ArticlePDF Available

Pramipexole in patients with Parkinson's disease and marked drug resistant tremor: A randomised, double blind, placebo controlled multicentre study

Authors:

Abstract and Figures

To compare the tremorlytic properties of pramipexole, a non-ergoline dopamine agonist to those of placebo as add on medication in patients with Parkinson's disease. Eighty four patients with early or advanced Parkinson's disease and marked, drug resistant tremor under a stable and optimised antiparkinsonian medication were included in a double blind, randomised, placebo controlled, multicentre study and assigned to add on treatment (7 week dose titration interval, 4 week maintenance period) with either pramipexole (n=44) or placebo (n=40) as adjunct. The primary end point was the absolute change in tremor score, defined as the sum of tremor related items (16, 20, 21) of the unified Parkinson's disease rating scale (UPDRS) in "on" periods. Secondary end points included the percentage change in tremor score, the absolute and percentage changes in long term EMG tremor registration, and the change in tremor self rating scales. Safety and tolerability were assessed on the basis of adverse events, laboratory tests, ECG, and vital signs. Pramipexole was significantly superior to placebo with a difference between treatment groups in the mean absolute change in tremor score of -4.4 (95% confidence interval (95% CI) -6.2 to -2.5) (p<0.0001), corresponding to a difference in the mean percentage change of -34.7% in favour of pramipexole. The secondary end points were consistent with the significant change in tremor score and provided further evidence for the benefit of pramipexole compared with placebo. Long term EMG registration as an objective measure showed a difference in mean absolute change in tremor occurrence of -15.2% (95%CI -21.4 to -9.0) (p<0.0001), and a difference in the mean percentage change of -45.7% in favour of pramipexole. The treatment effects increased during dose titration and remained stable during the 4 week maintenance dose period until the end of the study. The average daily pramipexole dose during maintenance was 4.1 (SD 0.9) mg. Safety analysis showed an increased rate of fatigue, insomnia, nausea, abdominal pain, and headache under pramipexole, comparable with previous studies. Pramipexole proved to be an effective agent for patients with Parkinson's disease and drug resistant tremor.
Content may be subject to copyright.
PAPER
Pramipexole in patients with Parkinson’s disease and
marked drug resistant tremor: a randomised, double
blind, placebo controlled multicentre study
O Pogarell, T Gasser, J J van Hilten, S Spieker, S Pollentier, D Meier, W H Oertel
.............................................................................................................................
J Neurol Neurosurg Psychiatry
2002;72:713–720
Objective
:
To compare the tremorlytic properties of pramipexole, a non-ergoline dopamine agonist to
those of placebo as add on medication in patients with Parkinson’s disease.
Methods
:
Eighty four patients with early or advanced Parkinson’s disease and marked, drug resistant
tremor under a stable and optimised antiparkinsonian medication were included in a double blind,
randomised, placebo controlled, multicentre study and assigned to add on treatment (7 week dose
titration interval, 4 week maintenance period) with either pramipexole (n=44) or placebo (n=40) as
adjunct. The primary end point was the absolute change in tremor score, defined as the sum of tremor
related items (16, 20, 21) of the unified Parkinson’s disease rating scale (UPDRS) in “on” periods. Sec-
ondary end points included the percentage change in tremor score, the absolute and percentage
changes in long term EMG tremor registration, and the change in tremor self rating scales. Safety and
tolerability were assessed on the basis of adverse events, laboratory tests, ECG, and vital signs.
Results: Pramipexole was significantly superior to placebo with a difference between treatment groups
in the mean absolute change in tremor score of 4.4 (95% confidence interval (95% CI) 6.2 to 2.5)
(p<0.0001), corresponding to a difference in the mean percentage change of 34.7% in favour of
pramipexole. The secondary end points were consistent with the significant change in tremor score and
provided further evidence for the benefit of pramipexole compared with placebo. Long term EMG
registration as an objective measure showed a difference in mean absolute change in tremor
occurrence of 15.2% (95%CI 21.4 to 9.0) (p<0.0001), and a difference in the mean percentage
change of 45.7% in favour of pramipexole. The treatment effects increased during dose titration and
remained stable during the 4 week maintenance dose period until the end of the study. The average
daily pramipexole dose during maintenance was 4.1 (SD 0.9) mg. Safety analysis showed an
increased rate of fatigue, insomnia, nausea, abdominal pain, and headache under pramipexole, com-
parable with previous studies.
Conclusion
:
Pramipexole proved to be an effective agent for patients with Parkinson’s disease and
drug resistant tremor.
T
remor at rest is one of the cardinal symptoms of
Parkinson’s disease and the first sign in about 75% of
patients. A minority of patients will present with tremor
as the predominant symptom throughout the course of the
disease (tremor dominant Parkinson’s disease).
1
Antiparkin-
sonian drugs usually provide effective treatment for bradyki-
nesia and rigidity, whereas drug treatment of tremor may be
more difficult: parkinsonian tremor is sometimes resistant to
cur rently available medication or sufficient pharmacotherapy
may cause intolerable side effects and lead to discontinuation
of treatment (drug resistant tremor).
2
Stereotactic neurosur-
gery with deep brain stimulation (nucleus ventralis interme-
dius thalami, subthalamic nucleus) or thalamotomy may lead
to excellent tremor reduction, but is—at least in the short
ter m—an expensive approach with rare but potentially severe
side effects,
3–6
and is limited to specialised centres. Therefore,
new antiparkinsonian compounds are to be investigated with
respect to their efficacy on predominant and drug resistant
parkinsonian tremor.
Pramipexole is a novel non-ergoline dopamine agonist with
high binding specificity for the dopamine D2 receptor family
and with preferential affinity to the dopamine D3 receptor
subgroup.
7–9
Clinical trials with pramipexole as monotherapy
and as an adjunct to levodopa have shown the compound to be
safe, well tolerated, and efficacious.
10–14
Recently, sudden onset
of somnolence (sleep attacks) was reported to occur in a few
patients with pramipexol.
15
This topic is presently under
further investigation.
Compared with placebo, pramipexole treated patients
showed an improvement in both activities of daily living
(ADL) and motor scores as assessed by the unified Parkinson’s
disease rating scale (UPDRS) in early and advanced Parkin-
son’s disease, and a reduction of duration and severity of “off”
periods in advanced Parkinson’s disease.
10–14
In most of these
trials, tremor items were not analysed separately or there were
no systematic investigations of patients with predominant
tremor. However, descriptive analyses of placebo controlled
trials of pramipexole showed that the incidence of tremor,
reported as a newly occurring symptom during the studies
(507 patients, 251 under pramipexole, 256 under placebo) was
less frequent in patients taking pramipexole compared with
placebo.
16
Based on an exploratory subanalysis of patients
from one study centre of a placebo controlled multicentre trial
in advanced Parkinson’s disease, pramipexole seemed to have
potent tremorlytic activity: besides overall antiparkinsonian
.............................................................
Abbreviations: UPDRS, Unified Parkinson’s disease rating scale; ADL,
activities of daily living; LOCF, last observation carried forward; SAEs,
serious adverse events
See end of article for
authors’ affiliations
.......................
Correspondence to:
Dr O Pogarell, Department
of Psychiatry, University
Hospital,
Ludwig-Maximilians-
University of Munich,
Section of Clinical
Neurophysiology,
D-80336 Munich,
Germany; oliver.pogarell@
psy.med.uni-muenchen.de
Received 13 April 2001
In final revised form
7 November 2001
Accepted
20 November 2001
.......................
713
www.jnnp.com
effects, pramipexole led to a statistically significant improve-
ment in UPDRS tremor scores in a subgroup of 16 patients
with previously drug resistant tremor.
17
As a consequence of these findings, the present trial was
conducted to investigate the tremorlytic properties of prami-
pexole compared with placebo as an add on therapy in
patients with early or advanced Parkinson’s disease presenting
with marked and previously drug resistant tremor.
METHODS
Study design
The study followed a double blind, placebo controlled,
randomised, multicentre, parallel group comparative design.
The trial was conducted according to the principles of good
clinical practice and was approved by local ethics committees
at each centre. Written informed consent had to be given by all
participants. A total of four study sites were involved in two
European countries. Trial duration was up to 12 weeks. The
treatment groups were pramipexole and placebo as add on
therapy to a stable and optimised antiparkinsonian medi-
cation. The study dr ugs were administered as tablets with
identical appearance and taste. A computer generated
randomisation plan that included stratification by centre and
block (with a block size of four) was used to ensure a balanced
distribution (1:1) of treatment groups within centres. No per-
son directly involved in management or analysis of the trial
had access to the treatment assignment during the conduct.
After a blinded repor t planning meeting where the analysis
plan was finalised, the database was locked, treatment assign-
ment was added, and no further changes were made to the
database.
Inclusion/exclusion criteria
Patients of both sexes with Parkinson’s disease (Hoehn and
Yahr stage I-IV as assessed after at least 12 hours off
medication) according to the United Kingdom Parkinson’s
disease brain bank criteria were included.
