11
Original article Efficacy and safety of pramipexole in idiopathic restless legs syndrome: A polysomnographic dose-finding study—The PRELUDE study * Markku Partinen a,e, * ,1 , Kari Hirvonen a,b,2 , Leni Jama a,2 , Anniina Alakuijala a,c,2 , Christer Hublin d,e,2 , Ilkka Tamminen f,2 , Juergen Koester g,2 , Juergen Reess h,2 a Skogby Sleep Clinic, Rinnekoti Research Centre, Kumputie 3, FI-02980 Espoo, Finland b Neurotest Tampere Oy, Hatanpa ¨a ¨n Valtatie 1, FIN-33100 Tampere, Finland c Department of Clinical Neurophysiology, University Hospital of Helsinki, P.O. Box 340, FI-00029 Helsinki, Finland d Finnish Institute of Occupational Health, Brain at Work Research Center, Topeliuksenkatu 41 a A, FI-00250 Helsinki, Finland e Department of Neurology, University of Helsinki, FI-00014 Helsinki, Finland f Medical Division, Boehringer Ingelheim Finland Ky, Tammasaarenkatu 5, FI-00180 Helsinki, Finland g Medical Division, Boehringer Ingelheim International GmbH, Binger Str. 173, 55216 Ingelheim, Germany h Department of Clinical Research CNS, Boehringer Ingelheim Pharma GmbH and Co. KG, Birkendorferstr.65, 88397 Biberach, Germany Received 22 November 2005; received in revised form 16 February 2006; accepted 6 March 2006 Abstract Background and purpose: To evaluate the effects of pramipexole (0.125–0.75 mg/d) on polysomnographic (PSG) measures and patient and clinician ratings of restless legs syndrome (RLS). Patients and methods: Patients (nZ109) with moderate to severe RLS were randomized to placebo or fixed doses of pramipexole during a 3-week, double-blind, placebo-controlled, dose-finding study. Results: In each pramipexole dose group, the periodic limb movements during time in bed index (PLMI) decreased significantly, compared with placebo (adjusted mean difference in log-transformed data: 0.125 mg, K1.54; 0.25 mg, K1.93; 0.50 mg, K1.89; and 0.75 mg, K1.52; P!0.0001). At all doses, International RLS Study Group Rating Scale (IRLS) scores were also significantly reduced, with the greatest adjusted mean reduction in the 0.50 mg group (K17.01). At all but the lowest pramipexole dose, the percentage of responders (R50% reduction of IRLS score) was substantially higher than for placebo (61.9–77.3, vs 33.3%). In the pramipexole groups, 50.0–77.3% of patients rated their condition as ‘much better’ or ‘very much better’, compared with 38.1% of patients in the placebo group (PZ0.0139 for the 0.50 mg dose). Clinical global impressions (CGI) scale ratings of ‘much improved’ or ‘very much improved’ were given to 61.9–86.4% of patients in the pramipexole groups, compared with 42.9% in the placebo group (P!0.05 for the 0.25, 0.50, and 0.75 mg groups). Pramipexole was well tolerated and did not produce somnolence at any dose. Conclusion: Pramipexole is effective and safe in the treatment of both objective and subjective facets of RLS. q 2006 Elsevier B.V. All rights reserved. Keywords: Restless legs syndrome; RLS; Periodic limb movements; Sleep; Polysomnography; Pramipexole; Placebo-controlled Sleep Medicine 7 (2006) 407–417 www.elsevier.com/locate/sleep 1389-9457/$ - see front matter q 2006 Elsevier B.V. All rights reserved. doi:10.1016/j.sleep.2006.03.011 * PRELUDE, Pramipexole for RLS: efficacy and tolerability of the usage of dopamine agonists. * Corresponding author. Address: Skogby Sleep Clinic, Rinnekoti Research Centre, Kumputie 3, FI-02980 Espoo, Finland. Tel.:C358 9 8551345; fax: C358 9 8551375. E-mail address: markku.partinen@rinnekoti.fi (M. Partinen). 1 Dr Partinen has received a grant from the Rinnekoti Research Foundation for his studies on restless legs syndrome (RLS), and he has received honoraria, totaling less than USD10,000 per year, for his presentations in postgraduate symposia on RLS. 2 The author has no conflict of interest related to this article.

Efficacy and safety of pramipexole in idiopathic restless legs syndrome: A polysomnographic dose-finding study—The PRELUDE study

Embed Size (px)

Citation preview

Original article

Efficacy and safety of pramipexole in idiopathic restless legs syndrome:

A polysomnographic dose-finding study—The PRELUDE study*

Markku Partinen a,e,*,1, Kari Hirvonen a,b,2, Leni Jama a,2, Anniina Alakuijala a,c,2,

Christer Hublin d,e,2, Ilkka Tamminen f,2, Juergen Koester g,2, Juergen Reess h,2

a Skogby Sleep Clinic, Rinnekoti Research Centre, Kumputie 3, FI-02980 Espoo, Finlandb Neurotest Tampere Oy, Hatanpaan Valtatie 1, FIN-33100 Tampere, Finland

c Department of Clinical Neurophysiology, University Hospital of Helsinki, P.O. Box 340, FI-00029 Helsinki, Finlandd Finnish Institute of Occupational Health, Brain at Work Research Center, Topeliuksenkatu 41 a A, FI-00250 Helsinki, Finland

e Department of Neurology, University of Helsinki, FI-00014 Helsinki, Finlandf Medical Division, Boehringer Ingelheim Finland Ky, Tammasaarenkatu 5, FI-00180 Helsinki, Finland

g Medical Division, Boehringer Ingelheim International GmbH, Binger Str. 173, 55216 Ingelheim, Germanyh Department of Clinical Research CNS, Boehringer Ingelheim Pharma GmbH and Co. KG, Birkendorferstr.65, 88397 Biberach, Germany

Received 22 November 2005; received in revised form 16 February 2006; accepted 6 March 2006

Abstract

Background and purpose: To evaluate the effects of pramipexole (0.125–0.75 mg/d) on polysomnographic (PSG) measures and patient and

clinician ratings of restless legs syndrome (RLS).

Patients and methods: Patients (nZ109) with moderate to severe RLS were randomized to placebo or fixed doses of pramipexole during a

3-week, double-blind, placebo-controlled, dose-finding study.

