Skip to main content
medRxiv
  • Home
  • About
  • Submit
  • ALERTS / RSS
Advanced Search

A randomized double-blind placebo-controlled trial of Guanfacine Extended Release for aggression and self-injurious behavior associated with Prader-Willi Syndrome

Deepan Singh, Michael Silver, View ORCID ProfileTheresa Jacob
doi: https://doi.org/10.1101/2024.08.22.24312419
Deepan Singh
1Department of Psychiatry, Maimonides Medical Center, Brooklyn, New York
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: desingh{at}maimo.org
Michael Silver
2Research Administration, Maimonides Medical Center, Brooklyn, New York
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Theresa Jacob
1Department of Psychiatry, Maimonides Medical Center, Brooklyn, New York
3Clinical and Translational Research Labs, Maimonides Medical Center, Brooklyn, New York
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Theresa Jacob
  • Abstract
  • Full Text
  • Info/History
  • Metrics
  • Data/Code
  • Preview PDF
Loading

Abstract

Introduction Prader-Willi Syndrome (PWS), a rare genetic disorder, affects development and behavior, frequently resulting in self-injury, aggression, hyperphagia, oppositional behavior, impulsivity and over-activity causing significant morbidity. Currently, limited therapeutic options are available to manage these neuropsychiatric manifestations. The aim of this clinical trial was to assess the efficacy of guanfacine-extended release (GXR) in reducing aggression and self-injury in individuals with PWS.

Trial Design Randomized, double-blind, placebo-controlled trial conducted under IRB approval.

Methods Subjects with a diagnosis of PWS, 6-35 years of age, with moderate to severe aggressive and/or self-injurious behavior as determined by the Clinical Global Impression (CGI)-Severity scale, were included in an 8-week double-blind, placebo-controlled, fixed-flexible dose clinical trial of GXR, that was followed by an 8-week open-label extension phase. Validated behavioral instruments and physician assessments measured the efficacy of GXR treatment, its safety and tolerability.

Results GXR was effective in reducing aggression/agitation and hyperactivity/noncompliance as measured by the Aberrant Behavior Checklist (ABC) scales (p=0.03). Overall aberrant behavior scores significantly reduced in the GXR arm. Aggression as measured by the Modified Overt Aggression Scale (MOAS) also showed a significant reduction. Skin-picking lesions as measured by the Self Injury Trauma (SIT) scale decreased in response to GXR. No serious adverse events were experienced by any of the study participants. Fatigue /sedation was the only adverse event significantly associated with GXR. The GXR group demonstrated significant overall clinical improvement as measured by the CGI-Improvement (CGI-I) scale. (p<0.01).

Conclusion Findings of this pragmatic trial strongly support the use of GXR for treatment of aggression, skin picking, and hyperactivity in children, adolescents, and adults with PWS.

Trial Registration ClinicalTrials.gov Identifier: NCT05657860

Introduction

Prader-Willi syndrome (PWS) is a rare, complex multisystem genetic disorder, which includes hypothalamic dysfunction, cognitive and behavioral problems, increased anxiety, and compulsive behaviors. 1, 2 It is the most common genetic cause of morbid obesity in children. 2 While hyperphagia is seen as the sine qua non of PWS, other behavioral symptoms such as aggression, oppositional behavior, and temper tantrums, are common and cause significant distress to patients and caregivers. 3, 4, 5 PWS also has a high prevalence of self-injurious behavior such as skin-picking, as well as repetitive motor activity, impulsivity, and hyperactivity.6, 7, 8, 9 Despite the high prevalence of behavioral disturbances in PWS, evidence-based pharmacological treatment options for them remain limited. 10 In fact, at the time of this writing, there are no published results from any randomized controlled clinical trials evaluating the efficacy of any agent against the common behavioral disturbances seen in PWS except for hyperphagia. This is particularly concerning given that close to half of all children between ages 11-17 and over 65% of all adults with PWS are on at least one prescribed psychotropic medication with most of them requiring more than one medication.11

Currently, serotonin reuptake inhibitors (SRIs) and atypical antipsychotics are the most frequently utilized medications to manage behavioral problems in PWS. 11 Both these classes of medications are associated with the potential for developing or worsening metabolic syndrome. 12, 13, 14, 15 Due to the high risk of obesity in PWS, it is important to avoid weight gain-inducing agents as much as possible. Furthermore, there have been reports of the development of psychotic symptoms such as auditory/visual hallucinations and paranoid delusions with the use of SRIs and serotonin-agonists, making serotonergic agents potentially dangerous in this population. 16, 17 Impulsivity and hyperactivity are well-known presenting signs of attention-deficit/hyperactivity disorder (ADHD), and as many as 25% of the patients with PWS exhibit these symptoms.7 Stimulants for the management of impulsivity and hyperactivity may sometimes worsen mood, and irritability, and chronic use of stimulants may lead to an increase in skin-picking behavior. 18 Although N-acetyl cysteine and oxytocin have been suggested for the management of skin-picking behaviors, safe and effective modalities remain sparse for the management of behavioral symptoms in PWS. 19, 20, 21, 1

