Sei sulla pagina 1di 9

Review

Endocrine & Metabolic

Enhancement of fracture healing


with parathyroid hormone:
1. Introduction
preclinical studies and potential
2. Evaluation of research so far clinical applications
3. Expert opinion
Byron Chalidis, Christopher Tzioupis, Eleftherios Tsiridis &
Peter V Giannoudis†
†University
Expert Opin. Investig. Drugs Downloaded from informahealthcare.com by York University Libraries on 11/14/14

of Leeds, Academic Unit, Clarendon Wing, Leeds Teaching Hospitals NHS Trust,
Great George Street, Leeds, LS1 3EX, UK

Parathyroid hormone (PTH) has become the most widely studied hormone
with regard to its administration to various species and their respective skele-
tal responses. Beyond its affirmative effect in osteoporosis treatment, sys-
temic PTH administration seems to stimulate bone formation in the fracture
healing process. According to preclinical experimental studies, once-daily
administration of PTH enhances the morphometric and mechanical pro-
perties of fracture calluses and accelerates the normal fracture healing. Its
anabolic effect is obvious even in low doses, as well as in cases of implant fix-
ation and distraction osteogenesis. There is little evidence of toxic effects and
For personal use only.

there are only a few reports of adverse events related to its use. The inci-
dence of bone neoplasms in animal studies depends on the dose and dura-
tion of treatment. However, it is not prognostic of an equivalent risk
potential of carcinogenesis in humans. In summary, the clinical promise of
parathyroid hormone is very high and a positive effect in fracture healing
should be anticipated.

Keywords: fracture callus, IGF-I, mesenchymal cells, osteoblast, osteosarcoma, teriparatide

Expert Opin. Investig. Drugs (2007) 16(4):441-449

1. Introduction

Parathyroid hormone (PTH), an 84 amino acid polypeptide, is secreted by the para-


thyroid glands and it is a major regulator of Ca2+ and phosphate homeostasis [1]. The
main function of PTH is to maintain the Ca2+ ion concentration of the extracellular
fluids within physiologic limits [1]. Although high, sustained levels of PTH are cata-
bolic to bone tissue, intermittent low-dose administration is potentially anabolic
and leads to increased osteoblastic activity and recovery of bone mass [2-4].
Recent animal experimental studies have demonstrated that intermittent treat-
ment with recombinant human PTH (hPTH [1-34] and hPTH (1-84)) promotes
osteogenesis in fracture healing and enhances the size and the mechanical properties
of calluses [5-19].
At present, no published clinical trials have investigated the effects of PTH
treatment on human fractures. Based on the anabolic effect of PTH on osteoporotic
bone [2,3] and the encouraging preclinical results in fracture healing [5-20], it is
subsequently hypothesized that PTH may be successfully used in selected human
fracture cases.
This review primarily aims to summarize the present preclinical evidence on
the effects of PTH in fracture healing, its safety and the potential efficacy in
clinical practice.

10.1517/13543784.16.4.441 © 2007 Informa UK Ltd ISSN 1354-3784 441


Enhancement of fracture healing with parathyroid hormone: preclinical studies and potential clinical applications

2. Evaluation of research so far Local delivery of PTH at the fracture site using the
gene-activated matrix technology has been shown to promote
2.1 Mechanism of action bone regeneration [13,20]. Chen et al. [13] compared the results
The cellular mechanism that is responsible for the anabolic of local, systemic and combined (both systemic and local)
impact of intermittent PTH treatment on fracture healing is application of PTH (1-34) in a rat osteotomized model. The
not fully known. The mitogenic effect of PTH may be medi- combination treatment group, when compared with the other
ated by regulators of bone remodeling such as TGF-β [21], groups, showed significantly higher bone mineral density,
which is a known potent mitogen for osteoprogenitor cells BMC and a trend for greater bone area in microradiographic
and it is regulated partly through PTH [21-23]. PTH may also analysis. The authors concluded that there is an additive effect
enhance the synthesis of IGF-I and the secretion of of systemic intermittent PTH subcutaneous injection and
IGF-I-binding proteins in osteoblast-like cells [24,25]. This local PTH application in promoting bone regeneration in rat
hypothesis was also suggested by an experiment from femurs and hypothesized that the above combination may be
Expert Opin. Investig. Drugs Downloaded from informahealthcare.com by York University Libraries on 11/14/14

Watson et al. [26] in PTH-treated rats. In situ hybridization considered a powerful adjunct in treating local bony defects
techniques have demonstrated that the anabolic effect of PTH such as non-union fractures.
is associated with increased IGF-I gene expression in Regarding the safety and tolerability of various PTH doses,
trabecular osteoblasts [23]. Alkhiary et al. [15] recorded the skeletal effect of daily sub-
β-Catenin is thought to be a further important factor in cutaneous injection of PTH (1-34) 5 or 30 µg/kg of in
osteogenic activity of PTH [27]. It collaborates with endothelial Sprague-Dawley rats. A significant increase was found over
(E)-cadherin at the bone cell surface to keep the cell attached the vehicle group with respect to torsional strength, stiffness,
to its neighbours as it can also modulate gene expression when BMC, bone mineral density and cartilage volume. The
inserting into the nucleus. PTH operates via the β-cat- authors stated that even low doses of PTH have an anabolic
enin-sparing mechanism that is driven by the paracrine Wnt effect throughout the remodeling phase of fracture healing
glycoprotein that promotes osteogenesis and bone growth [28]. process without inducing bone resorption at low doses.
In addition, Nakazawa et al. [5] have shown that PTH has The duration of intermittent treatment of PTH on fracture
For personal use only.

