Download as pdf or txt
Download as pdf or txt
You are on page 1of 7

RESEARCH ARTICLE

A Randomized, Double-Blind, Placebo-Controlled Trial of Safinamide


as Add-on Therapy in Early Parkinson’s Disease Patients
Fabrizio Stocchi, MD,1* Rupam Borgohain, DM,2 Marco Onofrj, MD,3 Anthony H.V. Schapira, MD,4
Mohit Bhatt, MD,5 Valentina Lucini, MD,6 Rodolfo Giuliani, MD,6 Ravi Anand, MD,7 and for the Study 015 Investigators8

1
Department of Neuroscience, IRCCS San Raffaele, Rome, Italy
2
Department of Neurology, Nizam’s Institute of Medical Sciences, Hyderabad, India
3
Department of Neurology, Chieti-Pescara University, Chieti, Italy
4
Department of Clinical Neurosciences, UCL Institute of Neurology, London, United Kingdom
5
Movement Disorder Clinic, Mumbai, India
6
Development Department, Newron Pharmaceuticals, Milan, Italy
7
APC, St. Moritz, Switzerland
8
Investigators are listed at the end of the manuscript.

A B S T R A C T : Safinamide is an a-aminoamide with ference between safinamide 200 mg and placebo was
both dopaminergic and nondopaminergic mechanisms not significant [point estimate: 20.4; 95% confidence
of action evaluated as an add-on to dopamine agonist interval (CI): 22.3–1.4; P 5 0.6504]. Although the differ-
(DA) therapy in early-stage PD. In this 24-week, double- ence between 100 mg/day and placebo was significant
blind study, patients with early PD receiving a stable (point estimate: 21.9; 95% CI: 23.7 to 20.1; P 5
dose of a single DA were randomized to once-daily 0.0419), these results are considered exploratory. No
safinamide 100 mg, safinamide 200 mg, or placebo. clinically meaningful differences from placebo were
The primary efficacy variable was UPDRS part III (motor observed for any safety variables. This study did not
examination) total score. Analysis was hierarchical: demonstrate a significant improvement of the primary
200 mg of safinamide versus placebo was tested first; endpoint for safinamide 200 mg/day. Exploratory analy-
the success of safinamide 100 mg versus placebo was sis of the primary endpoint for 100 mg/day demon-
contingent on this. Two hundred sixty-nine patients strated that the addition of safinamide to a stable dose
received safinamide 100 mg (n 5 90), safinamide of DA improves motor symptoms in early PD and war-
200 mg (n 5 89), or placebo (n 5 90); 70, 81, and 81 rants further investigation. V
C 2011 Movement Disorder

patients, respectively, completed the study. Mean Society


improvements from baseline to week 24 in UPDRS III
total scores were 23.90 for safinamide 200 mg, 26.0 Key Words: safinamide; Parkinson’s disease; add-on;
for safinamide 100 mg and 23.60 for placebo. The dif- motor function; a-aminoamide; dopamine

------------------------------------------------------------------------------------------------------------------------------
*Correspondence to: Dr. Fabrizio Stocchi, Department of Neuroscience, IRCCS San Raffaele, Via Della Pisana, 235, 00163 Rome, Italy; fabstocc@tin.it

Funding agencies: The sponsor, Newron Pharmaceuticals S.p.A., was responsible for the study design, investigator selection, general oversight of the
trial, and identification of the contract research organization responsible for day-to-day study management, monitoring, data management, and statistical
analysis. Professor Pablo Martinez-Martin provided training for the UPDRS. The interpretation of the data and preparation of the manuscript were
performed by the authors with the assistance of an independent medical writing agency, funded by Merck Serono. The decision to submit to Movement
Disorders was made by the authors.

Relevant conflicts of interest/financial disclosures: F.S. and all the other participants of the clinical study received payment according to the number
of patients included in the trial. F.S. has received honoraria for educational symposia and consultancy for Newron and Merck Serono. A.H.V.S. has
received honoraria from Merck Serono for medical education symposia and for providing advice in the development of safinamide. R.A. has received
honoraria for acting as a consultant for Newron Pharmaceuticals in the development of safinamide, in the conception and organization of this clinical trial,
in the statistical design and critical review of the analysis, and in the conception, review, and critique of the manuscript for this article. M.O. has received
honoraria for acting as a consultant for Newron Pharmaceuticals and from Merck Serono for an educational symposium. V.L. and R.G. are employees of
Newron. R.B. and M.B. have nothing else to disclose.

