You are on page 1of 8

Calcified Tissue International

https://doi.org/10.1007/s00223-019-00615-z

ORIGINAL RESEARCH

Pretreatment with Pamidronate Decreases Bone Formation


but Increases Callus Bone Volume in a Rat Closed Fracture Model
Alyson Morse1,2 · Michelle M. McDonald1,3 · Kathy Mikulec1 · Aaron Schindeler1,2 · Craig F. Munns2,4 ·
David G. Little1,2

Received: 18 June 2019 / Accepted: 5 August 2019


© Crown 2019

Abstract
Clinical concerns have been raised over prior exposure to bisphosphonates impairing fracture healing. To model this, groups
of male Wistar rats were assigned to saline control or treatment groups receiving 0.15 mg/kg (low dose), 0.5 mg/kg (medium
dose), and 5 mg/kg (high dose) Pamidronate (PAM) twice weekly for 4 weeks. At this point, closed fractures were made
using an Einhorn apparatus, and bisphosphonate dosing was continued until the experimental endpoint. Specimens were
analyzed at 2 and 6 weeks (N = 8 per group per time point). Twice weekly PAM dosing was found to have no effect on early
soft callus remodeling at 2 weeks post fracture. At this time point, the highest dose PAM group gave significant increases in
bone volume (+ 10%, p < 0.05), bone mineral content (+ 30%, p < 0.01), and bone mineral density (+ 10%, p < 0.01). This
PAM dosing regimen showed more substantive effects on hard callus at 6 weeks post fracture, with PAM treatment groups
showing + 46–79% increased bone volume. Dynamic bone labeling showed reduced calcein signal in the PAM-treated cal-
luses (38–63%, p < 0.01) and reduced MAR (32–49%, p < 0.01), suggesting a compensatory reduction in bone anabolism.
These data support the concept that bisphosphonates lead to profound decreases in bone turnover in fracture repair, however,
this does not affect soft callus remodeling.

Keywords  Bisphosphonate · Fracture · Pamidronate · Endochondral ossification · Osteogenesis imperfecta · Atypical


femoral fracture

Introduction and at higher doses to prevent secondary skeletal-related


events associated with metastatic cancers. In pediatrics, they
Bisphosphonates (BPs) are a class of anti-resorptive bone are routinely prescribed off-label to manage osteogenesis
drugs that are widely used in the management of osteoporo- imperfecta, idiopathic low bone density, hypercalcemia, and
sis and metabolic bone disorders in adults and children [1, heterotopic calcifications. Despite their widespread use in
2]. In adult patients, they are widely used to reduce fracture these clinical settings, it is unclear how BPs may interact
risk and associated morbidity and mortality for osteoporosis, with the process of fracture repair, and whether it fundamen-
tally differs from their usage in BP-naïve patients.
Fracture repair is a complex process responsible for facili-
* David G. Little tating the regeneration of bone without the development of
david.little@health.nsw.gov.au
scar tissue. Endochondral bone repair has been historically
1
Orthopaedic Research & Biotechnology Unit, The Children’s described as a sequence of events: inflammation, soft (car-
Hospital at Westmead, Locked Bag 4001, Westmead, tilaginous) callus formation, replacement with hard (bony)
NSW 2145, Australia callus, and bone remodeling, but subsequent models of frac-
2
Discipline of Child and Adolescent Health, Faculty ture healing have focused on anabolic-catabolic processes
of Medicine and Health, University of Sydney, Sydney, [3] or cellular drivers [4]. BPs are known to profoundly
NSW, Australia
interfere with the remodeling phase of fracture repair, inter-
3
Bone Biology Division, The Garvan Institute for Medical fering with osteoclastic bone resorption [5].
Research, Sydney, Australia
Numerous preclinical studies featuring fracture models
4
Department for Endocrinology and Diabetes, The Children’s concomitantly dosed with BPs have confirmed impaired
Hospital at Westmead, Westmead, NSW, Australia

