You are on page 1of 11

ORIGINAL ARTICLE

Differential Effects of Teriparatide and Denosumab


on Intact PTH and Bone Formation Indices: AVA
Osteoporosis Study

David W. Dempster, Hua Zhou, Robert R. Recker, Jacques P. Brown,


Christopher P. Recknor, E. Michael Lewiecki, Paul D. Miller, Sudhaker D. Rao,
David L. Kendler, Robert Lindsay, John H. Krege, Jahangir Alam,

Downloaded from https://academic.oup.com/jcem/article/101/4/1353/2804498 by guest on 10 February 2021


Kathleen A. Taylor, Boris Janos, and Valerie A. Ruff*
Context: Denosumab-induced PTH elevation may stimulate early bone formation.

Objective: Our objective was to evaluate whether denosumab-induced changes of intact PTH
(iPTH) result in early anabolic effects according to histomorphometry and bone turnover markers
(BTMs) compared with teriparatide, an established anabolic agent.

Design: This open-label, randomized study used quadruple labeling to label bone before/after
treatment, with a transiliac bone biopsy at 3 months.

Setting: This study took both in both US and Canadian sites.

Participants: Sixty-nine postmenopausal women with osteoporosis were included.

Interventions: Teriparatide (20 ␮g/day) for 6 months and denosumab (60 mg once) were used in
this study.

Main Outcome Measure: Between-treatment comparison of change from baseline to month 3 in


cancellous mineralizing surface/bone surface, histomorphometric indices in four bone envelopes,
and BTM and iPTH at baseline, 1, 3, and 6 months was undertaken.

Results: After denosumab, iPTH peaked at month 1 (P ⬍ .001), then declined, remaining above baseline
through month 6 (P ⱕ .01); after teriparatide, iPTH declined at all time points (P ⬍ .001). From baseline
to month 3, cancellous mineralizing surface/bone surface increased with teriparatide and decreased
with denosumab and at month 3, was higher with teriparatide. Similar results were observed in other
bone envelopes. BTMs increased from baseline in teriparatide-treated subjects (procollagen type 1
N-terminal propeptide at month 1 and carboxyterminal cross-linking telopeptide of type 1 collagen at
month 3); procollagen type 1 N-terminal propeptide and carboxyterminal cross-linking telopeptide of
type 1 collagen decreased from baseline at all time points in denosumab-treated subjects.

Conclusions: Denosumab treatment increased iPTH but inhibited bone formation indices. In con-
trast, teriparatide treatment decreased iPTH but stimulated bone formation indices. These findings
are not consistent with the hypothesis of early indirect anabolic effect with denosumab. (J Clin
Endocrinol Metab 101: 1353–1363, 2016)

steoporosis drugs can be grouped into either anabolic analog [1–34]), is currently the only US Food and Drug
O or anticatabolic (antiresorptive) classes of bone-ac-
tive agents (1). Teriparatide (recombinant human PTH
Administration–approved agent in the anabolic class (1,
2). The anticatabolic/antiresorptive drug class consists of

ISSN Print 0021-972X ISSN Online 1945-7197 *Author affiliations listed at the bottom of the next page.
Printed in USA Abbreviations: BFR, bone formation rate; BMD, bone mineral density; BS, bone surface;
Copyright © 2016 by the Endocrine Society BTM, bone turnover marker; BV, bone volume; CTX, carboxyterminal cross-linking telo-
Received December 8, 2015. Accepted February 1, 2016. peptide of type 1 collagen; ES, eroded surface; FAS, full analysis set; iPTH, intact PTH; MAR,
First Published Online February 10, 2016 mineral apposition rate; MOA, mechanism of action; MS, mineralizing surface; P1NP,
procollagen type 1 N-terminal propeptide; TEAE, treatment-emergent adverse event;
W.Th, wall thickness.

doi: 10.1210/jc.2015-4181 J Clin Endocrinol Metab, April 2016, 101(4):1353–1363 press.endocrine.org/journal/jcem 1353
1354 Dempster et al Teriparatide or Denosumab Bone Effects J Clin Endocrinol Metab, April 2016, 101(4):1353–1363

bisphosphonates, estrogens, selective estrogen receptor lism; history of malignancy in the previous 5 years; signifi-
modulators, calcitonin, and denosumab (3–5). cantly impaired hepatic or renal function; treatment with sys-
temic glucocorticoids (ⱖ5 mg/day prednisone or equivalent)
Although denosumab has been characterized as an an-
in the previous 6 months; and prior or current use of teripa-
tiresorptive agent by histomorphometric and bone turn- ratide, PTH, PTH analog, or denosumab or any IV bisphos-
over marker (BTM) assessments (6, 7), some effects of this phonate. Oral bisphosphonate use required a wash-out period
drug have led to speculation that denosumab may have before screening (6 months off for use ⬎2 weeks but ⱕ2
distinctive properties with respect to its mechanism of ac- months; 1 year off for use ⬎2 months but ⱕ1 year; 2 years off
tion (MOA). Studies have reported that intact PTH (iPTH) for use ⬎1 year).
levels are increased in the 1–3 months after denosumab
Study design and treatment
administration (8 –13). It has been suggested that eleva-
This was an open-label, randomized phase IV study of 6
tion of PTH may result in indirect stimulation of bone months’ duration comparing teriparatide (20 ␮g/d) with deno-

Downloaded from https://academic.oup.com/jcem/article/101/4/1353/2804498 by guest on 10 February 2021


