throbber
Tetrabenazine as antichorea therapy in
`Huntington disease
`A randomized controlled trial
`
`Huntington Study Group*
`
`Abstract—Background: Tetrabenazine (TBZ) selectively depletes central monoamines by reversibly binding to the type 2
`vesicular monoamine transporter. Open-label reports indicate TBZ is effective in treating chorea. Objective: To examine
`the safety, efficacy, and dose tolerability of TBZ for treating chorea in Huntington disease (HD). Methods: The authors
`randomized 84 ambulatory patients with HD to receive TBZ (n ⫽ 54) or placebo (n ⫽ 30) for 12 weeks. TBZ was increased
`over 7 weeks up to a maximum of 100 mg/day or until the desired antichoreic effect occurred or intolerable adverse effects
`supervened. The primary outcome was the change from baseline in the chorea score of the Unified Huntington’s Disease
`Rating Scale (UHDRS) Results: TBZ treatment resulted in a reduction of 5.0 units in chorea severity compared with a
`reduction of 1.5 units on placebo treatment (adjusted mean effect size ⫽ ⫺3.5 ⫾ 0.8 UHDRS units [mean ⫾ SE]; 95% CI:
`⫺5.2, ⫺1.9; p ⬍ 0.0001). There was also a significant benefit on ratings of clinical global improvement. There were five
`study withdrawals in the TBZ group and five serious adverse events (SAEs) in four subjects (drowning suicide, compli-
`cated fall, restlessness/suicidal ideation, and breast cancer) compared with one withdrawal and no SAEs in the placebo
`group. Conclusion: Tetrabenazine (TBZ), at adjusted dosages of up to 100 mg/day, effectively lessens chorea in ambulatory
`patients with Huntington disease. TBZ should be dosed individually based on ongoing assessment of possible adverse side
`effects.
`NEUROLOGY 2006;66:366–372
`
`Although there is no established treatment to delay
`the onset or forestall the progression of illness asso-
`ciated with Huntington disease (HD), symptomatic
`treatment of chorea may be beneficial in selected
`individuals.1,2 Neuroleptics reduce chorea but are as-
`sociated with extrapyramidal side effects. Several re-
`ports have suggested that tetrabenazine (TBZ)
`ameliorates hyperkinetic movement disorders,3-9 but
`controlled studies have been limited.10-12 TBZ is cur-
`rently marketed in nine countries including Canada,
`but it has been available in the United States only
`for research purposes.
`TBZ binds with high affinity (Kd ⫽ 2.4 nM) and
`selectivity to the CNS vesicular monoamine trans-
`porter (VMAT2),13-18 effectively depleting monoamines
`and serotonin (5HT) from nerve terminals by inhibiting
`their transport into presynaptic vesicles.14,19-21 TBZ
`depletes dopamine (IC50 ⫽ 0.4 mg/kg) preferentially
`
`Additional material related to this article can be found on the Neurology
`Web site. Go to www.neurology.org and scroll down the Table of Con-
`tents for the February 14 issue to find the title link for this article.
`
`over norepinephrine or 5HT (IC50 ⫽ 2 mg/kg), whereas
`reserpine, the only currently available monoamine de-
`pleter, is less selective and also binds to VMAT1 ex-
`pressed in the periphery.22-25 The highest binding
`density for TBZ is in the caudate nucleus, putamen,
`and nucleus accumbens, areas known to bear the brunt
`of pathology in HD.26,27 VMAT binding and mono-
`amine depletion by TBZ are reversible, last hours,
`and are not modified by chronic treatment,
`whereas those by reserpine are irreversible and
`last days to weeks.28,29 Unlike reserpine, TBZ has
`therefore not been associated with troublesome pe-
`ripheral side effects such as hypotension.
`We report the first multicenter, prospective,
`double-blind, placebo-controlled dose-finding study of
`TBZ for the treatment of chorea in HD.
`
`Methods. Subjects. Eligible subjects had HD as confirmed by
`the presence of a characteristic movement disorder (chorea), a
`family history, and an expanded CAG repeat (n ⱖ 37). All partici-
`pants were required to be independently ambulatory, to have a
`screening total functional capacity (TFC) of ⬎5,30 and a total max-
`imal chorea of ⱖ10 (sum of the maximal chorea scores for facial,
`
`*See the Appendix for a complete listing of authors.
`From the Clinical Trials Coordination Center, Department of Neurology, University of Rochester School of Medicine and Dentistry, NY.
