structure of haemato­poietic tissue and bone

NATURE REVIEWS | RHEUMATOLOGY ADVANCE ONLINE PUBLICATION  |  1


© 2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

Key points or through the direct actions of these cells on immune-


cell populations, including T cells.25,26 Such effects suggest
■■ Fractures are the most common large-organ, traumatic injuries in humans
that these cells impart immune tolerance throughout the
and approximately 10% do not heal properly
■■ Fracture healing involves an anabolic phase of increasing tissue volume, which early stages of endochondral bone formation and provide
forms new skeletal tissues, followed by a prolonged catabolic phase in which protection to the developing tissues by suppressing allo-
tissue is remodelled to the original structure proliferation of T cells during stem-cell recruitment
■■ Fracture healing is regulated by the nature and extent of trauma, the stability and cartilage formation.19 At different stages of fracture
of fracture fixation and biological processes, including immunological and healing, cytokines with inflammatory and immune func-
developmental processes associated with skeletal ontology tions, including IL‑1β,19 IL‑6,27,28 IL‑17F,19,29 IL‑2319 and
■■ Multiple strategies, involving biophysical, local and systemic cell-based
TNF,12,30 are variably expressed and have different effects.
systemic therapies that manipulate the morphogenetic processes that control
skeletal development are used to promote healing
During the period of acute inflammation immediately
■■ The two most widely examined therapies for biologically enhancing fracture after injury, TNF and IL‑6 recruit cells needed for tissue
healing are bone morphogenetic proteins, which act locally, and parathyroid regeneration and their complete absence has been shown
hormone, which acts systemically to delay skeletogenic mesenchymal stem cell differentia-
■■ To advance the field of skeletal healing and to set the stage for developing new tion.12,27–30 On the other hand, if inflammation remains
local and systemic therapies, conditions for skeletogenic stem cell recruitment unresolved, such as with a bacterial infection at the injury
and differentiation need to be optimized site, healing can fail.31 In the context of chronic inflamma-
tion, such as in streptozotocin-induced diabetes mellitus
a b c in mice, the cartilage callus is prematurely removed during
fracture healing,32,33 and in mouse models of systemic
lupus erythe­matosus (SLE), osteoclast activity and bone
turnover are high.19 In both of these disease models, less
primary and secondary bone accumulation occurs during
fracture healing than in control mice. Anti-inflammatory
biologic agents and antiresorptive agents might therefore
have therapeutic value in fracture healing for patients with
Figure 1 | Histology of early stages of mouse femur fracture repair. a | The either SLE or diabetes mellitus.34
inflammatory stage of fracture repair 24 h after injury. Sections were immunoreacted
Evidence for direct T‑cell involvement in the control of
with anti-TNF antibodies to show innate immune responses to the injury of both the
periosteum and marrow cell populations (brown) with haematoxylin counterstaining
fracture healing comes from studies that showed nega­tive
(blue). b | Late inflammatory stage 3 days after injury, cellular streaming of undefined effects after lymphocyte depletion.35–38 Studies of fracture
fibrous cells and early angiogenic cells forming small vessels are evident at the healing in Rag1–/– mice, which lack T cells and B cells,
fracture site. Stained with haematoxylin and eosin. c | The late endochondral stage, showed that although cartilage maturation and replace-
14 days after injury. The section was stained for tartrate-resistant acid phosphatase ment was delayed and overall less mineralized tissue accu-
to show the recruitment of resorptive osteoclasts (bright red). mulated,29 these mice recovered mechanical function earlier
than wild-type mice.39 γδ T cells, which can detect the prod-
is regenerated. In the final period of the catabolic phase, ucts of stressed or damaged cells even in the absence of
extensive vascular remodelling takes place in which the antigen presentation,40 are also involved in fracture repair.
increased vascular bed regresses and the high vascular flow In other studies, mice deficient in γδ T cells have shorter
rate returns to its pre-injury level.13,14 Although these pro- times to fracture union and earlier development of mature
cesses take place consecutively, they overlap substantially fracture calluses than γδ‑T‑cell-sufficient mice.41 Finally, in
and are a continuum of changing cell populations and sig- a model of SLE, in which mice have defective Fas receptors
nalling processes within the regenerating tissue. A tempo­ (TNF receptor superfamily member 6), the percentage of
ral overview of the biological events of fracture healing, and iTREG cells was increased in both callus and bone tissues
the cell types involved at each stage of fracture healing, are during the period of active cartilage formation, while at the
presented in Figure 2. same time the number of activated T cells in these tissues
decreased, despite increased numbers of activated T cells
Control of fracture healing in the spleen, indicative of an autoimmune condition.19
Innate and adaptive immune functions These results suggest that activated iTREG cells are chon-
Both innate and adaptive immune processes are essen- droprotective and are consistent with studies showing
tial during the anabolic and catabolic phases of fracture that mesenchymal stem cells induce both the differen-
healing. In the initial inflammatory stage after injury, tiation and immunosuppressive function of iTREG cells.42,43
specific cell-mediated immune functions remove necro­ Pharmacological factors that alter immune function and
tic tissues, promote angiogenesis and initiate repair.15–17 inflammation might have negative and positive effects
Interestingly, fracture leads to suppression of the immune on fracture healing and are important to consider in the
system,18 with a local increase in the number of induced context of specific comorbidities that affect regeneration.34
T regulatory (iTREG) cells that suppress active adaptive
immune responses within the fracture callus.19 Studies Stem cell origins
have further shown that mesenchymal stem cells actively The extent of external soft-tissue and hard-tissue
maintain a hypoimmunogenic state20,21 through the pro- trauma,44,45 and the mechanical strain produced by thera-
duction of immunosuppressive paracrine factors,22–24 peutic interventions, will each effect the origin of the stem

2  |  ADVANCE ONLINE PUBLICATION www.nature.com/nrrheum


© 2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

Periosteum Marrow Cortical bone

Day 0–3 Day 3–5 Day 5–10 Day 10–16 Day 16–21 Day 21–35

Soft callus/ Hard callus/


unmineralized Fibrous Hard callus/ remodelled
Haematoma Haematoma cartilage tissue secondary bone bone
Monocyte PMN Hypertrophic Myelopoietic
chondrocyte cell
T cell
Chondrocyte
Osteoclast Osteocyte

Haematopoietic
Macrophage B cell Stem cell Osteoblast cell
Anabolic phase
Catabolic phase
Inflammatory stage
Endochondral stage
Coupled remodelling stage

Figure 2 | Femur fracture repair. The major metabolic phases (blue bars) of fracture healing overlap with biological stages
(brown bars). The primary metabolic phases (anabolic and catabolic) of fracture healing are presented in the context of
three major biological stages (inflammatory, endochondral bone formation and coupled remodelling) that encompass these
phases. The primary cell types that are found at each stage, and the time span of their prevalence in each stage, are
denoted. The time scale of healing is equivalent to a mouse closed femur fracture fixed with an intramedullary rod.
Abbreviations: BMP, bone morphogenetic protein; BMPR, bone morphogenetic protein receptor; DKK1, Dickkopf-related
protein 1; LRP, LDL-receptor-related protein; MSC, mesenchymal stem cell; PMN, polymorphonuclear leukocyte; PTH,
parathyroid hormone; PTHrP, parathyroid-hormone-related protein; RANKL, receptor activator of nuclear factor κB ligand.