Full financial disclosures and author roles may be found in the online version of this article.
Received: 25 March 2011; Revised: 29 July 2011; Accepted: 14 August 2011
Published online 12 September 2011 in Wiley Online Library (wileyonlinelibrary.com). DOI: 10.1002/mds.23954

106 Movement Disorders, Vol. 27, No. 1, 2012


S A F I N A M I D E A S A D D - O N T H E R A P Y I N E A R L Y P D

Dopamine agonists (DAs) are commonly used as ini- dose wearing off, on/off phenomena, disabling dyski-
tial therapy in patients with newly diagnosed PD,1–3 nesia, or wide fluctuations; previous safinamide treat-
because they are associated with a low risk of motor ment; history or current diagnosis of psychosis or
complications, such as dyskinesia.4,5 The efficacy of depression; hypersensitivity to anticonvulsants or anti-
DAs declines with disease progression, and most parkinsonian agents; severe postural hypotension; use
patients eventually require additional therapy.6 Fur- of other PD medications (except for a single DA) dur-
thermore, DAs may be associated with side effects, ing the 4 weeks before screening; or concomitant
such as somnolence, peripheral edema, cognitive dis- use of MAO inhibitors. Based on its selectivity for
turbances, and impulse control disorders (ICDs).7,8 MAO-B and results indicating a lack of tyramine
Some of these may be improved by a reduction in potentiation by safinamide, there were no restrictions
dose, although at the risk of a decline in motor con- on tyramine intake.14
trol. Levodopa (L-dopa) is commonly added to DAs
when an improvement in motor function is required.
However, once L-dopa therapy is initiated, the risk of Procedures
dyskinesia is similar to that for patients initially After a 10-day screening and run-in period, eligible
treated with L-dopa.9,10 The addition of alternative patients were randomized using a computer-generated
adjunct therapy to DAs is an attractive strategy code (1:1:1) to receive safinamide 100 mg/day,
that may allow the use of a lower dose of DA while 200 mg/day, or placebo as add-on therapy to a single
maintaining efficacy and delaying the introduction of DA. An interactive voice-response system was used to
L-dopa. assign blinded medication and make dose changes.
Safinamide is an a-aminoamide with both dopami- The patients, caregivers, and raters remained blinded
nergic and nondopaminergic mechanisms of action, to the identity of the assigned medications throughout
including monoamine oxidase-B (MAO-B) and dopa- the study. Randomization was carried out on a coun-
mine reuptake inhibition, activity-dependent sodium- try-specific basis; a minimization procedure was used
channel antagonism, and inhibition of glutamate to ensure that the range of Hoehn and Yahr scores
release in vitro.11–13 were evenly distributed across treatment groups. Daily
This article reports on the results of a phase III doses were achieved using combinations of 50-mg cap-
trial (Study 015) evaluating the efficacy, safety, and sules of safinamide and matching placebo capsules
tolerability of safinamide as add-on therapy in early (both manufactured by MiPharm S.p.A., Milan, Italy),
PD patients treated with a stable dose of a single with 4 capsules being taken orally, once-daily, with
DA. breakfast.
Patients assigned to 100 mg/day of safinamide
received 50 mg/day for 2 weeks, before being titrated
Patients and Methods up to their target dose of 100 mg/day, whereas
patients assigned to 200 mg/day of safinamide initially
Patients received 100 mg/day and had their dose increased to
This 24-week, randomized, double-blind, placebo- 150 mg/day on day 7, and to the target dose of
controlled, parallel-group trial was conducted at 200 mg/day on day 14. Dose reductions were permit-
26 centers in Italy, Spain, the United Kingdom, India, ted for patients who could not tolerate their assigned
Argentina, Chile, and Colombia. The study protocol dose. The dose of DA was to remain constant
was approved by all ethics committees and regulatory throughout the trial; however, a dose increase or addi-
authorities. The study was conducted in accord with tion of another PD medication was permitted for
the International Conference on Harmonization Good patients who were receiving the maximum dose
Clinical Practice guidelines and the Declaration of of study medication, but whose motor symptoms
Helsinki. All patients provided written informed con- were worsening. All final efficacy and safety evalua-
sent before their participation. tions were performed before this intervention. A
Eligible patients were 30 to 80 years in age, with a decrease in the dose of DA was permitted, if clinically
clinical diagnosis of idiopathic PD <5 years in dura- indicated.
tion, and Hoehn and Yahr stage I to III. Patients were Patients returned for scheduled visits at 2, 4, 8, 12,
required to be on a stable dose of a single DA for at 18, and 24 weeks. Patients who completed 24 weeks
least 4 weeks before screening. Recruitment was car- of treatment, and those who discontinued prematurely
ried out by the principal and coinvestigators at each but returned for scheduled efficacy evaluations at
center and took place between December 21, 2004 weeks 12 and 24, were eligible to continue treatment
and July 22, 2005; the last patient completed the trial in a 52-week, double-blind, extension study (Study
on January 23, 2006. 017; NCT00642889).
Exclusion criteria included the following: current Blood samples were taken before the administration
diagnosis or recent history of substance abuse; end-of- of the first dose of study medication, 5 hours