18
Patients had to
fullfill the cr iteria of marked and drug resistant tremor during
their medical history—subjects either failed to experience a
clinically relevant and useful improvement in tremor under an
optimised antiparkinsonian therapy with various agents, or
side effects encountered under an effective antitremor therapy
were intolerable. Marked tremor was clinically defined by the
presence of a sum score of at least eight of 32 of the UPDRS
tremor items 16, 20, and 21 (referred to as tremor score) or, if
the tremor was present on one side only, by the presence of a
tremor score of at least six of 32. The generation of this sum
score is based on the validity of the UPDRS regarding
tremor,
19
and the clinical experience, that the UPDRS tremor
items reflect different but similarly important aspects of Par-
kinsonian tremor, including the impairment of activities of
daily living by tremor.
Tremor was assessed during “on” periods—that is, 2 to 3
hours after the last intake of antiparkinsonian medication.
Patients with atypical parkinsonian syndromes (multiple
system atrophy, progressive supranuclear palsy, etc), severe
dementia, epilepsy, previous neurosurgery, electroconvulsive
therapy within 90 days before randomisation, or severe other
physical diseases were excluded. Also excluded were patients
with symptomatic orthostatic hypotension—that is, a decline
in systolic (diastolic) blood pressure at 1 minute after standing
by 20 mm Hg or more compared with supine blood pressure
obtained after 5 minutes of quiet rest. Female patients of
childbearing potential were required to use medically accepted
means of contraception.
The patients had to receive a stable antiparkinsonian medi-
cation with levodopa/decarboxylase inhibitor preparations,
and/or selegiline, and/or amantadine for at least 30 days
before randomisation. Before study inclusion, the medication
had to be optimised regarding the symptomatic effects on
parkinsonian signs and symptoms and the dose related side
effects in each individual patient. Optimisation was mainly
perfor med by adjusting the levodopa dose either by increasing
the number of doses or increasing the dosage in each dose.
Concomitant treatment with the following agents was not
allowed during the past month (neuroleptic drugs and meto-
clopramide: the past 2 months) before randomisation:
dopamine agonists, MAO inhibitors except for selegiline, anti-
cholinergic drugs, budipine, reserpine, (classic) neuroleptic
drugs, metoclopramide, methylphenidate hydrochloride, am-
phetamine derivatives, α methyldopa, cinnarizine, and fluna-
rizine.
Efficacy criteria
The primary end point was the absolute change in tremor
score during “on” periods from baseline to the end of mainte-
nance. Secondary end points were (1) absolute and relative
(percentage) changes in separate tremor score items, and
other UPDRS scores, (2) changes in two tremor self rating
scales based on a patient’s diary, (3) absolute and relative
(percentage) changes in “tremor occurrence” as measured by
a long term EMG registration, and (4) a global assessment of
the effect on tremor.
Tremor score
Tremor score was calculated as the sum of UPDRS items 16,
20, and 21. Evaluation was done during an “on” period.
“On”periods were defined as 2 to 3 hours after the last intake
of study medication together with levodopa preparations
and/or other antiparkinsonian drugs, if applicable.
UPDRS scores
UPDRS part II (ADL) was evaluated for “on” and “off” periods
and the average calculated, a procedure consistent with other
Parkinson’s disease studies.
10 11
UPDRS par t III (motor exam-
ination) was evaluated during “on” periods. The sum of both
was calculated as the combined UPDRS II/III score.
Patient’s diary
The patient’s diary consisted of two tremor self rating scales
19
:
(1) Impairment of daily living by tremor and (2) severity of
tremor.
(1) Impairment of daily living by tremor is a 21 item check-
list of daily living tasks (for example, cutting with a knife,
using a spoon, brushing teeth etc). The best performance for
each item per day is rated using a four point scale (0, no diffi-
culty; 1, slight difficulty; 2, considerable effort; and 3, cannot
be fulfilled). The maximum score is 63 points.
(2) Severity of tremor is rated on a five point scale (0, miss-
ing; 1, mild; 2, moderate but occasionally occurring; 3, moder-
ate but per sisting; and 4 severe) with the items rest tremor,
postural tremor, and impairment by tremor (maximum score
12 points). Ratings were performed every 2 hours of the wak-
ing day. In both scales a reduction in total score indicates
improvement. Each scale was used on the 3 consecutive days
preceding each visit. The scores of these 3 days were averaged.
Long term EMG
A long term EMG tremor reg istration at baseline and end of
maintenance was used as an objective measure to quantify the
occur rence of tremor, expressed as a time percentage of a
registration period of 10 waking hours. The tremor recording
was performed with a long term EMG appliance via skin elec-
trodes placed bilaterally over the muscle venters of extensor
carpi radialis and flexor carpi ulnaris muscles with reference
electrodes placed 10 cm distally. Registration and tremor
analysis were performed according to a standardised protocol
of EMG tremor registration.
20 21
Only the side with the most
pronounced tremor was evaluated. The analysed time periods
were identical at baseline and end of maintenance in
individual patients.
714 Pogarell, Gasser, van Hilten, et al
www.jnnp.com
Global assessment
The effect on tremor at the end of the trial was assessed
globally on a three point scale (clear improvement, no clear
effect, clear deterioration) by both investigators and patients,
with the end points clear improvement versus no clear effect
or deterioration used in the analysis.
Safety criteria
Safety was assessed by physical examination, supine and
standing blood pressure measurements, laboratory tests (red
blood cells, white blood cells, enzymes, electrolytes, urinaly-
sis), 12 lead ECG, documentation of adverse events, and the
investigators’ global impression of tolerance.
Study procedures
At the screening visit medical history, physical examination,
vital signs, laboratory tests, ECG, modified Hoehn and Yahr
scale (“on” and “off”), UPDRS part II and III with derived
tremor scores were performed and the patients’ diaries with
self rating scales distributed.
Aftera2weekscreening period the patients were randomly
assigned to either pramipexole or placebo under double blind
conditions in a 1:1 ratio (baseline visit). Baseline assessments
comprised those for all end points, adverse events, and
changes in concomitant medication.
Subsequently the patients underwent an ascending dose
interval up to 7 weeks (weekly visits) with individual dose
adjustments from 0.375 mg to 4.5 mg/day pramipexole or
matching placebo using a three times a day regimen (3×0.125,
0.25, 0.5, 0.75, 1.0, 1.25, and 1.5 mg). The dosage was
increased in weekly steps until the patient showed a stable
and optimal improvement or received the maximally tolerated
dose (up to a maximum dose of 4.5 mg daily). Dose
adjustment was followed bya4weekmaintenance period. At
the end of maintenance there was a 1 week dose reduction
period to gradually withdraw study medication.
Additional antiparkinsonian medication (levodopa/
decarboxylase inhibitor preparations and/or selegiline and/or
amantadines) had to remain stable from 30 days before
randomisation and throughout the course of the study.
The following assessments were performed at each visit
after baseline—that is, at weekly visits during the ascending
dose interval, at the end of the maintenance period, and after
dose reduction: tremor score, UPDRS II (“on” and “off”),
UPDRS III (“on”), evaluation of patients’ diaries, adverse
events, changes in concomitant non-antiparkinsonian medi-
cation. Just before the end of maintenance there was a second
EMG registration of at least 10 hours of waking time. After
dose reduction physical examination, vital signs, laboratory
tests, and ECG were reassessed.
Statistical analysis
Baseline comparability of treatment groups was exploratively
assessed by χ
2
test (sex), two way analysis of variance
(ANOVA; age, UPDRS), Mantel-Haenszel test (Parkinson’s
disease duration, Hoehn and Yahr stage) and Wilcoxon-Mann-
Whitney test (levodopa dose) with factors treatment and cen-
tre.
Analysis of variance with f actors treatment and centre (for
nor mally distributed data), and the Wilcoxon-Mann-Whitney
test as alternative were applied for statistical evaluation of
differences in efficacy between the treatment groups regard-
ing the primary end point, the UPDRS derived secondary end
points, the self rating scales, and EMG tremor registration. As
the interaction treatment×centre was not significant (p=0.12)
this factor was removed from the ANOVA and only the main
effects remained. 95% Confidence intervals and p values were
calculated for differences in the mean absolute changes
between the treatment groups. The patients’and investigators’
global assessments of the effect on tremor of pramipexole or
placebo were compared using the Mantel-Haenszel test (clear
improvement v deterioration or no clear improvement), confi-
dence intervals for the differences of proportions were calcu-
lated using normal approximation. The null hypothesis for
statistical tests was that there is no difference between the
mean change from baseline to the end of maintenance in the
pramipexole and placebo treatment groups (intent to treat
population) and was tested for primary and secondary end
points. Post hoc subgroup analyses to determine the influence
of antiparkinsonian drugs other than study medication,
adverse events (fatigue), presence of “off” periods at baseline,
low or high tremor scores were performed for the primary end
point. Incidences of adverse events were descriptively evalu-
ated using Fisher’s exact test for the occurrence per treatment
group.
The intent to treat population included all patients who
were randomised to treatment, received at least one dose of
medication, and had at least one postbaseline efficacy assess-
ment. Missing data of the intent to treat population were esti-
mated using the “last observation carried forward” technique.