Guanfacine extended-release (GXR) is a postsynaptic α2-adrenoceptor agonist approved by the Food and Drug Administration (FDA) for the management of ADHD. 22, 23 Guanfacine is proposed to work by moderating the activity of the left dorsolateral prefrontal cortex associated with the emotional biasing of response execution, thus reducing impulsivity. 24 Medications that operate as α2-adrenoceptor agonists such as guanfacine are beneficial in treating disruptive and aggressive behaviors. 25 In addition, GXR has been demonstrated to be safe and effective for reducing hyperactivity, impulsivity, and distractibility in patients with autism. 26, 27 Most importantly, GXR has had no reports of significant weight gain, worsening hyperphagia, or other metabolic side effects. 28, 29 A retrospective cohort study has demonstrated that GXR led to improvement in symptoms of skin picking, aggression/agitation, and ADHD in patients with PWS.30 Given the significant morbidity caused by these behavioral problems in this already vulnerable population, novel evidence-based approaches are warranted for the management of aggression, impulsivity, and self-injury in PWS. 31 It was postulated that GXR is a safer treatment option for self-injurious and aggressive behaviors in PWS and we wanted to test this with a pragmatic approach.

The objective of this randomized, double-blind, placebo-controlled trial was to determine the efficacy of guanfacine extended release (GXR) in reducing aggression and self-injury in individuals with PWS, and to assess its safety and tolerability.

Methods

Study Design

This pragmatic two-phase 16-week study was designed to test the hypothesis that GXR is effective in reducing aggression and self-injury in individuals with PWS with moderate to severe aggressive and/or self-injurious behavior. In the first phase, the randomized controlled trial part, eligible subjects were randomly assigned in a 1:1 ratio, without stratification, to GXR or placebo for 8 weeks. This was the double-blind, placebo-controlled, fixed-flexible dose phase of the clinical trial. Immediately following the blinded randomized trial period, an 8-week open-label extension was pursued to further define the efficacy and tolerability of GXR, as well as to establish its safety with a specific focus on metabolic profile.

The study was performed in accordance with all current applicable regulations, the International Conference on Harmonization of Good Clinical Practice, the principles of the Declaration of Helsinki and local ethical and legal requirements. It was approved by the Institutional Review Board (# 2020-11-03-MMC) and registered at ClinicalTrials.gov (identifier: NCT05657860). It was conducted in a mental health clinic setting of a large, urban, community-based teaching hospital in a major metropolis of the United States. The study recruitment period began March 1, 2021 and ended October 31, 2023. Study participants received complete information regarding the protocol during the consent process before enrollment. Written, IRB-approved informed consent was obtained from each participant’s parent or legal guardian, and assent was obtained from each participant, as applicable.

Inclusion/Exclusion Criteria

To be eligible for inclusion, subjects had to be between 6 and 35 years of age with a diagnosis of PWS confirmed by genetic testing. In addition, a rating of moderate or above on the Clinical Global Impression - Severity scale (CGI-S) was required for inclusion. Females of childbearing potential had to have a negative urine pregnancy test at screening and baseline and to comply with any protocol contraceptive requirements. In addition, participants and their parent/legal guardian had to be willing, able, and likely to fully comply with the study procedures and restrictions defined in the protocol. Exclusionary criteria included a positive pregnancy test, swallowing difficulty, or the presence of clinically significant bradycardia or hypotension. Those currently on GXR, actively enrolled in other studies, or with a lactose intolerance or allergy were also excluded. Subjects receiving antipsychotic medications due to a documented history of psychosis or bipolar disorder were allowed to continue taking the medication without dosage modification. The pragmatic approach of this trial also allowed growth hormone, thyroid hormone replacement treatment, and non-psychiatric medicines, to continue albeit without dosage modifications. Similarly, N-Acetyl Cysteine (NAC) and anticonvulsant medications if prescribed for seizures were allowed, with specific instructions to refrain from making any dosage changes during the trial period.

Study Drug Administration

The starting dose for all subjects was 1 mg per day. Subjects weighing less than 25 kg remained on the 1 mg dose until day 14. If the medication was well tolerated, the dose was raised to 2 mg until day 28 and increased to 3mg for the remaining 4 weeks in the trial. Subjects weighing 25 kg or more were eligible for an increase to 2 mg at day 7, 3 mg at day 14, and 4 mg at day 21 or day 28. The dose schedule was not fixed; the treating clinician could delay a planned increase or lower the dose to manage adverse effects. Medication was dispensed at the baseline visit and at the unblinding visit for the subsequent dosing period. GXR was administered as 1 mg capsules; placebo group received matching placebo capsules.

