You are on page 1of 6

Research

JAMA Neurology | Original Investigation

Safety of Converting From Tetrabenazine


to Deutetrabenazine for the Treatment of Chorea
Samuel Frank, MD; David Stamler, MD; Elise Kayson, MS, RNC, ANP; Daniel O. Claassen, MD; Amy Colcher, MD;
Charles Davis, PhD; Andrew Duker, MD; Shirley Eberly, MS; Lawrence Elmer, MD, PhD; Erin Furr-Stimming, MD;
Mark Gudesblatt, MD; Christine Hunter, RN; Joseph Jankovic, MD; Sandra K. Kostyk, MD, PhD; Rajeev Kumar, MD;
Clement Loy, MBBS, FRACP, PhD; William Mallonee, MD; David Oakes, PhD; Burton L. Scott, MD, PhD;
Victor Sung, MD; Jody Goldstein, BS; Christina Vaughan, MD; Claudia M. Testa, MD, PhD; for the Huntington Study
Group/Alternatives for Reducing Chorea in Huntington Disease Investigators

Supplemental content
IMPORTANCE Tetrabenazine is efficacious for chorea control; however, tolerability concerns
exist. Deutetrabenazine, a novel molecule that reduces chorea, was well tolerated in a
double-blind, placebo-controlled study.

OBJECTIVES To evaluate the safety and explore the efficacy of conversion from tetrabenazine
to deutetrabenazine in patients with chorea associated with Huntington disease (HD).

DESIGN, SETTING, AND PARTICIPANTS In this ongoing, open-label, single-arm study that
started on December 21, 2013, 37 patients at 13 Huntington Study Group sites in the United
States and Australia who were taking stable doses of tetrabenazine that provided a
therapeutic benefit were switched overnight to deutetrabenazine therapy. After week 1, the
deutetrabenazine dose was titrated on a weekly basis for optimal chorea control.

INTERVENTIONS Deutetrabenazine administration at a dosage thought to provide


comparable systemic exposure to the active metabolites of the prior, stable tetrabenazine
regimen.

MAIN OUTCOMES AND MEASURES Safety measures included adverse events (AEs), clinical
laboratory tests, vital signs, electrocardiograms, and validated scales. Changes in the Unified
Huntington’s Disease Rating Scale total maximal chorea score and total motor score were
efficacy end points.

RESULTS Of the 53 patients with HD screened for the study, 37 ambulatory patients with
manifest HD (mean [SD] age, 52.4 [11.5] years; 22 [59%] male and 15 [41%] female; 36 white
[97.3%]) were enrolled. Deutetrabenazine was generally well tolerated, with low rates of
neuropsychiatric AEs. Safety scales did not reveal subclinical toxicity with deutetrabenazine
treatment. Rates of dose reduction or suspension attributable to AEs were also low. Chorea
control, as measured by the total maximal chorea score, was maintained at week 1 and
significantly improved at week 8 (mean [SD] change from baseline, 2.1 [3.2]; P < .001).

CONCLUSIONS AND RELEVANCE In patients with chorea, overnight conversion to


deutetrabenazine therapy provided a favorable safety profile and effectively maintained
chorea control.

Author Affiliations: Author


affiliations are listed at the end of this
article.
Group Information: The Huntington
Study Group/Alternatives for
Reducing Chorea in Huntington
Disease Investigators are listed at the
end of this article.
Corresponding Author: Samuel
Frank, MD, Harvard Medical School,
330 Brookline Ave, Kirstein Bldg,
JAMA Neurol. doi:10.1001/jamaneurol.2017.1352 Room 228, Boston, MA 02215
Published online July 10, 2017. (sfrank2@bidmc.harvard.edu).

(Reprinted) E1
© 2017 American Medical Association. All rights reserved.

