You are on page 1of 16

Journal of the Endocrine Society, 2023, 7, 1–16

https://doi.org/10.1210/jendso/bvad043
Advance access publication 4 April 2023
Clinical Research Article

Long-Term Safety and Efficacy of Recombinant Human


Parathyroid Hormone (1-84) in Adults With Chronic
Hypoparathyroidism
Nelson B. Watts,1 John P. Bilezikian,2 Henry G. Bone,3 Bart L. Clarke,4 Douglas Denham,5
Michael A. Levine,6 Michael Mannstadt,7 Munro Peacock,8 Jeffrey G. Rothman,9

Downloaded from https://academic.oup.com/jes/article/7/5/bvad043/7103305 by guest on 09 January 2024


Tamara J. Vokes,10 Mark L. Warren,11 Shaoming Yin,12 Nicole Sherry,12
and Dolores M. Shoback13,14
1
Osteoporosis and Bone Health Services, Mercy Health, Cincinnati, OH 45236, USA
2
Division of Endocrinology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
3
Michigan Bone and Mineral Clinic, PC, Detroit, MI 48236, USA
4
Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Mayo Clinic, Rochester, MN 55905, USA
5
Clinical Trials of Texas, Inc., San Antonio, TX 78229, USA
6
Division of Endocrinology and Diabetes and Center for Bone Health, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
7
Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
8
Department of Medicine, Division of Endocrinology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
9
UPG-Endocrine (Research Division), Staten Island, NY 10301, USA
10
Section of Endocrinology, University of Chicago Medicine, Chicago, IL 60637, USA
11
Endocrinology and Metabolism, Physicians East, PA, Greenville, NC 27834, USA
12
Takeda Pharmaceuticals USA, Inc., Lexington, MA 02421, USA
13
Endocrine Research Unit, San Francisco Veterans Affairs Medical Center, San Francisco, CA 94121, USA
14
Department of Medicine, University of California, San Francisco, CA 94143, USA
Correspondence: Nelson B. Watts, MD, Mercy Health Osteoporosis and Bone Health Services, 4760 East Galbraith Road, Suite 212, Cincinnati, OH 45236, USA.
Email: nelson.watts@hotmail.com.

Abstract
Context: Chronic hypoparathyroidism is conventionally treated with oral calcium and active vitamin D to reach and maintain targeted serum
calcium and phosphorus levels, but some patients remain inadequately controlled.
Objective: To assess long-term safety and efficacy of recombinant human parathyroid hormone (1-84) (rhPTH(1-84)) treatment.
Methods: This was an open-label extension study at 12 US centers. Adults (n = 49) with chronic hypoparathyroidism were included. The
intervention was rhPTH(1-84) for 6 years. The main outcome measures were safety, biochemical measures, oral supplement doses, bone indices.
Results: Thirty-eight patients (77.6%) completed the study. Throughout 72 months, mean albumin-adjusted serum calcium was within 2.00 to
2.25 mmol/L (8.0-9.0 mg/dL). At baseline, 65% of patients with measurements (n = 24/37) were hypercalciuric; of these, 54% (n = 13/24) were
normocalciuric at month 72. Mean serum phosphorus declined from 1.6 ± 0.19 mmol/L at baseline (n = 49) to 1.3 ± 0.20 mmol/L at month
72 (n = 36). Mean estimated glomerular filtration rate was stable. rhPTH(1-84)-related adverse events were reported in 51.0% of patients
(n = 25/49); all but 1 event were mild/moderate in severity. Mean oral calcium supplementation reduced by 45% ± 113.6% and calcitriol by 74%
± 39.3%. Bone turnover markers declined by month 32 to a plateau above pretreatment values; only aminoterminal propeptide of type 1
collagen remained outside the reference range. Mean bone mineral density z score fell at one-third radius and was stable at other sites.
Conclusion: 6 years of rhPTH(1-84) treatment was associated with sustained improvements in biochemical parameters, a reduction in the
percentage of patients with hypercalciuria, stable renal function, and decreased supplement requirements. rhPTH(1-84) was well tolerated; no
new safety signals were identified.
Key Words: recombinant human parathyroid hormone (1-84), hypoparathyroidism, active vitamin D, calcium, bone turnover, mineral homeostasis
Abbreviations: 1,25(OH)2D, 1,25-dihydroxyvitamin D; 25(OH)D, 25-hydroxyvitamin D; BAP, bone alkaline phosphatase; BMD, bone mineral density; BTM, bone
turnover marker; CKD, chronic kidney disease; CTX, cross-linked C-telopeptide of type 1 collagen; eGFR, estimated glomerular filtration rate; EOT, end of
treatment; ITT, intent-to-treat; PTH, parathyroid hormone; P1NP, aminoterminal propeptide of type 1 collagen; rhPTH(1-84), recombinant human parathyroid
hormone (1-84); TEAE, treatment-emergent adverse event.

Hypoparathyroidism is a rare endocrine disorder characterized parathyroid hormone (PTH) despite low circulating concentra­
by absent or inappropriately low circulating concentrations of tions of calcium [1]. The consequences of loss of PTH action on

Received: 30 November 2022. Editorial Decision: 24 March 2023. Corrected and Typeset: 21 April 2023
© The Author(s) 2023. Published by Oxford University Press on behalf of the Endocrine Society.
This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-NoDerivs licence (https://creativecommons.
org/licenses/by-nc-nd/4.0/), which permits non-commercial reproduction and distribution of the work, in any medium, provided the original work is not altered
or transformed in any way, and that the work is properly cited. For commercial re-use, please contact journals.permissions@oup.com
2 Journal of the Endocrine Society, 2023, Vol. 7, No. 5

the skeleton, kidney, and gastrointestinal tract typically mani­ the long-term safety and tolerability of rhPTH(1-84) for the
fest as hypocalcemia, hyperphosphatemia, hypercalciuria, de­ treatment of adult patients with chronic hypoparathyroidism.
creased bone turnover, and increased bone mineral density The secondary objectives were (1) to evaluate the impact of dif­
(BMD) [2, 3]. Management goals for patients with hypopara­ ferent preparations of calcium on the response to rhPTH(1-84);
thyroidism include maintenance of serum calcium in the lower (2) to establish whether dosing with rhPTH(1-84) across a
part of or slightly below the reference range, control of urinary range of 25 to 100 μg could be implemented safely and effect­
calcium excretion to within normal limits, normalization of se­ ively and maintained throughout long-term treatment; and
rum phosphorus levels, and improved quality of life [4-6]. (3) to evaluate the impact of thiazide diuretics on serum and
Conventional therapy for hypoparathyroidism includes oral urinary calcium in response to rhPTH(1-84). Detailed study
calcium and active vitamin D (eg, calcitriol) [1, 7]. In addition, methods for the assessment of safety and biochemical parame­
thiazide diuretics are sometimes prescribed for hypercalciuria ters have been reported previously [22].
[2, 8]. Conventional therapy increases serum calcium through
enhanced gastrointestinal absorption but does not replace other Participants
actions of PTH, including control of renal tubular reabsorption
Patients were eligible to participate in RACE if they completed

Downloaded from https://academic.oup.com/jes/article/7/5/bvad043/7103305 by guest on 09 January 2024


