You are on page 1of 13

The Journal of Clinical Endocrinology & Metabolism, 2023, 108, 1696–1708

https://doi.org/10.1210/clinem/dgad015
Advance access publication 12 January 2023
Clinical Research Article

Intranasal Carbetocin Reduces Hyperphagia, Anxiousness,


and Distress in Prader-Willi Syndrome: CARE-PWS
Phase 3 Trial
Elizabeth Roof,1 Cheri L. Deal,2 Shawn E. McCandless,3 Ronald L. Cowan,4 Jennifer L. Miller,5

Downloaded from https://academic.oup.com/jcem/article/108/7/1696/6986298 by guest on 20 November 2023


Jill K. Hamilton,6,7 Elizabeth R. Roeder,8,9 Shana E. McCormack,10,11 Tamanna R. Roshan Lal,12
Hussein D. Abdul-Latif,13 Andrea M. Haqq,14 Kathryn S. Obrynba,15,16 Laura C. Torchen,17
Alaina P. Vidmar,18,19 David H. Viskochil,20,21 Jean-Pierre Chanoine,22 Carol K.L. Lam,22
Melinda J. Pierce,23 Laurel L. Williams,24 Lynne M. Bird,25,26 Merlin G. Butler,27
Diane E. Jensen,28,29 Susan E. Myers,30 Oliver J. Oatman,31 Charumathi Baskaran,32,33
Laura J. Chalmers,34 Cary Fu,1 Nathalie Alos,2 Scott D. McLean,8 Ajay Shah,24
Barbara Y. Whitman,30 Brent A. Blumenstein,35 Sarah F. Leonard,36 Jessica P. Ernest,36
Joseph W. Cormier,36 Sara P. Cotter,36 and Davis C. Ryman36
1
Vanderbilt University, Nashville, TN 37240, USA
2
Department of Pediatrics, Centre Hospitalier Universitaire Sainte-Justine Centre de Recherche, Montréal, Québec H3T 1C5, Canada
3
Department of Pediatrics, Section of Genetics and Metabolism, University of Colorado School of Medicine and Children’s Hospital Colorado,
Aurora, CO 80309, USA
4
Department of Psychiatry, The University of Tennessee Health Science Center College of Medicine, Memphis, TN 37996, USA
5
Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL 32611, USA
6
Division of Endocrinology, The Hospital for Sick Children, Toronto M5G 1X8, Canada
7
Department of Pediatrics, University of Toronto, Toronto M5G 1X8, Canada
8
Department of Pediatrics, Baylor College of Medicine, San Antonio, TX 78207, USA
9
Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
10
Neuroendocrine Center, The Children’s Hospital of Philadelphia Division of Endocrinology and Diabetes, Philadelphia, PA 19104, USA
11
Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
12
Genetics and Metabolism, Children’s National Hospital, Washington, DC 20010, USA
13
Division of Pediatric Endocrinology and Diabetes, Children’s of Alabama, Birmingham, AL 35233, USA
14
Department of Pediatrics, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
15
Division of Endocrinology and Diabetes, Nationwide Children’s Hospital, Columbus, OH 43205, USA
16
Department of Pediatrics, The Ohio State University, Columbus, OH 43205, USA
17
Division of Endocrinology, Ann and Robert H. Lurie Children’s Hospital of Chicago, Feinberg School of Medicine, Northwestern University,
Chicago, IL 60208, USA
18
Diabetes & Obesity Program, Center for Endocrinology, Diabetes and Metabolism, Children’s Hospital Los Angeles Department of Pediatrics,
Los Angeles, CA 90027, USA
19
Keck School of Medicine of USC, Los Angeles, CA 90033, USA
20
Department of Pediatrics, Division of Medical Genetics, The University of Utah School of Medicine, Salt Lake City, UT 84112, USA
21
Shriners Hospital for Children, Salt Lake City, UT 84112, USA
22
Department of Pediatrics, Endocrinology and Diabetes Unit, The University of British Columbia, Vancouver V6H 3V4, Canada
23
Diabetes & Endocrinology, Children’s Minnesota—St Paul, St Paul, MN 55404, USA
24
Menninger Department of Psychiatry & Behavioral Sciences, Baylor College of Medicine, Houston, TX 77030, USA
25
Department of Pediatrics, University of California San Diego, San Diego, CA 92037, USA
26
Rady Children’s Hospital, San Diego, CA 92123, USA
27
Department of Psychiatry & Behavioral Sciences, University of Kansas Medical Center, Kansas City, KS 66160, USA
28
Children’s Health Queensland Hospital and Health Services, South Brisbane, Queensland 4101, Australia
29
Centre for Children’s Health Research, University of Queensland, Brisbane, Queensland 4101, Australia
30
Department of Pediatrics, Saint Louis University School of Medicine, Cardinal Glennon Children’s Hospital, Saint Louis, MO 63104, USA
31
Division of Endocrinology and Diabetes, Phoenix Children’s Hospital, Phoenix, AZ 85016, USA
32
Division of Endocrinology, Boston Children’s Hospital, Boston, MA 02115, USA
33
Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA

Received: 30 September 2022. Editorial Decision: 9 January 2023. Corrected and Typeset: 3 February 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 properly cited. For commercial re-use, please contact journals.permissions@oup.com
The Journal of Clinical Endocrinology & Metabolism, 2023, Vol. 108, No. 7 1697

34
Department of Pediatrics, The University of Oklahoma School of Community Medicine, Tulsa, OK 73117, USA
35
Trial Architecture Consulting, Chevy Chase, MD 20814, USA
36
Levo Therapeutics, Inc., Skokie, IL 60077, USA
Correspondence: Davis C. Ryman, MD, PhD, Aeovian Pharmaceuticals, Inc., 156 2nd St, San Francisco, CA 94105, USA. Email: dryman@aeovian.com.

Abstract
Context: Prader-Willi syndrome (PWS) is a rare genetic disorder characterized by endocrine and neuropsychiatric problems including
hyperphagia, anxiousness, and distress. Intranasal carbetocin, an oxytocin analog, was investigated as a selective oxytocin replacement therapy.
Objective: To evaluate safety and efficacy of intranasal carbetocin in PWS.
Design: Randomized, double-blind, placebo-controlled phase 3 trial with long-term follow-up.
Setting: Twenty-four ambulatory clinics at academic medical centers.
Participants: A total of 130 participants with PWS aged 7 to 18 years.

Downloaded from https://academic.oup.com/jcem/article/108/7/1696/6986298 by guest on 20 November 2023


Interventions: Participants were randomized to 9.6 mg/dose carbetocin, 3.2 mg/dose carbetocin, or placebo 3 times daily during an 8-week
placebo-controlled period (PCP). During a subsequent 56-week long-term follow-up period, placebo participants were randomly assigned to
9.6 mg or 3.2 mg carbetocin, with carbetocin participants continuing at their previous dose.
Main outcome measures: Primary endpoints assessed change in hyperphagia (Hyperphagia Questionnaire for Clinical Trials [HQ-CT]) and
obsessive-compulsive symptoms (Children’s Yale-Brown Obsessive-Compulsive Scale [CY-BOCS]) during the PCP for 9.6 mg vs placebo, and
the first secondary endpoints assessed these same outcomes for 3.2 mg vs placebo. Additional secondary endpoints included assessments
of anxiousness and distress behaviors (PWS Anxiousness and Distress Behaviors Questionnaire [PADQ]) and clinical global impression of
change (CGI-C).
Results: Because of onset of the COVID-19 pandemic, enrollment was stopped prematurely. The primary endpoints showed numeric
improvements in both HQ-CT and CY-BOCS which were not statistically significant; however, the 3.2-mg arm showed nominally significant
improvements in HQ-CT, PADQ, and CGI-C scores vs placebo. Improvements were sustained in the long-term follow-up period. The most
common adverse event during the PCP was mild to moderate flushing.
Conclusions: Carbetocin was well tolerated, and the 3.2-mg dose was associated with clinically meaningful improvements in hyperphagia and
anxiousness and distress behaviors in participants with PWS.
Clinical Trials Registration Number: NCT03649477
Key Words: Prader-Willi syndrome, carbetocin, oxytocin, vasopressin, hyperphagia, anxiety
Abbreviations: CARE-PWS, Phase 3 Study of Intranasal Carbetocin (LV-101) in Patients With Prader-Willi Syndrome; CGI-C, clinical global impression of
change; CGI-S, clinical global impression of severity; CY-BOCS, Children’s Yale-Brown Obsessive-Compulsive Scale; EC50, half maximal effective
concentration; LTFU, long-term follow-up; HQ-CT, Hyperphagia Questionnaire for Clinical Trials; PADQ, PWS Anxiousness and Distress Behaviors
Questionnaire; PAS, primary analysis set; PCP, placebo-controlled period; PWS, Prader-Willi syndrome; SAS, safety analysis set; V1aR, vasopressin 1A
receptor.