Randomization and Blinding

Subjects were randomly assigned to either the placebo or experimental (GXR) group in a 1:1 ratio via a computer-generated randomization scheme with randomly permuted blocks of varying sizes, done by Pharmacy. As the first phase of this study was double-blinded, the study pharmacist was the only team member aware of patient treatment assignments. Randomization occurred at baseline (visit 1). Unblinding was at week 8, and subjects continued treatment for 8 more weeks (open-label phase).

Assessments

Baseline and follow-up assessments included the Aberrant Behavior Checklist, (ABC), Self-Injury Trauma (SIT) scale, physician-administered Clinical Global Impression – Severity (CGI-S; only at baseline), and CGI-Improvement (CGI-I) scales, Hyperphagia Questionnaire for use in PWS clinical trials (HQ-CT), Modified Overt Aggression Scale (MOAS), and the Epworth Sleepiness Scale (ESS) at biweekly study visits. The ABC includes an overall score and sub-scores to monitor irritability, lethargy, inappropriate speech, hyperactivity and stereotypies.

In addition, laboratory tests, medical and psychiatric histories, physical examination, electrocardiography (ECG), and vital signs evaluation were conducted at study visits. Daily monitoring of vitals and problematic behavior was performed by caregivers.

Safety Monitoring

Blood pressure and pulse rate were measured at each visit. Laboratory tests were conducted at baseline and were repeated at week 8 and week 16. An ECG was obtained at baseline and at week 8 to assess the impact of GXR on cardiac conduction. Adverse events were systematically reviewed and documented. Side effects were measured using open-ended clinician inquiry with questions about drowsiness, fatigue, decreased appetite, emotional/tearful, dry mouth, irritability, anxiety, headache, increased energy, mid-sleep wakening, stomachache, constipation, increased repetitive behavior, aggression, depressed mood, cough/congestion, self-injury, nausea, trouble falling asleep, dizziness, silly, weakness, diarrhea, vomiting, increased appetite, excessive talking, blurred vision, skin rash/eczema, nightmares, enuresis, motor tics, skin-picking, and pyrexia.

Subjects who were randomly assigned to placebo group and did not show a positive response by 8 weeks, were offered open-label treatment with GXR for 8 weeks. All subjects in this continuation phase were seen every 2 weeks for monitoring. Those unblinded and started on GXR were monitored weekly for the first 4 weeks of the continuation phase.

Data Analysis

Data were analyzed using an intent-to-treat approach. Patient baseline data were summarized via descriptive statistics such that continuous variables were summarized by means (SD) or medians (IQR) and categorical data by counts (%). Baseline factors (CGI-S, age, gender) as well as all study observations were presented within each group. In the analysis, distributions were compared between treatment groups at baseline via the Wilcoxon rank sum test. Finally, we created a repeated measures linear regression model for each outcome scale, using Generalized Estimating Equations (GEE) to account for within-subject variability, with the predictor of treatment, to assess the impact of treatment across the entire timeline of the study. An unstructured correlation matrix was assumed. All models were then plotted in a Forest Plot for ease of comparison. Two-sided p-values < 0.05 were considered statistically significant. All analyses were conducted using SAS/STAT version 9.4 (SAS Institute Inc., Cary N.C. USA).

Results

Demographics

Over the study period, 65 patients with a clinical diagnosis of PWS were screened. Of these, 14 underwent randomization. Participant disposition in the study is shown in the CONSORT flow diagram (Fig 1). The study population included 11 males and 3 females, aged between 6 to 35 years, carrying genetically confirmed diagnoses of PWS. Of these subjects, 78.5% were Caucasian, and 7% were Asian. Table 1 reviews the demographics of the participants. Subtype distribution showed that 29% of the subjects carried paternal deletion and 43% had maternal uniparental disomy. Further, the submitted test reports did not distinguish subtype in 28% of the participants.

Fig 1.
  • Download figure
  • Open in new tab
Fig 1. CONSORT flow diagram.

Participant disposition in double-blind, randomized placebo-controlled trial for GXR (guanfacine-extended release)

View this table:
  • View inline
  • View popup
  • Download powerpoint
Table 1. Baseline Demographics and Clinical Characteristics of Study Participants.

Patient Baseline Characteristics

As seen in Table 2, there were no differences among the groups at baseline for any measures, including severity of illness as measured by the CGI-S.

View this table:
  • View inline
  • View popup
  • Download powerpoint
Table 2. Subject Behavioral Measure Scores at Index Visit.

