Summary Basis of Decision for Azilect ®
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:
Azilect®
Rasagiline mesylate, 0.5 mg, 1 mg, Tablet, Oral
Teva Pharmaceutical Industries Ltd.
Submission control no: 101846
Date issued: 2008-06-22
Health Products and Food Branch
Our mission is to help the people of Canada
maintain and improve their health.
Health Canada
HPFB's Mandate is to take an integrated approach to managing the health-related risks and benefits of health products and food by:
- minimizing health risk factors to Canadians while maximizing the safety provided by the regulatory system for health products and food; and,
- promoting conditions that enable Canadians to make healthy choices and providing information so that they can make informed decisions about their health.
Health Products and Food Branch
Également disponible en français sous le titre : Sommaire des motifs de décision (SMD), PrAZILECTMD, Rasagiline mesylate, 0.5 mg et 1 mg, comprimés, Teva Pharmaceutical Industries Ltd., No de contrôle de la présentation 101846
Foreword
Health Canada's Summary Basis of Decision (SBD) documents outline the scientific and regulatory considerations that factor into Health Canada regulatory decisions related to drugs and medical devices. SBDs are written in technical language for stakeholders interested in product-specific Health Canada decisions, and are a direct reflection of observations detailed within the evaluation reports. As such, SBDs are intended to complement and not duplicate information provided within the Product Monograph.
Readers are encouraged to consult the 'Reader's Guide to the Summary Basis of Decision - Drugs' to assist with interpretation of terms and acronyms referred to herein. In addition, a brief overview of the drug submission review process is provided in the Fact Sheet entitled 'How Drugs are Reviewed in Canada'. This Fact Sheet describes the factors considered by Health Canada during the review and authorization process of a drug submission. Readers should also consult the 'Summary Basis of Decision Initiative - Frequently Asked Questions' document.
The SBD reflects the information available to Health Canada regulators at the time a decision has been rendered. Subsequent submissions reviewed for additional uses will not be captured under Phase I of the SBD implementation strategy. For up-to-date information on a particular product, readers should refer to the most recent Product Monograph for a product. Health Canada provides information related to post-market warnings or advisories as a result of adverse events (AE).
For further information on a particular product, readers may also access websites of other regulatory jurisdictions. The information received in support of a Canadian drug submission may not be identical to that received by other jurisdictions.
Other Policies and Guidance
Readers should consult the Health Canada website for other drug policies and guidance documents. In particular, readers may wish to refer to the 'Management of Drug Submissions Guidance'.
1 Product and submission information
Brand name:
Manufacturer/sponsor:
Medicinal ingredient:
International non-proprietary Name:
Strength:
Dosage form:
Route of administration:
Drug identification number(DIN):
- 02284642
- 02284650
Therapeutic Classification:
Non-medicinal ingredients:
Submission type and control no:
Date of Submission:
Date of authorization:
2 Notice of decision
On August 17, 2006, Health Canada issued a Notice of Compliance to Teva Pharmaceutical Industries Ltd. for the drug product Azilect.
Azilect contains the medicinal ingredient rasagiline mesylate which is an antiparkinson agent.
Azilect is indicated for the treatment of the signs and symptoms of idiopathic Parkinson's disease as initial monotherapy and as adjunct therapy to levodopa. Azilect is a potent, selective, irreversible, monoamine oxidase type B (MAO-B) inhibitor. The dose at which rasagiline selectively inhibits only MAO-B (and not MAO-A) in humans and the sensitivity to tyramine during rasagiline treatment at doses higher than 1 mg have not been sufficiently characterized. The precise mechanisms of action of rasagiline are unknown.
The market authorization was based on quality (chemistry and manufacturing), non-clinical, and clinical data. The safety and effectiveness of Azilect for the treatment of Parkinson's disease were established in three 18- to 26-week randomized, placebo-controlled trials. The effectiveness of Azilect was demonstrated in patients with early Parkinson's disease who were receiving Azilect as monotherapy and who were not receiving any concomitant dopaminergic therapy. The effectiveness of Azilect as an adjunct therapy was demonstrated in patients with Parkinson's disease who were treated with levodopa.
Azilect (0.5 mg and 1.0 mg, rasagiline mesylate) is presented as oral tablets. The recommended and maximum dose in both monotherapy and adjunct therapy is 1.0 mg once daily. Rasagiline should not be used at daily doses exceeding the maximum recommended dose (1.0 mg/day) due to the risks associated with non-selective inhibition of MAO. Adequate studies above this dose have not been conducted. In the absence of restrictions being placed on diet and concomitant drug use, it is critical to adhere to this maximum dosage. When used in combination with levodopa, a reduction of the levodopa dosage may be considered based upon individual response. Patients who are initiating adjunct therapy, those with mild hepatic impairment, or those who are taking concomitant ciprofloxacin or other CYP1A2 inhibitors should use 0.5 mg Azilect daily. Dosing guidelines are available in the Product Monograph.