Results: In each pramipexole dose group, the periodic limb movements during time in bed index (PLMI) decreased significantly, compared

with placebo (adjusted mean difference in log-transformed data: 0.125 mg,K1.54; 0.25 mg,K1.93; 0.50 mg,K1.89; and 0.75 mg,K1.52;

P!0.0001). At all doses, International RLS Study Group Rating Scale (IRLS) scores were also significantly reduced, with the greatest

adjusted mean reduction in the 0.50 mg group (K17.01). At all but the lowest pramipexole dose, the percentage of responders (R50%

reduction of IRLS score) was substantially higher than for placebo (61.9–77.3, vs 33.3%). In the pramipexole groups, 50.0–77.3% of patients

rated their condition as ‘much better’ or ‘very much better’, compared with 38.1% of patients in the placebo group (PZ0.0139 for the

0.50 mg dose). Clinical global impressions (CGI) scale ratings of ‘much improved’ or ‘very much improved’ were given to 61.9–86.4% of

patients in the pramipexole groups, compared with 42.9% in the placebo group (P!0.05 for the 0.25, 0.50, and 0.75 mg groups).

Pramipexole was well tolerated and did not produce somnolence at any dose.

Conclusion: Pramipexole is effective and safe in the treatment of both objective and subjective facets of RLS.

q 2006 Elsevier B.V. All rights reserved.

Keywords: Restless legs syndrome; RLS; Periodic limb movements; Sleep; Polysomnography; Pramipexole; Placebo-controlled

Sleep Medicine 7 (2006) 407–417

www.elsevier.com/locate/sleep

1389-9457/$ - see front matter q 2006 Elsevier B.V. All rights reserved.

doi:10.1016/j.sleep.2006.03.011

* PRELUDE, Pramipexole for RLS: efficacy and tolerability of the usage of dopamine agonists.* Corresponding author. Address: Skogby Sleep Clinic, Rinnekoti Research Centre, Kumputie 3, FI-02980 Espoo, Finland. Tel.:C358 9 8551345; fax:

C358 9 8551375.

E-mail address: [email protected] (M. Partinen).1 Dr Partinen has received a grant from the Rinnekoti Research Foundation for his studies on restless legs syndrome (RLS), and he has received honoraria,

totaling less than USD10,000 per year, for his presentations in postgraduate symposia on RLS.2 The author has no conflict of interest related to this article.

M. Partinen et al. / Sleep Medicine 7 (2006) 407–417408

1. Introduction

Restless legs syndrome (RLS) is a sensorimotor disorder

affecting approximately 5–10% of the population [1–3]. Its

prevalence increases with advancing age and is higher in

women than in men [2,4,5]. Other risk factors have also

been identified, including positive family history, preg-

nancy, iron deficiency, and renal disease [1,6–8].

The primary clinical manifestations are a strong urge to

move the legs, and also abnormal sensations in the legs,

which become worse at night or during periods of inactivity

and are relieved by movement [9]. Periodic limb move-

ments during sleep (PLMS) occur in 70–80% of cases and

are defined as repetitive movements, usually extension of

the big toe and/or flexion of the ankle, knee and hip, of

which patients are often unaware [10,11]. Not surprisingly,

these night time symptoms often cause disruption of sleep,

insomnia, and sleepiness during the day, and it is often

sleep-related complaints that bring patients to the attention

of a physician [12]. RLS is often missed or misdiagnosed,

and its symptoms can be disabling [11,12]. Fortunately,

when properly diagnosed and managed, RLS is typically

very responsive to dopaminergic treatment [13].

The pathophysiology of RLS is not well understood, but

ongoing research suggests that dysregulation of dopamine

function plays a role [10,14]. Consistent with this

hypothesis, medications that enhance dopamine function

are considered to be appropriate treatments for the disorder.

For patients with intermittent symptoms, the dopamine

precursor levodopa may be utilized, although its pharma-

cokinetic and pharmacodynamic characteristics (e.g. short

half-life and augmentation) limit its use in patients with

regular symptoms [13]. In patients with daily symptoms,

dopamine agonists have been suggested as the treatment of

choice [13] and, in fact, are considered first-line treatment in

the guidelines established by the American Academy of

Sleep Medicine (AASM) [15]. Other, nondopaminergic

medications are also used clinically, but they carry risks of

abuse, dependence, or daytime somnolence.

Pramipexole, a second-generation, non-ergot dopamine

agonist with selectivity for the D2/D3 receptor [16], has

been marketed for several years for treatment of all stages of

Parkinson’s disease (PD). Meanwhile, some small studies

have shown that pramipexole can reduce the symptoms of

RLS. In a double-blind, placebo-controlled, 10-week,

crossover trial (4 weeks of treatment, 2 weeks of washout,

and 4 weeks of alternate treatment), Montplaisir and

colleagues [17] examined the effects of pramipexole

(0.375–1.5 mg/d salt) in 10 patients with RLS. Pramipexole

produced an 84% reduction in subjective leg restlessness

relative to placebo, compared with a 39% reduction for

levodopa in a historical control group. PLMS was reduced

by 98%, and limb movements during wakefulness were

reduced by more than 80%. The investigators followed

seven patients for up to 8 months and found persistent

benefit [18]. In addition, two open-label studies have shown

that pramipexole reduces RLS symptoms, with effects of a

similar magnitude to those observed in the Montplaisir

study [19–21]. A recently published open-label trial by

Stiasny-Kolster and Oertel [22] reports that low doses of

pramipexole reduced polysomnographic (PSG) measures of

RLS symptomatology, even in patients whose symptoms

were inadequately controlled by levodopa.

The present trial was designed to replicate and extend

earlier findings by exploring the effects of different doses of

pramipexole in a larger group of patients after short-term

treatment.