cells that contribute to healing, the extent of cartilage-cell that can be repaired is limited; too much damage to the
versus bone-cell differentiation and the progression of muscle and periosteum can overcome the supply of tissue-­
fracture healing.46–48 Results from transgenic lineage track- regenerative stem cells. Also important is to consider the
ing have shown that the fracture callus is formed mostly extent to which the vascular tissues in the surrounding
of cells from the periosteum.49 Other studies showed that muscle sheath are compromised; failure of angiogenesis
periosteal cells specifically respond to bone morphogenetic after f­racture or osteotomy can lead to nonunion.44
protein 2 (BMP‑2) to promote both chondrogenesis and
osteogenesis, whereas cells in the marrow space will form Stability
only bone in response to BMP‑2.50 Mechanisms controlling The overall stability of the fixation and immobilization
cortical bone repair might, therefore, be different to those of the fracture will also affect the patterns of skeleto­genic
that repair and remodel trabecular bone, and those that stem cell differentiation into chondrocytes or osteo­blasts,
take place in the medullary space. with more-extensive cartilage tissue formation associ­ated
The extent and type of trauma that accompanies a frac- with less stability, and increasing stability with more bone
ture will also affect the tissue source from which stem tissue.48 Interestingly, when fractures are not stably fixed,
cells that contribute to the callus are derived. Transgenic angiogenesis is initially increased.52–54 Excessive inter-
mouse studies using a reporter gene conditionally acti- fragmentary instability, however, will impede carti­lage
vated in muscle stem cells showed that these cells do not replacement, diminish angiogenesis and prevent bone
contribute substantially to callus formation in a model of from bridging the fracture gap.54 Therefore, an optimal
closed tibial fracture. By contrast, studies of open tibial ‘window’ of interfragmentary motion seems to be needed
fractures, in which the periosteum was surgically stripped to enable normal calluses to develop and stably bridge a
from the bone with additional muscle fenestration, showed fracture. Together, these mouse studies have considerable
that almost half the cells in the callus were derived from bearing on the potential application of treatments, such
the surrounding muscle.51 Thus, the extent of injury as BMP therapy, to human fractures. The data indicate

NATURE REVIEWS | RHEUMATOLOGY ADVANCE ONLINE PUBLICATION  |  3


© 2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

Box 1 | Tested methods of enhancing fracture healing Review focuses on those strategies that have undergone
rigorous scientific testing in preclinical animal studies as
Biophysical enhancement
well as clinical trials or case series. A summary of experi-
Electromagnetic field stimulation72–76
mentally tested approaches to promote fracture healing is
Low-intensity pulsed ultrasound stimulation77–79
presented in Box 1.
Locally applied biological enhancement71
Osteogenic materials
■■ Autologous bone81 Biophysical enhancement
■■ Autologous bone marrow71 Electromagnetic fields have been investigated as a means
Osteoconductive materials of skeletal repair for more than 60 years.71 Primarily
■■ Calcium phosphates applied to the treatment of nonunited fractures,72 they
■■ Calcium hydroxyapetites have had up to 80% success rates in clinical studies.73,74 In
■■ Calcium sulphates one study that demonstrated success in the treatment of
■■ Calcium phosphate/collagen composites delayed union, however, the effects of the electromagnetic
■■ Allogeneic bone
field on fresh fractures were unclear.75 In a meta-analysis of
■■ Demineralized bone matrix
randomized controlled trials by Mollon et al.,76 2,546 cita-
Tissue repair factors
tions were reviewed, 11 articles met the inclusion criteria:
Fibroblast growth factors57,58,60,82
criteria were the use of a random allocation of treatments;
Platelet-derived growth factors83
inclusion of patients presenting with a long-bone lesion;
Osteoinductive and morphogenetic factors
a treatment arm receiving electromagnetism of any wave-
■■ Bone morphogenetic proteins84,86–88,91,92
■■ Wnt proteins92 form to affect bone-healing; a treatment arm receiving no
active intervention; and report of the effect of electromag-
Systemic biological enhancement
Parathyroid hormone61,62,67,70 netic stimulation on direct bone-healing. Evidence from
Anti-sclerostin antibodies64,96–98 four trials reporting on delayed or nonunited fractures
Anti-Dickkopf-related protein 1 antibodies95 demon­strated an overall nonsignificant pooled relative
risk of 1.76 (95% CI, 0.8–3.8; P = 0.15; I = 60%) in favour of
electromagnetic stimulation. The authors concluded that
that the number of stem cells, the tissue source of these although the pooled analysis did not show a significant
cells and their ability to be recruited are dependent on the effect of electromagnetic stimulation on delayed unions or
extent of injury and the stability of the fracture union. nonunited long-bone fractures, the methodo­logical limita-
tions and heterogeneity of studies creates uncertainty as to
Mechanisms of ontogeny and postnatal healing this conclusion.
Both fracture healing and endochondral bone forma- Low-intensity pulsed ultrasonography has also been
tion are directly regulated by BMPs,55,56 fibroblast growth studied in clinical settings, including in patients with long-
factor 2 (FGF‑2),57,58 hedgehog proteins,59,60 parathyroid bone fractures of the upper and lower extremities,77,78 in
hormone (PTH), PTH-related protein,61,62 transforming patients having osteotomies, and in smokers with fractures
growth factor β (TGF‑β),63 protein wingless morpho­ (smoking is a negative risk factor for fracture healing).
genetic factors, Wnt proteins and Wnt signalling antago­ A meta-analysis of randomized controlled trials identified
nists.64,65 Several of these morphogenetic processes form 13 reports, of which five investigated outcomes relevant
interactive feedback loops, including coregulation of to fracture healing.79 These five studies used conservative
BMPs and Wnt signalling proteins;66,67 of PTH or PTH- nonoperative approaches. Two of the studies were of mod-
related protein67 and FGF‑2;69 and of PTH-related protein erate quality, one of which reported improved functional
and Wnt proteins.67–70 Furthermore, some of these factors recovery after treatment of clavicle fracture, and the other
regulate interaction between different cell and tissue types reported no improvement in functional recovery after
during skeletal healing. Therefore, how therapeutic drugs treatment of stress fractures; the other three trials were
coordinate the temporal and spatial inter­actions of dif- low quality and reported improved functional recovery
ferent tissues of the skeletal organ (vascular, skeletal and after nonoperative treatment of long-bone fractures.
haematopoietic) must be understood before their appli- Quality assessment was based on grades of recommen-
cation in the treatment of delayed healing or n­onunion dation according to Wright et al.80 The investigators con-
of fractures. cluded, and we agree with their assessment, that the effect
of low-intensity pulsed ultrasono­graphy on the healing of
Therapy fractures is moderate to very low in quality and the data
Several strategies have been developed to clinically are conflicting.79
enhance fracture healing. In general, fracture healing can
be enhanced by either biophysical or biological means. Local biological enhancement
With regard to biophysical enhancement, substantial Local strategies for the repair and regeneration of bone
research has been conducted on the use of electro­magnetic include the use of osteogenic materials, including
fields and low-intensity pulsed ultrasono­graphy. In the autologous bone marrow, peptide signalling molecules
case of biological enhancement, strategies can be sub­ (FGF‑2 and platelet-derived growth factors [PDGFs])
divided into local and systemic. Although a multitude of and morphogenetic factors (BMPs and Wnt proteins).
methods, materials and factors have been studied, this Although many other metabolites and proteins have

4  |  ADVANCE ONLINE PUBLICATION www.nature.com/nrrheum


© 2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

been investigated, those discussed in this article are, we The investigators concluded that, in patients requiring
believe, the most extensively studied and have the great- hind­foot or ankle arthrodesis, treatment with PDGF has
est potential to be therapeutically targeted to enhance similar fusion rates, less pain and fewer adverse effects in
skeletal repair. comparison with autografting.