Downloaded From: http://archneur.jamanetwork.com/ by a Columbia University User on 07/11/2017


Research Original Investigation Tetrabenazine to Deutetrabenazine Conversion for Chorea Treatment

H
untington disease (HD) is a hereditary, progressive,
neurodegenerative disorder characterized by chorea Key Points
and other motor symptoms, cognitive dysfunction,
Question Is overnight treatment conversion from tetrabenazine
and psychiatric symptoms. Up to 90% of patients with HD have to deutetrabenazine safe in patients with Huntington disease?
chorea that can interfere with daily function and may cause
Findings In this open-label, single-arm study, 37 patients with
injury.1,2 Tetrabenazine was the first drug approved by the US
chorea associated with Huntington disease switched overnight
Food and Drug Administration for chorea associated with HD.3
from a stable, therapeutic tetrabenazine regimen to
High peak plasma concentrations and large plasma fluctua- deutetrabenazine. Deutetrabenazine was generally safe and well
tions may contribute to the tolerability issues observed in some tolerated, with chorea scores maintained at the end of week 1 of
patients with tetrabenazine use.4 Deutetrabenazine, a novel deutetrabenazine therapy and improved at week 8.
molecule that contains 6 deuterium atoms in place of 6 hy-
Meaning Chorea control may be safely maintained after an
drogen atoms in specific positions in the tetrabenazine mol- overnight switch from tetrabenazine to deutetrabenzine.
ecule, was approved for treating chorea associated with HD in
2017.5 Clinical evaluation in the First-HD trial found that deu-
tetrabenazine provides significant chorea and motor improve- tures and an expanded CAG repeat sequence (≥37) and were
ment while maintaining a favorable safety profile when com- receiving a stable tetrabenazine regimen for 8 weeks or lon-
pared with placebo.6 The efficacy and excellent safety profile ger that provided a therapeutic benefit for chorea control. Pa-
were attributed to the unique pharmacokinetic profile of deu- tients had a total functional capacity score of 5 or higher at
tetrabenazine, which enables comparable systemic exposure screening. The total functional capacity score is a 13-point stan-
at lower doses, lower peak concentrations, and reduced plasma dardized disease staging scale that assesses an individual’s abil-
fluctuations.5 The current open-label study evaluated the ity to perform tasks in 5 functional areas: to perform chores,
safety and explored the efficacy of an overnight conversion perform activities of daily living, work, manage finances, and
from tetrabenazine to deutetrabenazine therapy with subse- live at home. Higher scores indicate earlier disease and better
quent optional dose adjustment in patients with HD receiv- functional status. In early HD, the mean (SD) decline is 0.63
ing a stable tetrabenazine regimen for chorea. (0.75) points per year.7 Enrolled patients were required to have
daily access to reliable caregiver support to oversee study drug
administration, ensure attendance at study visits, and pro-
vide another level of oversight for safety. Patients with more
Methods severe functional impairment as indicated by a total func-
Study Design tional capacity score of 5 to 7 at screening were required to have
This is an ongoing, open-label, single-arm study that started a live-in caregiver.
on December 21, 2013, at 13 Huntington Study Group sites in Patients with serious undertreated psychiatric illness were
the United States and Australia. In-person study visits were excluded; however, concomitant stable antidepressant therapy
conducted at baseline (day 0), on the last day of tetrabena- was permitted during the study. Patients with active or past
zine use, and at weeks 1, 4, and 8 after overnight conversion suicidal ideation, thoughts, or behavior were excluded. Pa-
to deutetrabenazine therapy. Telephone consultations were tients were excluded if they scored 11 or higher on the Hospi-
made at weeks 2, 3, and 7. Written approvals from an inde- tal Anxiety and Depression Scale, 11 or higher on the Swallow-
pendent ethics committee or institutional review board were ing Disturbance Questionnaire, or 3 or higher on the dysarthria
received before the initiation of this study. Western Institu- score of the Unified Parkinson’s Disease Rating Scale. Con-
tional Review Board was used at all sites except for the comitant use of dopamine receptor antagonists, dopamine ago-
following individual institutional review boards: Western Syd- nists, levodopa, reserpine, N-methyl-D-aspartate receptor an-
ney Local Health District Human Research Ethics Commit- tagonists, or monoamine oxidase inhibitors within 30 days of
tee, University of Alabama Birmingham, Duke University screening was also exclusionary.
Health System, The Cooper Health System, Vanderbilt Uni-
versity Medical Center, and Rocky Mountain Movement Dis- Dosing
orders Center. Institutional review board approval was also ob- Patients were required to have been receiving a stable tetra-
tained at the Chesapeake Insitutional Review Board and benazine regimen (≥8 weeks) to be eligible. Each patient’s in-
centrally at the Research Subjects Review Board at the Uni- dividualized tetrabenazine treatment regimen was deter-
versity of Rochester. An independent, qualified health care pro- mined by his or her primary health care professional
fessional evaluated all patients to determine patient capacity independently of this study and before study participation.
to consent to participate. If patients lacked the capacity to Masked CYP2D6 (OMIM 124030) genotyping was conducted
provide informed consent, a legally authorized representa- at screening and remained masked throughout the study.
tive provided written informed consent, in addition to the Therefore, subsequent dosing decisions were made without
patients’ provided written assent. knowledge of the participants CYP2D6 metabolism status. The
initial deutetrabenazine daily dose was approximately half
Patients the prior tetrabenazine daily dose determined to profile
Patients were eligible if they were ambulatory adults with mani- comparable systemic exposure (area under the curve) to ac-
fest HD indicated by characteristic motor examination fea- tive metabolites (eTable in the Supplement). One week after