2. Methods

2.1. Patients

The study population consisted of 28 male and 79 female

patients, aged 27–76 years, who were experiencing mainly

moderate to severe idiopathic RLS, according to criteria of

the International Restless Legs Syndrome Study Group

(IRLSSG), with scores of at least 15 on the International

RLS Study Group Rating Scale (IRLS) [9,23]. All

participants were required to have PLMS at least five

times per hour, as documented by baseline polysomno-

graphy, and also weekly RLS symptoms that had disrupted

sleep within the previous 3 months. Females of childbearing

potential and males were required to use adequate contra-

ception and females who were pregnant or breast-feeding

were excluded. Potential participants were also excluded for

medical contraindications to use of pramipexole, for

medical conditions or prescriptions that might influence

disease course (including but not limited to diabetes

mellitus, anemia, renal or hepatic disease), and for comorbid

conditions that may cause or complicate symptoms of RLS.

Patients who were currently (within the previous week)

receiving treatment for RLS were not enrolled, nor were

those who had been in an investigational drug study within

the previous 60 days. All patients provided written informed

consent prior to participating, and the study was approved

by the Ethics Committee of the Helsinki and Uusimaa

Hospital District.

2.2. Study design

The study was a 3week, double-blind, placebo-con-

trolled, parallel-group, fixed-dose trial designed to evaluate

the dose effects of pramipexole salt (0.125, 0.25, 0.50, and

0.75 mg/d, where 0.125 mg salt is equivalent to 0.088 mg

base) on objective and subjective ratings of RLS sympto-

matology. After completing baseline assessments, patients

were randomly assigned to 1 of 4 dose levels of pramipexole

or to placebo in a 1:1:1:1:1 ratio. All participants

randomized to active drug were started on 0.125 mg/d and

titrated up to their assigned dose in 4-day intervals. They

stayed on their assigned dose until the end of week 3. Doses

M. Partinen et al. / Sleep Medicine 7 (2006) 407–417 409

were taken once daily 2–3 h before bedtime. The primary

endpoint was change from baseline in the periodic limb

movements during time in bed index (PLMI). Secondary

assessments included additional PSG measures (described

in the following paragraph), along with changes from

baseline in subjective ratings on the IRLS and on clinician-

rated (clinical global impressions (CGI)) and patient-rated

(patient global impression (PGI)) scales. Another secondary

objective was to assess the effect of pramipexole on quality

of sleep and daytime well-being, as evaluated by self-

reported ratings of sleep quality, daytime somnolence, and

quality of life (QOL).

2.3. PSG measures

As an objective measure of periodic limb movements

during time in bed (asleep or awake) PLMI was assessed

by polysomnography, according to AASM guidelines, at

days 0 and 21. The procedure was performed in single

sound- and light-isolated bedrooms. Recordings were

started at the time of ‘lights off.’ After 8 h, the patients

were awakened (if asleep), and the recording was

stopped (‘lights on’). Several PSG measures were

evaluated as secondary efficacy endpoints, namely the

periodic limb movements during sleep index (PLMSI),

the periodic limb movements during wakefulness index

(PLMWI), the periodic limb movements during sleep

with arousal index (PLMAI), the total number of periodic

limb movements (PLM), the total number of PLMS, the

total number of periodic limb movements during sleep

with arousal (PLMSA), the total number of awakening-

s/arousals during sleep, and also sleep latency (SL), sleep

efficiency (SE), total sleeping time (TST), percentage of

delta sleep, and percentage of stage rapid eye movement

(sREM) sleep.

2.4. Subjective assessments

2.4.1. RLS Symptoms

By interview, patients completed the IRLS, formulated

by the IRLSSG [23], at baseline and at week 3. The

instrument is a 10-item self-reported assessment of the

severity of RLS symptoms over the preceding week in

5 degrees (e.g. ‘none’ to ‘very severe’); the maximum score

is 40. Change from baseline to the end of treatment was

taken as a measure of efficacy.

2.4.2. Sleepiness and sleep quality

The Epworth sleepiness scale (ESS) [24] is a self-

reported measure of daytime sleepiness. At baseline and at

the end of the 3 weeks of treatment, this instrument was used

to assess the likelihood of dozing off or falling asleep in

eight different situations, using a 4-point verbal rating scale

(‘no chance’ to ‘high chance’); items were summed to yield

a total score (range 0–24). Patients were asked to respond to

items with respect to the preceding week. Participants were

also asked to estimate their sleep quality, using the

subjective sleep quality scale (SSQ), (during the previous

night), on a scale of 1–10 in which 1 signifies ‘very poor

sleep quality’ and 10 ‘excellent sleep quality,’ and their

tiredness after waking up in the morning, on a scale from 1

to 10 in which 1 represents ‘very tired’ and 10 denotes ‘fully

awake’.

2.4.3. Quality of life

Health-related quality of life (QOL) was assessed by the

Short Form 36 Health Survey questionnaire (SF-36) [25] as

change from baseline to week 3. The SF-36 consists of four

physical domains (i.e. physical functioning, role physical,

bodily pain, and general health) and four nonphysical

domains (i.e. vitality, social functioning, role emotional,

and mental health index). A global evaluation of health is

also included. Each dimension is transformed to a scale

ranging from 0 to 100, with higher scores indicating better

health. The number of response alternatives per item varies

from 2 to 6. The SF-36 is designed to assess QOL during the

day and does not include parameters assessing well-being

during the night.

2.4.4. PGI and CGI scales

Patient and clinician ratings of improvement were

obtained by PGI and CGI [26]. In seven categories, these

instruments assess a patient’s overall condition at a given

timepoint in relation to baseline on a scale ranging from

‘very much better/improved’ to ‘very much worse’.

2.5. Adverse events

Adverse events (AEs) were assessed by non-probing

questions at each study visit and also by examination of

clinical test results (vital signs, electrocardiogram, blood

count and/or chemistry, and urinalysis) at regular intervals.

Events were recorded whether or not they were believed to

be causally related to the study drug. A serious adverse

event (SAE) was defined as any AE that resulted in death,

was immediately life-threatening, resulted in persistent or

significant disability/incapacity, required or prolonged

patient hospitalization, was a congenital anomaly/birth

defect, or was deemed serious for any other reason

representing a significant hazard. Sudden onset of sleep

(SOOS) during daily activities was considered to be a

significant AE.

2.6. Data analysis

The intent-to-treat principle was used to include as many

patients as possible in the analyses, according to protocol.

The analyses therefore encompassed all randomized

patients with at least baseline data and post-treatment

polysomnography. Continuous or discrete parameters were

analyzed by descriptive statistics or by frequency tables,

respectively.