Azilect is contraindicated for use with cyclobenzaprine; dextromethorphan; meperidine; St. John's wort; sympathomimetic amines, including amphetamines as well as cold products and weight-reducing preparations that contain vasoconstrictors; the analgesic agents tramadol, methadone, and propoxyphene; antidepressants; or other selective MAO-B or non-selective MAO inhibitors. Patients taking Azilect should not undergo elective surgery requiring general anesthesia and they should not be given local anesthesia containing cocaine or sympathomimetic vasoconstrictors. Azilect is also contraindicated in patients with pheochromocytoma. Azilect should be administered under the conditions stated in the Product Monograph taking into consideration the potential risks associated with the administration of this drug product. Detailed conditions for the use of Azilect are described in the Product Monograph.
Based on the Health Canada review of data on quality, safety, and effectiveness, Health Canada considers that the benefit/risk profile of Azilect is favourable for the treatment of the signs and symptoms of idiopathic Parkinson's disease as initial monotherapy and as adjunct therapy to levodopa.
3 Scientific and Regulatory Basis for Decision
The original submission for Azilect (Control No. 087283) was filed for review on October 8, 2003 and included only the 1 mg dose. The submission underwent a first review and was issued a Notice of Non-Compliance (NON) due to an outstanding safety concern. A response was filed in April 2005 and in September 2005 the sponsor was informed that an additional strength (0.5 mg tablet) would be required as a starting dose which was omitted from the original NON. As a result, it was suggested that the sponsor withdraw the submission and immediately refile to include the new strength. This new submission for Azilect (Control No. 101846) was filed for review on October 17, 2005. The issue of concern in the NON was resolved, and a Notice of Compliance (NOC) for Azilect was issued on August 17, 2006.
3.1 Quality Basis for Decision
3.1.1 Drug Substance (Medicinal Ingredient)
Manufacturing Process and Process Controls
Rasagiline mesylate is manufactured by a non-chiral selective synthesis and involves three steps. Each step of the manufacturing process is considered to be controlled within acceptable limits:
- The sponsor has provided information on the quality and controls for all materials used in the manufacture of the drug substance.
- The drug substance specifications are found to be satisfactory. Impurity limits meet ICH requirements.
- The processing steps have been evaluated and the appropriate ranges for process parameters have been established.
Characterization
Rasagiline mesylate is a white to off-white crystalline powder and is highly soluble. The structure of rasagiline mesylate has been adequately elucidated and the representative spectra have been provided. Physical and chemical properties have been described and are satisfactory.
The sponsor has provided a summary of all drug-related impurities. Impurities arising from manufacturing were reported and characterized. These products were found to be within ICH established limits, and therefore considered to be acceptable.
Control of Drug Substance
Validation reports are considered satisfactory for all analytical procedures used for in-process and release testing of the drug substance, and to justify the specifications of the drug substance.
Data from the batch analyses were reviewed and are considered to be acceptable according to the specifications of the drug substance.
The proposed packaging components are also considered acceptable.
Stability
Stability study results based on accelerated, long-term, and stress testing show that rasagiline mesylate is a stable compound when packaged as proposed over the suggested storage period. Rasagiline mesylate was shown to be photostable.
3.1.2 Drug Product
Description and Composition
Azilect (rasagiline mesylate tablets) is presented in two strengths, 0.5 mg and 1 mg (expressed as rasagiline base). Azilect tablets are provided as white to off-white, round, flat, bevelled tablets, debossed with either 'GIL' 1 or 'GIL' 0.5 on one side and plain on the other. The tablets are packaged in 30 mL high-density polyethylene (HDPE) containers with a threaded neck. Each container contains 30 tablets.
Two different closures are approved:
- white, round polypropylene child-resistant tamper-evident screw caps fitted with a silica desiccant insert, or
- white, round (non-child-resistant) polypropylene tamper-evident screw caps fitted with a silica desiccant insert
Tablets are also available in aluminum-silver/aluminum-soft blister packaging of 7-10 tablets for physician/promotional samples only.
Each Azilect tablet contains the non-medicinal ingredients mannitol, colloidal silicon dioxide, starch, pregelatinized starch, stearic acid, and talc. Due to the BSE/TSE risk, the stearic acid was replaced during formulation with vegetable-derived stearic acid.
All excipients found in the drug product are acceptable for use in drugs by the Canadian Food and Drug Regulations. The compatibility of rasagiline mesylate with the excipients is demonstrated by the stability data presented for the proposed commercial formulation.
