Sie sind auf Seite 1von 9

Articles

Efficacy and safety of recombinant human parathyroid


hormone (184) in hypoparathyroidism (REPLACE):
a double-blind, placebo-controlled, randomised,
phase 3 study
Michael Mannstadt, Bart L Clarke, Tamara Vokes, Maria Luisa Brandi, Lakshminarayan Ranganath, William D Fraser, Peter Lakatos,
Laszlo Bajnok, Roger Garceau, Leif Mosekilde, Hjalmar Lagast, Dolores Shoback, John P Bilezikian

Summary
Background Hypoparathyroidism results in impaired mineral homoeostasis, including hypocalcaemia and hyper- Lancet Diabetes Endocrinol
phosphataemia. Treatment with high-dose oral calcium and active vitamin D does not provide adequate or consistent 2013; 1: 27583

control of biochemical indices and can lead to serious long-term complications. We aimed to test the ecacy, safety, Published Online
October 7, 2013
and tolerability of once-daily recombinant human parathyroid hormone 184 (rhPTH[184]) in adults with
http://dx.doi.org/10.1016/
hypoparathyroidism. S2213-8587(13)70106-2
This online publication has
Methods In this double-blind, placebo-controlled, randomised phase 3 study (REPLACE), we recruited patients with been corrected.
hypoparathyroidism (18 months duration) aged 1885 years from 33 sites in eight countries. After an optimisation The corrected version rst
appeared at thelancet.com/
period, during which calcium and active vitamin D doses were adjusted to achieve consistent albumin-corrected
diabetes-endocrinology on
serum calcium, patients were randomly assigned (2:1) via an interactive voice response system to 50 g per day of Dec 5, 2013
rhPTH(184) or placebo for 24 weeks. Active vitamin D and calcium were progressively reduced, while rhPTH(184) See Comment page 260
could be titrated up from 50 g to 75 g and then 100 g (weeks 05). The primary endpoint was the proportion of Endocrine Unit, Massachusetts
patients at week 24 who achieved a 50% or greater reduction from baseline in their daily dose of oral calcium and General Hospital and Harvard
active vitamin D while maintaining a serum calcium concentration greater than or the same as baseline concentrations Medical School, Boston, MA,
and less than or equal to the upper limit of normal, analysed by intention to treat. This trial is registered with USA (M Mannstadt MD);
Division of Endocrinology,
ClinicalTrials.gov, number NCT00732615. Diabetes, Metabolism and
Nutrition, Mayo Clinic,
Findings Between June 23, 2009, and Feb 28, 2011, 134 eligible patients were recruited and randomly assigned to Rochester, MN, USA
rhPTH(184) (n=90) or placebo (n=44). Six patients in the rhPTH(184) group and seven in the placebo group (B L Clarke MD); Section of
Endocrinology, University of
discontinued before study end. 48 (53%) patients in the rhPTH(184) group achieved the primary endpoint compared Chicago Medicine, Chicago, IL,
with one (2%) patient in the placebo group (percentage dierence 511%, 95% CI 399623; p<00001). The USA (T Vokes MD); Department
proportions of patients who had at least one adverse event were similar between groups (84 [93%] patients in the of Internal Medicine,
University of Florence,
rhPTH[184] group vs 44 [100%] patients in the placebo group), with hypocalcaemia, muscle spasm, paraesthesias,
Florence, Italy
headache, and nausea being the most common adverse events. The proportions of patients with serious adverse (Prof M L Brandi MD);
events were also similar between the rhPTH(184) group (ten [11%] patients) and the placebo group (four [9%] Department of Clinical
patients). Biochemistry and Metabolic
Medicine, Royal Liverpool
University Hospital, Liverpool,
Interpretation 50 g, 75 g, or 100 g per day of rhPTH(184), administered subcutaneously in the outpatient setting, UK (L Ranganath MD);
is ecacious and well tolerated as a PTH replacement therapy for patients with hypoparathyroidism. Department of Medicine,
Norwich Medical School,
University of East Anglia,
Funding NPS Pharmaceuticals.
Norwich, UK
(Prof W D Fraser MD);
Introduction metabolites or analogues, an approach that does not 1st Department of Internal
Hypoparathyroidism is a rare disorder characterised by always provide adequate or consistent control of Medicine, Semmelweis
University, Budapest, Hungary
absent or decient production of parathyroid hormone biochemical and clinical aspects of the disease. Adverse
(P Lakatos MD); 1st Department
(PTH). PTH deciency is responsible for impaired short-term and long-term complications include large of Internal Medicine,
mineral homoeostasis, which is disrupted in several swings in serum calcium concentrations and risks of University of Pcs Medical
ways: calcium absorption is impaired due to reduced calcications in the kidney, brain, and elsewhere.4 School, Pcs, Hungary
(L Bajnok MD); NPS
renal conversion of 25-hydroxyvitamin D to active Without the calcium-conserving eects of PTH in the Pharmaceuticals, Bedminster,
1,25-dihydroxyvitamin D; hypercalciuria results from distal renal tubule, hypercalciuria, kidney stones, and NJ, USA (R Garceau MD,
reduced PTH-dependent renal calcium reabsorption; reduced renal function can occur.5,6 H Lagast MD); Department of
and bone turnover is decreased.13 Hormone deciency Human PTH(134), an active fragment of full length Endocrinology and Internal
Medicine, Aarhus University
states are usually treated by replacing the decient endogenous PTH(184), has been studied as a PTH Hospital, Aarhus, Denmark
hormone; however, this disorder is currently managed replacement therapy in hypoparathyroidism.712 Winer and (Prof L Mosekilde MD);
with large doses of oral calcium and active vitamin D colleagues712 have shown the feasibility of PTH Endocrine Research Unit,