Previous power calculations had suggested that a sample size
of 42 in each treatment arm would be required to detect a sta-
tistically significant difference with 80% probability at the 5%
Received pramipexole:
n = 44
Did not receive pramipexole
as allocated: n = 0
Not randomised: n = 6
• Withdrawal of consent
before randomisation n = 1
• Violation of inclusion-exclusion criteria
during screening period n = 5
Registered: n = 90
Follow up: n = 44 (ITT)
Timing: baseline,
on treatment at week:
• 1–7 (ascending dose)
• 11 (end of maintenance)
• 12 (after dose reduction)
(EMG recording at baseline
and week 11 only)
Withdrawn: n = 0
Completed pramipexole
treatment: n = 44
(ITT population n = 44)
Received placebo: n = 40
Did not receive placebo
as allocated: n = 0
Follow up: n = 39 (ITT)
Timing: baseline,
on treatment at week:
• 1–7 (ascending dose)
• 11 (end of maintenance)
• 12 (after dose reduction)
(EMG recording at baseline
and week 11 only)
Withdrawn: n = 2
• Intervention ineffective n = 1
• Before follow up due to
preexisting disease n = 1
Completed placebo
treatment: n = 38
(ITT population n = 39)
Randomisation:
n = 84
Figure 1 Profile of the randomised trial: flow diagram with the
progress of the patients throughout the trial.
Pramipexole in patients with Parkinson’s disease with predominant tremor 715
www.jnnp.com
level of significance, if the difference in the mean change of
tremor score between treatment groups was 2.2 units.
RESULTS
Treatment population
Eighty four patients gave informed consent, were included,
and were randomised. One patient was withdrawn after the
first treatment dose (placebo) without postbaseline efficacy
measurements, as immediate cardiac surgery for an hitherto
undetected aortic valve stenosis was required. Thus the inten-
tion to treat population consisted of 83 patients (60 male, 23
female), 44 in the pramipexole group, 39 in the placebo group.
Eighty two (44 pramipexole, 38 placebo) patients completed
the trial according to protocol. One placebo patient discontin-
ued during the ascending dose period because of an
unsatisfactory therapeutic effect (fig 1).
The mean age of the intention to treat population was 63.6
(SD 8.9) years with a range from 35 to 80 years and a mean
disease duration of 6.3 (SD 3.8) years. The mean duration of
drug treatment was 3.8 (SD 3.3) years. Seventy three patients
were treated with levodopa with a median daily dose of 300
mg. Forty three patients (52%) received daily dosages of 300
mg levodopa or less, and 30 (36%) of more than 300 mg. Sel-
egiline and amantadine were used by 12 (10 pramipexole, two
placebo) and 14 (nine pramipexole, five placebo) patients,
respectively.
The sever ity of Parkinson’s disease was estimated at screen-
ing by Hoehn and Yahr staging dur ing “on” and “off” periods.
In “on” periods most of the patients were in Hoehn and Yahr
stages 2 and 2.5. Thirty seven patients had no “off” periods at
screening and most of the 46 patients with fluctuations were
in Hoehn and Yahr “off” stages 2.5 and 3.
At baseline the mean UPDRS II scores (average of “on” and
“off”) were 13 (SD 6.4) in the pramipexole group and 11.5 (SD
4.6) in patients on placebo. Mean UPDRS III (“on”) baseline
scores were 34.2 (SD 15.3) compared with 32.1 (SD 11),
pramipexole and patients with placebo, respectively. The mean
tremor score in “on” was 11.9 (SD 5) in the pramipexole ver-
sus 10.9 (SD 3.5) in the placebo group with a mean proportion
of the tremor score relative to the UPDRS II and III sum scores
of 28% and 27%, respectively. Both treatment groups were
comparable with respect to demographic and clinical features.
Exploratory and descriptive analyses were not suggestive of
significant differences at baseline (table 1).
Table 1 Demographics and baseline characteristics of the intent to treat population
(n=83) with mean (SD), n (%), or median, where appropriate
Pramipexole
(n=44)
Placebo
(n=39) Total (n=83)
Sex (n (%)):
Male 30 (68) 30 (77) 60 (72)
Female 14 (32) 9 (23) 23 (28)
Age (y):
Mean (SD) 62.0 (10.1) 65.4 (7.1) 63.6 (8.9)
Range 35–80 47–80 35–80
Duration of PD (y):
Mean (SD) 6.5 (4.0) 6.0 (3.5) 6.3 (3.8)
Range 0.9–17 2.0–16 0.9–17
Duration of drug treatment (y):
Mean (SD) 3.9 (3.1) 3.6 (3.5) 3.8 (3.3)
Levodopa dose (mg):
Median 300 300 300
Range 50–700 100–1700 50–1700
Patients (n (%)):
With no levodopa 5 (11) 5 (13) 10 (12)
<300 mg 25 (57) 18 (46) 43 (52)
>300 mg 14 (32) 16 (41) 30 (36)
Patients without “off” periods (at baseline) (n (%)) 22 (50) 16 (41) 38 (46)
Hoehn and Yahr Stage: in “on” periods (n (%)):
I 6 (14) 4 (10) 10 (12)
I.5 4 (9) 5 (13) 9 (11)
II 22 (50) 20 (51) 42 (51)
II.5 10 (23) 7 (18) 17 (20)
III 2(4) 3(8) 5(6)
IV 000
Hoehn and Yahr Stage: in “off” periods (n (%)):
I 0 1(3) 1(1)
I.5 0 2 (5) 2 (2)
II 3 (7) 6 (15) 9 (11)
II.5 12 (27) 10 (26) 22 (27)
III 7 (16) 4 (10) 11 (13)
IV 0 1 (3) 1 (1)
Tremor score (mean (SD)) 11.9 (5) 10.9 (3.5) 11.4 (4.3)
Item 16 (reported) 2.2 (0.8) 2.1 (0.7) 2.2 (0.7)
Item 20 (rest) 6.3 (3.4) 5.8 (2.2) 6.1 (2.9)
Item 21 (action/postural) 3.3 (1.6) 3.0 (1.8) 3.2 (1.7)
UPDRS (mean (SD)):
UPDRS II/III sum score 47.2 (21.1) 43.6 (14.4) 45.5 (18.2)
UPDRS II(mean “on/off”) 13 (6.4) 11.5 (4.6) 12.3 (5.7)
UPDRS III (“on”) 34.2 (15.3) 32.1 (11) 33.2 (13.4)
Patient’s diary (mean (SD)):
Daily living 20.2 (14.9) 17.3 (10.8) 18.8 (13.1)
Severity 6.1 (2.9) 5.7 (2.3) 5.9 (2.6)
Long term EMG:
Tremor occurrence (%, mean (SD)) 41.8 (21.5) 49.5 (21.6) 45.5 (21.8)
716 Pogarell, Gasser, van Hilten, et al
www.jnnp.com
Efficacy
Primary end point
The change in tremor score showed a statistically significant
difference between pramipexole and placebo group
(p<0.0001). The difference in the mean absolute change
between the treatment groups was 4.4 (95% confidence
interval 6.2 to 2.5), corresponding to a difference in the
mean relative change of 34.7% in favour of pramipexole
(table 2, fig 2). The visit by visit analysis of the change in
tremor score showed that the improvement under pramipex-
ole increased in a dose dependent manner during the ascend-
ing dose interval and seemed to remain stable between the
beginning and end of maintenance (fig 3).
Single tremor items
The separate analysis of the single UPDRS tremor items
(UPDRS II item 16 “on”: reported tremor in the ADL, UPDRS
III items 20 and 21: tremor at rest and action or postural
tremor, resp) disclosed similar results, with a statistically sig-
nificant difference in favour of pramipexole for rest tremor
and postural tremor (p<0.0001), as well as for reported
tremor (p<0.01). The differences in the mean absolute change
between the treatment groups were 2.6 (95% CI 3.8 to 1.4)
and 1.2 (95% CI 1.8 to 0.6) for items 20 and 21,
respectively, and 0.6 (95% CI 0.9 to 0.2) for item 16.
Subgroup analyses
The change in tremor score (primary end point) was further
evaluated according (1) to the presence or absence of “off”
periods at baseline, (2) to low or high baseline tremor scores,
and (3) to the presence or absence of fatigue (table 3). In
addition, the influence of antiparkinsonian medication other
than pramipexole or placebo was analysed.