of calcium, excretion of phosphorus and regulation of bone
the 24-week REPLACE study and/or the 8-week RELAY study.
turnover [2, 9]. When hypoparathyroidism is not adequately
Daily subcutaneous injections of rhPTH(1-84) were self-
controlled by conventional therapy, chronic hypocalcemia, hy­
administered. The starting dose of rhPTH(1-84) was 50 µg for
perphosphatemia, and hypercalciuria can become troubling
patients with total serum calcium ≤2.37 mmol/L (≤9.5 mg/dL)
clinical issues [10-12] and may increase the risk of complica­
and for patients with total serum calcium >2.37 mmol/L
tions, including central nervous system calcifications, nephro­
(>9.5 mg/dL) taking ≥500 mg of supplemental calcium and/or
calcinosis and nephrolithiasis, and impaired renal function
any calcitriol. Starting dose was 25 µg for patients with total se­
[10, 13-16].
rum calcium >2.37 mmol/L taking <500 mg of supplemental
Recombinant human parathyroid hormone (1-84),
calcium and no calcitriol. Investigators could increase or de­
rhPTH(1-84), is full-length PTH approved in the United
crease the dose of rhPTH(1-84) at any time during the study in
States and Europe as an adjunct to calcium and active vitamin
stepwise adjustments of 25 µg, to a maximum of 100 µg, with
D for adults with hypoparathyroidism. Safety and efficacy of
the goal of achieving or maintaining total serum calcium levels
rhPTH(1-84) for patients with hypoparathyroidism have been
in the target range of 2.00 to 2.25 mmol/L (8.0-9.0 mg/dL).
evaluated in several prospective clinical studies [17-20].
Patients not on thiazide diuretics at baseline were allowed
RACE (ClinicalTrials.gov identifier, NCT01297309) was an
to initiate treatment with a thiazide diuretic after week 16
open-label extension study of the REPLACE (NCT00732615)
to manage hypercalciuria if they were on a stable dose of
[18, 21] and RELAY (NCT01268098) [17] studies and was de­
rhPTH(1-84) and had a 24-hour urine calcium >7.5 mmol
signed to assess long-term safety and efficacy of rhPTH(1-84).
(>300 mg) for men or >6.25 mmol (>250 mg) for women.
An analysis of 5-year results from RACE demonstrated a safety
Compliance with dosing was assessed using patient diary
profile consistent with those of REPLACE and RELAY. In add­
data. Patients were considered compliant if they reported
ition, mean serum calcium levels remained within the target
taking ≥80% of their expected doses.
range, mean 24-hour urinary calcium declined to within the ref­
The study was conducted in accordance with International
erence range, and skeletal catabolic and anabolic activity, as
Council for Harmonisation guidelines, Good Clinical Practice,
measured by levels of bone turnover markers (BTMs), stabilized
and the World Medical Association Declaration of Helsinki; it
within reference ranges after an initial increase; however, ami­
was approved by local institutional review boards or ethics
noterminal propeptide of type 1 collagen (P1NP) levels re­
committees. All patients provided written informed consent.
mained above the reference range [22]. In another long-term,
open-label study [20], treatment with rhPTH(1-84) over 8 years
led to sustained reduction in requirements for oral calcium and Outcome Measures and Safety Evaluations
calcitriol and decreases in urinary calcium, while serum calcium Outcome measures included albumin-adjusted serum cal­
levels were maintained slightly below the lower limit of the ref­ cium, 24-hour urinary calcium and phosphorus excretion, se­
erence range in accordance with current guidelines [4-6]. In rum phosphorus, serum creatinine, treatment-emergent
addition, BTMs increased during the first 12 months of treat­ adverse events (TEAEs), prescribed doses of oral calcium
ment, then declined to new steady-state levels that were above and calcitriol, serum 25-hydroxyvitamin D (25[OH]D) and
pretreatment values [23, 24]. 1,25-dihydroxyvitamin D (1,25[OH]2D), BTMs, and BMD.
Because rhPTH(1-84) therapy for hypoparathyroidism is a Samples for serum albumin, calcium, phosphorus, and cre­
chronic, life-long treatment, assessment of long-term safety atinine were collected under fasting conditions. Calcium-
and efficacy is important. In addition, the collection of detailed phosphorus product, estimated glomerular filtration rate
and granular data on biochemical parameters enables a deeper (eGFR), rhPTH(1-84) dosing, and urinary calcium normal­
understanding of the impact of treatments on the disease course. ized to body weight were evaluated in post hoc analyses.
Here we report final, 6-year safety- and efficacy-related out­ eGFR was calculated using the Chronic Kidney Disease
comes of treatment with rhPTH(1-84) from the RACE study. Epidemiology Collaboration (CKD-EPI) equation [25]. The
composite efficacy outcome was the proportion of patients
who met all of the following 3 criteria: ≥ 50% reduction
Materials and Methods
from baseline in oral calcium dose (or calcium ≤500 mg/
Study Design day), ≥ 50% reduction from baseline in calcitriol dose (or cal­
The open-label RACE extension study was conducted between citriol ≤0.25 μg/day), and normalized or maintained
April 2011 and February 2018 and enrolled patients from 12 albumin-adjusted serum calcium compared with baseline val­
centers in the United States. The primary objective was to assess ue and not exceeding the upper limit of normal for the central
Journal of the Endocrine Society, 2023, Vol. 7, No. 5 3

laboratory (2.55 mmol/L; 10.2 mg/dL). TEAEs were defined Statistical Analyses
as any unfavorable and unintended sign (including an abnor­ SAS version 9.4 (SAS Institute Inc., Cary, NC, USA) was used
mal laboratory finding), symptom, or disease that occurred for all statistical analyses. Because no formal hypothesis test­
during rhPTH(1-84) treatment, whether or not the event ing was performed, the sample size of this extension study was
was considered by the investigator to be related to not a consideration. Statistical testing was precluded by the
rhPTH(1-84). TEAEs were classified as mild, moderate, or se­ open-label study design and lack of comparator. Continuous
vere in intensity. TEAEs classified as severe were those that re­ variables were summarized using descriptive statistics, includ­
quired vigorous therapeutic intervention and interrupted ing number of patients, mean, and SD; categorical variables
usual activities. Serious TEAEs were defined as any medical were summarized with the number of patients and percen­
event that required hospitalization or prolonged hospitaliza­ tages. Visits were summarized based on the analysis visit win­
tion, caused persistent or significant disability, was life- dows. If more than 1 nonmissing observation was recorded
threatening, resulted in death, or was judged by the investigator within the same analysis visit window, the value of the obser­
as an important medical event. vation closest to the target day was used. If observations were
All assessments were carried out as reported previously to equidistant in days from the target day, the later measurement

Downloaded from https://academic.oup.com/jes/article/7/5/bvad043/7103305 by guest on 09 January 2024


month 60 [22]. From month 60 onward, the same schedule of based on measurement date and time was used. Responder
assessments and procedures was applied. Total serum calcium, rate and calcium and calcitriol supplementation endpoints
serum phosphorus, and serum 25-hydroxyvitamin D (25(OH) were based on investigator-prescribed data. Missing safety
D) were evaluated every 2 months. Twenty-four–hour urine cal­ and efficacy parameters were not imputed. For some parame­
cium and phosphorus, serum creatinine, eGFR, and BTMs were ters, patient numbers reported here differ from those reported
measured every 4 months and serum 1,25-dihydroxyvitamin D previously [22] because data were not stable at the 5-year
(1,25(OH)2D) every 6 months. BMD was assessed annually mark and were subsequently updated before database lock
and reported as percentage change from baseline. BMD was ac­ for the 6-year analysis. Data are presented as mean ± SD un­
quired using Hologic (Marlborough, MA, USA) and GE Lunar less otherwise specified.
(Madison, WI, USA) dual-energy x-ray absorptiometry instru­
ments. No weight adjustment was made to z scores calculated
on GE instruments.
Results
The analysis populations were the safety population, which
included all patients who received ≥1 dose of rhPTH(1-84), Patient Population
and the intent-to-treat (ITT) population, which included all We enrolled a total of 49 patients in the RACE extension study,
patients who received ≥1 dose of rhPTH(1-84) and had ≥1 ef­ including 48 patients who completed the RELAY and/or
ficacy measurement. In addition, in post hoc analyses, data REPLACE studies. In addition, 1 patient who enrolled in
were generated for the subset of patients who completed 72 REPLACE but discontinued during optimization met the eligi­
months of rhPTH(1-84) treatment (completer population). bility criteria and participated in RACE. Of the 49 patients
In this study, the safety and ITT populations were identical. treated with rhPTH(1-84), 38 patients (77.6%) completed
Baseline values for this study were defined as follows: for pa­ the study (Fig. 1). One patient discontinued within the analysis
tients who completed REPLACE and RELAY, or only com­ visit window for the month 72 visit and was therefore consid­
pleted REPLACE, the baseline value of REPLACE was used. ered to have completed the study; however, this patient did
For patients who only completed RELAY, the baseline value not contribute data to all study outcomes at month 72.
of RELAY was used. For patients who enrolled in Therefore, the ITT and safety populations consist of all 49 en­
REPLACE and dropped out during optimization and for those rolled patients, and the completer population comprised 37 pa­
new to rhPTH(1-84) treatment, the baseline visit of this study tients (75.5%) who completed 72 months of rhPTH(1-84)
was used. Patients whose hypoparathyroidism was not treatment and had month 72 endpoint data available.
adequately controlled at baseline were defined as those The mean duration of rhPTH(1-84) exposure was 2038 ±
with ≥1 biochemical parameter outside the following refer­ 675 days (∼5.6 years; median 2333 days; range 41-2497
ence ranges: albumin-adjusted serum calcium, 2.13 to days; n = 49). Eleven patients withdrew from the study. Six
2.63 mmol/L (8.5-10.5 mg/dL); serum phosphorus, 0.81 to discontinued based on the patient’s decision: 2 patients were
1.45 mmol/L (2.5-4.5 mg/dL); calcium-phosphorus product, able to receive rhPTH(1-84) by prescription (1437 and 1996
<4.4 mmol2/L2 (<55 mg2/dL2); serum creatinine, 51.3 to days in study, respectively), 2 were tired of daily injections
112.3 µmol/L (0.58-1.27 mg/dL); eGFR, 15 to 150 mL/min/ of study medication (393 and 1393 days in study), 1 found
1.73 m2; 24-hour urinary calcium, men <7.5 mmol the number of pills inconvenient (107 days in study), and 1
(<300 mg), women <6.25 mmol (<250 mg). had difficulty swallowing calcium supplements and maintain­
The end-of-treatment (EOT) visit was defined as the last ing study drug compliance (68 days in study). Two patients
available measurement during the treatment period. Patients discontinued based on the investigator’s decision: 1 patient
received rhPTH(1-84) until January 2018 or completion of was unable to attend clinic visits (750 days in study), and
the last study visit at month 72, whichever came later. the second experienced health deterioration in conjunction
Therefore, some patients completed the study with more with difficulty traveling to the study site (1698 days in study).
than 72 months of rhPTH(1-84) and so had their final visit The remaining 3 patients discontinued for the following rea­
after month 72 labeled as the EOT visit. Data for EOT out­ sons: 1 patient had a TEAE of metastatic lung carcinoma
comes were summarized and are reported separately. that was considered unrelated to treatment (study 618 days
Patients were contacted by phone 4 weeks after the EOT or in study), 1 patient died from cardiac arrest that was consid­
early termination visit to monitor for and follow up on any ered unrelated to treatment (2015 days in study), and 1 de­
serious TEAEs or TEAEs that were considered by the investi­ clined to continue the extension study after the first 12
gator to be related to rhPTH(1-84) treatment. months (347 days in study).
4 Journal of the Endocrine Society, 2023, Vol. 7, No. 5