13
Vol.:(0123456789)
A. Morse et al.

remodeling of the hard callus [6–8]. It was initially specu- Hospital Animal Ethics Committee with approval number
lated that BPs may impede chondroclast activity [9, 10], thus 5024.06-07. Rats were randomly allocated into four treat-
negatively impacting on bony union. However, a seminal 2013 ment groups and doses were administered subcutaneously
study revealed that BP treatment enhanced woven bone callus commencing at 5 weeks of age. Control animals received
size without impeding soft callus removal [6]. It was specu- twice weekly saline injections. Pamidronate disodium
lated that soft callus breakdown was enabled by non-osteo- (Novartis Pharma AG, Basil, Switzerland) was prepared
clastic cells able to express matrix metalloproteases (MMPs). in sterile water. The three PAM treatment groups received
Consistent with this concept, MMP inhibitor treatment hin- low, medium, and high doses of 0.15 mg/kg, 0.5 mg/kg,
dered cartilage resorption [6], a finding also supported by data and 5 mg/kg PAM twice weekly (biweekly/biw). Following
from MMP-9 and MMP-13 knockout models [11, 12]. To date, 4 weeks of PAM treatment, fracture surgery was performed
preclinical models have not substantively addressed the effect when rats were 9 weeks of age.
of chronic BP dosing on fracture healing. Prolonged high-dose
BP treatment was employed in a rat model of cyclic loading Fracture Surgery, Monitoring, and Specimen
induced stress fractures [13]. The group reported impaired Collection
stress fracture remodeling at high-dose BPs, but even this
study featured concomitant treatment with risedronate during Using a combination of 75 mg/kg ketamine and 4 mg/kg
cyclic loading rather than pretreatment. xylazine for anesthesia, a small incision was made ante-
The issue of chronic exposure is one with broad relevance rior to the patella of the right distal femur. The patellar was
to the clinical use of BPs, as their effects on bone healing may moved slightly lateral to reveal the distal femoral condyles
be divergent when comparing acute use during bone repair and a 1.1-mm Kirschner wire was inserted into the medul-
with patients on long-term BP dosing schedules. Atypical fem- lary canal from the femoral condyles until it slightly pro-
oral fractures have been associated with long-term BP dosing truded proximal to the greater trochanter. A model of the
and represent spontaneous or low-energy fractures with poor Einhorn 3-point drop weight fracture apparatus was used to
subsequent bone healing [14, 15]. In osteogenesis imperfecta, create a clean transverse mid-diaphyseal closed femoral frac-
it has been suggested that delayed union can occur in a subset ture [18]. The MX20 digital Faxitron X-ray system (Faxitron
of children treated with Pamidronate (PAM) [16]. A recent X-ray Corporation, Illinois, USA), was used to visualize all
Cochrane review further highlights the need for additional fractures. Rats with fractures that were not mid-diaphyseal
studies to examine the safety and efficacy of PAM in osteo- or comminuted were euthanized and excluded (N = 1). For
genesis imperfecta [17]. Mechanistically, coupling between pain relief buprenorphine (Temgesic, Reckitt Benckiser) was
bone catabolism and anabolism suggests that prolonged anti- administered as a subcutaneous dose of 0.05 mg/kg post-
resorptive treatments can lead to a generalized depression of operatively and daily monitoring of rats was carried out
bone formation necessary for bone healing. This effect and throughout the course of the study. During the study, three
its medical impact can be challenging to explore in a clinical animals were culled due to worsening health and/or evidence
setting. Hence, we speculated that a rodent model of BP pre- of infection and were excluded from subsequent analysis.
dosing could provide important insights into its bearing on the Calcein was administered at a dose of 10 mg/kg subcuta-
fracture repair process. neously at 10 and 3 days prior to harvest to allow for analy-
Closed fractures were produced using an Einhorn appa- sis of dynamic bone formation. Harvest time points were at
ratus in rats pre-treated for 4 weeks with PAM administered either 2 or 6 weeks after fracture surgery to examine soft and
twice weekly at low (0.15 mg/kg), medium (0.5 mg/kg), and hard callus remodeling stages, respectively. Left and right
high (5 mg/kg) doses. It was hypothesized that (1) no PAM femora and tibiae were harvested and fixed in 10% formalin
dose would impair of endochondral ossification, (2) hard for 4 h at room temperature followed by 24 h at 4 °C, and
callus remodeling would be impaired by PAM in a dose- then stored in 70% ethanol at 4 °C.
dependent manner, and (3) pre-dosing with the higher doses
of PAM may result in a significant corresponding decrease Radiological Analysis
in new bone formation.
Longitudinal radiographs were performed using the digital
MX 20 Faxitron system on both right and left femora. The
Methods fractured femora were graded as bridged or not united at
6 weeks by a blinded observer to assess radiological fracture
Animals and PAM Administration union. Femoral length was measured on all intact limbs.
Quantitative Computerized Tomography (QCT) was per-
Seventy male Wistar rats were housed from the age of formed on both right and left femora of all samples using a
5 weeks and all procedures were approved by the Westmead Stratec xQCT research scanner (Norland Stratec, Durlacher,