formation during a time of inhibited osteoclast activity sumab (60 mg once) in postmenopausal women with osteopo-
(12, 14) and in combination with denosumab-induced re- rosis. The primary endpoint was comparison of change from
ductions in cortical porosity (7, 12, 14, 15), may poten- baseline to 3 months in mineralizing surface/bone surface (MS/
BS) in the cancellous envelope of transiliac bone biopsies. Biop-
tially play a role in continuous long-term bone mineral
sies were taken at 3 months because iPTH is known to be elevated
density (BMD) gains (16) reported with denosumab. at this time point after treatment with denosumab (8 –13). The
The study objectives were to characterize denosumab- iPTH increase may be maximal with the first dose of denosumab
induced changes in iPTH levels in conjunction with bone (11–13); therefore, any anabolic properties that are observable
histomorphometric indices and BTMs and compare these by histomorphometric analyses should be apparent by 3 months
effects with those induced by teriparatide, an anabolic after the first dose. The effect of both teriparatide and deno-
sumab on remodeling also is apparent by this time based on
agent. To achieve these goals, this study used quadruple- biochemical marker data (11, 19). Finally, with quadruple-la-
labeled histomorphometry (17) of transiliac biopsies ob- beled histomorphometry, the possibility was considered that
tained from postmenopausal women with osteoporosis findings in subjects treated with teriparatide might not be reliable
treated with either denosumab or teriparatide. This is the beyond 3 months because some of the initial labels could have
first study to use quadruple-labeled histomorphometry been lost with remodeling. It was thought that loss of the first set
of labels by this mechanism should not be a factor if the biopsy
with an antiresorptive agent and to compare directly the
was obtained at 3 months; however, in a study using quadruple
MOAs of an antiresorptive agent and an anabolic agent. labeling, the first set of labels was demonstrated to be intact after
Quadruple labeling allowed both baseline and posttreat- 3 months of PTH(1– 84) treatment (20). MS/BS was chosen as
ment information to be obtained from a single biopsy from the primary endpoint because it is a robust variable that readily
each subject. distinguishes between anabolic and antiresorptive MOAs (19).
Other objectives were to characterize changes in: 1) serum iPTH;
2) a panel of histomorphometric indices in 4 bone envelopes
(cancellous, endocortical, intracortical, periosteal), and 3) BTMs
Subjects and Methods following denosumab or teriparatide treatment, comparing
treatment effects.
Subjects To assure a balance in bone turnover between the treatment
Ambulatory postmenopausal women with osteoporosis, groups at baseline, subjects were stratified based on their value
aged 55– 89 years, were included if their BMD T-score was at of nonfasting procollagen type 1 N-terminal propeptide (P1NP)
least 2.5 at the lumbar spine, femoral neck, or total hip; or less at screening: less than 30 ng/ml, at least 30 ng/ml to less than 50
than 1.5 with a prevalent vertebral or nonvertebral fragility frac- ng/ml, or more than 50 ng/ml. Using a computer-generated ran-
ture. Subjects had laboratory values within reference ranges for dom sequence generated by an interactive voice-response system,
serum calcium, PTH, and alkaline phosphatase. subjects were randomly assigned (1:1) to either teriparatide or
Subjects were excluded if they had any condition specified denosumab treatment groups based on their stratified baseline
in the teriparatide boxed warning or any treatment contrain- P1NP value. Subjects received elemental calcium (approximately
dications for teriparatide or denosumab (2, 18). Exclusion 1000 mg/d) and vitamin D3 or equivalent (800 –1200 IU/day)
criteria included 25-hydroxyvitamin D concentration of less throughout the study.
than 10 ng/ml; treatment with drugs or any active or suspected The study received ethical review board approval at all sites,
disease, except osteoporosis, known to affect bone metabo- and subjects gave informed written consent. The study was con-

Regional Bone Center (D.W.D., H.Z., R.L.), Helen Hayes Hospital, West Haverstraw, New York; Department of Pathology and Cell Biology (D.W.D.), College of Physicians and Surgeons
of Columbia University, New York; Department of Medicine (R.R.R.), Division of Endocrinology, School of Medicine, Creighton University, Omaha, Nebraska; Rheumatology and Bone
Diseases Research Group (J.P.B.), CHU de Quebec (CHUL) Research Centre and Department of Medicine, Laval University, Quebec City, Quebec, Canada; United Osteoporosis Centers
(C.P.R.), Gainesville, Georgia; New Mexico Clinical Research & Osteoporosis Center (E.M.L.), Albuquerque, New Mexico; Department of Medicine (P.D.M.), Colorado Center for Bone
Research, Lakewood, Colorado; Bone & Mineral Research Laboratory (S.D.R.), Henry Ford Hospital, Detroit, Michigan; Department of Medicine (Endocrinology) (D.L.K.), University of British
Columbia, Vancouver, British Columbia, Canada; Department of Medicine (R.L.), College of Physicians and Surgeons of Columbia University, New York; Lilly Research Laboratories (J.H.K.,
J.A.), Eli Lilly and Company, Indianapolis, Indiana; Musculoskeletal and Men’s Health (K.A.T., V.A.R.), Lilly USA, LLC, Indianapolis, Indiana; Research and Development – Bio-Medicines
(B.J.), Eli Lilly Canada Inc., Toronto, Ontario, Canada
doi: 10.1210/jc.2015-4181 press.endocrine.org/journal/jcem 1355

ducted in accordance with the Declaration of Helsinki and ap- the exception of osteoid volume/BV, which was assessed in only
plicable laws and guidelines. the cancellous envelope.

Assessments Biochemical markers of bone turnover and intact


PTH
Bone biopsy procedure
Blood (fasting) was collected at baseline and months 1, 3, and
Quadruple labeling using the fluorochromes, demeclocy-
6 for determination of serum BTM (P1NP and carboxyterminal
cline, and tetracycline was performed (17). Beginning 18 –23
cross-linking telopeptide of type 1 collagen [CTX]) and iPTH.
days before randomization, subjects took 150 mg demeclocy-
cline 4 times daily following a 3:12:3 day sequence (baseline
Safety
labeling); (Supplemental Appendix 1). One to 5 days after the last
demeclocycline dose, subjects were randomized to either deno- Clinical chemistry and hematology measurements were per-
formed at the screening visit and vital signs were collected at each
sumab or teriparatide. Beginning 22–25 days before the bone

Downloaded from https://academic.oup.com/jcem/article/101/4/1353/2804498 by guest on 10 February 2021


study visit. Treatment-emergent adverse events (TEAEs) and se-
biopsy, subjects took 250 mg tetracycline (posttreatment label-
rious adverse events were recorded at each study visit and at early
ing) following the same dosing schedule as demeclocycline. The
discontinuation, if applicable.
biopsy was performed 5– 8 days following the last tetracycline
dose.
Transiliac bone biopsies were performed using a Rochester Sample size
needle or similar large-bore (6 – 8 mm) manual trephine. Sample Twenty completers per group with adequate biopsies would
preparation and histomorphometric analyses were performed as provide approximately 80% power to detect a significant dif-
described (19, 21). The central histomorphometry reader was ference with a two-sided alpha level of 0.05 using a two-sample
blinded to treatment assignment and all indices were measured, t test. Assuming a 40% dropout or unevaluable biopsy rate, the
calculated, and expressed following American Society for Bone total number of randomized subjects needed was 68.
and Mineral Research Histomorphometric Nomenclature Com-
mittee recommendations (22). Statistical analyses
All prespecified efficacy and safety analyses were performed
Histomorphometry on the full analysis set (FAS) using a modified intent-to-treat
Demeclocycline can overestimate the length of fluorochrome principle. The FAS included data from all randomized subjects
labels relative to tetracycline (23, 24). This artifactual difference receiving at least one dose of study drug according to the actual
can potentially affect interpretation of data from indices derived received treatment. Tests of treatment effects were conducted at
from label length, such as MS/BS, which could be overestimated. a two-sided alpha level of 0.05. No adjustment for multiplicity
To assess treatment effects accurately and minimize label length was made for primary and secondary analyses. Demographics
artifacts, demeclocycline and tetracycline were administered for and baseline characteristics were summarized by treatment
group. Categorical variables were summarized by frequencies
the first and second labeling periods, respectively (23). The
and percentages. Continuous variables were summarized by
length of the tetracycline labels in both the teriparatide and de-
means and standard deviations. Categorical variables were an-
nosumab treatment groups was multiplied by a correction factor
alyzed using Fisher’s exact test, and continuous variables were
(23) of 1.34, which was empirically determined by the histomor-
analyzed using t test. Efficacy variables were analyzed using an
phometry laboratory assessing the biopsies.
analysis of covariance model, with treatment group as the main
For biopsy samples missing labels, the value of MS/BS was
effect and baseline measurement as a covariate. Paired t tests
zero. Mineral apposition rate (MAR) was reported as missing,
were used to assess changes from baseline within each treatment
and MAR-derived indices, such as bone formation rate, were
group. If the normality assumption of efficacy variable was not
assigned a value of zero. For biopsies with a single label only,
satisfied at the 0.01 significance level and if visual inspection of
MAR and MAR-derived indices were evaluated by two methods:
the data deemed it necessary, an appropriate nonparametric test,
1) assigned an imputed value of 0.3 ␮m/day or 2) counted as
such as the Wilcoxon rank-sum test, was performed to assess
missing (25). Results from the imputation method only are re-
treatment differences; the Wilcoxon signed-rank test was used to
ported here as results from the 2 methods were similar.
assess changes from baseline within each treatment group.
Baseline and 3-month data were obtained for MS/BS, bone
formation rate (BFR)/bone surface (BS), MAR, single-label sur-
face/BS, and double-label surface/BS in all four bone envelopes
defined as described (26). Only 3-month data were obtained for Results
BFR/bone volume (BV) (cancellous envelope only), activation
frequency, active formation period, adjusted apposition rate, Baseline demographics and characteristics
mineralization lag time, osteoid maturation time, and total for- Overall, 109 subjects were included from the United
mation period (cancellous, endocortical, and intracortical enve- States and Canada; of these, 69 subjects were random-
lopes). Data were only available at month 3 because they require ized beginning in February 2013 (33 teriparatide; 36
static components in their derivation, which cannot be separately
denosumab) (Supplemental Appendix 2). Thirty teripa-
assessed at baseline. Static indices measured were osteoid sur-
face/BS, osteoid volume/BV, osteoid thickness, wall thickness ratide-treated and 34 denosumab-treated subjects com-
(W.Th), and eroded surface (ES)/BS. These indices were assessed pleted the study. The last subject completed the study in
in the cancellous, endocortical, and intracortical envelopes with June 2014. Reasons for discontinuation in the teripa-
1356 Dempster et al Teriparatide or Denosumab Bone Effects J Clin Endocrinol Metab, April 2016, 101(4):1353–1363