Prader-Willi syndrome (PWS) is a rare neurodevelopmental Many of the hallmark features of PWS are thought to involve
genetic disorder with a complex phenotype and no approved deficits in neuroendocrine feedback loops that maintain energy
therapies to treat its most serious, life-threatening, and life- homeostasis and influence psychosocial, physical, and sexual
limiting symptoms. It occurs in approximately 1 in 10 000 development (2). PWS is associated with dysfunction and dys­
to 1 in 30 000 births (1, 2). Genetically, PWS is caused by a de­ regulation of multiple hormones and neuropeptides (reviewed
fect in paternal expression of imprinted genes in the chromo­ in 10), which may be causally related to a deficiency in prohor­
some 15q11-q13 region (3). mone convertase 1, resulting in impairment of normal process­
PWS is characterized by profound hyperphagia, which is an ing of multiple prohomones, including the hypothalamic
intense persistent sensation of hunger accompanied by food neuropeptide hormone oxytocin (11). In post mortem brain tis­
preoccupations, an extreme drive to consume food, and sue from individuals with PWS, a 42% decrease in number and
food-related behavior problems. Overt hyperphagia typically a 54% decrease in volume of oxytocin-expressing neurons have
presents by 7 to 8 years of age and is associated with the lack been reported in the paraventricular nucleus of the hypothal­
of a normal satiety response and a low basal metabolic rate amus (12). Mice that are null for Magel2 (one of the genes in
(4-6). Despite adequate energy reserves, these individuals are the 15q11-13 PWS locus that is not expressed in PWS) similarly
subjected to a false state of starvation (7) that can lead to epi­ have decreased levels of oxytocin in the paraventricular nucleus
sodes of food gorging associated with choking or gastric rup­ (11), and perinatal treatment with oxytocin has been shown to
ture if access to food is not carefully controlled. rescue postnatal lethality and social and learning deficits in this
Other psychiatric symptoms in PWS include increased lev­ model (13, 14). Although better known for its role in repro­
els of anxiousness, obsessive-compulsive tendencies, and ductive health, oxytocin is also involved in regulating social
maladaptive behaviors. An analysis of data from 4039 chil­ and emotional behaviors and is a potent anorexigenic hormone
dren and adolescents with genetic disorders associated with involved in the normal satiety response (15).
intellectual disability showed that the prevalence of psychi­ Carbetocin is an oxytocin analog designed to have im­
atric symptoms was the highest for PWS (74%) among the proved oxytocin receptor selectivity and longer half-life com­
10 disorders evaluated (8). To manage hyperphagia and psy­ pared with endogenous oxytocin. It has been approved in
chiatric symptoms, caregivers impose strict environmental more than 100 countries to prevent uterine atony following
controls and constant monitoring of individuals with PWS, cesarean delivery and has been administered to more
significantly limiting their independence. Assessments of than 11 million women since its first approval in 1997.
caregiver burden in PWS exceed those for conditions includ­ Compared with native oxytocin, carbetocin has substantially
ing Alzheimer’s disease and traumatic brain injury (9). greater selectivity to binding to oxytocin receptors compared
1698 The Journal of Clinical Endocrinology & Metabolism, 2023, Vol. 108, No. 7

with vasopressin receptors. In addition to its anorexigenic ef­ endpoint assessed efficacy of the lower, 3.2 mg 3 times daily,
fects, selective modulation of the oxytocin pathway by carbe­ dose of carbetocin vs placebo using these same instruments.
tocin may offer prosocial and anxiolytic benefits to Additional secondary endpoints evaluated efficacy of each
individuals with PWS (16) by virtue of reducing off-target dose (9.6 mg or 3.2 mg 3 times daily) vs placebo in improving
binding to vasopressin receptors and thereby widening the PWS-specific anxiousness and distress behaviors, measured by
therapeutic window for treatment in this population. the PWS Anxiousness and Distress Behaviors Questionnaire
The results of a phase 2 trial (ClinicalTrials.gov identifier: (PADQ), and evaluated efficacy in improving overall clinical
NCT01968187) in 37 individuals with PWS showed 9.6 mg symptoms of PWS, assessed by clinician-rated clinical global
of intranasal carbetocin reconstituted in saline administered impression of change (CGI-C) scores. Prespecified secondary
3 times per day with meals to be safe and well-tolerated endpoints also further evaluated effects on hyperphagia using
(17). Efficacy results over the 14 days of the trial demonstrated 2 different prespecified subsets of the HQ-CT: a subset focus­
significant improvements in hyperphagia and other behavioral ing on items less affected by food-related environmental con­
symptoms in PWS vs the placebo arm, warranting further trols (the HQ-CT subset, including questions 1, 2, 5, 6, 8, and
study as a potential therapeutic option (17). 9) and analysis of scores on HQ-CT question 9 in isolation.

Downloaded from https://academic.oup.com/jcem/article/108/7/1696/6986298 by guest on 20 November 2023