www.thelancet.com/diabetes-endocrinology Vol 1 December 2013 275


Articles

San Francisco Veterans Aairs replacement therapy; however, PTH(134) has a short half- patient preference. Active vitamin D (calcitriol or
Medical Center, University of life and has not been approved for this indication. By alfacalcidol, provided by the sites pharmacies or by
California, San Francisco, CA,
USA (Prof D Shoback MD); and
contrast, recombinant human (rh)PTH(184), which is prescription) and oral calcium doses were adjusted to
Division of Endocrinology, identical in structure to the full-length endogenous achieve consistent albumin-corrected serum calcium
College of Physicians and hormone, is associated with a longer calcaemic eect concentrations between 187 mmol/L and the laboratory
Surgeons, Columbia University, when injected into the thigh13 than is rhPTH(134) injected upper limit of normal range, but ideally within the target
New York, NY, USA
(Prof J P Bilezikian MD)
into the abdomen.14 Serum calcium returns to baseline range of 20225 mmol/L. Any deciencies in serum
concentrations 24 h after administration of rhPTH(184), 25-hydroxyvitamin D (<75 nmol/L) or magnesium
Correspondence to:
Dr Michael Mannstadt, making this compound suitable for once-daily (<065 mmol/L) concentrations were corrected. After
Endocrine Unit Thier 1051, administration for hypoparathyroidism.1518 calcium and active vitamin D doses were optimised and
Massachusetts General Hospital, In the REPLACE study, we aimed to test the ecacy, had remained stable for 2 weekswhich established
Boston, MA 02114, USA
safety, and tolerability of a once-daily exible dose (50 g, baseline dosesrandomisation took place. Data was
mmannstadt@partners.org
75 g, or 100 g) regimen of rhPTH(184) in adults with originally obtained and analysed using conventional
hypoparathyroidism, to assess whether rhPTH(184) is units before being converted to SI units. For equivalent
an eective replacement therapy in this population of conventional units, please see the appendix.
patients.
Randomisation and masking
Methods Eligible participants were randomly assigned via an
Study design and patients interactive voice response system in a 2:1 ratio to receive
In this randomised, placebo-controlled, double-blind 50 g of rhPTH(184) (NPS Pharmaceuticals) once daily
registration trial, we recruited patients aged 1885 years or placebo. Randomisation was centrally administered by
who had well documented hypoparathyroidism for ClinPhone (Perceptive Informatics, East Windsor, NJ,
18 months or longer from 33 outpatient sites in eight USA), using a randomisation list generated by Quintiles
countries: USA (20), Canada (3), Denmark (3), Hungary (Raleigh/Durham, NC, USA). Simple block ran-
(3), Belgium (1), France (1), Italy (1), and the UK (1). domisation was applied without use of stratication
Hypoparathyroidism was dened as hypocalcaemia factors. Patients and investigators, including those
(calcium concentration below the lower limit of normal) administering the care and those assessing the outcomes,
and documented PTH concentrations below the lower were masked to treatment allocation throughout the
limit of the normal range, recorded on at least two 24 week dosing period. Laboratory results for bone
occasions within the previous 12 months. Additional marker and PTH concentrations were not accessible to
eligibility criteria were: a requirement for active vitamin the medical monitors, sponsor personnel, or study site
D and oral calcium (1000 mg daily) treatment, normal personnel. To maintain blinding, each multidose
thyroid-stimulating hormone concentrations if not on injection pen cartridge contained a clear, colourless
thyroid hormone replacement therapy (or if on therapy, solution with 14 doses of an identical injection volume
the dose had to have been stable for 3 months), and (007 mL) for each dose, irrespective of treatment group
normal magnesium and serum 25-hydroxyvitamin D or assigned dose. None of the blinding was broken in
concentrations. Creatinine clearance needed to be either this study.
greater than 30 mL per min on two separate measure-
ments, or greater than 60 mL per min (one measurement) Procedures
with an accompanying serum creatinine concentration The prescribed dose of rhPTH(184) or placebo was self-
of less than 1326 mol/L. We excluded patients with a administered subcutaneously in the thigh every morning
known activating mutation in the calcium-sensing with a multidose injection pen device. Blood tests were
receptor gene; additional exclusion criteria are listed in done at each scheduled assessment before injecting the
See Online for appendix the appendix. study drug or taking oral medication.
The protocol and any amendments were approved by The 24 week treatment period began with a 12 week
the institutional review boards of all participating titration phase, during which the doses of active
institutions, and written informed consent was obtained vitamin D were reduced and, if possible, eliminated,
from all patients. An external data and safety monitoring followed by reduction in oral calcium doses, while
board was assigned to assess data throughout the study. maintaining serum calcium at or above the
The study consisted of three periods: optimisation of concentration recorded at baseline. During the titration
oral calcium and active vitamin D doses, correction of phase, the investigator could increase the daily dose of
vitamin D and magnesium deciencies, and rhPTH(184) at week 2 to 75 g and again at week 4 up
discontinuation of thiazides (216 weeks); treatment to the maximum dose of 100 g to allow active vitamin D
(24 weeks); and follow-up (4 weeks; appendix). During and oral calcium doses to be reduced until active
optimisation, oral calcium was replaced with calcium vitamin D could be eliminated and oral calcium could
citrate or calcium carbonate (both provided by NPS be reduced to 500 mg per day or less. Details of the
Pharmaceuticals, Bedminster, NJ, USA) according to titration protocol are presented in the appendix.