M. Partinen et al. / Sleep Medicine 7 (2006) 407–417410

The primary analysis was planned and performed using

an analysis of covariance (ANCOVA) model to compare the

efficacy of different doses of pramipexole for reducing the

PLMI after 3 weeks of randomized treatment. For

application of ANCOVA, the distribution of PLMI indices

required log transformation. Therefore, the final PSG

assessment was taken as the response parameter (instead

of change from baseline), and the log-transformed baseline

PLMI score was used as the covariate. Hierarchical testing

was utilized, sequentially comparing placebo with the

highest to lowest pramipexole dose. All tests were

conducted as 2-tailed at the 5% level of significance.

Nonparametric analyses (Wilcoxon–Mann–Whitney tests)

of the original data were performed as well. The hierarchical

testing allows inferences to be made on all primary endpoint

comparisons, while preserving an overall type I error

probability of 0.05.

Every effort was made to collect PSG and other data at

each visit. However, PSG and other data were collected

prematurely for patients wishing to drop out of double-blind

treatment. Randomly missing data (due, for instance, to a

mechanical failure) were estimated either by linear

interpolation of adjacent data or, if no subsequent data

were available, by a last observation carried forward

(LOCF) approach. For patients discontinuing due to

unexpected worsening of RLS, the missing data were

imputed by the least favorable data prior to discontinuation.

For patients who missed a visit due to other reasons, the

missing data were estimated by LOCF. The evaluability of

patients with protocol deviations likely to confound

treatment response was either predefined by the protocol

or decided before unblinding. For secondary endpoints,

there were no multiplicity adjustments.

All subjects who received at least one dose of study

medication were included in the analysis of safety

(NZ109). The occurrence of AEs was analyzed both by

randomized dose group (to permit a comparison of the

Fig. 1. Patient d

different pramipexole target doses) and by ‘treatment at

onset’ (to elucidate the AE pattern at the different

pramipexole dose levels).

3. Results

3.1. Patients and treatment

One hundred and forty-one patients were recruited for

the study. Among them, 32 patients were screening failures

and were not entered, leaving 109 patients who were

randomized to active treatment or placebo (Fig. 1). Two of

them withdrew prematurely: one patient in the placebo

group was lost to follow-up, without providing a second

PSG assessment, and one patient in the 0.125 mg group was

withdrawn due to an AE. For inclusion in the intent-to-treat

population, patients had to provide two PSG assessments (at

baseline and at the end of 3 weeks of treatment). One patient

from the 0.75 mg group was assessed 48 h after discontinu-

ation of treatment.

All 107 patients included in the intent-to-treat population

were Caucasian, and most (73.8%) were female. The gender

distribution was similar among treatment groups, although

the 0.75 mg group had fewer females (61.9%) and the

placebo and 0.50 mg groups had more females (81.0 and

81.8%, respectively) than the overall study population did.

Patients were a mean age of 56.2 years (standard deviation

(SD)Z10.9), except that the 0.125 mg group was slightly

but not significantly older (60.0 years, SDZ10.1). Patient

groups did not differ with respect to body mass index or

proportion previously treated (vs de novo), except in the

0.50 mg group, where 59.1% of participants had received

prior treatment (vs 41.1% overall). Table 1 summarizes

demographic and disease-related characteristics of the study

participants.

isposition.

Table 1

Demographic and disease-related characteristics in the intent-to-treat population

PBO N (%) PPX 0.125 mg N

(%)

PPX 0.25 mg N

(%)

PPX 0.50 mg N

(%)

PPX 0.75 mg

N (%)

Total N (%)

Number of patients 21 21 22 22 21 107

Gender

Male 4 (19.0) 6 (28.6) 6 (27.3) 4 (18.2) 8 (38.1) 28 (26.2)

Age (Mean [SD] years) 53.3 [11.1] 60.0 [10.1] 54.8 [10.9] 58.4 [9.5] 54.5 [12.2] 56.2 [10.9]

RLS duration (Mean [SD] years) 2.7 [10.1] 5.1 [11.1] 6.1 [10.6] 5.3 [10.6] 4.5 [10.3] 4.8 [10.4]

Previously treated for RLS 7 (33.3) 7 (33.3) 8 (36.4) 13 (59.1) 9 (42.9) 44 (41.1)

IRLS score (Mean, [SD]) 22.9 [4.2] 22.4 [4.7] 23.0 [3.4] 23.6 [3.7] 21.7 [4.6] 22.7 [4.1]

PLMI (median) 51.75 63.10 36.85 35.90 40.30

PLMS (median) 42.85 22.30 29.40 24.75 29.68 28.60

Total sleeping time (median, h) 6.4 5.7 6.8 6.1 6.6 6.3

Sleep efficiency (median, %) 84.55 84.00 89.65 82.55 88.20 85.10

PBO, placebo; PPX, pramipexole; SD, standard deviation; IRLS, International RLS Study Group Rating Scale; PLMI, periodic limb movements during time in

bed index; PLMS, periodic limb movements during sleep.

M. Partinen et al. / Sleep Medicine 7 (2006) 407–417 411

3.2. Objective assessments

Primary and secondary objective measures of efficacy

showed pramipexole to be superior to placebo. In particular,

the ANCOVA analysis of log-transformed PLMI showed

that the adjusted means at the end of week 3 were

significantly smaller in all active-dose groups than in the

placebo group (P!0.0001). In adjusted mean differences

from placebo, the largest effects (log-transformed data)

were in the 0.25 and 0.50 mg groups (Table 2). Nonpara-

metric analyses of PLMI changes from baseline (original

data) confirmed the ANCOVA results with P!0.001 for all

comparisons vs placebo. Pramipexole dose-independently

reduced PLMI at endpoint; the median change in each

treatment group was K3.00 for placebo, K52.70 for

Table 2

Median changes from baseline in PLM parameters

Parameter PBO PPX

0.125 mg

P

0

Number of patients 20 21

PLM during time in

bed index (PLMI)K3.00 K52.70b

PLM during sleep

index (PLMSI)K3.45 K20.90b

PLM during

wakefulness

index (PLMWI)

K11.00 K41.20a

PLM during sleep

with arousal

index (PLMAI)

K1.85 5.20

Total number of

awakenings/arousalsK22.50 8.00

Total number of PLM K24.00 K404.000b KTotal number of PLM

during sleepK19.50 K126.00b K

Total number of

PLM during sleep

with arousal

K9.00 K30.00

PLM, periodic limb movements; PBO, placebo; PPX, pramipexole.a P for difference from placebo !0.05 by Wilcoxon–Mann–Whitney test.b P for difference from placebo !0.01 by Wilcoxon–Mann–Whitney test.c P for difference from placebo !0.001 by Wilcoxon–Mann–Whitney test.