This phase 3 study of intranasal carbetocin (LV-101) in pa­ Prespecified exploratory analyses evaluated improvements
tients with Prader-Willi syndrome (CARE-PWS trial; in clinical global impression of severity (CGI-S) scores and
ClinicalTrials.gov identifier: NCT03649477) was designed long-term assessments of efficacy observed through the week
to evaluate the safety and efficacy of intranasal carbetocin on 64 LTFU trial visit.
behavioral aspects of PWS. CARE-PWS evaluated a novel for­ Exploratory efficacy objectives included assessments of the
mulation of the 9.6 mg dose tested in the phase 2 trial and add­ proportion of participants who exhibited clinically meaning­
itionally evaluated a lower 3.2 mg dose, as a strategy to avoid ful responses to carbetocin vs placebo, and post hoc analyses
potential off-target effects at higher doses. Efficacy of each included within-participant efficacy comparisons of partici­
dose arm was compared with a matching placebo (1:1:1) dur­ pants assigned to placebo and then later to receive 3.2 mg of
ing an 8-week placebo-controlled period (PCP). The trial col­ carbetocin during the LTFU.
lected safety and efficacy data during a subsequent 56-week Safety and tolerability objectives were to establish the safety
long-term follow-up (LTFU) period during which all partici­ profile of carbetocin in individuals with PWS. Assessments in­
pants received either 9.6 mg or 3.2 mg of carbetocin. After cluded evaluations of reported TEAEs and frequency of and
completion of the LTFU, participants were given the option reasons for trial discontinuation.
to continue to receive carbetocin during an extension period.
Participants
Methods Key eligibility criteria included genetically confirmed diagnoses
of PWS and age between 7 and 18 years at screening with a total
Trial Design HQ-CT score ≥13 (18) and total CY-BOCS score ≥9 (19). These
The CARE-PWS trial was a phase 3, randomized, double- thresholds were selected to ensure the presence of sufficient base­
blind, placebo-controlled, parallel-arm, multicenter trial con­ line scores to detect a potential treatment effect and were not
ducted in participants with PWS to evaluate the efficacy and communicated publicly during the study. Participants were re­
safety of intranasal carbetocin compared with placebo. The quired to be under the care and observation of a consistent care­
trial was conducted at 24 hospitals and clinics in the United giver; accordingly, those living in group homes were not eligible.
States, Canada, and Australia, following approval by inde­ Chronic concomitant medications or supplements were required
pendent ethics committees or institutional review boards at to remain stable for at least 3 months before the trial.
each trial center. The trial was conducted in accordance Participants were disqualified from participating if they had tak­
with Good Clinical Practice and the Declaration of Helsinki. en prostaglandins, prostaglandin analogs or agonists, and/or
Informed consent was obtained in writing from the partici­ weight loss medications within 6 months of screening.
pant’s parent or legally acceptable representative, and child Anti-inflammatory medications were allowed. Participants
assent was obtained, as necessary, before the first trial proced­ with a history of oxytocin or carbetocin use were required to
ure. Efficacy analyses were completed using a combination of have abstained from use of these medications within 3 months
clinician-reported and caregiver-reported assessments further of screening. Participants were excluded if they had a history
detailed later in this article. Safety was assessed by evaluation of psychotic symptoms, venous thrombosis or pulmonary em­
of incidence and severity of treatment-emergent adverse bolism, nasal surgery within 6 months of screening, or nasal dis­
events (TEAEs), laboratory assessments, electrocardiograms, orders that could significantly affect drug administration.
vital signs, physical examinations, and nasal assessments. Participants were prohibited from adding new food-related in­
terventions (including environmental or dietary restrictions)
Objectives and Endpoints within 1 month of screening and during the trial, as well as
use of nasal saline, irrigation, or other nasal medications for
The overall objective of the trial was to evaluate the efficacy,
2 weeks before the baseline visit and during the 8-week PCP
safety, and tolerability of 2 different doses of intranasal carbe­
of the trial. Participants who completed the 8-week PCP were
tocin in participants with PWS. The primary endpoints as­
given the option to continue to the LTFU portion of the trial.
sessed the efficacy of 9.6 mg of carbetocin 3 times daily vs
placebo in improving hyperphagia and obsessive-compulsive
symptoms during the 8-week PCP, measured by change in to­ Randomization and Masking
tal scores on the Hyperphagia Questionnaire for Clinical Trial enrollment began on December 6, 2018, and was closed
Trials (HQ-CT) and Children’s Yale-Brown Obsessive- prematurely on March 12, 2020, before the target enrollment
Compulsive Scale (CY-BOCS), respectively. A secondary number of 175 participants because of the impact of
The Journal of Clinical Endocrinology & Metabolism, 2023, Vol. 108, No. 7 1699

COVID-19. Participants were randomized through an inter­ for an overall assessment of improvement, and was also pre­
active web response system. Randomization was performed specified as a secondary endpoint.
before baseline to give adequate time to thaw vials for the first Obsessive-compulsive symptoms were assessed using the
dose administered at the baseline visit. The randomization ra­ CY-BOCS. This scale is a validated, clinician-administered,
tio was defined to be 2:2:1:1 into 4 trial arms: 9.6 mg carbeto­ semistructured inventory of specific symptoms and severity
cin 3 times daily, during PCP and LTFU; 3.2 mg carbetocin 3 with a total score range of 0 to 36. Reductions in scores reflect
times daily, during PCP and LTFU; placebo 3 times daily dur­ improvements in obsessions and compulsions. The scale is va­
ing the PCP and 9.6 mg carbetocin 3 times daily during LTFU; lidated for diagnosis and evaluation of pediatric obsessive-
and placebo 3 times daily during the PCP and 3.2 mg carbeto­ compulsive disorder (19). It has not been validated specifically
cin 3 times daily during LTFU. Efficacy analyses of the PCP for use in the PWS population.
pooled the 2 placebo arms together, resulting in a 1:1:1 ran­ The PADQ was administered to assess PWS-specific anx­
domization ratio. The sponsor (Levo Therapeutics), investiga­ iousness and distress behaviors. The PADQ is a validated
tors, caregivers, and participants were blinded to the assigned 15-item caregiver-reported instrument designed to capture
treatments during both the PCP and LTFU. To attempt to min­ the observable signs of anxiousness and distress that are com­

Downloaded from https://academic.oup.com/jcem/article/108/7/1696/6986298 by guest on 20 November 2023


imize placebo effects to the extent possible, standardized video mon among individuals with PWS. The total score of 0 to 56 is
training modules aimed at accurate reporting and placebo re­ based on responses to the first 14 items, with higher scores in­
sponse reduction for caregivers and clinicians were developed dicating greater levels of anxiousness and psychological dis­
by an organization specialized in mitigating placebo response tress. The fifteenth question is an overall summary of
in clinical trials (Analgesic Solutions, Wayland, MA). caregivers’ observations and is not included in the total score.
Following completion of the placebo-controlled period and The Clinical Global Impression (CGI) scale is among the
unblinding of efficacy results, all participants were transi­ most widely used brief assessment tools in psychiatry. The
tioned to the 3.2-mg dose in October 2020. CGI provides a clinician rating relative to the clinician’s experi­
The sponsor completed blinded safety reviews throughout ence with other individuals with the same diagnosis (20). This
the trial. An independent data safety monitoring committee scale measures the current severity of illness (CGI-S) and global
periodically reviewed unblinded safety data. No safety events change (CGI-C) in the severity of illness from baseline, each on
were observed during the trial that required unblinding of a a 7-point scale. The CGI-S uses a range of responses from 1
trial participant’s treatment assignment. (normal) to 7 (among the most severely ill). The CGI-C uses
a range of responses from 1 (very much improved) to 7 (very
much worse). The CGI-C instrument was used as a secondary
Procedures endpoint and the CGI-S was a tertiary endpoint.
Investigational product (carbetocin and placebo) was admin­ Safety was monitored throughout the trial. Safety assessments
istered intranasally 3 times per day with meals with a min­ included incidence and severity of adverse events, as well as as­
imum of 2.5 hours between doses. The placebo and both sessment of vital signs, electrocardiograms, physical examina­
doses of carbetocin were matched for taste and odor to pre­ tions, nasal examinations, and laboratory assessments
vent unblinding and were identical in all properties except (including chemistry, hematology, coagulation, and urinalysis).
for the absence of the active ingredient in the placebo vials.
Each vial provided 3 doses of study drug or placebo that
was to be used with an attachable nasal spray pump. Each Statistical Analyses
dose consisted of 2 sprays in each nostril, for a total of Results from the phase 2 trial of carbetocin in PWS were used
4 sprays. Administration was assisted by the caregiver or a as the basis for powering this trial (17). Planned enrollment of
helper (in the event the participant was at school or other ac­ 175 participants would confer ≥90% power for the HQ-CT
tivities during administration). and ≥99% power for the CY-BOCS to detect a treatment
Efficacy and safety were assessed at baseline, during the arm difference in the total score change from baseline to
PCP trial visits at week 2 and week 8, and during the LTFU week 8 in the 9.6-mg arm vs placebo. The trial type I error
period trial visits at weeks 10, 16, 28, 40, 52, and 64. probability (alpha) was specified to be 2-sided 0.05, with
Caregivers or designees were required to record all doses of multiplicity control for the 2 primary outcomes using the
study drug administration on a paper dosing diary and return Hochberg step-up procedure (21, 22), thereby potentially
diaries to the site staff at trial visits for confirmation of treat­ leading to primary outcome success if either or both outcomes
ment compliance. During the COVID-19 pandemic, trial visits met statistical criteria.
were conducted remotely via telemedicine, if necessary. Analyses included three analysis sets: the primary analysis
Participants’ hyperphagia symptoms were assessed using set (PAS), the full analysis set, and the safety analysis set
the HQ-CT, which is currently considered the consensus in­ (SAS). Following consultation with the Food and Drug
strument for evaluation of hyperphagia behavior in PWS. Administration and before unblinding, the PAS was defined
The HQ-CT is a 9-item validated questionnaire (total score to include all participants who completed at least 1 post-base­
range, 0-36) designed to be completed by caregivers of indi­ line visit (ie, week 2 or week 8) before March 1, 2020, and was
viduals with PWS and which has been used in other PWS clin­ used for all efficacy analyses in the PCP. This data cutoff date
ical trials (18). Reductions in scores reflect improvements in was used to minimize the potential confounding effects of the
hyperphagia. A subset of the HQ-CT items (including ques­ onset of the COVID-19 pandemic on behavioral measures in
tions 1, 2, 5, 6, 8, and 9), which are less susceptible to envir­ this sensitive population, resulting in 68% of the intended trial
onmental controls imposed by caregivers, was used as a size. Data from those withdrawing before the cutoff date were
secondary efficacy endpoint. HQ-CT question 9 asks the care­ included in the PAS even if no postbaseline visits were re­
giver to rate the overall level of interference that hyperphagia corded. The full analysis set included all participants who
causes in the daily life of the individual with PWS, can be used were randomized and dosed with study medication and was
1700 The Journal of Clinical Endocrinology & Metabolism, 2023, Vol. 108, No. 7

