throbber
ORIGINAL ARTICLE
`
`Dextromethorphan Plus Ultra Low-Dose
`Quinidine Reduces Pseudobulbar Affect
`
`Erik P. Pioro, MD, PhD,1 Benjamin Rix Brooks, MD,2 Jeffrey Cummings, MD,3
`Randolph Schiffer, MD,1 Ronald A. Thisted, PhD,4 Daniel Wynn, MD,5
`Adrian Hepner, MD,6 and Randall Kaye, MD6 for the Safety, Tolerability, and Efficacy
`Results Trial of AVP 923 in PBA Investigators
`
`Objective: To evaluate dextromethorphan combined with ultra low dose quinidine (DMq) for treating pseudobulbar
`affect (PBA) in patients with amyotrophic lateral sclerosis (ALS) or multiple sclerosis (MS).
`Methods: In a 12 week randomized, double blind trial, ALS and MS patients with clinically significant PBA (a baseline
`score 13 on the Center for Neurologic Studies Lability Scale [CNS LS]) were maintained, twice daily, on placebo,
`DMq at 30/10mg (DMq 30), or DMq at 20/10mg (DMq 20).
`Results: In 326 randomized patients (of whom 283, or 86.8%, completed the study), the PBA episode daily rate was
`46.9% (p < 0.0001) lower for DMq 30 than for placebo and 49.0% (p < 0.0001) lower for DMq 20 than for placebo
`by longitudinal negative binomial regression, the prespecified primary analysis. Mean CNS LS scores decreased by
`8.2 points for DMq 30 and 8.2 for DMq 20, vs 5.7 for placebo (p¼ 0.0002 and p¼ 0.0113, respectively). Other
`endpoints showing statistically significant DMq benefit included, for both dosage levels, the likelihood of PBA
`remission during the final 14 days and, for the higher dosage, improvement on measures of social functioning and
`mental health. Both dosages were safe and well tolerated.
`Interpretation: DMq markedly reduced PBA frequency and severity, decreasing the condition’s detrimental impact
`on a patient’s life, with satisfactory safety and high tolerability. The findings expand the clinical evidence that DMq
`may be an important treatment for patients suffering from the socially debilitating symptoms of PBA.
`ANN NEUROL 2010;68:693 702
`
`Introduction
`
`Pseudobulbar affect (PBA) is a neurologic condition
`
`laughing
`characterized by involuntary outbursts of
`and/or crying incongruous or disproportionate to the
`patient’s emotional state.1 The condition, hypothesized to
`arise from disconnection of brainstem structures from
`cortical inhibition, is associated with underlying central
`including stroke,2 traumatic
`nervous system disorders,
`brain injury,3 Alzheimer disease,4 amyotrophic lateral scle
`rosis (ALS),5–7 and multiple sclerosis (MS).8 Prevalence
`studies have reported that it affects 11% of patients 1 year
`after a stroke,2 11% of patients during the first year after
`traumatic brain injury,9 18% of patients with Alzheimer
`disease,4 10% of patients with MS,8 and 49% of patients
`with ALS.10 In addition to the effects of the underlying
`disorder, PBA can have a severe impact on well being and
`
`social functioning and can be highly disabling, owing in
`part to the stigma attached to loss of emotional control.11
`Yet even with such a significant burden of illness, PBA
`appears to be poorly recognized and consequently is
`undertreated.11,12
`In settings of ALS or MS, dextromethorphan plus
`quinidine (DMQ) has been found to be beneficial in reduc
`ing PBA.13,14 Dextromethorphan (DM) is known to be a
`low affinity, noncompetitive antagonist of the N methyl d
`aspartate glutamate receptor,15 and also a sigma receptor
`agonist.16 To block its first pass metabolism, it was originally
`coadministered with low dose quinidine (Q), a potent cyto
`chrome P450 2D6 inhibitor,17 at DMQ dosage of 30/30mg
`in a capsule taken twice daily. Without such blockade, DM
`blood levels in some ALS patients have been undetectably
`low even following DM dosage as high as 750mg/day.17 In
`
`View this article online at wileyonlinelibrary.com. DOI: 10.1002/ana.22093
`
`Received Feb 24, 2010, and in revised form Apr 30, 2010. Accepted for publication May 20, 2010.
`
`Address correspondence to Dr Pioro, Director, Section of ALS and Related Disorders, Department of Neurology, Neurological Institute, Cleveland
`Clinic, Desk S90, 9500 Euclid Avenue, Cleveland, OH 44195. E-mail: PIOROE@ccf.org
`
`From the 1Cleveland Clinic, Cleveland, OH; 2Carolinas Medical Center, Charlotte, NC; 3David Geffen School of Medicine at University of California at Los
`Angeles, Los Angeles, CA; 4University of Chicago, Chicago IL; 5Consultants in Neurology, Northbrook, IL; and 6Avanir Pharmaceuticals, Aliso Viejo, CA.
`
`VC 2010 American Neurological Association 693
`
`Page 1 of 10
`
`Biogen Exhibit 2028
`Coalition v. Biogen
`IPR2015-01993
`
`