an additional key effect on mesenchymal (chondroprogenitor) healing has been investigated by Komatsubara et al. [6] in a rat
cells by stimulating their proliferation and increasing their model experimental study. Subcutaneous injection of
recruitment into the chondrocyte lineage. As a result, a higher hPTH (1-34) 10 or 30 µg/kg/day improved the mechanical
proportion of progenitor cells achieve full osteoblast strength of fractured bone by accelerating the remodeling of
differentiation [29]. woven bone to lamellar bone and the formation of a new cor-
In general terms, it is postulated that inflammatory media- tical shell without increasing callus size. Moreover, it was evi-
tors may stimulate IGF-I expression by periosteal cells follow- dent that 3 weeks pre-treatment with PTH prior to fracture
ing a fracture. In turn, IGF-I stimulates chondrogenesis but alone did not affect fracture healing. However, the healing
PTH enhances ossification in an autocrine (PTH) or para- process was accelerated when PTH was administered 3 weeks
crine (PTH-related protein [PTHrP]) fashion [30]. It is consid- before and 12 weeks after the fracture. According to the data,
ered that IGF-I mediates the effect of PTH on PTH application should be continued when fragility fracture
osteoblast-induced bone formation and mesenchymal cells occurs in osteoporotic patients under PTH treatment.
proliferation [5]. Recently, evidence has been proposed that low-dose
hPTH (1-34) could enhance chondrogenesis and accelerate
2.2 Efficacy in fracture calluses cartilage differentiation. Nakazawa et al. [5] found that inter-
Several preclinical investigations have described the mittent low-dose treatment with recombinant hPTH (1-34)
enhancement of fracture healing and osteointegration in increased the proliferation of chondroprogenitor cells and the
animals after systemic [5-19] and local [13,20] administration synthesis of type II collagen, resulting in the formation of a
of PTH (Table 1). Andreassen et al. [9] studied the results of larger cartilaginous callus. The PTH-induced cartilaginous
intermittent administration of teriparatide (PTH [1-34]) callus was then rapidly replaced with bone. The latter effect
200 µg/kg/day on the density and mechanical strength of was speculated by the authors to be a potential important
callus in a 27-month-old rat tibial fracture model during advantage to support the future clinical application of this
the third and the eighth week of healing process. During peptide in the treatment of skeletal injuries.
weeks 3 – 8, the increase of the bone mineral content
(BMC) of the callus was 169% in the PTH-treated animal 2.3 Efficacy in implant fixation
compared with 60% in the vehicle-injected animals. At Addressing the issue of PTH administration and ortho-
3 weeks, PTH increased maximum load and external callus pedic implant fixation, Skripitz et al. [18,19] compared the
volume by 160 and 208%; at 8 weeks, these were increased effects of PTH in the implant–bone fixation of stainless
by 270 and 135%, respectively. In the contralateral intact steel screws in terms of new bone formation and normal
tibia, PTH treatment increased BMC by 18 and 21% at 3 baseline remodeling in rats. Histologic examination
and 8 weeks, respectively. revealed that PTH increased the density of surrounding

442 Expert Opin. Investig. Drugs (2007) 16(4)


Expert Opin. Investig. Drugs Downloaded from informahealthcare.com by York University Libraries on 11/14/14
For personal use only.

Table 1. Preclinical studies of PTH application in fracture healing process .

Study Year Animal Fracture model PTH dose: treatment duration Major findings
[20] 1996 Sprague-Dawley rats Mid-diaphyseal femoral Local PTH (1-34) and/or BMP-4 delivery Implantation of a gene-activated matrix containing either BMP-4 or
fracture PTH (1-34) resulted in a biologic response of new bone filling the
gap
Implantation of a two-plasmid gene-activated matrix encoding
BMP-4 and PTH (1-34) fragment acted synergistically
[8] 1999 3-month-old male Mid-diaphyseal femoral Daily subcutaneous injection injection of In the PTH-treated fractures, statistically significant increase in callus
Sprague-Dawley rats fracture PTH (1-34) 80 µg/kg versus saline vehicle area and strength
for 21 days Histologic examination of the calluses: increase in the amount of
new bone formed
Statistically significant increase in callus area and strength after
21 days of treatment of PTH-treated group (p < 0.05)
[10] 1999 3-month-old female Wistar Tibial fracture Daily subcutaneous injection of either 60-µg group: significant increase in BMC, external callus volume
rats PTH (1-34) 60 or 200 µg/kg for 20 or and ultimate load after 40 days
40 days versus vehicle control 200-µg group: significant increase in BMC, external callus volume
and ultimate load after 20 and 40 days
Both PTH groups: greater increases in contralateral intact tibia BMC
compared with vehicle after 40 days
[17] 2000 Adult male New Zealand Standard osteotomy in Daily injections of prednisone At 6 weeks, all of the animals treated with both PTHrP and
white rabbits both ulna of each rabbit (0.15 mg/kg) beginning 2 months prior prednisone showed complete union at the osteotomy site bilaterally.
to surgery and continuing until killing In contrast, no control osteotomies achieved union at 6 weeks
Daily injection of PTHrP analog RS-66271 Ulna in the PTHrP analog-treated rabbits showed greater
(0.01 mg/kg s.c.) starting 1 day after radiographic intensity (20 – 40%), larger callus area (209%