Downloaded from https://academic.oup.com/jcem/article/108/7/1696/6986298 by guest on 20 November 2023


Figure 1. CARE-PWS study CONSORT diagram. Participants were screened, randomized, and enrolled in an 8-week placebo-controlled study period
followed by a subsequent 56-week long-term follow-up period. A total of 130 participants received carbetocin or placebo.

used for efficacy analyses in the LTFU period. TEAE incidence but before enrollment confirmation at the baseline visit.
and severity, discontinuations, and other safety measures used Eight randomized participants failed screening criteria or
the SAS, which included all participants who received at least withdrew consent at the baseline visit and were therefore ex­
1 dose of study drug. cluded from enrollment before receiving the investigational
Efficacy analyses used the constrained longitudinal data product, for a total of 130 participants receiving at least
analysis mixed model with repeated measures (23-25) without 1 dose of the study medication. Of those 130 participants,
imputation. Sensitivity analyses were performed to confirm 128 completed their week 8 visit (2 participants in the
robustness of results to missing data and demonstrate consist­ 9.6-mg dose arm discontinued before week 8) and 103 com­
ency in results and conclusions. Statistical comparisons were pleted the trial (17 participants in the 9.6-mg LTFU dose
conducted using SAS, version 9.4 (SAS Institute, Cary, NC). arm and 8 participants in the 3.2-mg LTFU dose arm discon­
Within-participant response to treatment for the HQ-CT tinued before their week 64 visit). The PAS included 119
was evaluated as the proportion of participants that achieve evaluable participants with at least 1 postbaseline visit before
a 7.7-point reduction in total score, a responder threshold de­ the March 1, 2020, data cutoff date, 104 of which had com­
fined when validating the HQ-CT instrument (18) and con­ pleted their week 8 visit before the cutoff date.
sistent with anchor-based analyses of the CARE-PWS data. Randomization resulted in treatment arms that were gener­
Within-participant response to treatment for the PADQ was ally well-balanced with respect to baseline demographics as
similarly evaluated as the proportion of participants that well as baseline PWS characteristics (Table 1).
achieve a 10.5-point reduction in total score (the responder
threshold identified when validating the PADQ instrument
and using anchor-based analyses of the CARE-PWS data). Efficacy Outcomes
For placebo-crossover analyses, mean changes in HQ-CT Participants taking carbetocin showed numerical improve­
and PADQ from baseline to week 8 were compared with ments vs placebo in 11 of the 12 specified primary and second­
mean changes from week 8 to week 16 in participants that ary endpoints (Table 2). The 2 primary endpoints assessing
transitioned from placebo to active treatment arms during change in HQ-CT and CY-BOCS for the 9.6-mg carbetocin
the LTFU period. arm showed numeric improvements vs placebo that did not
All efficacy results analyzed participants according to the reach statistical significance; therefore, the trial did not meet
dose arm to which they were originally assigned. Analyses its primary endpoint and all other statistical analyses are con­
of safety data included assessments of TEAE incidence rates sidered nominal. Results for the 3.2 mg dose arm showed con­
and percentages of participants in each arm who experienced sistent and nominally significant improvements in multiple
a TEAE, including severity and relatedness to treatment. efficacy assessments compared with placebo (Table 2 and
Fig. 2). Specifically, benefits were observed in symptoms of hy­
perphagia (HQ-CT −3.1, P = 0.016; HQ-CT subset −2.4, P =
Results 0.011), anxiousness and distress (PADQ −3.8, P = 0.027), and
A total of 176 participants entered screening and 138 of those clinical global impressions of change (CGI-C −0.5, P = 0.027).
participants were randomized to receive study treatment Responder analyses for the 3.2-mg dose further supported the
(Fig. 1). Because intranasal carbetocin required overnight existence of clinically meaningful improvements in participants’
thawing before use, randomization occurred after screening, hyperphagia, anxiousness, and distress symptoms, with
The Journal of Clinical Endocrinology & Metabolism, 2023, Vol. 108, No. 7 1701

Table 1. Baseline demographics and PWS characteristics (primary analysis set)

Placebo-controlled period

3.2 mg 9.6 mg All placebo


(N = 39) (N = 40) (N = 40) P value

Age, y
Mean (SD) 12.3 (3.12) 11.7 (3.45) 11.8 (3.52) 0.675
Sex, n (%)
Female 24 (61.5) 19 (47.5) 23 (57.5) 0.432
Male 15 (38.5) 21 (52.5) 17 (42.5)
Race, n (%)
American Indian/Alaska Native 0 1 (2.5) 0 0.820

Downloaded from https://academic.oup.com/jcem/article/108/7/1696/6986298 by guest on 20 November 2023


Asian 2 (5.1) 2 (5.0) 0
Black/African American 2 (5.1) 1 (2.5) 1 (2.5)
White 33 (84.6) 33 (82.5) 35 (87.5)
Other 2 (5.1) 3 (7.5) 4 (10.0)
Ethnicity, n (%)
Hispanic or Latino 5 (12.8) 3 (7.5) 3 (7.5) 0.643
Not Hispanic or Latino 34 (87.2) 37 (92.5) 37 (92.5)
Baseline weight, kg
Mean (SD) 58.98 (24.5) 61.36 (32.5) 53.59 (25.5) 0.347
Baseline BMI
Mean (SD), kg/m2 26.43 (8.3) 26.30 (9.2) 24.35 (8.8) 0.453
z-score mean (SD) 1.45 (0.96) 1.52 (0.86) 1.14 (1.00) 0.164
Genetic subtype of PWS, n (%)
Deletion type 1 5 (12.8) 2 (5.0) 3 (7.5) 0.858
Deletion type 2 2 (5.1) 5 (12.5) 5 (12.5)
Deletion type unknown 16 (41.0) 19 (47.5) 14 (35.0)
UPD 9 (23.1) 8 (20.0) 9 (22.5)
ID 1 (2.6) 1 (2.5) 3 (7.5)
Unknown 6 (15.4) 5 (12.5) 6 (15.0)
Baseline total scores, mean (SD)
HQ-CT 22.1 (5.1) 23.4 (5.7) 22.4 (4.7) 0.536
CY-BOCS 25.5 (4.1) 28.4 (4.0) 27.8 (6.0) 0.606
PADQ 43.1 (6.9) 42.6 (7.2) 43.9 (6.7) 0.706