Bone marrow grafting BMPs


The field of biotechnology continues to investigate and BMPs are possibly the most extensively investigated candi­
develop new molecules for skeletal repair, such as the dates for the enhancement of skeletal repair, particu­larly
safe and effective method of percutaneous autologous recombinant human BMP‑2 and BMP‑7. Although these
bone-marrow grafting for the treatment of atrophic therapies were developed for similar purposes, their
tibial diaphysis nonunions. In a prospective case series, testing and regulation by the FDA has been different.
Hernigou et al.81 used this method to treat 60 patients Friedlaender et al.84 selected recombinant human BMP‑7
with uninfected aseptic nonunions. Patients underwent to treat tibial nonunions in a prospective random­ized con-
bone-marrow aspiration from both iliac crests. Samples trolled trial of 124 patients. All nonunions were at least
were concentrated on a cell separator and then injected 9 months old with no improvement in the 3 months prior
into the nonunion, resulting in fracture unions in 53 of to each patient enrolling in the study. 63 patients were
the 60 patients. In vitro cellular analysis of aliquots taken treated with a statically locked intramedullary nail plus
prior to injection showed positive correlations between the BMP‑7 with type I collagen as a carrier. The 61 individuals
number of colony-forming units detected and the volume in the control group were treated with an autologous bone
of mineralized callus at 4 months. The seven patients with graft. At 9 months, 81% and 75% of the BMP‑7-treated
nonunited fractures also had fewer colony-forming units patients and 85% and 85% of the autologous bone-graft-
in their grafts than those patients whose fractures united. treated patients were ‘healed’ (P = 0.524), as judged by lack
These results suggest that autologous bone marrow might of pain at the fracture site and radiographic assessment,
be an effective material for the enhancement of skeletal respectively. Although the investigators concluded that
repair, and that the quality of the harvesting technique and BMP‑7 treatment for tibial nonunion is safe and effective,
cell preparation might affect the efficacy of the graft. the data showed no improvement compared with autolo-
gous bone grafting; therefore, the FDA did not provide
FGF‑2 premarket approval for this ‘device’. Composite recombi-
Kawaguchi et al.82 investigated the use of recombinant nant human BMPs with their delivery vehicles, such as a
human FGF‑2 to enhance tibial-shaft fracture healing. type I collagen carrier, are regulated as a device. Instead, a
70 patients with a transverse or short-oblique fracture of ‘humanitarian device exemption’ was issued,85 allowing
the tibial shaft were randomized to one of three groups and a limited distribution to 4,000 patients per year. As part
were assessed for 24 weeks. Patients in each group were of this agreement, the institutions where the surgeries are
injected, into their fracture, with a placebo (gelatin hydro- performed must have an institutional review board to
gel), or with a low dose (0.8 mg) or high dose (2.4 mg) of monitor use of the device.
FGF‑2 hydrogel. The cumulative percentages of patients A different clinical setting was selected for the study of
with radiographically proven fracture union were higher recombinant human BMP‑2; a prospective randomized
in the FGF‑2-treated groups than in the placebo-treated controlled trial of open tibial-shaft fractures was con-
group, and no differences between the high-dose and low- ducted in which patients (n = 450) received initial irriga-
dose FGF‑2 groups were reported. None of the patients tion and debridement by the surgeon as well as a statically
underwent a secondary intervention, and no differences locked intramedullary nail (standard of care) or standard
in the number or type of adverse events were detected of care plus, at the time of wound closure, either 0.75 mg/kg
between the three groups. or 1.50 mg/kg BMP‑2 embedded in a type I collagen
sponge.86 After 12 months, the risk of secondary inter-
PDGF ventions was reduced by 44% in the group treated with
Another peptide signalling molecule that has been exten- the high dose of BMP‑2 compared with standard of care
sively studied for the enhancement of skeletal repair is alone (P = 0.005). 58% of the BMP‑2 group was ‘healed’,
recombinant human homodimeric PDGF subunit B as determined by radiographic evidence and lack of pain
(PDGF‑BB). DiGiovanni et al.83 enrolled 434 (with 397 at the fracture site, compared with 38% of the group
completing the study) patients in a prospective rand- treated by usual care (P = 0.001). Compared with usual
omized controlled (2:1) noninferiority trial of patients care, those patients treated with the high dose of BMP‑2
requiring hindfoot or ankle arthrodesis. The investiga- had fewer failures of the nail, fewer infections and faster
tors tested the hypothesis that PDGF‑BB, combined with wound-healing. The FDA granted premarket approval for
a β‑tricalcium phosphate matrix (n = 260; 394 joints), recombinant human BMP‑2,85 and this treatment is now
would be safe and effective as an alternative to the current available in several c­ountries for the treatment of fresh,
standard of care, an autologous bone graft from the iliac open tibial fractures.
crest (n = 137; 203 joints). CT showed that bones from Since the FDA-approval of BMP therapy, several
159 patients (262 joints) in the PDGF group and 85 (127 inves­tigations have re-examined their clinical use and
joints) in the autograft group were fused at 6 months. The reported sobering results. In a double-blind, randomized,
PDGF group had less pain and an improved safety profile. controlled phase II–III trial, the efficacy and safety of

NATURE REVIEWS | RHEUMATOLOGY ADVANCE ONLINE PUBLICATION  |  5


© 2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

Cartilage Wnt
PTHrP/PTH
PTH PTH

Committed Proliferative BMP/Wnt Hypertrophic


chondrocyte chondrocyte chondrocyte

BMP Wnt
(canonical) Sclerostin

PTH/Wnt Wnt
Skeletogenic
stem cell
(MSC) DKK1
Sclerostin/DKK1

Osteocyte
Osteoprogenitor Osteoblast Osteoprotegerin
RANKL

BMPRI/II
Monocyte/
PTH1R osteoclast
LRP5/LRP6 precursor Osteoclast
Frizzled proteins Bone