`Disclosure: This study was funded by a grant from Prestwick Pharmaceuticals, Inc., to the University of Rochester and in turn through subcontracts to the
`participating research sites. The Huntington Study Group (HSG) is a nonprofit consortium of Huntington’s disease investigators (http://www.huntington-
`study-group.org/). None of the HSG investigators or staff had equity interests with Prestwick Pharmaceuticals, Inc. Dr. Fahn received consulting fees of less
`than $10,000. Dr. Marshall presented data at national meetings for which he received travel reimbursement from Prestwick Pharmaceuticals, Inc. Dr.
`Clarence-Smith, Dr. O’Brien, and Ms. Wilson are employees of Prestwick Pharmaceuticals, Inc. The HSG Coordination and Biostatistics Centers at the
`University of Rochester independently compiled and analyzed the data for this study.
`Received April 4, 2005. Accepted in final form October 18, 2005.
`Address correspondence and reprint requests to Dr. F.J. Marshall, Clinical Trials Coordination Center, Department of Neurology, University of Rochester
`School of Medicine and Dentistry, 1359 Mt. Hope Ave., Suite 223, Rochester, NY 14620; e-mail: fred.marshall@ctcc.rochester.edu
`
`366 Copyright © 2006 by AAN Enterprises, Inc.
`
`Apotex Ex. 1010
`
`Apotex v. Auspex
`IPR2021-01507
`
`

`

`buccal–oral–lingual, trunkal, and each extremity from the motor
`subscale of the Unified Huntington’s Disease Rating Scale
`[UHDRS]).31 Patients were excluded if they had disabling depres-
`sion, dysphagia, or dysarthria. Eligible participants could not
`have been treated in the past with TBZ or currently with
`dopamine-depleting medications, dopamine D2 receptor blockers,
`selective or nonselective monoamine oxidase inhibitors, levodopa,
`dopamine agonists, amantadine, or memantine. Patients previ-
`ously treated with dopamine D2 receptor blockers were enrolled,
`provided they had been off these medications for at least 4 weeks.
`Patients were permitted to take existing antidepressant or benzo-
`diazepine medication if receiving stable dosages for at least 8
`weeks prior to the randomization visit. Subjects needed to be
`accompanied by a caregiver.
`Study design. This
`randomized, double-blind, placebo-
`controlled study was carried out at 16 Huntington Study Group
`(HSG) sites in the United States. The primary purpose was to
`study the efficacy, dosing, tolerability, and safety of TBZ in two
`parallel groups of HD subjects with clinically manifest chorea,
`allocated 2:1 to receive TBZ or placebo. The study protocol re-
`ceived institutional review board approval at each participating
`center prior to subject enrollment.
`A data and safety monitoring committee consisting of two phy-
`sicians experienced in clinical research and a biostatistician, who
`were not otherwise involved in the conduct of the study, convened
`on three occasions during the course of the trial to assess safety,
`including unblinded analysis of accrued adverse events and labo-
`ratory tests.
`Study procedures. Consenting research participants who sat-
`isfied the eligibility criteria were randomized (2:1) to receive ei-
`ther TBZ or placebo for 12 weeks. Treatment assignment was
`concealed from subjects and investigators, and randomization was
`performed through a computerized module developed in the De-
`partment of Biostatistics at the University of Rochester. TBZ was
`formulated in 12.5-mg tablets and was identical in appearance to
`matching placebo tablets. During the first 7 weeks of the study,
`dosage was titrated in blinded fashion by providing 1 tablet on the
`first day, then 1 tablet twice daily for the remainder of the first
`week. Subsequently, the number of tablets was increased by 1 per
`week up to 8 tablets per day in three divided doses or until a
`desired antichoreic effect was achieved or intolerable adverse ef-
`fects occurred. To reduce intolerable side effects, the number of
`tablets was reduced to the participant’s previously well-tolerated
`level or lower if necessary. Participants were permitted one sus-
`pension of study drug for up to 7 days. By the end of the first 7
`weeks of the study, participants were on their “best dose,” and
`during the last 5 weeks of the study, the dosage remained con-
`stant unless reduced because of intolerable adverse effects. After
`12 weeks, study drugs were withdrawn, and subjects returned for
`a follow-up visit 1 week later.
`Assessments. Subjects were examined at baseline and at the
`end of weeks 1, 3, 5, 7, 9, 12, and 13. At each examination, the
`investigator rated the UHDRS total maximal chorea score. The
`Clinical Global Impression (CGI) was done at weeks 1, 3, 5, 7, 9,
`and 12. A full UHDRS, including motor, cognitive, behavioral, and
`functional components, was completed at baseline, at the end of
`the titration phase, and at the end of the maintenance phase.31
`Complete blood count and general chemistry profiles were ob-
`tained at screening and at the end of week 12.