E2 JAMA Neurology Published online July 10, 2017 (Reprinted) jamaneurology.com

© 2017 American Medical Association. All rights reserved.

Downloaded From: http://archneur.jamanetwork.com/ by a Columbia University User on 07/11/2017


Tetrabenazine to Deutetrabenazine Conversion for Chorea Treatment Original Investigation Research

overnight conversion to deutetrabenazine, the investigator, in


Figure 1. Flow of Study Patients
consultation with the patient and caregiver, could begin weekly
dose adjustments of deutetrabenazine, if needed, to achieve
53 Assessed for eligibility
optimal chorea control. Dose adjustments were made on the
basis of assessment of tolerability and chorea control by the 16 Excluded
investigator. The maximum total daily dose of deutetrabena- 6 Used exclusionary medication
4 Did not meet other inclusion criteria
zine that could be used was 72 mg; however, if the patient was 6 Other
receiving a strong CYP2D6 inhibitor (eg, bupropion, parox-
etine, fluoxetine), the maximum total daily dose permitted was 37 Enrolled
42 mg.
1 Withdrew (investigator decision)
Assessments
Safety measures included assessment of adverse events 36 Converted to deutetrabenazine,
ongoing participation
(AEs), clinical laboratory tests, physical and neurologic
examinations, vital signs, electrocardiograms, and the fol- 36 Included in efficacy analysis
lowing scales: Unified Huntington’s Disease Rating Scale 37 Included in safety analysis
(UHDRS),8 Hospital Anxiety and Depression Scale,9 Colum-
bia Suicide Severity Rating Scale,10 Barnes Akathisia Rating After screening for eligibility, a total of 37 patients were enrolled. Of the 37
Scale,11 Swallowing Disturbance Questionnaire,12 Unified patients, 1 patient withdrew and all others continued in the ongoing study.