Downloaded from https://academic.oup.com/jes/article/7/5/bvad043/7103305 by guest on 09 January 2024


Figure 1. Flow diagram showing patients enrolled in the study. *One patient dropped out during optimization in REPLACE but met inclusion and
exclusion criteria for RACE. †Among enrolled patients, 48 of 49 had chronic hypoparathyroidism that was not adequately controlled at baseline for RACE
based on biochemical measurements. ‡One patient discontinued at around month 72; however, the patient’s last study visit fell within the analysis visit
window for the month 72 visit, and the patient was therefore deemed to have completed the study. ITT = intent to treat.

The demographic and baseline clinical characteristics of the whom data were available had hypercalciuria at baseline, and
patients were reported previously [22]. The study population 8 of 26 women (30.8%) and 2 of 5 men (40%) for whom data
was primarily female (n = 40; 81.6%) and White (n = 46; were available had hypercalciuria at month 72. Among the 24
93.9%). At baseline, mean age was 48.1 ± 9.78 years, dur­ completers with hypercalciuria at baseline, 7 (29.2%) re­
ation of hypoparathyroidism was 15.9 ± 12.49 years and mained hypercalciuric at month 72 (data were missing for 3
55% (n = 27) of patients had a history of thyroid surgery. patients [12.5%]).
At baseline, mean 24-hour urinary calcium excretion was
10.3 ± 6.67 mmol/24 hours (412 ± 267 mg/24 hours) for
Biochemical Parameters men (n = 9) and 8.6 ± 4.60 mmol/24 hours (344 ± 184 mg/
Biochemical parameters are reported for the ITT population 24 hours) for women (n = 39). At month 72, mean urinary
(n = 49) and for patients who completed 72 months of treat­ calcium had declined to 6.9 ± 4.66 mmol/24 hours (276 ±
ment with rhPTH(1-84) (completer population; n = 37). 186 mg/24 hours) for men (n = 5) and 5.4 ± 2.93 mmol/
Overall, outcomes for these 2 study populations were similar. 24 hours (216 ± 117 mg/24 hours) for women (n = 27).
Mean albumin-adjusted serum calcium concentration was When adjusted for body weight, mean urine calcium excretion
within the target range at baseline and through month 72 at baseline was higher than desirable, 0.11 ± 0.061 mmol/kg/
for both the ITT and completer populations (Fig. 2A). 24 hours (4.3 ± 2.44 mg/kg/24 hours; n = 48), and gradually
We defined hypercalciuria as ≥6.25 mmol/24 hours decreased to 0.07 ± 0.039 mmol/kg/24 hours by month 72
(250 mg/24 hours) for women and ≥7.5 mmol/24 hours (2.7 ± 1.55 mg/kg/24 hours; n = 32) (Fig. 2B). Data for pa­
(300 mg/24 hours) for men. In the ITT population, for pa­ tients who completed the study were almost identical: 0.11
tients with available data, at baseline, 27 of 39 women ± 0.066 mmol/kg/24 hours (4.3 ± 2.64 mg/kg/24 hours; n = 37)
(69.2%) and 6 of 9 men (66.7%) had hypercalciuria; the pro­ at baseline and 0.07 ± 0.039 mmol/kg/24 hours (2.7 ±
portions had reduced by more than half by month 72 (n = 9/ 1.57 mg/kg/24 hours; n = 31) at month 72 (Fig. 2B).
27 [33.3%] for women and n = 2/5 [40.0%] for men, n = Twelve patients used thiazide diuretics. Mean serum cal­
11/32 [34%] overall). cium concentrations and mean 24-hour urinary calcium ex­
A similar pattern was observed in the completer population, cretion at baseline were higher in patients using thiazide
18 of the 28 women (64.3%) and 6 of the 9 men (66.7%) for diuretics than in patients not using them (Table 1). At month
Journal of the Endocrine Society, 2023, Vol. 7, No. 5 5

Downloaded from https://academic.oup.com/jes/article/7/5/bvad043/7103305 by guest on 09 January 2024


Figure 2. Change over time in (A) albumin-adjusted serum calcium, (B) 24-hour urinary calcium excretion corrected for body weight, (C) serum
phosphorus, and (D) 24-hour urinary phosphorus excretion for the ITT and completer populations. Completers are patients who completed 72 months of
treatment. ITT, intent to treat; rhPTH(1-84), recombinant human parathyroid hormone (1-84).

72, the absolute change from baseline in serum and urinary In concert with declines in serum phosphorus, mean cal­
calcium was larger in patients receiving than not receiving cium-phosphorus product also declined from baseline with
thiazide diuretics. rhPTH(1-84). Calcium-phosphorus product was 3.4 ±
At baseline, mean serum phosphorus concentration was 0.51 mmol2/L2 at baseline (n = 49) and decreased to 2.7 ±
1.6 ± 0.19 mmol/L (4.8 ± 0.58 mg/dL; n = 49), above the ref­ 0.41 mmol2/L2 at month 72 (n = 36). The mean change
erence range (0.81-1.45 mmol/L [2.5-4.5 mg/dL]). After initi­ from baseline in calcium-phosphorus product was −0.8 ±
ation of treatment with rhPTH(1-84), mean serum 0.50 mmol2/L2 at month 72 (n = 36). For the completer popu­
phosphorus levels declined and then remained at a lower level lation, calcium-phosphorus product was 3.5 ± 0.51 mmol2/L2
through month 72 (Fig. 2C). At month 72, mean change from at baseline (n = 35) and 2.7 ± 0.41 mmol2/L2 at month 72
baseline in serum phosphorus was −0.3 ± 0.22 mmol/L (−1.0 (n = 35).
± 0.70 mg/dL; n = 36). At baseline, 31 of 49 patients (63.3%)
were hyperphosphatemic (ie, serum phosphorus >1.45 mmol/L
[>4.5 mg/dL]). Of the 24 patients who were hyperphosphate­ Renal Function
mic at baseline and had month 72 data available, 18 (75%) Mean serum creatinine level was steady and remained within
had normal serum phosphorus and 6 (25%) had high serum the reference range throughout the study (Fig. 3). Mean
phosphorus at month 72. Mean 24-hour urinary phosphorus eGFR was stable over 72 months of treatment; mean values
level also trended downward from baseline over the course of were 78.1 ± 17.75 mL/min/1.73 m2 at baseline (n = 49) and
the study (Fig. 2D), consistent with a reduced renal filtered 81.5 ± 18.27 mL/min/1.73 m2 at month 72 (n = 38). For
load of phosphorus due to the decrease in serum phosphorus the completer population, mean eGFR was 75.16 ±
levels. 17.05 mL/min/1.73 m2 at baseline (n = 37) and 84.39 ±
6 Journal of the Endocrine Society, 2023, Vol. 7, No. 5

Table 1. Change in serum calcium and 24-hour urinary calcium excretion among patients using and not using thiazide diuretics

Parameter Baseline Month 72

n Value n Value

Serum calcium, mmol/L (mg/dL), mean ± SD


Patients receiving thiazide diuretics
Absolute value 12 2.3 ± 0.18 (9.2 ± 0.71) 11 2.1 ± 0.13 (8.4 ± 0.53)
Change from baseline − 11 −0.21 ± 0.232 (−0.84 ± 0.930)
Patients not receiving thiazide diuretics
Absolute value 37 2.1 ± 0.17 (8.5 ± 0.67) 26 2.2 ± 0.19 (8.6 ± 0.76)
Change from baseline − 26 0.02 ± 0.164 (0.08 ± 0.658)
24-hour urinary calcium, mmol (mg), mean ± SD

Downloaded from https://academic.oup.com/jes/article/7/5/bvad043/7103305 by guest on 09 January 2024


Patients receiving thiazide diuretics
Absolute value 12 10.2 ± 4.78 (409.2 ± 191.03) 10 6.4 ± 3.60 (254.3 ± 143.98)
Change from baseline − 10 −4.6 ± 4.91 (−181.90 ± 196.38)
Patients not receiving thiazide diuretics
Absolute value 36 8.5 ± 5.07 (339.2 ± 202.93) 22 5.3 ± 3.04 (212.0 ± 121.71)
Change from baseline − 22 −2.4 ± 5.39 (−94.0 ± 215.45)

Figure 3. Change over time in serum creatinine for the ITT and completer populations. Completers are patients who completed 72 months of treatment.
ITT, intent to treat; rhPTH(1-84), recombinant human parathyroid hormone (1-84).