`Participants were also evaluated for adverse events, parkin-
`sonism subscore of the UHDRS (sum of finger taps, pronate/
`supinate hands, rigidity arms, bradykinesia–body, gait, tandem
`walking, and retropulsion pull test), akathisia as measured by the
`Barnes Akathisia Scale,32 speech and swallowing as measured by
`Unified Parkinson’s Disease Rating Scale Part II, item 5 (UPDRS
`speech) and item 7 (UPDRS swallowing),33 depression as mea-
`sured by the 17-item Hamilton Depression Scale (HAM-D),34
`sleepiness as measured by the Epworth Sleepiness Scale (ESS),35
`vital signs (systolic and diastolic blood pressure, radial artery
`pulse rate) while sitting, and a 12-lead EKG. We also piloted a
`new instrument, the Functional Impact Scale (FIS), to assess the
`degree of difficulty with bathing, dressing, feeding, social isola-
`tion, and toileting. FIS information was obtained from the accom-
`panying caregiver, and each item was graded on a scale of 0 to 3
`(see appendix E-1 on the Neurology Web site; go
`to
`www.neurology.org).
`An amendment to the initial protocol, adding a videotape to
`
`document subjects’ chorea at the end of the maintenance phase
`(week 12) and after withdrawal (week 13), was instituted during
`the course of the study. An independent movement disorder ex-
`pert who was blinded to subjects’ treatment assignments, as well
`as to whether a given tape had been done at the conclusion of the
`maintenance phase or the conclusion of the washout phase, rated
`each tape as to chorea severity using the UHDRS and overall
`clinical improvement using the CGI Improvement Scale.
`Statistical analysis. The primary prespecified efficacy out-
`come measure was the difference between the baseline total max-
`imal chorea score and the average of the score at week 9
`(midmaintenance phase) and week 12 (end of maintenance phase).
`Analysis of covariance (ANCOVA) was used to assess the treat-
`ment effect. The model included the site and baseline score. The
`treatment-by-site interaction was not significant and was there-
`fore not included in the final model. Primary analyses were based
`on intention to treat. If either the week 9 or the week 12 data
`were missing for a given subject, the missing data were imputed
`as the one available score. If both week 9 and week 12 data were
`missing, the subject’s last available score (after baseline) was car-
`ried forward.
`Power calculations were based on the results obtained in the
`subgroup of patients whose baseline total maximal chorea score
`was at least 10 in a previous 12-week HSG trial. Given a 2:1
`randomization (TBZ/placebo), an estimated dropout rate of 15%,
`and an estimated baseline chorea score of 14.5 ⫾ 3.5 (mean ⫾ SD),
`a total sample size of 72 subjects gave ⬎80% power to detect an
`effect size of at least 2.7-unit change in the total maximal chorea
`score. The study sample provided a probability of ⬎90% for detect-
`ing an adverse event that occurred with a frequency of 10% or
`more in the population from which the participants were drawn
`and a power of ⬎99% for detecting an absolute difference in toler-
`ability of 50% between the placebo group and the TBZ group.
`Because of the large number of potential outcome measures,
`the analysis plan prespecified that four secondary endpoints (CGI
`Global Improvement score, change in total motor score, change in
`functional checklist, change in gait score) be analyzed in descend-
`ing hierarchy, with definitive analyses ceasing when the signifi-
`cance level reached ␣ ⱖ 0.05. The CGI Global Improvement score
`was analyzed using only data from week 12, as prespecified in the
`analysis plan. ANCOVA procedures comparable with those in the
`primary efficacy analysis were used for the change from baseline
`in the other measures. Exploratory analyses of other outcome
`measures were undertaken after the hierarchical analyses
`reached a stopping point. Baseline characteristics were compared
`using t tests or ␹2 tests, as appropriate. Group comparisons on
`tolerability measures and adverse events were made with
`continuity-corrected ␹2 tests or F tests.
`
`Results. Baseline comparability.