Parkinson’s Disease Rating Scale dysarthria item,13 Montreal


Cognitive Assessment,14 and Epworth Sleepiness Scale.15
The changes from baseline in the UHDRS total maximal Results
chorea (TMC) score and total motor score (TMS) were evalu-
ated as exploratory efficacy end points. The TMC score is a sub- Baseline Characteristics
set of the TMS; for the TMC and TMS, higher scores indicate A total of 53 patients were assessed for eligibility in the study.
greater motor signs. Of these, 14 patients were excluded (6 were taking exclusion-
ary medications, 4 did not meet inclusion criteria, 2 had se-
Safety vere suicidal ideation or depression, 1 had abnormal labora-
Adverse events were tabulated, and treatment-emergent tory values, and 1 was excluded for other reasons) and 2 patients
AEs were defined as events that began after initiation of declined to participate in the study. Thus, 37 ambulatory pa-
study drug treatment that were not present at baseline tients with manifest HD (mean [SD] age, 52.4 [11.5] years; 22
or, if present at baseline, that worsened in severity. Two- [59%] male and 15 [41%] female; 36 white [97.3%]) were en-
sided paired t tests were used for descriptive analysis of rolled (Figure 1). Table 1 lists baseline characteristics, includ-
changes from baseline for the Epworth Sleepiness Scale, ing the baseline TMC scores and TMSs. A total of 26 patients
Swallowing Disturbance Questionnaire, Unified Parkinson’s (70%) were being treated with antidepressants at baseline.
Disease Rating Scale dysarthria, and Barnes Akathisia Rating
Scale. Dosage
The initial median deutetrabenazine daily dose was 18.0 mg,
Efficacy approximately half the prior median tetrabenazine daily dose
Descriptive statistics were conducted for the UHDRS TMC score (37.5 mg). After dose adjustment, the median deutetrabena-
and TMS for the actual data presented by visit and changes from zine daily doses were 30.0 mg at week 4 (n = 37) and 36.0 mg
baseline. Two-sided paired t tests were performed to analyze at week 8 (n = 35). Eighteen of 37 participants (49%) were tak-
changes from baseline. Baseline values used for calculating ing the same dose at the week 8 visit as they were taking or
change from baseline for TMC score and TMS were the mean assigned at week 4.
of the values from the screening and day 0 baseline visits.
Lower scores on the TMC and TMS indicate less severe impair- Safety Outcomes
ment. Participants were receiving a stable tetrabenazine A total of 20 patients (54%) reported at least 1 treatment-
regimen for at least 8 weeks before the screening visit and con- emergent adverse event (Table 2). No AEs of worsening of cho-
tinued this regimen through the day 0 baseline visit before the rea were reported in this study. Adverse events leading to dose
day 1 change to deutetrabenazine. reduction (4 [11%]) or dose suspension (1 [3%]) were ob-
served, and no patient withdrew because of an AE. There were
Statistical Analysis no clinically significant differences in laboratory values, vital
Because this was an open-label safety study, statistical con- signs, body weight, or electrocardiogram findings between
siderations did not determine sample size. All analyses in- baseline and week 8. Two patients (5%) experienced mild de-
clude observed data, with no imputation of missing values. pression; the association with deutetrabenazine was rated as
P < .05 (2-sided) was considered statistically significant. Sta- possible by the site investigators. There were 9 reported cases
tistical analyses were performed using SAS statistical soft- of somnolence, of which 6 (67%) were mild; among the re-
ware (SAS Institute Inc). maining 3, 1 case required dose reduction and 1 required dose

jamaneurology.com (Reprinted) JAMA Neurology Published online July 10, 2017 E3

© 2017 American Medical Association. All rights reserved.

Downloaded From: http://archneur.jamanetwork.com/ by a Columbia University User on 07/11/2017


Research Original Investigation Tetrabenazine to Deutetrabenazine Conversion for Chorea Treatment

Table 1. Baseline Characteristics of the Study Patientsa Table 2. TEAEs Occurring in at Least 4% of Patients
Finding No. (%) of Patients
Characteristic (N = 37) TEAE (N = 37)
Patient demographic characteristics Any 20 (54)
Age, y 52.4 (11.5) Somnolence 9 (24)
Female, No. (%) 15 (41) Fall 4 (11)
White, No. (%) 36 (97) Nasopharyngitis 3 (8)
Patient clinical characteristics Anxiety 2 (5)
CAG repeat length 44.5 (3.3) Diarrhea 2 (5)
Body mass indexb 23.9 (4.7) Constipation 2 (5)
Weight, kg 71.4 (16.6) Dry mouth 2 (5)
UHDRS total functional capacity 8.3 (2.1) Depression 2 (5)
UHDRS total maximal chorea 12.5 (5.2) Irritability 2 (5)
UHDRS total motor score 38.5 (18.7) Abbreviation: TEAE, treatment-emergent adverse event.
Abbreviation: UHDRS, Unified Huntington’s Disease Rating Scale.
a
Data are presented as mean (SD) unless otherwise indicated.
b
Calculated as weight in kilograms divided by height in meters squared. Figure 2. Mean Change in Total Maximal Chorea (TMC) Score

Mean Change in TMC Score From Baseline


suspension. One patient (3%) experienced a serious AE of de- 2
hydration, possibly related to the study drug; the patient con-
tinued in the study without change in dose, and dehydration 1

resolved. There were no deaths reported in this study. Dys- 0


phagia was not reported as an AE, and Swallowing Distur-
bance Questionnaire scores did not change significantly dur- –1 a

ing the study.