Abbreviations: BMI, body mass index; CY-BOCS, Children’s Yale-Brown Obsessive-Compulsive Scale; HQ-CT, Hyperphagia Questionnaire for Clinical
Trials; ID, imprinting defect; PADQ, PWS Anxiousness and Distress Behaviors; PWS, Prader-Willi syndrome; UPD, uniparental disomy.

meaningful responses occurring at significantly higher rates in 8 weeks on active drug when compared with their experi­
participants on carbetocin than those on placebo. Responder ence during their 8 weeks on placebo (Table 3), consistent
thresholds for these instruments were defined using an anchor- with the observations seen during the PCP for active vs pla­
based approach considering changes in CGI-C and correlating cebo. The corresponding comparisons for participants tran­
those changes to changes in questionnaire total scores. In the ag­ sitioning to 9.6 mg were unremarkable and therefore
gregate, these methods provide a range of 4- to 9-point improve­ consistent with what was observed in the PCP for 9.6 mg
ments for HQ-CT and a range of 6- to 13-point improvements participants.
in PADQ. Conclusions from responder analyses for HQ-CT to­ Improvements from baseline in efficacy assessments per­
tal scores are robust across a range of potential thresholds sisted throughout the long-term follow-up period (Fig. 4).
(Fig. 3A). Specifically, responder rates for the 3.2-mg dose con­ Notably, although the 3.2 mg dose continued to provide
sistently favored active treatment with odds ratios exceeding 3 equivalent or superior efficacy to that of the 9.6 mg dose,
for all thresholds and P values (2-sided Fisher exact test) below even participants taking the 9.6 mg dose who chose to stay
0.05 for all but the highest threshold. Responder analyses for in the trial continued to experience substantial, clinically
PADQ total scores are shown in Fig. 3B, and again consistently meaningful, and sustained improvements from baseline
favored the 3.2-mg dose with P values (2-sided Fisher exact test) throughout the LTFU period. These observations were con­
below 0.05 for all but 2 thresholds of improvement. sistent across both doses and across multiple efficacy assess­
Participants transitioning from placebo to the 3.2 mg dose ments including the HQ-CT (Fig. 4A), CY-BOCS (Fig. 4B),
arm at the end of the placebo-controlled period exhibited and PADQ (Fig. 4C). Although there appeared to be a numer­
substantial improvements in hyperphagia scores in their first ically greater improvement in the placebo-to-9.6-mg
1702 The Journal of Clinical Endocrinology & Metabolism, 2023, Vol. 108, No. 7

Table 2. Primary and secondary efficacy results

3.2-mg carbetocin 9.6-mg carbetocin Placebo


(N = 39) (N = 40) (N = 40)

HQ-CT
Baseline mean (SD) 22.1 (5.09) 23.4 (5.67) 22.4 (4.74)
Change from baseline to week 8
LS mean (SE) −5.4 (0.96) −3.4 (0.95) −2.2 (0.94)
LS mean difference vs placebo −3.1 −1.2 —
(95% CI of LS mean differences) (−5.7 to −0.6) (−3.7 to 1.3) —
Two-sided P value vs placebo 0.016 0.349 —
CY-BOCS

Downloaded from https://academic.oup.com/jcem/article/108/7/1696/6986298 by guest on 20 November 2023


Baseline mean (SD) 25.5 (4.10) 28.4 (3.97) 27.8 (6.01)
Change from baseline to week 8
LS mean (SE) −3.1 (0.87) −3.0 (0.86) −2.4 (0.86)
LS mean difference vs placebo −0.8 −0.6 —
(95% CI of LS mean differences) (−3.1 to 1.5) (−2.9 to 1.7) —
Two-sided P value vs placebo 0.514 0.600 —
PADQ
Baseline mean (SD) 43.1 (6.94) 42.6 (7.23) 43.9 (6.74)
Change from baseline to week 8
LS mean (SE) −8.3 (1.26) −4.3 (1.26) −4.5 (1.24)
LS mean difference vs placebo −3.8 0.2 —
(95% CI of LS mean differences) (−7.2 to −0.4) (−3.2 to 3.5) —
Two-sided P value vs placebo 0.027 0.914 —
CGI-C
Week 8 mean (SD) 3.4 (0.95) 3.6 (0.98) 3.9 (0.87)
Week 8
LS mean (SE) 3.4 (0.17) 3.6 (0.17) 3.9 (0.17)
LS mean difference vs placebo −0.5 −0.3 —
(95% CI of LS mean differences) (−0.9 to −0.1) (−0.7 to 0.1) —
Two-sided P value vs placebo 0.027 0.160 —
HQ-CT subset
Baseline mean (SD) 17.4 (3.66) 17.8 (3.76) 17.3 (3.66)
Change from baseline to week 8
LS mean (SE) −4.6 (0.71) −3.3 (0.70) −2.2 (0.69)
LS mean difference vs placebo −2.4 −1.1 —
(95% CI of LS mean differences) (−4.3 to −0.5) (−2.9 to 0.8) —
Two-sided P value vs placebo 0.011 0.248 —
HQ-CT question 9
Baseline mean (SD) 2.8 (1.33) 3.3 (0.88) 3.1 (1.08)
Change from baseline to week 8
LS mean (SE) −0.9 (0.18) −0.5 (0.18) −0.5 (0.18)
LS mean difference vs placebo −0.4 −0.1 —
(95% CI of LS mean differences) (−0.9 to 0.1) (−0.5 to 0.4) —
Two-sided P value vs placebo 0.114 0.832 —

Primary efficacy endpoints were HQ-CT and CY-BOCS for the 9.6-mg dose; the first secondary endpoints were HQ-CT and CY-BOCS for the 3.2-mg dose. P
values < 0.05 are bolded. Abbreviations: CGI-C, clinician-rated clinical global impression of change; CY-BOCS, Children’s Yale-Brown Obsessive-Compulsive
Scale; HQ-CT = Hyperphagia Questionnaire for Clinical Trials; LS, least squares; PADQ, Prader-Willi Syndrome Anxiousness and Distress Behaviors
Questionnaire; SE, standard error.

subgroup vs the placebo-to-3.2-mg subgroup late in the LTFU participants, including those initially randomized to placebo
period, these data should be interpreted with caution because (Fig. 4).
of small sample sizes in these subgroups and the confounding
effect of survivor bias, and in particular because the number of Safety Results
discontinuations was disproportionate among those random­ In the PCP, 63.2% of participants receiving carbetocin, in­
ized to 9.6 mg. Overall, results were broadly similar for all cluding 60.5% of participants (n = 26) in the 3.2-mg arm
The Journal of Clinical Endocrinology & Metabolism, 2023, Vol. 108, No. 7 1703

Downloaded from https://academic.oup.com/jcem/article/108/7/1696/6986298 by guest on 20 November 2023


Figure 2. Effects of 3.2 mg/dose carbetocin vs placebo across efficacy assessments in the placebo-controlled period. Changes from baseline to week
8 in study efficacy assessments are shown for the carbetocin 3.2-mg group compared with placebo. Note: questionnaires are of differing scales.
Abbreviations: CGI-Change, clinical global impression of change; CY-BOCS, Children’s Yale-Brown Obsessive-Compulsive Scale; HQ-CT, Hyperphagia
Questionnaire for Clinical Trials; PADQ, PWS Anxiousness and Distress Behaviors Questionnaire.