Movement Disorders, Vol. 27, No. 1, 2012 107


S T O C C H I E T A L .

postdosing, and at each subsequent visit for the mea- The primary efficacy analysis consisted of two com-
surement of plasma concentrations of safinamide and parisons: safinamide 200 mg versus placebo followed
its main metabolites (i.e., safinamide acid and N-deal- by safinamide 100 mg versus placebo, with the second
kylated acid) using liquid chromatography with tan- comparison being dependent on the success of the
dem mass spectrometry (LC-MS/MS). first. If both primary efficacy comparisons were signifi-
The primary efficacy measure was the change in cant, then the secondary endpoints were analyzed in a
UPDRS part III (motor examination) total score from hierarchical order in the same sequence as the dose
baseline to endpoint (week 24). Secondary efficacy comparisons for the primary endpoint. If any of the
measures included the following: responder analysis, tests were not significant (alpha ¼ 0.05), then the sub-
that is, the proportion of patients showing improve- sequent tests were considered exploratory analyses.
ment on the Clinical Global Impression–Change from The analysis of change from baseline in UPDRS part
baseline (CGI-C) at endpoint and the proportion of III total score between safinamide 200 mg versus pla-
patients with at least 30% improvement on UPDRS cebo and safinamide 100 mg versus placebo was per-
part III from baseline to endpoint with no worsening formed using a mixed linear model analysis of
of UPDRS part II (activities of daily living; ADL) and covariance (ANCOVA), with treatment, visit, and
part IV (complications of therapy) total scores; and treatment by visit interaction as fixed effects, baseline
mean change from baseline to endpoint on the UPDRS UPDRS part III total score as covariate, and country
part II total score and for CGI-C and CGI–Severity of as a random effect. The primary efficacy analysis was
illness (CGI-S). Mean changes from screening/baseline performed without imputation of missing values. Point
to endpoint on the total scores for the tertiary efficacy estimates and 95% confidence intervals (CIs) for the
variables, Hamilton Rating Scale for Depression mean difference in UPDRS part III total scores were
(HAMD),15 UPDRS part I (mentation, behavior, and calculated. Statistical comparisons were two-sided and
mood), UPDRS part IV, and Mini–Mental State Ex- were conducted at the 5% significance level.
amination (MMSE), were also analyzed. A secondary efficacy analysis of change from base-
Safety was assessed by the incidence of adverse line in UPDRS part III total score between safinamide
events (AEs) reported by the patient or observed by 200 mg versus placebo and safinamide 100 mg versus
the investigator (at all postscreening visits), vital signs placebo was performed for the intent-to-treat popula-
(at all visits), laboratory tests (at screening and all tion using the following imputation schemes: last ob-
postbaseline visits), 12-lead electrocardiogram (ECG; servation carried forward (LOCF); observed case
at screening, baseline, and 12 and 24 weeks), and (OC); retrieved dropout (RDO); and OC and RDO.
physical and neurologic examinations (at screening Subsequent secondary analyses of UPDRS parts II, III,
and 24 weeks). All safety data were monitored by an and IV and CGI scores utilized LOCF or OC imputa-
independent safety monitoring board. tion schemes, whereas tertiary analyses implemented
The trial is registered with the U.S. National Insti- LOCF only.
tutes of Health clinical trials database (ClinTrials.gov; Mean changes from baseline to endpoint for tertiary
no. NCT00643045). Two amendments to the trial variables were analyzed using an ANCOVA model,
protocol took place after commencement; the first with baseline value as covariate and treatment group
introduced additional measures to monitor the ocular and country as main effects. The proportion of
safety of patients, and the second was to designate patients showing improvement on the CGI-C or meet-
UPDRS part III as the only primary outcome. ing responder criteria for the UPDRS was analyzed
using the Cochran-Mantel-Haenszel method, weighted
by country. The Wilcoxon rank sum test was used to
Statistical Analysis
compare mean CGI-C scores at endpoint.
Two primary analysis populations were defined: Safety data were summarized for all patients by
intent to treat (all randomized patients) and the safety treatment group using appropriate summary statistics.
population (all patients who received at least one dose
of study medication and had a postbaseline safety
assessment). The sample size was calculated based on Results
a single primary endpoint, the UPDRS part III, and
assumed a two-tailed probability of type I error of Patient Disposition and Baseline
0.05. The planned sample size of 192 subjects had a Characteristics
90% power to detect a difference of 3.3 points in the A total of 270 patients were randomized (Fig. 1).
UPDRS part III, assuming an estimated standard devi- The first patient was enrolled on December 21, 2004,
ation (SD) of 5.5. Based on an anticipated drop-out and the last patient completed the study on January
rate of 20% during treatment, and a screen failure 23, 2006. All patients were receiving a single dopa-
rate of 10%, at least 267 patients were to be screened mine agonist at a stable dose for 4 weeks before
to ensure a sufficient number of randomized patients. screening, except 1 patient who had received a dose