276 www.thelancet.com/diabetes-endocrinology Vol 1 December 2013


Articles

Patients then entered a 12 week maintenance phase,


when oral calcium and active vitamin D doses were 196 screened

adjusted as appropriate to maintain serum calcium and


avoid hypercalciuria or raised concentrations of serum 62 not randomised
38 screen failure
calciumphosphate product (calcium concentration 20 consent withdrawal
phosphate concentration). The daily dose of rhPTH(184) 4 optimisation failure
established during titration could be reduced, but not
increased, in this phase. 134 randomised (2:1)
Assessment visits during the treatment period took
place weekly from weeks 06 and then at weeks 8, 12, 16,
20, and 24. Two visits took place during follow-up on 90 assigned to rhPTH(184) 44 assigned to placebo
weeks 25 and 28. At each visit, patients were seen by the
study physician and the research assistant. Laboratory
6 discontinued 7 discontinued
values, symptoms described by the patient, adverse 3 adverse events* 3 patient decision
events, and medication changes since the last visit were 2 patient/investigator decision 3 investigator decision
recorded. When doses of oral calcium, active vitamin D, 1 lost to follow-up 1 non-compliance

or rhPTH(184) were changed, a safety calcium


measurement was obtained 35 days afterwards. 84 completed study and follow-up 37 completed study and follow-up
Albumin-corrected serum calcium concentrations were
measured at every visit throughout the treatment period Figure 1: Trial prole
(weekly for weeks 06, and then at weeks 8, 12, 16, 20, rhPTH(184)=recombinant human parathyroid hormone 184. *Due to hypertension (n=1), stroke (n=1), and
several events (n=1, including injection site erythema, arthralgia, asthenia, blepharospasm, decreased appetite,
and 24). Total serum calcium was corrected for albumin
depression, headache, nausea, pain in extremity, and hypercalcaemia).
concentration using the following formula: serum
calcium in mmol/L + (002 [40 serum albumin in g/L])
or serum calcium in mg/dL + (08 [40serum albumin maintenance (weeks 1624). Symptoms of hypo-
in g/dL]). 24 h urinary calcium was measured at baseline calcaemia were identied by the investigator and
(week 0) and weeks 3, 5, 6, 8, 12, 16, and 24. At week 24, included paraesthesias, muscle cramps, hypoaesthesia,
rhPTH(184) was discontinued and patients resumed tetany, back pain, myalgia, muscle twitching, throat
their earlier, optimised doses of oral calcium and active tightness, musculoskeletal pain, anxiety, and seizures.
vitamin D. Serum calcium concentrations were measured A comprehensive list of all symptoms used to identify
1 week and 4 weeks after treatment was stopped (weeks 25 hypocalcaemic episodes for this ecacy endpoint is
and 28 after initiation of treatment). presented in the appendix.
We assessed compliance by reviewing the Prespecied exploratory endpoints included changes
rhPTH(184) self-dosing records in patients diaries in 24 h urine calcium excretion from baseline, and the
and by checking, at each visit, the amount of drug proportion of patients who had a calciumphosphate
injected, wasted, and returned (if any), computed as product in the normal range (44 mmol/L) at week 24.
(number of doses administered/[last dose date rst The safety analysis focused on the reports of adverse
dose date + 1]) 100%. events and laboratory data, including reports of
The primary endpoint was the proportion of patients at hypocalcaemia or an increase in blood calcium. For
week 24 who achieved all three of the following criteria: laboratory values, the protocol dened the serum calcium
50% or greater reduction from baseline of oral calcium target range to be 187 mmol/L to the upper limit of
dose; 50% or greater reduction from baseline of active normal. The investigator decided whether values outside
vitamin D dose; and maintenance of a stable albumin- that range were of clinical signicance and therefore
corrected total serum calcium concentration greater than reported as hypocalcaemia or hypercalcaemia.
or equal to baseline concentration and less than or equal
to the upper limit of normal, but ideally within the target Statistical analysis
range of 20225 mmol/L. As components of the The statistical analysis was done by two employees of
primary ecacy endpoint, daily doses of oral calcium NPS Pharmaceuticals. On the basis of 84 patients
and of the active vitamin D analogue used were obtained completing the study in a 2:1 ratio (rhPTH[184] vs
from site investigator prescription data and patient diary placebo) with an expected proportion of 40% and 10%,
data. respectively, of patients achieving the primary endpoint,
Secondary endpoints were the proportion of patients the study had 80% power to detect a dierence according
who achieved independence from active vitamin D to a two-tailed Fishers exact test and an alpha error of
while taking 500 mg per day or less of oral calcium at 005. The intention-to-treat population, for which the
week 24, percentage change from baseline in prescribed primary ecacy analysis was done, included all randomly
oral calcium dose by week 24, and changes in frequency assigned patients who received at least one dose of
of clinical symptoms of hypocalcaemia during study drug and had at least one postbaseline ecacy