19.00 mL/min/1.73 m2 at month 72 (n = 37). At baseline, 2 stage 3a, but no patients had progressed to CKD stage 3b,
of 41 patients (4.9%) had CKD stage 3a (eGFR 45-59 mL/ 4, or 5.
min/1.73 m2), and none had CKD stages 3b (eGFR 30−44
mL/min/1.73 m2), 4 (eGFR 15–29 mL/min/1.73 m2), or 5
(end-stage renal disease; eGFR <15 mL/min/1.73 m2 or dia­ Adverse Events
lysis). Both patients with CKD stage 3a at baseline completed Overall, TEAEs were reported in 48 patients (98.0%) and are
the study. At month 72, 4 of 38 patients (10.5%) had CKD summarized in Table 2. In addition, nephrolithiasis was
Journal of the Endocrine Society, 2023, Vol. 7, No. 5 7

Table 2. Treatment-emergent adverse events occurring in ≥20% of Based on patient diary data, 48 patients (98.0%) had ≥80%
patients treatment compliance during the first 12 months of the study,
as did 42 patients (85.7%) between 12 months and EOT.
TEAE, preferred term, n (%) Patients treated Events, n As shown in Fig. 4, doses of oral calcium and calcitriol sup­
with rhPTH(1-84)
plements were reduced rapidly during the first year of
(n = 49)
rhPTH(1-84) treatment. Thereafter, oral calcium supplement
Hypocalcemia 19 (38.8) 51 doses showed an upward trend from month 12 through
Muscle spasms 19 (38.8) 46 month 72, whereas calcitriol doses remained stable. The
Sinusitis 16 (32.7) 31 mean percentage change in oral calcium supplementation
was −69%±34.1% at month 12 (n = 46), and −45%
Paresthesia 15 (30.6) 38
±113.6% at month 72 (n = 37). The mean percentage change
Nausea 15 (30.6) 20
in calcitriol supplementation was −72%±44.6% at month 12
Headache 13 (26.5) 22 (n = 46) and −74%±39.3% at month 72 (n = 37).
Arthralgia 13 (26.5) 19 Reflecting the action of rhPTH(1-84) on the conversion of

Downloaded from https://academic.oup.com/jes/article/7/5/bvad043/7103305 by guest on 09 January 2024


Bronchitis 12 (24.5) 16 25(OH)D to 1,25(OH)2D, serum concentrations of
Nasopharyngitis 11 (22.4) 22 1,25(OH)2D increased over the course of the study. Mean se­
Back pain 11 (22.4) 16 rum 1,25(OH)2D concentration was 30.6 ± 12.21 pg/mL at
Pain in extremity 10 (20.4) 17
baseline (n = 29) and increased to 45.0 ± 25.94 pg/mL at
month 12 (n = 44), and 44.3 ± 17.37 pg/mL at month 72
Urinary tract infection 10 (20.4) 14
(n = 37). Mean change from baseline was 17.0 ± 29.62 pg/
Constipation 10 (20.4) 11 mL at month 12 (n = 27) and 11.6 ± 16.34 pg/mL at month
Diarrhea 10 (20.4) 11 72 (n = 24). Mean serum 25(OH)D concentration declined
Influenza 10 (20.4) 11 from a baseline value of 45.7 ± 16.86 ng/mL (n = 49) to
33.4 ± 10.24 ng/mL at week 24 (n = 47) and then remained
Abbreviations: rhPTH(1-84), recombinant human parathyroid hormone relatively stable over the remainder of the study (Fig. 5). Per
(1-84); TEAE, treatment-emergent adverse event.
protocol, supplements of ergocalciferol and cholecalciferol
could be altered at any time during the study, which would
likely influence the serum 25(OH)D results; however, data re­
reported in 6 patients (12.2%); nephrocalcinosis was not re­ garding changes in doses of these supplements are not
ported. TEAEs assessed by the investigator as related to treat­ available.
ment with rhPTH(1-84) were reported in 25 patients (51.0%);
all were mild or moderate in severity except for 1 event of se­
Composite Efficacy Outcome
vere lower back pain. The most common TEAEs that were
considered to be related to treatment were nausea (n = 7, The composite efficacy outcome was defined as a ≥50% re­
14.3%), hypercalcemia (n = 6, 12.2%), hypocalcemia (n = 5, duction in oral calcium (or ≤500 mg/day) and calcitriol (or
10.2%), tetany (n = 4, 8.2%), constipation (n = 3, 6.1%), ≤0.25 μg/day) doses and albumin-adjusted serum calcium
and paresthesia (n = 3, 6.1%). Thirty serious TEAEs were re­ concentration that were normalized or maintained compared
ported in 13 patients (26.5%); none were reported in more with baseline and did not exceed the upper limit of normal.
than 1 patient, and none were assessed by the investigator as The composite efficacy outcome was achieved by 35 of 46 pa­
related to treatment with rhPTH(1-84). Nine events of bone tients (76.1%; 95% CI 61.2-87.4%) at month 12 and by 25 of
fracture were reported in 8 patients during the study: 3 events 38 patients (65.8%; 95% CI 48.6-80.4%) at month 72. The
of ankle fracture; 1 event each of fibula, radius, rib, thumb, composite efficacy response rate was similar regardless of
and ulna fracture; and 1 event of stress fracture of the foot. the source of the oral calcium supplementation. Among pa­
None of the fracture events were considered by the investiga­ tients who were prescribed calcium as calcium citrate, the
tor to be related to treatment. No cases of osteosarcoma were composite efficacy response rate was 75.0% at month 12
reported. Two deaths occurred during the study; neither was (n = 21 of 28 patients) and 75.0% at month 72 (n = 18 of
considered by the investigator to be related to rhPTH(1-84) 24 patients). Among patients who were prescribed calcium
treatment. As reported previously, a 52-year-old woman as calcium carbonate, the composite efficacy response rate
with a history of hyperlipidemia, diabetes mellitus type 2, car­ was 77.8% at month 12 (n = 7 of 9 patients) and 66.7% at
diomyopathy, and hypertension died of congestive heart fail­ month 72 (n = 4 of 6 patients).
ure [22]. In addition, a 65-year-old woman with a history of
beta-thalassemia and mitral valve prolapse died from cardiac Bone Turnover Markers, Bone Mineral Density, and
arrest. There were no clinically significant changes in vital Z Score
signs or electrocardiogram parameters over the treatment For both the ITT and completer analysis populations, levels of
period. BTMs initially increased with rhPTH(1-84) treatment and
then declined by month 32 to steady-state values that re­
mained above pretreatment levels (Fig. 6). For the ITT popu­
rhPTH(1-84) Dosing, Oral Supplement Dosing, and lation, mean percentage change in BMD at month 72 was less
Serum 25(OH)D and 1,25(OH)2D Levels at lumbar spine (−1.9% ± 9.30%; n = 32), total hip (−2.8%
Most patients (n = 46; 93.9%) initiated the study on 50 µg per ± 7.09%; n = 31), and femoral neck (−3.1% ± 8.01%; n =
day of rhPTH(1-84), but by month 72, most (n = 26/37; 31) than at one-third radius (−7.5% ± 9.04%; n = 32), which
70.3%) were receiving 100 µg per day of rhPTH(1-84). declined progressively from baseline (Fig. 7). Baseline mean z
Administration of rhPTH(1-84) was generally well tolerated. scores were well above average at all sites (lumbar spine, +2.1
8 Journal of the Endocrine Society, 2023, Vol. 7, No. 5

Downloaded from https://academic.oup.com/jes/article/7/5/bvad043/7103305 by guest on 09 January 2024


Figure 4. Percentage change from baseline in dose of oral calcium and calcitriol over time for the ITT and completer populations. Completers are patients
who completed 72 months of treatment. ITT, intent to treat; rhPTH(1-84), recombinant human parathyroid hormone (1-84). *n = 38 for the overall study
(ITT) population.

