You are on page 1of 9

Parathyroid Hormone for Bone Regeneration

Samantha J. Wojda,1 Seth W. Donahue2


1
Department of Mechanical Engineering, Colorado State University, Fort Collins, Colorado, 2Department of Biomedical Engineering, University of
Massachusetts, Amherst, Massachusetts
Received 22 February 2018; accepted 18 June 2018
Published online 21 June 2018 in Wiley Online Library (wileyonlinelibrary.com). DOI 10.1002/jor.24075

ABSTRACT: Delayed healing and/or non-union occur in approximately 5–10% of the fractures that occur annually in the United States.
Segmental bone loss increases the probability of non-union. Though grafting can be an effective treatment for segmental bone loss,
autografting is limited for large defects since a limited amount of bone is available for harvest. Parathyroid hormone (PTH) is a key
regulator of calcium homeostasis in the body and plays an important role in bone metabolism. Presently PTH is FDA approved for use
as an anabolic treatment for osteoporosis. The anabolic effect PTH has on bone has led to research on its use for bone regeneration
applications. Numerous studies in animal models have indicated enhanced fracture healing as a result of once daily injections of PTH.
Similarly, in a human case study, non-union persisted despite treatment attempts with internal fixation, external fixation, and
autograft in combination with BMP-7, until off label use of PTH1-84 was utilized. Use of a biomaterial scaffold to locally deliver PTH to
a defect site has also been shown to improve bone formation and healing around dental implants in dogs and drill defects in sheep.
Thus, PTH may be used to promote bone regeneration and provide an alternative to autograft and BMP for the treatment of large
segmental defects and non-unions. This review briefly summarizes the unmet clinical need for improved bone regeneration techniques
and how PTH may help fill that void by both systemically and locally delivered PTH for bone regeneration applications. ß 2018
Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:2586–2594, 2018.

Keywords: bone tissue engineering and repair; bone/bone biology; bone fracture; repair/therapeutics

Bone generally has effective capacity to heal and, due limited to; bone grafting, distraction osteogenesis, and
to continuous remodeling of bone throughout adult bone transport, Masquelete technique, low intensity
life, healing often occurs with no scar.1 However, pulsed ultrasound (LIPUS) and pulsed electromagnetic
delayed healing and/or non-union occur in an esti- field therapy (PEMF). Open fractures, non-unions
mated 5–10% of the approximately 7.9 million frac- complicated by segmental defects, or cases where
tures that occur annually in the United States.2,3 If removal of tissue at a site of non-union is necessary
the bone fracture healing process exceeds 3 months, for treatment,17 are typically addressed by shortening
healing is considered to be delayed. If healing is not the bone or filling the defect.18 Shortening the bone
achieved by 9 months with no evidence of new healing has a limit of 3 cm in the upper limb and 1 cm in the
in the previous 3 months, the fracture is considered to lower limb before inducing a functional deficit for that
be a non-union. Non-union incidence varies (ranging limb. Filling the gap is traditionally achieved via bone
from 1.9% to 30%) depending on fracture location, graft or bone transport.18 The gold standard for bone
configuration, and associated soft tissue damage, as grafting is autograft. Autograft has the major down-
well as other treatment and lifestyle factors.4,5 Factors falls of donor site pain and morbidity as well as limited
that may disrupt healing and put patients at higher availability,19–21 as it is impractical to harvest a
risk for non-union include, but are not limited to, sufficient amount of bone for repairing a critical size
inadequate immobilization, obesity, smoking, age, mal- defect. As a result, allografts are a common alterna-
nutrition, and multiple injury.6–10 Segmental bone tive. However, allografts have a higher rate of failure
loss, infection, and complications from previous sur- (delayed or non-union, infection, or fracture) compared
geries may also contribute to non-union. Open frac- to autograft as well as potential for immunogenicity
tures, like those potentially caused by high energy problems.18,22–25 Regardless of the downfalls, grafting
trauma, or large bone defects caused by trauma or is presently the most effective option and approxi-
tumor resection have a higher relative risk for non- mately 2.2 million orthopaedic procedures utilize auto-
union.11–14 Indirect costs, like productivity losses due graft or allograft each year to treat large orthopaedic
to the injury, account for more than half of the total defects.26
cost of long bone non-union.15 Average cost for treat-
The limitations of grafting, and other currently
ment of a long bone non-union was reported as US
FDA approved methods, have led to progress in the
$11,333.16
exploration of bioactive molecules for promotion of
Current treatment options are dependent on loca-
bone regeneration. The cellular mechanisms behind
tion and type of non-union. For those complicated by
bone regeneration are not completely understood,
segmental bone loss options include, but are not
presenting challenges in optimizing alternatives to
bone grafts. However, the components that are under-
stood can be expanded upon as potential treatments.
Grant sponsor: NIH/NCRR Colorado CTSI; Grant number: UL1 For example, stem cells, gene therapy, and growth and
RR025780.
Correspondence to: Seth W. Donahue (T: 413-545-6366; differentiation factors may all have potential for
F: 413-545-0724; E-mail: swdonahue@umass.edu) complex healing applications. Thus far, bone morpho-
# 2018 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. genetic proteins (BMPs) have shown the most promise.

2586 JOURNAL OF ORTHOPAEDIC RESEARCH1 OCTOBER 2018


1554527x, 2018, 10, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/jor.24075 by Universidad Autonoma De Yucatan, Wiley Online Library on [08/11/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
PARATHYROID HORMONE FOR BONE REGENERATION 2587