Figure 3 | Crosstalk between Wnt, PTH and BMP signalling in cartilage and bone cell lineages. Wnt ligands mediate
canonical signalling through β‑catenin and BMP signalling can be mediated by SMAD1, SMAD3 and SMAD5. The LRP5 and
LRP6 antagonists sclerostin and DKK1 are a central focus of targeted antibody-based therapeutics. The primary stages in
the lineage progression of cartilage and bone cells as they differentiate from skeletogenic stem cells are depicted. Major
stimulatory and inhibitory effects on the differentiation and proliferation of the two lineages are denoted. Primary effects
of the BMP, PTH and Wnt pathway on osteoclast differentiation are indirectly mediated by differing pathway activities that
regulate paracrine factor expression in osteocytes, which in turn regulate osteoclast differentiation and function.
Abbreviations: BMP, bone morphogenetic protein; BMPR, bone morphogenetic protein receptor; DKK1, Dickkopf-related
protein 1; LRP, LDL receptor-related protein; MSC, mesenchymal stem cell; PTH, parathyroid hormone; PTH1R, parathyroid
hormone 1 receptor; PTHrP, parathyroid-hormone-related protein; RANKL, receptor activator of nuclear factor κB ligand;
SMAD, mothers against decapentaplegic homologue.

recombinant human BMP‑2 for the treatment of closed between treatment with BMP‑2 and an iliac crest bone-
tibial diaphysial fractures was studied in comparison graft. BMP‑2 had similar complication rates to iliac bone
with the standard of care alone.87 Co-primary endpoints grafting, but had higher rates during off-label procedures.
of the study were the time to fracture union and the The risk of cancer, for example, was slightly higher with
time to return to normal function. However, the study the use of BMP‑2 (relative risk 1.98; 95% CI 0.86–4.54).
was termi­nated after 6 months when an interim analysis Both recombinant human BMP‑2 and BMP‑7 are available
of the results from 180 patients revealed no shortening under various regulatory conditions, but the development
in the time to fracture union in the active groups of the of safer and more effective materials is an important goal.
study compared with standard of care. The median time
to pain-free, full-weight bearing was also not substantially Systemic biological enhancement
different between groups. The investi­gators concluded In the future, hopefully, a patient with a bone fracture will
that the times to fracture union and full-weight bearing be treated with an injection or pill that would enhance,
in patients with closed tibial fractures treated with intra­ accelerate or otherwise augment skeletal healing as an
medullary nail fixation were not substantially reduced by adjunct to surgical treatment. Several strategies might
BMP‑2 treatment. In a related study, the healing of open achieve this goal. Candidate therapies include the use of
tibial fractures treated with intramedullary nail fixation PTH or humanized monoclonal anti-sclerostin or anti-
was not substantially acceler­ated by the addition of an Dickkopf-related protein 1 antibodies. The cross-talk of
absorbable type I collagen sponge c­ontaining BMP‑2.88 these therapies with the BMP pathway, their function in
Although the focus of this article is fracture healing, endochondral bone formation and intramembranous
as opposed to spinal arthrodesis, the findings of the Yale bone formation, and their production during coupled
Open Data Access (YODA) project 89 should be men- remodelling are presented in Figure 3.65 In addition, the
tioned because they affect the use of BMP‑2 in the field common observation that patients with a sustained head
of musculoskeletal care. By performing a safety and effec- or spinal cord injury can have enhanced skeletal healing
tiveness study of recombinant human BMP‑2 for spinal suggests that a circulating factor, or perhaps a unique
fusion, a meta-analysis of individual participant data neuro­logical mechanism, could be induced to enhance
showed no evidence of clinically meaningful differences bone repair.

6  |  ADVANCE ONLINE PUBLICATION www.nature.com/nrrheum


© 2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

Parathyroid hormone diverse developmental processes. Activation of Wnt sig-


PTH is a naturally occurring hormone that modulates nalling blocks preadipocyte differentiation and stimulates
mineral homeostasis and has been developed as a drug osteoblastogenesis. Data show that some Wnts, such as
for the treatment of osteoporosis. The enhancement of protein Wnt10b, shift cells towards the osteoblastic lineage
fracture healing using both the active-site portion of the by induction of the transcription factors runt-related tran-
molecule (amino acids 1–34, also known as teriparatide) scription factor 2 (RUNX2), homeobox protein DLX5 and
and the full-length molecule (amino acids 1–84) have been transcription factor Sp7 (also known as osterix), and also
studied. Alkhiary et al.70 investigated standard closed dia- by suppression of adipogenic transcription factors.92
physial femoral fractures in 270 male Sprague Dawley LDL receptor-related proteins are a family of cell-­
rats administered a daily (up to 35 days) subcutaneous surface receptors involved in diverse biological processes,
injection of vehicle or of 5 μg/kg or 30 μg/kg PTH (1–34). inclu­ding lipid metabolism, retinoid uptake and neu-
By 21 days, calluses from the group treated with 30 μg of ronal migration. LDL receptor-related protein 5 (LRP5)
PTH (1–34) had significant (P <0.05) increases in torsional is required for signalling of genes in the Wnt family by
strength, stiffness, bone mineral content, bone mineral acting as a co-receptor. Activation of Wnt signalling in
density and cartilage volume, compared with controls. osteoblasts normally stimulates bone formation, and
No changes in osteoclast density were detected, suggesting antagonism of Wnt signalling by secreted proteins from
that treatment with PTH (1–34) enhanced bone forma- the Dickkopf family prevents formation of the active
tion, but did not induce bone resorption. The investiga- complex of LRP5 and thus modulates bone mass. Loss-
tors concluded that daily systemic administration of PTH of-function mutations in LRP5 also impair activation of
(1–34) produces a sustained anabolic effect throughout Wnt signalling and reduce bone mass; LRP5 Gly171Val
the bone-modelling phase of fracture healing. These data impairs the ability of Dickkopf-related proteins to antag-
support a clinical trial in which postmenopausal women onize the Wnt pathway, and unopposed Wnt signalling
with a distal radius fracture requiring closed reduction and leads to increased bone mass (Figure 3).93 Sclerostin,
immobilization, but not surgery, were randomly assigned the SOST gene product expressed exclusively by osteo-
to 8 weeks of once-daily injections of placebo, or of 20 μg cytes, inhibits LRP5 and thus inhibits the Wnt signalling
or 40 μg PTH (1–34) within 10 days of fracture (n = 34 in pathway.94–96 Indeed, the sclerosing bone dysplasias van
each group).90 The estimated median time from fracture to Buchem disease (also known as hyperostosis corticalis
first radiographic evidence of complete cortical bridging generalisata famili­aris) and sclerosteosis are character-
in three of four cortices was 9.1, 7.4, and 8.8 weeks in the ized by thick skulls, square jaws and finger abnormali-
three groups, respectively (overall P = 0.015). Statistically ties, and are associated with loss-of-function mutations
significant differences between the two doses of PTH in SOST.94,95 Several studies have demonstrated that
(1–34) were not found, but the time to healing was shorter anti-sclerostin antibody treatment enhances metaphysial
in the 20 μg PTH (1–34) group than in the placebo group bone-healing in rats97 and enhances proximal tibial defect
(P = 0.006). The investigators concluded that fracture healing in ovariectomized rats.97 Increased serum levels
repair can be accelerated by 20 μg PTH (1–34), but that of sclerostin during human fracture healing have also
these results warrant further study. been detected.98 Furthermore, a study of postmenopau-
In a prospective randomized controlled study to evalu- sal women with osteoporosis, although not a definitive
ate the effects of PTH (1–84) on pelvic fracture healing clinical trial of fracture treatment, has shown that anti-
and functional outcome in postmenopausal women, sclerostin antibody (romosozumab) therapy can increase
65 patients had radiographic and CT examination of bone mineral density and bone formation.99 These studies
pelvic fractures.91 21 patients were treated with a once- suggest that members of the Wnt signalling pathway, and
daily injection of 100 μg of PTH (1–84), beginning 2 days sclerostin in particular, might be therapeutic targets for
after admission to hospital, and the other 44 patients the enhancement of s­keletal repair.
were injected with saline. All patients were treated with
1,000 mg of calcium and 800 IU of vitamin D. CT was Recovery of biological and physical function
repeated at 4‑week increments until radiographic evi- Skeletal healing is defined functionally by the recovery
dence of cortical bridging at the fracture site was visible. of weight-bearing by fractured bone, and is dependent
The results showed a mean time to fracture healing on structural and material features of the tissue. Because
of 7.8 weeks for the PTH (1–84) group compared with aspects of tissue structure and material properties of
12.6 weeks for the control group (P <0.001). The investi- the tissue are variably affected by multiple biological
gators concluded that, in postmenopausal patients with pro­cesses, and each of these processes will be affected
osteoporosis, the PTH (1–84) fragment accelerates pelvic uniquely by a given therapy, each therapy will facilitate
fracture healing, and improves the functional outcome this recovery of function through differing processes.
assessed with a visual analogue scale for pain and a ‘timed Therapeutics for fracture healing should therefore be
up and go’ test.91 developed and administered according to the biological
processes they modify, how these actions affect tissue
The Wnt family material composition and structure, and the relation-
The Wnt family of signalling molecules is only now begin- ships between composition and structure to the recovery
ning to be the focus of studies to enhance skeletal healing. of mechanical function. The use of specific therapeutics
Wnt proteins are a family of secreted proteins that regulate in a situation in which healing has been compromised