(1) Pramipexole treatment was significantly superior to
placebo in both patients with and without “off periods at
baseline, with a difference in the mean change in tremor score
between the treatment groups by 5.8 (p=0.0001), and 3.2
(p=0.006), respectively. Interaction tests (ANOVA with factors
treatment and “off” status) disclosed that the reduction in
Table 2 Synopsis of primary and secondary end points for the intent to treat population (last observation carried
forward technique): absolute changes from baseline to end of maintenance
Pramipexole
(n=44)
Placebo
(n=39)
Difference between
groups (95% CI)
Difference in mean relative
changes (%)
Tremor scores (UPDRS):
Tremor score 5.8 (5) 1.5 (3.2) 4.4 (6.2 to 2.5) 34.7 p<0.0001
Item 16 0.7 (0.9) 0.2 (0.7) 0.6 (0.9 to 0.2) 21.5 p<0.01
Item 20 3.6 (3.3) 1.0 (2.2) 2.6 (3.8 to 1.4) 37.9 p<0.0001
Item 21 1.5 (1.6) 0.3 (1.1) 1.2 (1.8 to 0.6) 35.6 p<0.0001
UPDRS scores:
UPDRS II/III (sum score) 18.8 (13.9) 3.8 (8.3) 15 (20 to 10) 30.9 p<0.0001
UPDRS II (average “on”-”off”) 3.6 (3.8) 0.1 (2.6) 3.5 (4.9 to 2.0) 25.2 p<0.0001
UPDRS III (“on”) 15.2 (11.6) 3.7 (6.8) 11.5 (15.7 to 7.4) 34.1 p<0.0001
Patient’s diary:
Daily living 4.7 (8.6) 3.4 (8.0) 8.1 (11.7 to 4.5) 43.4 p<0.0001
Severity 1.6 (2.1) 0.6 (2.2) 2.2 (3.1 to 1.3) 39.5 p<0.0001
Long term EMG:
Tremor occurrence (%) 19.3 (14.8) 4.1 (13.2) 15.2 (21.4 to 9.0) 45.7 p<0.0001
Global assessment (patients with improvement (%)):
Investigator’s assessment 56.8 12.8 44.0 (26.1 to 61.9) NA p<0.0001
Patient’s assessment 56.8 17.9 38.9 (20.0 to 57.7) NA p<0.0001
Values are mean (SD) or n (%) for pramipexole and placebo group; differences in the mean absolute changes between groups (mean and 95% confidence
intervals (95% CIs); and differences in mean relative changes between groups ((%)); p values are given for the differences in the mean absolute changes
(controlled for differences between study centres); NA, not applicable.
14
10
6
8
4
0
2
–2
–4
–8
–6
End of
maintenance
– baseline
End of
maintenance
Pramipexole
Tremor score
Baseline
12
11.9 (5)
10.9 (3.5)
6.1 (5)
9.4 (4.1)
–5.8 (5)
–1.5 (3.2)
Placebo
p < 0.0001
Figure 2 Mean tremor score (SD) at baseline (left) and end of
maintenance (middle) of pramipexole (n=44) and placebo (n=39)
group. Right: mean change from baseline to end of maintenance.
14
12
8
10
6
4
0
2
12978645
Weeks on treatment
Pramipexole (n = 44)
Placebo (n = 39)
Tremor score
3201 10 11
End of
maintenance
Figure 3 Development of the mean tremor score per week on
treatment with pramipexole (ppx) and placebo (pbo) (intent to treat,
last observation carried forward), at week 0 (baseline), weeks 1–7
(ascending dose interval), weeks 7–11 (maintenance period), week
11–12 (dose reduction).
60
40
50
30
10
20
0
–10
–30
–20
End of
maintenance
– baseline
End of
maintenance
Pramipexole
% Time with tremor
Baseline
41.8 (21.5)
49.5 (21.6)
22.5 (20)
45.4 (21.6)
–19.3 (14.8)
–4.1 (13.2)
Placebo
p < 0.0001
Figure 4 Mean occurrence of tremor by long term EMG
registration (% time of waking hours (SD)) at baseline (left) and end
of maintenance (middle) of pramipexole and placebo group. Right:
Mean change from baseline to end of maintenance.
Pramipexole in patients with Parkinson’s disease with predominant tremor 717
www.jnnp.com
tremor was significantly greater in the subgroup with “off”
periods at baseline (p=0.038, “with off” v “without off”).
(2) As the median tremor score at baseline was 11, a high
score was defined as>11 and a low score as<11. The
difference between the treatment groups in the mean change
in tremor score was 6.1 (p=0.0001) in the high score group,
and 2.2 (p=0.03) in the low score group, both in favour of
pramipexole. The interaction between the factors treatment
and baseline tremor score was statistically significant
(p=0.01, “high” v “low score”)—that is, the improvement
under pramipexole was stronger in patients with a high base-
line tremor score.
(3) To investigate whether the reduction in tremor was
influenced by side effects such as increased tiredness or
fatigue, the pramipexole treated patients without these
adverse events (n=34) were descriptively compared to those
patients, who experienced fatigue or increased tiredness dur-
ing treatment (n=10). There was no significant difference in
the mean change in tremor score from baseline to end of
maintenance between these subgroups (p=0.73). The im-
provement was even slightly more pronounced in the group
without compared with the group with fatigue/tiredness
(mean change by 6 (SD 4.8) v 5.4 (SD 5.6)), so there is no
evidence that additional fatigue during the trial contributed to
a reduction in tremor.
Further post hoc analyses indicated that levodopa, sel-
egiline, or amantadine use did not influence treatment
outcome; neither did levodopa dose, when stratified as>300
mg versus<300 mg.
UPDRS parts II and III
The mean improvement in the combined UPDRS II/III sum
score was significantly superior under pramipexole
(p<0.0001). The difference in the mean absolute change
between the treatment groups was 15 (95% CI 20 to 10)—
that is, a percentage difference in UPDRS II/III sum scores of
30.9% in favour of pramipexole. The single UPDRS II and III
scores also showed a significant improvement (p<0.0001) in
the pramipexole group compared with the placebo group. The
differences between groups were 3.5 (95% CI 4.9 to 2.0) for
UPDRS II and 11.5 (95% CI 15.7 to 7.4) for UPDRS III,
indicating an overall efficacy of pramipexole on activities of
daily living and motor performance.
Patients’ diaries
The tremor self rating with respect to impairment of daily liv-
ing and severity of tremor, as based on the patients’ diaries
disclosed a difference in the mean improvement between the
treatment groups of 8.1 (95% CI 11.7 to 4.5) and 2.2 (95%
CI 3.1 to 1.3) in favour of pramipexole (p<0.0001), respec-
tively.
Long term EMG
The EMG recordings of tremor activity were consistent with
the patients’ subjective estimation (fig 4): Long term EMG
during waking hours displayed a mean occurrence of tremor
(as percentage of a time interval of 10 consecutive hour s) at
baseline of 41.8 (SD 21.5)% in the pramipexole group and 49.5
(SD 21.6)% in the placebo group. To the end of maintenance
the mean tremor occur rence decreased with a group
difference of 15.2 (95% CI 21.4 to 9.0) (p<0.0001), corre-
sponding to a difference in the mean relative change of -45.7%
in favour of pramipexole.
Global assessment
The investigators’ assessment of the effect on tremor showed
a clear improvement in 25 (56.8%) pramipexole patients ver-
sus five (12.8%) placebo patients. This corresponded with the
patients’ estimation: clear improvement was reported by 25
patients (56.8%) of the pramipexole and seven patients
(17.9%) of the placebo group. The differences between prami-
pexole and placebo groups were 44.0% (95% CI 26.1 to 61.9)
and 38.9% (95% CI 20.0 to 57.7) in favour of the pramipexole
group (p<0.0001), respectively.
Safety
The safety population consisted of all 84 patients who have
received at least one administration of the test dose (44
pramipexole, 40 placebo). Safety and tolerance were generally
assessed as good in most cases. Global clinical impression of
tolerance was rated as good in 94% of the patients and did not
differ between treatment groups. No deaths were reported.
The mean duration of treatment was comparable in the
treatment groups: 48.8 (SD 9.5) versus 46.4 (SD 9.1) days
ascending dose interval, 31.2 (SD 4.8) versus 29.4 (SD 3.9)
days maintenance, and 4.9 (SD 1.4) versus. 4.7 (SD 1.3) days
dose reduction phase, pramipexole and placebo group, respec-
tively. The average daily dose in the pramipexole group was 4.1
(SD 0.9) mg and 4 (SD 0.8) mg in the placebo group.
A descriptive analysis of adverse events showed a higher
overall incidence of side effects in patients treated with
pramipexole. A total of 72 patients (85.7%) reported at least
one adverse event, 41 (93.2%) and 31 (77.5%) in the
pramipexole and placebo group, respectively (p=0.06). Com-
mon treatment emergent adverse events—that is, with an
incidence of at least 10% in either treatment group were
Table 3 Subgroup analyses of primary end point data (tremor score): stratification
of the intent to treat population according to the presence or absence of “off”periods
at baseline, high or low tremor scores, and patients with and without fatigue,
reported as adverse event
Pramipexole Placebo
Mean difference
between groups
(p value)
Interaction test
(treatment×subgroup
qualifier)
“Off” status (yes/no) 22/22 patients 23/16 patients p=0.038
Patients with “off” 8.1 (5.4) 2.3 (3.6) 5.8 (p=0.0001)
Patients without “off” 3.6 (3.3) 0.4 (2.3) 3.2 (p=0.006)
Tremor score (high/low) 24/20 patients 18/21 patients p=0.01
Tremor score >11 8.0 (5.4) 1.9 (3.7) 6.1 (p=0.0001)
Tremor score <11 3.3 (2.6) 1.1 (2.8) 2.2 (p=0.03)
Fatigue (yes/no) 10/34 patients 4/35 patients NA
Patients with fatigue 5.4 (5.6)* 0.8 (3.3) NA
Patients without fatigue 6 (4.8)* 1.6 (3.3) NA
*p=0.73 for pramipexole treated patients.