Response to GXR

In repeated measures linear regression models, using GEE and an unstructured correlation matrix, there was a decrease in the Aberrant Behavior Checklist score of 9.962 points (95% CI: 19 - 0.8), p=0.03, in subjects receiving GXR treatment as seen in Table 3. A decrease in ABC subscale IV (Hyperactivity) of 5.27 points (95% CI: 1.23 – 9.30), p=0.01, was also observed in this group. While on GXR, subjects demonstrated a reduction in aggressive behaviors as reflected in the decrease in MOAS score of 3.65 points (95% CI: 0.50 – 6.79), p=0.02. The number of skin-picking lesions also decreased in response to GXR, as indicated by the SIT scale scores in the treatment arm; however, this decrease was not significant in comparison to that observed in the placebo arm. There was a highly significant improvement in the overall behavioral symptoms of participants on GXR, as reflected by the lower scores in the psychiatrist-rated CGI-I scales [1.25 points, (95% CI: 0.47 – 2.04), p=0.002]. There were no other statistical differences observed in outcome measures. Fig 2 demonstrates a schematic forest plot of the different effect sizes across measures.

Fig 2.
  • Download figure
  • Open in new tab
Fig 2. Forest plot.

A schematic of the different effect sizes across measures in the two study groups based on GEE Models (Generalized Estimating Equations)

View this table:
  • View inline
  • View popup
  • Download powerpoint
Table 3. GEE Models for Additional Outcomes and Scales.

Side Effects and Tolerability

There were no serious adverse events (SAEs) reported by subjects in either of the study groups. However, subjects on GXR were more likely to experience fatigue 29% vs 93%, p = 0.012. Table 4 shows all the treatment-emergent adverse events observed among trial subjects in GXR and placebo groups. There were no other differences in the rate of adverse events amongst the groups.

View this table:
  • View inline
  • View popup
  • Download powerpoint
Table 4. Incidence of Adverse Events

There were no GXR-related clinically meaningful changes in ECG and QTc-related parameters. No significant changes in vital signs, or lab values were observed between the groups.

The average stable dose of GXR at time of completion of the study was 2.75 mg daily.

Discussion

Behavioral and psychiatric symptoms are common in the PWS patient population. Individuals with PWS may exhibit impulsive behavior, which can impede daily functioning and lead to risky behavior. 6, 7, 32 Furthermore, patients can exhibit aggression and temper tantrums, which leads to significant caregiver burden. 5, 33 Skin-picking is another behavioral symptom commonly seen in this patient population, which may get complicated with infections and scarring at the site of damage. 19 The overall burden of illness over the lifetime of an individual with PWS is compounded by these behavioral problems.

Currently, the commonly used pharmacological treatment options for behavioral management in individuals with PWS are SRIs, stimulants, and atypical antipsychotics. Their use in PWS is widespread despite the lack of empirical placebo-controlled studies to demonstrate efficacy. These medications are associated with significant risks including weight gain for antipsychotics, psychosis for SRIs, and increased skin excoriation with stimulants. With the exception of NAC, options are limited for the treatment of skin-picking behavior. 19, 20, 21, 1, 34 Therefore, investigating relatively safe medication options that can effectively manage several impairing behavioral symptoms seen in PWS is warranted to positively impact the lives of patients and to reduce caregiver fatigue.

This randomized double-blind, placebo-controlled, fixed-flexible dose clinical trial with open-label continuation, was designed to explore the efficacy, safety, and tolerability of guanfacine extended release in the management of aggression and self-injurious behavior (skin picking) in individuals with PWS. The results of this study demonstrate that GXR ameliorates overall behavioral disturbances as measured by the ABC scale. GXR was particularly effective against hyperactivity in PWS. This finding is expected since GXR has established evidence for the treatment of ADHD in the general population. 35, 36 Additionally, GXR led to a significant reduction in aggressive behavior as measured by the MOAS. Our study also showed a positive response to GXR in terms of skin picking, with a reduction in the number of lesions in the treatment group, although it was not statistically significant as compared to that in the placebo group. Notably, subjects on GXR showed a significant improvement in their overall presentation as measured by the CGI-I scale.

GXR was generally well tolerated with the most commonly reported side effect being fatigue, commensurate with the findings in the general population. 37 It is important to note that despite this, the overall level of daytime sleepiness as measured by the ESS did not show any statistical difference from placebo. This finding is of particular salience given the high rates of daytime sleepiness experienced in this population. 38 There were no serious adverse events reported in the GXR group. Another key factor to consider in this study is that, none of the subjects exposed to GXR experienced weight gain. There was no difference noted between the GXR and placebo groups with respect to ECG changes, blood pressure, or heart rate.

Several outcomes trended towards a positive response but failed to reach a statistical significance level. In particular, a positive signal was observed for hyperphagia and skin picking. These need to be confirmed in future studies with a larger sample size.