NATURE REVIEWS | RHEUMATOLOGY ADVANCE ONLINE PUBLICATION  |  7


© 2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

Table 1 | Timing and effects of therapies for fracture healing


Therapy Optimal Biological effects Structural effects Mechanical Accelerated
therapeutic effects fracture
window union?
PTH Throughout all Chondrocyte and osteoblast proliferation Increased callus Increased Yes
stages Delayed chondrocyte hypertophy size, bone mass stiffness
Increased coupled remodelling and mineral content and strength
Anti- Late endochondral Osteprogenitor expansion Increased callus Increased Yes
sclerostin or phase Chondrocyte hypertrophy size, bone mass stiffness
anti-DKK1 Throughout bone Delayed cartilage resorption and mineral content and strength
antibodies remodelling Decreased coupled remodelling
BMP‑2 Inflammatory Stem cell commitment of chondrogenic Increased callus Increased No
period and osteogenic lineages size stiffness
Chondrocyte hypertophy and coupled
remodelling
Abbreviations: BMP-2, bone morphogenetic protein 2; DKK1, Dickkopf-related protein 1; PTH, parathyroid hormone.

should also be considered in the context of how comorbid- marrow space.101 Consistent with these results are data
ities can affect different biological processes and impede showing that BMPs selectively target periosteal stem-cell
healing. Importantly, some therapeutics can affect fracture differentiation.42 Also of interest is that inhibition of BMP
healing by their actions on multiple biological processes. signalling, taking place constitutively with the homeostatic
Therefore, when considering the use of a therapeutic one maintenance of bone, leads to increased bone mass; this
should consider which biological process would be com- effect is mediated by the loss of BMP regulation of expres-
promised by a given comorbidity and which therapeutic sion of SOST and Dickkopf-related proteins by osteogenic
might best modify the compromised state. Additionally, cells within the medullary space.66 Such divergent biologi-
timing the use of a therapeutic to have its maximal effect cal effects of both PTH and BMPs suggest, therefore, that
on a specific biological process is important to improve careful consideration must be given to both the timing
the progression of healing. Two examples are presented to and duration of their clinical use and to the therapeutic
demonstrate these relationships and Table 1 summarizes applications for which they are most efficacious.
how various therapeutics facilitate the recovery of f­unction
in comparison with their biological effects. Conclusions
In the first example, PTH improved healing in a mouse Optimizing conditions for the harvest, selection, expan-
femoral allograft model by promoting external callus for- sion and formulation of osteogenic stem cell preparations
mation and cartilage development, thereby facilitating is needed to advance the field of skeletal healing and to
the bridging of the graft with the surrounding bone.100 set the stage for developing new local and systemic thera-
Furthermore, PTH can promote intramedullary second- pies. We also need to develop better delivery systems for
ary remodelling of the graft, enabling new bone forma- stem cells, growth factors and osteoinductive substances,
tion to replace the graft.62 Consistent with these studies and to explore systemic applications of osteogenic agents.
are prior findings that daily systemic administration of Identification of appropriate experimental settings and
PTH (1–34) enhanced fracture-healing both by promoting measurable, meaningful clinical endpoints for human
early callus formation through endochondral bone forma- clinical trial design are also required. These objectives, if
tion as well as by producing a sustained anabolic effect based on a strong foundation of basic science knowledge
throughout the remodelling period of fracture healing.70 of skeletal healing, will lead to new methods to improve
Therefore, with different biological activities, increased the care of patients with skeletal injuries.
cartilage formation and increased coupled remodelling,
which separately increase both the cross-sectional area
and the amount of mineralized tissue, PTH improves the Review criteria
two separate phases of fracture healing. PubMed, MEDLINE, Google Scholar and the personal
In a different study, of tibial metaphysial fracture heal­ reference lists of the authors were searched for review
ing in rabbits, treatment with both PTH and recombi- and primary research articles published since 1990.
nant human BMP‑7 increased bone volume at the site of The primary search terms were “fracture healing” and
injury, but neither treatment alone improved mechanical “fracture repair”. Search terms, used singly and in
combination, included “fracture healing” and “fracture
function.101 Combination treatment improved mechani-
repair”, “inflammation”, “immune function”, “T‑cells”,
cal function, the quantity of bone tissue in the defect and “macrophages”, “inflammatory cytokines”, “vascular
integration between new and old bone tissues within the function”, “angiogenesis”, “fixation”, “systemic
injury site and the surrounding tissues. Interestingly, histo­ therapies”, “pharmacological therapies”, “BMP”, “Wnt”,
logical examination showed that recombinant human “FGF”, “TGF”, “PTH”, “LIPUS”, “electrical stimulation”,
BMP‑7 seemed to be strictly anabolic and only facilitated “cell based therapies”, “anabolic therapies”, “nonunion”
cortical bone repair, whereas PTH functioned in the and “clinical trial”. Additional references were identified
from individual articles.
context of coupled remodelling within the underlying