± 1.47 [n = 40]; total hip, +1.6 ± 1.12 [n = 40]; femoral neck, efficacy endpoint and rhPTH(1-84) dosing values at EOT
+1.4 ± 1.24 [n = 40]; one-third radius, +0.9 ± 0.83 [n = 40]). may be less robust than values at month 72 because patients
At month 72, mean z scores were slightly better than baseline could have discontinued treatment before escalation to full
in the lumbar spine and stable at the total hip and femoral dose occurred.
neck. Mean change of z score at the one-third radius was a de­
crease of 0.5 z score units (Fig. 7). Similar results were ob­
tained when BMD and z scores were determined for the Discussion
completer population (Fig. 7). Outcomes of this study confirm and extend results of previous
analyses of the safety and efficacy of rhPTH(1-84) for chronic
hypoparathyroidism and demonstrate that beneficial treat­
Summary of Efficacy Endpoints, rhPTH(1-84) ment effects were maintained over 6 years. All but 1 study pa­
Dosing, Biochemical Parameters, and Bone Indices tient (48 of 49) had chronic hypoparathyroidism that was not
Data collected at the last study visit (ie, at EOT) for study out­ adequately controlled at baseline of RACE, as defined by key
comes reported in the ITT population were collated and are biochemical measurements. Over 72 months of treatment
summarized in Tables 3 and 4. At EOT, the composite efficacy with rhPTH(1-84), biochemical parameters improved or
outcome was achieved by 30 of 49 patients (61.2%; 95% CI were maintained within reference ranges, and mean urinary
46.2–74.8%), which was similar to the response rate at month calcium excretion decreased to within the reference range
72. In addition, there was no substantial difference in the re­ for men and women. These outcomes demonstrate that
sponse rate between patients prescribed calcium citrate and patients treated long-term with rhPTH(1-84) can achieve im­
those prescribed calcium carbonate (Table 3). At EOT, 30 portant management goals for hypoparathyroidism as out­
of 49 patients (61.2%) were receiving 100 µg per day of lined in treatment guidelines [4, 5, 26]. The safety profile of
rhPTH(1-84) as their final dose level. Results for composite rhPTH(1-84) was consistent with results of the shorter-term
Journal of the Endocrine Society, 2023, Vol. 7, No. 5 9

Downloaded from https://academic.oup.com/jes/article/7/5/bvad043/7103305 by guest on 09 January 2024


Figure 5. Change over time in serum 25(OH)D for the ITT and completer populations. Completers are patients who completed 72 months of treatment.
ITT, intent to treat; rhPTH(1-84), recombinant human parathyroid hormone (1-84).

antecedent studies REPLACE and RELAY, as well as the 72 months, although considerable interpatient variability
5-year analysis of data from RACE, and another long-term, was observed. Among patients who had hypercalciuria at
single-center study of rhPTH(1-84) [17, 18, 22, 24]. No cases baseline, 39.4% achieved normal urine calcium excretion at
of osteosarcoma were reported during the study, and no new month 72. Similar beneficial effects on urinary calcium excre­
safety signals were identified. Although a small number of tion and preservation of renal function were also seen in an­
fractures occurred, none were considered related to other open-label, long-term extension study of rhPTH(1-84)
rhPTH(1-84). Overall, 76% of patients completed 72 months [20]. Furthermore, in retrospective analyses, patients treated
of rhPTH(1-84) treatment, and 86% maintained ≥80% treat­ with rhPTH(1-84) in clinical trials (including patients from
ment compliance between month 12 through EOT, indicating RACE) had a lower annual rate of eGFR decline and a lower
that the dosing regimen can be sustained over long-term risk of developing CKD compared with a historical control co­
treatment. hort not treated with rhPTH(1-84) [34-36]. Cumulatively,
Based on several retrospective studies, chronic hypopara­ these data suggest that therapy with rhPTH(1-84) may miti­
thyroidism has been associated with an increased risk of renal gate some of the risks of adverse renal outcomes for patients
complications, including nephrolithiasis, nephrocalcinosis, with chronic hypoparathyroidism. Nonetheless, for patients
eGFR decline, and CKD [10, 15, 27-29]. In RACE, renal func­ with hypercalciuria, nephrocalcinosis, and/or kidney stones,
tion was preserved throughout the 6 years of treatment with it would be advisable to follow 24-hour urine calcium excre­
rhPTH(1-84). Mean serum creatinine and eGFR were stable, tion during rhPTH(1-84) treatment.
and no patients experienced substantial eGFR decreases, end- Mean serum phosphorus level was above the upper limit of
stage renal disease, or the need for renal replacement therapy. the reference range at baseline, consistent with the absence of
The prevalence of nephrolithiasis in this study (12.2%) was the phosphaturic effect of endogenous PTH [9]. With
within the wide range (0-35.5%) reported in the literature rhPTH(1-84) treatment, serum phosphorus declined to within
for patients with chronic hypoparathyroidism [29-31]. In add­ the reference range by week 4 and was maintained at lower
ition, no events of nephrocalcinosis were reported; however, levels throughout the remainder of the study. Notably, the de­
routine ultrasound imaging was not mandated in the protocol, crease in serum phosphorus was not accompanied by a corre­
therefore the rate of asymptomatic nephrocalcinosis could not sponding increase in urine phosphorus, possibly because of
be ascertained. Furthermore, hypocitraturia, which was not the establishment of a new rhPTH(1-84)-driven steady state
recorded in this study, has been associated with renal stone in which the renal filtered load of phosphorus was reduced,
formation in patients with hyperparathyroidism [32] and or to individual dietary changes in phosphorus content not
has been reported in patients with hypoparathyroidism who captured in the trial. It is also possible that the daily injection
received synthetic rhPTH (rhPTH[1-34]) [33]. Mean of rhPTH(1-84), by promoting increased renal clearance of
24-hour urinary calcium levels were reduced from above the phosphorus during the initial hours after an injection (when
reference range at baseline to within the normal range over the circulating levels of PTH are high), reduces serum
10 Journal of the Endocrine Society, 2023, Vol. 7, No. 5

Downloaded from https://academic.oup.com/jes/article/7/5/bvad043/7103305 by guest on 09 January 2024


Figure 6. Change over time in serum bone turnover markers (A) BAP, (B) P1NP, and (C) CTX for the ITT and completer populations. Completers are
patients who completed 72 months of treatment. BAP, bone alkaline phosphatase; CTX, cross-linked C-telopeptide of type 1 collagen; ITT, intent to treat;
P1NP, aminoterminal propeptide of type 1 collagen; rhPTH(1-84), recombinant human parathyroid hormone (1-84).

phosphorus levels sufficiently that the overall renal filtered In this study, long-term daily treatment with rhPTH(1-84)
load of phosphorus is decreased over the 24-hour treatment improved important measures of bone health for patients
period, as was seen in the pharmacokinetics/pharmacodynam­ with hypoparathyroidism. As expected, BTMs were in the
ics study by Clarke et al [37]. In addition, treatment with lower part of the reference range at baseline because of low
rhPTH(1-84) may help restore the complex series of interac­ bone turnover in this condition [1]. With rhPTH(1-84) treat­
tions among PTH, fibroblast growth factor 23, and ment, bone formation and resorption markers increased in
1,25(OH)2D that together act to regulate serum and urine parallel, peaked at 9 to 16 months, and then declined by
phosphorus levels [38, 39]. Indeed, we note that during the month 32 but remained above pretreatment values at a
study, serum concentrations of 1,25(OH)2D increased while new steady state that continued through 72 months of ther­
serum concentrations of 25(OH)D decreased, a physiological apy. Rubin and colleagues reported a similar pattern of
response that is typical of PTH exposure [40]. This effect is the change in bone formation and resorption markers in another
result of induction of CYP27B1 by PTH, which increases con­ long-term study of rhPTH(1-84) in patients with chronic
version of 25(OH)D to 1,25(OH)2D, and induction of hypoparathyroidism [24]. The time course of changes in
CYP24A1 by 1,25(OH)2D, which increases inactivation of BTM levels with rhPTH(1-84) treatment may have impacted
25(OH)D [40, 41]. patients’ oral supplement requirements. As BTM levels
Journal of the Endocrine Society, 2023, Vol. 7, No. 5 11

Downloaded from https://academic.oup.com/jes/article/7/5/bvad043/7103305 by guest on 09 January 2024