108 Movement Disorders, Vol. 27, No. 1, 2012


S A F I N A M I D E A S A D D - O N T H E R A P Y I N E A R L Y P D

TABLE 1. Baseline Characteristics for the


Intent-to-Treat/Safety Population

Safinamide Safinamide
200 mg/day 100 mg/day Placebo
Characteristic (n ¼ 89) (n ¼ 90) (n ¼ 90)

Age (yr) 58.5 (11.7) 56.5 (11.3) 57.3 (10.8)


Males (%) 54 (61) 59 (66) 56 (62)
Duration of PD (yr) 2.30 (1.32) 2.64 (1.42) 2.41 (1.20)
Race
White (%) 52 (58) 51 (57) 55 (61)
Asian (Indian) (%) 35 (39) 35 (39) 34 (38)
Weight (kg) 68.0 (12.5) 72.3 (13.8) 69.3 (13.9)
Hoehn and Yahr stage (%)
I 18 (20) 14 (16) 10 (11)
I.5 11 (12) 16 (18) 17 (19)
II 43 (48) 45 (50) 48 (53)
II.5 14 (16) 10 (11) 10 (11)
III 3 (3) 5 (6) 5 (6)
IV 0 0 0
V 0 0 0
CGI–Severity of illness 3.1 (0.85) 3.1 (0.79) 3.1 (0.76)
UPDRS part I (mentation, 1.3 (1.23) 1.2 (1.26) 1.3 (1.24)
behavior, and mood)
UPDRS part II (ADL) 7.3 (4.71) 8.2 (4.92) 8.1 (5.32)
UPDRS part III (motor) 19.3 (9.80) 22.0 (10.15) 20.7 (9.63)
UPDRS part IV 0.4 (0.83) 0.4 (0.73) 0.6 (0.95)
(complications
of therapy)
MMSE total score 28.3 (1.54) 28.9 (1.21) 28.4 (1.56)
HAMD total score 4.2 (3.11) 4.0 (3.43) 4.3 (3.22)