`

`ANNALS of Neurology
`
`atrial fibrillation and flutter, Q is utilized for conversion as
`well as reduction in frequency of relapse. However, Q doses
`are often in excess of 1,000 to 1,600mg/day and may affect
`cardiac function in ways that include prolongation of the
`QTc interval,18 which in turn may be associated with risk of
`ventricular arrhythmias.19 In the treatment of PBA, a formal
`pharmacokinetic/pharmacodynamic analysis has predicted
`that the Q dosage can be reduced to 10mg per capsule (ultra
`low dosage, q), as a treatment referred to as DMq, with
`maintained efficacy and a decreased potential for proarrhyth
`mic risk.20 The present 12 week trial was designed to evalu
`ate DMq at 30/10mg and at 20/10mg twice daily versus pla
`cebo for treating PBA in patients with ALS or MS. An
`additional objective was to determine the pharmacokinetic
`parameters of each DMq formulation in a subset of the
`study population, as will be reported separately.
`
`Patients and Methods
`Design
`This was a 12-week, randomized, double-blind, placebo-con-
`trolled, 3-arm, parallel-group study conducted at 60 centers in the
`United States and South America between December 2007 and
`March 2009. Patients completed screening procedures 1 to 4
`weeks before their baseline visit. At the screening visit, those meet-
`ing all inclusion/exclusion criteria (see below) were randomized
`(1:1:1) to receive placebo, DM 30mg þ Q 10mg (DMq-30), or
`DM 20mg þ Q 10mg (DMq-20). For the first treatment week,
`patients took a single capsule of study drug in the morning. Dur-
`ing weeks 2 through 12, they took study drug once in the morn-
`ing and once in the evening. Follow-up visits occurred at 2, 4, 8,
`and 12 weeks. In addition, for 1 week prior to baseline and
`throughout the trial, patients were required to maintain a diary
`recording the daily number of laughing and/or crying episodes
`experienced, the medications they took, and any adverse experien-
`ces. Patients completing the study were eligible to continue treat-
`ment in a 12-week open-label phase with DMq-30 twice daily.
`The study protocol was approved by local institutional
`review boards or independent ethics committees and was con-
`ducted in accordance with Good Clinical Practice Consolidated
`Guidance, as approved by the International Conference on Har-
`monization (1997), and also with local or national
`laws and
`regulations. Prior to entry, study procedures and risks were
`explained to each subject, and written informed consent was
`obtained. The study’s randomization code (blocked by center
`and by underlying neurological disorder) was computer-gener-
`ated, and study drug was supplied in blister packs of identical-
`looking capsules. The sponsor, all patients, and all investigators
`were blind to treatment identification and allocation.
`
`Patients
`For entry, men or women 18 to 80 years old were required to
`have clinically significant PBA, with a score 13 on the Center
`for Neurologic Study Lability Scale (CNS-LS),21 and a diagno-
`sis either of ALS (by El Escorial criteria22) within the past 30
`
`FIGURE 1: Subjects’ disposition. DMq 30 5 dextromethorphan
`combined with ultra low dose quinidine at 30/10mg; DMq 20 5
`DMq at 20/10mg.
`
`months or of MS or probable MS (by McDonald criteria23).
`Patients were excluded for any evidence of clinically significant
`abnormality on screening electrocardiogram, a family history of
`congenital QT-interval prolongation syndrome, a resting respi-
`ratory rate outside the range of 12 to 20/min, or a resting diur-
`nal oxygen saturation <95%. Patients were also excluded for
`any presence or history of major psychiatric disturbance, includ-
`ing current symptoms of a depressive disorder (or a score >19
`on the Beck Depression Inventory II [BDI-II]24); major sys-
`temic disease or organ dysfunction capable of interfering with
`study assessments or putting the patient at risk; and exacerba-
`tion of the patient’s underlying ALS or MS within the previous
`2 months. Women with childbearing potential were required to
`use a medically acceptable form of birth control; pregnant or
`lactating women were excluded.
`
`Efficacy Assessments
`The primary efficacy outcome was a patient’s change from base-
`line in the number of PBA episodes (laughing and/or crying)
`per day, as recorded in the patient’s diary. Diary data also
`yielded, as secondary outcomes, a responder analysis (the pro-
`portion of patients with an improvement from baseline PBA
`rate, assessed across all degrees of improvement); number of
`episode-free days; and occurrence of
`remission from PBA
`(defined by absence of episodes during the study’s final 14
`days). Additional secondary outcomes were a patient’s change
`from baseline on CNS-LS, which was administered at baseline
`and at each follow-up visit, and on BDI-II, the Neuropsychiat-
`ric Inventory (NPI),25 and the Medical Outcomes Study 36-
`Item Short-Form Health Survey Version 1.0 (SF-36),26 which
`were administered at baseline and at 12 weeks.
`The CNS-LS is a 7-item self-assessment of PBA severity,
`validated for measuring PBA in ALS21 and MS.27 Total scores
`range from 7 to 35; a score 13 is the instrument’s range for
`clinical PBA. The BDI-II is a 21-item self-assessment of symp-
`toms of depression. A total score of 14 19 is considered mild,
`20 28 is moderate, and 29 63 is severe. The NPI is a question-
`naire
`covering 12 neuropsychiatric
`symptom domains;
`it
`
`694
`
`Volume 68, No. 5
`
`Page 2 of 10
`
`