Expert Opin. Investig. Drugs (2007) 16(4)


surgery for 6 weeks anteroposterior view; 417% lateral view), greater stiffness (64%)
Control animals received subcutaneous and torque (87%) when compared with controls
normal saline
Study at 6 and 10 weeks
[11] 2000 4-month-old ovariectomized Bilateral tibial shaft Daily subcutaneous injection of either Increase in mechanical and morphometric fracture callus parameters
Sprague-Dawley rats fracture PTH (1-84) 15 or 150 µg/kg for 30 days (dose dependent) in the PTH-treated group
versus 17 β-estradiol No advantages in 17 β-estradiol-treated group
[18] 2000 Adult male Sprague-Dawley Implantation of bone Daily subcutaneous injection of human Cancellous density was increased by 90, 132 and 173% in the
rats conduction chamber at PTH (1-34) 15, 60 or 240 µg/kg for groups given PTH in a dose of 15, 60 or 240 µg/kg/day, respectively
the proximal tibial 6 weeks versus vehicle Linear correlation between bone density and the log PTH doses
metaphysis (r2 = 0.6)
BMP: Bone morphogenetic protein; BMC: Bone mineral content; BMD: Bone mineral density; C10: PTH 10 µg/kg before and after surgery; C30: PTH 30 µg/kg before and after surgery; CNT: Vehicle control; hPTH: Human PTH;
P10: PTH 10 µg/kg before surgery; P30: PTH 30 µg/kg before surgery; PTH: Parathyroid hormone.
Chalidis, Tzioupis, Tsiridis & Giannoudis

443
Expert Opin. Investig. Drugs Downloaded from informahealthcare.com by York University Libraries on 11/14/14
For personal use only.

444
Table 1. Preclinical studies of PTH application in fracture healing process (continued).

Study Year Animal Fracture model PTH dose: treatment duration Major findings
[9] 2001 27-month-old female Wistar Tibial fracture Daily subcutaneous injection of At 3 weeks: increased maximum load, external callus volume,
rats PTH (1-34) 200 µg/kg for 3 or 8 weeks BMC by 160, 208 and 190%, respectively, in the PTH group
versus vehicle At 8 weeks: increased maximum load, external callus volume,
BMC by 270, 135 and 388%, respectively, in the PTH group
Week 3 to week 8: callus BMC increased by 60% in the
vehicle-injected animals and by 169% in the PTH-treated animals
Contralateral intact tibia: PTH treatment increased BMC by 18 and
21% (3 and 8 weeks)
[19] 2001 Sprague-Dawley rats Implantation of one Daily subcutaneous injection of human PTH increased the screw mean removal torque from 1.1 to 3.5 Ncm
screw in either one or in PTH (1-34) 60 µg/kg/day over a period of (p = 0.001) and the mean pull-out strength from 66 to 145 N
both proximal tibia 4 weeks versus vehicle (p = 0.002)
Histologic examination: higher density of trabecular bone around
the implant in the PTH group
[12] 2002 2-month-old male Closed mid-diaphyseal Daily subcutaneous injection of On day 28 after fracture: BMC, BMD and ultimate load to failure of
Sprague-Dawley rats femur fracture PTH (1-34) 10 µg/kg for 4 or 6 weeks the calluses were significantly increased in the PTH-treated group by
versus vehicle 61, 46 and 32%, respectively, compared with the control group
On day 42 after fracture: similar increases of 119, 74 and 55%,
respectively
PTH (1-34) increases production of bone matrix proteins and
enhances osteoclastogenesis during the phase of callus remodeling

Expert Opin. Investig. Drugs (2007) 16(4)


[13] 2003 Adult male rats Bilateral femoral Daily subcutaneous injections of Combination of systemic and local PTH administration leads to
segmental osteotomy 40 µg/kg or local PTH (1-34) delivery or higher bone mineral density and mineral content
combination of the 2 for 6 weeks versus Local PTH gene therapy enhances the anabolic activity of systemic
vehicle PTH administration during fracture healing
[14] 2004 3-month-old male Mid-diaphyseal femoral Every second day: 60 µg/kg of human In 20 days: PTH (1-34) increased ultimate load (56%), stiffness
Sprague-Dawley rats osteotomy and PTH (1-34)/kg versus vehicle until (117%), total regenerate callus volume (58%), callus BMC (24%)
lengthening of the right consolidation occurs (20 or 40 days after and histologic bone density (35%) compared with untreated
femur using an external distraction was completed) distraction osteogenesis specimens
fixator In 40 days: PTH (1-34) increased ultimate load (54%), stiffness
(55%), callus BMC (33%) and bone density (23%) compared with
untreated distraction osteogenesis specimens
BMP: Bone morphogenetic protein; BMC: Bone mineral content; BMD: Bone mineral density; C10: PTH 10 µg/kg before and after surgery; C30: PTH 30 µg/kg before and after surgery; CNT: Vehicle control; hPTH: Human PTH;
P10: PTH 10 µg/kg before surgery; P30: PTH 30 µg/kg before surgery; PTH: Parathyroid hormone.
Enhancement of fracture healing with parathyroid hormone: preclinical studies and potential clinical applications
Expert Opin. Investig. Drugs Downloaded from informahealthcare.com by York University Libraries on 11/14/14
For personal use only.