8  |  ADVANCE ONLINE PUBLICATION www.nature.com/nrrheum


© 2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

1. Bolander, M. E. Regulation of fracture repair by cells and immune tolerance. Leuk. Lymphoma 40. Born, W. K., Reardon, C. L. & O’Brien, R. L.
growth factors. Proc. Soc. Exp. Biol. Med. 200, 48, 1283–1289 (2007). The function of γδ T cells in innate immunity.
165–170 (1992). 22. Rizzo, R. et al. A functional role for soluble Curr. Opin. Immunol. 18, 31–38 (2006).
2. Einhorn, T. A. The cell and molecular biology HLA‑G. antigens in immune modulation 41. Colburn, N. T., Zaal, K. J., Wang, F. & Tuan, R.
of fracture healing. Clin. Orthop. Relat. Res. mediated by mesenchymal stromal cells. A role for gamma delta T‑cells in a mouse model
355 (Suppl.), S7–S21 (1998). Cytotherapy 10, 364–375 (2008). of fracture healing. Arthritis Rheum. 60,
3. Ferguson, C., Alpern, E., Miclau, T. & Helms, J. A. 23. Morandi, F. et al. Immunogenicity of human 1694–1703 (2009).
Does adult fracture repair recapitulate mesenchymal stem cells in HLA-class I‑restricted 42. Melief, S. M. et al. Multipotent stromal cells
embryonic skeletal formation? Mech. Dev. 87, T‑cell responses against viral or tumor-associated induce human regulatory T cells through a novel
57–66 (1999). antigens. Stem Cells 26, 1275–1287 (2008). pathway involving skewing of monocytes toward
4. Gerstenfeld, L. C., Cullinane, D. M., Barnes, G. L., 24. Montespan, F., Deschaseaux, F., Sensébé, L., anti-inflammatory macrophages. Stem Cells 31,
Graves, D. T. & Einhorn, T. A. Fracture healing as Carosella, E. D., Rouas-Freiss, N. 1980–1991 (2013).
a post-natal developmental process: molecular, Osteodifferentiated mesenchymal stem cells from 43. Yan, Z., Zhuansun, Y., Chen, R., Li, J. & Ran, P.
spatial, and temporal aspects of its regulation. bone marrow and adipose tissue express HLA‑G. Immunomodulation of mesenchymal stromal
J. Cell Biochem. 88, 873–884 (2003). and display immunomodulatory properties in HLA- cells on regulatory T cells and its possible
5. Vortkamp, A. et al. Recapitulation of signals mismatched settings: implications in bone repair mechanism. Exp. Cell Res. 15, 65–74 (2014).
regulating embryonic bone formation during therapy. J. Immunol. Res. 2014, 23034 (2014). 44. Utvåg, S. E., Grundnes, O., Rindal, D. B. &
postnatal growth and in fracture repair. 25. Najar, M. et al. Immune-related antigens, surface Reikerås, O. Influence of extensive muscle injury
Mech. Dev. 71, 65–76 (1998). molecules and regulatory factors in human- on fracture healing in rat tibia. J. Orthop. Trauma
6. Bais, M. et al. Transcriptional analysis of fracture derived mesenchymal stromal cells: the 17, 430–435 (2003).
healing and the induction of embryonic stem expression and impact of inflammatory priming. 45. Claes, L. et al. The effect of both a thoracic
cell-related genes. PLoS ONE 4, e5393 (2009). Stem Cell Rev. 8, 1188–1198 (2012). trauma and a soft-tissue trauma on fracture
7. Phillips, A. M. Overview of the fracture healing 26. Uccelli, A., Pistoia, V. & Moretta, L. healing in a rat model. Acta Orthop. 82,
cascade. Injury 36 (Suppl. 3), 55–57 (2005). Mesenchymal stem cells: a new strategy for 223–227 (2011).
8. Buckwalter, J. A., Einhorn, T. A., Bolander, M. E. & immunosuppression? Trends Immunol. 28, 46. Gardner, T. N., Stoll, T., Marks, L., Mishra, S. &
Cruess, R. L. in Rockwood and Green’s Fractures in 219–226 (2007). Knothe Tate, M. The influence of mechanical
Adults (Bucholz, R. W. & Heckman, J. D.) 27. Yang, X. et al. Callus mineralization and stimulus on the pattern of tissue differentiation
245–271 (Lippincott, Williams and Wilkins, 2001). maturation are delayed during fracture healing in in a long bone fracture—an FEM study.
9. Hausman, M. R., Schaffler, M. B. & interleukin‑6 knockout mice. Bone 41, 928–936 J. Biomech. 33, 415–425 (2000).
Majeska, R. J. Prevention of fracture healing in (2007). 47. Claes, L. E. & Heigele, C. A. Magnitudes of local
rats by an inhibitor of angiogenesis. Bone 29, 28. Wallace, A., Cooney, T. E., Englund, R. & stress and strain along bony surfaces predict
560–564 (2001). Lubahn, J. D. Effects of interleukin‑6 ablation the course and type of fracture healing.
10. Kurdy, N. M., Weiss, J. B. & Bate, A. Endothelial on fracture healing in mice. J. Orthop. Res. 29, J. Biomech. 32, 255–266 (1999).
stimulating angiogenic factor in early fracture 1437–1442 (2011). 48. Morgan, E. F. et al. Correlations between local
healing. Injury 27, 143–145 (1996). 29. Nam, D. et al. T‑lymphocytes enable osteoblast strains and tissue phenotypes in an
11. Young, L. F., Choi, Y. W., Behrens, F. F., maturation via IL‑17F during the early phase of experimental model of skeletal healing.
DeFouw, D. O. & Einhorn, T. A. Programmed fracture repair. PLoS ONE 7, e40044 (2012). J. Biomech. 43, 2418–2424 (2010).
removal of chondrocytes during endochondral 30. Glass, G. E. et al. TNF-α promotes fracture repair 49. Colnot, C. Skeletal cell fate decisions within
fracture healing. J. Orthop. Res. 6, 144–149 by augmenting the recruitment and differentiation periosteum and bone marrow during bone
(1998). of muscle-derived stromal cells. Proc. Natl Acad. regeneration. J. Bone Miner. Res. 24, 274–282
12. Gerstenfeld, L. C. et al. Impaired fracture healing Sci. USA 108, 1585–1590 (2011). (2009).
in the absence of TNF-α signaling: the role of 31. Motsitsi, N. S. Management of infected 50. Yu, Y. Y., Lieu, S., Lu, C. & Colnot, C. Bone
TNF-α in endochondral cartilage resorption. nonunion of long bones: the last decade morphogenetic protein 2 stimulates
J. Bone Miner. Res. 18, 1584–1592 (2003). (1996–2006). Injury 39, 55–60 (2007). endochondral ossification by regulating
13. Melnyk, M., Henke, T., Claes, L. & Augat, P. 32. Alblowi, J. et al. High levels of tumor necrosis periosteal cell fate during bone repair. Bone
Revascularisation during fracture healing with factor-α contribute to accelerated loss of 47, 65–73 (2010).
soft tissue injury. Arch. Orthop. Trauma Surg. cartilage in diabetic fracture healing. Am. J. 51. Liu, R. et al. Myogenic progenitors contribute
128, 1159–1165 (2008). Pathol. 175, 1574–1585 (2009). to open but not closed fracture repair.
14. Holstein, J. H. et al. Endostatin inhibits callus 33. Kayal, J. et al. TNF-α mediates diabetes BMC Musculoskelet. Disord. 12, 288 (2011).
remodeling during fracture healing in mice. enhanced chondrocyte apoptosis during fracture 52. Olerud, S. & Strömberg, L. Intramedullary
J. Orthop. Res. 31, 1579–1584 (2013). healing and stimulates chondrocyte apoptosis reaming and nailing: its early effects on cortical
15. Kon, T. et al. Expression of osteoprotegerin, through FOXO‑1. J. Bone Miner. Res. 25, bone vascularization. Orthopedics 9,
receptor activator of NF‑κB ligand (osteoprotegerin 1604–1615 (2010). 1204–1208 (1986).
ligand) and related proinflammatory cytokines 34. Claes, L., Recknagel, S. & Ignatius, A. Fracture 53. McCarthy, I. D. & Hughes, S. P. The vascular
during fracture healing. J. Bone Miner. Res. 16, healing under healthy and inflammatory response to fracture micromovement.
1004–1014 (2001). conditions. Nat. Rev. Rheumatol. 8, 133–143 Clin. Orthop. Relat. Res. 301, 281–290 (1994).
16. Ozaki, A., Tsunoda, M., Kinoshita, S. & Saura, R. (2012). 54. Claes, L., Eckert-Hübner, K. & Augat, P. The effect
Role of fracture hematoma and periosteum 35. Santavirta, S. et al. Immunologic studies of of mechanical stability on local vascularization
during fracture healing in rats: interaction of nonunited fractures. Acta Orthop. Scand. 63, and tissue differentiation in callus healing.
fracture hematoma and the periosteum in the 579–586 (1992). J. Orthop. Res. 20, 1099–1105 (2002).
initial step of the healing process. J. Orthop. Sci. 36. Askalonov, A. A. Changes in some indices of 55. Axelrad, T. W. & Einhorn, T. A. Bone
5, 64–70 (2005). cellular immunity in patients with uncomplicated morphogenetic proteins in orthopaedic surgery.
17. Timlin, M. D. et al. Fracture hematoma is a and complicated healing of bone fractures. Cytokine Growth Factor Rev. 20, 481–488
potent proinflammatory mediator of neutrophil J. Hyg. Epidemiol. Microbiol. Immunol. 25, (2009).
function. J. Trauma 58, 1223–1229 (2005). 307–310 (1991). 56. Khosla, S., Westendorf, J. J. & Oursler, M. J.
18. Meert, K. L., Ofenstein, J. P. & Sarnaik, A. P. 37. Askalonov, A. A., Gordienko, S. M., Building bone to reverse osteoporosis and repair
Altered T cell cytokine production following Avdyunicheva, O. E., Bondarenko, A. V. & fractures. J. Clin. Invest. 118, 421–428 (2008).
mechanical trauma. Ann. Clin. Lab. Sci. 28, Voronkov, S. F. The role of T system immunity in 57. Du, X., Xie, Y., Xian, C. J. & Chen, L. Role of
283–288 (1998). reparatory regeneration of the bone tissue in FGFs/FGFRs in skeletal development and bone
19. Al-Sebaei, M. O. et al. Role of Fas and TREG cells animals. J. Hyg. Epidemiol. Microbiol. Immunol. regeneration. J. Cell Physiol. 227, 3731–3734
in fracture healing as characterized in the Fas- 31, 219–224 (1987). (2012).
deficient (lpr) mouse model of lupus. J. Bone 38. Hauser, C. J. et al. The immune 58. Fei, Y., Gronowicz, G. & Hurley, M. M. Fibroblast
Miner. Res. 29, 1478–1491 (2014). microenvironment of human fracture/soft-tissue growth factor‑2, bone homeostasis and fracture
20. Nauta, A. J. & Fibbe, W. E. Immunomodulatory hematomas and its relationship to systemic repair. Curr. Pharm. Des. 19, 3354–3363
properties of mesenchymal stromal cells. Blood immunity. J. Trauma 42, 895–903 (1997). (2013).
110, 3499–3506 (2007). 39. Toben, D. et al. Fracture healing is accelerated in 59. Edwards, P. C. et al. Sonic hedgehog gene-
21. Noël, D., Djouad, F., Bouffi, C., Mrugala, D. & the absence of the adaptive immune system. enhanced tissue engineering for bone
Jorgensen, C. Multipotent mesenchymal stromal J. Bone Miner. Res. 26, 113–124 (2011). regeneration. Gene Ther. 12, 75–86 (2005).

NATURE REVIEWS | RHEUMATOLOGY ADVANCE ONLINE PUBLICATION  |  9


© 2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

60. Song, K., Rao, N. J., Chen, M. L., Huang, Z. J. fracture-healing: a meta-analysis of randomized 89. Simmonds, M. C. et al. Safety and effectiveness
& Cao, Y. G. Enhanced bone regeneration with controlled trials. J. Bone Joint Surg. Am. 90, of recombinant human bone morphogenetic
sequential delivery of basic fibroblast growth 2322–2330 (2008). protein‑2 for spinal fusion: a meta-analysis of
factor and sonic hedgehog. Injury 42, 796–802 77. Heckman, J., Ryaby, J. McCabe, J., Frey, J. J. individual-participant data. Ann. Intern. Med. 158,
(2011). & Kilcoyne, R. F. Acceleration of tibial fracture- 877–879 (2013).
61. Barnes, G. L. et al. Stimulation of fracture- healing by non-invasive, low-intensity pulsed 90. Aspenberg, P. et al. Teriparatide for acceleration
healing with systemic intermittent parathyroid ultrasound. J. Bone Joint Surg. Am. 76, 26–34 of fracture repair in humans: a prospective,
hormone treatment. J. Bone Joint Surg. Am. (1994). randomized, double-blind study of 102
90 (Suppl. 1), 120–127 (2008). 78. Kristiansen, T. K. The effect of low power postmenopausal women with distal radial
62. Jørgensen, N. R. & Schwarz, P. Effects of anti- specifically programmed ultrasound on the fractures. J. Bone Miner. Res. 24, 404–414
osteoporosis medications on fracture healing. healing time of fresh fractures using a Colles’ (2010).
Curr. Osteoporos. Rep. 9, 149–145 (2011). model. J. Orthop. Trauma 4, 227–228 (1990). 91. Peichl, P., Holzer, L. A., Maier, R. & Holzer, G.
63. Patil, A. S., Sable, R. B. & Kothari, R. M. An 79. Busse, J. W. et al. Low intensity pulsed Parathyroid hormone 1–84 accelerates fracture-
update on transforming growth factor‑β (TGF‑β): ultrasonography for fractures: systematic review healing in pubic bones of elderly osteoporotic
sources, types, functions and clinical of randomized controlled trials. BMJ 338, women. J. Bone Joint Surg. Am. 93, 1583–1587
applicability for cartilage/bone healing. J. Cell. 351–360 (2009). (2011).
Physiol. 226, 3094–3103 (2011). 80. Wright, J. G., Einhorn, T. A. & Heckman, J. D. 92. Canalis, E., Giustina, A. & Bilezikian, J. P.
64. Virk, M. S. et al. Systemic administration of Grades of recommendation. J. Bone Joint Mechanisms of anabolic therapies for
sclerostin antibody enhances bone repair in Surg. Am. 87, 1909–1910, (2005). osteoporosis. N. Engl. J. Med. 357, 905–916
a critical-sized femoral defect in a rat model. 81. Hernigou, P. Poignard, A., Beaujean, F. & (2007).
J. Bone Joint Surg. Am. 95, 694–701 (2013). Rouard, H. Percutaneous autologous bone- 93. Boyden, L. M. et al. High bone density due to a
65. Ke, H. Z., Richards, W. G. Li, X. & Ominsky, M. S. marrow grafting for nonunions. Influence of the mutation in LDL‑receptor‑related protein 5.
Sclerostin and Dickkopf‑1 as therapeutic targets number and concentration of progenitor cells. N. Engl. J. Med. 346, 1513–1521 (2002).
in bone diseases. Endocr. Rev. 33, 747–783 J. Bone Joint Surg. Am. 87, 1430–1437 (2005). 94. Staehling-Hampton, K. et al. A 52‑kb deletion
(2012). 82. Kawaguchi, H. et al. A local application of in the SOST-MEOX1 intergenic region on
66. Kamiya, N. The role of BMPs in bone anabolism recombinant human fibroblast growth factor 2 17q12‑q21 is associated with van Buchem
and their potential targets SOST and DKK1. for tibial shaft fractures: a randomized, placebo- disease in the Dutch population. Am. J. Med.
Curr. Mol. Pharmacol. 5, 153–163 (2012). controlled trial. J. Bone Miner. Res. 12, Genet. 110, 144–152 (2002).
67. Kakar, S. et al. Enhanced chondrogenesis and 2735–2743 (2010). 95. Hamersma, H., Gardner, J. & Beighton, P. The
Wnt signaling in PTH treated fractures. J. Bone 83. DiGiovanni, C. W. et al. Recombinant human natural history of sclerosteosis. Clin. Genet. 63,
Miner. Res. 22, 1903–1912 (2007). platelet-derived growth factor-BB and β‑tricalcium 192–197 (2003).
68. Baron, R. & Kneissel, M. Wnt signaling in bone phosphate (rhPDGF‑BB/β‑TCP): an alternative 96. Agholme, F., Li, X. Isaksson, H., Ke, H. Z. &
homeostasis and disease: from human to autogenous bone graft. North American Aspenberg, P. Sclerostin antibody treatment
mutations to treatments. Nat. Med. 19, 179–192 Ortrhopedic Foot and Ankle Study Group. J. Bone enhances metaphyseal bone healing in rats.
(2013). Joint Surg. Am. 95, 1184–1192 (2013). J. Bone Miner. Res. 24, 2412–2418 (2012).
69. Fei, Y. & Hurley, M. M. Role of fibroblast growth 84. Friedlaender, G. E. et al. Osteogenic protein‑1 97. McDonald, M. M. et al. Inhibition of sclerostin by
factor 2 and Wnt signaling in anabolic effects of (bone morphogenetic protein‑7) in the systemic treatment with sclerostin antibody
parathyroid hormone on bone formation. J. Cell. treatement of tibial nonunions. J. Bone Joint enhances healing of proximal tibial defects in
Physiol. 227, 3539–3545 (2012). Surg. Am. 83 (Suppl. 1), S151–S158 (2001). ovariectromized rats. J. Orthop. Res. 30,
70. Alkhiary, Y. M. et al. Enhancement of 85. Blue Cross & Blue Shield of Mississippi. 1541–1548 (2012).
experimental fracture-healing by systemic Bone morphogenetic protein [online], http:// 98. Sarahrudi, K., Thomas, A., Albrecht, C. &
administration of recombinant human www.bcbsms.com/com/bcbsms/apps/ Aharinejad, S. Strongly enhanced levels of
parathyroid hormone (PTH 1–34). J. Bone Joint policysearch/views/viewpolicy.php?&noprint= sclerostin during human fracture healing.
Surg. Am. 87, 731–741 (2005). yes&path=%2Fpolicy%2Femed%2Fbone_ J. Orthop. Res. 30, 1549–1155 (2012).
71. Yasuda, I. Fundamental aspects of fracture morphogenetic_protein.html (2014). 99. McClung, M. R. Romosozumab in
treatment. J. Kyoto Med. Soc. 4, 395–406 86. Govender, S. et al. Recombinant human bone postmenopausal women with low bone mineral
(1953). morphogenetic protein‑2 for treatment of open density. N. Engl. J. Med. 370, 412–420 (2014).
72. Brighton, C. T. et al. A multicenter study of the tibial fractures: a prospective, controlled, 100. Takahata, M., Schwarz, E. M., Chen, T.,
treatment of non-union with constant direct randomized study of four hundred and fifty O’Keefe, R. J. & Awad, H. A. Delayed short-course
current. J. Bone Joint Surg. Am. 63, 2–13 (1981). patients. BMP‑2 Evaluation in Surgery for Tibial treatment with teriparatide (PTH(1–34)) improves
73. Brighton, C. T. Current concepts review. The Trauma (BESTT) study group. J. Bone Joint femoral allograft healing by enhancing
treatment of non-unions with electricity. J. Bone Surg. Am. 84, 2123–2134 (2002). intramembranous bone formation at the graft-host
Joint Surg. Am. 63, 847–851 (1981). 87. Lyon, T. et al. Efficacy and safety of recombinant junction. J. Bone Miner. Res. 27, 26–37 (2012).
74. Scott, G. & King, J. B. A prospective, double-blind human bone morphogenetic protein‑2/calcium 101. Morgan, E. F. et al. Combined effects of
trial of electrical capacitive coupling in the phosphate matrix for closed tibial diaphyseal recombinant human BMP‑7 (rhBMP‑7) and
treatment of non-union of long bones. J. Bone fracture: a double-blind, randomized, controlled parathyroid hormone (1–34) in metaphyseal
Joint Surg. Am. 76, 820–826 (1994). phase I–II/III trial. J. Bone Joint Surg. Am. 95, bone healing. Bone 43, 1031–1038 (2008).
75. Sharrard, W. J. A double-blind trial of pulsed 2088–2096 (2013).
electromagnetic fields for delayed union of tibial 88. Aro, H. T. et al. Recombinant human bone Author contributions
fractures. J. Bone Joint Surg. Am. 72, 347–355 morphogenetic protein‑2: a randomized trial in T.A.E. and L.C.G. contributed equally to researching
(1990). open tibial fractures treated with reamed nail data for the article, substantially contributing
76. Mollon, B., da Silva, V., Busse, J. W., Einhorn, T. A. fixation. J. Bone Joint Surg. Am. 93, 801–808 to discussion of the content, writing the article and to
& Bhandari, M. Electrical stimulation for long-bone (2011). review/editing of the manuscript before submission.

10  |  ADVANCE ONLINE PUBLICATION www.nature.com/nrrheum


View publication stats © 2014 Macmillan Publishers Limited. All rights reserved

You might also like