NA; not applicable; change in tremor score from baseline to end of maintenance; difference in the mean
change between treatment groups (p value for the difference between treatment groups); and interaction tests
with factors treatment×subgroup qualifiers (“off” status, tremor score). Change in tremor score in patients with
or without fatigue per treatment group.
718 Pogarell, Gasser, van Hilten, et al
www.jnnp.com
fatigue (10 pramipexole/4 placebo patients), dizziness (8/6),
insomnia (9/3), nausea (7/3), aggravated parkinsonism (4/4),
abdominal pain (6/1), tremor (2/5), and headache (6/1).
Except for aggravated parkinsonism and tremor each of these
adverse events were reported more often in the pramipexole
group (table 4). Adverse events with a risk difference in excess
of 5% (more than two patients) between treatment groups,
were fatigue, insomnia, nausea, abdominal pain, and head-
ache. Tremor was reported in more placebo patients (12.5%
versus 4.5%). The differences between the treatment groups in
the incidence of these adverse events did not reach statistical
significance. In this trial, new occurrence of dyskinesias was
not seen in the pramipexole group and hallucinations were
reported in two (4.5%) patients on pramipexole. Serious
adverse events (SAEs) occurred in four (4.8%) patients, two in
each treatment group. Only one SAE (orthostatic hypotension
with a short loss of consciousness in a pramipexole treated
patient) was assessed as drug related. One patient (placebo
group) dropped out due to a serious adverse event (cardiac
surgery for hitherto undetected aortic valve stenosis), but this
was not related to the study drug. No other adverse events led
to discontinuation. Laboratory assessments, ECG, vital signs,
and physical findings seemed not to be systematically changed
by pramipexole treatment.
DISCUSSION
The present study shows that pramipexole is not only an
effective antiparkinsonian agent with respect to improvement
in ADL or UPDRS motor scores as a whole, but also leads to a
statistically significant reduction of parkinsonian tremor
when added to a stable antiparkinsonian medication. With
respect to the tremor scores, the difference in mean relative
improvement between the treatment groups was 34.7% in
favour of pramipexole. The corresponding difference in the
change in UPDRS II/III sum score was 30.9%. Accordingly,
there could be a slightly pronounced efficacy of pramipexole
on tremor in these patients, and it seems that the reduction in
tremor is not merely due to an overall antiparkinsonian effect.
However, our findings are only descriptive, and in the
literature there are no comparable data on selective efficacy of
antiparkinsonian agents.
The long term EMG recordings as an objective measure of
tremor activity over time were consistent with the change in
tremor score, showing a difference in the mean relative reduc-
tion in tremor occurrence by 45.7% in favour of pramipexole.
Furthermore, both patients’ and investigators’ assessments as
well as the patients’ self rating scales showed superiority of
pramipexole in this trial and thus underlined the clinical rel-
evance and benefit in daily living of the observed effects.
The differences between pramipexole and placebo increased
dose dependently during titration and were maintained until
the end of the study. As indicated by the analysis of the single
tremor score items, pramipexole favorably influenced not
only—as might have been anticipated—rest tremor, but also
both action and postural components of tremor.
Subgroup analyses disclosed that patients with “off”
periods, indicating an advanced stage of the disease, and
patients with higher tremor scores at baseline derived best
benefit from pramipexole treatment. However, the treatment
effect of pramipexole remained significant in the subgroups
without “off” periods, or with lower baseline tremor scores.
Pramipexole has been reported to cause “sleep attacks”.
15
A
non-specific sedative effect could, conceivably, be responsible
for the improvement of tremor. As there was no correlation
between the reduction in tremor and the presence or absence
of fatigue or increased tiredness in the subgroup analysis, effi-
cacy end points do not seem to be influenced by this side effect
profile of the study drug.
Two points should be made concerning the inclusion crite-
ria and study population. Firstly, there is, unfortunately, no
accepted UPDRS derived measure to define “tremor domi-
nant”, so that certainly some patients with mixed type
Parkinson’s disease and severe tremor were included. By clini-
cal impression, though, most patients were characterised as
tremor dominant Parkinson’s disease. As the mean tremor
score was 11.4 at baseline, all patients had at least marked
parkinsonian tremor in “on”, regardless of being tremor
dominant or not.
Secondly, patients were required to receive an optimised
antiparkinsonian therapy before inclusion. As the median
levodopa dose was relatively low (300 mg in all study arms),
this condition does not seem to be fulfilled at first glance.
However, most patients had Hoehn and Yahr scores of 2 in the
“on” periods and only 2.5 in the “off” periods indicating a suf-
ficient global therapeutic response, and a mild to moderate
disease severity of the study population. In addition, before
study inclusion, antiparkinsonian therapy was optimised by
balancing the best possible ratio of symptomatic benefit to
side effects provided at an individual base, which might favour
lower levodopa doses.
The improvement in tremor in this patient group is remark-
able, as the patients enrolled into the trial had shown a
marked to severe parkinsonian tremor throughout their
medical history, despite an otherwise optimised medication.
Add on therapy with pramipexole led to a further significant
improvement in parkinsonian symptoms and particularly in
tremor throughout the study. Upon discontinuation of the
trial medication, the tremor scores rapidly returned to baseline
values (fig 3), consistent with a drug relation of the observed
effect. A maintenance period of 4 weeks as in this study is
relatively short. However, other clinical trials with longer
observation periods proved a sustained treatment effect of
pramipexole.
10 11 13
The safety data showed a good overall tolerability, although
the number of patients with at least one adverse event was
higher in the pramipexole group (93.2% v 77.5%, p=0.06). The
most prominent adverse events were fatigue, insomnia,
nausea, abdominal pain, and headache, which is in line with
previous studies and similar to those of other dopamine
agonists.
2101214
None of these single adverse events showed a
statistically significant difference between treatment groups;
however, these exploratory analyses are limited by the few
patients and should not be overrated. There were no
unexpected safety results or deaths. “Sleep attacks”, recently
reported in patients under non-ergoline dopamine agonists,
15
did not occur during this trial, and increased tiredness or
fatigue did not contribute to the treatment effects. Pramipex-
ole was regarded as safe and tolerable within the tested dose
range from 0.375 to 4.5 mg daily with an average daily dose of
4.1 mg in the population studied.
Table 4 Occurrence of adverse events
(AEs)/treatment group
pramipexole
(n=44)
placebo
(n=40) p Value
Any AE 41 (93.2) 31 (77.5) 0.06
Fatigue 10 (22.7) 4 (10) 0.15
Dizziness 8 (18.2) 6 (15) 0.78
Insomnia 9 (20.5) 3 (7.5) 0.12
Nausea 7 (15.9) 3 (7.5) 0.32
Aggravation of parkinsonism 4 (9.1) 4 (10) 1.00
Abdominal pain 6 (13.6) 1 (2.5) 0.11
Tremor 2 (4.5) 5 (12.5) 0.25
Headache 6 (13.6) 1 (2.5) 0.11
Serious AE 2 (4.5) 2 (5) 1.00
Assessed as drug related 1 (2.3) 0 1.00
Values are number of patients (%) with at least one AE; common
treatment emergent adverse events, occurring in at least 10% of
patients in either treatment group; and serious adverse events; p
values (Fisher’s exact test) for the differences between treatment
groups
Pramipexole in patients with Parkinson’s disease with predominant tremor 719
www.jnnp.com
Positive effects of dopamine agonists as adjunct to levodopa
therapy on parkinsonian tremor have been reported
earlier.
22–24
Pergolide improved parkinsonian tremor in an open
label trial in patients unresponsive to bromocriptine therapy
(retrospective analysis),
22
and in a placebo controlled double
blind trial
23
as part of an overall antiparkinsonian effect. Both
lisuride and bromocriptine showed a significant improvement
in tremor when added to long term levodopa treatment in
patients with wear ing off phenomena.
24
None of these studies
were designed to investigate the effects on tremor separately
in a homogeneous sample of patients.
The reported trial is the first controlled, double blind study
of a dopamine agonist focusing on this cardinal symptom,
including tremor scales and objective EMG recordings. The
study was planned to test previous—methodologically
limited—findings that have suggested tremorlytic properties
of pramipexole and was designed to compare pramipexole
with placebo.
17
As no other dopamine agonist has been inves-
tigated with a special attention to tremor, the magnitude of
the tremorlytic response of pramipexole cannot be directly
compared with other substances. Data available on budipine, a
butyldiphenylpiperidine with affinities to various neurorecep-
tors and an antiparkinsonian agent, suggested to be tremor-
lytic, have shown an improvement of 33.7%
25
and 40%
26
as
measured with long term EMG. Our result of 45.7% difference
in relative tremor reduction between pramipexole and placebo
is even more pronounced. Thus further active comparator
studies are warranted to directly compare pramipexole to
other substances with tremorlytic potential. Nevertheless, we
conclude that in patients with early or advanced Parkinson’s
disease and insufficient control of tremor, add on pramipexole
is an effective dopamine agonist and thus seems to provide a
therapeutic option for this subgroup of patients with Parkin-
son’s disease.