`Figure 1 outlines the
`flow of participants in the study. Ninety-one individuals
`were screened and 84 eligible subjects were randomized
`between July and December 2003. Table E-1 lists the base-
`line demographic and outcome variable scores for all par-
`ticipants. The groups were comparable with regard to age,
`CAG repeat length, gender, duration of illness, and propor-
`tion with a history of depression. Subjects randomized to
`TBZ scored worse at baseline on the Symbol Digit Test of
`the UHDRS cognitive battery (p ⫽ 0.018), as well as on the
`pilot FIS (p ⫽ 0.035). Although the other individual
`UHDRS measures were not statistically different across
`treatment groups, subjects randomized to TBZ tended to
`score somewhat more poorly at baseline on most UHDRS
`measures of severity with the notable exception of chorea
`score (see table E-1). There was a strong inverse correla-
`tion between CAG repeat length and the age at symptom
`onset in our study sample (r ⫽ ⫺0.63, p ⬍ 0.0001), as
`observed in other studies of HD patients.36,37
`Outcome summaries. Table 1 shows the adjusted mean
`changes in treatment-related outcome measures from
`baseline to the prespecified study endpoint (average of
`week 9 and week 12 scores), including the adjusted treat-
`February (1 of 2) 2006 NEUROLOGY 66 367
`
`Apotex Ex. 1010
`
`

`

`Primary efficacy outcome: Impact of TBZ on cho-
`rea. Although there was a mild reduction in total maxi-
`mal chorea in the placebo group between baseline and the
`average of the week 9 and week 12 assessments (⫺1.5 ⫾
`0.7 UHDRS units; adjusted mean ⫾ SE), the impact of
`TBZ on chorea severity was larger: ⫺5.0 ⫾ 0.5 UHDRS
`units (p ⬍ 0.0001). The adjusted effect size of ⫺3.5 UH-
`DRS units (95% CI: ⫺5.2, ⫺1.9), represents a 23.5% aver-
`age reduction in baseline chorea severity due to TBZ
`(figure 2). Whereas only 20% of placebo subjects had a
`reduction in chorea of at least 3 UHDRS units, 69% of
`TBZ-treated subjects had a reduction of at least this mag-
`nitude (adjusted odds ratio ⫽ 9.9; 95% CI: 3.2, 29.9; p ⬍
`0.0001). The TBZ-related reduction in chorea was not re-
`lated to age, gender, trinucleotide repeat length, gender of
`affected parent, baseline CGI of severity, or baseline cho-
`rea score.
`Secondary efficacy outcomes. TBZ was superior to pla-
`cebo on the CGI Global Improvement Scale, with an ad-
`justed effect size (improvement) of ⫺0.7 CGI unit (95% CI:
`⫺1.3, ⫺0.2) on this 7-point scale. Figure 3 shows the distri-
`bution of CGI Global Improvement scores at week 12 by
`treatment group. Twenty-four percent of subjects in the
`placebo group achieved a CGI Global Improvement score of
`ⱕ3 (corresponding to at least minimal global improve-
`ment) compared with 69% of TBZ subjects (p ⫽ 0.0001).
`Only two participants receiving placebo (6.9%) had more
`than minimal global improvement, whereas 23 TBZ partic-
`
`Figure 1. Flow of study subjects.
`
`ment effect with 95% CIs, the effect sizes as a function of
`the average baseline scores, and the effect sizes as a func-
`tion of the total scale range for each variable.
`
`Table 1 Change in outcome variables (adjusted mean ⫾ SE) from baseline to average of week 9 and week 12 scores
`(ITT/LOCF analyses)
`
`Direction of
`favorable
`change
`
`Placebo,
`n ⫽ 30
`
`TBZ,
`n ⫽ 54
`
`p Value
`⬍ 0.05
`
`Adjusted
`mean TE,
`scale units
`
`TE 95%
`CI, scale
`units
`
`TE,
`% baseline
`mean score
`
`TE,
`% scale
`range
`
`Primary outcome variable