Individuals with PWS suffer from growth hormone (GH) deficiency, which contributes to the short stature and the central obesity. GH replacement therapy has shown beneficial effects in aiding linear growth, and restoring normal body composition, bone density, as well as lipid profile values in individuals with PWS. 39,40 At the time of this writing, GH is the only medication that has an FDA approved indication for the management of PWS. 41 Clonidine is an α2-adrenoceptor agonist similar to GXR, and both medications have been shown to increase GH levels in a dose-dependent manner. However, GXR is less sedating compared to clonidine and may be a better alternative in this setting. 42 Although not measured in the current study, the possible physical benefits of increased GH could indicate an additional advantage of GXR in comparison to other medications used for the behavioral management of individuals with PWS. This could be further elucidated in prospective trials.

Limitations

The multiple limitations of this study need to be highlighted. Although there were no gender differences between the active and placebo arms, the majority of patients enrolled were male. A larger study could reduce the possibility of there being a gender based moderating effect on the results. A larger sample size would also allow stratification of the results by PWS genotype. Similarly, the participants were predominantly Caucasian hence limiting generalizability of findings. The study was conducted at a single site, limiting the diversity in participants that can be achieved from a multicenter study. It is challenging to establish external validity in a single-site study, due to limitations of accounting for the variance in environmental and practitioner-based factors. Another factor that must be considered is the relatively high prevalence of ADHD and autism in PWS.6 Therefore, there is a possibility that the observed effects may be moderated by these co-existing conditions.

Despite these challenges to the study, the results of this clinical trial, along with prior knowledge of the side effects posed by the alternatives available to practitioners, suggests that GXR should be the first line treatment for behavioral problems, specifically aggression, irritability, and hyperactivity in individuals with PWS.

Conclusions

This randomized, fixed-flexible dose placebo-controlled clinical trial, with open label extension, aimed to provide a supporting framework for the utilization of GXR in the management of behavioral symptoms comorbid with PWS. The assessed symptoms included aggression, irritability, inattention/hyperactivity, and skin-picking. GXR treatment leads to global improvement in symptoms, particularly those related to behavioral disturbance in persons with PWS. Specifically, GXR reduces aggression and hyperactivity in this population. Additionally, there are possible beneficial effects on skin picking behavior. The most commonly reported side effect was fatigue, but overall daytime sleepiness was not worse than placebo. There were no serious adverse effects reported. GXR did not lead to any change in weight, heart rate, blood pressure, or cardiac rhythm, as compared to placebo. The results of this clinical trial suggests that GXR is an effective and relatively safe treatment modality for behavioral problems in patients with PWS.

Clinical Significance

Individuals with PWS often present with challenging behavioral disturbances such as aggression/agitation, skin-picking, and ADHD symptoms like impulsivity and inattention. Current management strategies for behavioral disturbances in PWS patients show limited efficacy and can have significant side effects. Although other studies have been conducted to assess treatment options against hyperphagia, this is the first randomized placebo-controlled trial examining the efficacy of a psychotropic medication against the behavioral manifestations of PWS. This randomized placebo-controlled clinical trial demonstrated that GXR is effective in the management of behavioral disturbances in PWS while being well tolerated. The authors recommend that GXR be considered as a first line treatment strategy for the management of behavioral problems, especially aggression and hyperactivity, associated with PWS.

Data Availability

All relevant data are within the manuscript and its Supporting Information files and will be available upon its publication.

Supporting Information

S1 Checklist. CONSORT Checklist

Acknowledgments

The authors would like to thank our hospital pharmacists Jason Brady and Rina Evans for their invaluable contribution. We would also like to thank our clinical research coordinators Otuwe Anya and Hasan Mustafic, without whom this study would not have been possible.