–2

Deutetrabenazine Deutetrabenazine Deutetrabenazine


Efficacy Outcomes –3
(18.0 mg) (30.0 mg) (36.0 mg)
The mean (SD) TMS at baseline was 37.7 (18.6), and the mean (n = 37) (n = 37) (n = 36)
–4
(SD) TMC score was 12.5 (5.2). The mean (SD) reduction in TMC 0 1 2 3 4 5 6 7 8 9
score from baseline to week 1 was 0.7 (2.6) UHDRS units Length of Deutetrabenazine Treatment, wk

(P = .10) (Figure 2). The mean (SD) reduction in TMS from base-
The TMC score was assessed during 8 weeks. The TMC assessments at baseline
line was 2.1 (5.4) UHDRS units (P = .02) at week 1 after dose con-
(blue dotted line) and weeks 1, 4, and 8 are represented for the median daily
version (Figure 3). The mean (SD) reduction in TMC score from doses of deutetrabenazine. Error bars represent SEM.
baseline to week 4 was 0.6 (3.0) units (P = .21) (Figure 2). Cho- a
P < .001.
rea control improved compared with baseline at week 8 (2.1
[3.2]; P < .001). The mean (SD) TMS was unchanged com- pants up to week 8 after dose conversion. Although patients
pared with baseline at week 4 (1.7 [8.3]; P = .22) and week 8 with active or undertreated psychiatric symptoms, such as de-
(2.4 [8.7]; P = .10). pression, were excluded according to the study design, pa-
tients receiving stable antidepressant treatment were en-
rolled. There was a notable absence of suicidal ideation and
parkinsonism during this 8-week study. Because this cohort
Discussion
is a part of an ongoing, long-term safety study, the safety pro-
Overnight treatment conversion from tetrabenazine 3 times file of those patients with impaired CYP2D6 metabolism has
daily to deutetrabenazine twice daily can safely maintain cho- not been included in this early safety analysis because CYP2D6
rea control in patients with HD. Deutetrabenazine therapy was genetic variant status remains masked to investigators and par-
generally well tolerated by patients after the overnight con- ticipants. In addition, the results of this open-label study should
version from tetrabenazine. Adherence with a twice-daily be interpreted with caution because this study was not ran-
medication was excellent and may be attributable to simpli- domized or masked.
fied medication regimens compared with existing therapies.
The favorable safety profile observed in this study is reflec- Limitations
tive of the findings in the First-HD study, which supports the This study was performed with an open-label design without
hypothesis that deuterium substitution–mediated attenua- a control group by unmasked site investigators determining
tion of drug metabolism allows for maximal efficacy through chorea and TMSs in unmasked patients who had treatment con-
dose adjustment while maintaining tolerability in patients with verted from tetrabenazine to deutetrabenazine, factors that
HD.6 The safety of deutetrabenazine is further highlighted by may impart treatment bias. Therefore, all efficacy findings re-
the low rates of depression and anxiety observed in partici- ported in this study should be considered to be exploratory.

E4 JAMA Neurology Published online July 10, 2017 (Reprinted) jamaneurology.com

© 2017 American Medical Association. All rights reserved.

Downloaded From: http://archneur.jamanetwork.com/ by a Columbia University User on 07/11/2017


Tetrabenazine to Deutetrabenazine Conversion for Chorea Treatment Original Investigation Research

patients thought that the combination of chorea level, effect


Figure 3. Mean Change in Total Motor Score (TMS) After Switch
of chorea on the specific patient, and drug tolerance justified
3 further dose adjustments, up or down, to maximize overall pa-
tient status. The TMC score improved at week 8 compared with
2
Mean Change in TMS From Baseline

baseline, suggesting that patients need to be reassessed for tol-


Deutetrabenazine Deutetrabenazine Deutetrabenazine erability and chorea control after initial treatment conversion
1
(18.0 mg) (30.0 mg) (36.0 mg)
(n = 37) (n = 37) (n = 36) and any subsequent dose adjustments. The improvement in
0
chorea raises the possibility that some patients were not able
–1 a to tolerate higher doses of tetrabenazine or, despite being
stable, may have had an incomplete response on tetrabena-
–2
zine. Patients in either circumstance may be more likely to en-
–3 roll in this type of study compared with other patients receiv-
ing a stable tetrabenazine regimen, creating an ascertainment
–4 bias. The starting dose for conversion may also have been too
0 1 2 3 4 5 6 7 8 9
Length of Deutetrabenazine Treatment, wk low in some patients.