`⌬ UHDRS tot max. chorea
`Secondary outcome variables
`CGI Global Improvement‡
`⌬ UHDRS total motor
`Exploratory outcome variables
`⌬ UHDRS functional checklist
`⌬ 17-item HAM-D
`⌬ Epworth Sleepiness
`⌬ Stroop test
`Word reading
`Color naming
`Interference
`
`⫺
`
`⫺
`
`⫹
`
`⫺
`
`⫺
`
`⫹
`
`⫹
`
`⫹
`
`⫺1.5 ⫾ 0.7 ⫺5.0 ⫾ 0.5
`
`0.0001*
`
`⫺3.5†
`
`⫺5.2, ⫺1.9
`
`⫺23.5
`
`⫺12.5
`
`3.0 ⫾ 0.2
`3.7 ⫾ 0.2
`⫺3.5 ⫾ 1.5 ⫺6.8 ⫾ 1.1
`
`0.007*
`
`0.4 ⫾ 0.4 ⫺0.8 ⫾ 0.3
`⫺2.4 ⫾ 0.4 ⫺0.7 ⫾ 0.3
`⫺0.3 ⫾ 0.6
`1.5 ⫾ 0.5
`
`0.02§
`0.003§
`0.02§
`
`1.8 ⫾ 2.1 ⫺4.8 ⫾ 1.5
`1.3 ⫾ 1.7 ⫺1.7 ⫾ 1.2
`1.5 ⫾ 1.2 ⫺1.5 ⫾ 0.9
`
`0.01§
`
`⫺0.7
`⫺3.3
`
`⫺1.2
`1.6
`1.8
`
`⫺6.6
`⫺2.9
`⫺3.0
`
`⫺1.3, ⫺0.2
`⫺7.0, 0.3
`
`⫺2.2, ⫺0.2
`0.6, 2.7
`0.3, 3.4
`
`⫺11.8, ⫺1.5
`⫺7.3, 1.4
`⫺6.0, 0.0
`
`⫺7.1
`
`⫺6.3
`33.8
`47.7
`
`⫺12.1
`⫺6.6
`⫺12.9
`
`⫺10.0
`⫺2.7
`
`⫺4.8
`3.1
`7.5
`
`—
`—
`—
`
`Per the prespecified hierarchical analysis plan (see text), only changes in total maximal chorea and CGI Global Improvement scores
`achieved definitive significance.
`
`* Favors TBZ over placebo.
`† The study was powered to detect an effect size of 2.7 UHDRS units.
`‡ CGI Global Improvement analysis based on week 12 rating per analysis plan. For CGI Global Improvement, a score of ⬍4 ⫽ improve-
`ment, 4 ⫽ no change, ⬎4 ⫽ worsening.
`§ Favors placebo over TBZ.
`
`ITT ⫽ intention-to-treat analysis; LOCF ⫽ last observation carried forward analysis; TBZ ⫽ tetrabenazine; TE ⫽ treatment effect (TBZ
`compared with placebo); UHDRS ⫽ Unified Huntington’s Disease Rating Scale; HAM-D ⫽ Hamilton Depression Scale; CGI ⫽ Clinical
`Global Impression; UPDRS ⫽ Unified Parkinson’s Disease Rating Scale.
`
`368 NEUROLOGY 66 February (1 of 2) 2006
`
`Apotex Ex. 1010
`
`

`

`Figure 2. Mean change from baseline
`in Unified Huntington’s Disease Rating
`Scale total maximal chorea score by
`treatment group (last observation car-
`ried forward [LOCF] except for week
`13). Change from baseline to week 12
`favors tetrabenazine (p ⫽ 0.0001; anal-
`ysis of covariance, intention to treat,
`LOCF, side panel). There was blinded
`washout of study drug after week 12.
`Scale range: 0 to 28.
`
`ipants (45.1%) were more than minimally improved (p ⫽
`0.0004). There was a correlation between improvement in
`chorea and the CGI Global Improvement score at week 12
`(r ⫽ 0.58, p ⬍ 0.0001, n ⫽ 80). A similar correlation was
`found between improvement in total motor score and CGI
`Global Improvement score at week 12 (r ⫽ 0.41, p ⫽
`0.0001).
`Although there was a trend toward an improvement,
`the impact of TBZ on the UHDRS total motor score did not
`reach significance (adjusted mean treatment effect ⫽ ⫺3.3
`UHDRS units; 95% CI: ⫺7.0, 0.3; p ⫽ 0.08). Per the se-
`quential hierarchy of analyses prespecified in the study
`protocol, all further analyses of secondary outcomes were
`considered exploratory rather than definitive once this re-
`sult was established.
`Exploratory outcome measures. There was an adverse
`impact of TBZ on the UHDRS Functional Checklist, with
`the placebo group improving by 0.4 unit on this 25-point
`scale and the TBZ worsening by 0.8 unit (p ⫽ 0.02). There
`was no impact of TBZ on UHDRS gait assessment,
`UHDRS parkinsonism score, or any of the other explor-
`atory outcome measures (TFC, FIS, Independence Scale).
`There was a small but significant correlation between
`worsening UHDRS Functional Checklist scores and wors-
`ening UHDRS parkinsonism scores (r ⫽ 0.24; p ⫽ 0.027).