www.thelancet.com/diabetes-endocrinology Vol 1 December 2013 277


Articles

patients who achieved the primary endpoint, the two-


rhPTH(184) Placebo
(n=90) (n=44) sided Fishers exact test was used. Dierences between
the two groups are presented as their two-sided
Age, years 470 (122) 485 (137)
asymptotic 95% CI. Alfacalcidol dose was converted to
Sex
calcitriol dose on the basis that 2 g of alfacalcidol equals
Women 69 (77%) 36 (82%)
1 g of calcitriol.
Men 21 (23%) 8 (18%)
This trial is registered with ClinicalTrials.gov, number
Race
NCT00732615.
White 85 (94%) 43 (98%)
Other 5 (6%) 1 (2%)
Role of the funding source
Geographic region*
The sponsor of the study participated in study design,
North America 49 (54%) 25 (57%) data collection, data analysis, data interpretation, and
Europe 41 (46%) 19 (43%) review and approval of the report, and supplied the study
Duration of hypoparathyroidism, years 141 (1114) 110 (798) drug. All authors had full access to all the data in the
5 years 19 (21%) 12 (27%) study and all had nal responsibility for the decision to
510 years 28 (31%) 14 (32%) submit for publication.
10 years 43 (48%) 18 (41%)
Cause of hypoparathyroidism Results
Postsurgical 68 (76%) 31 (70%) Between June 23, 2009, and Feb 28, 2011, 196 patients
Idiopathic 14 (16%) 8 (18%) were screened, of whom 134 were eligible for random
Autoimmune disease 5 (6%) 4 (9%) assignment to the rhPTH(184) (n=90) or placebo groups
Known genetic disorder 2 (2%) 1 (2%) (n=44; gure 1). All 134 patients were included in the
Radiation 1 (1%) 0 primary ecacy and safety analyses. Patient
Prescribed active vitamin D metabolite/analogue demographics, baseline characteristics, and laboratory
Low dose 6 (7%) 4 (9%) values were similar between the rhPTH(184) and
Medium dose 23 (26%) 12 (27%) placebo groups (table 1). Most patients were women
High dose 61 (68%) 28 (64%) (78%), and the mean age of all patients was 475 years
Prescribed calcium (SD 1271). The most common cause of hypopara-
02000 mg per day 61 (68%) 31 (70%) thyroidism was postsurgical hypoparathyroidism (74%),
>2000 mg per day 29 (32%) 13 (30%) with a mean duration of 13 years (SD 1029). At baseline
Laboratory variables after optimisation, mean oral calcium dose was 2101 mg
Albumin-corrected total serum Ca, mmol/L 212 (020) 215 (015) per day (SD 1286, range 100012 000) and mean calcitriol
Serum phosphate, mmol/L 047 (007) 048 (007) dose was 089 g per day (048, range 02530).
Serum magnesium, mmol/L 082 (008) 082 (008) Baseline mean albumin-corrected total serum calcium
Serum 25-hydroxyvitamin D, nmol/L 10808 (4593) 11032 (4518) concentrations were similar in the two groups (table 1).
Serum 1,25-dihydroxyvitamin D, pmol/L 8233 (5016) 7825 (2760) Mean urinary calcium excretion exceeded 300 mg per
Urine calcium, mg per 24 h 3566 (1890) 3454 (1707)
24 h in both groups. At baseline, one patient in the
Creatinine, mol/L 8752 (1848) 8310 (2175)
rhPTH(184) group and no patients in the placebo group
Calcium-to-creatinine ratio, mg/mg 0271 (01) 0287 (01)
had a calciumphosphate product that was greater than
44 mmol/L.
Calciumphosphate product >44 mmol/L 1 (11%) 0
Compliance with injection was excellent for both
Data are mean (SD), median (IQR), or number (%), unless otherwise stated. rhPTH(184)=recombinant human groups, with 80% or higher compliance in 88 (98%) of
parathyroid hormone 184. *North America consists of Canada and the USA; Europe consists of Belgium, Denmark,
90 patients in the rhPTH(184) group and 42 (96%) of
France, Hungary, Italy, and the UK. For calcitriol (as dened in the study), low dose is 025 g per day, medium dose is
>02505 g per day, and high dose is >05 g per day; for alfacalcidol low dose is 050 g per day, medium dose is 44 patients in the placebo group. 47 (52%) patients in the
>05010 g per day, and high dose is >10 g per day. rhPTH(184) group completed the study on a
rhPTH(184) dose of 100 g per day, 24 (27%) on a dose
Table 1: Patient demographics and baseline characteristics
of 75 g per day, and 19 (21%) on a dose of 50 g per day.
A higher proportion of patients in the rhPTH(184)
measurement. If a patient did not have week 24 data, the group achieved all three criteria for the composite
ecacy assessment was made using the last observation primary endpoint than did those in the placebo group
carried forward. The safety population included all (48 [53%] patients vs one [2%] patient; percentage
randomly assigned patients who received at least one dierence 511%, 95% CI 399623; p<00001). Of the
dose of study drug. All statistical procedures were patients for whom we had week 24 data, 36 (43%) of
completed using SAS version 9.1. 84 patients who received rhPTH(184) were able to stop
The site investigators prescription records were the taking active vitamin D and were receiving 500 mg per
primary source for documenting amounts of oral calcium day of oral calcium at week 24, compared with two (5%)
and active vitamin D used. To assess the proportion of of 37 patients who received placebo (p<00001).

278 www.thelancet.com/diabetes-endocrinology Vol 1 December 2013


Articles

Analysis of self-dosing data recorded in the patient vitamin D dose decreased by 78% and 30% in the
diaries corresponded with the prescribed dosing data rhPTH(184) and placebo groups, respectively
(gure 2). (p<00001).
Between-group dierences in the mean decrease from During maintenance (weeks 1624), a smaller
baseline for prescribed doses of both oral calcium proportion of patients in the rhPTH(184) group reported
(p=00016) and active vitamin D (p=00035) were clinical symptoms associated with hypocalcaemia than did
apparent from week 3 and continued until week 24 those in the placebo group (30 [33%; 95% CI 237441]
(gure 3); similar results were seen with patient diary patients vs 18 [41%; 263568] patients), although the
data (not shown). Oral calcium and active vitamin D dierence was not statistically signicant (p=039).
doses throughout the trial are summarised in the At week 24, mean 24 h urinary calcium excretion rates
appendix. Additionally, the rhPTH(184) group had a had decreased by 736 mg per 24 h (SD 1902) in the
mean percentage decrease from baseline in oral calcium rhPTH(184) group and by 838 mg per 24 h (1691) in the
dose of 52% compared with a 6% mean percentage placebo group (p=057). The mean urinary calcium
increase in the placebo group (p<00001), and the active excretion rate in the placebo group declined by a

A Primary outcome B Independence from active vitamin D and reduction in oral calcium
70 Placebo (n=44)
rhPTH(184) (n=90)

60

50
Patients (%)

40

30

20

10

0
0 4 8 12 16 20 24 0 4 8 12 16 20 24
Weeks Weeks

Figure 2: Achievement of primary outcome and independence from active vitamin D and reduction in oral calcium during treatment period, according to patient diary data
(A) Proportion of patients achieving the criteria for the primary endpoint throughout the 24 week treatment period. (B) Proportion of patients who were able to stop taking active vitamin D and to
reduce their dose of oral calcium dose to 500 mg per day throughout the 24 week treatment period. rhPTH(184)=recombinant human parathyroid hormone 184.