13
Pretreatment with Pamidronate Decreases Bone Formation but Increases Callus Bone Volume in…

Germany). Right (fractured) femora underwent scans at a Resin-processed callus sections were Von Kossa stained
voxel-size of 0.07 mm and a scan speed of 5 s with 91-mm- to allow analysis of trabecular and cortical bone in the cal-
thick slices taken with a slice separation of 1 mm through lus. Bone volume within the primary trabecular regions was
the entire fracture callus. For analysis of the callus, cortical then determined and BV/TV of the primary trabecular callus
bone was manually excluded and bone selected at a consist- was calculated. Adjacent sections were cleared for fluores-
ent threshold for all specimens. Left (unfractured) femora cent analysis, which included quantification of the percent-
were scanned with the same voxel size and scan speed but age fluorescent labeled bone in the total fracture callus. This
only two slices were measured, with one slice placed 8 mm was determined using the total bone volume of each callus
from the distal femoral boundary, measuring metaphyseal as measured using adjacent Von Kossa-stained sections. The
bone, and the other placed in the mid-diaphyseal region. distance between double labeling in the trabecular bone of
Data from the diaphyseal slice of the left femora were multi- the callus was also measured in the 6W samples, generat-
plied by nine to extrapolate data for the comparison with the ing data on mineral apposition rate (MAR). At 2 weeks, the
contralateral limb. Data for Bone Mineral Content (BMC), ossifying callus is not mature enough to show clear double
Bone Volume (BV), Bone Mineral Density (BMD), and labels.
Moment of Inertia (MMI) were generated for each sample.
Cortical Thickness, Periosteal Circumference, and Endosteal Left (Contralateral) Femora Specimens
Circumference are reported for the diaphysis of intact legs.
Resin sections of the left femoral diaphyses were cleared
for analysis of MAR on both the periosteal and endosteal
Histology and Histomorphometry
surfaces of each sample. Mineralizing Surface was also
determined for both the periosteal and endosteal surfaces
Right and left femora were bisected into lateral and medial
after measuring the length of the single label surface (SLS),
halves. The lateral halves were decalcified along with the
double label surface (DLS), and bone surface. The formula
left tibia in 0.39M EDTA in 1.25% formalin (pH 8.0) at
[(DLS + SLS/2)/bone surface] was used.
room temperature (RT) for 3 days, with further decalcifica-
tion in 0.34M EDTA (pH 8.0) for 2 weeks at RT, before
Statistics
being processed and embedded in paraffin wax. The medial
halves of fractures and right tibia were processed through
For all data, means and standard deviations were calculated
graded acetones and embedded in methyl methacrylate resin
and a one-way analysis of variance (ANOVA) applied with
for un-decalcified histology. Sagittal sections of fractures
post hoc t tests by the least squares difference method (SPSS
and coronal sections of tibia, 5 µm thick, were produced for
Inc Chicago, IL). The cutoff for statistical significance was
each sample on a Leica RM2155 semi-automated microtome
p < 0.05.
(Leica, Germany).