FIG. 1. Study profile. Data are presented as mean (SD) or number (%) of patients.
ADL, activities of daily living; CGI, Clinical Global Impression; HAMD,
Hamilton Rating Scale for Depression; MMSE, Mini–Mental State
Examination; SD, standard deviation.
adjustment 3 weeks before screening. The majority of
patients (>90%) reached the target doses of 100 or
200 mg/day and remained at that dose throughout the cebo showed improvement from baseline to week 24 in
study. Withdrawal of consent and AEs were the most UPDRS part III total score (6.0 versus 3.6; point
common reasons for premature withdrawal from the estimate: 1.9; 95% CI: 3.7 to 0.1; P ¼ 0.0419).
study.
There were no meaningful differences among treat-
ment groups in any of the key demographic and base- TABLE 2. Concomitant Medication for PD
line characteristics (Table 1), except for the finding (Intent-to-Treat Population)
that the mean UPDRS part III score at baseline was
Safinamide Safinamide
2.7 points higher in the safinamide 100 mg/day group,
200 mg/day 100 mg/day Placebo
compared with the 200 mg/day group (not Medication (n ¼ 89) (n ¼ 90) (n ¼ 90)
significant).
Dopamine agonist usea:
Bromocriptine (%) 10 (11) 16 (22) 13 (14)
Efficacy Cabergoline (%) 3 (3) 9 (10) 9 (10)
Lisuride (%) 3 (3) 0 0
The primary efficacy endpoint of mean change from Pramipexole (%) 27 (30) 21 (23) 23 (26)
baseline in UPDRS part III total score at week 24 was Pergolide (%) 2 (2) 1 (1) 0
3.9 (SD 6 6.01), 6.0 (SD 6 7.18), and 3.6 Piribedil (%) 5 (6) 2 (2) 4 (4)
(SD 67.08) for safinamide 200 mg, safinamide Ropinirole (%) 40 (45) 43 (48) 41 (46)
Dopa and dopa 1 (1) 2 (2) 1 (1)
100 mg, and placebo, respectively (Fig. 2). For the first derivatives (%)
comparison of safinamide 200 mg versus placebo, Amantadine (%) 0 1 (1) 1 (1)
there was no significant difference in change from Anticholinergic agents (%) 2 (2) 2 (2) 1 (1)
baseline to week 24 in UPDRS part III total score
between treatment groups (3.9 versus 3.6; point a
Concomitant dopamine agonist use for PD includes any dopamine agonist
that was started on or after the first administration of safinamide and any
estimate: 0.4; 95% CI: 2.3–1.4; P ¼ 0.65). The dopamine agonist started before the study, but continued after safinamide
second comparison of safinamide 100 mg versus pla- administration.

Movement Disorders, Vol. 27, No. 1, 2012 109


S T O C C H I E T A L .

FIG. 2. Mean change from baseline in the UPDRS part III total scores–mixed linear model (intent-to-treat population).