Table 1. Baseline Characteristics


Teriparatide n ⴝ 33 Denosumab n ⴝ 36
Age, y (mean [SD]) 61.6 (5.8) 65.2 (8.3)a
Race, n (%)
Asian 0 (0) 2 (5.6)
Black or African American 1 (3.0) 2 (5.6)
Multiple 1 (3.0) 1 (2.8)
White 31 (93.9) 31 (86.1)
Body mass index, kg/m2 (mean [SD]) 24.7 (4.6) 24.0 (3.5)
Previous osteoporosis therapy, n (% yes) 11 (33.3) 14 (38.9)
Prevalent clinical fracture, n (% yes) 17 (51.5) 16 (44.4)
Lumbar spine T-score, mean (SD) ⫺2.58 (0.91) ⫺2.45 (0.93)
⫺2.24 (0.92) ⫺2.39 (0.63)

Downloaded from https://academic.oup.com/jcem/article/101/4/1353/2804498 by guest on 10 February 2021


Femoral neck T-score, mean (SD)
Total hip T-score, mean (SD) ⫺1.68 (0.93) ⫺1.92 (0.76)
Serum P1NP strata, n (%)
⬍30 ng/ml 0 (0) 2 (5.6)
ⱖ30 ng/ml to ⱕ50 ng/ml 13 (39.4) 14 (38.9)
⬎50 ng/ml 20 (60.6) 20 (55.6)
25-hydroxyvitamin D (mean [SE]), ng/mlb 35.5 (2.5) (n ⫽ 32) 38.0 (3.1) (n ⫽ 35)
iPTH (mean [SE]), pg/mlc 34.8 (2.4) (n ⫽ 33) 33.7 (2.4) (n ⫽ 34)
a
P ⬍ .05 using Fisher’s exact test for categorical variables and using t test for continuous variables.
b
Reference range: ⱖ29 ng/ml for ⬎18 y of age.
c
Reference range: 14 –72 pg/ml for ⬎18 y of age.

ratide group were adverse event (n ⫽ 1) and sponsor Of the five significant protocol violations (one each in
decision (n ⫽ 2), and in the denosumab group, subject three teriparatide-treated subjects; one denosumab-
decision (n ⫽ 2). Biopsies were collected from 66 sub- treated subject with two violations), all involving viola-
jects (31 teriparatide; 35 denosumab); all biopsies were tions of inclusion/exclusion criteria, one subject was re-
evaluable for all indices with the exception of one bi- moved from the study. Analyses excluding these subjects
opsy with a missing cortex. were performed for the primary endpoint; inclusion of
Baseline characteristics between the two treatment these subjects in the FAS did not significantly change the
groups were similar with the exception of mean age, which result or conclusions.
was higher in the denosumab group than in the teripa-
ratide group (65.2 years vs 61.6 years; P ⫽ .041) (Table 1); iPTH and BTMs
this difference did not affect any treatment outcomes (data In the denosumab group, iPTH was significantly in-
not shown). The groups were balanced with respect to creased from baseline (89.6 mean percent change; P ⬍
P1NP stratification. .001) after 1 month of treatment (mean absolute values,

A B

Figure 1. Changes in serum bone turnover markers and intact parathyroid hormone. (A) Denosumab; (B) teriparatide. Mean percent change ⫾ SE
is shown. †P ⬍ .001 for within treatment group comparison from baseline to each time point. *P ⫽ .01 for within treatment group comparison
from baseline to each time point using t test.
doi: 10.1210/jc.2015-4181 press.endocrine.org/journal/jcem 1357

baseline: 38.1 pg/ml; month 1: 63.5 pg/ml; reference (5.22%) and denosumab (4.78%) (Figure 2A, Table 2). At
range: 14 –72 pg/ml) (Figure 1A). Thereafter, iPTH levels month 3, median MS/BS was significantly higher in the
declined but were still significantly greater than baseline at teriparatide group (18.73%) than in the denosumab group
month 3 (68.0%, 51.5 pg/ml) and at month 6 (31.9%, (0.96%) (P ⬍ .001). From baseline to month 3, MS/BS in-
47.9 pg/ml). However, despite the elevated iPTH levels, creased with teriparatide treatment (P ⬍ .001) and decreased
both P1NP and CTX levels were significantly reduced
with denosumab treatment (P ⬍ .001) (median of change,
(P ⬍ .001 vs baseline at all time points). In contrast, after
⫹12.4% vs ⫺2.5%; P ⬍ .001 for between-group compari-
1 month of teriparatide treatment, there was a significant
reduction from baseline (⫺27.1 mean percent change; P ⬍ son), indicating increased extent of new bone formation with
.001) in iPTH levels (mean absolute values, baseline: 38.6 teriparatide and decreased extent with denosumab. Similar
pg/ml; month 1: 27.1 pg/ml), continuing through month 6 results for MS/BS were observed in the endocortical, intra-
(month 3: ⫺22.4%, 25.1 pg/ml; month 6: ⫺35.3%, 22.2