Right (Fractured) Femora Specimens Results

Decalcified callus sections were stained with Alcian Blue/ Chronic PAM Dosing Does not Impair Early
Picrosirius Red for cartilaginous and ossified callus mor- Endochondral Bone Repair
phology, and adjacent sections were stained with Safranin O
and counterstained with 0.4% Light Green to analyze carti- At 2 weeks post fracture only high-dose PAM produced
lage. Adjacent sections were also stained for tartrate-resist- minor but significant differences in radiographic callus size
ant acid phosphatase (TRAP) expression, and counterstained compared to saline controls. In the high-dose PAM group,
with Hematoxylin, to highlight and analyze osteoclasts. QCT showed that callus BMC was increased by + 30%
Image capture and analysis were performed using BIO- (p < 0.01), callus bone volume by + 10% (p < 0.05), and
QUANT measure 32 Nova prime (Nashville, Texas) image callus BMD by + 10% (p < 0.01) (Table 1). However, this
analysis system connected to a QICAM Fast 1394 color 12 did not translate to a significant increase in callus moment
bit camera (Quantitative Imaging Corporation, Canada). of inertia.
Osteoclast number was counted within a controlled region Histological analysis was performed on the 2  week
of interest in the callus adjacent to the fracture site in sam- fracture specimens. The BV/TV of the ossified callus was
ples at 6 weeks post fracture only. The total bone surface significantly increased in all of the PAM treatment groups
within this region was also measured, as well as the bone (Fig. 1a). PAM treatment was associated with no change
surface in contact with osteoclasts, allowing calculation of in the % of bone containing fluorescent label in the callus
a percent osteoclast surface compared to total bone surface. (Fig. 1b); however, the average femur length was decreased

13
A. Morse et al.

with higher doses of PAM (Fig. 1c). At the 2 week time in soft callus area (21–28%, N.S.) between the saline con-
point, cartilaginous tissue was present in the soft callus for trols and the 3 PAM treatment groups.
all groups (Fig. 1d), and quantification showed no difference
Chronic PAM Dosing Increases Callus Size at 6 Weeks
but Reduces the Formation of New Bone
Table 1  QCT parameters in fractured femora
2W right femora 6W right femora Radiographic analysis by an orthopedic surgeon blinded
Mean SD Mean SD to treatment group confirmed that all fractures were radio-
graphically united by 6 weeks. The effects of PAM on cal-
Callus BMC
lus size and density were examined by QCT (Table 1). By
 Saline 128.53 8.24 140.04 16.34
6 weeks post fracture, callus BV and BMD were signifi-
 PAM 0.15 mg/kg 146.35 25.43 217.61* 37.56
cantly increased with all levels of PAM treatment over saline
 PAM 0.5 mg/kg 144.54 21.35 231.24* 41.54
by 46–79% and 55–94%, respectively (p < 0.01). Calcula-
 PAM 5 mg/kg 166.68* 30.86 272.22* 47.13
tions of moment of inertia values similarly showed increases
Callus BMD
with high dose PAM over saline of + 127% (p < 0.01).
 Saline 717.9 22.2 917.4 105.9
Histological analysis of the 6-week specimens showed
 PAM 0.15 mg/kg 738.8 61.4 956.6 45.8
ossified callus BV/TV was increased with PAM treat-
 PAM 0.5 mg/kg 729.1 57.1 1030.9* 67.3
ment (Fig. 2a), similar to the 2-week samples. However,
 PAM 5 mg/kg 790.8* 51.9 985.3 47.0
by 6 weeks, the fluorescent label in the callus was signifi-
Callus BV
cantly reduced in all PAM treatment groups, indicating a
 Saline 182.26 11.02 156.01 31.64
compensatory decease in bone anabolism by this time point
 PAM 0.15 mg/kg 200.05 28.53 229.72* 45.42
(Fig. 2b). Despite suppressed bone formation, histological
 PAM 0.5 mg/kg 200.82 25.48 228.42* 55.38
staining with Alcian blue/Picrosirius red showed no evi-
 PAM 5 mg/kg 214.3 42.01 279.34* 54.04
dence of disrupted soft callus removal and retention of
MMI
cartilage (Fig. 2d). Nevertheless, hard callus remodeling
 Saline 102.1 15.8 78.8 33.5
was clearly disrupted by PAM, particularly in the PAM 5
 PAM 0.15 mg/kg 121.7 34.4 124.9 47.6
group with retention of a larger less remodeled hard callus.
 PAM 0.5 mg/kg 124.5 32.6 120.5 58.3
A trend was seen for decreased femur length in PAM-treated
 PAM 5 mg/kg 133.2 06.3 177.1* 62.0
groups, however, only PAM 0.5 showed a significant reduc-
*p < 0.05 tion (Fig. 2c).