BMPs plays a role in bone regeneration and are known secretion.42,43 The pulsatile component is highly sensi-
to robustly promote osteogenic differentiation. BMPs tive to changes in ionized calcium and calcitrol levels.
have, therefore, been combined with materials like When extracellular calcium concentrations are outside
hydroxyapatite, Di-calcium phosphate dehydrate of the normal range the parathyroid glands change
(DCPD), collagen, PLGA, and poly(propylene fuma- secretion of PTH to return serum calcium levels to
rate) (PPF), many others in order to create an implant normal (using the skeleton as a calcium reservoir).
to promote bone healing.27–30 In 2001 the FDA The classic actions of PTH, including stimulating bone
approved OP-1TM, a BMP-7 implant, as a potential resorption and increasing calcium reabsorption, reside
alternative to autograft in recalcitrant long bone non- in the N-terminal 1–34 region of the protein.44 PTH
union situations. A short time later, InfuseTM, bovine acts through a g-protein coupled receptor, type 1 PTH
collagen with BMP-2, was FDA approved. Since then receptor (PTHR1). When PTH binds to the receptor,
BMP-2 has become a prominent treatment option for adenylate cyclase, or phospholipase C is activated
spinal fusion applications, tibial fractures, and sinus starting a cascade that activates protein kinase A
repair. BMP- 2 and BMP-7 have been shown to (PKA) or protein kinase C (PKC), respectively.45
perform as well as iliac crest bone grafts (ICBG) for Activation of the PKA pathway is mediated by the Gas
spinal fusion applications as well as long bone non- part of the receptor, this cascade stimulates cAMP
union applications. BMP has also been effectively used production, and subsequently PKA. Activation of the
to enhance chances of union after failed grafting in PKC pathway is a Gaq mediated response. PKC is
both tibial and femur non-unions,31,32 and in upper activated through phospholipase C dependent and
limb-non unions when used in conjunction with autolo- independent pathways, which leads to activation of
gous bone graft.33 BMP use may also be efficacious for osteoblastic bone formation.46 Through these pathways
some trauma applications.34–37 However, BMP use PTH can mediate both bone formation and bone
comes with the possibility of an increased risk of resorption. Thus, these pathways are generally cred-
cancer at 24 months.38 When BMP is used in non- ited with control of the classic actions of PTH.47
spinal orthopaedic procedures, serious adverse events However, the evidence suggesting PTH induces MSC
may lead to additional surgeries.39 Like ICBG, the differentiation to the osteoblast lineage is not neces-
success of BMP is somewhat dependent on the type of sarily explained through the traditional pathway.
fracture as well as other patient predispositions to Anabolic effects may also stem from the effect PTH
non-union and is similarly, if not more, financially has on local factors like BMPs, Wnts, or TGFb through
burdensome than ICBG.40,41 Thus, there is a need for recruitment of their receptors to PTHR1 and/or endo-
additional alternative biologics to promote bone regen- cytosis.46,48,49 PTH has been shown to induce MSC
eration when complications arise. differentiation to the osteoblast lineage through a
Parathyroid hormone (PTH) is a molecule that has signaling cascade that enhances BMP signaling.50
good potential for enhancing bone regeneration in Similarly, T lymphocytes express functional PTHR1
large bone defects. This potential lies in the anabolic and PTH has been shown to increase the production of
effect PTH has on bone. Daily injections of PTH are an Wnt10b by bone marrow CD8þ T cells and induce
effective FDA approved treatment for osteoporosis that lymphocytes to activate canonical Wnt signaling in
results in increases in both bone mineral density and preosteoblasts.51,52 Further supporting the role of T
bone volume. Thus, PTH may promote bone regenera- cells role in increasing capacity of stromal cells to
tion and provide an alternative to autograft and BMP support PTH induced effects, continuous infusion of
use for the treatment of large segmental defects and PTH (mimicking hyperparathyroidism) fails to induce
non-unions. In a case study, treatment attempts with bone resorption and cortical bone loss in mice lacking
internal fixation, external fixation, and autograft in T cells.53 However, as the receptor for PTH is not
combination with BMP-7, non-union persisted until off found in osteoclasts osteoclast response to PTH is
label use of PTH1-84 was utilized.3 This review briefly mediated through osteoblasts.54,55 The overall re-
summarizes how PTH may help fill the clinical need sponse of osteoblasts to PTH stimulation, however, is
for improved bone regeneration techniques. dependent on concentration and timing. Both tonic
and pulsatile secretions play a role in normal bone
BIOLOGICAL MECHANISMS OF PTH MEDIATED maintenance, though it is not fully clear what role
BONE FORMATION each plays. In glucocorticoid induced osteoporosis
Parathyroid hormone, an 84 amino acid protein, is a endogenous tonic PTH levels are decreased, and the
key regulator of calcium homeostasis in the body. amount released in pulsatile fashion is increased.42 It
Normal secretion (from the parathyroid gland) is an has been proposed that the increase in pulsatile
ultradian rhythm with tonic and pulsatile components release is perhaps an attempt at compensation for the
(70% tonic, 30% pulsatile), and is dependent on serum decrease in tonic release, however an osteoporotic
calcium and phosphorous. The half-life of PTH in state still develops. This raises the possibility of an
humans is approximately 2–5 min and the pulsatile underappreciated role of tonic (continuous) PTH secre-
component is in low amplitude high frequency tions in normal bone maintenance that could be used
bursts every 10–20 min superimposed on the tonic to develop novel bone regeneration strategies.

JOURNAL OF ORTHOPAEDIC RESEARCH1 OCTOBER 2018


1554527x, 2018, 10, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/jor.24075 by Universidad Autonoma De Yucatan, Wiley Online Library on [08/11/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
2588 WOJDA AND DONAHUE

Generally, continuously elevated serum PTH levels as well as co-existing deficiencies (like serum 25-
have catabolic effects in bone and intermittently hydroxyvitamin D (25OHD)) with severity of PHPT
elevated PTH is anabolic. This effect is observed in as well as the impact on BMD and bone
both animal and human models. Treatment of rats turnover.66–70 However, no clear explanation stands
with continuous infusion of PTH 1–84 resulted in an out. Perhaps there is a threshold above which
increase in bone formation comparable to that associ- continuously elevated PTH is catabolic and below
ated with intermittent injection, but a more marked which it is anabolic. As the preservative effects seem
calcemic effect and thus a net decrease in trabecular limited to sites of higher remodeling (like trabecular
bone.56 Similarly, continuously administered PTH 1– bone), continuously elevated PTH may also play a
34 caused a dose dependent decrease in dry weight of role in bone regeneration. Thus, it is possible that
the femur and a hyperparathyroidism-like condition in local delivery of PTH to a bone defect may promote
the highest administered dose (214 ng/kg/h) in rats.57 bone healing while minimizing potential negative
This effect is also observed at the cellular level. systemic effects.
Continuous PTH 1–84 treatment for 7 days in mice Contrary to continuously elevated PTH, when inter-
resulted in an increase in osteoclast number at day 2 mittently administered, both PTH 1–34 and 1–84 have
followed by an increase in osteoblast number at day 4. a consistent anabolic effect on bone.59,71–76 When PTH
Whereas daily injections of PTH 1–84 did not affect 1–34 and PTH 1–84 were administered intermittently
osteoclast number and the increase in osteoblast (as once daily injections) at an equivalent molar dose,
number was significantly larger than that of continu- no difference in the anabolic effect on bone was
ous treatment.58 Interestingly, in male Sprague Daw- observed.73 Presently, recombinant PTH 1–34 is FDA
ley rats continuous infusion of PTH 1–34 for 12 days approved in the United States for the use of osteoporo-
(4 mg/100 g/day) had an inconsistent effect. Upon re- sis treatment (administered as daily injections), as
peating the experiment three times, one of the three FORTEO1 (Eli Lilly). Recombinant PTH 1–84 was
experiments showed bone formation was increased approved in Europe in 2006 for the treatment of
with continuous PTH treatment (n ¼ 7 per group) over osteoporosis (but was withdrawn in 2014), as Preo-
that of vehicle treated animals. However, the other tact1. Recombinant PTH 1–84 is also approved in the
two experiments showed no difference between ani- United States for treatment of hypocalcemia in per-
mals receiving vehicle treatment versus continuous sons with hyperparathyroidism (Natpara1). Intermit-
PTH 1–34 treatment.59 This variable response is tently administered PTH decreases the risk of fragility
somewhat consistent with what is observed in humans fractures, increases cortical thickness and improves
with primary hyperparathyroidism (PHPT). Cortical trabecular bone architecture in the ilium of post-
bone loss is characteristic of primary hyperparathy- menopausal women,74,77 and increases bone mineral
roidism, however trabecular bone may be preserved or density (BMD) in the lumbar spine in both men and
even enhanced.60,61 women.78 In osteoporotic women, bone mineral density
In the classic presentation of PHPT, transiliac of the lumbar spine increases 6.3% with PTH treat-
biopsies exhibit increased cortical porosity as well as ment compared to an increase of 4.6% with alendro-
reduced cortical width, but preservation, or enhance- nate (an anti-resorptive) treatment.79 Similarly, a 14%
ment, of trabecular bone architecture.61 These find- increase in bone volume was observed in women with
ings have been corroborated by studies in, post- osteoporosis that received daily injections of PTH 1–34
menopausal women with mild primary hyperparathy- compared to the 24% loss in bone volume in women
roidism (PTH levels: 116 þ/ 19 pg/ml, normal: 10– with osteoporosis who were not treated with PTH.77
65 pg/ml). Post-menopausal women with mild PHPT These changes occur due to an unbalance in remodel-
showed preservation of trabecular bone whereas ing, favoring formation, caused by intermittent PTH
postmenopausal women without PHPT exhibited tra- (daily subcutaneous injection) therapy.46,80
becular bone loss.62,63 Similarly, a study in patients It is important to note PTH treatment in humans is
with asymptomatic PHPT indicated bone mineral limited to 2 years and is associated with a “black box”
density was reduced at cortical sites, but at more warning for potential tumorigenic effects. Surveillance
cancellous sites (like the lumbar spine) it was of human osteosarcoma patients has not yet detected a
preserved.64 Other studies showed no correlation causal association between PTH therapy and osteosar-
between mild asymptomatic PHPT and risk factor for coma.81 However, studies in F344 rats indicated a
vertebral fractures.65 However, more recent studies significant percentage of subjects developed osteosar-
indicated even if bone mineral density was preserved, coma following two years of PTH 1–34 treatment.82
the risk of vertebral fracture was higher in postmen- Interestingly, in F344 rats, PTH 1–84 has an osteosar-
opausal patients with mild PHPT. The risk of verte- coma incidence curve shifted to the right of PTH 1–34
bral fracture appears independent of severity of indicating a lower incidence of osteosarcoma in rats
PHPT. These observations on PHPT indicate there is given PTH 1–84 than 1–34.83 Low dose (5–10 mg/kg)
some inconsistency in response to continuously ele- PTH resulted in no neoplastic changes, however high
vated PTH. The reason for this is unclear. Studies dose (>30 mg/kg) administered for 20–24 months (70–
have examined the correlation of demographic factors 80% of lifespan) resulted in significant increases in