References

  1. ↵
    Miller JL, Tamura R, Butler MG, Kimonis V, Sulsona C, Gold JA et al. Oxytocin treatment in children with Prader-Willi syndrome: A double-blind, placebo-controlled, crossover study. Am J Med Genet A. 2017;173A:1243–50.
    OpenUrl
  2. ↵
    Butler MG. Prader-Willi Syndrome: Obesity due to Genomic Imprinting. Curr Genomics. 2011;12:204–15.
    OpenUrlCrossRefPubMedWeb of Science
  3. ↵
    Ogura K, Shinohara M, Kousaku O, Mori E. Frontal behavioral syndromes in Prader-Willi syndrome. Brain Dev. 2008;30:469–76.
    OpenUrlCrossRefPubMed
  4. ↵
    Arron K, Oliver C, Moss J, Berg K, Burbidge C. The prevalence and phenomenology of self-injurious and aggressive behaviour in genetic syndromes. J Intellect Disabil Res. 2011;55:109–20.
    OpenUrlCrossRefPubMed
  5. ↵
    Tunnicliffe P, Woodcock K, Bull L, Oliver C, Penhallow J. Temper outbursts in Prader-Willi syndrome: causes, behavioral and emotional sequence and responses by carers. J Intellect Disabil Res. 2014;58:134–50.
    OpenUrl
  6. ↵
    Ogata H, Ihara H, Murakami N, Gito M, Kido Y, Nagai T. Autism spectrum disorders and hyperactive/impulsive behaviors in Japanese patients with Prader-Willi syndrome: a comparison between maternal uniparental disomy and deletion cases. Am J Med Genet A. 2014;164A:2180–6.
    OpenUrlCrossRef
  7. ↵
    Wigren M, Hansen S. ADHD symptoms and insistence on sameness in Prader-Willi syndrome. J Intellect Disabil Res. 2005;49:449–56.
    OpenUrlCrossRefPubMed
  8. ↵
    Greaves N, Prince E, Evans DW, Charman T. Repetitive and ritualistic behavior in children with Prader-Willi syndrome and children with autisum. J Intellect Disabil Res. 2006;50:92–100.
    OpenUrlCrossRefPubMedWeb of Science
  9. ↵
    Banga A, Connor DF. Effectiveness of naltrexone for treating pathologic skin picking behavior in an adolescent with Prader-Willi syndrome. J Child Adolesc Psychopharmacol. 2012;22:396–8.
    OpenUrlPubMed
  10. ↵
    Bonnot O, Cohen D, Thuilleaux D, Consoli A, Cabal S, Tauber M. Psychotropic treatments in Prader Willi syndrome: a critical review of published literature. Eur J Pediatr. 2016;175:9-18.
    OpenUrl
  11. ↵
    Matesevac L, PATH for PWS Study 2021 Update: A Non-Interventional, Observational, Natural History Study of Serious Medical Events in Prader-Willi Syndrome. Presented in the PWSA-USA Medical & Scientific Conference 2021, Virtual. https://www.pwsausa.org/2021-national-convention-session-recordings/
  12. ↵
    Kohn Y, Weizman A, Apter A. Aggravation of food-related behavior in an adolescent with Prader-Willi syndrome treated with fluvoxamine and fluoxetine. Int J Eat Disord. 2001;30:113–7.
    OpenUrlCrossRefPubMedWeb of Science
  13. ↵
    Kraemer S, Minarzyk A, Forst T, Kopf D, Hundemer HP. Prevalence of metabolic syndrome in patients with schizophrenia, and metabolic changes after 3 months of treatment with antipsychotics—results from a German observational study. BMC Psychiatry. 2011;11:173.
    OpenUrlCrossRefPubMed
  14. ↵
    Hellings JA. Self-injurious behavior and serotonin in Prader-Willi syndrome. Psychopharmacol Bull. 1994;20:245–50.
    OpenUrl
  15. ↵
    Warnock JK, Kestenbaum T. Pharmacologic treatment of severe skin-picking behaviors in Prader-Willi syndrome. Two case reports. Arch Dermatol. 1992;128:1623–5.
    OpenUrlCrossRefPubMedWeb of Science
  16. ↵
    Herguner S, Mukaddes NM. Psychosis associated with fluoxetine in Prader-Willi syndrome. J Am Acad Child Adolesc Psychiatry. 2007;46:944–45.
    OpenUrlPubMed
  17. ↵
    Dennison EA, Raidoo BM, Cruz R, Raidoo D. A case of high-dose venlafaxine-related psychosis. J Clin Psychopharmacol. 2013;33:134–6.
    OpenUrl
  18. ↵
    Kara T, Akaltun I. Newly Developed Skin Picking After Methylphenidate Treatment in Attention Deficit Hyperactivity Disorder: Possible Mechanisms. Clin Neuropharmacol. 2018;41:28–30.
    OpenUrl
  19. ↵
    Miller JL, Angulo M. An open-label pilot study of N-acetylcysteine for skin-picking in Prader-Willi syndrome. Am J Med Genet A. 2014;164A:421–4.
    OpenUrl
  20. ↵
    Tauber M, Mantoulan C, Copet P, Jauregui J, Demeer G, Diene G, et al. Oxytocin may be useful to increase trust in others and decrease disruptive behaviours in patients with Prader-Willi syndrome: a randomised placebo-controlled trial in 24 patients. Orphanet J Rare Dis. 2011;6:47.
    OpenUrlPubMed
  21. ↵
    Kuppens RJ, Donze SH, Hokken-Koelega AC. Promising effects of oxytocin on social and food-related behaviour in young children with Prader-Willi syndrome: a randomized, double-blind, controlled crossover trial. Clin Endocrinol (Oxf). 2016;85:979–87.
    OpenUrlPubMed
  22. ↵
    Website: “Drugs@FDA: FDA Approved Drug Products.” Accessdata.fda.gov, www.accessdata.fda.gov/scripts/cder/daf/index.cfm?event=overview.process&applno=200881. (Access date: July 09, 2018)
  23. ↵
    Cruz MP. Guanfacine Extended-Release Tablets (Intuniv), a Nonstimulant Selective Alpha(2A)-Adrenergic Receptor Agonist For Attention-Deficit/Hyperactivity Disorder. P T. 2010;35:448–51.
    OpenUrlPubMed
  24. ↵
    Schulz KP, Clerkin SM, Fan J, Halperin JM, Newcorn JH. Guanfacine modulates the influence of emotional cues on prefrontal cortex activation for cognitive control. Psychopharmacology (Berl). 2013;226:261–71.