Fig. 1  2W histology. Measurements of a BV/TV, b callus fluorescence, and c femur length were quantified from tissue sections. *p < 0.05,
**p < 0.01. d Histological images of closed fractures stained with Alcian blue/Picrosirius red

13
Pretreatment with Pamidronate Decreases Bone Formation but Increases Callus Bone Volume in…

Fig. 2  6W histology. Measurements of a BV/TV, b callus fluorescence, and c femur length were quantified from tissue sections. *p < 0.05,
**p < 0.01. d Histological images of closed fractures stained with Alcian blue/Picrosirius red

More detailed bone histomorphometry was performed in all three PAM treatment groups compared to saline
for further cellular mechanisms. PAM at 0.25, 0.5, and 5.0 (p < 0.01), but cortical thickness was significantly increased
reduced the callus MAR at 6 weeks by 32%, 38%, and 49%, by 16–22%. This increased cortical thickness reflected
respectively (p < 0.01, Fig. 3a). Analysis of TRAP + osteo- decreases in endosteal circumference rather than increases
clast staining within an ROI of the callus showed increased in periosteal circumference. However, a statistically signifi-
osteoclast number with PAM treatment (Fig. 3b), which has cant increase in periosteal circumference was still seen in the
been previously reported for BPs [19]. However, measures high-dosed PAM group.
that consider the osteoclast surface show a proportional
decrease in osteoclasts lining the bone relative to osteoclast
number (Figs. 3c, d). Discussion

The Effect of PAM Treatment on Intact Bones This study examined a preclinical fracture model that fea-
tured bone pre-treated with increasing doses of PAM and
In order to appreciate the effects of chronic BP treatment on tested the effects on subsequent bone repair. Consistent with
bone healing, the contralateral femora and the tibiae were prior reports looking at concomitant BP dosing [6–8, 20],
measured radiographically. we saw no negative effect on bone union and soft callus
PAM treatment resulted in small changes in femur length removal. While to date a significant research has focus has
that were nonetheless statistically significant. At the study been placed on the utility of BPs to augment bone healing,
endpoint (10 weeks of dosing), femur length was decreased the effect of chronic BP treatment on fracture repair remains
by an average of 2% with medium-dosed PAM (p < 0.05) under-explored. In the context of the elderly osteoporotic
and 3% with high-dosed PAM (p < 0.01) compared to saline. patient, there are concerns that long-term BP treatment can
Growth plate height was assessed in all proximal tibiae and lead to atypical femoral fractures, potentially through a
revealed no changes with PAM treatment (data not shown). reduction in global bone anabolism and a capacity to repair
Radiographic (QCT) analysis performed on the contralat- microdamage. However, there are little data to support or
eral femora at the study endpoint showed a significant + 9% reject extending these concerns to a pediatric setting, which
increase in BMC with high-dose PAM compared to saline is relevant to patient populations at high risk of childhood
(p < 0.05) (Table 2). Bone volume was also increased by fracture who routinely receive off-label BPs.
+ 14% with low-dose PAM (p < 0.05) and 19% with high- The physiology of bone growth can be profoundly differ-
dose PAM compared to Saline (p < 0.01). Notably, cortical ent between children and adults. This may also lead to dif-
bone BMD was significantly decreased 5–6% by 6 weeks ferences in the localization and action of BPs as the bone to

13
A. Morse et al.

Fig. 3  6W callus histomor-


phometry. Measurements of a
MAR, b N.Oc, c Oc.S/N.Oc,
and d Oc.S/BS were quantified
from tissue sections. *p < 0.05,
**p < 0.01