`

`Pioro et aI: Pseudobulbar Affect
`
`TABLE 1: Patients’ Baseline Characteristis (ITT Population)
`
`Characteristic
`
`No.
`
`Age, mean yr (SD)
`
`Females. No. (%)
`
`Ethnic origin, No. (%)
`White
`
`Hispanic
`
`Black
`
`Other
`
`Diagnosis, No. (%)
`
`A15
`
`MS
`
`Time since ALS diagnosis, mean mon (SD)
`
`PBA episodeslday, mean (SD)
`
`CNS LS smre, mean (SD)
`
`BDI H score, mean (SD)
`
`NPI score, mean (SD)
`
`Frequency
`
`Severity
`
`SF 36 score, mean (SD)
`
`Mental Summary
`
`Physical Summary
`
`DMq-30
`
`DMq—20
`
`Placebo
`
`110
`
`53.1 (11.0)
`
`64 (58.2)
`
`80 (72.7)
`
`21 (19.1)
`
`6 (5.5)
`
`3 (2.7)
`
`65 (59.1)
`
`45 (40.9)
`
`22.7 (29.8)
`
`4.7 (9.5)
`
`1.7 (3.4)
`
`3.0 (6.7)
`
`19.8 (4.9)
`
`9.4 (6.1)
`
`6.2 (6.3)
`
`5.8 (3.9)
`
`107
`
`50.8 (11.1)
`
`54 (50.5)
`
`80 (74.8)
`
`21 (19.6)
`
`2 (1.9)
`
`4 (3.7)
`
`68 ((33.6)
`
`39 (36.4)
`
`16.3 (22.9)
`
`6.8 (12.9)
`
`4.1 (11.8)
`
`2.8 (4.2)
`
`21.0 (5.0)
`
`10.9 (5.8)
`
`7.8 (6.7)
`
`7.0 (4.5)
`
`109
`
`50.3 (11.9)
`
`59 (54.1)
`
`83 (76.1)
`
`21 (19.3)
`
`4 (3.7)
`
`1 (0.9)
`
`64 (58.7)
`
`45 (41.3)
`
`13.4 (18.0)
`
`4.5 (7.6)
`
`2.5 (7.4)
`
`2.0 (20)
`
`19.9 (4.7)
`
`10.5 (5.4)
`
`7.0 (6.7)
`
`6.3 (4.5)
`
`44.0 (10.9)
`
`40.1 (10.1)
`
`44.6 (11.2)
`
`37.0 (10.4)
`
`44.9 (10.6)
`
`38.5 (9.8)
`
`1'IT = intent-to-treat; DMq—30 = dexttomethorphan combined with ultra low-dose quinidine at 30/101115 DMq-20 = DMq at
`20I10mg: SD = standard deviation; ALS = amyotrophic lateral sclerosis; MS = multiple scletosk; PBA = pseudobulbar affect;
`CNS-I.S = Center for Neurologic Study Lability Scale; BDI-II = Beck Depression Inventory Second Edition; NP1 = Neuro-
`psychiatric Inventory; SF-36 = Medical Outcomes Study 36—Itun Short-Form Health Survey.
`
`provides a brief, inlbrmant-based assesment of neutopsychiatric
`symptoms and caregiver distres. The SF-36 "B a 36-item
`health-status assessment, with subdomains for Physical Func-
`tioning, Role-Physical, Bodily Pain, General Hmlth, Vitality,
`Social Ftmctioning, Role-Emotional, and Mental Health. Each
`of 2 summary scores (Mental Component and Physical Com-
`ponent) is standardized so that 50 represents the US general
`population norm (for 1998).
`
`Safety/Tolerability Assessments
`At all visits, vital
`were measured, 12-lead electrocardiography
`was performed, and reports of adverse events (AE5) were obtained
`Serious AEs were defined as final,
`life-threatening, significantly
`disabling, or requiring hospitalimtion. Resting diurnal oxygn sat-
`
`uration and nocturnal oxygen saturation were measured (with
`pulse oximetry) at screening and at 2 weelcs.
`diumal oxy-
`gensaturarionwasalsomeasuredat12weJs.Clini<allaboratory
`testingwasperfi1rmedatsa‘eeningandat4and12weelts.
`
`Statistical Analyses
`In the intent-to-treat population, comprising all randomized
`patients, change from baseline in
`epkode rate
`was analymd using longitudinal negative binomial regression,”
`with adjustment for baseline rate, diagnosis, and study site. As
`a sensitivity analysis, change in epkode rate was also assessed by
`a nonlongitudinal negative binomial model. In addition, 12-
`week change in episode rate was analyzed using the Wilamon
`rank sum test. For number of episode-free days, a 2-sample t
`
`November, 2010
`
`Page 3 of 10
`
`695
`
`