Figure 6. Continued

decline and less calcium is released into the blood in the latter postmenopausal women in Norway, elevated endogenous
part of the rhPTH(1-84) treatment period, dosages of oral PTH concentration was associated with increased cortical
calcium were increased. porosity in the proximal femur [45], while iliac crest bone bi­
It is important that the long-term implications of PTH re­ opsy data from a study of 5 patients with hypoparathyroid­
placement on bone are well established. A case study regard­ ism showed a significant increase in cortical porosity after 12
ing a patient with hypoparathyroidism treated with months of treatment with rhPTH(1-34), with no correspond­
rhPTH(1-34) for 3 years reported upregulated bone uptake ing increase in cortical thickness [46]. The modest change in z
in the peripheral joints and in the axial skeleton [42], and score (−0.5) observed in this study; however, suggests that
an increase in BMD was reported in a patient with autosomal most of the decrease at one-third radius observed in this
dominant hypoparathyroidism secondary to calcium recep­ study was expected. It should be noted that the patients in
tor mutation who received rhPTH(1-34) for 13.5 years this study had longstanding hypoparathyroidism, and their
[43]. Furthermore, diffuse calvarial thickening was observed BMD was better than expected for age, race, and sex.
in a patient with chronically elevated PTH levels, with the ac­ Furthermore, the radius is a known site of catabolic actions
tivation of cyclic adenosine monophosphate signaling path­ of PTH [47] and a site where endosteal resorption and perios­
ways proposed as a plausible mechanism [44]. Although teal apposition of new bone could maintain skeletal strength
BMD decreased at all sites measured over the course of the despite the decline in BMD. Further and more detailed
study, changes in the spine and hip were no more than ex­ assessments of this site would shed light on those possibilities.
pected for age, race, and sex. BMD decline was somewhat BMD changes over time at the spine and hip were much
greater at the one-third radius. At least part of the decrease smaller quantitatively and are unlikely to be clinically con­
at the one-third radius was due to treatment. In a study of cerning in the majority of patients. Moreover, rhPTH(1-84)
12 Journal of the Endocrine Society, 2023, Vol. 7, No. 5

Downloaded from https://academic.oup.com/jes/article/7/5/bvad043/7103305 by guest on 09 January 2024


Figure 7. Change from baseline in z score over time. Filled symbols with solid lines represent the ITT population, and open symbols with dashed lines
represent the completer population. The table below the graph reports percentage change from baseline in BMD at month 72 and z score at baseline and
month 72. Completers are patients who completed 72 months of treatment. BMD, bone mineral density; ITT, intent to treat; rhPTH(1-84), recombinant
human parathyroid hormone (1-84). *Excludes results from 6 patients who were assessed by different dual-energy x-ray absorptiometry machines at
baseline vs month 72 (Hologic and GE Lunar instruments, respectively).

Table 3. Summary of efficacy endpoints and rhPTH(1-84) dosing (ITT population))

Parameter rhPTH(1-84)
(n = 49)

Composite efficacy outcome n Value at EOT


Composite efficacy outcome, % (95% CI) 30 61.2 (46.2-74.8)
Patients prescribed calcium citratea 20 69.0 (49.2-84.7)
Patients prescribed calcium carbonateb 7 63.3 (30.8-89.1)
rhPTH(1-84) dose n Value at EOT
rhPTH(1-84) dose level, %
25 µg 1 2.0
50 µg 10 20.4
75 µg 8 16.3
100 µg 30 61.2

Abbreviations: EOT, end of treatment; ITT, intent to treat; rhPTH(1-84), recombinant human parathyroid hormone (1-84).
a
Among 29 patients.
b
Among 11 patients.

administration in hypoparathyroidism is known to affect in life may already have low BMD or osteoporosis by the
skeletal microstructure in ways that could mitigate concerns time they develop hypoparathyroidism. How treatment
that are typically associated with reductions in BMD [41]. with rhPTH(1-84) would affect BMD in these situations is
However, patients who develop hypoparathyroidism later unknown.
Journal of the Endocrine Society, 2023, Vol. 7, No. 5 13

Table 4. Summary of biochemical parameters, oral supplementation, and bone indices (ITT population)

Biochemical parameters

Parameter, mean ± SD Baseline EOT

n Value n Value

Serum calcium, mmol/L (mg/dL) 49 2.1 ± 0.17 (8.4 ± 0.70) 49 2.0 ± 0.18 (8.1 ± 0.71)
Change from baseline, mmol/L (mg/dL) − 49 −0.1 ± 0.19 (−0.2 ± 0.75)
Change from baseline, % − 49 −2.5 ± 8.90
Patients receiving thiazide diuretics
Absolute value, mmol/L (mg/dL) 12 2.3 ± 0.18 (9.2 ± 0.71) 12 2.1 ± 0.13 (8.4 ± 0.51)
Change from baseline, mmol/L (mg/dL) − 12 −0.2 ± 0.19 (−0.9 ± 0.78)

Downloaded from https://academic.oup.com/jes/article/7/5/bvad043/7103305 by guest on 09 January 2024


Patients not receiving thiazide diuretics
Absolute value, mmol/L (mg/dL) 37 2.1 ± 0.17 (8.5 ± 0.67) 37 2.1 ± 0.21 (8.4 ± 0.84)
Change from baseline, mmol/L (mg/dL) − 37 −0.04 ± 0.200 (−0.2 ± 0.80)
Urinary calcium, mmol/24 hours (mg/24 hours) 48 8.9 ± 5.01 (356.7 ± 200.37) 48 6.5 ± 3.71 (258.2 ± 148.23)
Change from baseline, mmol/24 hours (mg/24 hours) − 47 −2.4 ± 5.81 (−94.7 ± 232.36)
Patients receiving thiazide diuretics
Absolute value 12 10.2 ± 4.78 (409.2 ± 191.03) 12 6.2 ± 2.24 (246.5 ± 89.56)
Change from baseline − 12 −4.1 ± 6.00 (−162.7 ± 240.00)
Patients not receiving thiazide diuretics
Absolute value 36 8.5 ± 5.07 (339.2 ± 202.93) 36 6.6 ± 4.10 (262.1 ± 164.08)
Change from baseline − 35 −1.8 ± 5.71 (−71.4 ± 228.53)
Serum phosphorus, mmol/L (mg/dL) 49 1.6 ± 0.19 (4.8 ± 0.58) 49 1.3 ± 0.24 (4.0 ± 0.74)
Change from baseline, mmol/L (mg/dL) − 49 −0.3 ± 0.29 (−0.8 ± 0.91)
Change from baseline, % − 49 −15.2 ± 18.90
Urinary phosphorus, mmol/24 hours (mg/24 hours) 48 31.9 ± 14.50 (1.0 ± 0.45) 48 24.6 ± 10.80 (0.8 ± 0.33)
Change from baseline, mmol (mg/dL) − 47 −7.6 ± 14.14 (−0.2 ± 0.44)
Change from baseline, % − 47 −13.8 ± 44.32
Creatinine, µmol/L (mg/dL) 49 84.7 ± 18.16 (1.0 ± 0.21) 48 79.8 ± 21.60 (0.9 ± 0.24)
Change from baseline, µmol/L (mg/dL) − 48 −5.3 ± 16.88 (−0.1 ± 0.19)
Change from baseline, % − 48 −5.6 ± 19.60
eGFR, mL/min/1.73 m2 49 78.1 ± 17.75 48 80.3 ± 20.55
25(OH)D, ng/mL 49 45.7 ± 16.86 49 38.2 ± 10.58
Change from baseline, ng/mL − 49 −7.6 ± 18.53
1,25(OH)2D, pg/mL 29 30.6 ± 12.21 46 36.2 ± 13.66
Change from baseline, pg/mL − 28 6.4 ± 17.62

Oral supplementation

Baseline EOT

Parameter, mean ± SDa n Value n Value

Oral calcium, mg/day 49 2194 ± 1732.3 49 1032 ± 1352.8


Change from baseline, % − 49 −40.4 ± 106.98
Oral calcitriol, µg/day 49 0.67 ± 0.446 49 0.18 ± 0.414
Change from baseline, % − 49 −73.7 ± 39.35
Supplement use at EOT, %
Patients off oral calcium − 14 28.6
Patients off calcitriol − 32 65.3
Patients with oral calcium <600 mg/day and off calcitriol − 19 38.8
Patients off oral calcium and calcitriol − 10 20.4

(continued)
14 Journal of the Endocrine Society, 2023, Vol. 7, No. 5

Table 4. Continued

Bone indices

Baseline EOT

Parameter, mean ± SD n Value n Value

Bone indices

Baseline EOT

Parameter, mean ± SD n Value n Value

BAP, µg/L 48 9.6 ± 3.32 48 14.6 ± 11.54

Downloaded from https://academic.oup.com/jes/article/7/5/bvad043/7103305 by guest on 09 January 2024