which BPs bind becomes buried more rapidly in a pediatric hypotheses for the coupling of osteoclastic and osteoblast
setting. Currently, there are no robust data to measure and crosstalk [28], including paracrine signals from osteoclasts
describe how BP embedding affects its long-term retention as well as paracrine factors released from the bone by osteo-
in children. At a functional level, the long-term risks of BP clastic bone resorption. The observation of increased osteo-
use on growth, modeling, microdamage repair, fracture risk, clast numbers with nitrogen-containing BP treatment (simi-
and fracture healing are poorly described. It must neverthe- lar to previous reports [19]) supports the latter model. The
less be acknowledged that the best clinical evidence to date progressive suppression of bone turnover by anti-resorptives
shows that the context of osteogenesis imperfecta BPs do likely underlies conditions such as atypical femoral fractures
not significantly impair growth [21, 22], increase the risk of that are sporadically seen with long-term BP treatment. This
atypical type fractures [23], or impair fracture repair [24]. has been attributed to a decreased capacity to repair micro-
Yet, the focus of previous studies testing extended BP damage, leading to stress fractures and low-energy fractures
treatment has been in an adult osteoporosis setting. In ova- [13]. A recent study has suggested that the subsequently
riectomized rats treated with alendronate following femoral impaired bone repair may be improved by PTH or teripara-
shaft fractures, investigators reported increased callus size tide treatment [29]. Teriparatide has also been suggested to
and density, but also a decreased mineral apposition rate at improve cortical bone formation and turnover in patients
6 and 16 weeks post fracture [25]. A corresponding clinical on long-term alendronate therapy in a clinical setting [30].
report indicated that osteoporotic fracture patients treated There are a number of caveats associated with the model
with alendronate post-operatively showed no significant design and analysis that leave scope for future research.
worsening or improvement with treatment [26]. Other anti- Firstly, the length of pretreatment was not specifically
resorptive agents have shown similar outcomes in rodent conceived to model the potentially years of BP treatment
models, such as anti-RANKL antibody (denosumab), which that individuals with osteoporosis or OI may be exposed to
produces larger callus sizes after fracture [27]. before developing a poorly healing fracture. Secondly, when
Our model shows local reductions in callus MAR with interpreting the data of this study and attempting to apply it
PAM treatment; however, small but significant reductions to clinical situations, it must be acknowledged that this study
in bone formation were also seen in contralateral intact did not feature models of osteoporosis or OI. Future studies
limbs with high-dose PAM (data not shown). The mecha- could attempt these experiments in ovariectomized rats or
nism underlying the corresponding decrease in bone forma- rodent models of osteogenesis imperfecta. Thirdly, there are
tion with BPs remains unclear. There are currently multiple practical limitations with the specific QCT calculations: BP