Table 1. Preclinical studies of PTH application in fracture healing process (continued).

Study Year Animal Fracture model PTH dose: treatment duration Major findings
[7] 2004 3-month-old female Wistar Closed fracture above Daily subcutaneous injection of 60 µg/kg After 8 weeks: increased fracture strength and callus volume in
rats tibiofibular junction in intermittent hPTH or vehicle PTH-treated rats (ultimate load 66%, ultimate stiffness 58%, callus
the right tibia Duration of PTH application is 8 weeks volume 28%)
and rats investigated in 8 or 16 weeks After 16 weeks: no differences in fracture strength, callus volume or
callus tissue mechanical quality between PTH and vehicle
Comparing PTH-treated animals at 8 and 16 weeks: fracture
strength and callus tissue mechanical quality continued to increase
after the withdrawal of PTH (ultimate load 23%, ultimate stress
88%, elastic modulus 87%) and external callus volume declined
during this period (27%)
[6] 2005 Female Sprague-Dawley rats Transverse fracture in Daily subcutaneous injection of PTH for New cortical shell significantly higher in C10 and C30 groups
bilateral femur 3, 6 and 12 weeks in four groups versus compared with the other groups
vehicle Ultimate load in mechanical testing significantly higher in C30
Group I: P10 group than in CNT, P10 and P30 groups
Group II: C10 Intermittent PTH treatment at 30 µg/kg before and after osteotomy
Group III: P30 accelerated the healing process (earlier replacement of woven bone
Group IV: C30 to lamellar bone, increased new cortical shell formation, increased
the ultimate load up to 12 weeks after osteotomy)
[5] 2005 2-month-old male Closed mid-diaphyseal Daily subcutaneous of PTH (1-34) Low-dose treatment with PTH (1-34) increased the recruitment of
Sprague-Dawley rats fracture in the right 10 µg/kg for 2, 4, 7, 14, 21 and 28 days mesenchymal cells into the chondrocyte lineage
femur versus vehicle Proliferation of mesenchymal (chondroprogenitor) cells and

Expert Opin. Investig. Drugs (2007) 16(4)


synthesis of type II collagen
Formation of a larger cartilaginous callus
The PTH-induced cartilaginous callus was then rapidly replaced with
bone
[15] 2005 Male Sprague-Dawley rats Standard, closed femoral Daily subcutaneous injections of PTH Day 21: the 30-mg group had a significant increase over the
fracture (1-34) 5 or 30 µg/kg in 3 groups for controls with respect to torsional strength, stiffness, BMC, BMD and
35 days versus vehicle cartilage volume
Further PTH group division into 3 groups, Day 35: both groups treated with PTH showed significant increase in
killed on days 21, 35 and 84 after the bone BMC, density total osseous tissue volume and significant
fracture decrease in void space and cartilage volume
Day 84: the 30-mg group had significant increase over the controls
with respect to torsional strength and bone mineral density
BMP: Bone morphogenetic protein; BMC: Bone mineral content; BMD: Bone mineral density; C10: PTH 10 µg/kg before and after surgery; C30: PTH 30 µg/kg before and after surgery; CNT: Vehicle control; hPTH: Human PTH;
P10: PTH 10 µg/kg before surgery; P30: PTH 30 µg/kg before surgery; PTH: Parathyroid hormone.
Chalidis, Tzioupis, Tsiridis & Giannoudis

445
Enhancement of fracture healing with parathyroid hormone: preclinical studies and potential clinical applications

bone and mounted the implant–bone contact. The results 2.6 Clinical relevance
of these studies indicate that intermittent PTH treatment There is a very significant and continually growing interest in
may be helpful in the augmentation of early fixation of the therapeutic efficacy and safety of PTH on human bone heal-
orthopedic implants. ing [25,35]. PTH seems to enhance both the mechanical strength
and the bone mass of the calluses, producing a sustained ana-
2.4 Distraction osteogenesis bolic effect throughout the remodeling phase of fracture heal-
The act of PTH (1-34) treatment was also examined in dis- ing. According to the experimental studies, it seems that the
traction osteogenesis [14]. Distraction osteogenesis is used for effect of PTH is strongest on repairing, cancellous, loaded bone
both leg lengthening and bone transportation for the with hematopoietic marrow [49]. As such, conditions are more
treatment of fractures and non-unions. Using this method, abundant in fracture sites (hematoma formation, angiogenesis
the time needed for the new formed bone to consolidate and and cell proliferation) than the rest of the skeleton: one could
become strong enough for weight bearing can be expect PTH to have an even stronger effect in orthopedic and
Expert Opin. Investig. Drugs Downloaded from informahealthcare.com by York University Libraries on 11/14/14

≤ 12 months. trauma surgery than in the treatment of osteoporosis. However,


Seebach et al. [14] performed an experimental study on rats some considerations exist regarding how the potential frac-
investigating whether intermittent PTH (1-34) admin- ture-healing effects of PTH in humans can be examined. As nei-
istration could accelerate the consolidation of new formed ther morphologic appearance nor mineral content correlate with
bone after distraction osteogenesis. The results indicated a sig- mechanical bone strength, it is conspicuous that clinical trials
nificant increase in BMC and histologic bone density of the will be designed with relevant end points.
femoral distraction callus in treated models. The contralateral Skripitz et al. [49] stated that PTH cannot induce bone for-
femur became stiffer and denser as well, but to a lesser degree. mation from uncommitted cells in comparison with other
The authors concluded that PTH (1-34) may be useful as a anabolic agents (such as bone morphogenetic proteins) and,
stimulator of bone formation to improve regenerated bone therefore, it cannot be expected to be useful in non-unions.
stability when consolidation occurs. Conversely, Whitfield [25] noted that clinical application of
such bone-building peptides will speed up the mending of
For personal use only.