Pharmaceutical Development
Pharmaceutical development data, including container closure system and microbiological attributes, were considered acceptable. Studies which justified the type and proposed concentration of excipients to be used in the drug product were reviewed and considered to be acceptable.
Manufacturing Process and Process Controls
The manufacturing process for Azilect includes dry mixing, wet granulation, drying, milling, blending, tablet compression, and packaging. All equipment, operating parameters, in-process tests and detailed instructions are adequately defined in the documentation. The manufacturing process is considered to be acceptable and adequately controlled within justified limits.
Control of Drug Product
Azilect batches are tested to verify appearance, identification, content uniformity, dissolution, assay, weight, thickness, hardness, disintegration, friability, and the presence of impurities and degradation products. The test specifications and analytical methods are considered acceptable; the shelf-life and the release limits, for individual and total degradation products are within acceptable limits.
Validation reports are satisfactory for all analytical procedures used for in-process and release testing of the drug product, and to justify the specifications of the drug product.
Data from final batch analyses were reviewed and considered to be acceptable according to the specifications of the drug product.
Stability
Stability data show that HDPE bottles capped with polypropylene child- or non-child-resistant caps and the aluminum-silver/aluminum-soft physician sample blister packs are acceptable container/closure systems for the drug product. Azilect tablets are photostable.
Based upon the real-time and accelerated stability study data submitted, the proposed 36- and 24-month shelf-lives at 25 ºC for the commercial product and physician samples, respectively, are considered acceptable.
3.1.3 Facilities and Equipment
The design, operations, and controls of the facility and equipment that are involved in the production are considered suitable for the activities and products manufactured. All of the proposed manufacturing sites comply with the requirements of Division 2 of the Food and Drug Regulations.
3.1.4 Summary and Conclusion
This New Drug Submission is considered to meet the requirements of Division C.08.002 of the Food and Drug Regulations insofar as the Quality (Chemistry and Manufacturing) information is concerned. The Chemistry and Manufacturing information submitted for Azilect has demonstrated that the drug substance and drug product can be consistently manufactured to meet the specifications agreed upon. Proper development and validation studies were conducted, and adequate controls are in place for the commercial processes.
3.2 Non-Clinical Basis for Decision
3.2.1 Pharmacodynamics
The primary pharmacological activity of rasagiline was investigated in various in vitro and in vivo systems. Rasagiline was found to be a highly potent, selective, irreversible monoamine oxidase type B (MAO-B) inhibitor. Inhibition of the MAO-B enzyme causes an increase in extracellular levels of dopamine in the striatum, which in turn increases dopaminergic activity. It is likely that this is the main mechanism by which rasagiline improves models of dopaminergic motor dysfunction. Rasagiline does not inhibit other regulatory enzymes or bind to other receptors.
The ability of rasagiline to restore normal motor activities was studied in the rat for drug- and hypoxia-induced dopaminergic dysfunctions. Effects on learning and memory were also tested using models of passive avoidance, active avoidance, and the Morris water maze. Rasagiline treatment was found to improve passive avoidance, learning, and memory in both postnatal and adult rats.
In addition, results from non-clinical studies have indicated that rasagiline improved cognition in animal models and was neuroprotective in both in vitro and in vivo models. The neuroprotective activity of rasagiline is independent of its MAO inhibitory activity, and may be mediated by other mechanisms. One such mechanism may be the influence of aminoindan, a major metabolite of rasagiline. Aminoindan is not a MAO inhibitor, but in several animal models of impaired motor and cognitive functions it was demonstrated to ameliorate dopaminergic motor dysfunction and hypoxia-induced cognitive deficits, thus potentially contributing to rasagiline's beneficial effect.
The neuroprotective effects of rasagiline were compared to the effect of selegiline, another MAO-B inhibitor, using in vivo and in vitro models. Rasagiline exhibited more potent neuroprotective and neurotrophic effects compared to selegiline on dopaminergic neurons in primary mesencephalic cultures. Rasagiline also exhibited some protective effects against neurotoxin MPTP, which can induce parkinsonism in human and non-human primates. The required potency of a drug to prevent MPTP-induced neurotoxicity is indicative of its MAO-B inhibitory activity. In cell cultures, the drug effects were interpreted as inhibition of apoptotic cell death which was not due to the MAO inhibitory activity of these compounds. In vivo, the neuroprotective action of rasagiline was clearly evident and increased animal survival rates were noted. Increased neuronal survival was confirmed by histological, immunocytochemical, and MRI analyses. These results are currently considered preliminary however, and further investigation is required to draw definitive conclusions and establish clinical relevance.
The effects of rasagiline on the cardiovascular and central nervous system in animal models were investigated, however the safety pharmacological studies are considered to be incomplete. According to ICH guidelines, the core testing battery should also include analysis of the respiratory system and if there is a base of concern, other physiological systems such as the renal and gastrointestinal systems, should also be investigated.
No overt changes in arterial blood pressure or heart rates were seen in any of the four animals tested. There were no large or sustained changes in ECG waveform intervals or morphology. Rasagiline administered as a single dose up to 2 mg/kg produced no effect on motor activity, behaviour, or response to stimuli. At doses at least 10-fold higher than the proposed human therapeutic dose, no observable adverse effects in the cardiovascular or central nervous system were found.
ECG evaluation in free-moving dogs by telemetry, using a single dose (3 mg/kg), gave negative findings with respect to QT interval prolongation. Some increases in QT intervals were observed in toxicity studies with dogs treated with levodopa/carbidopa with or without rasagiline; however, there was no prolongation in the group treated with rasagiline alone. A study to test the potential of rasagiline to interact with potassium ion channels (hERG test) was performed and it did not indicate any potential to induce QT prolongation.
3.2.2 Pharmacokinetics
The pharmacokinetic studies indicated that absorption of rasagiline was rapid and substantial in all animal species studied. Despite these findings, oral bioavailability was relatively low, mainly due to intestinal and liver first-pass metabolism. The systemic exposure to rasagiline was evaluated in toxicokinetic studies, as part of each toxicity study in three animal species. Over the range of doses used in toxicity studies, exposure to rasagiline increased linearly with dose at low doses (up to 5 mg/kg/day) and non-linearly at high doses (above 5 mg/kg). Plasma protein binding was relatively high in all species tested but is not considered to affect pharmacological action. Volume of distribution was also high, indicating that the drug is distributed to all tissues.
The metabolism of rasagiline was investigated in the mouse, rat, and dog after single oral and intravenous doses. Rasagiline is primarily biotransformed by CYP450 enzymes to various metabolites. Similar metabolites were identified in all species studied.
In all species tested, rasagiline was excreted in the urine, mainly as polar metabolites. Excretion was rapid, and only small residues were noted 24-hours post-dosing. This may be related to the irreversible binding of rasagiline at MAO sites.
It should be emphasized that although rasagiline did not accumulate in the tissues in rats, the drug was retained at higher concentrations in pigmented cells, mainly in the eyes and skin, than in non-pigmented cells. It is possible that substantial accumulation of rasagiline in melanocytes may cause some interaction with endogenous and/or exogenous compounds that have adverse effects, such as induction of cell proliferation, genotoxic effects, or inhibition of the apoptotic process.
3.2.3 Toxicology
All toxicity studies used acceptable, standard protocols designed according to ICH Guidelines. Most of the studies were carried out in accordance with Good Laboratory Practices.
Short-Term
General toxicity studies did not reveal any remarkable or unexpected toxicity findings in any of the animal species tested. Rasagiline demonstrated low acute toxicity; acute toxicity is not anticipated at the human therapeutic dose or even at much higher doses. The lack of toxicity in many of the subchronic studies suggests that the dosages used were too low, although the toxicokinetic data indicate adequate exposure. The reduction of body weight in treated animals as observed in all species tested, clinical observations, and clinical pathological and histopathological findings, could be associated with the dopaminergic activity of rasagiline.
Signs of immunotoxic effects in both rat and dog studies were noted, primarily atrophy of lymphoid tissues at the highest tested dose levels. These were not followed up by functional testing. The sponsor considered that the changes in the lymphoid tissues were related to reduced body weight gains.
Genotoxicity
The genotoxic potential of rasagiline was evaluated in a number of in vitro and in vivo tests that examined bacterial and mammalian gene mutations, chromosomal aberrations in mammalian cell cultures, and micronucleus induction in the bone marrow of mice.
Rasagiline produced negative results in the bacterial mutation assays and in the mouse micronucleus assay. However, there were positive responses in the chromosomal aberration assay in lymphocytes and the mouse lymphoma cell assay for gene mutation, which together suggest that rasagiline has genotoxic potential.
The results obtained from experiments where changes were made to modify the culture medium were difficult to interpret.
Carcinogenicity
Carcinogenicity bioassays were conducted in rats at oral (gavage) doses of 0.3, 1, and 3 mg/kg (males) and 0.5, 2, 5, and 17 mg/kg (females) and mice at oral (gavage) doses of 1. 15, and 45 mg/kg. In rats, there was no increase in tumours at any dose tested. The sponsor's overall conclusion was that rasagiline has equivocal carcinogenic activity, not related to drug effects, despite the fact that positive results were obtained in the mouse study (the exposure ratio of mice at 15 mg/kg/day relative to humans treated with 1 mg/day is greater than 100-fold higher).
The sponsor argued that the tumours observed in mice at the two anatomical sites (lung and Harderian gland) were common tumours and thus the trend test for tumour rates should only be considered statistically significant at a p-value of equal or less than 0.005. This is based on a decision criterion developed by the FDA in evaluating common tumour incidences (Draft Guidance for Industry Statistical Aspects of the Design, Analysis, and Interpretation of Chronic Rodent Carcinogenicity Studies of Pharmaceutical, FDA, 2001). Based on the FDA's criterion for significance in incidence rates, the sponsor claimed that only the combined alveolar/bronchial adenoma/carcinoma of the lung in male mice (with a p = 0.002) should be considered a significant positive trend while the trend tests for all other sites, including tumour incidences in female mice of the lung (p = 0.0113) and Harderian tumours in male mice (p = 0.0194), should all be considered non-significant.
It should be noted that although the FDA's draft document regarding the statistical analysis of tumour data has been in existence for more than three years, it has not yet been finalized. Furthermore, in other carcinogenicity studies, notably in the studies conducted by the National Toxicology Program (NTP), different p-values for accepting significance in trend tests are being used (e.g., p-value of 0.05 and 0.01 for rare and common tumour incidences, respectively) than those suggested in the FDA's draft document.
Another important point to consider in demonstrating significant tumour rates is that, in addition to the trend test, pairwise comparison of tumour incidences between the control and a treated group should also be used. This is generally recognised as a test for significance and is also advocated in the FDA's document. The tumour rates in pairwise comparison are considered to be significant at p-value of 0.01 and 0.05 for common and rare tumours, respectively. By comparing the high dose and control groups in the mouse study a significant increase in tumour rates for adenomas/carcinomas in the lung of males can be demonstrated (p = 0.0046 for 45 mg/kg/d dose and p = 0.036 for 15 mg/kg/d dose).
As both statistical analyses (trend test and pairwise comparison), resulted in significant positive tumour incidences for lung tumours in male mice, it is concluded that there is clear potential for carcinogenic activity of rasagiline at high multiples of the clinical dose.
The increased incidences of Harderian gland adenomas/carcinoma with a p-value of 0.0131 when the high dose vs. control groups were compared, provide additional support to consider rasagiline carcinogenic in male mice.
In female mice, a significant increase in the adenoma/carcinoma incidences of the lung (p = 0.0237) was shown by pairwise comparison of the high dose vs. control groups. This result, together with a positive trend test for this type of tumour, provides some evidence that rasagiline also has carcinogenic activity in female mice.
Further information regarding carcinogenicity risks in humans can be found in section 3.3.5 Assessment of Melanoma Risk.
Reproduction and Development
Rasagiline administered to rats and rabbits of both sexes did not produce adverse effects on fertility. The no observed adverse effect level (NOAEL) was 3 mg/kg/day and 7 mg/kg/day for rats and rabbits, respectively. These doses represent a more than 50-fold higher dose than the human therapeutic dose, based on AUC ratios.
In combination treatment with levodopa (LD) and carbidopa (CD), rasagiline induced maternal toxicity in rabbits at doses above 0.1 mg/kg/day. Exposure to a 0.1/80/20 mg/kg/day combination (LD/CD/rasagiline) dose was similar to the exposure received by humans at the 1 mg/day clinical dose. No teratogenic effects were seen in rabbits. In rats however, exposure to combination treatment at a dose of 1/80/20 mg/kg/day, produced a high incidence of skeletal malformations in the form of wavy ribs. The sponsor argued that although the wavy ribs appeared to be treatment-related, these are reversible effects found in rodents only. Nevertheless, wavy ribs are considered to be a minor skeletal malformation. From these studies it is uncertain whether rasagiline alone is able to induce this malformation, or if their formation is due to the presence of LD or CD.
Based on these findings, it is recommended that rasagiline not be given to women during pregnancy. A warning has been included in the Product Monograph.
3.2.4 Summary and Conclusion
Based on studies of the pharmacological activity of rasagiline, the data demonstrate conclusively that it is an effective inhibitor of MAO-B, and at therapeutic doses it produces relatively minimal inhibitory activity of MAO-A. The interpretation of data
related to the neuroprotective effects of rasagiline is considered too speculative, and further investigations are needed to draw definitive conclusions. More importantly, further investigations are required to establish clinical relevance.
The most critical finding in the non-clinical assessment of this submission is the carcinogenic activity of rasagiline in mice. Due to the positive responses in some of the genotoxicity tests, including chromosomal aberrations and gene mutation in cultured mammalian cells, the carcinogenic activity of rasagiline is thought most likely to occur through a genotoxic mechanism. Together with the positive results of the mouse carcinogenicity study, the non-clinical data suggest that rasagiline could pose a potential carcinogenic risk to humans. Further analysis of the carcinogenic potential of rasagiline can be found in section 3.3.5 Assessment of Melanoma Risk.
3.3 Clinical basis for decision
3.3.1 Pharmacodynamics
The risk of hypertensive crisis with non-selective MAO inhibitors (or irreversible MAO-A inhibitors) following intake of indirectly-acting sympathomimetic amines such as tyramine is well recognized, and can be potentiated in subjects receiving concomitant levodopa (LD). Reversible MAO inhibitors or doses of irreversible MAO-B inhibitors (such as rasagiline) would not be expected to interact with tyramine. Therapeutic doses of the irreversible MAO-B inhibitor, selegiline however, have been associated with a fall in standing blood pressure in PD patients and diminished cardiovascular autonomic responses. Concomitant administration of selegiline with LD in late-stage patients can further exacerbate orthostatic hypotension, particularly if the dose of LD is not reduced. Rasagiline is a potent, highly selective, irreversible MAO-B inhibitor. Four studies were conducted to examine the effects of rasagiline with oral administration of tyramine. Tyramine exposure from fasting capsular administration can be over 4-fold higher than that achieved from a comparable amount of tyramine obtained from dietary sources. In all four studies with rasagiline, a moderate to high dose of tyramine was given in capsular form either under fasting conditions or with a light meal. These conditions are not equivalent to those expected when having a tyramine-rich/lipid-rich meal. Therefore, the amount of tyramine in such a meal is not anticipated to cause a substantial increase in blood pressure for patients treated with rasagiline at the intended dose.
Based on data from these four studies, as well as cardiovascular safety data obtained across the clinical program and literature review, no dietary tyramine restriction is proposed for the use of rasagiline in PD patients according to the prescribing information. A precaution has been included in the Product Monograph not to exceed the recommended rasagiline daily dose. This is especially important when used as adjunct therapy to LD.
3.3.2 Pharmacokinetics
Rasagiline is widely distributed, with a mean apparent volume of distribution (Vd) of approximately 243 L following a single intravenous (IV) administration of 2 mg. This large Vd is related to the irreversible binding of rasagiline to MAO in the body and is consistent with the findings in the population PK studies. There are no clinical data on tissue distribution of rasagiline in humans. However, results from in vitro binding studies show that rasagiline is over 90% protein-bound with slightly higher binding in males (92.4%) than in females (91.1%). In an ex vivo study at therapeutic rasagiline concentrations, about 70% of radioactivity (rasagiline plus metabolites) was protein-bound 1 hour post-dosing, declining to about 63% during 3 to 12 hours post-dosing. The ex vivo data in conjunction with in vitro protein binding data for human albumin (range 61.4% to 66.2%) and human alpha-1-acid glycoprotein (range 33.8% to 64.1%) indicate moderate protein binding of rasagiline. For this reason, rasagiline is unlikely to affect the protein binding of other drugs, and its disposition should not be affected by drug-drug interactions associated with protein displacement. The blood cell-to-plasma ratio for rasagiline-derived radioactive material ranged from 0.1 to 1.2, with a mean ratio across time of 0.2 to 0.7, indicating that association and/or distribution of rasagiline and/or its metabolites into blood cells is not extensive.
3.3.3 Clinical Efficacy
Three pivotal studies were submitted to support this application. Two of the studies were North American trials (TEMPO and PRESTO) and the third one was a European, Argentinian, and Israeli study (LARGO). TEMPO was a monotherapy trial with rasagiline alone, and PRESTO and LARGO were adjunctive therapy trials with rasagiline as an add-on to levodopa. In addition, the LARGO trial utilized entacapone as a positive control. All were multicentre, double-blind, placebo-controlled, parallel-group, Phase III studies. Study participants received either 0.5, 1.0, or 2.0 mg rasagiline daily, entacapone, or placebo. Efficacy measurements were based on the Unified PD Rating Scale (UPDRS) and on diaries measuring ON and OFF duration.
No formal dose-response studies were performed to measure the efficacy of Azilect and no studies utilizing doses less than 0.5 mg/day were performed for either monotherapy or adjunct therapy. All doses utilized in the pivotal clinical trials showed statistically significant efficacy, therefore no minimally effective dose for either monotherapy or adjunct therapy has been identified.
Treatment with 1.0 and 2.0 mg rasagiline per day for 6 months showed significant reductions in adjusted mean change from baseline for Total UPDRS score in early stage PD patients. These results were primarily driven by the Activities of Daily Living (ADL) and Motor sub-scales. Treatment was also associated with clinical benefit in terms of other relevant PD endpoints (symptomatic or treatment), and maintenance of baseline Quality of Life (QoL) level. This is in contrast to the deterioration in QoL level observed in the placebo group.
Treatment for over one year resulted in sustained clinical improvement in Parkinsonian symptoms. Patients treated with 1.0 mg per day tended to have greater clinical benefit at 1 year than those treated with 2.0 mg per day for 6 months.
Daily dosing of 0.5 mg and 1.0 mg rasagiline was found to be effective in decreasing total daily OFF-time in LD-treated subjects experiencing motor fluctuations. OFF-time is the period during which patients with PD experience partial or total immobility. The 1.0 mg dose provided a larger and more consistent benefit than 0.5 mg, with a readily manageable side effect profile. The decrease in motor fluctuations was apparent as early as 6 weeks. Total UPDRS and UPDRS Motor scores were also improved in patients previously optimized on LD therapy. These data suggest that rasagiline may extend the duration of LD benefit and also enhance its maximum antiparkinson effect.
3.3.4 Clinical Safety
Azilect generally appeared to be safe and well-tolerated at doses of 0.5-2.0 mg/day. The vast majority of adverse events (AEs) were considered to be mild or moderate in severity and few were considered to be treatment-related. A low incidence of dopaminergic AEs was observed, including incidences of dizziness, confusion, hallucinations, orthostatic/postural hypotension, and dyskinesia. Ataxia, accidental injury, arthralgia, and vomiting were also observed. There appeared to be a trend to dose-response for several of the AEs, including those considered dopaminergic, in both monotherapy and adjunct therapy trials. Even at the lowest dose, several AEs occurred with a higher frequency in rasagiline-treated groups than in the placebo/active comparator groups. There were no significant differences between groups in percent of premature discontinuations, percent of discontinuations due to AEs, or in time to discontinuation.
When rasagiline 1 mg/day was given as adjunct to levodopa, the data indicated a higher incidence of cardiovascular (CV)-related AEs for rasagiline-exposed patients compared to placebo or active comparator-treated patients, including first degree atrioventricular (AV) block. When rasagiline was given as monotherapy, the placebo group had a higher incidence of CV-related AEs in levodopa-treated patients.
There was no evidence of rasagiline interaction with tyramine.
The safety issues described above are considered mild. A separate analysis of the risk of melanoma from treatment with rasagiline is presented below.
3.3.5 Assessment of Melanoma Risk
A total of 20 cases of malignant melanoma in 19 patients (8 invasive and 12 in situ) were diagnosed and reported during the clinical program for rasagiline. Fifteen of these patients were treated with rasagiline, one patient was on placebo, and an additional three were diagnosed prior to treatment. The majority of cases (14) were diagnosed following the implementation of active skin examinations in all rasagiline clinical trials worldwide. Eleven of the 15 patients were noticed in studies without a concurrent placebo group. After the occurrence of the fourth case, the sponsor began investigating the occurrence of melanoma cases in the rasagiline development program. A "Dear Doctor" letter was sent to all the investigators participating in the rasagiline clinical trials in North America. Following the sixth case, the sponsor took active steps. The first action was the implementation of periodical skin examinations for all patients participating in clinical trials, as well as the addition of melanoma language in the Investigator's brochure and informed consent. It was, therefore, expected that this proactive step would inflate the number of diagnosed cases owing to the increased awareness that had been created. It was well recognized that there is no reliable database for a comparison to the general population and that under-reporting is likely to be substantial. Several activities were initiated to gain further understanding of the occurrence of melanoma cases seen in rasagiline trials.
It has been previously suggested however, that PD patients may be at increased risk for melanoma. To further investigate this hypothesis, two cohort studies in PD patients not treated with rasagiline, were initiated. Each of the studies carried out in North America and in Israel, was aimed at enrolling up to 2000 patients. Interim results from both studies show that the prevalence range of melanoma in PD patients (1 in 76 to 1 in 143) is higher than that found in the general population (1 in 160 to 1 in 700). An additional cohort study performed in the Danish population demonstrated a two fold increase in the relative risk for melanoma in PD patients compared with the general population. Another study has looked at the inverse issue, the rate of PD in patients with malignant melanoma. Here too, the rate of PD patients among melanoma patients exceeded the PD proportion in the general population. These studies provided substantial evidence and a more solid basis for the implied association between PD and melanoma. A molecular, genetic, or pathophysiological basis for this association can also be suggested in view of possible common pathways for the two diseases.
A third factor suggested as having a possible association with PD and melanoma is social class. There are assumptions that melanoma is more frequent in people of higher class. This is also true for PD patients (via inverse association with tobacco smoking). All attempts were made to collect data on melanoma risk factors from patients participating in the rasagiline clinical trials. Indeed, it can clearly be seen that the risk factor profile of patients in North America is considerably different than that of the European population that participated in rasagiline clinical trials. Irrespective of the stage of the disease (early or advanced), a higher proportion of patients in North America have risk factors, and the mean number of risk factors per patient is higher. Results obtained from the skin examinations and biopsies taken in rasagiline trials showed that other skin cancers and skin lesions were also more frequent in the North American patient population. The same was true for the percent of patients with a history of skin cancers.
Despite an additional European study conducted in Italy that found PD patients to have a lesser risk of melanoma, and the possibility that rasagiline could act to promote melanoma in susceptible individuals or via the drug's anti-apoptotic activity, there is substantial evidence to suggest that the risk of melanoma is higher in patients with PD than the general population. Therefore, it is equivocal whether the increased incidence of melanoma observed in rasagiline-treated patients is treatment-related.
A general Class Statement regarding the increased risk of melanoma for PD patients has been included in the Precautions section of the Product Monograph for all drugs indicated for Parkinson's disease.
3.4 Benefit/Risk Assessment and Recommendation
3.4.1 Benefit/risk assessment
The observed incidence of melanoma in the Azilect clinical development program was of concern and Health Canada recommended that a scientific advisory panel be convened to further examine this issue. However, due to logistical and scheduling problems, the formation of this committee was delayed. In the meantime, through further FDA-requested analyses of the clinical study data, as well as the recent submission of the second periodic safety update report (PSUR, July 3, 2005 to January 2, 2006), Health Canada concluded that sufficient data were available to perform a risk-benefit assessment.
Based on additional information from clinical study data and on epidemiologic evidence of increased melanoma in Parkinson's disease, it was concluded that Azilect did not appear to pose a significantly increased risk for the development of melanoma when compared to patients with Parkinson's disease who were not taking Azilect.
As a result of the information provided in the NDS for Azilect, a Class Statement, as follows, for all drugs indicated for Parkinson's disease, was developed to be included in the Product Monograph indicating that there is an increased risk of melanoma, and to advise on frequent monitoring by a dermatologist:
Epidemiological studies have shown that patients with Parkinson's disease have a higher risk (2- to approximately 6-fold higher) of developing melanoma than the general population. Whether the increased risk observed was due to Parkinson's disease or other factors, such as drugs used to treat Parkinson's disease, is unclear. For the reasons stated above, patients and providers are advised to monitor for melanomas frequently and on a regular basis when using TRADE NAME for any indication. Ideally periodic skin examinations should be performed by appropriately qualified individuals (e.g. dermatologists).
With the addition of this statement to the Product Monograph, Health Canada considers the benefit/risk assessment of Azilect to be favourable.
3.4.2 Recommendation
Based on the Health Canada review of data on quality, safety and efficacy, Health Canada considers that the benefit/risk profile of Azilect is favourable in the treatment of the signs and symptoms of idiopathic Parkinson's disease as initial monotherapy and as adjunct therapy to levodopa. The 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.
4 Submission Milestones
Submission Milestones: Azilect®
Submission Milestone | Date |
---|---|
Original Submission No. 087283 | |
Submission filed | 2003-10-08 |
Screening 1 | |
Screening Deficiency Notice issued | 2003-11-03 |
Response filed | 2003-11-20 |
Screening Acceptance Letter issued | 2003-11-26 |
Review 1 | |
Quality Evaluation complete | 2004-09-23 |
Clinical Evaluation complete | 2004-12-22 |
NON issued by Director General | 2004-12-24 |
Response filed | 2005-04-15 |
Screening 2 | |
Screening Acceptance Letter issued | 2005-05-16 |
Review 2 | |
Clinical Evaluation complete | 2005-06-24 |
Withdrawal/Cancellation by sponsor | 2005-10-11 |
Refiled Submission No. 101846 | |
Submission filed | 2005-10-17 |
Screening | |
Screening Acceptance Letter issued | 2005-12-01 |
Review | |
Biostatistics Evaluation complete | 2006-06-28 |
Quality Evaluation complete | 2006-05-18 |
Clinical Evaluation complete | 2006-08-09 |
Labelling Evaluation complete | 2006-08-08 |
NOC issued by Director General | 2006-08-17 |
Related Drug Products
Product name | DIN | Company name | Active ingredient(s) & strength |
---|---|---|---|
AZILECT | 02284642 | TEVA CANADA LIMITED | RASAGILINE (RASAGILINE MESYLATE) 0.5 MG |
AZILECT | 02284650 | TEVA CANADA LIMITED | RASAGILINE (RASAGILINE MESYLATE) 1 MG |