Similar results for UPDRS part III total scores were pain (upper), vomiting, pyrexia, cough, hypertension,
obtained using the alternative imputation schemes. blurred vision, gastritis, peripheral edema, nasophar-
The proportion of responders at week 24 in each yngitis, dizziness, back pain, and tremor. The inci-
treatment group is shown in Figure 3. A greater propor- dence of these TEAEs was <10% in each group.
tion of patients in the safinamide 200 mg and 100 mg TEAEs were generally of mild or moderate intensity.
groups experienced benefit, compared with those in the The number of patients with TEAEs rated as severe
placebo group (P < 0.05 for CGI-C responders in was 2 (2.2%) for the safinamide 100 mg group,
the OC analysis). 9 (10.1%) for the safinamide 200 mg group, and
Mean (6 SD) CGI-C from baseline score at week 24 6 (6.7%) for the placebo group. Of these, incidences
for the safinamide 200 mg and 100 mg groups and of blurred vision and leg pain in the safinamide
placebo groups were 3.1 (0.98; n ¼ 70), 3.1 (1.06; 200 mg group and vomiting and dizziness in the safi-
n ¼ 80), and 3.4 (1.04; n ¼ 82), respectively, indicating namide 100 mg group were considered to be related
an improvement in CGI score for both active treatment to treatment. Four patients experienced serious
groups versus placebo (P ¼ 0.0325 and P ¼ 0.0293
for safinamide 200 mg and 100 mg versus placebo,
respectively). The CGI-C from baseline score and CGI-S
mean change at endpoint were not statistically different
between groups.
Mean change from baseline to week 24 in UPDRS
part II indicated an improvement in ADL for the safi-
namide 100 mg group versus placebo (2.2 versus
1.2, respectively; P ¼ 0.0248); there was no signifi-
cant difference between the safinamide 200 mg group
and placebo (1.4 versus 1.2, respectively). Changes
from baseline to week 24 in HAMD, UPDRS I,
UPDRS IV, and MMSE did not show a significant
benefit for treatment with safinamide.

Safety
The most common treatment-emergent adverse FIG. 3. Secondary efficacy variables: responders at endpoint (week 24;
events (TEAEs) were nausea, headache, abdominal intent-to-treat population).

110 Movement Disorders, Vol. 27, No. 1, 2012


S A F I N A M I D E A S A D D - O N T H E R A P Y I N E A R L Y P D

TEAEs. These included retinal vein occlusion and In addition, analyses of safinamide 100 mg/day dem-
iron-deficiency anemia (100 mg group) and gastroen- onstrated improvements in CGI-C and UPDRS part II
teritis (200 mg group). There was one death during (ADL) scores. The proportion of patients considered
the study (gunshot wound). None of these serious to be ‘‘responders’’ was higher in both safinamide
TEAEs were considered by the investigators to be 100 mg and 200 mg groups, compared with the pla-
related to the study medication. Overall, 21.3% of cebo group.
patients discontinued treatment in the safinamide The reason for the lack of efficacy of the higher
200 mg group, compared with 10.0% each for safina- dose of safinamide is unknown. The baseline demo-
mide 100 mg and placebo. Six patients discontinued graphics and characteristics of the two safinamide
treatment as a result of TEAEs: 1 in the safinamide groups were similar. It is possible that the higher inci-
100 mg group, 3 in the safinamide 200 mg group, and dence of discontinuations in the safinamide 200 mg
2 in the placebo group. These were all considered to group (21.3% versus 10.0% each for safinamide
be possibly or probably related to study medication 100 mg and placebo) may have prevented the demon-
and included nausea/anxiety, head tremor/nausea/ stration of a significant clinical benefit. It is also possi-
gastroesophageal reflux disease, hypotension/diarrhea/ ble that the higher rate of discontinuation in the