and 65.9% of participants (n = 29) in the 9.6-mg arm, re­ trial discontinuation (2 of the discontinuations occurred in the
ported at least 1 TEAE, compared with 55.8% of participants PCP and 1 occurred in the LTFU period).
(n = 24) in the placebo arm (Table 4). Most TEAEs were mild Although the rates of adverse events were generally similar
to moderate in intensity. Only 1 severe TEAE was reported between dose arms, discontinuations from behavior-related
during the placebo-controlled period in the 9.6-mg arm adverse events among participants taking 9.6 mg carbetocin
(2.3%); the participant experienced an episode of severe sinus were higher than those taking 3.2 mg (n = 6 vs 2, respectively),
pain during landing of an airplane flight while suffering from a and all reported TEAEs of aggression in both the PCP and
sinus infection, which subsequently resolved with oral LTFU period were in participants in the 9.6 mg/dose arm
antibiotics. (n = 1 and 3, respectively). This is consistent with discontinu­
The most frequently reported TEAE by preferred term dur­ ations from the trial generally, which were also more common
ing the PCP was flushing (17.2%), which was mild to moder­ among participants receiving 9.6 mg carbetocin vs those re­
ate in severity. The incidence of flushing was lower in ceiving 3.2 mg (n = 19 and 8, respectively; Fig. 1).
participants in the 3.2 mg arm (14.0%) compared with the Evaluation of additional safety data including laboratory
9.6 mg arm (20.5%), and no participants in the placebo arm assessments (hematology, coagulation, clinical chemistry,
reported a TEAE of flushing. The occurrence of transient and urinalysis), physical examination findings, Tanner sta­
flushing is consistent with previous reports that oxytocin ging, electrocardiograms, weight, body mass index, and vital
and oxytocin agonists can be associated with transient periph­ signs did not reveal clinically relevant trends associated with
eral vasodilation (26). Flushing was also the most frequently study medication.
reported TEAE considered to be possibly related to study
drug by the investigator during the placebo-controlled period.
In all participants, flushing was transient, mild in intensity, Discussion
not serious, and self-limited (generally resolving spontaneous­ The therapeutic rationale for investigating carbetocin as a po­
ly within 30 minutes). tential treatment for PWS is based on its role as a selective oxy­
The most frequently reported TEAEs during the LTFU and tocin receptor agonist with prolonged half-life relative to
extension periods were nasopharyngitis (10.9% of all partic­ native oxytocin itself, potentially enabling replacement of
ipants, including 9.4% and 12.1% of participants in the the functional oxytocin deficiency in PWS while minimizing
3.2 mg and the 9.6 mg arms, respectively), headache (11.7% off-target effects related to oxytocin’s lack of selectivity that
of all participants, including 15.6% and 7.8% of participants may have limited previous attempts at treatment.
in the 3.2 mg and the 9.6 mg arms, respectively), and epistaxis To date, 5 clinical studies have reported the effects of intra­
(10.9% of all participants and for both dose arms). In com­ nasal oxytocin in PWS (27-31). Oxytocin replacement has
parison, during the placebo-controlled period, the incidence shown varying degrees of efficacy, but has also been associ­
of nasopharyngitis was 1.1% in the LV-101 all active arm ated with an increase in temper outbursts after 8 weeks of
(0 in the 3.2-mg/dose arm and 2.3% in the 9.6 mg/dose treatment, driven by significant increases in outbursts ob­
arm) and 7.0% in the placebo arm. served in participants with PWS receiving higher doses
No deaths or treatment-emergent serious adverse events (27). It has been hypothesized that the off-target effects of
were reported during the PCP. No study drug discontinua­ oxytocin binding to vasopressin receptors may be limiting
tions were reported during the PCP by participants in the pla­ its therapeutic window in PWS (27). Because of oxytocin’s
cebo arm or the 3.2 mg arm. A total of 3 participants (6.8%) lack of selectivity, off-target activation of the vasopressin
in the 9.6-mg arm reported TEAEs during the PCP that led to 1A receptor (V1aR) with high doses of oxytocin may
1704 The Journal of Clinical Endocrinology & Metabolism, 2023, Vol. 108, No. 7

Downloaded from https://academic.oup.com/jcem/article/108/7/1696/6986298 by guest on 20 November 2023


Figure 3. HQ-CT and PADQ responders to carbetocin. Proportions of participants reaching various thresholds of change from baseline to week 8 in (A)
HQ-CT and (B) PADQ scores are shown for the carbetocin 3.2-mg group compared with placebo. Note: all participants with missing week 8 data are
considered nonresponders at all thresholds (even if because of the March 1, 2020, COVID-19 cutoff date). Abbreviations: HQ-CT, Hyperphagia
Questionnaire for Clinical Trials; PADQ, PWS Anxiousness and Distress Behaviors Questionnaire.

Table 3. Placebo-crossover participants

Placebo-to-3.2-mg carbetocin Placebo-to-9.6-mg carbetocin

Baseline to week 8 (n = 20) Week 8 to week 16 (n = 18) Baseline to week 8 (n = 20) Week 8 to week 16 (n = 17)

HQ-CT, LS mean (SE) 0.3 (1.37) −9.1 (1.64) −4.8 (1.41) −5.0 (1.69)
vs first 8 weeks, LS mean −9.3 −0.2
(95% CI) (−14.5 to −4.2) (−5.5 to 5.1)
P value <0.001 0.944

Abbreviations: LS, least squares; HQ-CT, Hyperphagia Questionnaire for Clinical Trials; SE, standard error.

exacerbate PWS behavioral symptoms and mute or negate vasopressin type 2 receptor can be associated with increased
any positive efficacy derived from oxytocin receptor agonism risks of antidiuresis and hyponatremia, which together can
because the V1aR has been implicated in anxiety and depres­ have significant adverse health consequences. The therapeut­
sion (16, 32). Additionally, oxytocin activation of the ic utility of native oxytocin may be further limited by its short
The Journal of Clinical Endocrinology & Metabolism, 2023, Vol. 108, No. 7 1705

Downloaded from https://academic.oup.com/jcem/article/108/7/1696/6986298 by guest on 20 November 2023

Figure 4. Long-term efficacy data from week 8 through week 64. Data from study efficacy questionnaires (A) HQ-CT, (B) CY-BOCS, and (C) PADQ
throughout the long-term follow-up period are shown for each treatment group. Abbreviations: CY-BOCS, Children’s Yale-Brown
Obsessive-Compulsive Scale; HQ-CT, Hyperphagia Questionnaire for Clinical Trials; PADQ, PWS Anxiousness and Distress Behaviors Questionnaire.

half-life of approximately 3 to 5 minutes, making sustained CARE-PWS is the first phase 3 trial of intranasal carbetocin
and selective action at the oxytocin receptor extremely chal­ in individuals with PWS, and is one of the largest intervention­
lenging to achieve. al trials conducted in this population to date. This trial builds
1706 The Journal of Clinical Endocrinology & Metabolism, 2023, Vol. 108, No. 7

Table 4. TEAEs reported during the PCP by ≥5% of participants in any long-term treatment, with a substantially lower discontinu­
treatment by preferred term ation rate observed in this arm.
As with all clinical studies of potential treatments for PWS,
MedDRA 3.2-mg 9.6-mg All active All placebo statistical power is limited by the number of participants that
preferred term carbetocin carbetocin (N = 87) (N = 43)
can reasonably be enrolled from this rare population.
(N = 43) (N = 44) n (%) n (%)
n (%) n (%) Although the active arms did appear to numerically separate
from placebo, a placebo effect was present, as is commonly
At least 1 TEAE 26 (60.5) 29 (65.9) 55 (63.2) 24 (55.8) observed in clinical trials with behavioral endpoints, making
Flushing 6 (14.0) 9 (20.5) 15 (17.2) 0 statistical significance more challenging to achieve. The sam­
Headache 7 (16.3) 4 (9.1) 11 (12.6) 3 (7.0) ple size of CARE-PWS, specifically, was smaller than planned
Epistaxis 1 (2.3) 6 (13.6) 7 (8.0) 1 (2.3) because of needing to prematurely close enrollment at the out­
break of the COVID-19 pandemic. In general, smaller sample
Diarrhea 4 (9.3) 2 (4.5) 6 (6.9) 1 (2.3)
sizes make it more difficult to detect treatment differences be­
Upper respiratory 3 (7.0) 2 (4.5) 5 (5.7) 2 (4.7)
tween drug and placebo and increase the probability that a