ACKNOWLEDGEMENTS
This study was supported by Boehringer Ingelheim Pharma KG,
Ingelheim, Germany. The following coinvestigators were involved in
patient recruitment and data collection: A Ihsche, S Redies, and F
Schnorpfeil, Marburg, Germany; M Singer, and C Macher, Munich,
Germany;WJTvandeBeek, Leiden, The Netherlands; S Breit,
Tuebingen, Germany. We thank Dr J Koester, Ingelheim, Germany, for
statistical planing, analysis, and review. DM and SP are employees of
Boehringer Ingelheim, the manufacturer of pramipexole. JJvH has
received a fund from Boehringer Ingelheim for research. TG and WHO
have been reimbursed by Boehr inger Ingelheim for attending several
symposia, at which they gave presentations. In addition WHO received
an unrestricted grant of US$15 000 for studying sudden onset
somnolence in Parkinson’s disease in the year 2000.
.....................
Authors’ affiliations
O Pogarell, W H Oertel, Department of Neurology, Philipps-University
of Marburg, Marburg, Germany
T Gasser, Department of Neurology, University of Munich, Munich,
Germany
J J van Hilten, Department of Neurology, Leiden University Medical
Center, Leiden, The Netherlands
S Spieker, Department of Neurology, University of Tuebingen,
Tuebingen, Germany
S Pollentier, D Meier, Boehringer Ingelheim Pharma KG, Clinical
Research, Ingelheim, Germany
REFERENCES
1 Paulson HL, Stern MB. Clinical manifestations of Parkinson’s disease. In:
Watts RL, Koller WC, eds.
Movement disorders: neurologic principles
and practice
. New York: McGraw-Hill, 1997:183–99.
2 Oertel WH, Quinn N. Parkinsonism. In: Brandt T, Diener HC, Caplan LR,
et al
,eds.
Neurological disorders: course and treatment.
San Diego,
California: Academic Press, 1996:715–72.
3 Benabid AL, Pollak P, Gao DM,
et al
. Chronic electrical stimulation of
the ventralis intermedius nucleus of the thalamus as a treatment of
movement disorders.
J Neurosurg
1996;84:203–14.
4 Koller W, Pahwa R, Busenbark K,
et al
. High frequency unilateral
thalamic stimulation in the treatment of essential and parkinsonian tremor.
Ann Neurol
1997;42:292–9.
5 Krack P, Benazzouz A, Pollak P,
et al
. Treatment of tremor in Parkinson’s
disease by subthalamic nucleus stimulation.
Mov Disord
1998;13:907–14.
6 Limousin P, Krack P, Pollak P,
et al
. Electrical stimulation of the
subthalamic nucleus in advanced Parkinson’s disease.
N Engl J Med
1998;339:1105–11.
7 Mierau J, Schingnitz G. Biochemical and pharmacological studies on
pramipexole, a potent and selective dopamine D
2
receptor agonist.
Eur J
Pharmacol
1992;215:161–70.
8 Mierau J, Schneider FJ, Ensinger HA,
et al
. Pramipexole binding and
activation of cloned and expressed dopamine D2, D3 and D4 receptors.
Eur J Pharmacol
1995;290:29–36.
9 Piercey MF. Pharmacology of pramipexole, a dopamine D3-preferring
agonist useful in treating Parkinson’s disease.
Clin Neuropharmacol
1998;21:141–51.
10 Guttman M, and the International Pramipexole-bromocriptine Study
Group. Double-blind comparison of pramipexole and bromocriptine
treatment with placebo in advanced Parkinson’s disease.
Neurology
1997;49:1060–5.
11 Liebermann A, Ranhosky A, Korts D. Clinical evaluation of pramipexole
in advanced Parkinson’s disease: results of a double-blind,
placebo-controlled, parallel-group study.
Neurology
1997;49:162–8.
12 Parkinson Study Group. Safety and efficacy of pramipexole in early
Parkinson’s disease: a randomized dose-ranging study.
JAMA
1997;278:125–30.
13 Shannon KM, Bennett JP, Friedman JH,
et al
. Efficacy of pramipexole, a
novel dopamine agonist, as monotherapy in mild to moderate Parkinson’s
disease.
Neurology
1997;49:724–8.
14 Pinter MM, Pogarell O, Oertel WH. Efficacy, safety, and tolerance of
the non-ergoline dopamine agonist pramipexole in the treatment of
advanced Parkinson’s disease: a double blind, placebo controlled,
randomised, multicentre study.
J Neurol Neurosurg Psychiatry
1999;66:436–41.
15 Frucht S, Rogers JD, Greene PE,
et al
. Falling asleep at the wheel: motor
vehicle mishaps in persons taking pramipexole and ropinirole.
Neurology
1999;52:1908–10.
16 Pogarell O, Künig G, Oertel WH. A non-ergot dopamine agonist,
pramipexole, in the therapy of advanced Parkinson’s disease:
improvement of Parkinsonian symptoms and treatment associated
complications: a review of three studies.
Clin Neuropharmacol
1997;20:s28–35.
17 Künig G, Pogarell O, Möller JC,
et al
. Pramipexole, a non-ergot
dopamine agonist, is effective against rest tremor in intermediate to
advanced Parkinson’s disease.
Clin Neuropharmacol
1999;22:301–5.
18 Gibb WR, Lees AJ. The relevance of Lewy body to the pathogenesis of
idiopathic Parkinson’s disease.
J Neurol Neurosurg Psychiatry
1988;51:745–52.
19 Zimmermann R, Deuschl G, Hornig A,
et al
. Tremors in Parkinson’s
disease: symptom analysis and rating.
Clin Neuropharmacol
1994;17:303–14.
20 Boose A, Spieker S, Jentgens CH,
et al
. Assessing tremor severity with
long-term tremor recordings.
J Neurol Neurosurg Psychiatry
1994;57:397.
21 Spieker S, Jentgens C, Boose A,
et al
. Reliability, specificity and
sensitivity of long-term tremor recordings.
Electroencephalogr Clin
Neurophysiol
1995;97:326–31.
22 Factor SA, Sanchez-Ramos JR, Weiner WJ. Parkinson’s disease: an open
label trial of pergolide in patients failing bromocriptine therapy.
J Neurol
Neurosurg Psychiatry
1998;51:529–33.
23 Olanow CW, Fahn S, Muenter M,
et al
. A multicenter double-blind
placebo-controlled trial of pergolide as an adjunct to sinemet® in
Parkinson’s disease.
Mov Disord
1994;9:40–7.
24 Laihinen A, Rinne UK, Suchy I. Comparison of lisuride and
bromocriptine in the treatment of advanced Parkinson’s disease.
Acta
Neurol Scand
1992;86:593–5.
25 Spieker S, Löschmann P, Jentgens C,
et al
. Tremorlytic activity of
budipine: a quantitative study with long-term tremor recordings.
Clin
Neuropharmacol
1995;18:266–72.
26 Spieker S, Eisebitt R, Breit S,
et al
. Tremorlytic activity of budipine in
Parkinsons disease.
Clin Neuropharmacol
1999;22:115–19.
720 Pogarell, Gasser, van Hilten, et al
www.jnnp.com
... По данным отечественных авторов, АДР уменьшают дрожательный гиперкинез при БП на 29% [14]. Прамипексол, апоморфин и другие АДР могут усиливать эффект лево-допы, обеспечивая больший контроль тремора [30,31]. Их можно использовать в качестве начальной монотерапии или как адъювантное дополнение к леводопе. ...
... Их можно использовать в качестве начальной монотерапии или как адъювантное дополнение к леводопе. Добавление прамипексола к леводопе приводит к снижению показателей тремора примерно на 45% по UPDRS и значительно снижает частоту возникновения тремора в часы бодрствования, что регистрируется с помощью долговременной электромиографии [30]. ...
Article
Objective : to evaluate the efficacy of antiparkinsonian drugs of different groups considering the frequency-amplitude characteristics in the correction of tremulous hyperkinesis in Parkinson's disease (PD) using tremorography. Material and methods . 172 patients with a confirmed diagnosis of PD were observed. Taking into account age and contraindications, patients were divided into four groups receiving one of four drugs in monotherapy: dopamine receptor agonists (ADR; pramipexole), levodopa (Tidomet forte), anticholinergics (biperiden), amantadines (amantadine sulfate). Before starting treatment and after completion of the selected therapy, patients underwent tremorography to analyse the frequency, amplitude and duration of tremor over time in four standard tests. Results . All antiparkinsonian drugs reduced rest tremor, and the efficacy of pramipexole in treating tremor was comparable to that of levodopa in the early stages of the disease. As the severity of the disease increased, the efficacy of ADRs decreased, whereas levodopa became more important. Amantadine sulfate showed the lowest efficacy in rest tremor. The efficacy of anticholinergics against tremor is high but inconsistent. Conclusion . We demonstrated feasibility of using tremorography in the selection of an individualised treatment programme for patients with shaking hyperkinesis in PD , and the high efficacy of levodopa in shaking hyperkinesis was confirmed.
... Pramipexole, apomorphine, and other DAs can augment the effect of levodopa, hence, providing greater tremor control [63,64]. They can be used as an initial monotherapy, or as adjuvant addition to levodopa [50,65]. ...