JOURNAL OF ORTHOPAEDIC RESEARCH1 OCTOBER 2018


1554527x, 2018, 10, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/jor.24075 by Universidad Autonoma De Yucatan, Wiley Online Library on [08/11/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
PARATHYROID HORMONE FOR BONE REGENERATION 2589

osteosarcoma incidence.83,84 Similar studies in cyno- did increase callus volume, bone mineral content, and
molgus macaques indicated PTH 1–34 treatment (5 mg/ bone volume fraction.95 Intermittent injections of PTH
kg/d) for 18 months did not result in neoplastic 1–34 have also been shown to enhance guided bone
changes over the course of the 18 months of treatment healing in rat calvarial defects.96 And more recently,
or the 3-year follow-up observation.85 Though there is treatment with 100 mg/kg 3 per week for 8 weeks
the potential tumorigenic effect with long-term high increased union rate and accelerated healing in a
doses of PTH, the ability of PTH to increase bone refractory fracture model in rats.97 Subcutaneous
formation makes it a molecule of interest for short- injections of 40 mg/kg/day PTH in mice resulted in
term low dose bone regeneration applications. enhanced healing (increased bone volume and BMC)
in an open defect reconstructed with a PLA/bTCP
EFFICACY OF SYSTEMIC PTH FOR BONE scaffold.98 Likewise, rats treated with injections of
REGENERATION AND DEFECT REPAIR PTH 1–34 every other day during distraction osteogen-
Based on the success of PTH as an anabolic osteoporo- esis showed increased callus volume and mechanical
sis treatment, it follows that there may also be strength.99 Bone graft healing in spinal fusion models
implications for its use for promoting bone regenera- is also enhanced by intermittently administered PTH
tion in large bone defects. Though it may seem the 1–34.100–102 Daily systemic injections of PTH 1–34
benefits of utilizing PTH for bone regeneration are (40 mg/kg) enhance osseointegration of devitalized fem-
limited to intermittent administration, some of the oral allografts in a critical size femoral defect model in
aforementioned findings suggest continuously elevated mice.103 Daily injections of PTH 1–34 (5 mg/kg/day)
PTH may be anabolic in certain circumstances, such also enhanced allograft incorporation in adult female
as in mild PHPT64 and tonic PTH secretions.42,43 The mongrel hounds.104
ability of PTH to increase remodeling in bone gives it Though it is clear that PTH has positive impacts on
great potential in the realm of fracture healing.86 bone regeneration, the ideal administration regime is
Incorporation of PTH into a treatment process for yet to be determined. When the duration of PTH
segmental defects could enhance healing. Research treatment (as daily injections) was investigated in
thus far has examined the potential of systemically mice, it was found that immediate treatment for only
administered PTH and PTH delivered locally (directly 2 weeks was sufficient to increase the ultimate torque
to the defect site). of femurs with diaphyseal allografts, but 4 weeks of
Daily injections of PTH seem to have a consistently treatment showed no increased benefit in terms of
beneficial impact on healing of fractures, graft incorpo- bone strength.105 Delaying the initiation of PTH
ration, implant fixation, distraction osteogenesis, and treatment until 4 weeks after surgery was completely
spinal fusion in animal models.87–91 Treatment of rats ineffective, whereas delaying only 1 or 2 weeks
with PTH 1–34 (60 mg/kg/day) resulted in a dense resulted in equivalent biomechanical properties as
network of trabeculae in bone chambers as compared immediate treatment.105 When PTH was administered
to control.86 Similarly, treatment with PTH 35 mg/kg/ at 15 mg/kg daily, 35 mg/kg 3 days/week, or 105 mg/kg
day or 105 mg/kg 3 per week resulted in a dose once per week, all PTH treatments resulted in more
dependent increase in bone ingrowth into bone cham- bone formation than controls; however, PTH 105 mg/kg
bers in rat calvaria.89 Studies in mice examining once per week resulted in greater bone formation than
treatment with 10, 40, and 200 mg/kg/day show similar the other PTH groups.106 Four weeks of PTH treat-
dose dependent increases in bone formation in the ment in mice examining differences in low dose/high
fracture callus.92 In male rats, 75 mg/kg/day PTH 1–84 frequency (3 mg/kg, 3 times/day), low dose/low fre-
for 12 weeks increased the healing of a femoral quency (9 mg/kg, once/day), high dose/high frequency
defect.90 Daily injections of PTH 1–34 (15 nmol/kg/day) (9 mg/kg, 3 times/day), and high dose/low frequency
and PTH 1–31 (15 nmol/kg/day) have been shown to (27 mg/kg, once/day) indicated high dose and/or high
increase the external volume and strength of closed frequency treatment increased callus bone volume but
fracture calluses.91 Similarly, closed fractures in rats did not have a significant impact on biomechanical
treated with daily injections of PTH showed an recovery whereas low dose/low-frequency therapy
increase in bone mineral content, bone mineral den- resulted in a lesser increase in callus bone volume but
sity, and cartilage volume.93 Accelerated healing was significant increases in biomechanical recovery mea-
observed with PTH treatment in a closed fracture sures.107 Likewise, studies by Sheyn et al.108 indicate
model in female cynomolgus monkeys.94 Systemic 0.4 mg/kg was more successful in accelerating and
intermittent injections of PTH have a consistently maintaining repair of bone defects in ostoportic rats
anabolic effect on healing of closed fractures, indicat- than higher doses. Current FDA approved treatment
ing similar effects may be observed in more complex is 20 mg/day (0.3–0.5 mg/kg) for up to 2 years when
healing scenarios like open fractures or segmental used in humans for treatment of osteoporosis. The
bone defects. dosages used in animal studies range from 0.4 to
An increase in union rate has not been observed as 40 mg/kg. Some studies show a dose response to PTH
a result of PTH treatment for open fractures in animal and some do not, however it should be acknowledging
models. However, like with closed fractures, treatment the higher doses in rodent studies are necessarily less