    OpenUrl
  25. ↵
    Pringsheim T, Hirsch L, Gardner D, Gorman DA. The pharmacological management of oppositional behaviour, conduct problems, and aggression in children and adolescents with attention-deficit hyperactivity disorder, oppositional defiant disorder, and conduct disorder: a systematic review and meta-analysis. Part 1: psychostimulants, alpha-2 agonists, and atomoxetine. Can J Psychiatry. 2015;60:42-51.
    OpenUrlCrossRefPubMed
  26. ↵
    Scahill L, McCracken JT, King BH, Rockhill C, Shah B, Politte L et al. Research Units on Pediatric Psychopharmacology Autism Network. Extended-Release Guanfacine for Hyperactivity in Children With Autism Spectrum Disorder. Am J Psychiatry. 2015;172:1197–206.
    OpenUrlCrossRefPubMed
  27. ↵
    Davis NO, Kollins SH. Treatment for co-occurring attention deficit/hyperactivity disorder and autism spectrum disorder. Neurotherapeutics. 2012;9:518–30.
    OpenUrlCrossRefPubMed
  28. ↵
    Hervas A, Huss M, Johnson M, McNicholas F, van Stralen J, Sreckovic S et al. Efficacy and safety of extended-release guanfacine hydrochloride in children and adolescents with attention-deficit/hyperactivity disorder: a randomized, controlled, phase III trial. Eur Neuropsychopharmacol. 2014;24:1861–72.
    OpenUrlPubMed
  29. ↵
    Singh D, Kadakia N, Pinkhasov A. Significant reduction of aggression with guanfacine extended release in an adolescent with Prader-Willi syndrome. J Child Adolesc Psychopharmacol. 2015;25:376–7.
    OpenUrl
  30. ↵
    Singh D, Wakimoto Y, Filangieri C, Pinkhasov A, Angulo M. Guanfacine Extended Release for the Reduction of Aggression, Attention-Deficit/Hyperactivity Disorder Symptoms, and Self-Injurious Behavior in Prader-Willi Syndrome-A Retrospective Cohort Study. J Child Adolesc Psychopharmacol. 2019;29:313–7.
    OpenUrl
  31. ↵
    Soni S, Whittington J, Holland AJ, Webb T, Maina E, Boer H, et al. The course and outcome of psychiatric illness in people with Prader-Willi syndrome: Implications for management and treatment. J Intellect Disabil Res. 2007;51:32–42.
    OpenUrlCrossRefPubMedWeb of Science
  32. ↵
    Dalsgaard S, Ostergaard SD, Leckman JF, Mortensen PB, Pedersen MG. Mortality in children, adolescents, and adults with attention deficit hyperactivity disorder: a nationwide cohort study. Lancet. 2015;385:2190–6.
    OpenUrlCrossRefPubMed
  33. ↵
    Mazaheri MM, Rae-Seebach RD, Preston HE, Schmidt M, Kountz-Edwards S, Field N et al. The impact of Prader-Willi syndrome on the family’s quality of life and caregiving, and the unaffected siblings’ psychosocial adjustment. J Intellect Disabil Res. 2013;57:861–73.
    OpenUrlCrossRefPubMed
  34. ↵
    Logan SL, Carpenter L, Leslie RS, Garrett-Mayer E, Hunt KJ, Charles J et al. Aberrant Behaviors and Co-occurring Conditions as Predictors of Psychotropic Polypharmacy among Children with Autism Spectrum Disorders. J Child Adolesc Psychopharmacol. 2015;25:323– 36.
    OpenUrl
  35. ↵
    Newcorn JH, Harpin V, Huss M, Lyne A, Sikirica V, Johnson M et al. Extended-release guanfacine hydrochloride in 6-17-year olds with ADHD: a randomised-withdrawal maintenance of efficacy study. J Child Psychol Psychiatry. 2016;57:717–28.
    OpenUrl
  36. ↵
    Butterfield ME, Saal J, Young B, Young JL. Supplementary guanfacine hydrochloride as a treatment of attention deficit hyperactivity disorder in adults: A double blind, placebo-controlled study. Psychiatry Res. 2016;236:136–41.
    OpenUrl
  37. ↵
    Wilens TE, Robertson B, Sikirica V, Harper L, Young JL, Bloomfield R, et al. A Randomized, Placebo-Controlled Trial of Guanfacine Extended Release in Adolescents With Attention-Deficit/Hyperactivity Disorder. J Am Acad Child Adolesc Psychiatry. 2015;54:916–25.e2.
    OpenUrlPubMed
  38. ↵
    Duis J, Pullen LC, Picone M, Friedman N, Hawkins S, Sannar E et al. Diagnosis and management of sleep disorders in Prader-Willi syndrome. J Clin Sleep Med. 2022;18:1687–96.
    OpenUrl
  39. ↵
    Bakker NE, Lindberg A, Heissler J, Wollmann HA, Camacho-Hubner C, Hokken-Koelega AC. Growth Hormone Treatment in Children With Prader-Willi Syndrome: Three Years of Longitudinal Data in Prepubertal Children and Adult Height Data From the KIGS Database. J Clin Endocrinol Metab. 2017;102:1702–11.
    OpenUrlPubMed
  40. ↵
    Sipila I, Sintonen H, Hietanen H, Apajasalo M, Alanne S, Viita A-M, et al. Long-term effects of growth hormone therapy on patients with Prader-Willi syndrome. Acta Paediatr. 2010;99:1712–8.
    OpenUrlCrossRefPubMed
  41. ↵
    Singh D, Miller JL, Wassman ER, Butler MG, Foley Shenk A, Converse M et al. The Arduous Path to Drug Approval for the Management of Prader-Willi Syndrome: A Historical Perspective and Call to Action. Int J Mol Sci. 2023;24:11574.
    OpenUrl
  42. ↵
    Balldin J, Berggren U, Eriksson E, Lindstedt G, Sundkler A. Guanfacine as an alpha-2-agonist inducer of growth hormone secretion--a comparison with clonidine. Psychoneuroendocrinology. 1993;18:45–55.
    OpenUrlCrossRefPubMedWeb of Science
Back to top
PreviousNext
Posted August 22, 2024.
Download PDF
Data/Code
Email