0.125 mg, K31.05 for 0.25 mg, K26.55 for 0.50 mg, and

K30.00 for 0.75 mg.

Data distributions for all other PSG parameters were

checked for normality. Since, all parameters except ‘SL’

and ‘percentage of time spent in sREM sleep’ showed a non-

normal distribution (and hence failed to meet the

assumption of normality required for a linear model such

as ANCOVA), natural logarithmic transformations were

used. In consequence, data were lost for a small number of

patients where baseline or on-treatment measurements were

zero. For all ANCOVA analyses, age was included as a

covariate. However, age was not found to have a significant

influence as a covariate for any of the parameters.

Pramipexole was significantly superior to placebo for

values of PLMI, PLMSI, PLMWI, total PLM, and total

PX

.25 mg

PPX

0.50 mg

PPX

0.75 mg

22 22 21

K31.05c K26.55c K30.00c

K26.65c K22.45b K17.00b

K36.50a K38.45a K21.30

K2.35 K3.25 K2.90

4.00a K4.00 1.00

244.00c K212.50b K180.00b

147.00c K135.50b K111.00c

K16.00 K21.50 K18.00

Table 3

Median changes from baseline in sleep parameters assessed by polysomnography

Parameter PBO PPX 0.125 mg PPX 0.25 mg PPX 0.50 mg PPX 0.75 mg

Number of patients 20 21 22 22 21

Sleep latency (min) K2.00 K9.50a K5.00 K11.75b K7.00a

Sleep efficiency (%) 5.85 3.60 2.95 8.30 5.30

Total sleeping time (min) 25.50 56.50 25.75 66.75a 34.50

Time spent in stages

2-4 and sREM (min)

26.75 50.00 40.00 68.00a 37.00

Percentage of time spent in

sREM sleep (%)

K1.45 K1.30 K2.15 K6.30 K4.20

Percentage of delta sleep (%) 1.30 K4.00 K4.45a K2.40 K6.10a

Amount of time spent in delta

sleep (min)

8.64 K1.81 K18.96 1.70 K17.20a

PBO, placebo; PPX, pramipexole; sREM, stage rapid eye movement.a P for difference from placebo !0.05 by Wilcoxon–Mann–Whitney test.b P for difference from placebo !0.01 by Wilcoxon–Mann–Whitney test.

M. Partinen et al. / Sleep Medicine 7 (2006) 407–417412

number of PLM. There was no statistical difference between

pramipexole and placebo for PLMAI, total number of PLM

during sleep with arousal, and total number of awakening-

s/arousals, with the exception of the 0.25 mg dose (P!0.05)

(Table 2).

Table 3 shows that pramipexole led to a significant

reduction in SL for pramipexole vs placebo at the 0.125,

0.50, and 0.75 mg doses. However, there was no statistical

difference found for SE and TST (except for the 0.50 mg

dose group). Interestingly, patients in the pramipexole

dose groups showed a reduced amount of time spent

in delta sleep, as well as a reduced percentage of delta

sleep.

Fig. 2. 3-Week change in International RLS S

3.3. Subjective assessments

3.3.1. RLS symptoms

For the IRLS total score, the assumption of normality

required for a linear model (ANCOVA) was satisfied.

Pramipexole significantly reduced the total score at 3 weeks.

The adjusted mean (with SE) change from baseline was

K6.08 (1.51) for placebo, K11.87 (1.51) for 0.125 mg,

K15.18 (1.47) for 0.25 mg,K17.01 (1.48) for 0.50 mg, and

K15.86 (1.52) for 0.75 mg. Adjusted mean changes from

baseline were statistically significant for all active doses

when compared with placebo. Mean values for baseline and

week 3 for each dose level are displayed in Fig. 2.

tudy Group Rating Scale (IRLS) score.

M. Partinen et al. / Sleep Medicine 7 (2006) 407–417 413

Data for IRLS ratings were also evaluated as proportion

of responders. In all but the 0.125 mg group, the proportion

of patients who experienced at least a 50% reduction in RLS

severity was significantly greater than in the placebo group.

Using categorical data-analysis techniques, pair-wise com-

parisons were performed for the number of responders in

each active-treatment group vs placebo. Using the Fisher’s

exact test method, statistically significant results were

obtained in the 0.25 mg group (PZ0.023), the 0.50 mg

group (PZ0.004), and the 0.75 mg group (PZ0.006). The

percentage of responders in each of the active-treatment

groups was 61.9% (0.125 mg group), 68.2% (0.25 mg

group), 77.3% (0.50 mg group), and 76.2% (0.75 mg

group). Thus, the higher-dose groups of 0.25 mg, 0.50 mg,

and 0.75 mg showed a larger clinical response than the

0.125 mg group.

3.3.2. Ratings of improvement and response to treatment

Responder rates in PGI (combined categories ‘much

better’ and ‘very much better’) were higher in the

pramipexole groups than in the placebo group. Using the

Fisher’s exact test method, statistically significant results

were obtained for the 0.50 mg group (PZ0.039) and the

0.75 mg group (PZ0.041). At week 3, the proportion of

patients who rated their current condition as ‘very much

better’ was 27.2% in the 0.50 mg group and 23.8% in the

0.75 mg group, compared with 4.8% in the placebo group.

Fifty percent of patients in the 0.50 mg group and 33.3% of

patients in the 0.75 mg group were classified as ‘much

better,’ compared with 33.3% in the placebo group. No

Fig. 3. Clinical global impressions scale (CG

patients in any treatment group assessed their condition as

‘much worse’ or ‘very much worse’.