13
Pretreatment with Pamidronate Decreases Bone Formation but Increases Callus Bone Volume in…

Table 2  QCT parameters in intact femora did lead to decreased bone turnover resulting in significant
2W left femora 6W left femora
increases in callus size and predicted strength. The effects
of PAM were dose-dependent, although all doses did have
Mean SD Mean SD
effects on new bone formation as well as resorption. These
BMC data support further investigation into the long-term dosing
 Saline 44.35 2.87 53.97 2.37 of BP, with a particular focus on their effects on bone turno-
 PAM 0.15 mg/kg 46.22 1.42 57.57 3.95 ver and in clinically relevant models, such as murine models
 PAM 0.5 mg/kg 45.85 2.15 55.96 3.76 of osteogenesis imperfecta.
 PAM 5 mg/kg 45.39 1.91 59.04 5.64
BMD
 Saline 1163.7 15.1 1227.0 10.6 Compliance with Ethical Standards 
 PAM 0.15 mg/kg 1158.9 18.8 1163.3 32.3
 PAM 0.5 mg/kg 1153.4 23.4 1168.6 16.8 Conflict of interest Dr. Little and Dr Schindeler report grants and
non-financial support from Novartis Pharma, grants from N8 Medical,
 PAM 5 mg/kg 1133.8 13.0 1151.6 29.1 grants from Celgene, grants and nonfinancial support from Amgen Inc.
Cortical BV outside the submitted work.
 Saline 38.07 2.33 43.95 1.84
 PAM 0.15 mg/kg 39.83 1.29 49.91 4.57 Human and Animal Rights and Informed Consent  All procedures
performed in studies involving human participants were in accord-
 PAM 0.5 mg/kg 39.81 2.57 48.35 3.75 ance with the ethical standards of the institutional research committee
 PAM 5 mg/kg 40.13 1.87 52.35 6.56 (Westmead Hospital Animal Ethics Committee, #5024.06-07) and with
Periosteal Cir. the 1964 Helsinki declaration and its later amendments or comparable
 Saline 11.01 0.28 11.76 0.30 ethical standards.
 PAM 0.15 mg/kg 11.19 0.26 12.10 0.52
 PAM 0.5 mg/kg 11.03 0.42 11.90 0.35
 PAM 5 mg/kg 11.14 0.24 12.30* 0.61
Endosteal Cir.
References
 Saline 4.96 0.20 5.23 0.44
1. Maraka S, Kennel KA (2015) Bisphosphonates for the prevention
 PAM 0.15 mg/kg 4.88 0.33 4.39* 0.74 and treatment of osteoporosis. BMJ 351:h3783
 PAM 0.5 mg/kg 4.56* 0.36 4.33* 0.46 2. Baroncelli GI, Bertelloni S (2014) The use of bisphosphonates in
 PAM 5 mg/kg 4.74 0.41 4.31* 0.67 pediatrics. Horm Res Paediatr 82(5):290–302
3. Little DG, Ramachandran M, Schindeler A (2007) The anabolic
Cortical thickness
and catabolic responses in bone repair. J Bone Joint Surg Br
 Saline 0.96 0.05 1.04 0.05 89(4):425–433
 PAM 0.15 mg/kg 1.00 0.04 1.23* 0.12 4. Schindeler A, McDonald MM, Bokko P, Little DG (2008) Bone
 PAM 0.5 mg/kg 1.03 0.03 1.21* 0.10 remodeling during fracture repair: the cellular picture. Sem Cell
Dev Biol 19(5):459–466
 PAM 5 mg/kg 1.02* 0.05 1.27* 0.16
5. Chambers TJ, Fuller K (2011) How are osteoclasts induced to
MMI resorb bone? Ann N Y Acad Sci 1240:1–6
 Saline 17.6 1.8 22.3 2.2 6. McDonald MM, Morse A, Mikulec K, Peacock L, Baldock PA,
 PAM 0.15 mg/kg 18.9 1.5 25.7 3.7 Kostenuik PJ, Little DG (2013) Matrix metalloproteinase-driven
endochondral fracture union proceeds independently of osteoclast
 PAM 0.5 mg/kg 18.2 2.7 24.1 2.7
activity. J Bone Miner Res 28(7):1550–1560
 PAM 5 mg/kg 18.7 1.5 28.2* 5.7 7. Hao Y, Wang X, Wang L, Lu Y, Mao Z, Ge S, Dai K (2015)
Zoledronic acid suppresses callus remodeling but enhances callus
*p < 0.05 strength in an osteoporotic rat model of fracture healing. Bone
81:702–711
8. Menzdorf L, Weuster M, Klüter T, Brüggemann S, Behrendt P,
treatment may lead to subtle changes in cortical porosity that Fitchen-Oestern S, Varoga D, Seekamp A, Purcz N, Glueer CC,
may alter the thresholds used to designate “bone,” which Pufe T, Lippross S (2016) Local pamidronate influences fracture
would affect measures of bone volume and BMD, although healing in a rodent femur fracture model: an experimental study.
BMC Musculoskelet Disord 17:255
not BMC. Additionally, MMI calculations in the high-dose
9. Takuma S (1962) Electron microscopy of cartilage resorption by
BP group be affected by the shortened bone length, but the chondroclasts. J Dent Res 41:883–889
substantive changes in moment of inertia are best explained 10. Knowles HJ, Moskovsky L, Thompson MS, Grunhen J, Cheng
by the substantive changes in cortical thickness rather than X, Kashima TG, Athanasou NA (2012) Chondroclasts are mature
osteoclasts which are capable of cartilage matrix resorption. Vir-
the relatively subtle changes in length.
chows Arch 461(2):205–210
In conclusion, this study confirms many of our original 11. Colnot C, Thompson Z, Miclau T, Werb Z, Helms JA (2003)
hypotheses. PAM pretreatment, even at doses far higher than Altered fracture repair in the absence of MMP9. Development
in clinical use, did not impair endochondral ossification but 130(17):4123–4133