`Subjects in the TBZ group reported more sleepiness on the
`ESS (adjusted mean effect size ⫽ 1.8 ESS units; 95% CI:
`0.3, 3.4; p ⫽ 0.02). There was no significant impact of TBZ
`on the Barnes Akathisia Scale, UHDRS Behavioral Assess-
`ment (calculated as the sum of all items), UPDRS swallow-
`
`ing, or UPDRS speech items. TBZ had an adverse impact
`on Stroop word reading, but not on other UHDRS
`measures of cognitive function. Subjects in both the pla-
`cebo and the TBZ groups improved on the 17-item HAM-D
`over the course of the trial, but those in the placebo group
`did so to a slightly greater degree, with a relatively smaller
`improvement seen in the TBZ group (adjusted mean effect
`size ⫽ 1.6 HAM-D units; 95% CI: 0.6, 2.7; p ⫽ 0.003). As
`the mean HAM-D score across all subjects at baseline was
`only 4.7, the net TBZ effect was clinically insignificant in
`the context of the threshold score of ⬎12 for a diagnosis of
`depression. There was, however, a small but significant
`correlation between worsening HAM-D scores and worsen-
`ing UHDRS Functional Checklist scores (r ⫽ 0.30;
`p ⫽ 0.006).
`Washout period. There were no differences between
`TBZ and placebo at the conclusion of the washout phase
`(week 13), compared with baseline, with regard to motor,
`cognitive, behavioral, or global measures of illness sever-
`ity. Chorea in subjects on TBZ worsened more than pla-
`cebo subjects in the period following withdrawal of
`medication at week 12 (adjusted effect size ⫽ 4.4 UHDRS
`units; 95% CI: 2.8, 6.0; p ⬍ 0.0001). These results were
`confirmed by analysis of chorea ratings and CGI ratings
`made by the independent movement disorder expert on a
`subset of 23 subjects for whom videotapes at weeks 12 and
`13 were available. Video chorea worsening attributable to
`the washout from TBZ vs placebo was estimated at 4.08
`UHDRS units (ANCOVA, 95% CI: 0.55, 7.62; p ⫽ 0.03).
`Seventy-one percent (10/14) of the TBZ patients were at
`
`Figure 3. Distribution of Clinical
`Global Impression Global Improvement
`ratings at week 12 (end of active treat-
`ment phase) by treatment group. Scores
`are as follows: 1 ⫽ very much im-
`proved, 2 ⫽ much improved, 3 ⫽ mini-
`mally improved, 4 ⫽ no change, 5 ⫽
`minimally worse, 6 ⫽ much worse, 7 ⫽
`very much worse. There is a shift of the
`curve to the left (improvement) in the
`tetrabenazine group (p ⫽ 0.0001 for
`proportion achieving score of ⱕ3; ␹2
`intention to treat).
`
`February (1 of 2) 2006 NEUROLOGY 66 369
`
`Apotex Ex. 1010
`
`

`

`Table 2 Tolerability analyses
`
`Variable
`
`Subjects withdrawn (see text)
`Subjects experiencing at least one SAE (see text)
`New AEs per subject, mean ⫾ SD
`All
`Excluding mild
`Subjects reporting AEs
`All
`Excluding mild
`Subjects with week 12 reduced dosage due to intolerability
`
`TBZ ⫽ tetrabenazine; SAE ⫽ serious adverse event.
`
`least minimally worse on the CGI after withdrawal of drug
`compared with 22% (2/9) of the placebo patients (Coch-
`ran—Armitage trend test, p ⫽ 0.01). There was strong
`correlation between the independent rater’s assessment of
`chorea severity by video and investigators’ reports of cho-
`rea severity in the clinic at both week 12 (r ⫽ 0.76; p ⬍
`0.0001) and at week 13 (r ⫽ 0.68; p ⫽ 0.0004).
`Tolerability and serious adverse events. Seventy-eight
`(93%) of the 84 subjects completed the full 13 weeks of the
`study. One subject in the placebo group was lost to follow-
`up. Five withdrawals occurred in the TBZ group: four due
`to serious adverse events (SAEs), and one due to akathisia.
`Four participants receiving TBZ experienced five SAEs.
`There was one death by drowning due to suicide. Neither
`the investigator nor the subject’s caregiver had detected
`signs of depression at a study visit conducted 2 weeks prior
`to the event, and no abnormality was detected on the
`HAM-D at that time. There was a premature disclosure of
`treatment assignment to the site investigator consequent
`to this SAE. One TBZ-treated subject had an intracerebral
`hemorrhage consequent to a fall and subsequently re-
`turned to baseline level of function. One participant was
`hospitalized for restlessness that resolved within 48 hours
`of dosage reduction and treatment with clonazepam, but 2
`weeks later developed depressive symptoms, irritability,
`and suicidal ideation when motor symptoms worsened.