`

`ANNALS of Neurology
`
`FIGURE 2: Twelve week time course of pseudobulbar affect
`weekly episode rate and Center for Neurologic Study
`Lability Scale (CNS LS) score (intent to treat population).
`Weekly rates (top chart) are shown as change from baseline
`at each visit in mean daily rates 3 7. CNS LS scores (bottom
`chart) are the means at each visit. DMq 30 5 dextromethor
`phan combined with ultra low dose quinidine at 30/10mg;
`DMq 20 5 DMq at 20/10mg.
`
`test was used. Changes on CNS-LS, SF-36, BDI-II, and NPI
`were analyzed with analysis of covariance, using the method of
`Frison and Pocock.29 Baseline value, study site, and diagnosis
`were covariates. Observed cases were used in the sensitivity
`analyses, with no imputation for missing data. All analyses were
`2-sided hypothesis tests at the 0.05 significance level.
`The safety population comprised all patients who took at
`least 1 dose of study medication. Their AE rates, for types reported
`by 5% of patients in any treatment group, were compared among
`groups, and mean change in resting nocturnal oxygen saturation
`(from baseline to day 15) was assessed by 2-sample t test.
`
`Sample Size Calculation
`Based on PBA episode rates in previous studies of DMq for
`PBA in ALS13 and in MS,14 a sample size of approximately 90
`
`patients (60 with ALS and 30 with MS) per treatment group
`was planned. This size was expected to be sufficient to detect a
`36% reduction in mean episode rate for DMq-30 vs placebo
`with at least 90% power. The study was not powered to test a
`difference between DMq-30 and DMq-20.
`
`Results
`Subjects
`In all, 332 patients were screened, and among them 326
`were randomized, 110 to DMq 30, 107 to DMq 20, and
`109 to placebo (Fig 1). The main reasons for screening
`failure were unwillingness to discontinue disallowed medi
`cations, CNS LS score <13, and BDI II score >19. In all,
`283 patients (86.8% of 326) completed the study, includ
`ing 101 (91.8% of 110)
`in the DMq 30 group, 88
`(82.2% of 107) in the DMq 20 group, and 94 (86.2% of
`109) in the placebo group. Demographically and in base
`line PBA characteristics, the treatment groups were well
`matched (Table 1), except for a higher baseline PBA epi
`sode rate in the DMq 20 group than in the other groups,
`and a longer time since ALS diagnosis in the DMq 30
`group. At entry, no patient had clinical depression.
`
`Efficacy
`Over the course of the study, all 3 groups showed sub
`stantial reduction in daily PBA episode rates relative to
`baseline. However, the reduction in daily PBA episode
`rate was significantly greater in each of the DMq groups
`than in the placebo group. By longitudinal negative bino
`mial model (predefined primary efficacy analysis), the
`
`FIGURE 3: Responder analysis by treatment group (intent
`to treat population). Each curve graphs the proportion of
`patients improved with a given degree of improvement
`from baseline pseudobulbar affect (PBA) rate at endpoint.
`DMq 30 5 dextromethorphan combined with ultra low dose
`quinidine at 30/10mg; DMq 20 5 DMq at 20/10mg.
`
`696
`
`Volume 68, No. 5
`
`Page 4 of 10
`
`