2.5 Safety severe non-union fractures and the installation of artificial hip
The published data from the systematic application of PTH and knees, expanding their potential targets far beyond osteo-
in osteoporosis treatment, or from cases with long-standing porosis. Recent evidence that PTH increases recruitment of
hyperparathyroidism [31], are reassuring regarding the fre- mesenchymal cells into the chondrocyte lineage, resulting in
quency and intensity of the side effects of the drug. The formation of a larger cartilaginous callus supports this
described symptoms are usually mild and transient in most of hypothesis and the potential use of the PTH peptide beyond
the cases and termination or discontinuation of treatment is the acute fracture-repairing processes [5,25].
not a common incident (Table 2) [2,3,32-44]. Alternative delivery systems and greater patient tolerance
However, some questions have been raised regarding the will enlarge the efficacy of PTH clinical application. Many
potential PTH carcinogenicity and its effect in inducing efforts have been made during the last years for the devel-
bone proliferative lesions [2,42,45,46]. Vahle et al. [46] noticed opment of non-injectable routes of PTH delivery including
that there is a dose of the peptide that is strongly osteogenic oral (buccal, sublingual and enteric–gastrointestinal),
but not carcinogenic in an oncogenicity long-term study in transdermal, nasal and pulmonary approaches [50]. If equiva-
female Fischer rats. PTH has not been proven to be lent bioavailability and anabolic action are achieved in com-
genotoxic and, therefore, it is virtually certain not to be parison with standard injection methods, patient acceptance
carcinogenic when applied for the very few times needed to and compliance will be increased. In particular, after the
enhance fracture healing [15]. Furthermore, there is enough trauma period, the aversion for self-injection could be over-
ambiguity whether tumor-related studies in rat models could come and the duration of treatment may be extended to offer
be connected to human physiology [47,48] as there are several the optimal result.
differences in the osteon remodeling system between human
and rat skeletons. Nevertheless, the same limitations regard- 3. Expert opinion
ing the appropriate selection of the patients must be followed
in a potential application of PTH in fracture cases. Paget’s PTH constitutes a promising skeletal anabolic agent, which
disease, previous bone radiation, history of cancer, bone has largely changed the mainstay of osteoporosis treatment.
metabolic diseases, use in children, high levels of Ca2+ and Since the FDA approval in 1992, several studies have taken
alkaline phosphatase, nursing and allergic reactions to PTH place to assess the therapeutic effect of PTH in post-
or to its ingredients must be considered as contraindications menopausal women and to compare its anabolic effect
of teriparatide treatment. Patients at high risk should be against or along with previous well-known antiresorptive
excluded from any therapeutic fracture protocol until agents such as bisphosponates and calcitonin. PTH seems
long-term safety data will re-establish the inclusion and not only to be effective in increasing the bone density, but it
exclusion criteria [33,48]. also has the potential to improve the microarchitecture of the

446 Expert Opin. Investig. Drugs (2007) 16(4)


Chalidis, Tzioupis, Tsiridis & Giannoudis

In addition to PTH studies, there has also been a constant


Table 2. Adverse effects of parathyroid hormone
influx of new information from all of the relevant disciplines
injection in humans.
of fracture healing. The contribution of many local and sys-
Adverse effect Studies temic regulatory factors such as hormones, cytokines,
Hypercalcemia [2,33,38,40,41,43,44]
growth factors and stem cells in bone regeneration is known
to be substantial and undisputable. The ability to visualize
Hypercalciuria [2,35,38,40,41]
and manipulate matters at the nanoscale, dealing with struc-
Serum creatinine increase [38] tures and devices with length scales in the range of
Creatinine clearance increase [37] 1 – 100 nm offers a new prospective of delivering bioactive
Headache [2,35] regulators (such as PTH) in a more accurate fashion. Drugs
such as PTH could be encapsulated into nanoparticles and
Dizziness [2]
be delivered to fracture site in a time-dependent fashion.
Expert Opin. Investig. Drugs Downloaded from informahealthcare.com by York University Libraries on 11/14/14

Hyperuricemia [2,35,40]
New bone formation in the fracture site after implantation
Allergic events [34] of a degradable gene-activated collagen matrix sponge
Injection site reaction [34] loaded with plasmid DNA encoding hPTH (1-34) or bone
Nausea [34] morphogenetic protein-4 has already been accomplished on
an experimental basis. Gene therapy is an attractive method
Leg cramps [2,40]
for drug delivery as it could be associated with efficient tar-
geting to cell surface receptors and less protein is necessary
bone. On the other hand, long-term results are still unknown to achieve a clinically useful effect.
and close monitoring of patients for potential late adverse In the next 10 years, more data will be revealed regarding
events is obvious and imperative as the use of PTH is still a the precise cellular and biologic pathways of bone-healing
new protocol. processes and the appropriate manner to intervene with safety
The concept of an anabolic skeletal agent that could in fracture environment. Cross-disciplinary research will eval-
For personal use only.