JOURNAL OF ORTHOPAEDIC RESEARCH1 OCTOBER 2018


1554527x, 2018, 10, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/jor.24075 by Universidad Autonoma De Yucatan, Wiley Online Library on [08/11/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
2590 WOJDA AND DONAHUE

clinically relevant. Still, the high dose may be utilized EFFECT OF LOCALLY DELIVERED PTH ON
for studies ranging from 4 to 12 weeks in duration as FRACTURE HEALING AND BONE DEFECT
opposed to up to 2 years of treatment in clinical REGENERATION
applications. Local delivery of PTH may allow for complete union in
PTH mediated enhancement of closed fracture repair what would otherwise be non-union scenarios while
has been associated with a larger cartilaginous cal- reducing the costs and logistics associated with sys-
lus,109 amplified chondrocyte recruitment, and matura- temic delivery. The effect of locally delivered PTH on
tion in early fracture callus, as well as an increase in fracture healing and bone regeneration has not been
canonical Wnt signaling.110,111 Intermittent PTH as extensively investigated as systemically delivered
(10 mg/kg/day) enhanced closed fracture healing in rats PTH. However, there are some promising results. PTH
by stimulating proliferation and differentiation of osteo- delivered via polyethylene glycol (PEG) matrix
progenitor cells, increased production of bone matrix resulted in a 1.9-fold increase in mineralized bone and
proteins, and enhanced osteoclastogenesis during callus 2.3-fold increase in the area of regeneration in bone
remodeling.112 In a closed fracture model in female chambers in rabbit calvaria.122 Similarly, PEG matri-
cynomolgus monkeys PTH accelerated fracture healing ces with PTH 1–34 (20 mg/ml) were used to fill a
by shrinking callus size and increasing the degree of cylindrical defect around a titanium implant in the
mineralization of the fracture callus (accelerating callus mandible between the first premolar and second molar
remodeling).94 However, when used in conjunction with in dogs. After 12 weeks, matrices infused with PTH
devitalized allografts, PTH 1–34 (40 mg/kg/day) in- had a significantly greater area fraction of newly
creased callus volume and mineral content, but did not formed bone within the defect compared to empty
affect allograft bone volume, osteoclasts, or resorption defects or defects filled with the PEG matrix alone.
surfaces suggesting no increase in graft resorption.103 The PEG matrix with PTH was as effective as an
PTH administration can also influence endothelial autogeneous bone graft when comparing newly formed
cells and stem cells which may aid in bone heal- bone volume between groups.123 PTH 1–34 linked to a
ing.113 The impact PTH has on vascular regeneration fibrin hydrogel with a plasmin sensitive link has also
as well as proliferation may aid in its anabolic been investigated as a method for enhanced bone
capabilities for bone regeneration applications. Not healing. Cylindrical drill defects in the femur of sheep
only are bone endothelial cells capable of responding (8 mm diameter, 13 mm depth) were filled with the
to PTH,114 PTH enhances migration of CD45þ/ hydrogel containing PTH at 50, 100, 400, or 1000 mg/
CD34þ (angiogenic progenitor cells) stem cells,115 ml. After 8 weeks the defects containing the hydrogel
and increases the number of hematopoietic stem cells with 100 mg/ml exhibited an increased bone volume
in the bone marrow that can be mobilized into the fraction as compared to the hydrogel control.124 Simi-
peripheral circulation.116 Patients with primary hy- larly implantation of a gene-activated matrix contain-
perparathyroidism (PHPT) show an increased num- ing a plasmid coding for PTH fragment 1–34 resulted
ber of circulating CD45/CD34 stem cells in the in new bone filling a segmental defect in a rat.125 PTH
peripheral blood.117 Similarly, PTH levels in patients 1–34 covalently linked to a fibrin hydrogel (1 mg/ml)
with untreated PHPT are correlated with elevated has also been shown to improve postoperative progno-
levels of circulating bone marrow-derived progenitor sis of subchondral bone cysts in the distal aspect of the
cells.117 The ability of PTH to facilitate stem cell proximal phalanx in horses.126 Local delivery of PTH
mobilization was corroborated by a phase 1 clinical treatment has also been achieved through use of gene-
trial where PTH enhanced CD34 cells in patients activated matrices (GAM) which induce local produc-
with inadequate CD34þ cell counts.118 PTH also has tion of PTH at the defect site. Treatment with PTH-
been shown to increase proliferation of bone marrow GAM and a collagen matrix has been shown to
stem cells and facilitate homing to sites of therapeu- promote bone regeneration in a craniomaxillofacial
tic need.119,120 In both athymic rats and pigs PTH bone defect.127 Though pulsatile release of PTH from
significantly enhanced mesenchymal stem cell (MSC) biomaterial scaffolds has been explored, they have not
migration to the lumbar region (location of cylindri- been tested extensively in vivo for bone regeneration
cal drill defects in two vertebral bodies) where they applications.128,129 One study showed PTH delivered
differentiated into bone forming cells.108 There also via a cell free biomimetic nanofibrous scaffold designed
is evidence for a synergistic effect of treatment with to deliver a daily pulse of PTH for 21 days. This
exogenous MSCs and PTH concomitantly when used treatment resulted in improved regeneration of a
to treat rib defects in athymic rats.121 The impact critical-size calvarial defect with negligible systemic
PTH has on homing of cells to injury sites could be side effects in a mouse model.130 These studies indi-
controlled outside of the already well-established cate that locally delivered PTH may prove to be
pathways resulting in the anabolic effect of PTH. effective and affordable substitutes for bone grafts for
Though classic actions are generally accepted to procedures such as the repair of critical size bone
occur through the aforementioned PKA and PKC defects. Though studies indicate locally delivered PTH
signaling resulting from binding to PTHR1, there are is a promising method for enhancing bone regenera-
other possibilities. tion, much research is still needed to determine the