`

`TABLE 2: Twelve-Week Mean Change on CNS-LS, NPI, and BDI-ll
`
`Pioro et aI: Pseudobulbar Affect
`
`DMq—30
`
`DMq—20
`
`Phcebo
`
`Endpoint
`
`CNS LS
`
`No.
`
`Mean change (p vs placebo)
`BDI II
`
`No.
`
`103
`
`-8.2 (0.0002)
`
`103
`
`96
`
`-8.2 (0.01 13)
`
`97
`
`-1.0 (0.2707)
`
`79
`
`-2.6 (0.0938)
`
`54
`
`— 1.6 (0.207)
`
`101
`
`-5.7
`
`101
`
`0.02
`
`66
`
`-1.3
`
`48
`
`— 1.0
`
`Mean change (p vs placebo)
`
`-1.6 (0.0368)
`
`NP1 (fiequency)
`No.
`
`74
`
`Mean change (p vs placebo)
`
`-1.6 (0.6558)
`
`NP1 (severity)
`No.
`
`46
`
`Mean change (p vs placebo)
`
`-0.7 (0.510)
`
`psychianic Inventory; BDI-11 = Beck Depression Invumory
`CNS-IS = Center fin Ncurologic Study-Iabiliry Sale; NPI = Ncuro
`Second Edition; DMq-30 = dextromethorphan combined with ultra low-dose quinidine at 30/10mg; DMq-20 = DMq at
`20/10mg.
`
`treatment effect in each DMq group over that seen in
`the placebo group was an incremental reduction in PBA
`episode rate of 46.9% (p < 0.0001) for DMq 30 com
`pared to placebo and 49.0% (p < 0.0001) for DMq 20
`compared to placebo. By nonlongitudinal negative bino
`rnial model with constant dispersion (predefined efficacy
`sensitivity analysis), the additional improvement over pla
`
`cebo at both dosage levels was also statistically significant
`(p < 0.0001 and p= 0.0370, respectively). The 12 week
`mean change in
`episode rate was -4.1 for DMq 30
`and -3.9 for DMq 20, vs -3.0 for placebo (p= 0.0099
`and p= 0.0048, respectively). Weekly rates (daily rates X
`7) showed signifiarnt decrease at all time points assessed,
`beginning with day 15 (Fig 2, top).
`the 12 week mean
`Among secondary outcomes,
`reduction from baseline CNS LS score was significantly
`greater at both DMq dosage levels than for placebo
`(Table 2), and for DMq 30, the man reduction was sig
`nificant at all time points assessed, beginning with day
`15 (see Fig 2, bottom). Among secondary outcomes
`derived from
`data, the proportion of patients with
`
`an improvement from their baseline PBA rate was higher
`for both DMq 30 and DMq 20 than for placebo, across
`all degrees of improvement (Fig 3). The proportion of
`patients’ episode free days was significantly greater for
`DMq 30 than fin placebo at all time points assessed, and
`for DMq 20 vs placebo at all time points except day 15
`
`(Fig 4, left). lastly, the proportion of patients reporting
`remission of PBA was significantly greater at both DMq
`dosage levels than for placebo (see Fig 4, right).
`
`November, 2010
`
`Page 5 of 10
`
`On BDI II, mean improvement was significantly
`greater for DMq 30 than for placebo (see Table 2). On
`NPI, total scores showed no significant change for either
`dosage vs placebo (see Table 2). On SF 36, improvement
`was significant for DMq 30 vs placebo on the Mental
`Summary score and on its subdomains for social func
`
`tioning and mental health (Table 3).
`
`Safety and Tolerability
`The proportion of patients reporting at least 1 AE was
`similar in all treatment groups, at 82.7% of DMq 30
`recipimts, 79.4% of DMq 20 recipients, and 82.6% of
`
`100
`
`90
`__ ao
`5 7.
`E 60

`'5
`so
`-
`so
`30
`20
`10
`O
`
`100
`
`so
`.. so
`i 7.
`3 60
`0
`*5
`so
`E
`.
`so
`30
`20
`10
`D
`
`
`
`Visit 5
`Vlsvt 4
`V51! 3
`VISII 2
`(DQY 35)
`(Dav-'17)
`(DEV 29)
`(Dav 15)
`-DMq-3D(N=H0) IDMQ-20(N=107) C]P|aoebo(N=10m
`
`"""'”'°"
`
`Flnal 14 Days
`'p<0D5vsD|uct-be
`“ptooosvspioocho
`
`FIGURE 4: Decrease of pseudobulbar affect. as assessed by
`freedom from episodes and by remission (intent to treat
`population). Freedom from episodes (left) was defined as
`the percentage of episodefree days since the preceding
`visit. Remission (right) was defined by absence of episodes
`throughout the study's final 14 days. DMq 30 = dextromethor
`phan corrbined wi1h ultra Iowdose qiriciie at 30/1Gng;
`DMq 20 = DMq at 20/1 Gng.
`
`697
`
`