A Calcium dose B Active vitamin D dose


4000 Placebo (n=44) 15
rhPTH(184) (n=90)
3500
Mean active vitamin D dose (g per day)

3000
10
Mean calcium dose (mg per day)

2500

2000
05
1500

1000
0
500

500 05
0 4 8 12 16 20 24 0 4 8 12 16 20 24
Weeks Weeks

Figure 3: Changes in calcium and active vitamin D doses throughout treatment period, by treatment group
Mean oral calcium dose (A) and mean active vitamin D dose (B) in the placebo group and rhPTH(184) group throughout the 24 week treatment period. Error bars show SD. rhPTH(184)=recombinant
human parathyroid hormone 184. From weeks 324, each mean measurement of calcium dose and active vitamin D dose diered signicantly between groups (all p<0005).

www.thelancet.com/diabetes-endocrinology Vol 1 December 2013 279


Articles

urinary calcium excretion showed a slight decrease


A Placebo (gure 4B). Post-hoc analysis gave similar results for
10 Placebo (n=44) serum calcium Serum target range (8090 mg/dL) 800 uncorrected total calcium concentrations (data not shown).
Placebo (n=44) urine calcium Normal urine excretion (50300 mg per 24 h) Mean serum phosphate concentrations were similar (at
rhPTH(184) (n=90) serum calcium
rhPTH(184) (n=90) urine calcium
the upper limit of normal) in both groups at baseline
Mean albumin-corrected total serum calcium (mg/dL)

(table 1), but fell substantially in the rhPTH(184) group

Mean 24 h urinary calcium excretion (mg per 24 h)


9 600 upon treatment and remained lower than in the placebo
group throughout treatment (p=00025 at all timepoints).
At week 24, mean serum phosphate concentrations had
400 decreased by 005 mmol/L (SD 008) in the rhPTH(184)
8 group and 001 mmol/L (007) in the placebo group
(p=000098). From week 5 to week 23, no patient in
either treatment group had greater than 44 mmol/L of
200 calciumphosphate product, until week 24, when one
7
patient in the placebo group had a raised calcium
phosphate product of 445 mmol/L. In the rhPTH(184)
0
group, the mean calciumphosphate product decreased
6 from 32 mmol/L (SD 06) at baseline to 28 mmol/L
(05) at week 24 (p<00001), and in the placebo group
0
from 33 mmol/L (05) to 32 mmol/L (04; p=011).
B rhPTH(184)
Results of a post-hoc analysis showed that the dierence
10 800
in the mean change from baseline to week 24 between
the rhPTH(184) and placebo groups was signicant
Mean albumin-corrected total serum calcium (mg/dL)

(p=00005).
Mean 24 h urinary calcium excretion (mg per 24 h)

9 600 Mean serum concentration of 25-hydroxyvitamin D


decreased in the rhPTH(184) group by 280 nmol/L
(SD 474, range 2796 to 1148) from baseline to
week 24; minimal change was seen in the placebo group
400
8 (35 nmol/L [327, range 1023 to 449]). Mean serum
1,25-dihydroxyvitamin D concentrations were maintained
in the normal range in both groups (823 pmol/L
200 [SD 502] at baseline and 794 pmol/L [300] at week 24
7 in the rhPTH[184] group vs 783 pmol/L [276] at
baseline and 790 pmol/L [442] at week 24 in the placebo
group).
0 The overall incidences of adverse events were similar
6
in both groups (table 2). By week 28, 84 (93%) patients in
0 the rhPTH(184) and 44 (100%) patients in the placebo
0 4 8 12 16 20 24
Weeks group had at least one adverse event, with hypocalcaemia,
muscle spasm, paraesthesias, headache, and nausea
Figure 4: Changes in albumin-corrected total serum calcium concentrations and 24 h urine calcium excretion
throughout treatment period, by treatment group being the most common adverse events (appendix). Ten
Mean albumin-corrected serum calcium concentration and mean 24 hr urinary calcium excretion in the placebo (11%) patients in the rhPTH(184) group and four (9%)
group (A) and rhPTH(184) group (B) throughout the 24 week treatment period. Error bars show SD. Every in the placebo group had serious adverse events; only
measurement of serum calcium from week 1 to week 16 diered signicantly between treatment groups
one serious adverse event (hypercalcaemia requiring a
(p=0018). Each measurement of urine calcium excretion from week 3 to week 8 diered signicantly between
groups (p=0015). rhPTH(184)=recombinant human parathyroid hormone 184. brief hospital stay) in the rhPTH(184) group was
regarded as treatment related. The episode of
hypercalcaemia occurred 32 days after starting
signicantly greater amount from baseline to week 8 than rhPTH(184), the study drug had been up-titrated from
did the urinary calcium concentration in the rhPTH(184) 50 g to 75 g 7 days before the event. The hypercalcaemia
group (p=0015; gure 4). In the placebo group, total resolved and the event did not lead to study
serum calcium concentrations fell rapidly and remained discontinuation. The patient enrolled in subsequent
close to the lower end of the target range for the duration extension studies and has been receiving rhPTH(184)
of the study; mean urinary calcium excretion rate also for more than 24 months without any further episodes of
decreased (gure 4A). By contrast, in the rhPTH(184) hypercalcaemia.
group, albumin-corrected serum calcium concentrations No signicant changes in mean cardiovascular
increased at the start of treatment, despite large reductions variables (blood pressure, heart rate, or QTc interval) or
in both oral calcium and active vitamin D doses, whereas renal variables (serum creatinine or estimated