Downloaded from https://academic.oup.com/jcem/article/101/4/1353/2804498 by guest on 10 February 2021


cortical, and periosteal envelopes (Figure 2, B–D).
pg/ml) (Figure 1B). However, both P1NP and CTX levels Results for supporting histomorphometric indices for
increased significantly starting at 1 and 3 months, respec- cancellous and other envelopes are shown in Tables 2 and
tively, and rose through 6 months with P1NP approxi- 3. In all bone envelopes, the median month 3 values for
mately 400% above baseline at 6 months. most dynamic and static indices of bone formation were
Bone histomorphometry significantly different between treatment groups and were
At baseline, in the cancellous envelope, median MS/BS greater for teriparatide vs denosumab. The static index of
values were not significantly different between teriparatide bone resorption, ES/BS, was significantly lower in the de-

A B

C D

Figure 2. Changes in mineralizing surface. Median and interquartile range (Q1, Q3) are shown for the four bone envelopes at baseline and 3
months: (A) cancellous; (B) endocortical; (C) intracortical; and (D) periosteal. For samples with no label, MS/BS was assigned a value of zero. *P ⬍
.001 for between treatment group comparison at baseline or 3 months in each envelope using Wilcoxon rank-sum test. †P ⬍ .001 for within
treatment group comparison from baseline to 3 months in each envelope using Wilcoxon signed-rank test. ¥P ⬍ .01 for within treatment group
comparison from baseline to 3 months in each envelope using Wilcoxon signed-rank test.
1358 Dempster et al Teriparatide or Denosumab Bone Effects J Clin Endocrinol Metab, April 2016, 101(4):1353–1363

Table 2. Histomorphometric Indices (Baseline to Month 3)a,b


Cancellous Endocorticalc

Indices, Baseline Month 3 Baseline Month 3


Median
(Q1, Q3) TPTD DMAb TPTD DMAb TPTD DMAb TPTD DMAb
MS/BS, %d 5.22 4.78 18.73e 0.96e,f 8.73 8.54 39.50e 5.42f,g
(2.63, 7.39) (2.68, 6.19) (11.13, 26.64) (0.44, 1.93) (5.90, 15.64) (5.62, 12.44) (23.92, 60.39) (2.63, 10.15)
n ⫽ 31 n ⫽ 35 n ⫽ 31 n ⫽ 35 n ⫽ 30 n ⫽ 35 n ⫽ 30 n ⫽ 35
BFR/BS, 0.0096 0.0091 0.0366e 0.0014e,f 0.0201 0.0169 0.0889e 0.0096e,f
mm3/mm2/yh (0.0051, 0.0148) (0.0048, 0.0115) (0.0226, 0.0543) (0.0005, 0.0033) (0.0096, 0.0353) (0.0092, 0.0241) (0.0445, 0.1501) (0.0029, 0.0193)
n ⫽ 31 n ⫽ 35 n ⫽ 31 n ⫽ 35 n ⫽ 30 n ⫽ 35 n ⫽ 30 n ⫽ 35
MAR, 0.55 0.52i 0.57 0.41e,f 0.56 0.50i 0.60j 0.45e,f
␮m/dayh (0.48, 0.58) (0.45, 0.54) (0.49, 0.59) (0.30, 0.48) (0.52, 0.61) (0.46, 0.55) (0.53, 0.63) (0.32, 0.50)
n ⫽ 31 n ⫽ 35 n ⫽ 31 n ⫽ 33 n ⫽ 29 n ⫽ 34 n ⫽ 30 n ⫽ 31

Downloaded from https://academic.oup.com/jcem/article/101/4/1353/2804498 by guest on 10 February 2021


sLS/BS, % 4.68 4.13 15.44e 1.28e,f 7.80 7.90 24.24e 7.18f
(2.71, 7.19) (2.90, 6.29) (10.16, 18.53) (0.66, 2.62) (5.93, 12.59) (5.69, 9.93) (14.65, 30.32) (4.13, 10.58)
n ⫽ 31 n ⫽ 35 n ⫽ 31 n ⫽ 35 n ⫽ 30 n ⫽ 35 n ⫽ 30 n ⫽ 35
dLS/BS, % 2.47 2.43 10.44e 0.22e,f 5.11 4.72 28.84e 1.68e,f
(1.54, 3.16) (1.19, 3.31) (6.00, 15.41) (0.00, 0.79) (2.21, 8.57) (3.33, 7.47) (12.83, 43.96) (0.00, 5.08)
n ⫽ 31 n ⫽ 35 n ⫽ 31 n ⫽ 35 n ⫽ 30 n ⫽ 35 n ⫽ 30 n ⫽ 35

Abbreviations: dLS/BS, double labels per bone surface; DMAb, denosumab; sLS/BS, single labels per bone surface; TPTD, teriparatide. Median
values are in bold.
a
Baseline values were derived from demeclocycline labels.
b
Month 3 values were derived from tetracycline labels.
c
One sample (teriparatide group) had a missing cortex and was not included in analysis of cortical envelopes.
d
For samples with no label, MS/BS was assigned a value of zero.
e
P ⬍ .001 for within treatment group comparison from baseline to 3 months in each envelope using Wilcoxon signed-rank test.
f
P ⬍ .001 for between treatment group comparison at baseline or 3 months in each envelope using Wilcoxon rank-sum test.
g
P ⬍ .01 for within treatment group comparison from baseline to 3 months in each envelope using Wilcoxon signed-rank test.
h
For MAR and BFR/BS derived from MAR, MAR measured on samples with double label and on samples with single label only that were imputed
to 0.3 ␮m/day (samples with no label reported as missing for MAR and assigned a value of zero for BFR/BS).
i
P ⬍ .05 for between treatment group comparison at baseline or 3 months in each envelope using Wilcoxon rank-sum test.
j
P ⬍ .05 for within treatment group comparison from baseline to 3 months in each envelope using Wilcoxon signed-rank test.