`

`ANNALS ofNeurology
`
`TABLE 3: Twelve-Week Mean Changes on SF-36 (I'lT Population)
`
`SF-36 Domain,
`
`Mean Change
`(p vs placebo)
`
`Mental Summary
`
`Vitality
`
`Social Functioning
`Role Emotional
`
`Mental Health
`
`Physiarl Summary
`
`Physical Functioning
`
`Role Physical
`
`Bodily Pain
`
`General Health
`
`DMq-30
`(n = 110)
`
`4.5 (00193)
`
`-0.9 (0.2972)
`
`9.3 (0.0033)
`
`11.6 (03658)
`
`5.5 (00028)
`
`-0.8 (05877)
`
`-0.9 (0.2972)
`
`3.5 (0.3063)
`
`4.1 (0.0740)
`
`-1.5 (0.8703)
`
`DMq-20
`(n = 107)
`
`1.8 (0.6792)
`
`-5.3 (0.75l0)
`
`1.4 (05544)
`
`-1.8 (0.6838)
`
`3.1 (0.4457)
`
`-1.0 (0.9967)
`
`-5.3 (0.7Sl0)
`
`-4.3 (0.2292)
`
`5.8 (0.0678)
`
`-3.0 (0.3583)
`
`Placebo
`
`(n = 109)
`
`0.3
`
`-4.1
`
`-3.1
`
`2.4
`
`-0.3
`
`-1.3
`
`-4.1
`
`-1.8
`
`-1.1
`
`-1.3
`
`SF-36 = Medical Outcomes Study 36-10:-.m Short-Form Halrh Survey; I'IT = intent-to-treat; DMq-30 = dcxtromethorphan
`combined with ultra low—dose quinidine at 30/10mg; DMq-20 = DMq at 20/ 10mg.
`
`placebo recipients. Overall, AE incidence was distributed
`evenly throughout
`the study, except for slightly higher
`rates in the DMq 30 and placebo groups during the ini
`tial treatment week. The proportion of patients reporting
`serious AES was also similar across groups, at 7.3% (8
`patients) in the DMq 30 group, 8.4% (9 patients) in the
`DMq 20 group, and 9.2% (10 patients) in the placebo
`group. Two serious AEs, both in the DMq 20 group,
`were reported as possibly treatment related. In 1 of these
`patients, the event was reported as respiratory depression
`and ALS progresion. The other patient had Iorsening
`musde spastrcrty. Seven deaths were reported, a
`In ALS
`patients: 3 In the DMq 30 group, 3 m the DMq 20
`group, and 1 in the placebo group. All deaths were classi
`fied by an independent mortality adjudication committee as
`having a respiratory cause likely to be the result of progres
`sion of the underlying neurologic disease. No acute decom
`pensation of respiratory function after iniliation of study
`drugwasobserved,andnodeathswereascribedtoacardiac
`muse. Discontinuations due to AEs were more frequent in
`11.: DMq 20 group, at 9.3% (10 patients), tlnn an the
`DMq 30 group, at 5.5% (6 patients), or the placebo group,
`at 1-8% (2 1miems>- Among freqtrndy reported AEs
`(r‘*“‘’ 4)’ ‘‘‘”"‘= “W """"‘°"~ ""1 “""“'>’ ‘W
`infection were more frequent for DMq 30 than for placebo,
`whereas falls, headadre, somnolence, fatigue, and other AES
`occurred at rates resembling those for placebo.
`resting
`Vital signs, physical examination findings,
`diurnal oxygen saturation, and clinical laboratory values
`showed no significant changes from their baseline means
`in any treatment group. For resting nocturnal oxygen sat
`uration, Table 5 compares baseline and day 15 findings.
`
`698
`
`Page 6 of 10
`
`At day 15, the mean change was -0.2 percentage points
`in the DMq 30 group and -0.7 percentage points in
`the DMq 20 group, vs -0.1 for placebo (p= 0.794 and
`
`TABLE 4: Adverse Events Reported by 25% of
`Any Group (Safety Population)‘
`
`five.“ Type’
`0' 0%)
`Fall
`Dizziness
`Headache
`
`Nausea
`Diarrhea
`Sornnolence
`
`Dlulrlailo DNEFIZO plafblo
`(n _
`) (n _ 07) (n _ 09)
`22 (20_())
`14 (13_1)
`22 (203)
`20 (182)
`11 (103) 6 (55)
`15 (13.6)
`15 (14.0)
`17 (15-6)
`
`14 (127)
`11 (10.0)
`11 (10.0)
`
`10 (92)
`8 (75)
`14 (13.1) 7 (6.4)
`9 (8.4)
`10 (9.2)
`
`Fatigue
`
`9 (8.2)
`
`11 (10.3)
`
`10 (9.2)
`
`Nasopharyngitis 9 (8.2)
`
`6 (5.6)
`
`8 (7.3)
`
`8 (7-3)
`
`Urinary tract
`inf°°ti°"
`7 (6-4)
`Constipation
`7 (6.4)
`Muscle spasms
`Muscle weakness 6 (5.5)
`_
`5 (45)
`Dysplugla
`Pain in extremity 5 (4.5)
`
`4 (3-7)
`
`3 (2-3)
`
`7 (6-5)
`4 (3.7)
`5 (4.7)
`
`6 6'6)
`2 (1.9)
`
`9 (8-3)
`10 (9.2)
`4 (3.7)
`
`4 6'7)
`8 (7.3)
`
`6 (5.5)
`1 (0.9)
`0
`Depression
`=3’. M¢.«1DRA Prcfinad term, lkred by fiequency in the
`DMq—30 group.
`DMq-30 = dcxuomcthorphan combined widr ultra low-
`dme quinidine at 30/10mg; DMq—20 = DMq at 20Il0rng.
`
`Volume 68, No. 5
`
`