... Both pramipexole and pergolide produce a similar degree of resting tremor suppression when used as monotherapy [66]. The addition of pramipexole to levodopa results in an estimated 45% reduction in the Unified Parkinson's Disease Rating Scale (UPDRS) tremor scores and significantly lower tremor occurrence during waking hours as recorded by longterm electromyography (EMG) [63]. Apomorphine is a potent, relatively short-acting DA that can be administered with a continuous subcutaneous infusion pump, or an intermittent sublingual, and subcutaneous injection [67,68]. ...
Article
Full-text available
Background: Tremor is one of the most prevalent symptoms in Parkinson's Disease (PD). The progression and management of tremor in PD can be challenging, as response to dopaminergic agents might be relatively poor, particularly in patients with tremor-dominant PD compared to the akinetic/rigid subtype. In this review, we aim to highlight recent advances in the underlying pathogenesis and treatment modalities for tremor in PD. Methods: A structured literature search through Embase was conducted using the terms "Parkinson's Disease" AND "tremor" OR "etiology" OR "management" OR "drug resistance" OR "therapy" OR "rehabilitation" OR "surgery." After initial screening, eligible articles were selected with a focus on published literature in the last 10 years. Discussion: The underlying pathophysiology of tremor in PD remains complex and incompletely understood. Neurodegeneration of dopaminergic neurons in the retrorubral area, in addition to high-power neural oscillations in the cerebello-thalamo-cortical circuit and the basal ganglia, play a major role. Levodopa is the first-line therapeutic option for all motor symptoms, including tremor. The addition of dopamine agonists or anticholinergics can lead to further tremor reduction. Botulinum toxin injection is an effective alternative for patients with pharmacological-resistant tremor who are not seeking advanced therapies. Deep brain stimulation is the most well-established advanced therapy owing to its long-term efficacy, reversibility, and effectiveness in other motor symptoms and fluctuations. Magnetic resonance-guided focused ultrasound is a promising modality, which has the advantage of being incisionless. Cortical and peripheral electrical stimulation are non-invasive innovatory techniques that have demonstrated good efficacy in suppressing intractable tremor.
... Among the 26 studies examining motor function with UPDRS, only 5 studies provided detailed information on the timing of levodopa administration in relation to the assessment. [37][38][39][40][41] These studies addressed a practically defined off and administration of a levodopa test-dose 2 hours before the assessment, respectively. In contrast, §Upgrading of the quality of evidence as the effect is larger than the minimally clinically important change. ...
Article
Full-text available
Background The aim of this manuscript is to review the evidence and compare the efficacy and safety of catechol-O-methyltransferase inhibitors (COMT-Is), dopamine receptor agonists (DRAs) and monoamine-oxidase B inhibitors (MAOB-Is) as adjunctive treatment to levodopa in patients with Parkinson’s disease (PD) experiencing motor complications. Methods In this systematic review and network meta-analysis, literature searches were performed in MEDLINE and Embase to identify eligible randomised controlled trials (RCTs) with a minimal follow-up of at least 4 weeks published in English between 1980 and 2021. RCTs were included if either a COMT-I, DRA or MAOB-I was evaluated as an adjunctive therapy to levodopa in patients with PD experiencing motor complications and dyskinesia. The main outcomes included daily off-medication time, motor and non-motor examination scales, and adverse events including dyskinesia. Results 74 RCTs reporting on 18 693 patients were included. All three studied drug classes decreased daily off-medication time compared with placebo (COMT-Is mean −0.8 hours (95% CI −1.0 to −0.6), DRAs −1.1 hours (95% CI −1.4 to −0.8), MAOB-Is −0.9 hours (95% CI −1.2 to −0.6)). Safety analysis showed an increased risk of dyskinesia for all three drug classes (COMT-Is OR 3.3 (95% CI 2.7 to 4.0), DRAs 3.0 (95% CI 2.5 to 3.5), MAOB-Is 1.6 (95% CI 1.2 to 2.2)). According to surface under the cumulative ranking curve scores, pramipexole IR was associated with the most favourable benefit–risk profile. Conclusions COMT-Is, DRAs and MAOB-Is effectively reduce motor complications and increase incidence of dyskinesia. In the network meta-analysis, adjunctive use of DRAs appeared most effective.
... Similarly, walking can be managed pharmacologically using dopamine agonists [25] and therapeutically with exercise [24]. Health domains were included if they were also potential side efects of treatments, such as fatigue [32]. ...
Article
Full-text available
Background: Parkinson's disease (PD) is a neurodegenerative condition, predominantly affecting older adults. Preference-based measures (PBMs) can be used to make decisions about the cost-utility of different treatments. There are currently no PBMs for health-related quality of life (HRQoL) for PD. A previous study identified important health domains for individuals with PD and developed an item pool from existing measures per domain. The current study aims to contribute to the development of a new disease-specific PBM of HRQoL for PD by reducing the current pool of items according to the preferences of individuals with PD. Methods: Fifty-three participants completed a visual analogue scale (VAS) of self-perceived health, the prototype PBM measure, and an item importance rating. To reduce the item pool, the following were calculated: (1) inter-item correlations; (2) impact of each item based on item performance and importance rating; (3) directionality of response options by comparing the VAS scores against each item. Results: Participants (male = 54.7%, age = 60.0 ± 10.2) had a median Hoehn and Yahr score of 2.5 (interquartile range = 1). Items supported for inclusion by this analysis were sleep, fatigue, tremor, mood, walking, memory, and dexterity. Items demonstrating a logical decrease in VAS score with each increasing severity level were sleep, memory, tremor, fatigue, and mood. Conclusion: This PBM will be critical for informing decisions about the cost-utility of PD treatments, guiding the resource allocation within our healthcare system. Future research will include cognitive debriefing with individuals with PD to refine item response options.
... Furthermore, combination therapy allows the L-dopa dose to be reduced (Biglan and Holloway 2002). Pramipexole has beneficial effects on motoric symptoms and can improve akinesia, rigor, and tremor (Möller et al. 2005;Pogarell et al. 2002). Some studies have shown that pramipexole has significantly less motoric late-stage complications in comparison to L-dopa (Biglan and Holloway 2002;PSG 2000), while another study demonstrated antidepressant effects . ...
Chapter
Many substance classes have been introduced for the treatment of motor symptoms in Parkinson’s disease within the last 50 years. The most important and effective medication is still levodopa in combination with a decarboxylase inhibitor. In addition, dopamine agonists, monoamine oxidase B (MAO-B) inhibitors, catechol-o-methyltransferase (COMT) inhibitors, amantadine (N-methyl-d-aspartate-antagonist), and less important anticholinergics are administered depending on the motor symptoms, age, and a multitude of other factors. Fortunately, within the substance classes there are various preparations, which also have different effects. In order that the advantages of the individual medications can be exploited and undesired effects and interactions can be avoided, one must come to terms with the complex data and the pharmacology of the medication. In addition, important aspects are interactions and reciprocal actions. The decision as to which medication to choose must be made individually for each patient.
Article
Introduction: Tremor is often perceived as severely disabling by patients with idiopathic Parkinson's disease (iPD) and yet ranges among the most difficult symptoms to treat. To date, no comprehensive analysis of non-lesional therapies to manage tremor in iPD exists to base recommendations upon. We therefore present a systematic literature review and meta-analysis assessing the efficacy/effectiveness and safety of non-lesional treatments for tremor in iPD. Methods: Three electronic databases were searched using a combination of title/abstract keywords complemented by hand-searching of reference lists. A random-effects meta-analysis of standardized mean change scores was conducted where appropriate. Results: 114 studies met inclusion criteria involving 8.045 patients. The meta-analysis revealed an overall reduction of standardized mean change scores by -0.93 [CI: -1.42; -0.43], p<0.001) by 14 different dopaminergic and non-dopaminergic classes of agents. No significant differences were identified between direct comparisons. Subgroup analysis comparing dopamine receptor agonists resulted in superior effects of pramipexole and rotigotine compared with ropinirole. There was little cumulative evidence to support the use of individual non-pharmacological interventions for tremor, except for electrical stimulation. Conclusions: The results of this meta-analysis suggest a large but nonspecific effect of established pharmacological therapies on tremor in iPD. Based on high-quality studies, there is sufficient evidence to support that levodopa, dopamine receptor agonists, and MAO inhibitors provide tremor relief in most patients, while evidence supporting other treatments is less well established. Sufficient evidence to draw conclusions on effects of non-lesional treatments in cases with refractory tremor is lacking.
Article
Full-text available
The pathophysiology of Parkinson’s disease (PD) tremor remains incompletely understood and there is a lack of clinical trials specifically addressing its pharmacological treatment. Levodopa is the most efficacious drug for most patients and should be used as primary approach to control troublesome tremor. While the efficacy of oral dopamine agonists on PD tremor has been demonstrated in controlled trials, there is no evidence of greater antitremor efficacy compared to levodopa. The magnitude of the antitremor effect of anticholinergics is generally lower than that of levodopa. Due to their adverse effects, anticholinergics have a limited role in selected young and cognitively intact patients. Propranolol may improve resting and action tremor and may be considered as an adjunct in patients with insufficient tremor response to levodopa and this also applies to clozapine, despite its unfavorable adverse effect profile. Treating motor fluctuations with MAO-B and COMT inhibitors, dopamine agonists, amantadine, or on-demand treatments such as subcutaneous or sublingual apomorphine and inhaled levodopa as well as with continuous infusions of levodopa or apomorphine will improve off period tremor episodes. For patients with drug-refractory PD tremor despite levodopa optimization deep brain stimulation and focused ultrasound are first-line considerations. Surgery can also be highly effective for the treatment medication-refractory tremor in selected patients without motor fluctuations. The present review highlights the clinical essentials of parkinsonian tremor, critically examines available trial data on the effects of medication and surgical approaches and provides guidance for the choice of treatments to control PD tremor in clinical practice.