Downloaded from https://academic.oup.com/jcem/article/108/7/1696/6986298 by guest on 20 November 2023


tract infection
false-negative trial result may be obtained.
Nasal discomfort 3 (7.0) 2 (4.5) 5 (5.7) 1 (2.3)
We note that the previous phase 2 study of 9.6 mg/dose car­
Fatigue 3 (7.0) 1 (2.3) 4 (4.6) 0 betocin showed a statistically significant improvement in hy­
Pyrexia 3 (7.0) 0 3 (3.4) 0 perphagia scores, whereas the phase 3 study showed a
Nasopharyngitis 0 1 (2.3) 1 (1.1) 3 (7.0) smaller trend for improvement in this dose group that did
not reach significance. Although the potential reasons for
Note: A TEAE is defined as an AE occurring after the initial dose of IP; this are difficult to determine definitively, contributing factors
participants may have more than 1 TEAE per SOC and/or PT. At each level
of participant summarization (SOC and PT), participants who experienced 1 may include greater variability because of increased numbers
or more AEs within that level are only counted once for that level; the PTs of study sites (24 vs 3) and differences in the measurement of
included are only those reported by ≥5% participants in any treatment, thus, hyperphagia (the HQ-CT vs a different hyperphagia question­
addition of the numbers in the SOC and/or PT rows does not equal the
number in the “at least 1 TEAE” row. naire, the Hyperphagia in PWS Questionnaire Responsiveness
Abbreviations: AE, adverse event; MedDRA, Medical Dictionary for [HPWSQ-R], which had more and differently worded
Regulatory Activities; IP, Investigational Product; PCP, placebo-controlled
period; PT, Preferred Term; SOC, System Organ Class; TEAE,
questions).
treatment-emergent adverse event. The current study also unexpectedly observed superior effi­
cacy in the 3.2 mg/dose arm relative to the 9.6 mg/dose arm.
on the encouraging results of an earlier phase 2 trial and sup­ Although the reasons for this are not known conclusively,
ports that carbetocin can meaningfully improve both hyper­ we note that relevant prior literature regarding use of exogen­
phagia and anxiousness and distress behaviors in individuals ous oxytocin in PWS suggest that increased off-target vaso­
with PWS while maintaining a favorable safety profile with pressin receptor agonism at high doses may be associated
chronic use. with increased emotional outbursts, which, in turn, may neg­
Two doses were tested in this phase 3 trial, using the higher ate the ability of an individual to experience the beneficial be­
dose of 9.6 mg for the primary endpoints and the lower 3.2 mg havioral effect of oxytocin receptor agonism (27).
dose for the first secondary endpoints. The primary analysis of Cell-based functional reporter assays reveal that carbetocin
the higher 9.6 mg dose arm demonstrated numeric improve­ has substantially greater affinity for oxytocin receptors com­
ments in HQ-CT and CY-BOCS scores vs placebo, which pared with vasopressin receptors (half maximal effective con­
did not reach statistical significance in this truncated trial; centration [EC50] of 0.7, 41, and 172 nM for the oxytocin,
therefore, the trial did not meet its primary endpoint. V1a, and V2 receptors, respectively), whereas oxytocin itself
However, the first secondary endpoint evaluating efficacy of has similar affinities for oxytocin receptors and the V1a and
the 3.2 mg dose arm demonstrated meaningful and nominally V2 vasopressin receptor subtypes (EC50 of 2.3, 10, and
significant improvements in hyperphagia vs placebo. The 7 nM, respectively). In addition to the observed differences
3.2 mg dose additionally showed consistent improvements in EC50, carbetocin also appears to have less functional activ­
vs placebo across multiple secondary endpoints, including as­ ity on the V1aR following receptor binding (maximum pos­
sessments of anxiousness and distress behaviors. These im­ sible effect of 24% vs 88% with oxytocin). Although the
provements were subsequently maintained throughout the enhanced selectivity of carbetocin may mitigate off-target ef­
observed 56-week follow-up period and were clinically mean­ fects, carbetocin nonetheless has some vasopressin receptor
ingful. As additional clinical context, in a natural history activity (eg, V1aR) at higher doses, which may mute efficacy
study of individuals with PWS living in the United States given the overlapping nature of the behaviors measured. As
(33), HQ-CT scores were not observed to change significantly another potential hypothesis, if higher doses were associated
between baseline and 6 months of follow-up, and most indi­ with greater desensitization of oxytocin receptors, this could
viduals’ HQ-CT score changes were between 0 to 2. also potentially contribute to the observed difference in effi­
The safety data support that intranasal carbetocin is safe cacy. Such potential unintended effects of oxytocin agonists
and well tolerated in participants with PWS, consistent with at high doses may explain why, although numerical improve­
the existing favorable safety profile of IV carbetocin in clinical ments were observed in the 9.6 mg/dose arm, they were lesser
use for obstetric indications. The most frequent TEAE ob­ than those observed in the 3.2 mg/dose arm and did not reach
served during the placebo-controlled period was flushing, statistical significance.
which was mild and self-limited. Consistent with the hypoth­ Despite limitations including an unplanned truncation of
esis that the lower 3.2 mg dose may be associated with lower enrollment because of the COVID-19 pandemic, the
off-target activity, trial data also suggest a more favorable CARE-PWS trial demonstrates that 3.2 mg intranasal carbeto­
long-term tolerability profile for 3.2 mg carbetocin during cin is associated with clinically meaningful and sustained
The Journal of Clinical Endocrinology & Metabolism, 2023, Vol. 108, No. 7 1707

improvements in hyperphagia and behaviors of anxiousness associated with intellectual disability. J Am Acad Child Adolesc
and distress in individuals with PWS and demonstrates a fa­ Psychiatry. 2020;59(9):1036-1048.
vorable safety profile. The highly positive benefit/risk relation­ 9. Kayadjanian N, Schwartz L, Farrar E, Comtois KA, Strong TV.
ship in the face of substantial unmet medical needs of High levels of caregiver burden in Prader-Willi syndrome. PLoS
One. 2018;13(3):e0194655.
individuals with PWS warrant continued investigation of car­
10. Correa-da-Silva F, Fliers E, Swaab D, Yi C. Hypothalamic neuro­
betocin as a potential treatment.
peptides and neurocircuitries in Prader Willi syndrome. J
Neuroendocrinol. 2021;33(7):e12994.
Acknowledgments 11. Burnett L, LeDuc C, Sulsona C, et al. Deficiency in prohormone
convertase PC1 impairs prohormone processing in Prader-Willi
Levo Therapeutics and the investigators would like to thank syndrome. J Clin Invest. 2017;127(1):293-305.
all of the clinical site support staff who assisted with coordin­ 12. Swaab DF. Prader-Willi syndrome and the hypothalamus. Acta
ation and collection of trial data and, most importantly, the Paediatr Suppl. 1997;86(S423):50-54.
individuals with PWS and their families who participated in 13. Schaller F, Watrin F, Sturny R, Massacrier A, Szepetowski P,
the trial, as well as Scientific Commercialization for their as­ Muscatelli F. A single postnatal injection of oxytocin rescues the le­