nosumab vs teriparatide group (P ⬍ .001) at month 3 in all ratide subject. Additionally, with teriparatide treatment,
envelopes. the posttreatment label is shifted downward with respect
For indices with baseline and month 3 data, the direc- to the baseline label (Figure 3C, see asterisk), confirming
tional change with treatment was generally consistent across that the mineralization front was moving laterally during
envelopes (increased with teriparatide; decreased with deno- the bone formation phase.
sumab), with the exception of MAR. In the cancellous en-
velope, there was no significant longitudinal change in me- Safety
dian MAR for the teriparatide group (P ⫽ .311), but a There were no deaths in this trial. Two subjects in each
significant decrease was observed for the denosumab group group reported serious adverse events (teriparatide: chest
(P ⬍ .001). Consequently, MAR was higher with teripa- pain and lung adenocarcinoma; denosumab: appendicitis
ratide than with denosumab at 3 months (P ⬍ .001). In the and deep vein thrombosis); none of these events were con-
endocortical and intracortical envelopes, MAR increased sidered related to teriparatide or denosumab treatment,
with teriparatide and decreased with denosumab. There was and none led to study discontinuation. One teriparatide-
no change in MAR with either treatment in the periosteal treated subject discontinued because of an adverse event of
envelope, perhaps because of a lower number of samples abdominal pain, which was considered by the investigator
with labels in this envelope than in the other envelopes. to be unrelated to teriparatide.
Figure 3 shows representative images from two subjects At least one TEAE was experienced by 27 (81.8%)
each in the teriparatide and denosumab groups. These teriparatide-treated and 23 (63.9%) denosumab-treated
subjects were chosen because their MS/BS values were sim- subjects. The most common TEAEs experienced by teripa-
ilar to the median value for their assigned treatment group. ratide-treated subjects were contusion related to the bi-
There are many more posttreatment labels (red arrows) in opsy procedure (30.3%); arthralgia (15.2%); and nausea,
the teriparatide subject (Figure 3A) than in the denosumab fatigue, and headache (12.1% each). The most common
subject (Figure 3B), and the labels are longer in the teripa- TEAEs experienced by denosumab-treated subjects were
doi: 10.1210/jc.2015-4181 press.endocrine.org/journal/jcem 1359

Table 2. Continued
Intracorticalc Periostealc

Indices, Baseline Month 3 Baseline Month 3


Median
(Q1, Q3) TPTD DMAb TPTD DMAb TPTD DMAb TPTD DMAb
MS/BS, %d 8.99 10.16 21.69e 3.05e,f 0.73 0.48 4.69e 0.00e,f
(5.74, 11.84) (6.85, 16.95) (15.61, 30.17) (2.03, 7.38) (0.00, 3.25) (0.00, 1.76) (1.24, 8.98) (0.00, 0.00)
n ⫽ 30 n ⫽ 35 n ⫽ 30 n ⫽ 35 n ⫽ 30 n ⫽ 35 n ⫽ 30 n ⫽ 35
BFR/BS, 0.0216 0.0229 0.0668e 0.0048e,f 0.0008 0.0005 0.0051e 0.0000e,f
mm3/mm2/yh (0.0127, 0.0295) (0.0144, 0.0376) (0.0465, 0.0950) (0.0025, 0.0099) (0.0000, 0.0033) (0.0000, 0.0023) (0.0014, 0.0111) (0.0000, 0.0000)
n ⫽ 30 n ⫽ 35 n ⫽ 30 n ⫽ 35 n ⫽ 30 n ⫽ 35 n ⫽ 30 n ⫽ 35
MAR, 0.63 0.63 0.84e 0.40e,f 0.30 0.30 0.30 0.30
␮m/dayh (0.54, 0.70) (0.54, 0.70) (0.78, 0.90) (0.30, 0.45) (0.30, 0.43) (0.30, 0.37) (0.30, 0.33) (0.30, 0.30)
n ⫽ 30 n ⫽ 35 n ⫽ 30 n ⫽ 34 n ⫽ 20 n ⫽ 25 n ⫽ 29 n⫽5

Downloaded from https://academic.oup.com/jcem/article/101/4/1353/2804498 by guest on 10 February 2021


sLS/BS, % 8.34 11.69 15.28e 4.66e,f 1.45 0.77 9.37e 0.00e,f
(5.75, 11.24) (7.09, 13.89) (11.17, 17.26) (2.54, 9.57) (0.00, 5.50) (0.00, 3.50) (2.47, 13.92) (0.00, 0.00)
n ⫽ 30 n ⫽ 35 n ⫽ 30 n ⫽ 35 n ⫽ 30 n ⫽ 35 n ⫽ 30 n ⫽ 35
dLS/BS, % 4.72 5.82 14.96e 0.83e,f 0.00 0.00 0.00 0.00i,g
(2.61, 6.36) (2.80, 9.73) (10.55, 19.69) (0.00, 1.87) (0.00, 0.39) (0.00, 0.29) (0.00, 0.62) (0.00, 0.00)
n ⫽ 30 n ⫽ 35 n ⫽ 30 n ⫽ 35 n ⫽ 30 n ⫽ 35 n ⫽ 30 n ⫽ 35

constipation, contusion, and postprocedural discomfort that PTH increases are compensatory responses to main-
(11.1% each). TEAEs were generally mild or moderate in tain serum calcium levels, which decrease with antiresorp-
severity with the exception of one subject in the teripa- tive treatment (8 –11, 13, 28, 29). In this study, we could
ratide group (lung adenocarcinoma) and two subjects in not address this hypothesis because serum calcium was
the denosumab group (appendicitis and carpal tunnel syn- assessed at screening only. Likewise, our finding that iPTH
drome), whose events were considered severe. levels decrease during teriparatide treatment has been re-
ported previously (28, 30, 31). This effect is expected and
is likely because of teriparatide-mediated suppression of
Discussion endogenous PTH release from the parathyroid glands.
Theoretically, to be defined as a bone anabolic agent,
Our results confirm previous observations that iPTH lev-
treatment must result in a significant increase in bone mass
els are elevated after a single administration of deno-
sumab. The response profile is consistent with these stud- because of an increase in bone remodeling with a positive
ies, with maximal iPTH elevations occurring at 1 month basic multicellular unit balance in which bone formation
and then declining toward baseline through 6 months is greater than resorption (1, 25). In the presence of an
(10 –13). Although we did not assess the kinetics of en- anabolic agent, indices of bone formation, including MS/
dogenous PTH in this study, it is unlikely that they would BS, BFR/BS, osteoid surface/BS, and W.Th, are expected to
be similar to the pulsatile increments that occur with daily increase relative to placebo or baseline because they reflect
subcutaneous injection of PTH peptides and have been an increase in osteoblast number and activity. Despite the
associated with bone anabolism (27). The increase in PTH significant elevation in serum iPTH levels during 6 months
that we observed in our study is not unique to denosumab of denosumab treatment, these four indices (except W.Th
and may be an attribute of the antiresorptive drug class in the intracortical envelope), along with others, including
because iPTH also increases in response to alendronate MAR (except in the periosteal envelope), were signifi-
and zoledronic acid treatment (11, 12, 28, 29). It is likely cantly lower with denosumab compared to teriparatide at
1360 Dempster et al Teriparatide or Denosumab Bone Effects J Clin Endocrinol Metab, April 2016, 101(4):1353–1363

Table 3. Histomorphometric Indices (Month 3)a


Cancellous Endocorticalb Intracorticalb
Indices, Median
(Q1, Q3)c TPTD DMAb TPTD DMAb TPTD DMAb
Dynamic
BFR/BV, % per y 0.66 0.03d
(0.45, 0.88) (0.01, 0.07) NM NM NM NM
n ⫽ 31 n ⫽ 35
Ac.f, cycle/y 1.41 0.06d 2.77 0.34d 1.88 0.12d
(0.88, 2.02) (0.02, 0.14) (1.48, 4.31) (0.09, 0.59) (1.13, 2.48) (0.06, 0.27)
n ⫽ 31 n ⫽ 35 n ⫽ 30 n ⫽ 35 n ⫽ 30 n ⫽ 35
a.FP, y 0.13 0.16d 0.15 0.19d 0.13 0.26d

Downloaded from https://academic.oup.com/jcem/article/101/4/1353/2804498 by guest on 10 February 2021


(0.12, 0.14) (0.14, 0.20) (0.14, 0.16) (0.16, 0.22) (0.11, 0.14) (0.22, 0.35)
n ⫽ 31 n ⫽ 33 n ⫽ 30 n ⫽ 31 n ⫽ 30 n ⫽ 34
Aj.AR, ␮m/day 0.58 0.14d 0.76 0.58e 1.03 0.35d
(0.46, 0.67) (0.08, 0.33) (0.56, 0.88) (0.22, 0.81) (0.88, 1.18) (0.19, 0.73)
n ⫽ 31 n ⫽ 33 n ⫽ 30 n ⫽ 30 n ⫽ 30 n ⫽ 33
Mlt, day 10.93 21.68f 9.28 9.15 7.21 12.04f
(9.66, 13.78) (12.20, 48.83) (7.37, 11.46) (5.50, 18.00) (5.92, 8.52) (7.08, 24.94)
n ⫽ 31 n ⫽ 33 n ⫽ 30 n ⫽ 31 n ⫽ 30 n ⫽ 34
Omt, day 10.88 9.86e 11.43 10.76 8.40 11.45d
(10.11, 11.97) (8.28, 11.80) (9.94, 12.87) (7.89, 12.92) (7.45, 9.48) (9.45, 13.73)
n ⫽ 31 n ⫽ 33 n ⫽ 30 n ⫽ 31 n ⫽ 30 n ⫽ 34
Tt.FP, y 0.12 0.47d 0.12 0.15 0.10 0.30d
(0.11, 0.15) (0.21, 0.91) (0.10, 0.15) (0.11, 0.38) (0.09, 0.12) (0.16, 0.52)
n ⫽ 31 n ⫽ 33 n ⫽ 30 n ⫽ 31 n ⫽ 30 n ⫽ 33
Static
OV/BV, % 2.58 0.39d
(1.67, 3.51) (0.17, 0.65) NM NM NM NM
n ⫽ 31 n ⫽ 35
OS/BS, % 20.51 3.46d 30.85 6.95d 17.81 4.34d
(12.50, 24.13) (2.05, 5.40) (21.47, 48.02) (2.82, 10.88) (11.89, 22.50) (2.07, 6.17)
n ⫽ 31 n ⫽ 35 n ⫽ 30 n ⫽ 35 n ⫽ 30 n ⫽ 35
O.Th, ␮m 6.12 3.78d 6.99 4.33d 7.08 4.68d
(5.52, 6.71) (3.49, 4.18) (5.61, 7.99) (3.49, 4.78) (6.15, 7.92) (3.68, 5.13)
n ⫽ 31 n ⫽ 35 n ⫽ 30 n ⫽ 35 n ⫽ 30 n ⫽ 35
W.Th, ␮m 26.29 24.03d 31.88 30.08e 38.25 37.80
(25.58, 27.84) (22.79, 25.23) (29.75, 32.99) (27.61, 32.03) (33.87, 39.84) (34.61, 40.15)
n ⫽ 31 n ⫽ 35 n ⫽ 30 n ⫽ 35 n ⫽ 30 n ⫽ 35
ES/BS, % 3.73 1.52d 3.79 1.65d 11.19 1.27d
(2.95, 6.05) (0.92, 2.87) (2.79, 6.25) (0.78, 2.47) (7.37, 13.71) (0.90, 3.82)
n ⫽ 31 n ⫽ 35 n ⫽ 30 n ⫽ 35 n ⫽ 30 n ⫽ 35
Abbreviations: a.FP, active formation period; Ac.f, activation frequency; Aj.AR, adjusted apposition rate; DMAb, denosumab; Mlt, mineralization
lag time; NM, not measured; O.Th, osteoid thickness; Omt, osteoid maturation time; OS, osteoid surface; OV, osteoid volume; Tt.FP, total
formation period; TPTD, teriparatide. Median values are in bold.
a
Month 3 values were derived from tetracycline labels.
b
One sample (teriparatide group) had a missing cortex and was not included in the analysis of cortical envelopes.
c
For dynamic parameters derived from MAR, MAR measured on samples with double label and on samples with single label only imputed to 0.3
␮m/day (samples with no label reported as missing with the exception of BFR/BV and Ac.f, which were assigned a value of zero).
d
P ⬍ .001 for between treatment group comparison using Wilcoxon rank-sum test.
e
P ⬍ .05 for between treatment group comparison using Wilcoxon rank-sum test.
f
P ⬍ .01 for between treatment group comparison using Wilcoxon rank-sum test.

3 months. In addition, with quadruple labeling, we dem- these results provide for the first time direct evidence in a
onstrated that MS/BS and BFR/BS decreased from baseline single biopsy for stimulation of bone formation in the peri-
to 3 months with denosumab treatment, whereas these osteal envelope with teriparatide. These same indices,
indices increased with teriparatide treatment. however, decreased with denosumab treatment.
The finding that teriparatide treatment resulted in sig- The median baseline value for MAR was significantly
nificant increases in MS/BS and BFR/BS in the periosteal different between treatment groups in the cancellous and
envelope is interesting and adds to the current body of endocortical envelopes, being higher with teriparatide
evidence for this effect (32, 33). Using quadruple labeling, treatment. This difference does not appear to be the result
doi: 10.1210/jc.2015-4181 press.endocrine.org/journal/jcem 1361

greater proportion of the formation


A B sites with denosumab treatment will
be at a later stage in the bone forma-
tion phase, and, therefore, MAR
would be expected to be lower. By
contrast, more remodeling units will
be initiated with teriparatide with a
greater proportion of formation sites
at an earlier stage in the bone forma-
tion phase, and MAR would be ex-
C D pected to be higher. Treatment ef-

Downloaded from https://academic.oup.com/jcem/article/101/4/1353/2804498 by guest on 10 February 2021