Chapter
An amino benzothiazole derivative pramipexole is a non-ergoline, dopamine D2 receptor agonist. Pharmacologically, within the dopamine D2 receptor family (D2, D3, and D4) it has the highest affinity for D3 receptors and exhibits no affinity for D1 and D5 receptors. It has a higher affinity for D3 receptors than ropinirole. The usual dosage as a Parkinson’s disease (PD) treatment is 3.0–4.5 mg per day. D3 receptors are involved in the sensations and cognitive functions of the limbic midbrain, as well as the transmission of pain sensations in the spinal cord. As such pramipexole is thought to utilize these receptors to provide effective treatment of Restless Leg Syndrome.
Article
Full-text available
Sixty-three patients with Parkinson's disease who failed bromocriptine therapy for various reasons were treated in an open-label trial of pergolide. The data were evaluated in a retrospective manner. Forty-six percent had a good response and tolerated the pergolide. A comparison of the outcomes regarding response and toxicity revealed that bromocriptine and pergolide act differently in individual patients. A trial of pergolide in Parkinsonian patients failing bromocriptine therapy may be therapeutically useful.
Article
Pramipexole, a non-ergot dopamine D2/D3-receptor agonist, has been investigated in multicenter phase II and III studies as an add-on drug for patients receiving L-dopa therapy with advanced Parkinson's disease (PD) and motor (on/off) fluctuations. In three double-blind, placebo-controlled, multicenter studies, 507 patients were assigned to add-on treatment with pramipexole (n = 251) or placebo (n = 256). The relative improvement of the pramipexole group in Unified Parkinson's Disease Rating Scale parts II and III (activities of daily living and motor examination) was highly significant in all three trials. Reduction in part IV (complications of therapy) was significant in two trials (438 patients). Concomitant L-dopa medication was reduced by 25% under pramipexole vs. 51% under placebo (statistically significant difference). There were no unexpected adverse safety results. The adverse event profile revealed high tolerability (not dose-dependent) in the dosage range of 1.5-4.5 mg/day and an additional potential for efficacy in drug-resistant resting tremor. There was, however, a slightly higher incidence of dyskinesia with pramipexole than with placebo during the ascending dose interval but not in the maintenance phase. Pramipexole may be an efficacious therapeutic alternative for patients with advanced PD and motor fluctuations (or drug-resistant resting tremor).
Article
Pramipexole (SND 919; 2-amino-4,5,6,7-tetrahydro-6-propyl-amino-benzthiazole- dihydrochloride) was tested for its agonistic activity at pre- and postsynaptic dopamine (DA) receptors. L-Dihydroxyphenylalanine (L-dopa) accumulation in the rat striatum and limbic system and the alpha-methyltyrosine-induced reduction of DA were inhibited. Both effects were fully antagonized by haloperidol but not by the selective DA D1 receptor antagonist SCH 23390. Pramipexole decreased the levels of DA metabolites dose dependently, whereas striatal DA levels remained unchanged. In mice, pramipexole (0.001-1 mg/kg s.c.) reduced exploratory locomotor activity. In rats with unilateral striatal lesions, only weak ipsilateral rotation was produced by pramipexole at the highest dose. However, in rats with unilateral lesions of the medial forebrain bundle, pramipexole potently induced contralateral circling (ED50 0.026 mg/kg s.c.). In the N-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) monkey model, pramipexole also had potent stimulatory effects. Finally, in haloperidol-sensitized monkeys, the substance did not elicit dyskinesia/dystonia when given alone, but rather inhibited those symptoms which had been induced by haloperidol (ED50 0.116 mg/kg i.m.). It is concluded that pramipexole has therapeutic potential for schizophrenic patients, as a result of its autoreceptor agonistic effects and its weak effects at normosensitive postsynaptic DA receptors. Furthermore, its potent stimulatory effects in DA-depleted animals suggest a possible use in the treatment of Parkinson's disease.
Article
Twenty patients with advanced idiopathic Parkinson's disease were studied, all having a deteriorating response to levodopa and suffering from daily fluctuations in disability. A double-blind randomized cross-over study was conducted. Basic levodopa and anticholinergic treatment was continued unchanged in all patients. The dose increment period of 4-8 weeks was followed by a 4 week treatment period on a fixed optimal dose. In both treatment groups the mean optimal daily dose of lisuride was 1.3 mg (range 0.2-2.4 mg) and that of bromocriptine about 15 mg (range 3.75-30.0), without any significant differences between the treatment groups. The addition of lisuride or bromocriptine to levodopa treatment resulted in a significant and equal further improvement of parkinsonian disability. The therapeutic profiles of both lisuride and bromocriptine were similar. There was significantly more improvement in tremor than in other parkinsonian symptoms. Both lisuride and bromocriptine elicited a significant improvement in fluctuations of disability. No significant differences between the treatments were observed. The occurrence of clinical side effects seemed to be similar with both treatment regimens. In advanced parkinsonian patients the therapeutic efficacy of lisuride seems to be equal to that of bromocriptine as far as parkinsonian disability and fluctuations in disability are concerned.
Article
The Lewy body is a distinctive neuronal inclusion that is always found in the substantia nigra and other specific brain regions in Parkinson's disease. It is mainly composed of structurally altered neurofilament, and occurs wherever there is excessive loss of neurons. It occurs in some elderly individuals and rarely in other degenerative diseases of the central nervous system. In 273 brains of patients dying from disorders other than Parkinson's disease, the age-specific prevalence of Lewy bodies increased from 3.8% to 12.8% between the sixth and ninth decades. Associated pathological findings suggest that these cases of incidental Lewy body disease are presymptomatic cases of Parkinson's disease, and confirm the importance of age (time) in the evolution of the disease. In view of the common and widespread occurrence of this disorder we propose that endogenous mechanisms operating in early life may be more important than environmental agents in the pathogenesis of Lewy bodies and Parkinson's disease.
Article
Pramipexole (SND 919; 2-amino-4,5,6,7-tetrahydro-6-propylamino-benzthiazole-dihydrochlor ide) is a potent dopamine autoreceptor agonist. We have carried out an analysis of the binding affinities of dopamine D2L, D2S, D3, and D4 receptors for pramipexole using both [3H]pramipexole and [3H]spiperone as radioligands at cloned and heterologously expressed receptors. Studies were carried out using rat and human D2L, D2S and D3 receptors with equivalent results. When the binding of pramipexole to the high affinity, guanine nucleotide-sensitive state of each receptor was analyzed, pramipexole is most selective for D3 compared to D2 and D4 receptors. These results indicate a 5-fold selectivity of pramipexole for D3 receptors, while quinpirole and bromocriptine are non-selective or more D2/D4 receptor selective. Two measurements of receptor activation for dopamine D2, D3, and D4 receptors also show that pramipexole is most potent for activation of D3 receptors. The dopamine D3 receptor selectivity of pramipexole may explain the previously described properties of this drug, including its potent autoreceptor preference.
Article
Three hundred and seventy-six subjects with advanced Parkinson's disease participated in a prospective, double-blind placebo-controlled study of the dopamine agonist pergolide mesylate as an adjunct to Sinemet. At 6 months, patients randomized to pergolide had a statistically significant improvement in total Parkinson's score, scores of activities of daily living, motor function, number of "off" hours, Hoehn and Yahr stage, and numerous parameters of parkinsonian function including bradykinesia, rigidity, gait, and dexterity. This benefit was obtained with the addition of a mean dose of 2.94 mg of pergolide, which permitted a 24.7% reduction in dose of levodopa. Adverse reactions were, for the most part, mild, reversible, and not of major clinical significance. No significant cardiac or electrocardiographic abnormalities were detected. This study demonstrates that pergolide mesylate, as an adjunct to levodopa, is an effective antiparkinsonian agent that provides clinical improvement while permitting a reduction in levodopa dose.
Article
We have developed a method of long-term EMG recording that has proven suitable for the quantification of pathological tremor. In the present paper we show that the principal parameters of the method (tremor occurrence, tremor intensity, tremor frequency) are highly reproducible and that the method is specific and sensitive for detection of pathological tremor. Twelve patients with essential tremor (ET) and 13 patients with Parkinson's disease (PD) were recorded repeatedly on 3 successive days. For each patient group and for each parameter the intersubject variability was much larger than the intersubject variability. The intraclass correlation coefficient "R" was in the order of 0.9 for each parameter and the mean of Pearson's correlation coefficient between successive days was also approximately 0.9. Recordings from normal controls demonstrated that the method's specificity for pathological tremor is 94.1% and its sensitivity is 96%.