Downloaded from https://academic.oup.com/jcem/article/108/7/1696/6986298 by guest on 20 November 2023


sistance with medical writing and author review collation. thal feeding behaviour in mouse newborns deficient for the im­
printed Magel2 gene. Hum Mol Genet. 2010;19(24):4895-4905.
14. Meziane H, Schaller F, Bauer S, et al. An early postnatal oxytocin
Disclosures treatment prevents social and learning deficits in adult mice defi­
E.R., C.L.D., S.E.M., R.L.C., J.L.M., J.K.K., E.R.R., S.E. cient for Magel2, a gene involved in Prader-Willi syndrome and aut­
McCandless, T.R.L., H.D.A., A.M.H., K.S.O., L.C.T., ism. Biol Psychiatry. 2015;78(2):85-94.
A.P.V., D.H.V., J.C., C.K.L., M.P., L.L.W., L.M.B., M.G.B., 15. Jurek B, Neumann ID. The oxytocin receptor: from intracellular
D.E.J., S.E. McCormack, O.J.O., C.B., L.J.C., C.F., N.A., signaling to behavior. Physiol Rev. 2018;98(3):1805-1908.
16. Neumann LC, Markaki Y, Mladenov E, Hoffmann D, Buiting K,
S.D.M., A.S., and B.Y.W. received research funding to their
Horsthemke B. The imprinted NPAP1/C15orf2 gene in the
institutions from Levo Therapeutics for the conduct of this
Prader-Willi syndrome region encodes a nuclear pore complex asso­
study. E.R., S.E. McCandless, C.L.D., and B.A.B. received ciated protein. Hum Mol Genet. 2012;21(18):4038-4048.
consulting fees from Levo Therapeutics. A.M.H. is an investi­ 17. Dykens EM, Miller J, Angulo M, et al. Intranasal carbetocin re­
gator for trials sponsored by Rhythm Pharmaceuticals. S.F.L., duces hyperphagia in individuals with Prader-Willi syndrome. JCI
J.P.E., J.W.C., D.C.R., and S.P.C. were employees of Levo Insight. 2018;3(12):e98333.
Therapeutics. 18. McCandless SE, Yanovski JA, Miller J, et al. Effects of MetAP2 in­
hibition on hyperphagia and body weight in Prader-Willi syn­
drome: a randomized, double-blind, placebo-controlled trial.
Data Availability Diabetes Obes Metab. 2017;19(12):1751-1761.
Restrictions apply to the availability of some or all data gen­ 19. Scahill L, Riddle MA, McSwiggin-Hardin M, et al. Children’s
erated or analyzed during this study to preserve patient confi­ Yale-Brown obsessive compulsive scale: reliability and validity. J
dentiality or because they were used under license. The Am Acad Child Adolesc Psychiatry. 1997;36(6):844-852.
corresponding author will on request detail the restrictions 20. Busner J, Targum SD. The clinical global impressions scale: apply­
and any conditions under which access to some data may be ing a research tool in clinical practice. Psychiatry (Edgmont).
2007;4(7):28-37.
provided.
21. Hochberg Y. A sharper Bonferroni procedure for multiple tests of
significance. Biometrika. 1988;75(4):800-802.
References 22. Huang Y. Hochberg’s step-up method: cutting corners off Holm’s
step-down method. Biometrika. 2007; 94(4):965-975.
1. Cassidy SB, Schwartz S, Miller JL, Driscoll DJ. Prader-Willi syn­
23. Liang K-Y, Zeger SL. Longitudinal data analysis of continuous and
drome. Genet Med. 2012;14(1):10-26.
discrete responses for pre-post designs. Sankhyā : Indian J Stat Ser
2. Driscoll DJ, Miller JL, Schwartz S, Cassidy SB. Prader-Willi syn­
drome. In: Adam MP, Everman DB, Mirzaa GM, Pagon RA, B. 2000; 62(B):134-148.
24. Liu GF, Lu K, Mogg R, Mallick M, Mehrotra DV. Should baseline
Wallace SE, Bean LJH, et al., eds. GeneReviews((R)) University
of Washington; 1993:1-37. be a covariate or dependent variable in analyses of change from
3. Butler MG, Miller JL, Forster JL. Prader-Willi syndrome—clinical baseline in clinical trials? Stat Med. 2009;28(20):2509-2530.
genetics, diagnosis and treatment approaches: an update. Curr 25. Coffman CJ, Edelman D, Woolson RF. To condition or not condi­
Pediatr Rev. 2019;15(4):207-244. tion? Analysing ‘change’ in longitudinal randomised controlled tri­
4. Alsaif M, Elliot SA, MacKenzie ML, Prado CM, Field CJ, Haqq als. BMJ Open. 2016;6(12):e013096.
AM. Energy metabolism profile in individuals with Prader-Willi 26. Petersson M. Cardiovascular effects of oxytocin. Prog Brain Res.
syndrome and implications for clinical management: a systematic 2002;139(1):281-288.
review. Adv Nutr. 2017;8(6):905-915. 27. Einfeld SL, Smith E, McGregor IS, et al. A double-blind randomized
5. Miller JL, Lynn CH, Driscoll DC, et al. Nutritional phases in controlled trial of oxytocin nasal spray in Prader Willi syndrome.
Prader-Willi syndrome. Am J Med Genet A. 2011;155A(5): Am J Med Genet A. 2014:164A(9):2232-2239.
1040-1049. 28. Kuppens RJ, Donze SH, Hokken-Koelega AC. Promising effects of
6. Butler MG, Theodoro MF, Bittel DC, Donnelly JE. Energy expend­ oxytocin on social and food-related behaviour in young children
iture and physical activity in Prader-Willi syndrome: comparison with Prader-Willi syndrome: a randomized, double-blind, con­
with obese subjects. Am J Med Genet A. 2007;143A(5):449-459. trolled crossover trial. Clin Endocrinol (Oxf). 2016;85(6):979-987.
7. Schwartz L, Caixàs A, Dimitropoulos A, et al. Behavioral features 29. Miller JL, Tamura R, Butler MG, et al. Oxytocin treatment in children
in Prader-Willi syndrome (PWS): consensus paper from the inter­ with Prader-Willi syndrome: a double-blind, placebo-controlled,
national PWS clinical trial consortium. J Neurodev Disord. crossover study. Am J Med Genet A. 2017;173(5):1243-1250.
2021;13(1):25. 30. Tauber M, Boulanouar K, Diene G, et al. The use of oxytocin to im­
8. Glasson EJ, Buckley N, Chen W, et al. Systematic review and meta- prove feeding and social skills in infants with Prader-Willi syn­
analysis: mental health in children with neurogenetic disorders drome. Pediatrics. 2017;139(2):e20162976.
1708 The Journal of Clinical Endocrinology & Metabolism, 2023, Vol. 108, No. 7

31. Tauber M, Mantoulan C, Copet P, et al. Oxytocin may be useful to behavior in vasopressin V1a receptor knockout mice.
increase trust in others and decrease disruptive behaviours in pa­ Neuropsychopharmacology. 2004;29(3):483-493.
tients with Prader-Willi syndrome: a randomised placebo- 33. Kayadjanian N, Vrana-Diaz C, Bohonowych J, et al.
controlled trial in 24 patients. Orphanet J Rare Dis. 2011;6(1):47. Characteristics and relationship between hyperphagia, anxiety, be­
32. Bielsky IF, Hu SB, Szegda KL, Westphal H, Young LJ. Profound havioral challenges and caregiver burden in Prader-Willi syndrome.
impairment in social recognition and reduction in anxiety-like PLoS One. 2021;16(3):e0248739.

Downloaded from https://academic.oup.com/jcem/article/108/7/1696/6986298 by guest on 20 November 2023

You might also like