vomiting (200 mg group), parasthesia (100 mg group), safinamide 200 mg group was the result of a lack of
and nausea and headache/dystonia (placebo group). efficacy; only 2 patients in this group were specifically
The incidence of clinically notable values for labora- withdrawn because of lack of efficacy, but 8 patients
tory parameters and vital signs was comparable were withdrawn for withdrawal of consent/other rea-
between groups. sons, and the possibility that these reflect a lack of ef-
ficacy cannot be ruled out. It has previously been
shown that different results may be obtained with dif-
Pharmacokinetics ferent doses of an MAO-B inhibitor.15 Safinamide has
both dopaminergic (including MAO-B inhibition) and
Plasma concentrations in both safinamide groups nondopaminergic mechanisms of action.11–13
were consistent with the dose level and showed a lin- In this study, an improvement in UPDRS part III
ear dose exposure and a relationship between dose total scores was observed in the placebo group, an
groups. However, in the blood samples from patients effect that may have contributed to the lack of efficacy
in the placebo group, safinamide and its metabolites observed with safinamide 200 mg/day. The influence
were detected in 155 of 588 (26%) plasma samples of placebo in clinical trials in PD patients that investi-
taken from 79 of 90 patients randomized to placebo. gate motor functional endpoints has been previously
This indicates that patients in the placebo group were documented.16 This influence may be caused by the
exposed, at some visits, to a certain amount of safina- release of neurotransmitters, such as dopamine, or
mide and this exposure was most frequently detected other physiological mechanisms, which may obscure
at weeks 8 and 12 (78% and 58% of patients, respec- the effects of the investigational therapy.16 The devel-
tively). At these time points, the average safinamide opment of rating scales of motor function that are
concentration in placebo samples with measurable lev- able to discern the effects of placebo would be a criti-
els was approximately 20% of that observed in sam- cal advantage for future clinical trials investigating do-
ples from patients receiving safinamide 100 mg. An paminergic therapies.17
audit revealed that 2 of 51 bulk placebo bottles con- Safinamide treatment was associated with a low
tained a mixture of placebo and safinamide capsules. incidence of serious TEAEs and discontinuations
No errors in randomization, labeling, or dispensing resulting from AEs. In addition, the incidence of the
were detected. most common AEs was low (<10%) and comparable
between the three groups, and the proportion of
patients with serious AEs was lowest in the safinamide
Discussion 100 mg group (2.2%). The extension study (Study
017) will provide additional safety data for safinamide
This phase III, randomized, double-blind, placebo- as add-on to DA therapy.
controlled study compared two once-daily oral dose Pharmacokinetic analysis revealed that some
ranges of safinamide with placebo as add-on therapy patients in the placebo group inadvertently received
to a stable dose of a single DA in patients with early study medication, because safinamide was detected in
PD. In the primary efficacy analysis, this study failed plasma at various time points. At the time points in
to demonstrate a significant difference between safina- which the greatest safinamide exposure was detected,
mide 200 mg/day and placebo for the change in motor the average concentrations in samples with measurable
symptoms. The subsequent exploratory analysis indi- safinamide did not exceed 20% of the levels in plasma
cated a significant improvement in UPDRS part III from patients randomized to the 100 mg/day dose
total scores for safinamide 100 mg/day versus placebo. group. These findings need to be considered when