280 www.thelancet.com/diabetes-endocrinology Vol 1 December 2013


Articles

creatinine clearance) were reported in either group.


rhPTH(184) Placebo
Three (3%) patients in the rhPTH(184) group
discontinued treatment because of adverse events, Patients (n=90) Events Patients (n=44) Events

including one patient who had several adverse events Any AE


(some of which were judged to be treatment related); No 6 (7%) NA 0 NA
the other two patients had adverse events (worsening Yes 84 (93%) 854 44 (100%) 354
hypertension, cerebrovascular accident) that were not AE of highest severity*
thought to be related to treatment or the concentration Mild 25 (28%) 15 (34%)
of serum calcium. No adverse events led to Moderate 44 (49%) 24 (55%)
discontinuation in the placebo group. Severe 15 (17%) 5 (11%)
Hypocalcaemia, which is a characteristic of hypopara- AEs leading to study discontinuation 3 (3%) 15 0 NA
thyroidism, was frequently reported as an adverse event in AEs leading to death 0 NA 0 NA
both groups throughout the study. During the treatment Serious AEs 10 (11%) 12 4 (9%) 5
period, hypocalcaemia was reported as an adverse event in Mild 1 (1%) 1 1 (2%) 1
23 (26%) patients in the rhPTH(184) group (43 events) Moderate 3 (3%) 5 2 (5%) 2
compared with nine (21%) patients in the placebo group Severe 6 (7%) 6 1 (2%) 2
(nine events). During follow-up, after rhPTH(184)
treatment ended and patients returned to baseline oral Data are number of patients (% of patients) and number of events. If a patient had more than one AE in a category, the
patient was counted only once in that category; each event is counted. AE=adverse event. rhPTH(184)=recombinant
calcium and active vitamin D doses, hypocalcaemia was human parathyroid hormone 184. *Patients with several adverse events were classied according to their event with
reported as an adverse event in a higher proportion of the highest severity. Includes hypocalcaemia (n=2), hypercalcaemia (n=1), pancreatitis (n=1), cerebrovascular
patients in the rhPTH(184) group than in the placebo accident (n=1), and diarrhoea (n=1); all cases resolved themselves except the case of cerebrovascular accident; none
was study drug related except the case of hypercalcaemia. One patient reported asthma or chronic obstructive
group (28 [31%] patients vs four [9%] patients). At week 12, pulmonary disease that was not study drug related and resolved itself.
mean peak increases of albumin-corrected total serum
calcium concentration measured before and after injection Table 2: Summary of adverse events
of rhPTH(184) were about 025 mmol/L or less from the
predose baseline 610 h after injection; no clear dose in both calcium and active vitamin D while maintaining
response relation was noted and these transient excursions serum calcium concentrations in the target range.
did not raise any safety concerns. Reports of hypo- Nevertheless, the results clearly favour the rhPTH(184)
calcaemia, hypercalcaemia, and hypercalciuria by regimen over placebo. Additional support for the
treatment period are summarised in the appendix. signicance of the primary ecacy outcomewhich was
based on investigator-prescribed doses of oral calcium
Discussion and active vitamin Dwas provided by analysis of data
This randomised controlled trial provides evidence that collected via patients electronic diaries, which
rhPTH(184) replacement therapy is eective in treating documented the doses that were actually taken.
hypoparathyroidism when compared with oral calcium Small changes in serum calcium concentrations lead to
and active vitamin D alone (placebo group). More than substantial changes in the ltered load of calcium.20
half the patients in the rhPTH(184) group (53%) reached Therefore, unsurprisingly, 24 h urinary calcium excretion
the primary endpoint, compared with only 2% in the in patients in the placebo group was lower at the end of
placebo group. the trial than at baseline, since their mean serum calcium
Patients recruited for this trial were representative of concentration was also lower at the end of the study.
the disorder in that even after optimisation of their Importantly, in the rhPTH(184) group, 24 h urinary
calcium and vitamin D regimen, 89 (66%) patients were calcium excretion also showed a slight decrease, whereas
taking high doses of active vitamin D (>05 g calcitriol serum calcium concentration remained relatively stable
per day) and 42 (31%) patients were taking high doses of (within the target range) despite reductions in oral
calcium (>2 g per day; table 1). A signicantly higher calcium and active vitamin D. This nding is probably
proportion of patients in the rhPTH(184) group were due to the potent renal calcium-conserving actions of
able to discontinue all active vitamin D therapy and to rhPTH(184) and represents a potential advantage of
reduce their oral calcium dose to 500 mg per day or less rhPTH(184) replacement therapy in hypoparathyr-
compared with the placebo group. Even though many oidism. The slight decrease in mean serum calcium
patients who received rhPTH(184) had a reduced need concentrations from week 12 to week 24 in the
for oral calcium and active vitamin D on treatment, not rhPTH(184) group is a result of continual adjustments
all patients who had this clinical eect met all three of oral calcium and active vitamin D, and down-titration
criteria of the primary endpoint. This nding is due, in of the rhPTH(184) dose in individual patients. Serum
part, to the strict protocol algorithm that focused on 1,25-dihydroxyvitamin D concentrations were maintained
eliminating all active vitamin D before reducing oral in the normal range in both groups throughout the trial
calcium during titration. This experimental design made despite dramatic reductions in active vitamin D dose
it more challenging to achieve 50% or greater reductions requirements in the rhPTH(184) group.