The TMS was evaluated during 8 weeks. The TMS assessments at baseline (blue
dotted line) and weeks 1, 4, and 8 are represented for the median daily doses of
deutetrabenazine. Error bars represent SEM.
Conclusions
a
P = .02. The patients enrolled in this study were experiencing chorea
and receiving tetrabenazine with clinical benefit before treat-
Because the deutetrabenazine starting dose was thought to ment conversion. This proof-of-concept study reveals that
match the systemic active metabolite exposure achieved with chorea control can be safely maintained when converting
the prior tetrabenazine dose, baseline levels of chorea were ex- from tetrabenazine to deutetrabenazine, with a favorable
pected to be maintained but not necessarily improve after the safety profile. Change from a nondeuterated to a deuterium
switch to deutetrabenazine therapy. Worsening of chorea was substituted compound can achieve treatment goals with
not observed. With optional continued dose adjustments dur- fewer doses given across the day, lower total daily dose, and
ing 8 weeks, chorea control improved. There are a few expla- equivalent or potentially improved tolerance, all important
nations for continued dose adjustment after 4 weeks in half considerations in populations with neurodegenerative
of the patients. Doses could be adjusted if site investigators and disease.

ARTICLE INFORMATION Medical University of South Carolina, Charleston receiving grants and nonfinancial support from HSG
Accepted for Publication: May 10, 2017. (Vaughan); Department of Neurology, Virginia on behalf of Auspex Pharmaceuticals during the
Commonwealth University, Richmond (Testa). conduct of the study and other support from
Published Online: July 10, 2017. Auspex Pharmaceuticals outside the submitted
doi:10.1001/jamaneurol.2017.1352 Author Contributions: Drs Frank and Testa had full
access to all the data in the study and take work. Dr Furr-Stimming reported serving as a
Author Affiliations: Department of Neurology, responsibility for the integrity of the data and the consultant for Cynapsus and on the speakers’
Harvard Medical School, Boston, Massachusetts accuracy of the data analysis. bureau for Lundbeck (Xenazine) and Teva (Azilect).
(Frank); Teva Pharmaceutical Industries, Frazer, Study concept and design: Frank, Stamler, Kayson, Dr Jankovic reported receiving research and/or
Pennsylvania (Stamler); Clinical Trials Coordination Davis, Oakes, Vaughan, Testa. training grants from Adamas Pharmaceuticals Inc,
Center, University of Rochester, Rochester, New Acquisition, analysis, or interpretation of data: Allergan Inc, CHDI Foundation, Civitas/Acorda
York (Kayson, Eberly, Goldstein); Department of Frank, Stamler, Claassen, Colcher, Davis, Duker, Therapeutics, Dystonia Medical Research
Neurology, Vanderbilt University Medical Center, Elmer, Furr-Stimming, Gudesblatt, Hunter, Foundation, Huntington Study Group, Ipsen
Nashville, Tennessee (Claassen); Department of Jankovic, Kostyk, Kumar, Loy, Mallonee, Oakes, Limited, Kyowa Haako Kirin Pharma Inc, Lundbeck
Neurology, Cooper University Hospital, Camden, Scott, Sung, Goldstein, Vaughan, Testa. Inc, Medtronic, Merz Pharmaceuticals, Michael J.
New Jersey (Colcher, Mallonee); CSD Biostatistics, Drafting of the manuscript: Frank, Stamler, Colcher, Fox Foundation for Parkinson Research, National
Tucson, Arizona (Davis); Department of Neurology, Davis, Duker, Furr-Stimming, Gudesblatt, Hunter, Institutes of Health, National Parkinson Foundation,
University of Cincinnati, Cincinnati, Ohio (Duker); Jankovic, Loy, Oakes, Vaughan, Testa. Parkinson Study Group, Pfizer, Prothena
The Center for Neurologic Health, Toledo, Ohio Critical revision of the manuscript for important Biosciences Inc, Psyadon Pharmaceuticals Inc, St.
(Elmer); Department of Neurology, The University intellectual content: All authors. Jude Medical, and Teva Pharmaceutical Industries
of Texas Medical School, Houston (Furr-Stimming); Statistical analysis: Davis, Oakes. Ltd. Dr Jankovic reported serving as a consultant or
South Shore Neurologic Associates, Islip, New York Obtained funding: Frank. as an advisory committee member for Adamas
(Gudesblatt); Department of Neurology, Baylor Administrative, technical, or material support: Pharmaceuticals Inc, Allergan Inc, and Teva
College of Medicine, Houston, Texas (Hunter, Kayson, Goldstein. Pharmaceutical Industries Ltd. Dr Jankovic
Jankovic); Department of Neurology, The Ohio Study supervision: Frank, Stamler, Kayson, reported receiving royalties from Cambridge,
State University, Columbus (Kostyk); Rocky Goldstein, Testa. Elsevier, Future Science Group, Hodder Arnold,
Mountain Movement Disorders Center, Englewood, Lippincott Williams and Wilkins, and
Colorado (Kumar); Department of Neurology, Conflict of Interest Disclosures: Dr Frank reported Wiley-Blackwell and serving on editorial boards for
University of Sydney, Sydney, Australia (Loy); receiving grants from the Huntington Study Group. Medlink Neurology, Expert Review of
Department of Statistics, University of Rochester, Dr Stamler reported being an employee of Auspex Neurotherapeutics, Neurology in Clinical Practice,
Rochester, New York (Oakes); Department of Pharmaceuticals. Ms Kayson reported receiving The Botulinum Journal, PeerJ, Therapeutic Advances
Neurology, Duke University, Durham, North honoraria and funding for a meeting from Raptor in Neurological Disorders, Neurotherapeutics,
Carolina (Scott); Department of Neurology, Pharmaceutical and serving as a consultant to Tremor and Other Hyperkinetic Movements, Journal
University of Alabama School of Medicine, Pfizer Inc. Dr Davis reported serving as a consultant of Parkinson's Disease, and UpToDate. Dr Kostyk
Birmingham (Sung); Department of Neurology, for Auspex Pharmaceuticals. Dr Duker reported reported receiving consultant fees from the