Generally, the clinicians’ assessment of patients’ improve-

ment was similar to the patients’ own assessment. For CGI

ratings of global improvement (Fig. 3), more than 60% of

patients in all pramipexole dose groups were rated by the

investigator as being ‘much improved’ or ‘very much

improved’ after 3 weeks of therapy, compared with 42.9%

of patients in the placebo group. A comparison of the lowest

active dose (0.125 mg) with placebo was not statistically

significant (PO0.31); however, the comparison for the highest

active doses (0.25, 0.50, and 0.75 mg) did reach statistical

significance (PZ0.022, 0.001, and 0.008, respectively).

Similar results were obtained for CGI ratings of severity of

illness. At baseline, the majority of patients were considered

either ‘markedly ill’ (38.3%) or ‘moderately ill’ (34.6%). At

week 3, in stark contrast, a substantial proportion (overall,

30.8%) of patients were evaluated as ‘not at all ill,’ including

9.5% of the placebo group and 19.0% (0.125 mg), 36.4%

(0.25 mg), 54.5% (0.50 mg), and 33.3% (0.75 mg) of the

pramipexole groups. Fig. 4 shows the percentage of patients in

each treatment group who exhibited improvement in severity

ratings at the end of week 3.

Moreover, pramipexole increased the percentage of

patients rated by clinicians as showing a therapeutic effect.

For 47.6% of patients in the placebo group, a ‘marked’ or

‘moderate’ therapeutic effect was seen, compared with

66.7–90.5% of patients in the pramipexole groups. A

comparison of the 0.125 mg group with placebo was not

statistically significant (PZ0.162), but for the higher-dose

groups (0.25, 0.50, and 0.75 mg), the comparisons with

I): global improvement after 3 weeks.

Fig. 4. Clinical global impressions scale (CGI): severity of illness after 3 weeks.

M. Partinen et al. / Sleep Medicine 7 (2006) 407–417414

placebo were statistically significant (PZ0.0127, 0.002, and

0.004, respectively). Pramipexole did not affect the number

of clinician-rated side effects reported at the end of week 3.

3.3.3. Sleep quality and sleepiness

Evaluated by ESS, pramipexole likewise showed no

effect on daytime sleepiness. All groups had mean baseline

ESS scores in the range of healthy persons and, after 3

weeks, all groups showed minimal change, with no

significant difference between placebo and any active

treatment (Table 4).

At baseline, subjective sleep quality (SSQ; median over 1

week) ranged from6.4 to 7.0 among the various groups. By the

end of week 3, it had improved in all groups, the improvement

being substantial in the higher-dose groups (1.01, 1.44, and

1.36 for the 0.25, 0.50, and 0.75 mg groups, respectively).

For placebo and for the 0.125 mg group, only small increases

(0.54 and 0.59, respectively) were observed.

For subjective tiredness after awakening, a baseline of

6.5–7.3 (median over 1 week) improved in all treatment

groups, with larger median increases for the 0.25 and

Table 4

Median [range] changes from baseline in Epworth Sleepiness Scale (ESS)

Parameter PBO PPX 0.125 mg

Number of patients 20 21

ESS median change, range K1.0 [K6, 3] 0.0 [K4, 3]

PBO, placebo; PPX, pramipexole.

0.75 mg groups (0.82 and 0.70, respectively) than for

placebo (0.50) (Table 5).

3.3.4. Quality of life

At baseline, the mean overall index for social

functioning (87.38G17.55) was comparable to a healthy

Finnish population (81.20G19.30) [27]. Changes in

health-related QOL, as measured by the generic SF-36

instrument, were not significantly different in the

pramipexole and placebo groups for 7 subscales, with

the exception of a positive effect of pramipexole on the

social functioning subscale. While the adjusted mean SF-

36 social functioning subscore decreased in the placebo

group, it increased in the pramipexole groups. Pair-wise

comparisons of pramipexole doses vs placebo showed

significant adjusted mean differences for the 0.125 mg

(PZ0.041), 0.50 mg (PZ0.031), and 0.75 mg groups

(PZ0.008).

3.3.5. Safety and tolerability

The overall incidence of AEs was similar between

placebo and pramipexole total (77.3 vs. 74.7%). Among

PPX 0.25 mg PPX 0.50 mg PPX 0.75 mg

22 22 21

0.0 [K5, 4] 0.0 [K4, 3] 1.0 [K13, 2]

Table 5

Subjective sleep quality assessment

PBO

(NZ21)

PPX 0.125 mg

(NZ21)

PPX 0.25 mg

(NZ22)

PPX 0.50 mg

(NZ22)

PPX 0.75 mg

(NZ21)

Total

(NZ107)

Sleep quality during the previous night

Baseline (visits 1–2)

median

7.00 6.43 6.62 6.50 6.71 6.67

Week 3 (visits 2–4,

maintenance) median

7.38 7.30 7.59 7.58 7.20 7.44

Change from baseline

median

0.54 0.59 1.01 1.44 1.36 0.77

Tiredness after waking up in the morning

Baseline (visits 1–2)

median

6.50 7.29 6.93 7.20 6.50 6.86

Week 3 (visits 2–4,

Maintenance) median

7.00 7.50 7.80 7.41 7.20 7.40

Change from baseline

median

0.50 0.22 0.82 0.18 0.70 0.50

PBO, placebo; PPX, pramipexole. For the evaluation of baseline, the last 7 days prior to randomization were used.

M. Partinen et al. / Sleep Medicine 7 (2006) 407–417 415

the different doses of pramipexole, the highest frequency

of AEs was reported for 0.125 mg (44.8%, compared

with 33.3% for 0.25 mg, 31.8% for 0.50 mg, and 27.3%

for 0.75 mg). Since all patients had started their

treatment at 0.125 mg/d, this pattern reflects a titration

of pramipexole during which, in general, the increasing

dosage led neither to new AEs nor to a worsening of

existing ones.

The most frequently reported AEs (O5%) more often

reported in the combined pramipexole groups than in the

placebo group were nausea (14.9 vs. 4.5%) and nasophar-

yngitis (6.9 vs. 0.0%). Conversely, fatigue (22.7 vs. 18.4%)

and headache (31.8 vs. 19.5%) were reported more

frequently in the placebo group than in the combined

pramipexole groups (Table 6).