`The subject was continued on TBZ, rehospitalized, and
`treated with mirtazapine, with resolution of suicidal ide-
`ation within a day of rehospitalization. One subject had
`been aware of a breast lump prior to screening but did not
`bring it to the investigator’s attention until after she was
`enrolled; she was diagnosed with breast cancer during the
`study. There were no SAEs in the placebo group.
`All AEs. Table 2 summarizes the number of with-
`drawals, dosage reductions due to intolerability, subjects
`experiencing at least one AE, and number of AEs per sub-
`ject. Twenty-one (70%) of placebo and 49 (91%) of TBZ
`participants experienced an AE (World Health Organiza-
`tion preferred coding; p ⫽ 0.01). Table E-2 lists AEs re-
`ported in four or more participants (approximately 5% of
`the total study population). By the conclusion of the main-
`tenance phase, when subjects were presumably on optimal
`dosage, there were no significant differences between TBZ
`and placebo with regard to specific AEs that had not been
`reported at baseline (coding based on the AE log using
`World Health Organization preferred term). Among sub-
`
`370 NEUROLOGY 66 February (1 of 2) 2006
`
`Placebo, n (%), n ⫽ 30
`
`TBZ, n (%), n ⫽ 54
`
`p Value
`
`1 (3.3)
`0 (0)
`
`1.5 ⫾ 1.8
`0.6 ⫾ 0.9
`
`21 (70.0)
`10 (33.3)
`1 (3.3)
`
`5 (9.3)
`4 (7.4)
`
`3.8 ⫾ 3.1
`1.9 ⫾ 2.0
`
`49 (90.7)
`37 (68.5)
`24 (44.4)
`
`NS
`NS
`
`0.0005
`0.0007
`
`0.01
`0.002
`⬍0.0001
`
`jects completing the study, the most common AE at week
`12 was fatigue, reported by seven subjects on TBZ (14.3%)
`and two subjects on placebo (6.9%).
`Dosage adjustments. Figure E-1 on the Neurology Web
`site gives the distribution of dosages achieved by each
`group at the end of the titration and maintenance phases.
`Twenty-seven (55%) of the 49 TBZ subjects and 4 (14%) of
`the 29 placebo subjects completing the study were taking
`less than the maximum allowed dosage at the end of the
`maintenance phase (week 12). Of these, one TBZ and two
`placebo subjects had early washouts during the mainte-
`nance phase (unrelated to adverse events), whereas two
`TBZ and one placebo subject achieved desirable anticho-
`reic effect at less than maximal dosage. Dose-limiting
`symptoms (based on the dosage adjustment log) in the
`TBZ-treated group included sedation in 13 (27%), akathi-
`sia in 4 (8%), parkinsonism in 2 (4%), depression as de-
`scription of mood rather than a formal diagnosis in 2 (4%),
`and other in 3 (6%). One placebo-treated subject (3%) re-
`ported light-headedness as a dose-limiting symptom.
`Vital signs and laboratory results. There were no sig-
`nificant treatment differences in blood pressure or weight
`between baseline and the end of the maintenance phase.
`There was an increase in pulse in the TBZ group (5.9 ⫾ 2.7
`beats/min, adjusted mean effect ⫾ SE; p ⫽ 0.03). There
`were no clinically relevant EKG changes during the course
`of the study. All participants had laboratory values less
`than grade 3 by National Cancer Institute guidelines at
`screening, with the exception of lipid profiles. Three sub-
`jects receiving TBZ developed alanine aminotransferase
`(ALT) increases that were not associated with elevated
`bilirubin values or clinical symptoms. Of these three pa-
`tients, two were classified as grade 2 and resolved with
`continued therapy in an open extension trial. The patient
`who developed a grade 3 increase had an abnormal value
`at baseline and acknowledged binge drinking at the time of
`the increase; this patient’s ALT normalized on withdrawal
`of therapy after which he tolerated retreatment at a lower
`dose in an open extension trial. There were no other clini-
`cally meaningful differences between the groups with re-
`gard to changes in laboratory values during the course of
`the study, and the groups did not differ statistically with
`regard to the occurrence of ALT elevation.
`Medications. The most common concomitant medica-
`tions at study entry included antidepressants (60%) and
`
`Apotex Ex. 1010
`
`

`

`benzodiazepines (17%). There was no difference between
`groups in the use of these medications.