Thank you for your interest in spreading the word about medRxiv.

NOTE: Your email address is requested solely to identify you as the sender of this article.

Enter multiple addresses on separate lines or separate them with commas.
A randomized double-blind placebo-controlled trial of Guanfacine Extended Release for aggression and self-injurious behavior associated with Prader-Willi Syndrome
(Your Name) has forwarded a page to you from medRxiv
(Your Name) thought you would like to see this page from the medRxiv website.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Share
A randomized double-blind placebo-controlled trial of Guanfacine Extended Release for aggression and self-injurious behavior associated with Prader-Willi Syndrome
Deepan Singh, Michael Silver, Theresa Jacob
medRxiv 2024.08.22.24312419; doi: https://doi.org/10.1101/2024.08.22.24312419
Twitter logo Facebook logo LinkedIn logo Mendeley logo
Citation Tools
A randomized double-blind placebo-controlled trial of Guanfacine Extended Release for aggression and self-injurious behavior associated with Prader-Willi Syndrome
Deepan Singh, Michael Silver, Theresa Jacob
medRxiv 2024.08.22.24312419; doi: https://doi.org/10.1101/2024.08.22.24312419

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Subject Area

  • Psychiatry and Clinical Psychology
Subject Areas
All Articles
  • Addiction Medicine (349)
  • Allergy and Immunology (668)
  • Allergy and Immunology (668)
  • Anesthesia (181)
  • Cardiovascular Medicine (2648)
  • Dentistry and Oral Medicine (316)
  • Dermatology (223)
  • Emergency Medicine (399)
  • Endocrinology (including Diabetes Mellitus and Metabolic Disease) (942)
  • Epidemiology (12228)
  • Forensic Medicine (10)
  • Gastroenterology (759)
  • Genetic and Genomic Medicine (4103)
  • Geriatric Medicine (387)
  • Health Economics (680)
  • Health Informatics (2657)
  • Health Policy (1005)
  • Health Systems and Quality Improvement (985)
  • Hematology (363)
  • HIV/AIDS (851)
  • Infectious Diseases (except HIV/AIDS) (13695)
  • Intensive Care and Critical Care Medicine (797)
  • Medical Education (399)
  • Medical Ethics (109)
  • Nephrology (436)
  • Neurology (3882)
  • Nursing (209)
  • Nutrition (577)
  • Obstetrics and Gynecology (739)
  • Occupational and Environmental Health (695)
  • Oncology (2030)
  • Ophthalmology (585)
  • Orthopedics (240)
  • Otolaryngology (306)
  • Pain Medicine (250)
  • Palliative Medicine (75)
  • Pathology (473)
  • Pediatrics (1115)
  • Pharmacology and Therapeutics (466)
  • Primary Care Research (452)
  • Psychiatry and Clinical Psychology (3432)
  • Public and Global Health (6527)
  • Radiology and Imaging (1403)
  • Rehabilitation Medicine and Physical Therapy (814)
  • Respiratory Medicine (871)
  • Rheumatology (409)
  • Sexual and Reproductive Health (410)
  • Sports Medicine (342)
  • Surgery (448)
  • Toxicology (53)
  • Transplantation (185)
  • Urology (165)