Overall, 45.0% of patients reported AEs related to study

drug, as assessed by an investigator. In the placebo group,

the proportion was 50.0% of patients, while in the

Table 6

Adverse events occurring in O5% of the population

MedDRA system organ

class/preferred term

PBO DB PPX PPX

N (%) N (%) N

Total treated 22 100.0 87 100.0 87

Total with any adverse event 17 77.3 65 74.7 39

Nervous system disorders 8 36.4 30 34.5 14

Headache 7 31.8 17 19.5 7

Gastrointestinal disorders 2 9.1 21 24.1 11

Nausea 1 4.5 13 14.9 7

General disorders and

administration-site conditions

6 27.3 17 19.5 14

Fatigue 5 22.7 16 18.4 14

Infections and infestations 3 13.6 15 17.2 3

Nasopharyngitis 0 0.0 6 6.9 2

Musculoskeletal and connective

tissue disorders

2 9.1 4 4.6 2

Psychiatric disorders 3 13.6 3 3.4 2

PBO, placebo; DB, double-blind; PPX, pramipexole.

pramipexole groups it ranged between 31.8% (0.75 mg)

and 61.9% (0.125 mg). The most frequent study-drug-

related AEs were fatigue (overall, 16.5%), nausea (overall,

12.8%), and headache (overall, 5.5%). All other drug-

related AEs occurred in less than 5.0% of all patients.

The majority of AEs had a maximum intensity of mild or

moderate; only three patients (3.4%) reported AEs with

severe intensity. Significant AEs that led to study

discontinuation or to a decrease in study drug dose were

reported for two patients receiving pramipexole and for no

patient in the placebo group. No patient in any group

experienced an SAE.

Aggravation of RLSwas observed in four patients (3.7%):

one at 0.125 mg, two at 0.25 mg, and one at 0.50 mg.

Somnolence was reported by three patients (2.8%), all at the

0.125 mg level.

No significant change in any laboratory parameter or

vital sign (blood pressure, pulse rate, or weight) was

0.125 mg PPX 0.25 mg PPX 0.50 mg PPX 0.75 mg

(%) N (%) N (%) N (%)

100.0 66 100.0 44 100.0 22 100.0

44.8 22 33.3 14 31.8 6 27.3

16.1 10 15.2 6 13.6 2 9.1

8.0 4 6.1 5 11.4 2 9.1

12.6 5 7.6 4 9.1 1 4.5

8.0 3 4.5 2 4.5 1 4.5

16.1 2 3.0 2 4.5 0 0.0

16.1 1 1.5 1 2.3 0 0.0

3.4 8 12.1 2 4.5 2 9.1

2.3 2 3.0 1 2.3 1 4.5

2.3 1 1.5 0 0.0 1 4.5

2.3 1 1.5 0 0.0 0 0.0

M. Partinen et al. / Sleep Medicine 7 (2006) 407–417416

observed in any pramipexole group, compared with the

placebo group. Only one patient reported symptomatic

orthostatic hypotension during the study. At screening, this

patient had exhibited asymptomatic orthostatic hypotension

(systolic change R20 mmHg in combination with a

diastolic change R10 mmHg from supine to standing).

4. Discussion

Overall, pramipexole was highly effective and safe

for key RLS parameters. In all active-treatment groups,

the objective measure of PLMI was significantly reduced

(P!0.001); the largest reduction was observed at a dosage

of 0.125 mg/d. This marked improvement in the primary

endpoint demonstrated that pramipexole rapidly controlled

RLS symptoms. Statistical improvement from baseline was

shown for pramipexole vs placebo for the total IRLS score

(P%0.001), the CGI global assessment (P!0.05), and the

CGI ‘severity of illness’ assessment (P!0.05) across all

dosing groups.

Secondary endpoints analyzed by polysomnography

evaluated statistical significant changes from baseline for

PLMSI, PLMWI, and total number of PLM with the

treatment of pramipexole. No statistical difference was

found for SE and TST. This is in line with findings

from PSG studies with other dopamine agonists [12,28].

Pramipexole numerically decreased the PLM with arousal

compared with placebo, but the difference did not reach

statistical difference. The total number of arousals

decreased with placebo and increased slightly in the

pramipexole groups. However, this is in contrast to the

improvements seen in the subjective sleep quality assess-

ment. The median change from baseline for ‘sleep quality

during the previous night’ and ‘tiredness after waking up in

the morning’, as assessed using the SSQ, favored

pramipexole over placebo for these endpoints.

A disconcordance of ‘objective sleep findings’ and the

subjective assessments was also reported with other

dopaminergic drugs [12,28]. There is no conclusive

explanation for this finding at the present time.

The safety and tolerability of pramipexole over 3 weeks

was comparable to placebo and comparable to that reported

for other agents in this class [28–30]. The incidence of new

AEs or aggravation of existing ones did not increase with

increasing doses of pramipexole. The rapid up-titration to

effective dosing was well tolerated.

Previous studies have shown that RLS has an impact on

QOL comparable to that of many other chronic diseases

[31,32]. In the present study, statistical improvement over

placebo in ‘social functioning’ was shown for pramipexole

using the SF-36 health questionnaire. The fact that the

SF-36 does not account for nighttime symptoms from RLS

bypasses an important aspect of this disorder.

In summary, the findings of the present trial support

previous, smaller-scale studies in showing that

pramipexole produced improvement in both objective

and subjective measures of RLS symptomatology at all

doses tested and was generally well tolerated. The effects

of pramipexole were robust and were apparent within 3

weeks of treatment.

The outcomes suggest that pramipexole has considerable

promise in patients with RLS in the dose range tested. The

lowest dose, 0.125 mg, was safe and effective and generally

indistinguishable from the higher doses, suggesting that it

may be a reasonable place to start when targeting maximum

dose for optimum efficacy.

Acknowledgements

This study was supported by Boehringer-Ingelheim

International GmbH. The authors wish to thank the main

research nurse, M. Halavaara, and the personnel of the

Haaga Neurological Research Centre in Helsinki, Finland,

for providing sleep laboratory facilities and help in the

polysomnographic recordings, and also L.H. Brauer, PhD,

for her help in the preparation of the article, and R. Morton

for his statistical contribution.

References

[1] Phillips B, Young T, Finn L, et al. Epidemiology of restless legs

symptoms in adults. Arch Intern Med 2000;160:2137–41.