jamaneurology.com (Reprinted) JAMA Neurology Published online July 10, 2017 E5

© 2017 American Medical Association. All rights reserved.

Downloaded From: http://archneur.jamanetwork.com/ by a Columbia University User on 07/11/2017


Research Original Investigation Tetrabenazine to Deutetrabenazine Conversion for Chorea Treatment

National Institutes of Health and the US Food and Columbus, Ohio; Daniel Claassen, MD, Department 4. Jankovic J. Dopamine depleters in the treatment
Drug Administration and has had meeting travel of Neurology, Vanderbilt University, Nashville, of hyperkinetic movement disorders. Expert Opin
costs covered through the Huntington's Study Tennessee; Rajeev Kumar, MD, and Vicki Segro, Pharmacother. 2016;17(18):2461-2470.
Group and Pfizer Pharmaceuticals. Dr Kumar MSN, C-ANP, Rocky Mountain Movement Disorders 5. AustedoTM [package insert]. North Wales, PA:
reported receiving personal fees from Teva Center, Englewood, Colorado; Clement Loy, MD, Teva Pharmaceuticals Inc; 2017.
Pharmaceuticals outside the submitted work. Dr Samuel Kim, MD, Elizabeth McCusker, Department
Mallonee reported receiving other support from of Neurology, Westmead Hospital, Sydney, 6. Frank S, Testa CM, Stamler D, et al; Huntington
HSG on behalf of Auspex Pharmaceuticals during Australia; William M. Mallonee, MD, Hereditary Study Group. Effect of deutetrabenazine on chorea
the conduct of the study. Dr Oakes reported Neurological Disease Centre, Wichita, Kansas; among patients with Huntington Disease:
receiving research support from Auspex for this and Andrew Duker, MD, University of Cincinnati, a randomized clinical trial. JAMA. 2016;316(1):40-50.
another study of SD809 in Huntington disease Cincinnati, Ohio; Fredy Revilla, MD, University 7. Feigin A, Kieburtz K, Bordwell K, et al. Functional
(HD), Vaccinex Inc and Prana Pharmaceuticals for Neurology Inc, Cincinnati, Ohio; Lawrence Elmer, decline in Huntington’s disease. Mov Disord. 1995;
studies in HD, Biogen Inc for a study in Parkinson MD, PhD, The University of Toledo 10(2):211-214.
disease, and the National Institutes of Health for Gardner-Mcmaster Parkinson Center, Toledo, Ohio; 8. Huntington Study Group. Unified Huntington’s
studies in HD and Parkinson disease and personal Burton Scott, MD, PhD, Duke Health Center, Duke Disease Rating Scale: reliability and consistency.
honoraria from Raptor Pharmaceuticals and University, Durham, North Carolina; Patrick Hickey, Mov Disord. 1996;11(2):136-142.
Voyager Inc. Dr Sung reported receiving personal DO, Duke University Medical Center, Durham,
fees from Lundbeck Inc. Dr Testa reported receiving North Carolina; Erin Furr-Stimming, MD, University 9. Zigmond AS, Snaith RP. The hospital anxiety and
grants from the Huntington Study Group, Teva of Texas at Houston, Houston; William Ondo, MD, depression scale. Acta Psychiatr Scand. 1983;67
Pharmaceuticals, and the CHDI Foundation and University of Texas Medical School at Houston, (6):361-370.
other support from Auspex Pharmaceuticals and Houston; Amy Colcher, MD, The Cooper University 10. Posner K, Brown GK, Stanley B, et al. The
Lundbeck Pharmaceuticals outside the submitted Health System, Camden, New Jersey; Andrew Columbia-Suicide Severity Rating Scale: initial
work. No other disclosures are reported. McGarry, MD, Cooper University Hospital, Camden, validity and internal consistency findings from three