Movement Disorders, Vol. 27, No. 1, 2012 111


S T O C C H I E T A L .

interpreting the results of the placebo group for this 2. Horstink M, Tolosa E, Bonuccelli U, et al. Review of the therapeu-
tic management of Parkinson’s disease. Report of a joint task force
study. However, the effects are likely to be minor, and of the European Federation of Neurological Societies and the
it is possible that differences between safinamide and Movement Disorder Society-European Section. Part I: early
(uncomplicated) Parkinson’s disease. Eur J Neurol 2006;13:
placebo may have been greater if the placebo group 1170–1185.
had remained free from active drug. 3. NICE. Parkinson’s disease diagnosis and management in primary
In a previous study in early PD, safinamide 1 mg/kg and secondary care. 2006. Available at: http://www.nice.org.uk/
nicemedia/live/10984/30088/30088.pdf. Accessed on July 2, 2010.
(median dose: 70 mg/day) was associated with signifi-
4. Rascol O, Brooks DJ, Korczyn AD, et al. A five-year study of the
cant improvements in motor function.18,19 In the cur- incidence of dyskinesia in patients with early Parkinson’s disease
rent study, treatment with safinamide 100 mg/day was who were treated with ropinirole or levodopa. N Engl J Med
2000;342:1484–1491.
associated with improvements in motor symptoms and
5. Holloway RG, Shoulson I, Fahn S, et al. Pramipexole vs levodopa
ADL, with no clinically meaningful differences from as initial treatment for Parkinson disease: a 4-year randomized
placebo for any safety variables. These findings sug- controlled trial. Arch Neurol 2004;61:1044–1053.
gest that the addition of safinamide 100 mg/day to 6. Bonuccelli U, Pavese N. Dopamine agonists in the treatment of
Parkinson’s disease. Expert Rev Neurother 2006;6:81–89.
DA therapy may be an effective treatment strategy
7. Singh A, Kandimala G, Dewey RB, Jr., O’Suilleabhain P. Risk
that could allow physicians to improve motor func- factors for pathologic gambling and other compulsions among
tion, prolong the use of DAs, and/or delay the intro- Parkinson’s disease patients taking dopamine agonists. J Clin Neu-
rosci 2007;14:1178–1181.
duction of L-dopa. Further studies are required to
8. Simuni T, Lyons KE, Pahwa R, et al. Treatment of early Parkin-
confirm this; indeed, safinamide is being evaluated fur- son’s disease. Part 2. Eur Neurol 2009;61:206–215.
ther in ongoing phase III trials, including a random- 9. Constantinescu R, Romer M, McDermott MP, Kamp C, Kieburtz
ized, double-blind study evaluating the efficacy and K. Impact of pramipexole on the onset of levodopa-related dyski-
nesias. Mov Disord 2007;22:1317–1319.
tolerability of safinamide 50 or 100 mg/day as add-on
10. Rascol O, Brooks DJ, Korczyn AD, et al. Development of dyskine-
therapy to a single DA (MOTION; NCT00605683). sias in a 5-year trial of ropinirole and L-dopa. Mov Disord 2006;
21:1844–1850.
11. Pevarello P, Bonsignori A, Dostert P, et al. Synthesis and anticon-
Conclusion vulsant activity of a new class of 2-[(arylalky)amino]alkanamide
derivatives. J Med Chem 1998;41:579–590.
12. Caccia C, Maj R, Calabresi M, et al. Safinamide: from molecular
Further investigation is important to evaluate the targets to a new anti-Parkinson drug. Neurology 2006;67:
potential benefits of safinamide add-on to DAs. It has S18–S23.
been shown that the risk of developing dyskinesia in 13. Caccia C, Salvati P, Rossetti S, Anand R. Safinamide: beyond
MAO-B inhibition. Parkinsonism Relat Disord 2007;13(Suppl 2):
patients on dopamine agonists treated with L-dopa/car- S99.
bidopa/entacapone is greater than that in patients initi- 14. Cattaneo C, Caccia C, Marzo A, Maj R, Fariello RG. Pressor
ated on L-dopa/carbidopa/entacapone.20 Furthermore, response to intravenous tyramine in healthy subjects after safina-
mide, a novel neuroprotectant with selective, reversible monoamine
the addition of safinamide may provide an alternative oxidase B inhibition. Clin Neuropharmacol 2003;26:213–217.
strategy to increasing the DA dose, thereby reducing the 15. Hamilton M. A rating scale for depression. J Neurol Neurosurg
risk of DA-related side effects, such as ICDs. Psychiatry 1960;23:56–62.
16. Lidstone SC, Schulzer M, Dinelle K, et al. Effects of expectation
on placebo-induced dopamine release in Parkinson disease. Arch
Acknowledgments: The authors thank Janet Dawson and Kirs- Gen Psychiatry 2010;67:857–865.
teen Munn of Complete Medical Communications (supported by Merck
Serono S.A. – Geneva, Switzerland, a branch of Merck Serono S.A., 17. Meissner WG, Frasier M, Gasser T, et al. Priorities in Parkinson’s
Coinsins, Switzerland, an affiliate of Merck KGaA, Darmstadt, Germany) disease research. Nat Rev Drug Discov 2011;10:377–393.
for their editorial support. 18. Stocchi F, Arnold G, Onofrj M, et al. Improvement of motor func-
tion in early Parkinson disease by safinamide. Neurology 2004;63:
746–748.
References 19. Onofrj M, Bonanni L, Thomas A. An expert opinion on safina-
mide in Parkinson’s disease. Expert Opin Investig Drugs 2008;17:
1. Miyasaki JM, Martin W, Suchowersky O, Weiner WJ, Lang AE.
1–11.
Practice parameter: initiation of treatment for Parkinson’s disease:
an evidence-based review: report of the Quality Standards 20. Stocchi F, Rascol O, Kieburtz K, et al. Initiating levodopa/carbi-
Subcommittee of the American Academy of Neurology. Neurology dopa therapy with and without entacapone in early Parkinson dis-
2002;58:11–17. ease: the STRIDE-PD study. Ann Neurol 2010;68:18–27.

112 Movement Disorders, Vol. 27, No. 1, 2012

You might also like