Change from baseline, µg/L − 47 5.3 ± 11.16
P1NP, µg/L 49 33.7 ± 19.72 48 124.9 ± 104.44
Change from baseline, µg/L − 48 91.9 ± 104.33
CTX, ng/L 48 213.0 ± 172.34 48 422.8 ± 282.83
Change from baseline, ng/L − 47 224.2 ± 295.86
BMD, change from baseline, %b
Lumbar spine − 39 −2.2 ± 8.83
Total hip − 39 −2.9 ± 6.55
Femoral neck − 39 −4.4 ± 7.76
One-third radius − 39 −6.9 ± 9.94
z scoreb
Lumbar spine 40 2.1 ± 1.47 42 2.2 ± 1.56
Total hip 40 1.6 ± 1.12 42 1.6 ± 1.10
Femoral neck 40 1.5 ± 1.24 42 1.4 ± 1.17
One-third radius 40 0.9 ± 0.83 42 0.5 ± 1.22

Abbreviations: BAP, bone alkaline phosphatase; BMD, bone mineral density; CTX, cross-linked C-telopeptide of type 1 collagen; eGFR, estimated glomerular
filtration rate; EOT, end of treatment; ITT, intent to treat; P1NP, aminoterminal propeptide of type 1 collagen; rhPTH(1-84), recombinant human parathyroid
hormone (1-84).
a
Unless otherwise indicated.
b
Excludes results from 6 patients who were assessed by different dual-energy x-ray absorptiometry machines at baseline vs month 72 (Hologic and GE Lunar
instruments, respectively).

In contrast to the decline in BMD at all sites in the current or BTMs were collected at prescribed times or under fasting
study, another long-term study of rhPTH(1-84)-treated pa­ conditions, which may limit data interpretation.
tients with hypoparathyroidism by Tay et al showed an in­
crease in BMD from baseline at the lumbar spine, total hip,
and femoral neck [20]. In both the current study and the Conclusions
Tay et al study, BMD decreased at the one-third radius, but Continuous use of rhPTH(1-84) over 6 years is an effective
the magnitude of the decline was less in the analysis by Tay long-term treatment for chronic hypoparathyroidism and
et al [20]. Patients in the study by Tay et al had a longer dur­ has a safety profile consistent with previous studies.
ation of hypoparathyroidism (29.9 ± 3.3 years for patients in Treatment with rhPTH(1-84) permitted reductions in oral
Tay et al vs 15.9 ± 12.5 years in RACE) and used a lower dose calcium and calcitriol, while albumin-adjusted serum cal­
of rhPTH(1-84) (50 µg daily was the most frequent dose in the cium was maintained in the target range. Improvements in
Tay study vs 100 µg daily in RACE) [20], either of which efficacy-related biochemical parameters were sustained, re­
could affect the BMD response to rhPTH(1-84) treatment. nal function was preserved, and mean urinary calcium excre­
Together, the available bone-related data suggest that long- tion declined to within the reference ranges for men and
term rhPTH(1-84) treatment may help restore the skeleton women.
closer to the euparathyroid state without an associated in­
crease in fracture risk.
A major strength of this study was that the cohort of patients
was followed longitudinally for 6 years, with few discontinua­ Acknowledgments
tions due to TEAEs. Long-term follow-up enabled the collec­ Under the direction of the authors, editorial support in the
tion of an extensive biochemical and safety data set for this preparation of this manuscript was provided by Alan Storey,
rare disease population. Limitations include the open-label de­ PhD, an employee of ICON (Reading, Berkshire, UK), and
sign and lack of control cohort. In addition, the protocol did funded by Takeda Development Center Americas Inc.,
not specify that serum samples for 25(OH)D, 1,25(OH)D2, Lexington, MA, USA.
Journal of the Endocrine Society, 2023, Vol. 7, No. 5 15

Funding 2. Bilezikian JP, Brandi ML, Cusano NE, et al. Management of hypo­
parathyroidism: present and future. J Clin Endocrinol Metab.
The clinical trial was funded by Takeda Pharmaceuticals 2016;101(6):2313-2324.
U.S.A., Lexington, MA, USA. 3. Shoback DM, Bilezikian JP, Costa AG, et al. Presentation of hypo­
parathyroidism: etiologies and clinical features. J Clin Endocrinol
Metab. 2016;101(6):2300-2312.
Disclosures 4. Bollerslev J, Rejnmark L, Marcocci C, et al. European Society of
N.B.W. has served as research investigator and speaker for Endocrinology clinical guideline: treatment of chronic hypopara­
thyroidism in adults. Eur J Endocrinol. 2015;173(2):G1-G120.
and received honoraria and research support from Takeda
5. Brandi ML, Bilezikian JP, Shoback D, et al. Management of hypo­
Pharmaceuticals USA, Inc. J.P.B. has served as advisory board
parathyroidism: summary statement and guidelines. J Clin
member, research investigator, and speaker for and has re­ Endocrinol Metab. 2016;101(6):2273-2283.
ceived consulting fees, research support, and honoraria from 6. Bollerslev J, Rejnmark L, Zahn A, et al. European Expert consensus
Takeda Pharmaceuticals USA, Inc. H.G.B. has served as re­ on practical management of specific aspects of parathyroid disor­
search investigator for and received research support from ders in adults and in pregnancy: recommendations of the ESE edu­

Downloaded from https://academic.oup.com/jes/article/7/5/bvad043/7103305 by guest on 09 January 2024


Takeda Pharmaceuticals USA, Inc. and has served as consult­ cational program of parathyroid disorders. Eur J Endocrinol.
ant for Entera Bio and BridgeBio Pharma. B.L.C. has served 2021;186(2):R33-R63.
as consultant and research investigator for Takeda 7. Rubin MR. Recent advances in understanding and managing hypo­
Pharmaceuticals USA, Inc., and has received research grant parathyroidism. F1000Res. 2020;9:766.
funding from Takeda Pharmaceuticals USA, Inc., Ascendis 8. Babey M, Brandi ML, Shoback D. Conventional treatment of hypo­
parathyroidism. Endocrinol Metab Clin North Am. 2018;47(4):
Pharma, and Chugai Pharmaceutical Co., Ltd. D.D. has re­
889-900.
ceived research support from Takeda Pharmaceuticals USA, 9. Shoback D. Hypoparathyroidism. N Engl J Med. 2008;359(4):
Inc. MAL has served as advisory board member, consultant, 391-403.
and research investigator for Takeda Pharmaceuticals USA, 10. Mitchell DM, Regan S, Cooley MR, et al. Long-term follow-up of
Inc. and as a consultant for Ascendis Pharma and Calcilytix. patients with hypoparathyroidism. J Clin Endocrinol Metab.
M.M. has served as an advisory board member, consultant, 2012;97(12):4507-4514.
and research investigator for Takeda Pharmaceuticals USA, 11. Underbjerg L, Sikjaer T, Rejnmark L. Long-term complications in
Inc.; as a consultant and research investigator for Chugai patients with hypoparathyroidism evaluated by biochemical find­
Pharmaceutical Co., Ltd; and as a consultant for ings: a case-control study. J Bone Miner Res. 2018;33(5):822-831.
Amolyt Pharma. M.P. has received research support from 12. Abate EG, Clarke BL. Review of hypoparathyroidism. Front
Endocrinol (Lausanne). 2017;7:172.
Takeda Pharmaceuticals USA, Inc. J.G.R. has served as a re­
13. Goswami R, Sharma R, Sreenivas V, Gupta N, Ganapathy A, Das S.
search investigator for and received research support from
Prevalence and progression of basal ganglia calcification and its
Takeda Pharmaceuticals, USA, Inc. T.J.V. has served as advis­ pathogenic mechanism in patients with idiopathic hypoparathyr­
ory board member, consultant, and research investigator for oidism. Clin Endocrinol. 2012;77(2):200-206.
and received consulting fees and research support from 14. David K, Moyson C, Vanderschueren D, Decallonne B. Long-term
Takeda Pharmaceuticals USA, Inc. M.L.W. has served as complications in patients with chronic hypoparathyroidism: a
speaker for and has received honoraria and research support cross-sectional study. Eur J Endocrinol. 2019;180(1):71-78.
from Takeda Pharmaceuticals USA, Inc. S.Y. is an employee 15. Underbjerg L, Sikjaer T, Mosekilde L, Rejnmark L. Cardiovascular
and N.S. is a former employee of Takeda Pharmaceuticals and renal complications to postsurgical hypoparathyroidism: a
USA, Inc., Lexington, MA, USA. D.M.S. has served as advis­ Danish nationwide controlled historic follow-up study. J Bone
ory board member, consultant, and research investigator for Miner Res. 2013;28(11):2277-2285.
16. Kim SH, Rhee Y, Kim YM, et al. Prevalence and complications of
and received consulting fees and research support from
nonsurgical hypoparathyroidism in Korea: a nationwide cohort
Takeda Pharmaceuticals USA, Inc. and has received institu­ study. PLoS One. 2020;15(5):e0232842.
tional research support from and served as a consultant for 17. Bilezikian JP, Clarke BL, Mannstadt M, et al. Safety and efficacy of
Ascendis Pharma. recombinant human parathyroid hormone in adults with hypo­
parathyroidism randomly assigned to receive fixed 25-μg or 50-μg
daily doses. Clin Therap. 2017;39(10):2096-2102.
Data Availability 18. Mannstadt M, Clarke BL, Vokes T, et al. Efficacy and safety of re­
Some or all data sets generated during and/or analyzed during combinant human parathyroid hormone (1-84) in hypoparathyr­
oidism (REPLACE): a double-blind, placebo-controlled,
the current study are not publicly available but are available
randomized, phase 3 study. Lancet Diabetes Endocrinol.
from the corresponding author on reasonable request.
2013;1(4):275-283.
19. Sikjaer T, Rejnmark L, Rolighed L, Heickendorff L, Mosekilde L,
Hypoparathyroid Study Group. The effect of adding PTH(1-84)
Clinical Trial Information to conventional treatment of hypoparathyroidism: a randomized,
NCT01297309, registered February 11, 2011. placebo-controlled study. J Bone Miner Res. 2011;26(10):
2358-2370.
20. Tay Y-KD, Tabacco G, Cusano NE, et al. Therapy of hypopara­
Current Affiliation thyroidism with rhPTH(1-84): a prospective, 8–year investigation
of efficacy and safety. J Clin Endocrinol Metab. 2019;104(11):
Nicole Sherry, Freeline Therapeutics, Boston, MA, USA. 5601-5610.
21. Clarke BL, Vokes TJ, Bilezikian JP, Shoback DM, Lagast H,
Mannstadt M. Effects of parathyroid hormone rhPTH(1-84) on
References phosphate homeostasis and vitamin D metabolism in hypopara­
1. Bilezikian JP. Hypoparathyroidism. J Clin Endocrinol Metab. thyroidism: REPLACE phase 3 study. Endocrine. 2017;55(1):
2020;105(6):1722-1736. 273-282.
16 Journal of the Endocrine Society, 2023, Vol. 7, No. 5

22. Mannstadt M, Clarke BL, Bilezikian JP, et al. Safety and efficacy of 35. Chen KS, Gosmanova EO, Curhan GC, et al. Five-year estimated
5 years of treatment with recombinant human parathyroid hor­ glomerular filtration rate in patients with hypoparathyroidism
mone in adults with hypoparathyroidism. J Clin Endocrinol treated with and without rhPTH(1-84). J Clin Endocrinol Metab.
Metab. 2019;104(11):5136-5147. 2020;105(10):e3557-e3565.
23. Cusano NE, Rubin MR, McMahon DJ, et al. Therapy of hypopara­ 36. Rejnmark L, Ayodele O, Lax A, Mu F, Swallow E, Gosmanova EO.
thyroidism with PTH(1-84): a prospective four-year investigation The risk of chronic kidney disease development in adult patients
of efficacy and safety. J Clin Endocrinol Metab. 2013;98(1): with chronic hypoparathyroidism treated with rhPTH(1-84): a
137-144. retrospective cohort study. Clin Endocrinol (Oxf). 2023;98(4):
24. Rubin MR, Cusano NE, Fan W-W, et al. Therapy of hypoparathyr­ 496-504.
oidism with PTH(1-84): a prospective six year investigation of effi­ 37. Clarke BL, Berg JK, Fox J, Cyran JA, Lagast H. Pharmacokinetics
cacy and safety. J Clin Endocrinol Metab. 2016;101(7):2742-2750. and pharmacodynamics of subcutaneous recombinant parathyroid
25. Levey AS, Stevens LA, Schmid CH, et al. A new equation to estimate hormone (1-84) in patients with hypoparathyroidism: an open-
glomerular filtration rate. Ann Intern Med. 2009;150(9):604-612. label, single-dose, phase I study. Clin Therap. 2014;36(5):722-736.
26. Khan AA, Koch CA, Van Uum S, et al. Standards of care for hypo­ 38. Bergwitz C, Juppner H. Regulation of phosphate homeostasis by
parathyroidism in adults: a Canadian and international consensus. PTH, vitamin D, and FGF23. Annu Rev Med. 2010;61(1):91-104.

Downloaded from https://academic.oup.com/jes/article/7/5/bvad043/7103305 by guest on 09 January 2024


Eur J Endocrinol. 2019;180(3):P1-P22. 39. Peacock M. Hypoparathyroidism and the kidney. Endocrinol
27. Underbjerg L, Sikjaer T, Mosekilde L, Rejnmark L. The epidemi­ Metab Clin North Am. 2018;47(4):839-853.
ology of nonsurgical hypoparathyroidism in Denmark: a nation­ 40. Goltzman D, Mannstadt M, Marcocci C. Physiology of the
wide case finding study. J Bone Miner Res. 2015;30(9):1738-1744.
calcium-parathyroid hormone-vitamin D axis. Front Horm Res.
28. Gosmanova EO, Chen K, Rejnmark L, et al. Risk of chronic kidney
2018;50:1-13.
disease and estimated glomerular filtration rate decline in patients
41. Christakos S, Li S, De La Cruz J, Bikle DD. New developments in
with chronic hypoparathyroidism: a retrospective cohort study.
our understanding of vitamin metabolism, action and treatment.
Adv Ther. 2021;38(4):1876-1888.
Metabolism. 2019;98:112-120.
29. Ketteler M, Chen K, Gosmanova EO, et al. Risk of nephrolithiasis
42. Goujard C, Salenave S, Briot K, Chanson P, Grimon G, Kamenicky
and nephrocalcinosis in patients with chronic hypoparathyroidism:
P. Treating hypoparathyroidism with recombinant human parathy­
a retrospective cohort study. Adv Ther. 2021;38(4):1946-1957.
roid hormone (1-34): long-term safety concerns. Lancet. 2020;395­
30. Gosmanova EO, Houillier P, Rejnmark L, Marelli C, Bilezikian JP.
(10232):1304.
Renal complications in patients with chronic hypoparathyroidism
43. Theman TA, Collins MT, Dempster DW, et al. PTH(1-34) replace­
on conventional therapy: a systematic literature review: renal dis­
ease in chronic hypoparathyroidism. Rev Endocr Metab Disord. ment therapy in a child with hypoparathyroidism caused by a spor­
2021;22(2):297-316. adic calcium receptor mutation. J Bone Miner Res. 2009;24(5):
31. Ridder LO, Harslof T, Sikjaer T, Underbjerg L, Rejnmark L. 964-973.
Determinants of hypercalciuria and renal calcifications in chronic 44. Theng EH, Weinstein LS, Collins MT. Calvarial hyperostosis in pri­
hypoparathyroidism: a cross-sectional study. Clin Endocrinol mary hyperparathyroidism and other settings of increased cAMP
(Oxf). 2021;95(2):286-294. signalling. Lancet. 2022;399(10328):956.
32. Alvarez-Arroyo MV, Traba ML, Rapado A, de la Piedra C. Role of 45. Osima M, Borgen TT, Lukic M, et al. Serum parathyroid hormone
citric acid in primary hyperparathyroidism with renal lithiasis. Urol is associated with increased cortical porosity of the inner transition­
Res. 1992;20(1):88-90. al zone at the proximal femur in postmenopausal women: the
33. Gafni RI, Langman CB, Guthrie LC, et al. Hypocitraturia is an un­ Tromso study. Osteoporos Int. 2018;29(2):421-431.
toward Side effect of synthetic human parathyroid hormone 46. Gafni RI, Brahim JS, Andreopoulou P, et al. Daily parathyroid hor­
(hPTH) 1-34 therapy in hypoparathyroidism that may increase re­ mone 1-34 replacement therapy for hypoparathyroidism induces
nal morbidity. J Bone Miner Res. 2018;33(10):1741-1747. marked changes in bone turnover and structure. J Bone Miner
34. Ayodele O, Rejnmark L, Sherry N, et al. Change in estimated glom­ Res. 2012;27(8):1811-1820.
erular filtration rate in adult patients with chronic hypoparathyr­ 47. Silva BC, Bilezikian JP. Skeletal abnormalities in hypoparathyroid­
oidism treated with rhPTH(1-84) compared with a historical ism and in primary hyperparathyroidism. Rev Endocr Metab
control cohort. J Endo Soc. 2021;5(Suppl 1):A257-A258. Disord. 2020;22(4):789-802.

You might also like