Funding/Support: This study was supported by New Jersey; and Mark Gudesblatt, MD, South Shore multisite studies with adolescents and adults. Am J
Auspex Pharmaceuticals, a wholly owned Neurologic Associates, Patchoque, New York. Psychiatry. 2011;168(12):1266-1277.
subsidiary of Teva Pharmaceutical Industries Ltd. Additional Contributions: Administrative, 11. Barnes TR. A rating scale for drug-induced
Role of the Funder/Sponsor: Auspex technical, and material support was provided by akathisia. Br J Psychiatry. 1989;154:672-676.
Pharmaceuticals aided in the design of the study; Jacquelyn Whaley, MS, Sherry Weston, AAS 12. Manor Y, Giladi N, Cohen A, Fliss DM, Cohen JT.
provided input on the oversight, conduct, and (University of Rochester, Rochester, New York), and Validation of a swallowing disturbance
management of the study; played no role in the Nupur Patel (Teva Pharmaceuticals, Frazer, questionnaire for detecting dysphagia in patients
collection of data but provided oversight with Pennsylvania); these individuals received with Parkinson’s disease. Mov Disord. 2007;22(13):
management; was involved with but did not play compensation for their work. We thank the patients 1917-1921.
the primary role in analysis and interpretation of and families who participated in this study.
13. Fahn S, Elton RL; Members of the UDC. Unified
the data; aided in the preparation of the manuscript Parkinson’s Disease Rating Scale. In: Fahn S,
(figures, references, and formatting) and reviewed REFERENCES
Marsden CD, Calne DB, Goldstein M, eds. Recent
content for intellectual property issues; and 1. Burgunder J-M, Guttman M, Perlman S, Developments in Parkinson’s Disease. Vol 2. Florham
provided approval of the final draft of the Goodman N, van Kammen DP, Goodman L. An Park, NJ: Macmillan Healthcare Information; 1987:
manuscript. The decision to submit the manuscript international survey-based algorithm for the 153-163.
for publication was based on Huntington Study pharmacological treatment of chorea in
Group principles with sponsor input through the Huntington’s disease. PLoS Curr. 2011;3:RRN1260. 14. Nasreddine ZS, Phillips NA, Bédirian V, et al.
steering committee. The Montreal Cognitive Assessment, MoCA: a brief
2. Jankovic J, Roos RA. Chorea associated with screening tool for mild cognitive impairment. J Am
Group Information: The Huntington Study Group/ Huntington’s disease: to treat or not to treat? Mov Geriatr Soc. 2005;53(4):695-699.
Alternatives for Reducing Chorea in Huntington Disord. 2014;29(11):1414-1418.
Disease Investigators are as follows: Joseph 15. Johns MW. A new method for measuring
3. Kenney C, Hunter C, Jankovic J. Long-term daytime sleepiness: the Epworth sleepiness scale.
Jankovic, MD, and Joohi Jimenez-Shahed, MD, tolerability of tetrabenazine in the treatment of
Parkinson’s Disease Center and Movement Sleep. 1991;14(6):540-545.
hyperkinetic movement disorders. Mov Disord.
Disorders Clinic, Houston, Texas; Sandra Kostyk, 2007;22(2):193-197.
MD, PhD, Ohio State University Medical Center,

E6 JAMA Neurology Published online July 10, 2017 (Reprinted) jamaneurology.com

© 2017 American Medical Association. All rights reserved.

Downloaded From: http://archneur.jamanetwork.com/ by a Columbia University User on 07/11/2017

You might also like