`

`Pioro et aI: Pseudobulbar Affect
`
`TABLE 5: Summary of Nocturnal Oxygen-Saturation Data (Safety Population)
`
`Data
`
`DMq—30
`(n = 108 or 106’),
`n = 110
`
`DMq-20
`(n = 102 or 100’),
`n = 107
`
`Placebo
`(n = 108 or 109‘),
`n = 109
`
`94.1 (5.4)
`
`94.4 (2.1)
`
`94.9 (2.0)
`
`94.1 (2.5)
`
`-0.2 (2.0) (0.794)
`
`-0.7 (2.0) (0.039)
`
`94.6 (2.2)
`
`94.4 (2.2)
`
`-0.1 (2.1)
`
`Saturation, mean % (SD)
`
`At baseline
`
`At day 15
`
`Mean change
`(p vs placebo)
`Number of events
`<88%. mean (SD)
`
`At baseline
`
`At day 15
`Total time in minutes
`<88%, mean (SD)
`
`At baseline
`
`At day 15
`
`11.2 (39.2)
`
`4.1 (19.7)
`
`9.9 (39.1)
`
`11.0 (41.7)
`
`9.1 (23.1)
`
`11.2 (20.9)
`
`‘For saturation and desaturation analyses, respectively.
`DMq—30 = dextromethorphan combined with ultra low-dose quinidine at 30/10 mg; DMq-20 = DMq at 20/10 mg; SD =
`standard deviation.
`
`p = 0.039, respectively). The differences between groups
`were not clinically signifimnt. Descriptive analyses of desa
`
`izcd in Table 6. At all time points assessed, no DMq recipi
`ent had a QTC interval absolute value >480 milliseconds
`
`turation data identified no substantial differences between
`groups (see Table 5). QTc interval changes are summar
`
`(with Fridericia correction) or a change from baseline >60
`milliseconds.
`
`TABLE 6: Summary of O.Tc-Interval Data (Safety Population)
`
`Data
`
`DMq-30 (n = 110)
`
`DMq-20 (n = 107)
`
`Placebo (n = 109)
`
`QTcB/QTcF at baseline, mean ms
`
`418.2/406.6
`
`QTcB/QTcF at day 84, mean ms
`
`420.6/411.8
`
`QTcB/QTcF change from baseline,
`H1631! HIS
`
`3.0/4.8
`
`Proportion of postbaseline ECGs with
`absolute QTCB/QTCF
`
`>450 ms
`
`>480 ms
`
`>500 ms
`
`Proportion of postbaseline ECGs with
`change from baseline QTCB/QTCF
`30 60 ms
`
`>60 ms
`
`>90 ms
`
`6.3%I1.9%
`
`0.2%/0.0%
`
`0.0%/0.0%
`
`7.0%/7.2%
`
`0.5%/0.0%
`
`0.0%I0.0%
`
`416.4/404.2
`
`413.8/405.1
`
`-1.9/1.0
`
`4.9%/ 1.2%
`
`0.0%/0.0%
`
`0.0%/0.0%
`
`3.9%/2.9%
`
`0.2%/0.0%
`
`0.0%/0.0%
`
`416.1/404.7
`
`416.8/405.8
`
`1.6/1.0
`
`6.1%/2.4%
`
`0.9%/0.0%
`
`0.2%/0.0%
`
`6.6%/3.5%
`
`0.5%/0.5%
`
`0.0%/0.0%
`
`DMq-30 = datromerhorphan combined with ultra low-dose quinidine at 30/10mg; DMq-20 = DMq at 20/10mg; QTcB = QT
`interval corrected for heart rate (Bamett’s formula); QTcF = QT interval corrected for heart rate (Fridericia’s formula); ECG =
`electrocardiogram.
`
`November, 2010
`
`Page 7 of 10
`
`699
`
`