fects on osteoblasts are not needed to
explain these findings; this could be
explained by different effects of de-
nosumab and teriparatide on activa-
tion frequency.
This study has several strengths.
The ability to measure baseline his-
Figure 3. Quadruple labeling in teriparatide- and denosumab-treated subjects in cancellous bone. tomorphometric data as a result of
Low- (top) and high-power (bottom) images of samples, each from a different subject, representative using quadruple-labeling allowed
of the month 3 median values for MS/BS for each treatment group (18.73% teriparatide vs 0.96%
for a head-to-head comparison of di-
denosumab). The white arrows indicate first labeling period (demeclocycline) at baseline; red arrows
indicate second labeling period (tetracycline) at 3 months. A, Teriparatide, low power; B, Denosumab rectional changes, providing a more
low power; C, Teriparatide, high power; D, Denosumab, high power. accurate and robust picture of drug
effects on bone metabolism and
MOA. Additionally, although this
of differences in baseline bone remodeling rate, because study was powered for MS/BS, a full panel of supporting
there were no differences in the distribution of subjects in histomorphometric indices was assessed across four bone
the two treatment groups among the P1NP strata at ran- envelopes providing a comprehensive assessment of the
domization. After adjusting for baseline MAR, results for effects of these drugs on bone at an early time point. More-
primary and secondary endpoints were similar to the orig- over, based on preclinical findings that suggest a role for
inal findings and conclusions remained the same. MAR modeling-based bone formation in the MOA of denosumab
increased with teriparatide in the endocortical and intra- (37), we have the ability to assess both modeling- and re-
cortical envelopes, whereas it decreased with denosumab modeling-based formation in all four envelopes in these hu-
in the cancellous, endocortical, and intracortical enve- man biopsy samples. This work is ongoing. Finally, changes
lopes. The current finding that teriparatide is able to stim- in iPTH, histomorphometric indices, and BTMs were as-
ulate MAR as previously reported (17, 32) and that de- sessed during the same time frame in the same subjects. This
nosumab reduced MAR is of interest. This is the first study study was, however, limited by a short duration with a bone
to use quadruple labeling to assess the effects of an anti- biopsy taken after 3 months of treatment.
resorptive agent, and it is the first prospective demonstra- In conclusion, denosumab-induced increases in iPTH,
tion of an inhibitory effect of an antiresorptive agent on when examined in parallel with changes in BTMs and a
MAR. It is important to note that this decrease in MAR full panel of histomorphometric indices across four bone
with denosumab remains significant when only the data envelopes, are not consistent with and do not support
from subjects with double label were used (data not early bone-forming activity of this drug. Our data suggest
shown). The mechanism for such an effect is unclear, but that the sustained and substantial increases in BMD seen
there are possible explanations. One could be because of with denosumab treatment are not explained by indirect
denosumab’s action to reduce osteoclast number and ac- anabolic action induced by increases in endogenous PTH
tivity, resulting in a decrease in osteoclast-derived osteo- and must be caused by other mechanism(s).
blast stimulating factors or “clastokines” (34). A second
possibility relates to the decline of MAR over time during
the bone formation phase (35, 36). Early in antiresorptive
Acknowledgments
treatment, there is a rapid reduction of initiation of new We thank the study investigators, site personnel, and patients
remodeling, and sites that were active before treatment and their families. Medical writing support was provided by Lori
complete their formation phase (1). Consequently, a Kornberg (INC Research, Raleigh, NC).
1362 Dempster et al Teriparatide or Denosumab Bone Effects J Clin Endocrinol Metab, April 2016, 101(4):1353–1363

Address all correspondence and requests for reprints to: Makras P. Comparative effect of zoledronic acid versus denosumab
David W. Dempster, Regional Bone Center, Helen Hayes Hos- on serum sclerostin and dickkopf-1 levels of naive postmenopausal
pital, Route 9W, West Haverstraw, New York 10993. E-mail: women with low bone mass: a randomized, head-to-head clinical
ddempster9@aol.com. trial. J Clin Endocrinol Metab. 2013;98:3206 –3212.
9. Bekker PJ, Holloway DL, Rasmussen AS, et al. A single-dose pla-
This work was sponsored by Eli Lilly and Company.
cebo-controlled study of AMG 162, a fully human monoclonal an-
Trial Registration: ClinicalTrials.gov: NCT01753856. tibody to RANKL, in postmenopausal women. J Bone Miner Res.
Disclosure Summary: D.W.D. has received research grants 2004;19:1059 –1066.
from and serves as a consultant and speaker for Eli Lilly and 10. Makras P, Polyzos SA, Papatheodorou A, Kokkoris P, Chatzifotia-
Company and Amgen, and is a consultant for Merck, Tarsa dis D, Anastasilakis AD. Parathyroid hormone changes following
Pharmaceuticals, and Radius Health. R.R.R. receives research denosumab treatment in postmenopausal osteoporosis. Clin Endo-
grants from and is a consultant for Eli Lilly and Company and crinol (Oxf). 2013;79:499 –503.
Merck. J.P.B. receives research grants from Amgen, Eli Lilly and 11. McClung MR, Lewiecki EM, Cohen SB, et al. AMG 162 Bone Loss
Company, and Novartis; serves as a consultant for Amgen, Eli Study Group. Denosumab in postmenopausal women with low

Downloaded from https://academic.oup.com/jcem/article/101/4/1353/2804498 by guest on 10 February 2021