13
A. Morse et al.

12. Behonick DJ, Xing Z, Lieu S et al (2007) Role of matrix metal- relationship with bisphosphonate treatment. J Bone Miner Res
loproteinase 13 in both endochondral and intramembranous ossi- 32(5):1034–1039
fication during skeletal regeneration. PLoS ONE 2(11):e1150 24. Munns CF, Rauch F, Zeitlin L, Fassier F, Glorieux FH (2004)
13. Kidd LJ, Cowling NR, Wu AC, Kelly WL, Forwood MR (2011) Delayed osteotomy but not fracture healing in pediatric osteogen-
Bisphosphonate treatment delays stress fracture remodeling in the esis imperfecta patients receiving pamidronate. J Bone Miner Res
rat ulna. J Orthop Res 29(12):1827–1833 19(11):1779–1786
14. Shane E, Burr D, Ebeling PR, Abrahamsen B, Adler RA, Brown 25. Fu LJ, Tang TT, Hao YQ, Dai KR (2013) Long-term effects of
TD et al (2010) Atypical subtrochanteric and diaphyseal femoral alendronate on fracture healing and bone remodeling of femoral
fractures: report of a task force of the American Society for Bone shaft in ovariectomized rats. Acta Pharmacol Sin 34(3):387–392
and Mineral research. J Bone Miner Res 25:2267–2294 26. Seo JB, Yoo JS, Ryu JW, Yu KW (2016) Influence of early bis-
15. Shane E, Burr D, Abrahamsen B, Adler RA, Brown TD, Cheung phosphonate administration for fracture healing in patients with
AM et al (2014) Atypical subtrochanteric and diaphyseal femoral osteoporotic proximal humerus fractures. Clin Orthop Surg
fractures: second report of a task force of the American Society 8(4):437–443
for Bone and Mineral research. J Bone Miner Res 29:1–23 27. Gerstenfeld LC, Sacks DJ, Pelis M, Mason ZD, Graves DT, Bar-
16. Alharbi M, Pinto G, Finidori G, Souberbielle JC, Guillou F, rero M, Ominsky MS, Kostenuik PJ, Morgan EF, Einhorn TA
Gaubicher S, Le Merrer M, Polak M (2009) Pamidronate treat- (2009) Comparison of effects of the bisphosphonate alendronate
ment of children with moderate-to-severe osteogenesis imperfecta: versus the RANKL inhibitor denosumab on murine fracture heal-
a note of caution. Horm Res 71(1):38–44 ing. J Bone Miner Res 24(2):196–208
17. Dwan K, Phillipi CA, Steiner RD, Basel D (2016) Bisphosphonate 28. Sims NA, Martin TJ (2015) Coupling signals between the osteo-
therapy for osteogenesis imperfecta. Cochrane Database Syst Rev. clast and osteoblast: how are messages transmitted between these
https​://doi.org/10.1002/14651​858.CD005​088.pub4 temporary visitors to the bone surface? Front Endocrinol (Laus-
18. Bonnarens F, Einhorn TA (1984) Production of a standard closed anne) 6:41
fracture in laboratory animal bone. J Orthop Res 2(1):97–101 29. Bakr MM, Kelly WL, Brunt AR, Paterson BC, Massa HM, Mor-
19. Schindeler A, Ramachandran M, Godfrey C, Morse A, McDonald rison NA, Forwood MR (2019) Single injection of PTH improves
M, Mikulec K, Little DG (2008) Modeling bone morphogenetic osteoclastic parameters of remodeling at a stress fracture site in
protein and bisphosphonate combination therapy in wild-type and rats. J Orthop Res 37(5):1172–1182
Nf1 haploinsufficient mice. J Orthop Res 26(1):65–74 30. Ma YL, Zeng QQ, Chiang AY, Burr D, Li J, Dobnig H, Fahrleit-
20. Amanat N, McDonald M, Godfrey C, Bilston L, Little D (2007) ner-Pammer A, Michalská D, Marin F, Pavo I, Stepan JJ (2014)
Optimal timing of a single dose of zoledronic acid to increase Effects of teriparatide on cortical histomorphometric variables in
strength in rat fracture repair. J Bone Miner Res 22(6):867–876 postmenopausal women with or without prior alendronate treat-
21. Munns CF, Rauch F, Travers R, Glorieux FH (2005) Effects of ment. Bone 59:139–147
intravenous pamidronate treatment in infants with osteogenesis
imperfecta: clinical and histomorphometric outcome. J Bone Publisher’s Note Springer Nature remains neutral with regard to
Miner Res 20(7):1235–1243 jurisdictional claims in published maps and institutional affiliations.
22. Biggin A, Munns CF (2014) Osteogenesis imperfecta: diagnosis
and treatment. Curr Osteoporos Rep 12(3):279–288
23. Trejo P, Fassier F, Glorieux FH, Rauch F (2017) diaphyseal
femur fractures in osteogenesis imperfecta: characteristics and

13

You might also like