www.thelancet.com/diabetes-endocrinology Vol 1 December 2013 281


Articles

while substantially reducing the need for active


Panel: Research in context vitamin D and calcium. Hyperphosphataemia was also
Systematic review improved, and hypercalciuria did not increase.
We searched PubMed for clinical trials published before Aug 22, 2013, with the terms Additionally, treatment with rhPTH(184) was well
hypoparathyroidism and parathyroid hormone. Seven clinical trials reported the eects tolerated, as shown by the high compliance rate.
of the human parathyroid hormone fragment (hPTH[134]), in patients with Benecial eects included increasing low serum calcium
hypoparathyroidism.712,19 These studies progressed from a proof-of-concept study with concentrations without a concomitant increase in
once-daily hPTH(134)7 to studies with dose-adjustable PTH(134) regimens.8,10 They calciuria, decreasing high serum phosphate, decreasing
showed that twice-daily dosing achieves better control than once-daily dosing8,10 and that calciumphosphate product, and activating endogenous
delivery with a pump achieves normalisation of mineral metabolism.12 Use of PTH(134) 1,25-dihydroxyvitamin D production.
was also shown to be eective in children, and long-term use for up to 3 years was shown to Overall, these ndings show that rhPTH(184) is
be safe.10 Six studies reported the eects of the full-length hormone recombinant ecacious as a replacement for endogenous PTH in
(r)hPTH(184) in patients with hypoparathyroidism.1518,21,22 rhPTH(184), when given at patients with hypoparathyroidism. This study also
100 g subcutaneously every other day, was eective and safe for up to 4 years.15 The only conrms that 50 g per day subcutaneously is an
double-blinded, placebo-controlled trial used rhPTH(184) as an add-on therapy at a xed acceptable starting dose of rhPTH(184), with possible
dose of 100 g once daily.17 With this regimen, initial rates of hypercalcaemia were higher up-titration by increments of 25 g up to 100 g, for the
than with placebo. PTH replacement therapy needs exible dosing to correspond to treatment of hypoparathyroidism in an outpatient
dierent needs in dierent patients. No placebo-controlled trial has been reported that uses setting.
such a design that is applicable to clinical practice. Contributors
MM, BLC, TV, RG, LM, HL, DS, and JPB contributed to the writing and
Interpretation review of the report. MM, BLC, TV, MLB, LR, WDF, PL, LB, LM, DS, and
Our clinical trial, the largest done so far in patients with hypoparathyroidism, is the rst JPB contributed to recruitment of patients and were study investigators.
MM, BLC, TV, RG, HL, DS, and JPB contributed to the data
phase 3 trial for PTH replacement therapy using the full length molecule, rhPTH(184), in
interpretation. RG, HL, and JPB contributed to the study design.
the outpatient setting. By showing the safety and ecacy of once-daily rhPTH(184) by
REPLACE principal investigators
subcutaneous injection at exible doses of 50100 g per day, this trial shows that
The following individuals were principal investigators at sites enrolling
rhPTH(184) could become a PTH replacement therapy for hypoparathyroidism by patients in the REPLACE study: D Crawford Allison (Waco, TX, USA),
addressing the underlying defect. Laszlo Bajnok (Pecs, Hungary), Albert Beckers (Liege, Belgium),
Jolene Berg (San Antonio, TX, USA), John P Bilezikian (New York, NY,
USA), Maria Luisa Brandi (Florence, Italy), Kim Brixen (Odense,
Denmark), Bart L Clarke (Rochester, MN, USA), William D Fraser
Clinical symptoms associated with hypocalcaemia were (Norwich, UK), David A Hanley (Calgary, AB, Canada), Pascal Houillier
assessed for both safety and ecacy. Despite substantial (Paris, France), Stephanie M Kaiser (Halifax, NS, Canada), Aliya Khan
(Oakville, ON, Canada), Peter Lakatos (Budapest, Hungary),
decreases in oral calcium and active vitamin D doses, Michael Levine (Philadelphia, PA, USA), Ivy-Joan Madu (Orange, CA,
rhPTH(184) was associated with fewer clinical USA), Michael Mannstadt (Boston, MA, USA), Shon Meek (Jacksonville,
symptoms of hypocalcaemia during maintenance than FL, USA), Leif Mosekilde (Aarhus, Denmark), Christina Orr (Vancouver,
was placebo, although during follow-up (after withdrawal WA, USA), Munro Peacock (Indianpolis, IN, USA), Michael Perley
(Lakewood, CA, USA), Lakshminarayan Ranganath (Liverpool, UK),
of study drug) hypocalcaemia was reported as an adverse Jerey Rothman (New York, NY, USA), Dolores Shoback (San Francisco,
event in a higher proportion of patients in the CA, USA), Zsuzsanna Valkusz (Szeged, Hungary), Tamara Vokes
rhPTH(184) group than in the placebo group. (Chicago, IL, USA), Mark Warren (Greenville, NC, USA), Nelson Watts
(Cincinnati, OH, USA), and Michael Whitaker (Scottsdale, AZ, USA).
The short-term design, which precluded a detailed
analysis of skeletal and renal eects of rhPTH(184) Conicts of interest
MM and TV have served as advisory group members for NPS
therapy, is a limitation of this study. Another limitation
Pharmaceuticals (MM through a contract with his institution). BLC, DS,
was that the dose schedule dened by the protocol did and JPB have received institutional research grants from and served as
not allow up-titration of study drug after week 5 or dose advisory group members for NPS Pharmaceuticals. RG and HL are
increases of greater than 100 g per day; therefore, we do employees of NPS Pharmaceuticals.
not know whether some patients might have been able to Acknowledgments
achieve the endpoint with higher doses. Statistical analysis was done by Zane Bai and Benjamin Li of NPS
Pharmaceuticals.
This is the rst study to use a exible dosing regimen
of rhPTH(184) and a rigorous algorithm for titration of References
1 Bilezikian JP, Khan A, Potts JT Jr, et al. Hypoparathyroidism in the
oral calcium and active vitamin D in a double-blinded adult: epidemiology, diagnosis, pathophysiology, target-organ
and placebo-controlled trial (panel). The exible dosing involvement, treatment, and challenges for future research.
and titration algorithm can be readily implemented in an J Bone Miner Res 2011; 26: 231737.
2 Shoback D. Clinical practice. Hypoparathyroidism. N Engl J Med
outpatient setting, which makes it immediately applicable 2008; 359: 391403.
for clinical practice. 3 Rejnmark L, Sikjaer T, Underbjerg L, Mosekilde L. PTH replacement
Other strengths are the large number of patients therapy of hypoparathyroidism. Osteoporos Int 2013; 24: 152936.
enrolled, since hypoparathyroidism is a rare disease. 4 Goswami R, Sharma R, Sreenivas V, Gupta N, Ganapathy A, Das S.
Prevalence and progression of basal ganglia calcication and its
The results show that replacement therapy with pathogenic mechanism in patients with idiopathic
rhPTH(184) can optimise and stabilise serum calcium hypoparathyroidism. Clin Endocrinol (Oxf) 2012; 77: 20006.