`
`Discussion. TBZ significantly reduced chorea bur-
`den, improved global outcome, and was generally
`safe and well tolerated in this 12-week study. The
`antichoreic impact of TBZ was clinically meaningful
`and statistically significant, with benefit in a global
`measure of clinical outcome (CGI Global Improve-
`ment Scale) that correlated well with the degree of
`chorea improvement. The 3.5-unit (23.5%) reduction
`in chorea severity seen in the TBZ group, compared
`with placebo, contrasts favorably with that seen in
`recently reported trials of remacemide (0.9 unit,
`9.8%)36 and riluzole (2.2 units, 18.0%).37 Amantadine
`was previously reported to improve chorea by 36%,1
`but a more recent double-blind controlled study
`showed no benefit of amantadine on chorea.38 A post-
`hoc analysis of chorea response by dosage in our
`study indicates that subjects whose TBZ dosage was
`50 mg/day or less had a greater improvement in cho-
`rea at week 12 (adjusted effect size compared with
`placebo: ⫺6.24; 95% CI: ⫺8.56, ⫺3.93) than did those
`receiving more than 50 mg/day (⫺3.65; 95% CI:
`⫺5.70, ⫺1.60). Differences in susceptibility to TBZ
`due to hepatic processing, avidity of VMAT-2, or
`other unknown factors may account for this finding.
`Whereas chorea is often treated with standard or
`atypical neuroleptics, there are no controlled clinical
`data available to judge the degree of chorea reduc-
`tion achieved with these agents. Adverse effects of
`neuroleptics as antichoreic treatment include par-
`kinsonism, decreased balance, akathisia, neuroleptic
`malignant syndrome, acute dystonic reactions, tar-
`dive dyskinesia, blunting of affect, and generalized
`apathy. Based on this study and the pharmacology of
`TBZ, the typical extrapyramidal effects seen with
`neuroleptics appear to be less common. Because our
`study was only 12 weeks long, we cannot draw infer-
`ences about adverse effects due to TBZ that may
`emerge after more prolonged exposure.
`The rapid recurrence of chorea during the wash-
`out phase is likely due to the relatively short half-life
`of the drug (approximately 4 hours), but a placebo
`effect cannot be completely excluded. A staggered
`washout study would be informative.
`An unexplained outcome of this study is the dis-
`cordant improvement of TBZ-treated patients on the
`CGI Global Improvement Scale in comparison with
`the slight worsening on the exploratory analysis of
`the UHDRS Functional Checklist. No adverse im-
`pact of TBZ on other measures of function (Indepen-
`dence Scale, FIS) was found. One explanation of this
`finding is that the UHDRS Functional Checklist
`failed to incorporate items of functional significance
`relevant to potentially beneficial effects of TBZ, such
`as the ability to carry out specific activities (e.g.,
`eating or dressing). We noted a significant correla-
`tion between deterioration on the HAM-D or on the
`UPDRS Parkinsonism Scale and worsening UHDRS
`Functional Checklist scores. The fact that improve-
`
`ment in chorea was significantly correlated to im-
`provement in the CGI Global Improvement Scale
`underscores the contribution that chorea makes to
`the overall clinical burden of HD and validates the
`use of the UHDRS chorea score as a clinical outcome
`measure.
`TBZ was associated with a significant increase in
`reports of drowsiness and insomnia, though only four
`subjects reported both symptoms. Other dose-
`limiting AEs included depressed mood (n ⫽ 2), par-
`kinsonism (n ⫽ 2), and akathisia (n ⫽ 4). These AEs
`generally resolved with dosage adjustment. There
`were no significant differences between TBZ and pla-
`cebo in the number of AEs present on optimized dos-
`ages at week 12. We detected no adverse impact at
`week 12 on the Barnes Akathisia Rating Scale or on
`measures of parkinsonism, dysphagia, or dysarthria.
`There was no evidence of TBZ-related depression as
`determined by the HAM-D,17 though patients in the
`placebo group had a greater improvement in this
`index of mood than did those in the TBZ group over
`the 12-week study.
`One subject in the TBZ group committed suicide,
`despite scoring within the normal range on the
`HAM-D 2 weeks prior to the event. Clinicians should
`be cautious about the heightened risk of suicide in
`HD regardless of depression indexes or use of mono-
`amine depleters, as overall completed suicide rates
`are estimated at 7.3% and up to 25% of patients may
`attempt suicide at some point in the illness.39,40 The
`increase in suicide in HD may be due to the high
`frequency of impulsivity seen in these patients, but
`the occurrence of the suicide by our subject cannot be
`absolutely attributed to impulsivity and may possi-
`bly be related to exposure to TBZ or to other aspects
`of the underlying di

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket