Summary Basis of Decision for Neupro

Review decision

The Summary Basis of Decision explains why the product was approved for sale in Canada. The document includes regulatory, safety, effectiveness and quality (in terms of chemistry and manufacturing) considerations.


Product type:

Drug

Summary Basis of Decision (SBD) documents provide information related to the original authorization of a product. The SBD for Neupro is located below.

Recent Activity for Neupro

SBDs written for eligible drugs approved after September 1, 2012 will be updated to include post-authorization information. This information will be compiled in a Post-Authorization Activity Table (PAAT). The PAAT will include brief summaries of activities such as submissions for new uses of the product, and whether Health Canada's decisions were negative or positive. PAATs will be updated regularly with post-authorization activity throughout the product's life cycle.

Post-Authorization Activity Table (PAAT) for Neupro

Updated:

2019-01-03

The following table describes post-authorization activity for Neupro, a product which contains the medicinal ingredient rotigotine. For more information on the type of information found in PAATs, please refer to the Frequently Asked Questions: Summary Basis of Decision (SBD) Project: Phase II and to the list of abbreviations that are found in PAATs.

For additional information about the drug submission process, refer to the Management of Drug Submissions Guidance.

Drug Identification Number (DIN):

  • DIN 02403897 - 1 mg/24 h patch, transdermal
  • DIN 02403900 - 2 mg/24 h patch, transdermal
  • DIN 02403919 - 3 mg/24 h patch, transdermal
  • DIN 02403927 - 4 mg/24 h patch, transdermal
  • DIN 02403935 - 6 mg/24 h patch, transdermal
  • DIN 02403943 - 8 mg/24 h patch, transdermal

Post-Authorization Activity Table (PAAT)

Activity/submission type, control numberDate submittedDecision and dateSummary of activities
NC # 2162982018-05-15Issued NOL
2018-09-14
Submission filed as a Level II (90 day) Notifiable Change (Risk Management Change) to update the PM to reflect revisions in the company core data sheet. As a result of the NC, modifications were made to the Adverse Reactions sections of the PM. The submission was reviewed and considered acceptable, and an NOL was issued.
NC # 2126562018-01-19Issued NOL
2018-04-06
Submission filed as a Level II (90 day) Notifiable Change (Risk Management Change) to update the PM to reflect revisions in the company core data sheet. As a result of the NC, modifications were made to the Warnings and Precautions, and Adverse Reactions sections of the PM, and corresponding changes were made to the PM Part III: Consumer Information. The submission was reviewed and considered acceptable, and an NOL was issued.
SNDS # 2037222017-03-13Issued NOC
2017-11-01
Submission filed as a Level I - Supplement for an additional drug substance manufacturing site and drug substance manufacturing process. The submission was reviewed and considered acceptable, and an NOC was issued.
SNDS # 2013372016-10-21Issued NOC
2017-08-11
Submission filed as a Level I - Supplement to support a formulation change. The submission was reviewed and considered acceptable, and an NOC was issued.
NC # 1755182014-06-10Issued No Objection Letter
2014-09-19
Submission filed as a Level II (90 day) Notifiable Change (Safety Change) to update the Adverse Reactions section of the Product Monograph. The submission was reviewed and a No Objection Letter was issued.
NC # 1700232013-11-14Issued No Objection Letter;
2014-02-24
Submission filed as a Level II (90 day) safety change to update the Adverse Reactions section of the Product Monograph based on post-market safety information. The submission was reviewed and a No Objection Letter was issued.
Drug Product (DINs 02403897, 02403919) market notificationNot applicableDate of first sale:
2013-12-19
The manufacturer notified Health Canada of the date of first sale pursuant to C.01.014.3 of the Food and Drug Regulations.
Drug product (DINs 02403900, 02403927, 02403935, 02403943) market notificationNot applicableDate of first sale:
2013-06-27
The manufacturer notified Health Canada of the date of first sale pursuant to C.01.014.3 of the Food and Drug Regulations.
NDS # 1455232012-03-30Issued NOC
2013-03-21
Notice of Compliance issued for New Drug Submission.
Summary Basis of Decision (SBD) for Neupro

Date SBD issued: 2013-06-25

The following information relates to the new drug submission for Neupro.

Rotigotine, 1 mg/24 h, 2 mg/24 h, 3 mg/24 h, 4 mg/24 h, 6 mg/24 h, 8 mg/24 h, patch, transdermal system

Drug Identification Number (DIN):

  • DIN 02403897 - 1 mg/24 h patch
  • DIN 02403900 - 2 mg/24 h patch
  • DIN 02403919 - 3 mg/24 h patch
  • DIN 02403927 - 4 mg/24 h patch
  • DIN 02403935 - 6 mg/24 h patch
  • DIN 02403943 - 8 mg/24 h patch

UCB Canada Inc.

New Drug Submission Control Number: 145523

On March 21, 2013, Health Canada issued a Notice of Compliance to UCB Canada Inc. for the drug product, Neupro (rotigotine) transdermal system.

The market authorization was based on quality (chemistry and manufacturing), non-clinical (pharmacology and toxicology), and clinical (safety and efficacy) information submitted. Based on Health Canada's review, the benefit/risk profile of Neupro is favourable for the treatment of the signs and symptoms of idiopathic Parkinson's disease, and the symptomatic treatment of moderate to severe idiopathic Restless Legs Syndrome (RLS) in adults.

1 What was approved?

Neupro is a transdermal system (patch) containing rotigotine, a non-ergolinic dopamine agonist that acts primarily on dopaminergic receptors. When applied to intact skin, Neupro is designed to continuously deliver rotigotine over a 24 hour period. Neupro was authorized for use in adults aged 18 years and older for the treatment of the signs and symptoms of idiopathic Parkinson's disease, and for the symptomatic treatment of moderate to severe idiopathic Restless Legs Syndrome (RLS).

For the treatment of the signs and symptoms of idiopathic Parkinson's disease, Neupro may be used both as early therapy, without concomitant levodopa, and as an adjunct to levodopa.

Neupro can be prescribed to elderly patients aged greater than 65 years, with dosing titration managed in a normal manner but may be individualized to accommodate potential age-related comorbidity.

The safety and efficacy of Neupro have not been established in children less than 18 years of age, therefore Neupro is not recommended in this patient population.

Neupro is contraindicated for patients who have hypersensitivity to the active substance or to any of the excipients. Neupro was approved for use under the conditions stated in the Neupro Product Monograph taking into consideration the potential risks associated with the administration of this drug product.

Neupro is available in six nominal strengths (1 mg/24 h, 2 mg/24 h, 3 mg/24 h, 4 mg/24 h, 6 mg/24 h, 8 mg/24 h rotigotine) and is presented as a transdermal patch composed of three layers. Each patch strength has a release surface area of 5, 10, 15, 20, 30, and 40 cm2 and contains 2.25, 4.5, 6.75, 9.0, 13.5, and 18.0 mg rotigotine, respectively. In addition to the medicinal ingredient rotigotine, the self-adhesive drug-matrix layer contains the following excipients: poly (dimethylsiloxane, trimethylsilyl silicate)-copolymerisate; povidone K90; sodium metabisulphite (E223); ascorbyl palmitate (E304) and DL-alpha-tocopherol (E307). The backing layer is a siliconized and aluminized polyester film, colour coated with a pigment [titanium dioxide (E171), pigment yellow 95, pigment red 166] and imprinted for identification (pigment red 144, pigment yellow 95, pigment black 7). The release liner is a transparent fluoropolymer coated polyester film.

For more information, refer to the Clinical, Non-Clinical, and Quality (Chemistry and Manufacturing) Basis for Decision sections.

Additional information may be found in the Neupro Product Monograph, approved by Health Canada and available through the Drug Product Database.

2 Why was Neupro approved?

Health Canada considers the benefit/risk profile of Neupro to be favourable for the treatment of the signs and symptoms of idiopathic Parkinson's disease, and for the symptomatic treatment of moderate to severe Idiopathic Restless Legs Syndrome (RLS) in adults.

Parkinson's disease is a chronic progressive neurodegenerative disease. Its prevalence is estimated to be 1-2 per 1,000 persons in the general population and 2 per 100 persons in people aged 65 years and older. The etiology of the disease currently remains unknown. Neuropathology findings in Parkinson's disease patients suggest that a substantial loss of dopaminergic neurons occurs in regions of the brain, including the substantia nigra pars compacta and the locus coeruleus. The progressive loss of dopaminergic neurons disrupts levels of dopamine and neurotransmission leading to motor symptoms of Parkinson's disease.

The clinical manifestations of Parkinson's disease include a characteristic combination of tremor, rigidity, slow movement (bradykinesia), and disturbance of gait and posture. In addition to these core symptoms, the patients may also have symptoms that indicate mood disorder, sleep disturbance, and cognitive impairment in varying degrees, especially as the disease progresses. As early-stage Parkinson's disease (EPD) patients progress to advanced-stage Parkinson's disease (APD), a combination therapy with the dopamine precursor drug levodopa and an adjuvant drug is required in order to reduce motor complications.

Idiopathic Restless Legs Syndrome (RLS) is described as a sensorimotor disorder with irresistible leg movements that are often accompanied by creeping sensations deep in the limbs. The disorder is commonly associated with periodic leg movements during sleep and sleep disturbances. The etiology is unclear. The prevalence of RLS is estimated to be approximately 4% in men and 8% in women in Europe and North America, in comparison to 2% in men and 4% in women in Asia. Some RLS patients have symptoms of anxiety and depression. Cases with significant severity require medical management. The use of a low dose non-ergolinic dopamine agonist has provided a favourable balance of effectiveness and safety for the symptomatic treatment of moderate to severe RLS.

Neupro (active ingredient rotigotine) has been studied in patients with EPD, APD, and RLS of at least moderate severity. The market authorization for Neupro was based primarily on six pivotal trials, in addition to several supportive clinical studies. Among the six pivotal trials, two studies (SP512 and SP513) conducted in EPD patients, two studies (SP650 and SP515) conducted in APD patients and two studies (SP790 and SP792) conducted in RLS patients, established the effectiveness of Neupro for the management of EPD, APD, and RLS.

The safety profile for Neupro appears to be similar to that of other non-ergolinic dopamine agonists in the same drug class. Adverse events (AEs) such as hallucination, orthostatic hypotension, tachycardia, and hypertension, are consistent with rotigotine's effect on the dopaminergic systems. Most of these AEs reported in the clinical trials were mild to moderate in severity. Several less frequent, but potentially more serious adverse events (SAEs) reported in clinical trials with Neupro included sudden onset of sleep, peripheral edema, impulse-control disorders, melanoma, fall, fibrotic complications, hyperglycaemia, heart failure, urinary retention and infection, constipation, and augmentation of RLS. Of the reported SAEs, sudden onset of sleep has been highlighted with a Black Box Warning in the Product Monograph.

Skin reactions were also observed at the site of patch application with Neupro. Application site reactions reported were generally mild to moderate in severity. In order to reduce potential skin injuries, Neupro is to be applied to the skin for 24 hours and then replaced with a new patch on another skin application site [for example (e.g.) abdomen, flank, hip, shoulder, thigh or upper arm] on a daily basis.

A Risk Management Plan (RMP) for Neupro was submitted by UCB Canada Inc. to Health Canada. Upon review, a revised RMP was requested. The RMP is designed to describe known and potential safety issues, to present the monitoring scheme and when needed, to describe measures that will be put in place to minimize risks associated with the product.

Overall, the therapeutic benefits of Neupro reported in the pivotal studies are positive and outweigh the potential risks. Neupro has an acceptable safety profile based on the data from non-clinical and clinical studies. The safety issues identified can be managed through labelling and pharmacovigilance surveillance. Appropriate warnings and precautions are described in the Neupro Product Monograph which addresses the safety concerns.

This New Drug Submission complies with the requirements of sections C.08.002 and C.08.005.1 and therefore Health Canada has granted the Notice of Compliance pursuant to section C.08.004 of the Food and Drug Regulations. For more information, refer to the Clinical, Non-Clinical, and Quality (Chemistry and Manufacturing) Basis for Decision sections.

3 What steps led to the approval of Neupro?

Submission Milestones: Neupro

Submission MilestoneDate
Pre-submission meeting:2010-11-04
Submission filed:2012-03-30
Screening
Screening Acceptance Letter issued:2012-05-25
Review
Biopharmaceutics Evaluation complete:2013-02-22
Quality Evaluation complete:2013-03-11
Clinical Evaluation complete:2013-03-19
Labelling Review complete:2013-03-18
Notice of Compliance issued by Director General, Therapeutic Products Directorate:2013-03-21

The Canadian regulatory decision on the non-clinical and clinical review of Neupro was based on a critical assessment of the Canadian data package. Foreign reviews completed by the European Union's centralized procedure European Medicines Agency (EMA), by the United States of America Food and Drug Administration (FDA), and the Australian Therapeutic Goods Administration (TGA) were consulted as an added reference.

For additional information about the drug submission process, refer to the Management of Drug Submissions Guidance.

4 What follow-up measures will the company take?

Requirements for post-market commitments are outlined in the Food and Drugs Act and Regulations.

6 What other information is available about drugs?

Up to date information on drug products can be found at the following links:

7 What was the scientific rationale for Health Canada's decision?
7.1 Clinical basis for decision

Clinical Pharmacology

Neupro (active ingredient rotigotine) is a non-ergolinic dopamine agonist for the treatment of signs and symptoms of idiopathic Parkinson's disease, and idiopathic Restless Legs Syndrome (RLS). Rotigotine is believed to elicit its beneficial effect on Parkinson's disease by activation of the dopaminergic receptors in the brain, including D3, D2, and D1 in a descending order of affinity. The precise mechanism of action of rotigotine as a treatment for RLS is unknown. As RLS is understood to be associated with a subtle central dopaminergic dysfunction, it is thought that rotigotine may exert its activity mainly via dopamine receptors. Rotigotine also acts as an agonist at the D5, D4.4 dopamine receptors at concentrations relevant to its therapeutic actions. In functional assays, rotigotine demonstrated antagonistic activity on the alpha2B and alpha2C adrenergic receptors and agonistic activity on the 5-HT1A and 5-HT1D serotonergic receptors. Rotigotine has no meaningful affinity to the 5-HT2B receptor at concentrations relevant to clinical use.

Approximately 45% of the rotigotine content in the patch is released within 24 hours. Rotigotine reaches steady-state plasma concentrations within 2 to 3 days of daily dosing. The binding of rotigotine to human plasma proteins is approximately 89.5% in vivo. Rotigotine is extensively metabolized by conjugation and N-dealkylation. Multiple cytochrome P450 (CYP) isoenzymes catalyze the metabolism of rotigotine. Plasma rotigotine delivered through the patch has a terminal half-life of 5 to 7 hours. Rotigotine is primarily excreted in urine (~71 %) and a smaller proportion is excreted in feces (~23 %).

A comprehensive Drug-Drug Interaction program was performed with rotigotine during its development program. No specific issue was identified with CYP inducers or inhibitors, nor with other concomitant medications likely used in the target populations.

In patients with moderate hepatic impairment, the plasma concentrations and exposure of unconjugated rotigotine are comparable to patients with normal hepatic function; however the plasma concentrations and exposure of the total rotigotine are increased by 38% and 14% respectively. Rotigotine has not been studied in patients with severe hepatic impairment. For patients with severe renal impairment, exposure to the inactive conjugates of rotigotine is doubled based on single-dose studies, while rotigotine exposure remains comparable to subjects without renal impairment. Rotigotine is not eliminated through dialysis.

The main safety issues of rotigotine identified in the pharmacology studies included orthostatic hypotension and lower prolactin concentrations and higher aldosterone concentrations. Moreover, heat application to the area where the patch is applied is expected to increase rotigotine release from the patch. Accordingly, appropriate warnings are in place in the Neupro Product Monograph to address the need of avoiding external sources of direct heat on the patch, including the other safety concerns.

For further details, please refer to the Neupro Product Monograph, approved by Health Canada and available through the Drug Product Database.

Clinical Efficacy

Clinical Trials in Parkinson's Disease

The efficacy of Neupro in the treatment of Parkinson's disease was evaluated in a multinational drug development program consisting of four randomized, double-blind placebo-controlled Phase III trials. Two trials (SP512 and SP513) were conducted in patients with early-stage Parkinson's disease (EPD) who were not receiving concomitant levodopa, and two studies (SP650 and SP515) were conducted in patients with advanced-stage Parkinson's disease (APD) who were receiving concomitant levodopa.

Clinical Trials in Patients with Early-Stage Parkinson's Disease (EPD)

Patients enrolled in the two Phase III EPD trials (SP512 and SP513) had limited or no prior exposure to levodopa. Patients were excluded from either trial if they had a history of pallidotomy, thalamotomy, deep brain stimulation, or fetal tissue transplant. Patients receiving selegiline, anticholinergic agents, or amantadine were required to be on a stable dose and able to maintain that dose for the duration of the trial.

Changes from baseline in the Unified Parkinson's Disease Rating Scale (UPDRS), Part II & III served as the primary outcome measures in both SP512 & SP513 trials. A "responder" for this EPD patient population was a patient with a 20% or greater decrease in the sum of scores from the Activities of Daily Living (ADL) and Motor Examination sections in the UPDRS (Parts II & III; a UPDRS subtotal) from the baseline visit to end of the double-blind maintenance period.

Study SP512 was a two-arm parallel group efficacy trial with randomized placebo-control to evaluate the efficacy of Neupro (2 mg/24 h, 4 mg/24 h, or 6 mg/24 h rotigotine) in reducing the symptoms of EPD. A total of 277 patients were randomized to treatment, with 96 on placebo and 181 on Neupro. After a run-in period of 4 weeks, patients underwent a weekly titration over 3 weeks up to a maximal dose of 6 mg/24 h rotigotine depending on efficacy and tolerability in achieving best symptomatic relief. Patients then received maintenance treatment over a 24-week period, followed by a 4-day dose de-escalation period and a 4-week safety follow-up period for patients who did not enroll in an open-label extension. The average age of the study population was 63 years with slightly more men (65%) than women enrolled.

In Study SP512, the Full Analysis Set (FAS) included 273 patients, with 96 on placebo and 177 on Neupro. The mean baseline subscore of the combined UPDRS (Part II & III) was similar in both groups (29.9 Neupro, 30.0 placebo). Neupro-treated patients experienced a mean change in the UPDRS (Parts II + III) subscore from baseline to end of treatment of 4.0, and the difference from placebo was statistically significant. Symptomatic improvement started to appear as titration progressed. The responder rates, based on a 20% reduction on the primary endpoint UPDRS (Part II + III), were 48% of the patients on Neupro versus (vs.) 19% on placebo.

Study SP513 was a three-arm parallel group efficacy trial with randomized placebo-control to evaluate the efficacy of Neupro (2 mg/24 h, 4 mg/24 h, 6 mg/24 h, or 8 mg/24 h) in reducing the symptoms of EPD. A total of 561 enrolled patients were randomized to treatment with 118 on placebo, 215 on Neupro and 228 on active oral comparator (ropinirole) for up to 39 weeks. After a run-in period of 4 weeks, the patients underwent a 13-week titration period to a maximal dose of 8 mg/24 h rotigotine depending on efficacy and tolerability in achieving best symptomatic relief. Patients then received maintenance treatment over a 24-week period, followed by a 12-day dose de-escalation period and a 4-week safety follow-up period for patients who did not enroll in an open-label extension. The average age of the study population was 61 years with slightly more men (58%) than women enrolled.

In Study SP513, the Full Analysis Set (FAS) included 557 patients, with 117 on placebo, 213 on Neupro, and 227 on active comparator. The mean baseline UPDRS (Parts II + III) sub-score was similar across all groups (33.2 Neupro, 31.3 placebo, 32.2 active comparator). Neupro-treated patients experienced a mean change in the UPDRS (Parts II + III) subscore from baseline to end of treatment of -6.8, and the difference from placebo was statistically significant. Symptomatic improvement started to appear as titration progressed. The responder rates, based on a 20% reduction on the primary endpoint UPDRS (Part II + III), were 52% of the patients on Neupro vs. 30% on placebo. The active comparator performed as expected.

Clinical Trials in Patients with Advanced-Stage Parkinson's Disease (APD)

Patients enrolled in the two Phase III APD trials (SP650 and SP515) had to be experiencing "on-off" periods at baseline, despite treatment with optimal doses of levodopa. Patients continued concomitant administration of levodopa (L-dopa) during the trial; however, reductions in the dosage of levodopa were allowed if patients experienced adverse events that the investigator considered related to dopaminergic therapy. Patients were excluded from the trials if they had a history of pallidotomy, thalamotomy, deep brain stimulation, or fetal tissue transplant. Patients receiving selegiline, anticholinergic agents, or amantadine were required to be on a stable dose and able to maintain that dose for the duration of the study. In the SP650 trial, catechol-O-methyltransferase (COMT)-inhibitors were not permitted.

Change from baseline in time spent "off" (hours) based on daily diaries was the primary outcome measure in these two APD trials. A "responder" for this APD patient population was defined as a patient with a ≥30% decrease in absolute time spent "off" from baseline to the end of the double-blind maintenance period.

Study SP650 was a three-arm, parallel group efficacy trial with randomized placebo-control to evaluate the efficacy of Neupro (8 mg/24 h or 12 mg/24 h) in reducing the symptoms of APD. A total of 351 patients were randomized, 120 on placebo, 120 on Neupro 8 mg/24, and 111 on Neupro 12 mg/24 h. After a run-in period of 4 weeks, the patients underwent a 3 to 5-week titration period with either placebo or Neupro (8 mg/24 h or 12 mg/24 h) where back titration was permitted to achieve optimal symptomatic relief. Patients then received maintenance treatment over a 24-week period. During the maintenance treatment, there was a permitted L-dopa reduction window of 1 to 3 weeks and then a stable L-dopa dose of 21 to 23 weeks. The maintenance period was then followed by an 8-day de-escalation period and a 4-week safety follow-up period for patients who did not enroll in an open-label extension. The average age of the study population was 66 years with slightly more men than women.

The Full Analysis Set (FAS) included 341 patients, 119 on placebo, 113 on Neupro 8 mg/24 h, and 109 on Neupro 12 mg/24 h. Mean baseline "off" times were similar among all treatment groups (6.4, 6.8, and 6.3 h for the placebo, Neupro 8 mg/24 h and 12 mg/24 h treatment groups, respectively). Neupro-treated patients experienced a mean change in "off" time from baseline to end of treatment of -2.7 h for the 8 mg/24 h treatment arm and -2.1 h for the 12 mg/24 h treatment arm, and the difference from placebo was statistically significant for both Neupro doses (8 mg/24 h, 12 mg/24 h). Symptomatic improvement started to appear as titration progressed. The responder rates, based on a 30% reduction on the primary endpoint ("off" time at the end of treatment) were 57% and 55% for the Neupro 8mg/24 h and 12mg/24 h groups respectively vs. 34% on placebo.

Study SP515 was a three-arm, parallel group trial using a double-dummy treatment to evaluate the efficacy of Neupro, up to a maximal dose of 16 mg/24 h with doses ranging from 4 mg/24 h to 16 mg/24 h in reducing the symptoms of APD. Enrolled patients were assigned to treatment with either a placebo or Neupro or an active oral comparator (pramipexole). A total of 506 patients were randomized, 101 on placebo vs. 204 on Neupro and 201 on pramipexole. After a run-in period of 4 weeks, the patients underwent an up to 7 week titration period with either Neupro, placebo or pramipexole to obtain best symptomatic relief. Patients then received maintenance treatment over a 16 week period, with a permitted L-dopa reduction window of 1 to 3 weeks, and stable L-dopa dose of 13 to 15 weeks. The maintenance period was then followed by a dose de-escalation period of up to 6 days and a 4-week safety follow-up period for patients who did not enroll in an open-label extension. The average age of the study population was 64 years with more men than women.

The Full Analysis Set (FAS) included 501 patients, 100 on placebo vs. 201 on Neupro, and 200 on pramipexole. Mean baseline "off" times were similar among all treatment groups (6.6, 6.2, and 6.0 h for the placebo, Neupro, and comparator treatment groups, respectively). Neupro-treated patients experienced a mean 2.5 hour decrease change in "off" time from baseline to end of treatment, and the difference from placebo was statistically significant. Symptomatic improvement started to appear as titration progressed. The responder rates, based on a 30% reduction on the primary endpoint ("off" time at the end of treatment) were 60% of the patients on Neupro vs. 35% on placebo.

Clinical Trials in Restless Legs Syndrome

The efficacy of Neupro was also evaluated for use in patients with moderate and severe idiopathic Restless Legs Syndrome (RLS) in two Phase III pivotal trials, SP790 and SP792. Both trials were fixed-dose, randomized, double-blind, placebo-controlled studies with maintenance periods of 6 months duration.

The primary outcome measurement (primary endpoint) used to assess treatment effect in both trials was the absolute change from baseline in the International Restless Legs Scale (IRLS Scale) sum score at the end of the maintenance period.

Across the two studies, the mean duration of RLS was 2.1 to 3.1 years, mean age was approximately 55 years (range of 19 to 78 years), approximately 67% were women, and 97% were Caucasian. A total of 746 patients were treated with Neupro in these two studies.

Study SP790 was a 4-arm, parallel-group, placebo-control efficacy trial conducted in moderate-to-severe RLS patients. Enrolled RLS patients were randomized to receive either placebo or one of the following three Neupro doses (1 mg/24 h, 2 mg/24 h, and 3 mg/24 h). A total of 458 patients were randomized (117 on placebo, 115 on Neupro 1 mg/24 h, 112 on Neupro 2 mg/24 h, and 114 on Neupro 3 mg/24 h). After a run-in period of 7 days, patients underwent a 3-week titration period up to their target daily dose. Patients then received maintenance treatment for 6 months, followed by a 7-day dose de-escalation phase and a 4-week safety follow-up period for patients who did not enroll in an open-label extension. The trial permitted one-step back-titration during the forced titration period, but none during the maintenance period. The average age of the study population was 58 years with more women than men.

The Full Analysis Set (FAS) included 447 patients: 114 on placebo; 112 on Neupro 1 mg/24 h; 109 on Neupro 2 mg/24 h; and 112 on Neupro 3 mg/24 h. Study results showed mean baseline IRLS sum scores were similar among all treatment groups (28.1, 28.1, 28.2, and 28.0 for the placebo, Neupro 1 mg/24 h, 2 mg/24 h, and 3 mg/24 h groups, respectively). Patients experienced a mean change in the IRLS sum score from baseline to the end of treatment for each of the 3 Neupro dose groups, and the difference from placebo was significant. The primary endpoint mean change from baseline in IRLS sum score at end of the maintenance period were as follows: -8.0 ± 9.7 (LS Mean ± SD) on placebo vs. -13.2 ± 10.0 on Neupro 1 mg/24 h; -15.6 ± 9.5 on Neupro 2 mg/24 h; and -16.1 ± 10.9 on Neupro 3 mg/24 h. The responder rates with a 50% cut-off on the IRLS were 25.4% on placebo vs. 51.8% on Neupro 1 mg/24 h; 57.8% on Neupro 2 mg/24 h; and 55.4% on Neupro 3 mg/24 h. The high dose at 3 mg/24 h did not result in an appreciable level of symptomatic relief than the mid-dose. The profiles of treatment-emergent adverse events and serious adverse events were similar to those observed in the patients with Parkinson's disease. The relatively higher rate of discontinuation, ranging from 22% to 42%, resulted mainly from the design of the trial (forced up-titration), inefficacy (in those on placebo), and adverse reactions to rotigotine (e.g. those on 3 mg/24 h).

Study SP792 was similar in design to the SP790 study; however this study was a 5-arm, parallel-group trial conducted in moderate-to-severe RLS patients. Enrolled RLS patients were randomized to receive either placebo or one of the following four Neupro doses (0.5 mg/24 h, 1 mg/24 h, 2 mg/24 h, and 3 mg/24 h). A total of 505 patients were randomized: 100 on placebo; 99 on Neupro; 0.5 mg/24 h; 101 on Neupro 1 mg/24 h; 99 on Neupro 2 mg/24 h; and 106 on Neupro 3 mg/24 h. After a run-in period of 7 days, the patients then underwent a 4-week titration period up to the target daily dose. The maintenance period was 6 months, followed by a 7-day dose de-escalation period and a 4-week safety follow-up period for patients who did not enroll in an open-label extension. The trial permitted one-step back-titration during the forced titration period, but none during the maintenance period. The average age of the study population was 52 years with slightly more women than men.

The Full Analysis Set (FAS) included 494 patients: 99 on placebo; 98 on Neupro 0.5 mg/24 h; 99 on Neupro 1 mg/24 h; 95 on Neupro 2 mg/24 h; and 103 on Neupro 3 mg/24 h. Study results showed mean baseline IRLS sum scores were similar among all treatment groups (23.5, 23.1, 23.2, 23.3, and 23.6 for the placebo, Neupro 0.5 mg/24 h, 1 mg/24 h, 2 mg/24 h, and 3 mg/24 h groups, respectively). Patients experienced a mean change in the IRLS sum score from baseline to the end of treatment for each of the 4 Neupro dose groups. The difference between the two highest treatment groups (Neupro 2 mg/24 h and 3 mg/24 h) and placebo was significant. The mean change from baseline in IRLS Scale sum score at end of the maintenance period were as follows: -9.0 ± 7.7 (LS Mean ± SD) on placebo vs. -10.9 ± 8.9 on Neupro 0.5 mg/24 h; -11.1 ± 9.3 on Neupro 1 mg/24 h; -13.4 ± 9.2 on Neupro 2 mg/24 h; and -14.3 ± 9.4 on Neupro 3 mg/24 h. The responder rates with a 50% cut-off on the IRLS were: 37% on placebo vs. 48% on Neupro 0.5 mg/24 h; 52% on Neupro 1 mg/24 h; 60% on Neupro 2 mg/24 h; and 67% on Neupro 3 mg/24 h.

Overall, six Phase III pivotal studies (two studies for each indication), demonstrated the efficacy of Neupro for use in the management of EPD, APD, and moderate to severe idiopathic RLS.

For additional information, refer to the Neupro Product Monograph, approved by Health Canada and available through the Drug Product Database.

Clinical Safety

The safety profile of Neupro was based primarily on pooled safety data from the four Phase III pivotal trials conducted in patients with Parkinson's disease, in addition to the two Phase III pivotal trials conducted in patients with RLS. These six pivotal trials have been previously described in Clinical Efficacy section. Additional safety data derived from Phase II studies were also assessed in establishing the safety profile of Neupro. The description of the safety profile is intended to inform the health-care provider and the patient for optimal safety management. Because of the complex scenarios of patient pathophysiology and therapeutics, the general safety profile makes no statement on causality.

Adverse Events in Early-Stage Parkinson's Disease (EPD)

The safety of Neupro was evaluated in 649 patients with EPD. The most frequently observed adverse events (AEs) reported with an incidence rate of ≥5% in Neupro-treated patients compared to placebo-treated patients were those of nausea, application and instillation site reactions, somnolence, dizziness, headache, vomiting, fatigue, insomnia, peripheral edema, and constipation.

Approximately 6.8% of Neupro-treated patients reported serious adverse events (SAEs), vs. 5.9% of patients on placebo. The most frequent SAE was application site reactions (0.5% on Neupro vs. 0.0% on placebo).

Approximately 13% of 649 Neupro-treated patients discontinued treatment because of AEs, compared with 6% of 290 patients who received placebo. The AEs causing most discontinuations of treatment were those of application site reaction, nausea, and vomiting.

Adverse Events in Advanced-Stage Parkinson's Disease (APD)

The safety of Neupro was evaluated in 658 patients with APD. The most frequently observed AEs reported with an incidence rate of ≥5% in Neupro-treated patients compared to placebo-treated patients were those of application site reactions, nausea, somnolence, dyskinesia, dizziness, vomiting, insomnia, and peripheral edema.

Approximately 7.4% of Neupro-treated patients reported SAEs vs. 6.9% of patients on placebo. The most frequent SAEs included nausea and application site dermatitis (both 0.6% Neupro vs. 0.0% placebo).

Approximately 11% of 658 Neupro-treated patients discontinued treatment because of AEs, compared with 8% of patients who received placebo. The AEs causing most discontinuations of treatment were those of nausea, vomiting, dizziness, and application site reactions.

Adverse Events in Restless Legs Syndrome (RLS)

The safety of Neupro was evaluated in 748 Neupro-treated patients with RLS. The most frequently observed AEs reported with an incidence rate of ≥5% in Neupro-treated patients compared to placebo-treated patients were those of application and instillation site reactions, nausea, headache, fatigue, nasopharyngitis, somnolence, dizziness, and pruritus.

Approximately 5.6% of Neupro-treated patients reported SAEs vs. 4.1% of patients on placebo. The most frequent SAE was application site and instillation reactions (0.8% on Neupro vs. 0.0% on placebo).

Approximately 18% of 748 Neupro-treated patients discontinued treatment because of AEs, compared with 6% of patients who received placebo. The AEs most commonly causing discontinuation of treatment were those of application site reactions, dizziness, and nausea.

Other Adverse Events

Other AEs, such as hallucination, orthostatic hypotension, tachycardia, and hypertension, have also been reported with use of Neupro. However, these AEs are known to be associated with use of a dopamine agonist, like that of Neupro, due to the drug's stimulation of the dopaminergic system.

Several less frequent but potentially serious adverse events (SAEs) reported with use of Neupro in clinical trials included sudden onset of sleep, peripheral edema, impulse-control disorders, melanoma, fall, fibrotic complications, hyperglycaemia, heart failure, urinary retention and infection, constipation, and augmentation of RLS. While the majority of these events are known to occur in the Parkinson's disease patient population irrespective of drug exposure, sudden onset of sleep, peripheral edema, impulse-control disorders and augmentation of RLS are known to occur with Neupro as well as with other non-ergolinic dopamine agonists.

A Black Box Warning describing serious warnings and precautions has been included in the Product Monograph for Neupro. Patients receiving treatment with Neupro and other dopaminergic agents have reported excessive daytime somnolence and episodes of sudden onset of sleep while engaged in activities of daily living. Although some of the patients reported somnolence while on Neupro, others perceived that they had no warning signs, such as excessive drowsiness, and believed that they were alert immediately prior to the event.

In clinical trials, the pattern of deaths and SAEs were in line with expectations for the disease and age strata. There was no peculiar finding in this set of data that could raise concern.

The post-market safety profile of Neupro is similar to that seen in clinical trials. Aggression and pruritus generalized are two AEs that have been identified in the post-market safety database. With the exception of application site skin reactions, the observed AEs are consistent with those seen with other dopaminergic agents in the same drug class.

In conclusion, the safety profile for Neupro appears to be consistent with its dopaminergic activity and associated mechanism of action, with the exception of its tendency to cause application site irritations. Neupro may result in SAEs such as hallucination, orthostatic hypotension, tachycardia, and hypertension. In addition, other potential SAEs include sudden onset of sleep, peripheral edema, impulse-control disorders, melanoma, fall, fibrotic complications, hyperglycaemia, heart failure, urinary retention and infection, constipation, and augmentation of RLS. Risk management wording, commensurate with the likelihood of the adverse reactions and their potential seriousness, has been included in the Product Monograph for Neupro.

For more information, refer to the Neupro Product Monograph, approved by Health Canada and available through the Drug Product Database.

7.2 Non-Clinical Basis for Decision

The non-clinical studies submitted for review are adequate to support the market authorization of Neupro (rotigotine). Results of non-clinical studies demonstrated that rotigotine is effective in animal models of Parkinson disease where it restores locomotor activity.

Toxicological studies identified the major effects observed as associated with induced dopaminergic stimulation and the consequent decrease of prolactin secretion. Rotigotine has no overt genotoxicity potential. Rotigotine was found to have no carcinogenicity potential in mice, rats, and minipigs. The neoplasticity finding in rats was believed to be a class effect of dopaminergic agents.

Reproductive studies have shown that rotigotine reduces mating success and foetal implantation in mice, rats, and rabbits. It retards foetal growth, due to trans-placental exposure, and neonatal growth, due to exposure to rotigotine concentrated in the milk of the treated mothers. Rotigotine also retards growth in young rodents. No teratogenicity was found.

The results of the non-clinical studies as well as the potential risks to humans have been included in the Neupro Product Monograph. Appropriate warnings and precautionary measures are in place in the Neupro Product Monograph to address the identified safety concerns.

For more information, refer to the Neupro Product Monograph, approved by Health Canada and available through the Drug Product Database.

7.3 Quality Basis for Decision

The Chemistry and Manufacturing information submitted for Neupro has demonstrated that the drug substance and drug product can be consistently manufactured to meet the approved acceptance criteria. Proper development and validation studies were conducted, and adequate controls are in place for the commercial processes. Changes to the manufacturing process and formulation made throughout the pharmaceutical development are considered acceptable upon review. Based on the stability data submitted, the proposed shelf-life of Neupro is acceptable.

The proposed limits of drug-related impurities are considered adequately qualified [that is within International Conference on Harmonisation (ICH) limits].

All facilities involved in production of Neupro (drug substance and drug product) are compliant with Good Manufacturing Practices.

All non-medicinal ingredients found in the drug product (described above) are acceptable for use in drugs according to the Food and Drug Regulations.

There are no excipients of human or animal origin used in the manufacture of the drug substance or drug product.