282 www.thelancet.com/diabetes-endocrinology Vol 1 December 2013


Articles

5 Weber G, Cazzu MA, Frisone F, et al. Nephrocalcinosis in 14 Satterwhite J, Heathman M, Miller PD, Marin F, Glass EV,
children and adolescents: sonographic evaluation during long-term Dobnig H. Pharmacokinetics of teriparatide (rhPTH[1-34]) and
treatment with 1,25-dihydroxycholecalciferol. Child Nephrol Urol calcium pharmacodynamics in postmenopausal women with
1988; 9: 27376. osteoporosis. Calcif Tissue Int 2010; 87: 48592.
6 Mitchell DM, Regan S, Cooley MR, et al. Long-term follow-up of 15 Cusano NE, Rubin MR, McMahon DJ, et al. Therapy of
patients with hypoparathyroidism. J Clin Endocrinol Metab 2012; hypoparathyroidism with PTH(1-84): a prospective four-year
97: 450714. investigation of ecacy and safety. J Clin Endocrinol Metab 2013;
7 Winer KK, Yanovski JA, Cutler GB Jr. Synthetic human parathyroid 98: 13744.
hormone 1-34 vs calcitriol and calcium in the treatment of 16 Rubin MR, Sliney J Jr, McMahon DJ, Silverberg SJ, Bilezikian JP.
hypoparathyroidism. JAMA 1996; 276: 63136. Therapy of hypoparathyroidism with intact parathyroid hormone.
8 Winer KK, Yanovski JA, Sarani B, Cutler GB Jr. A randomized, Osteoporos Int 2010; 21: 192734.
cross-over trial of once-daily versus twice-daily parathyroid hormone 17 Sikjaer T, Rejnmark L, Rolighed L, Heickendor L, Mosekilde L, for
1-34 in treatment of hypoparathyroidism. J Clin Endocrinol Metab the Hypoparathyroid Study Group. The eect of adding PTH(1-84)
1998; 83: 348086. to conventional treatment of hypoparathyroidism: a randomized,
9 Winer KK, Ko CW, Reynolds JC, et al. Long-term treatment of placebo-controlled study. J Bone Miner Res 2011; 26: 235870.
hypoparathyroidism: a randomized controlled study comparing 18 Sikjaer T, Amstrup AK, Rolighed R, Kjaer SG, Rejnmark L,
parathyroid hormone-(1-34) versus calcitriol and calcium. Mosekilde L. PTH (1-84) replacement therapy in hypoparathyroidism:
J Clin Endocrinol Metab 2003; 88: 421420. a randomized controlled trial on pharmacokinetic and dynamic
10 Winer KK, Sinaii N, Peterson D, et al. Eects of once versus eects following 6 months of treatment. J Bone Miner Res 2013;
twice-daily parathyroid hormone 134 therapy in children with published online May 3. DOI:10.1002/jbmr.1964.
hypoparathyroidism. J Clin Endocrinol Metab 2008; 93: 338995. 19 Gafni RI, Brahim JS, Andreopoulou P, et al. Daily parathyroid
11 Winer KK, Sinaii N, Reynolds J, et al. Long-term treatment of hormone 134 replacement therapy for hypoparathyroidism induces
12 children with chronic hypoparathyroidism: a randomized trial marked changes in bone turnover and structure. J Bone Miner Res
comparing synthetic human parathyroid hormone 134 versus 2012; 27: 181120.
calcitriol and calcium. J Clin Endocrinol Metab 2010; 95: 268088. 20 Macfadyen IJ, Nordin BE, Smith DA, Wayne DJ, Rae SL. Eect of
12 Winer KK, Zhang B, Shrader JA, et al. Synthetic human parathyroid variation in dietary calcium on plasma concentration and urinary
hormone 134 replacement therapy: a randomized crossover trial excretion of calcium. BMJ 1965; 1: 16164.
comparing pump versus injections in the treatment of chronic 21 Sikjaer T, Rejnmark L, Thomsen JS, et al. Changes in 3-dimensional
hypoparathyroidism. J Clin Endocrinol Metab 2012; 97: 39199. bone structure indices in hypoparathyroid patients treated with
13 Fox J, Wells DS, Garceau RJ. Relationships between PTH(1-84): a randomized controlled study. J Bone Miner Res 2012;
pharmacokinetic prole of human PTH (1-84) and serum calcium 27: 78188.
response in postmenopausal women following four dierent 22 Rubin MR, Dempster DW, Sliney J Jr, et al. PTH(184)
methods of administration. http://www.asbmr.org/Meetings/ administration reverses abnormal bone-remodeling dynamics and
AnnualMeeting/AbstractDetail.aspx?aid=cb1c7954-4672-41f1-8b55- structure in hypoparathyroidism. J Bone Miner Res 2011; 26: 272736.
9a70ec107b6f (accessed June 24, 2013).

www.thelancet.com/diabetes-endocrinology Vol 1 December 2013 283

Das könnte Ihnen auch gefallen