bone mineral density. N Engl J Med. 2006;354:821– 831.
Lilly and Company, and Radius; and is a speaker for Amgen and
12. Seeman E, Libanati C, Austin M, et al. The transitory increase in
Eli Lilly and Company. C.P.R. has received grant/research sup- PTH following denosumab administration is associated with re-
port from Amgen, Merck, and Eli Lilly and Company and No- duced intracortical porosity: a distinctive attribute of denosumab
vartis, and has served on scientific advisory boards for Amgen, therapy. J Bone Miner Res. 2011; 26(Suppl 1) (presentation 1064).
Merck, Eli Lilly and Company, NPS, and Novartis and receives 13. Sugimoto T, Matsumoto T, Hosoi T, et al. Three-year denosumab
honorarium and travel expenses for advisory boards. E.M.L. has treatment in postmenopausal Japanese women and men with os-
received institutional grant/research support from Amgen, teoporosis: results from a 1-year open-label extension of the Deno-
Merck, and Eli Lilly and Company; has served on scientific ad- sumab Fracture Intervention Randomized Placebo Controlled Trial
visory boards for Amgen, Merck, Eli Lilly and Company, Radius (DIRECT). Osteoporos Int. 2015;26:765–774.
14. Seeman E, Libanati C, Austin M, et al. Association between tran-
Health, AgNovos Healthcare, Alexion, NPS, and AbbVie; and
sitory increase in PTH following denosumab administration and
receives honorarium and travel expenses for advisory boards.
reduced intracortical porosity is a distinctive attribute of denosumab
P.D.M. serves on scientific advisory boards for Alexion, Amgen, therapy. Bone. 2012;50:S162.
AgNovos, Lilly, Merck, Radius Pharma, and Roche; receives 15. Zebaze RM, Libanati C, Austin M, et al. Differing effects of deno-
research grants from Amgen, Boehringer Ingelheim, Immunodi- sumab and alendronate on cortical and trabecular bone. Bone.
agnostics, Lilly, Merck, Merck Serrano, Novartis, Novo Nor- 2014;59:173–179.
disk, Radius Pharma, Roche Diagnostics, and Takeda; and is a 16. Papapoulos S, Lippuner K, Roux C, et al. The effect of 8 or 5 years
speaker for Alexion Pharmaceuticals and Radius Health. S.D.R. of denosumab treatment in postmenopausal women with osteopo-
receives research grants from Eli Lilly and Company. D.L.K. rosis: results from the FREEDOM Extension study. Osteoporos Int.
2015;26:2773–2783.
receives research grants, consultancies, and/or honoraria from
17. Lindsay R, Cosman F, Zhou H, et al. A novel tetracycline labeling
Amgen, Eli Lilly and Company, Pfizer, GSK, Astra Zeneca, and
schedule for longitudinal evaluation of the short-term effects of an-
Astalis. R.L. is a speaker for Eli Lilly and Company. J.H.K., J.A., abolic therapy with a single iliac crest bone biopsy: early actions of
K.A.T., B.J., and V.A.R. are employees of Eli Lilly and Company teriparatide. J Bone Miner Res. 2006;21:366 –373.
and own stock. H.Z. has nothing to declare. 18. Prolia Prescribing Information. Amgen. Thousand Oaks, CA, USA,
2015. http://pi.amgen.com/united_states/prolia/prolia_pi.pdf. Ac-
cessed November 30, 2015.
19. Dempster DW, Zhou H, Recker RR, et al. Skeletal histomorphom-
References etry in subjects on teriparatide or zoledronic acid therapy (SHOTZ)
study: a randomized controlled trial. J Clin Endocrinol Metab.
1. Riggs BL, Parfitt AM. Drugs used to treat osteoporosis: the critical 2012;97:2799 –2808.
need for a uniform nomenclature based on their action on bone 20. Rubin MR, Dempster DW, Sliney J Jr., Zhou H, et al. PTH(1– 84)
remodeling. J Bone Miner Res. 2005;20:177–184. administration reverses abnormal bone-remodeling dynamics and
2. Forteo Prescribing Information. Eli Lilly and Company. Indianap- structure in hypoparathyroidism. J Bone Miner Res. 2011;26:2727–
olis, IN, 2012. http://pi.lilly.com/us/forteo-pi.pdf. Accessed Novem- 2736.
ber 30, 2015. 21. Dempster DW. Bone quantification and dynamics of bone turnover
3. Recker RR, Armas L. The effect of antiresorptives on bone quality. by histomorphometric analysis. In: KL Becker, ed. Principles and
Clin Orthop Relat Res. 2011;469:2207–2214. Practice of Endocrinology and Metabolism. 2nd ed. Philadelphia:
4. Reginster JY, Neuprez A, Dardenne N, Beaudart C, Emonts P, J. B. Lippincott Co.; 1995:491– 498.
Bruyere O. Efficacy and safety of currently marketed anti-osteopo- 22. Dempster DW, Compston JE, Drezner MK, et al. Standardized no-
rosis medications. Best Pract Res Clin Endocrinol Metab. 2014;28: menclature, symbols, and units for bone histomorphometry: a 2012
809 – 834. update of the report of the ASBMR Histomorphometry Nomencla-
5. Tella SH, Gallagher JC. Prevention and treatment of postmeno- ture Committee. J Bone Miner Res. 2013;28:2–17.
pausal osteoporosis. J Steroid Biochem Mol Biol. 2014;142:155– 23. Lindsay R, Zhou H, Cosman F, Nieves J, Dempster D. Double and
170. quadruple tetracycline labeling of bone: impact of the label itself.
6. Cummings SR, San Martin J, McClung MR, et al. FREEDOM Trial. J Bone Miner Res. 2013;28:222–223.
Denosumab for prevention of fractures in postmenopausal women 24. Parfitt AM, Foldes J, Villanueva AR, Shih MS. Difference in label
with osteoporosis. N Engl J Med. 2009;361:756 –765. length between demethylchlortetracycline and oxytetracycline: im-
7. Reid IR, Miller PD, Brown JP, et al. Denosumab Phase 3 Bone His- plications for the interpretation of bone histomorphometric data.
tology Study Group. Effects of denosumab on bone histomorphom- Calcif Tissue Int. 1991;48:74 –77.
etry: the FREEDOM and STAND studies. J Bone Miner Res. 2010; 25. Recker RR, Kimmel DB, Dempster D, Weinstein RS, Wronski TJ,
25:2256 –2265. Burr DB. Issues in modern bone histomorphometry. Bone. 2011;
8. Anastasilakis AD, Polyzos SA, Gkiomisi A, Bisbinas I, Gerou S, 49:955–964.
doi: 10.1210/jc.2015-4181 press.endocrine.org/journal/jcem 1363

26. Dempster DW, Zhou H, Recker RR, et al. A longitudinal study of denosumab treatment for 3 months. Osteoporos Int. 2014;25:
skeletal histomorphometry at 6 and 24 months across four bone 1633–1642.
envelopes in postmenopausal women with osteoporosis receiving 32. Lindsay R, Zhou H, Cosman F, Nieves J, Dempster DW, Hodsman
teriparatide or zoledronic acid in the SHOTZ trial [published online AB. Effects of a one-month treatment with PTH(1–34) on bone
February 3, 2016]. J Bone Miner Res. doi:10.1002/jbmr.2804. formation on cancellous, endocortical, and periosteal surfaces of the
human ilium. J Bone Miner Res. 2007;22:495–502.
27. Dobnig H, Turner RT. The effects of programmed administration
33. Ma YL, Zeng Q, Donley DW, et al. Teriparatide increases bone
of human parathyroid fragment (1–34) on bone histomorphometry
formation in modeling and remodeling osteons and enhances IGF-II
and serum chemistry in rats. Endocrinology. 1997;138:4607– 4612. immunoreactivity in postmenopausal women with osteoporosis.
28. Anastasilakis AD, Polyzos SA, Makras P, et al. Circulating sema- J Bone Miner Res. 2006;21:855– 864.
phorin-4D and plexin-B1 levels in postmenopausal women with low 34. Shiwaku Y, Neff L, Nagano K, et al. The crosstalk between oste-
bone mass: the 3-month effect of zoledronic acid, denosumab or oclasts and osteoblasts is dependent upon the composition and
teriparatide treatment. Expert Opin Ther Targets. 2015;19:299 – structure of biphasic calcium phosphates. PloS One. 2015;10:
306. e0132903.
29. Polyzos SA, Anastasilakis AD, Terpos E. Transient secondary hy- 35. Eriksen EF. Normal and pathological remodeling of human trabec-

Downloaded from https://academic.oup.com/jcem/article/101/4/1353/2804498 by guest on 10 February 2021


perparathyroidism following intravenous infusion of zoledronic ular bone: three dimensional reconstruction of the remodeling se-
acid. Support Care Cancer. 2009;17:1329 –1330. quence in normals and in metabolic bone disease. Endocr Rev. 1986;
7:379 – 408.
30. Anastasilakis AD, Polyzos SA, Goulis DG, et al. Endogenous intact
36. Parfitt AM. The cellular basis of bone remodeling: the quantum
PTH is suppressed during Teriparatide (rhPTH 1–34) administra-
concept reexamined in light of recent advances in the cell biology of
tion in postmenopausal women with established osteoporosis. En- bone. Calcif Tissue Int. 1984;36:S37–S45.
docr J. 2008;55:613– 616. 37. Ominsky MS, Libanati C, Niu QT, et al. Sustained modeling-based
31. Anastasilakis AD, Polyzos SA, Makras P, et al. Circulating irisin is bone formation during adulthood in cynomolgus monkeys may con-
associated with osteoporotic fractures in postmenopausal women tribute to continuous BMD gains with denosumab. J Bone Miner
with low bone mass but is not affected by either teriparatide or Res. 2015;30:1280 –1289.

You might also like