`

`ANNALS of Neurology
`
`Discussion
`In this
`large, double blind, placebo controlled study,
`both dosage levels of DMq were significantly superior to
`placebo for
`reducing PBA episode frequency among
`patients with underlying ALS or MS, as assessed by lon
`gitudinal and nonlongitudinal statistical models and also
`by mean change in daily PBA episode rate. At both dos
`age levels, DMq also significantly reduced the severity of
`PBA, as represented by CNS LS score. For reduction in
`episode rate, a prespecified responder analysis showed, at
`both dosage levels, a substantial difference from placebo
`across all degrees of improvement, despite a strong pla
`cebo effect (resembling those seen in previous studies13).
`The differences between DMq and placebo included, at
`both dosage levels, a significantly higher likelihood of
`PBA remission on DMq than on placebo, suggesting that
`for large proportions of patients, the active treatment’s
`amelioration of PBA may be marked.
`Numerically, the responses to the higher DMq dos
`age were more robust than those to the lower dosage in
`several ways, including an earlier improvement in CNS
`LS score, an earlier time to significant difference vs pla
`cebo in number of episode free days, and a slightly
`greater 12 week mean change vs placebo in PBA daily
`episode rate. At the higher dosage, DMq was also associ
`ated with significant
`improvement
`in mental health
`measures, by BDI II and SF 36. Because none of the
`subjects
`in this
`study was clinically depressed, and
`because PBA can result in substantial reduction of quality
`of life,11 this improvement may have been in well being.
`Specific improvements on SF 36 subdomains for social
`functioning and mental health are further evidence that
`the social and psychological disability associated with
`PBA may have been reduced. However, the possibility
`that DMq may have direct antidepressant properties can
`not be excluded, and would require further study. Over
`all, the efficacy reported for DMq containing Q at ultra
`low dosage
`10mg per capsule
`resembled the benefits
`reported by measures including CNS LS scores and PBA
`episode counts in studies of DMq in its original formula
`tion, in which the Q content per capsule was 30mg.13,14
`In the present study, both dosage levels were safe.
`In particular, cardiovascular safety was satisfactory, with
`mild QTc prolongation and no proarrhythmic events.
`Respiratory findings appeared to be consistent with ALS
`progression. However, physicians should always exercise
`caution in managing a patient population that has com
`promised respiratory function. Both dosage levels were
`also well tolerated, with only 13% of DMq recipients
`discontinuing during 12 weeks of double blind treatment.
`The overall discontinuation rate was lower for DMq 30, at
`8%, than for DMq 20, at 18%. In studies of DMQ as
`
`originally formulated, with DM at 30mg per capsule and
`Q at 30mg, the 12 week discontinuation rate had been
`much higher, at 28% in ALS patients13 and 25% in MS
`patients.14 The implication is that the improved tolerability
`demonstrated in the present study may reflect ultra low
`dosing of Q. Usage of dose escalation (with once daily
`dosing during week 1) may also have contributed.
`A body of published evidence suggests that PBA
`may be ameliorated pharmacologically,1 but
`the trials
`assessing current agents, all of which are being utilized
`off label, have limitations. In 1979, dopaminergic treat
`ment, specifically L dopa, was reported to be effective for
`incontinence,’’30 but in a follow up uncon
`‘‘emotional
`trolled study of L dopa or amantadine, only 10 of 25
`recipients responded.31 Since then, reports have centered
`on antidepressants, notably tricyclic agents (eg, amitripty
`line32 or nortriptyline33) and selective serotonin reuptake
`inhibitors (eg, fluoxetine,34 citalopram,35,36 paroxetine,36
`or sertraline37). Overall, the trials have been hampered
`by small size (12 to 28 subjects, among those referenced
`above) and by methodological problems, such as their
`definitions of PBA improvement. Substantial placebo
`effects, as demonstrated in the present
`study, make
`uncontrolled findings all the more difficult to interpret.
`In brief, well controlled data to support current options
`are scarce, and no option is currently approved by the
`US Food and Drug Administration. In addition, antide
`pressants are associated with incompletely elucidated AE
`profiles (including QT interval prolongation19).
`In interpreting the present study’s findings, the trial’s
`limitations should be taken into account. Because its subjects
`were carefully selected, the findings should be generalized to
`a broader
`spectrum of PBA with caution. The study
`required, for instance, a baseline CNS LS score of at least
`13. Accordingly, the effects of DMq 30 or DMq 20 on
`milder forms of PBA are unknown. In addition, the study
`enrolled only patients with underlying ALS or MS. Because
`the pathophysiologic mechanisms causing PBA are probably
`similar regardless of the underlying CNS pathology, DMq
`will likely be effective in reducing symptoms of PBA arising
`in various brain disease or injury states, much in the same
`way that antispastic medication reduces spasticity, irrespective
`of the underlying condition. Even so, additional studies of
`the effect of DMq on PBA in various neurological disorders
`could provide enhanced safety, efficacy, and health outcome
`insight. Hence, further clinical studies of PBA are warranted.
`Nevertheless, the present study represents the largest
`and longest double blind, randomized, placebo controlled
`trial of DMq conducted to date in PBA, and also the
`first to test DMq in PBA patients at ultra low Q dosage.
`Its
`findings expand the clinical evidence that with
`
`700
`
`Volume 68, No. 5
`
`Page 8 of 10
`
`

`

`satisfactory safety and high tolerability, DMq markedly
`reduces PBA frequency and severity, decreasing the con
`dition’s detrimental impact on a patient’s life.
`
`Acknowledgment
`This study was supported by Avanir Pharmaceuticals.
`
`Potential Conflicts of Interest
`E.P.P. has received research support and compensation for
`consulting from Avanir Pharmaceuticals. B.R.B. has
`received compensation for consulting from Avanir Phar
`maceuticals, Bayer Healthcare Pharmaceuticals, Biogen
`Idec, Genentech, and Teva Neuroscience, and has received
`research support
`from Avanir Pharmaceuticals, Biogen
`Idec, National Institutes of Neurological Disorders and
`Stroke Clinical Research Consortium, Novartis, and Teva
`Neuroscience. J.C. has received compensation for consult
`ing from Abbott, Acadia, Accera, ADAMAS, Astellas,
`Avanir Pharmaceuticals, Bristol Myers Squibb, CoMentis,
`Eisai, Elan, EnVivo, Forest, GlaxoSmithKline, Janssen,
`Lilly, Lundbeck, Medivation, Merck, Merz, Myriad,
`Neuren, Novartis, Pfizer, Prana, Schering Plough, Sonexa,
`Takeda, Toyama, and Wyeth, and owns the copyright of
`the Neuropsychiatric Inventory. A.H. and R.K. are
`employees of and own stock options in Avanir Pharma
`ceuticals. R.S. has received compensation for consulting
`from Teva and Avanir Pharmaceuticals and owns 300
`shares of common stock in Johnson & Johnson Corpora
`tion.
`R.A.T.
`is a statistical consultant
`to Avanir
`Pharmaceutical

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