JOURNAL OF ORTHOPAEDIC RESEARCH1 OCTOBER 2018


1554527x, 2018, 10, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/jor.24075 by Universidad Autonoma De Yucatan, Wiley Online Library on [08/11/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
PARATHYROID HORMONE FOR BONE REGENERATION 2591

delivery materials, PTH concentrations, and PTH 4. Mills LA, Aitken SA, Simpson A. 2017. The risk of non-
delivery profiles that most efficaciously promote bone union per fracture: current myths and revised figures from
a population of over 4 million adults. Acta Orthop
regeneration.
88:434–439.
5. Tzioupis C, Giannoudis PV. 2007. Prevalence of long-bone
SUMM ARY, CONCLUSION , AND FUTURE non-unions. Injury 38:S3–S9.
DIRECTIONS 6. Green E, Lubahn JD, Evans J. 2005. Risk factors, treat-
Parathyroid hormone is an effective anabolic agent used ment, and outcomes associated with nonunion of the
in the treatment of osteoporosis. The evidence indicating midshaft humerus fracture. J Surg Orthop Adv 14:64–72.
beneficial effects of systemic administration of PTH on 7. Perumal V, Roberts CS. 2007. (ii) Factors contributing to
non-union of fractures. Current Orthopaedics 21:258–261.
fracture healing are difficult to dispute. However, the
8. Buckwalter J, Cruess R. 1991. Fractures in adults. Phila-
timing and dose of PTH treatment have yet to be delphia: J.B. Lippincott Company.
optimized. At least two studies have shown that PTH 9. Guarniero R, de Barros Filho TE, Tannuri U, et al. 1992.
injections administered once to three times a week (as Study of fracture healing in protein malnutrition. Rev Paul
opposed to daily) may be sufficient to promote heal- Med 110:63–68.
ing.97,106 However, the ideal window for PTH administra- 10. Dodds RA, Catterall A, Bitensky L, et al. 1986. Abnormali-
tion is unknown;105 for example, is PTH treatment ties in fracture healing induced by vitamin B6-deficiency in
rats. Bone 7:489–495.
necessary during the entire healing process or could it be 11. Livingston TL, Gordon S, Archambault M, et al. 2003.
limited to healing phases that are most responsive to Mesenchymal stem cells combined with biphasic calcium
PTH? Though positive outcomes result from systemically phosphate ceramics promote bone regeneration. J Mater
administered PTH, can the same (or better) anabolic Sci Mater Med 14:211–218.
effect be achieved with locally delivered PTH for bone 12. Karladani AH, Granhed H, Karrholm J, et al. 2001. The
regeneration applications? Studies thus far indicate prom- influence of fracture etiology and type on fracture healing:
a review of 104 consecutive tibial shaft fractures. Arch
ising outcomes for locally delivered PTH for bone regener-
Orthop Trauma Surg 121:325–328.
ation applications. However, it is unclear whether or not 13. Harris AM, Althausen PL, Kellam J, et al. 2009. Complica-
continuous or intermittent release is more efficacious. The tions following limb-threatening lower extremity trauma.
findings that tonic PTH secretion plays a role in normal J Orthop Trauma 23:1–6.
bone metabolism,42 and post-menopausal women with 14. Beaver R, Brinker MR, Barrack RL. 1997. An analysis of
mild primary hyperparathyroidism have a protective the actual cost of tibial nonunions. J La State Med Soc
effect against osteoporosis62 support the idea that low 149:200–206.
15. Hak DJ, Fitzpatrick D, Bishop JA, et al. 2014. Delayed
levels of continuously delivered PTH can be used to
union and nonunions: epidemiology, clinical issues, and
promote bone regeneration. Thus, future studies that financial aspects. Injury 45:S3–S7.
more closely examine the efficacy of locally delivered PTH 16. Busse JW, Bhandari M, Sprague S, et al. 2005. An
for bone regeneration applications could lead to improved economic analysis of management strategies for closed and
treatments for segmental defects in bone. open grade I tibial shaft fractures. Acta Orthop 76:
705–712.
17. Ring D, Barrick WT, Jupiter JB. 1997. Recalcitrant non-
AUTHORS’ CONTRIBUTION union. Clin Orthop Relat Res 340:181–189.
SJW: substantial contribution; completed literature 18. Mahendra A, Maclean AD. 2007. Available biological treat-
search, reviewed articles, wrote, and revised paper. ments for complex non-unions. Injury 38:S7–12.
SWD: substantial contribution; reviewed articles, 19. Coventry MB, Tapper EM. 1972. Pelvic instability: a
edited, and revised paper. Both authors approved the consequence of removing iliac bone for grafting. J Bone
Joint Surg Am 54:83–101.
submission and final version of this manuscript.
20. Damien CJ, Parsons JR. 1991. Bone graft and bone graft
substitutes: a review of current technology and applica-
ACKNOWLEDGMENTS tions. J Appl Biomater 2:187–208.
We thank Dr. Michael Yaszemski and Dr. Kristi Anseth for 21. Banwart JC, Asher MA, Hassanein RS. 1995. Iliac crest
helpful discussions on the clinical aspects of bone regenera- bone graft harvest donor site morbidity. A statistical
evaluation. Spine (Phila Pa 1976) 20:1055–1060.
tion and approaches for local PTH delivery to large bone
22. Meijer GJ, de Bruijn JD, Koole R, et al. 2007. Cell-based
defects. This publication was supported in part by funding
bone tissue engineering. PLoS Med 4:e9.
from NIH/NCRR Colorado CTSI Grant Number UL1
23. Stevenson S, Horowitz M. 1992. The response to bone
RR025780. The content is solely the responsibility of the
allografts. J Bone Joint Surg Am 74:939–950.
authors and does not necessarily represent the official views 24. Friedlaender GE. 1983. Immune responses to osteochondral
of the National Institutes of Health. allografts. Current knowledge and future directions. Clin
Orthop Relat Res (174):58–68.
REFERENCES 25. Tiyapatanaputi P, Rubery PT, Carmouche J, et al. 2004.
1. Einhorn TA. 1998. The cell and molecular biology of A novel murine segmental femoral graft model. J Orthop
fracture healing. Clin Orthop Relat Res (355 Suppl): S7–21. Res 22:1254–1260.
2. Marsell R, Einhorn TA. 2010. Emerging bone healing 26. Jahangir AA, Nunley RM, Mehta S, et al. 2008. Bone-graft
therapies. J Orthop Trauma 24:S4–S8. substitutes in orthopaedic surgery. AAOS Now: American
3. Paridis D, Karachalios T. 2011. Atrophic femoral bone Academy of Orhthopaedic Surgeons.
nonunion treated with 1–84 PTH. J Musculoskelet Neuro- 27. Tuominen T, Jamsa T, Oksanen J, et al. 2001. Composite
nal Interact 11:320–322. quiz 323. implant composed of hydroxyapatite and bone morphoge-

JOURNAL OF ORTHOPAEDIC RESEARCH1 OCTOBER 2018


1554527x, 2018, 10, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/jor.24075 by Universidad Autonoma De Yucatan, Wiley Online Library on [08/11/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
2592 WOJDA AND DONAHUE