provoke new bone formation and the promising results of the uate the interaction and sequence of all of the aforementioned
initial studies in the treatment of osteoporosis may extend the anabolic factors and will radically change the prospects for
indications of PTH in trauma cases and especially in bone regenerative medicine.
fractures. In the last 10 years, many experimental trials have So far, no systemic pharmacologic agent has been proven to
been made with the purpose of testifying the role of the have undoubted clear benefits in fracture healing and, if PTH
intermittent PTH in the fracture environment. Accelerating confirmed to be effective in human bone callus formation, it
bone healing, stimulation of osteogenesis (hard callus will offer optimum benefits.
formation) and improvement of the mechanical strength of In clinical practice, PTH could be a feasible therapeutic
callus mass have been documented with histologic and agent in well-selected trauma conditions, such as fragility frac-
histomorphometric analyses in animal models. tures in osteoporotic patients. Acceleration of the healing
Despite the fact that the results were in favor of using the response and solid implant–bone consolidation will minimize
PTH as an anabolic agent in the fracture-healing process in all the risks of re-operation and prolong immobilization and
the animal trials, many questions regarding the safety, the mortality.
appropriate dose, the duration of therapy and the cost-effec-
tiveness are pending. Therefore, enthusiasm for widespread use Disclosure
of PTH in orthopedic trauma cases must be tempered. Clini-
cal prospective blinded and randomized studies in different age This work is attributed to the Academic Unit, Trauma and
and gender groups will allow generation of adequate evidence Orthopaedic Surgery, Clarendon Wing, Leeds General
regarding the efficacy and safety of PTH in fracture repair. Infirmary, Great George Street, Leeds, LS1 3EX, UK.

Expert Opin. Investig. Drugs (2007) 16(4) 447


Enhancement of fracture healing with parathyroid hormone: preclinical studies and potential clinical applications

Bibliography increase. Calcif. Tissue Int. (2004) have a sustained anabolic effect to
Papers of special note have been highlighted as 74(4):351-356. fracture healing.
either of interest (•) or of considerable interest 8. HOLZER G, MAJESKA RJ, 16. KIM HW, JAHNG JS: Effect of
(••) to readers. LUNDY MW, HARTKE JR, intermittent administration of parathyroid
1. JUPPNER H, KRONENBERG H: EINHORN TA: Parathyroid hormone hormone on fracture healing in
Primer on the metabolic bone diseases and enhances fracture healing. A preliminary ovariectomized rats. Iowa Orthop. J. (1999)
disorders of mineral metabolism. In: report. Clin. Orthop. Relat. Res. (1999) 19:71-77.
Parathyroid Hormone. Favus M (Ed.), 366:258-263. 17. BOSTROM MP, GAMRADT SC,
American Society for Bone and Mineral 9. ANDREASSEN TT, FLEDELIUS C, ASNIS P et al.: Parathyroid
Research, Washington DC, USA EJERSTED C, OXLUND H: Increases in hormone-related protein analog RS-66271
(2003):117-124. callus formation and mechanical strength of is an effective therapy for impaired bone
2. NEER RM, ARNAUD CD, healing fractures in old rats treated with healing in rabbits on corticosteroid therapy.
Expert Opin. Investig. Drugs Downloaded from informahealthcare.com by York University Libraries on 11/14/14

ZANCHETTA JR et al.: Effect of parathyroid hormone. Acta Orthop. Scand. Bone (2000) 26(5):437-442.
parathyroid hormone (1-34) on fractures (2001) 72(3):304-307. 18. SKRIPITZ R, ANDREASSEN TT,
and bone mineral density in 10. ANDREASSEN TT, EJERSTED C, ASPENBERG P: Parathyroid
postmenopausal women with osteoporosis. OXLUND H: Intermittent parathyroid hormone (1-34) increases the density of rat
N. Engl. J. Med. (2001) hormone (1-34) treatment increases callus cancellous bone in a bone chamber.
344(19):1434-1441. formation and mechanical strength of A dose–response study. J. Bone Joint
3. DEMPSTER DW, COSMAN F, healing rat fractures. J. Bone Miner. Res. Surg. Br. (2000) 82(1):138-141.
KURLAND ES et al.: Effects of daily (1999) 14(6):960-968. 19. SKRIPITZ R, ASPENBERG P:
treatment with parathyroid hormone on 11. JAHNG JS, KIM HW: Effect of Implant fixation enhanced by intermittent
bone microarchitecture and turnover in intermittent administration of parathyroid treatment with parathyroid hormone.
patients with osteoporosis: a paired biopsy hormone on fracture healing in J. Bone Joint Surg. Br. (2001)
study. J. Bone Miner. Res. (2001) ovariectomized rats. Orthopedics (2000) 83(3):437-440.
16(10):1846-1853. 23(10):1089-1094. • The authors found that PTH application
For personal use only.

4. PIERROZ DD, BOUXSEIN ML, 12. NAKAJIMA A, SHIMOJI N, SHIOMI K enhances the implant–bone fixation of
RIZZOLI R, FERRARI SL: Combined et al.: Mechanisms for the enhancement of stainless steel screws and the incorporation
treatment with a β-blocker and intermittent fracture healing in rats treated with of orthopedic implants.
PTH improves bone mass and intermittent low-dose human parathyroid 20. FANG J, ZHU YY, SMILEY E et al.:
microarchitecture in ovariectomized mice. hormone (1-34). J. Bone Miner. Res. (2002) Stimulation of new bone formation by
Bone (2006) 39(2):260-267. 17(11):2038-2047. direct transfer of osteogenic plasmid genes.
5. NAKAZAWA T, NAKAJIMA A, 13. CHEN H, FRANKENBURG EP, Proc. Natl. Acad. Sci. USA (1996)
SHIOMI K et al.: Effects of low-dose, GOLDSTEIN SA, MCCAULEY LK: 93(12):5753-5758.
intermittent treatment with recombinant Combination of local and systemic 21. CENTRELLA M, MCCARTHY TL,
human parathyroid hormone (1-34) on parathyroid hormone enhances bone CANALIS E: Parathyroid hormone
chondrogenesis in a model of experimental regeneration. Clin. Orthop. Relat. Res. modulates transforming growth factor β
fracture healing. Bone (2005) (2003) 416:291-302. activity and binding in osteoblast-enriched
37(5):711-719. • This study demonstrates that local PTH cell cultures from fetal rat parietal bone.
•• This is the first study that clearly shows gene therapy enhances the anabolic effect Proc. Natl. Acad. Sci. USA (1988)
that PTH stimulates the proliferation of of systemic PTH. 85(16):5889-5893.
chondroprogenitor cells in vivo and 14. SEEBACH C, SKRIPITZ R, 22. CANALIS E, CENTRELLA M,
demonstrates that PTH enhances both ANDREASSEN TT, ASPENBERG P: BURCH W, MCCARTHY TL: Insulin-like
osteogenesis (hard callus formation) and Intermittent parathyroid hormone (1-34) growth factor I mediates selective anabolic
chondrogenesis (soft callus formation). enhances mechanical strength and density effects of parathyroid hormone in bone
6. KOMATSUBARA S, MORI S, of new bone after distraction osteogenesis in cultures. J. Clin. Invest. (1989) 83(1):60-65.
MASHIBA T et al.: Human parathyroid rats. J. Orthop. Res. (2004) 22(3):472-478. 23. MCCARTHY TL, CENTRELLA M,
hormone (1-34) accelerates the fracture • This study shows the anabolic effect of CANALIS E: Parathyroid hormone
healing process of woven to lamellar bone PTH injection in distraction osteogenesis. enhances the transcript and polypeptide
replacement and new cortical shell 15. ALKHIARY YM, GERSTENFELD LC, levels of insulin-like growth factor I in
formation in rat femora. Bone (2005) KRALL E et al.: Enhancement of osteoblast-enriched cultures from fetal rat
36(4):678-687. experimental fracture-healing by systemic bone. Endocrinology (1989)
7. ANDREASSEN TT, WILLICK GE, administration of recombinant human 124(3):1247-1253.
MORLEY P, WHITFIELD JF: Treatment parathyroid hormone (PTH 1-34). J. Bone 24. DEMPSTER DW, COSMAN F,
with parathyroid hormone hPTH(1-34), Joint Surg. (2005) 87(4):731-741. PARISIEN M, SHEN V, LINDSAY R:
hPTH(1-31), and monocyclic hPTH(1-31) •• In this paper, the authors proved that Anabolic actions of parathyroid hormone
enhances fracture strength and callus even low doses of PTH, which may be on bone. Endocr. Rev. (1993)
amount after withdrawal fracture strength more relevant to the clinically available 14(6):690-709.
and callus mechanical quality continue to dose used to treat osteoporosis, seem to

448 Expert Opin. Investig. Drugs (2007) 16(4)


Chalidis, Tzioupis, Tsiridis & Giannoudis

25. WHITFIELD JF: Parathyroid hormone 34. CRANNEY A, PAPAIOANNOU A, 43. MCCLUNG MR, SAN MARTIN J,
and leptin – new peptides, expanding ZYTARUK N et al.: Parathyroid hormone MILLER PD et al.: Opposite bone
clinical prospects. Expert Opin. for the treatment of osteoporosis: remodeling effects of teriparatide and
Investig. Drugs (2005) 14(3):251-264. a systematic review. CMAJ (2006) alendronate in increasing bone mass.
26. WATSON P, LAZOWSKI D, HAN V 175(1):52-59. Arch. Intern. Med. (2005)
et al.: Parathyroid hormone restores bone 35. TASHJIAN AH Jr, GAGEL RF: 165(15):1762-1768.
mass and enhances osteoblast insulin-like Teriparatide [human PTH(1-34)]: 2.5 years 44. COSMAN F, NIEVES J, ZION M et al.:
growth factor I gene expression in of experience on the use and safety of the Daily and cyclic parathyroid hormone in
ovariectomized rats. Bone (1995) drug for the treatment of osteoporosis. women receiving alendronate. N. Engl.
16(3):357-365. J. Bone Miner. Res. (2006) 21(3):354-365. J. Med. (2005) 353(6):566-575.
27. WHITFIELD JF: Osteoporosis-treating 36. HODSMAN AB, HANLEY DA, 45. VAHLE JL, SATO M, LONG GG et al.:
parathyroid hormone peptides: what are ETTINGER MP et al.: Efficacy and safety Skeletal changes in rats given daily
Expert Opin. Investig. Drugs Downloaded from informahealthcare.com by York University Libraries on 11/14/14