[2] Rothdach AJ, Trenkwalder C, Haberstock J, et al. Prevalence and risk

factors of RLS in an elderly population: the MEMO study. Memory

and morbidity in augsburg elderly. Neurology 2000;54:1064–8.

[3] Hening W, Walters AS, Allen RP, et al. Impact, diagnosis and

treatment of restless legs syndrome (RLS) in a primary care

population: the REST (RLS epidemiology, symptoms, and treatment)

primary care study. Sleep Med 2004;5:237–46.

[4] Lavigne GJ, Montplaisir JY. Restless legs syndrome and sleep

bruxism: prevalence and association among Canadians. Sleep 1994;

17:739–43.

[5] Berger K, Luedemann J, Trenkwalder C, et al. Sex and the risk of

restless legs syndrome in the general population. Arch Intern Med

2004;164:196–202.

[6] Earley CJ, Allen RP, Beard JL, Connor JR. Insight into the

pathophysiology of restless legs syndrome. J Neurosci Res 2000;62:

623–8.

[7] Earley CJ. Clinical practice. Restless legs syndrome. N Engl J Med

2003;348:2103–9.

[8] Sonka K, Kemlink D. Restless legs syndrome in 2004. Prague Med

Rep 2004;105:337–56.

[9] Allen RP, Picchietti D, Hening WA, et al. Restless legs syndrome:

diagnostic criteria, special considerations, and epidemiology. A report

from the restless legs syndrome diagnosis and epidemiology work-

shop at the national institutes of health. Sleep Med 2003;4:101–19.

[10] Allen RP, Earley CJ. Restless legs syndrome: a review of clinical and

pathophysiologic features. J Clin Neurophysiol 2001;18:128–47.

[11] Stiasny-Kolster K, Benes H, Peglau I, Hornyak M, et al. Effective

cabergoline treatment in idiopathic restless legs syndrome. Neurology

2004;63:2272–9.

[12] Allen R, Becker PM, Bogan R, et al. Ropinirole decreases periodic leg

movements and improves sleep parameters in patients with restless

leg syndrome. Sleep 2004;27:907–14.

M. Partinen et al. / Sleep Medicine 7 (2006) 407–417 417

[13] Silber MH, Ehrenberg BL, Allen RP, et al. An algorithm for the

management of restless legs syndrome. Mayo Clin Proc 2004;79:

916–22.

[14] Michaud M, Dumont M, Selmaoui B, et al. Circadian rhythm of

restless legs syndrome: relationship with biological markers. Ann

Neurol 2004;55:372–80.

[15] Chesson Jr AL, Wise M, Davila D, et al. Practice parameters for the

treatment of restless legs syndrome and periodic limb movement

disorder. An American academy of sleep medicine report. Standards

of practice committee of the American academy of sleep medicine.

Sleep 1999;22:961–8.

[16] Deleu D, Northway MG, Hanssens Y. Clinical pharmacokinetic and

pharmacodynamic properties of drugs used in the treatment of

Parkinson’s disease. Clin Pharmacokinet 2002;41:261–309.

[17] Montplaisir J, Nicolas A, Denesle R, Gomez-Mancilla B. Restless legs

syndrome improved by pramipexole: a double-blind randomized trial.

Neurology 1999;52:938–43.

[18] Montplaisir J, Denesle R, Petit D. Pramipexole in the treatment of

restless legs syndrome: a follow-up study. Eur J Neurol 2000;7(Suppl.

1):27–31.

[19] Lin SC, Kaplan J, Burger CD, Fredrickson PA. Effect of pramipexole

in treatment of resistant restless legs syndrome. Mayo Clin Proc 1998;

73:497–500.

[20] Lin S, Kaplan J, Burger C, Fredrickson P. Long-term use of

pramipexole in the treatment of restless legs syndrome: a two-year

follow-up. The sixth international congress of Parkinson’s disease and

movement disorders, Barcelona, Spain, 11–15 June, 2000.

[21] Becker PM, Ondo W, Sharon D. Encouraging initial response of

restless legs syndrome to pramipexole. Neurology 1998;51:1221–3.

[22] Stiasny-Kolster K, Oertel WH. Low-dose pramipexole in the

management of restless legs syndrome. Neuropsychobiology 2004;

50:65–70.

[23] Walters AS, LeBrocq C, Dhar A, et al. For the international restless

legs syndrome study group. Validation of the international restless

legs syndrome study group rating scale for restless legs syndrome.

Sleep Med 2003;4:121–32.

[24] Johns MW. A new method for measuring daytime sleepiness: the

Epworth sleepiness scale. Sleep 1991;14:540–5.

[25] Jenkinson C, Coulter A, Wright L. Short form 36 (SF36) health survey

questionnaire: normative data for adults of working age. Br Med J

1993;306:1437–40.

[26] National Institute of Mental Health (NIMH). Early clinical drug

evaluation unit (ECDEU). Clinical global impressions. In: Guy W,

editor. ECDEU assessment manual for psychopharmacology. rev.

Rockville, MD: NIMH USA; 1976.

[27] Solovieva S, Santavirta N, Santavirta S, Konttinen YT. Assessing

quality of life in individuals with hereditary blood coagulation

disorders. Qual Life Res 2004;13:987–1000.

[28] Trenkwalder C, Hundemer HP, Lledo A, et al. Efficacy of pergolide in

treatment of restless legs syndrome: the PEARLS study. Neurology

2004;62:1391–7.

[29] Benes H, Heinrich CR, Ueberall MA, Kohnen R. Long-term safety

and efficacy of cabergoline for the treatment of idiopathic restless legs

syndrome: results from an open-label 6-month clinical trial. Sleep

2004;27:674–82.

[30] Adler CH, Hauser RA, Sethi K, et al. Ropinirole for restless legs

syndrome. Neurology 2004;62:1405–7.

[31] Abetz L, Allen R, Follet A, et al. Evaluating the quality of life

of patients with restless legs syndrome. Clin Ther 2004;26:

925–35.

[32] Allen RP, Walters AS, Montplaisir J, et al. Restless legs syndrome

prevalence and impact: REST general population study. Arch Intern

Med 2005;165:1286–92.