netic protein in the healing of a canine ulnar defect. Ann 46. Jilka RL. 2007. Molecular and cellular mechanisms of the
Chir Gynaecol 90:32–36. anabolic effect of intermittent PTH. Bone 40:1434–1446.
28. Chu TM, Sargent P, Warden SJ, et al. 2006. Prelimi- 47. Silva BC, Bilezikian JP. 2015. Parathyroid hormone:
nary evaluation of a load-bearing BMP-2 carrier for anabolic and catabolic actions on the skeleton. Curr Opin
segmental defect regeneration. Biomed Sci Instrum Pharmacol 22:41–50.
42:42–47. 48. Kousteni S, Bilezikian JP. 2008. The cell biology of
29. Kempen DH, Lu L, Hefferan TE, et al. 2008. Retention of parathyroid hormone in osteoblasts. Curr Osteoporos Rep
in vitro and in vivo BMP-2 bioactivities in sustained 6:72–76.
delivery vehicles for bone tissue engineering. Biomaterials 49. Polo S, Di Fiore PP. 2006. Endocytosis conducts the cell
29:3245–3252. signaling orchestra. Cell 124:897–900.
30. Cowan CM, Soo C, Ting K, et al. 2005. Evolving concepts in 50. Yu B, Zhao X, Yang C, et al. 2012. Parathyroid hormone
bone tissue engineering. Curr Top Dev Biol 66:239–285. induces differentiation of mesenchymal stromal/stem cells
31. Zimmermann G, Wagner C, Schmeckenbecher K, et al. by enhancing bone morphogenetic protein signaling.
2009. Treatment of tibial shaft non-unions: bone morphoge- J Bone Miner Res 27:2001–2014.
netic proteins versus autologous bone graft. Injury 40: 51. Terauchi M, Li JY, Bedi B, et al. 2009. T lymphocytes
S50–S53. amplify the anabolic activity of parathyroid hormone
32. Kanakaris NK, Lasanianos N, Calori GM, et al. 2009. through Wnt10b signaling. Cell Metab 10:229–240.
Application of bone morphogenetic proteins to femoral non- 52. Pacifici R. 2013. Role of T cells in the modulation of PTH
unions: a 4-year multicentre experience. Injury 40: action: physiological and clinical significance. Endocrine
S54–S61. 44:576–582.
33. Singh R, Bleibleh S, Kanakaris NK, et al. 2016. Upper limb 53. Gao Y, Wu X, Terauchi M, et al. 2008. T cells potentiate
non-unions treated with BMP-7: efficacy and clinical PTH-induced cortical bone loss through CD40L signaling.
results. Injury 47:S33–S39. Cell Metab 8:132–145.
34. Govender S, Csimma C, Genant HK, et al. 2002. Recombi- 54. Teitelbaum SL. 2000. Bone resorption by osteoclasts.
nant human bone morphogenetic protein-2 for treatment of Science 289:1504–1508.
open tibial fractures: a prospective, controlled, randomized 55. McSheehy PM, Chambers TJ. 1986. Osteoblastic cells
study of four hundred and fifty patients. J Bone Joint Surg mediate osteoclastic responsiveness to parathyroid hor-
Am 84-A:2123–2134. mone. Endocrinology 118:824–828.
35. Aro HT, Govender S, Patel AD, et al. 2011. Recombinant 56. Tam CS, Heersche JNM, Murray TM, et al. 1982. Parathy-
human bone morphogenetic protein-2: a randomized trial roid-Hormone stimulates the bone apposition rate indepen-
in open tibial fractures treated with reamed nail fixation. dently of its resorptive action—Differential-Effects of
J Bone Joint Surg Am 93:801–808. intermittent and continuous administration. Endocrinology
36. Bhattacharyya T. 2011. Commentary on an article by 110:506–512.
Hannu T. Aro, MD, PhD, et al.: “Recombinant human bone 57. Uzawa T, Hori M, Ejiri S, et al. 1995. Comparison of the
morphogenetic protein-2: a randomized trial in open tibial effects of intermittent and continuous administration of
fractures treated with reamed nail fixation.” J Bone Joint human parathyroid hormone(1-34) on rat bone. Bone
Surg Am 93:e50. 16:477–484.
37. Study to Prospectively Evaluate Reamed Intramedullary 58. Bellido T, Ali AA, Plotkin LI, et al. 2003. Proteasomal
Nails in Patients with Tibial Fractures I Bhandari M, degradation of Runx2 shortens parathyroid hormone-in-
Guyatt G, et al. 2008. Randomized trial of reamed and duced anti-apoptotic signaling in osteoblasts. A putative
unreamed intramedullary nailing of tibial shaft fractures. explanation for why intermittent administration is needed
J Bone Joint Surg Am 90:2567–2578. for bone anabolism. J Biol Chem 278:50259–50272.
38. Fu R, Selph S, McDonagh M, et al. 2013. Effectiveness and 59. Hock JM, Gera I. 1992. Effects of continuous and intermit-
harms of recombinant human bone morphogenetic protein- tent administration and inhibition of resorption on the
2 in spine fusion: a systematic review and meta-analysis. anabolic response of bone to parathyroid hormone. J Bone
Ann Intern Med 158:890–902. Miner Res 7:65–72.
39. Woo EJ. 2013. Adverse events after recombinant human 60. Silverberg SJ, Shane E, de la Cruz L, et al. 1989. Skeletal
BMP2 in nonspinal orthopaedic procedures. Clin Orthop disease in primary hyperparathyroidism. J Bone Miner Res
Relat Res 471:1707–1711. 4:283–291.
40. Dahabreh Z, Calori GM, Kanakaris NK, et al. 2009. A cost 61. Parisien M, Silverberg SJ, Shane E, et al. 1990. The
analysis of treatment of tibial fracture nonunion by bone histomorphometry of bone in primary hyperparathyroid-
grafting or bone morphogenetic protein-7. Int Orthop ism: preservation of cancellous bone structure. J Clin
33:1407–1414. Endocrinol Metab 70:930–938.
41. Giorgio Calori M, Capanna R, Colombo M, et al. 2013. Cost 62. Dempster DW, Muller R, Zhou H, et al. 2007. Preserved
effectiveness of tibial nonunion treatment: a comparison three-dimensional cancellous bone structure in mild pri-
between rhBMP-7 and autologous bone graft in two Italian mary hyperparathyroidism. Bone 41:19–24.
centres. Injury 44:1871–1879. 63. Dempster DW, Parisien M, Silverberg SJ, et al. 1999. On
42. Chiavistelli S, Giustina A, Mazziotti G. 2015. Parathyroid the mechanism of cancellous bone preservation in postmen-
hormone pulsatility: physiological and clinical aspects. opausal women with mild primary hyperparathyroidism.
Bone Res 3:14049. J Clin Endocrinol Metab 84:1562–1566.
43. Schmitt CP, Schaefer F, Bruch A, et al. 1996. Control of 64. Silverberg SJ, Walker MD, Bilezikian JP. 2013. Asymptom-
pulsatile and tonic parathyroid hormone secretion by atic primary hyperparathyroidism. J Clin Densitom
ionized calcium. J Clin Endocrinol Metab 81:4236–4243. 16:14–21.
44. Potts JT, Jr., Kronenberg HM, Rosenblatt M. 1982. Para- 65. Wilson RJ, Rao S, Ellis B, et al. 1988. Mild asymptomatic
thyroid hormone: chemistry, biosynthesis, and mode of primary hyperparathyroidism is not a risk factor for
action. Adv Prot Chem 35:323–396. vertebral fractures. Ann Intern Med 109:959–962.
45. Karaplis AC, Goltzman D. 2000. PTH and PTHrP effects on 66. Grey A, Lucas J, Horne A, et al. 2005. Vitamin D repletion
the skeleton. Rev Endocr Metab Disord 1:331–341. in patients with primary hyperparathyroidism and coexis-

JOURNAL OF ORTHOPAEDIC RESEARCH1 OCTOBER 2018


1554527x, 2018, 10, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/jor.24075 by Universidad Autonoma De Yucatan, Wiley Online Library on [08/11/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
PARATHYROID HORMONE FOR BONE REGENERATION 2593

tent vitamin D insufficiency. J Clin Endocrinol Metab hormone 1–84 in a 2-year study in Fischer 344 rats. Toxicol
90:2122–2126. Pathol 34:929–940.
67. Stein EM, Dempster DW, Udesky J, et al. 2011. Vitamin D 84. Vahle JL, Long GG, Sandusky G, et al. 2004. Bone
deficiency influences histomorphometric features of bone in neoplasms in F344 rats given teriparatide [rhPTH(1-34)]
primary hyperparathyroidism. Bone 48:557–561. are dependent on duration of treatment and dose. Toxicol
68. Moosgaard B, Christensen SE, Vestergaard P, et al. 2008. Pathol 32:426–438.
Vitamin D metabolites and skeletal consequences in pri- 85. Vahle JL, Zuehlke U, Schmidt A, et al. 2008. Lack of bone
mary hyperparathyroidism. Clin Endocrinol (Oxf) neoplasms and persistence of bone efficacy in cynomolgus
68:707–715. macaques after long-term treatment with teriparatide
69. Walker MD, Nishiyama KK, Zhou B, et al. 2016. Effect of [rhPTH(1–34)]. J Bone Miner Res. 23:2033–2039.
low vitamin D on volumetric bone mineral density, bone 86. Skripitz R, Andreassen TT, Aspenberg P. 2000. Strong
microarchitecture, and stiffness in primary hyperparathy- effect of PTH (1–34) on regenerating bone: a time sequence
roidism. J Clin Endocrinol Metab 101:905–913. study in rats. Acta Orthop Scand 71:619–624.
70. Makras P, Anastasilakis AD. 2018. Bone disease in primary 87. Daugaard H, Elmengaard B, Andreassen T, et al. 2011.
hyperparathyroidism. Metabolism 80:57–65. Parathyroid hormone treatment increases fixation of ortho-
71. Fox J, Miller MA, Recker RR, et al. 2007. Effects of pedic implants with gap healing: a biomechanical and
treatment of ovariectomized adult rhesus monkeys with histomorphometric canine study of porous coated titanium
parathyroid hormone 1–84 for 16 months on trabecular and alloy implants in cancellous bone. Calcif Tissue Int
cortical bone structure and biomechanical properties of the 88:294–303.
proximal femur. Calcif Tissue Int 81:53–63. 88. Daugaard H, Elmengaard B, Andreassen TT, et al. 2012.
72. Jodar-Gimeno E. 2007. Full length parathyroid hormone Systemic intermittent parathyroid hormone treatment
(1-84) in the treatment of osteoporosis in postmenopausal improves osseointegration of press-fit inserted implants in
women. Clin Interv Aging 2:163–174. cancellous bone. Acta Orthop 83:411–419.
73. Mosekilde L, Sogaard CH, Danielsen CC, et al. 1991. The 89. Tsunori K, Sato S, Hasuike A, et al. 2015. Effects of
anabolic effects of human parathyroid hormone (hPTH) on intermittent administration of parathyroid hormone on
rat vertebral body mass are also reflected in the quality of bone augmentation in rat calvarium. Implant Dent
bone, assessed by biomechanical testing: a comparison 24:142–148.
study between hPTH-(1–34) and hPTH-(1–84). Endocrinol- 90. Hamann C, Picke AK, Campbell GM, et al. 2014. Effects of
ogy 129:421–428. parathyroid hormone on bone mass, bone strength, and
74. Neer RM, Arnaud CD, Zanchetta JR, et al. 2001. Effect of bone regeneration in male rats with type 2 diabetes
parathyroid hormone (1-34) on fractures and bone mineral mellitus. Endocrinology 155:1197–1206.
density in postmenopausal women with osteoporosis. 91. Andreassen TT, Willick GE, Morley P, et al. 2004. Treat-
N Engl J Med 344:1434–1441. ment with parathyroid hormone hPTH(1-34), hPTH(1-31),
75. Jerome CP, Burr DB, Van Bibber T, et al. 2001. Treatment and monocyclic hPTH(1-31) enhances fracture strength and
with human parathyroid hormone (1-34) for 18 months callus amount after withdrawal fracture strength and
increases cancellous bone volume and improves trabecular callus mechanical quality continue to increase. Calcif
architecture in ovariectomized cynomolgus monkeys Tissue Int 74:351–356.
(Macaca fascicularis). Bone 28:150–159. 92. Milstrey A, Wieskoetter B, Hinze D, et al. 2017. Dose-
76. Ejersted C, Andreassen TT, Oxlund H, et al. 1993. Human dependent effect of parathyroid hormone on fracture heal-
parathyroid hormone (1–34) and (1–84) increase the me- ing and bone formation in mice. J Surg Res 220:327–335.
chanical strength and thickness of cortical bone in rats. 93. Alkhiary YM, Gerstenfeld LC, Krall E, et al. 2005. En-
J Bone Miner Res 8:1097–1101. hancement of experimental fracture-healing by systemic
77. Jiang Y, Zhao JJ, Mitlak BH, et al. 2003. Recombinant administration of recombinant human parathyroid hor-
human parathyroid hormone (1–34) [teriparatide] improves mone (PTH 1-34). J Bone Joint Surg Am 87:731–741.
both cortical and cancellous bone structure. J Bone Miner 94. Manabe T, Mori S, Mashiba T, et al. 2007. Human
Res 18:1932–1941. parathyroid hormone (1-34) accelerates natural fracture
78. Dempster DW, Cosman F, Kurland ES, et al. 2001. Effects healing process in the femoral osteotomy model of cynomol-
of daily treatment with parathyroid hormone on bone gus monkeys. Bone 40:1475–1482.
microarchitecture and turnover in patients with osteoporo- 95. Tagil M, McDonald MM, Morse A, et al. Intermittent
sis: a paired biopsy study. J Bone Miner Res 16:1846–1853. PTH(1-34) does not increase union rates in open rat
79. Black DM, Greenspan SL, Ensrud KE, et al. 2003. The femoral fractures and exhibits attenuated anabolic effects
effects of parathyroid hormone and alendronate alone or in compared to closed fractures. Bone 46:852–859.
combination in postmenopausal osteoporosis. N Engl J Med 96. Andreassen TT, Cacciafesta V. 2004. Intermittent parathy-
349:1207–1215. roid hormone treatment enhances guided bone regenera-
80. Dempster DW, Zhou H, Ruff VA, et al. 2018. Longitudinal tion in rat calvarial bone defects. J Craniofac Surg
effects of teriparatide or zoledronic acid on bone modeling- 15:424–427. discussion 428-429.
and remodeling-based formation in the SHOTZ study. J 97. Kumabe Y, Lee SY, Waki T, et al. 2017. Triweekly
Bone Miner Res 33:627–633. administration of parathyroid hormone (1-34) accelerates
81. Andrews EB, Gilsenan AW, Midkiff K, et al. 2012. The US bone healing in a rat refractory fracture model. BMC
postmarketing surveillance study of adult osteosarcoma Musculoskelet Disord 18:545.
and teriparatide: study design and findings from the first 7 98. Jacobson JA, Yanoso-Scholl L, Reynolds DG, et al. 2011.
years. J Bone Miner Res 27:2429–2437. Teriparatide therapy and beta-tricalcium phosphate en-
82. Vahle JL, Sato M, Long GG, et al. 2002. Skeletal changes hance scaffold reconstruction of mouse femoral defects.
in rats given daily subcutaneous injections of recombinant Tissue Eng Part A 17:389–398.
human parathyroid hormone (1-34) for 2 years and rele- 99. Seebach C, Skripitz R, Andreassen TT, et al. 2004.
vance to human safety. Toxicol Pathol 30:312–321. Intermittent parathyroid hormone (1-34) enhances mechan-
83. Jolette J, Wilker CE, Smith SY, et al. 2006. Defining a ical strength and density of new bone after distraction
noncarcinogenic dose of recombinant human parathyroid osteogenesis in rats. J Orthop Res 22:472–478.