they? What do they do? How might they do of human parathyroid hormone-(1-84) in subcutaneous injections of recombinant
it? Curr. Opin. Investig. Drugs (2006) increasing bone mineral density in human parathyroid hormone (1-34) for
7(4):349-359. postmenopausal osteoporosis. J. Clin. 2 years and relevance to human safety.
28. KULKARNI NH, HALLADAY DL, Endocrinol. Metab. (2003) Toxicol. Pathol. (2002) 30(3):312-321.
MILES RR et al.: Effects of parathyroid 88(11):5212-5220. 46. VAHLE JL, LONG GG, SANDUSKY G
hormone on Wnt signaling pathway in 37. HODSMAN AB, FRAHER LJ, et al.: Bone neoplasms in F344 rats given
bone. J. Cell. Biochem. (2005) WATSON PH et al.: A randomized teriparatide [rhPTH(1-34)] are dependent
95(6):1178-1190. controlled trial to compare the efficacy of on duration of treatment and dose.
•• In this paper, the authors describe the role cyclical parathyroid hormone versus cyclical Toxicol. Pathol. (2004) 32(4):426-438.
of β-catenin in the PTH osteogenic parathyroid hormone and sequential •• In this paper, the authors show that the
response of PTH. calcitonin to improve bone mass in effects of teriparatide on bone tumor
29. WANG YH, LIU Y, ROWE DW: Effects of postmenopausal women with osteoporosis. incidence were clearly dependent on dose
transient PTH on early proliferation, J. Clin. Endocrinol. Metab. (1997) and duration of treatment.
For personal use only.

apoptosis, and subsequent differentiation of 82(2):620-628. 47. TASHJIAN AH Jr, CHABNER BA:
osteoblast in calvarial osteoblast cultures. 38. BLACK DM, GREENSPAN SL, Commentary on clinical safety of
Am. J. Physiol. (2007) 292:E594-E603. ENSRUD KE et al.: The effects of recombinant human parathyroid
30. OKAZAKI K, JINGUSHI S, IKENOUE T parathyroid hormone and alendronate alone hormone 1-34 in the treatment of
et al.: Expression of parathyroid or in combination in postmenopausal osteoporosis in men and postmenopausal
hormone-related peptide and insulin-like osteoporosis. N. Engl. J. Med. (2003) women. J. Bone Miner. Res. (2002)
growth factor I during rat fracture healing. 349(13):1207-1215. 17(7):1151-1161.
J. Orthop. Res. (2003) 21(3):511-520. 39. BODY JJ, GAICH GA, SCHEELE WH 48. RUBIN MR, BILEZIKIAN JP: Parathyroid
•• In this paper, the authors reveal et al.: A randomized double-blind trial to hormone as an anabolic skeletal therapy.
the interaction of PTH and IGF-I compare the efficacy of teriparatide Drugs (2005) 65(17):2481-2498.
during fracture healing in their [recombinant human parathyroid 49. SKRIPITZ R, ASPENBERG P: Parathyroid
experimental study. hormone (1-34)] with alendronate in hormone – a drug for orthopedic surgery?
31. PALMER M, ADAMI HO, postmenopausal women with osteoporosis. Acta Orthop. Scand. (2004) 75(6):654-662.
KRUSEMO UB, LJUNGHALL S: J. Clin. Endocrinol. Metab. (2002)
50. MORLEY P: Delivery of parathyroid
Increased risk of malignant diseases after 87(10):4528-4535.
hormone for the treatment of osteoporosis.
surgery for primary hyperparathyroidism. 40. FINKELSTEIN JS, HAYES A, Expert Opin. Drug Deliv. (2005)
A nationwide cohort study. Am. J. HUNZELMAN JL et al.: The effects of 2(6):993-1002.
Epidemiol. (1988) 127(5):1031-1040. parathyroid hormone, alendronate, or both
in men with osteoporosis. N. Engl. J. Med.
32. LANE NE, SANCHEZ S, MODIN GW Affiliation
et al.: Bone mass continues to increase at the (2003) 349(13):1216-1226. Byron Chalidis1 MD, Trauma Fellow,
hip after parathyroid hormone treatment is 41. KURLAND ES, COSMAN F, Christopher Tzioupis1 MD, Research Fellow,
discontinued in glucocorticoid-induced MCMAHON DJ et al.: Parathyroid Eleftherios Tsiridis1 FRCS PhD, Consultant
osteoporosis: results of a randomized hormone as a therapy for idiopathic Orthopaedic Surgeon &
controlled clinical trial. J. Bone Miner. Res. osteoporosis in men: effects on bone Peter V Giannoudis†1 BSc MB MD EEC
(2000) 15(5):944-951. mineral density and bone markers. J. Clin. (Ortho), Professor of Trauma and
33. MILLER PD, BILEZIKIAN JP, DEAL C, Endocrinol. Metab. (2000) Orthopaedic Surgery
85(9):3069-3076. †Author for correspondence
HARRIS ST, CI RP: Clinical use of
1University of Leeds, Academic Unit, Clarendon
teriparatide in the real world: initial 42. ORWOLL ES, SCHEELE WH, PAUL S
insights. Endocr. Pract. (2004) et al.: The effect of teriparatide [human Wing, Leeds Teaching Hospitals NHS Trust,
10(2):139-148. parathyroid hormone (1-34)] therapy on Great George Street, Leeds, LS1 3EX, UK
bone density in men with osteoporosis. Tel: +44 (0)113 392 2611;
J. Bone Miner. Res. (2003) 18(1):9-17. E-mail: pgiannoudi@aol.com

Expert Opin. Investig. Drugs (2007) 16(4) 449

Potrebbero piacerti anche