JOURNAL OF ORTHOPAEDIC RESEARCH1 OCTOBER 2018


1554527x, 2018, 10, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/jor.24075 by Universidad Autonoma De Yucatan, Wiley Online Library on [08/11/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
2594 WOJDA AND DONAHUE

100. Abe Y, Takahata M, Ito M, et al. 2007. Enhancement of cardiac repair by enhanced neovascularization and cell
graft bone healing by intermittent administration of human survival. Cardiovasc Res 77:722–731.
parathyroid hormone (1-34) in a rat spinal arthrodesis 116. Adams GB, Martin RP, Alley IR, et al. 2007. Therapeutic
model. Bone 41:775–785. targeting of a stem cell niche. Nat Biotechnol 25:238–243.
101. O’Loughlin PF, Cunningham ME, Bukata SV, et al. 2009. 117. Brunner S, Theiss HD, Murr A, et al. 2007. Primary
Parathyroid hormone (1-34) augments spinal fusion, fusion hyperparathyroidism is associated with increased circulat-
mass volume, and fusion mass quality in a rabbit spinal ing bone marrow-derived progenitor cells. Am J Physiol
fusion model. Spine (PhilaPa 1976) 34:121–130. Endocrinol Metab 293:E1670–E1675.
102. Lawrence JP, Ennis F, White AP, et al. 2006. Effect of daily 118. Ballen KK, Shpall EJ, Avigan D, et al. 2007. Phase I trial
parathyroid hormone (1-34) on lumbar fusion in a rat of parathyroid hormone to facilitate stem cell mobilization.
model. Spine J 6:385–390. Biol Blood Marrow Transpl 13:838–843.
103. Reynolds DG, Takahata M, Lerner AL, et al. 2011. 119. Calvi LM, Adams GB, Weibrecht KW, et al. 2003. Osteo-
Teriparatide therapy enhances devitalized femoral allograft blastic cells regulate the haematopoietic stem cell niche.
osseointegration and biomechanics in a murine model. Nature 425:841–846.
Bone 48:562–570. 120. Nishida S, Yamaguchi A, Tanizawa T, et al. 1994. In-
104. Nishitani K, Mietus Z, Beck CA, et al. 2017. High dose creased bone formation by intermittent parathyroid hor-
teriparatide (rPTH1-34) therapy increases callus volume mone administration is due to the stimulation of
and enhances radiographic healing at 8-weeks in a massive proliferation and differentiation of osteoprogenitor cells in
canine femoral allograft model. PLoS ONE 12:e0185446. bone marrow. Bone 15:717–723.
https://doi.org/10.1371/journal.pone.0185446 121. Cohn Yakubovich D, Sheyn D, Bez M, et al. 2017. Systemic
105. Takahata M, Schwarz EM, Chen T, et al. 2012. Delayed administration of mesenchymal stem cells combined with
short-course treatment with teriparatide (PTH1-34) parathyroid hormone therapy synergistically regenerates
improves femoral allograft healing by enhancing intramem- multiple rib fractures. Stem Cell Res Ther 8:51.
branous bone formation at the graft-host junction. J Bone 122. Jung RE, Hammerle CHF, Kokovic V, et al. 2007. Bone
Miner Res 27:26–37. regeneration using a synthetic matrix containing a para-
106. Tsunori K. 2015. Effects of parathyroid hormone dosage thyroid hormone peptide combined with a grafting mate-
and schedule on bone regeneration. J Oral Sci 57:131–136. rial. Int J Oral Max Impl 22:258–266.
107. Ota M, Takahata M, Shimizu T, et al. 2018. Optimal 123. Jung RE, Cochran DL, Domken O, et al. 2007. The effect of
administration frequency and dose of teriparatide for matrix bound parathyroid hormone on bone regeneration.
acceleration of biomechanical healing of long-bone fracture Clin Oral Implants Res 18:319–325.
in a mouse model. J Bone Miner Metab. https://doi.org/ 124. Arrighi I, Mark S, Alvisi M, et al. 2009. Bone healing
10.1007/s00774-018-0930-3. [Epub ahead of print] induced by local delivery of an engineered parathyroid
108. Sheyn D, Shapiro G, Tawackoli W, et al. 2016. PTH induces hormone prodrug. Biomaterials 30:1763–1771.
systemically administered mesenchymal stem cells to mi- 125. Fang J, Zhu YY, Smiley E, et al. 1996. Stimulation of new
grate to and regenerate spine injuries. Mol Ther 24: bone formation by direct transfer of osteogenic plasmid
318–330. genes. Proc Nat Acad Sci USA 93:5753–5758.
109. Nakazawa T, Nakajima A, Shiomi K, et al. 2005. Effects of 126. Fuerst A, Derungs S, von Rechenberg B, et al. 2007. Use of
low-dose, intermittent treatment with recombinant human a parathyroid hormone peptide (PTH(1-34))-enriched fibrin
parathyroid hormone (1-34) on chondrogenesis in a model hydrogel for the treatment of a subchondral cystic lesion in
of experimental fracture healing. Bone 37:711–719. the proximal interphalangeal joint of a warmblood filly.
110. Kakar S, Einhorn TA, Vora S, et al. 2007. Enhanced J Vet Med A Physiol Pathol Clin Med 54:107–112.
chondrogenesis and Wnt signaling in PTH-treated frac- 127. Lee PH, Yew TL, Lai YL, et al. 2018. Parathyroid hormone
tures. J Bone Miner Res 22:1903–1912. gene-activated matrix with DFDBA/collagen composite matrix
111. Chen Y, Alman BA. 2009. Wnt pathway, an essential role enhances bone regeneration in rat calvarial bone defects. J
in bone regeneration. J Cell Biochem 106:353–362. Chin Med Assoc. pii: S1726-4901(18)30050-9. https://doi.org/
112. Nakajima A, Shimoji N, Shiomi K, et al. 2002. Mechanisms 10.1016/j.jcma.2017.12.004. [Epub ahead of print]
for the enhancement of fracture healing in rats treated 128. Jeon JH, Puleo DA. 2008. Formulations for intermittent
with intermittent low-dose human parathyroid hormone release of parathyroid hormone (1-34) and local enhance-
(1–34). J Bone Miner Res 17:2038–2047. ment of osteoblast activities. Pharm Dev Technol
113. Langer M, Prisby R, Peter Z, et al. 2011. Simultaneous 3D 13:505–512.
imaging of bone and vessel microstructure in a rat model. 129. Liu X, Pettway GJ, McCauley LK, et al. 2007. Pulsatile
Ieee Trans Nucl Sci 58:139–145. release of parathyroid hormone from an implantable deliv-
114. Brandi ML, Collin-Osdoby P. 2006. Vascular biology and ery system. Biomaterials 28:4124–4131.
the skeleton. J Bone Miner Res 21:183–192. 130. Dang M, Koh AJ, Jin X, et al. 2017. Local pulsatile PTH
115. Zaruba MM, Huber BC, Brunner S, et al. 2008. Parathyroid delivery regenerates bone defects via enhanced bone
hormone treatment after myocardial infarction promotes remodeling in a cell-free scaffold. Biomaterials 114:1–9.

JOURNAL OF ORTHOPAEDIC RESEARCH1 OCTOBER 2018

You might also like