Summary Basis of Decision for Saphris ™

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
SaphrisTM

Asenapine, as asenapine maleate, 5 mg and 10 mg, Tablet, Sublingual

Merck Canada Inc.

Submission control no: 127427

Date issued: 2012-02-14

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:

SaphrisTM

Manufacturer/sponsor:

Merck Canada Inc.

Medicinal ingredient:

Asenapine, as asenapine maleate

International non-proprietary Name:

Asenapine

Strength:

5 mg and 10 mg

Dosage form:

Tablet

Route of administration:

Sublingual

Drug identification number(DIN):

  • 02374803 - 5 mg tablet
  • 02374811 - 10 mg tablet

Therapeutic Classification:

Antipsychotic

Non-medicinal ingredients:

Gelatin and mannitol

Submission type and control no:

New Drug Submission, Control Number 127427

Date of Submission:

2009-11-12

Date of authorization:

2011-10-07
2 Notice of decision

On October 7, 2011, Health Canada issued a Notice of Compliance to Merck Canada Inc. for the drug product, Saphris™.

Saphris™ contains the medicinal ingredient asenapine, as asenapine maleate, which is an antipsychotic drug.

Saphris™ is indicated for use in adults

  • for the treatment of schizophrenia.
  • for the acute treatment of manic or mixed episodes associated with bipolar I disorder. Saphris™ may be used as acute monotherapy or co-therapy with lithium or divalproex sodium.

Physicians who elect to use Saphris™ for extended periods should periodically re-evaluate the long-term risks and benefits of the drug for the individual patient. Saphris™ should be used with care in the elderly. Limited data on safety and efficacy are available in patients 65 years of age or older.

The mechanism of action of asenapine, as with other drugs having efficacy in schizophrenia and bipolar disorder, is not fully understood. However, based on its receptor pharmacology, it is proposed that the efficacy of asenapine is mediated through a combination of antagonist activity at dopamine 2 receptors and serotonin (5-HT) 5-HT2A receptors. Actions at other receptors, for example (e.g.) 5-HT1A, 5-HT1B, 5-HT2C, 5-HT6, 5-HT7, D3, and α2-adrenergic receptors, may also contribute to the clinical effects of asenapine.

The market authorization was based on quality, non-clinical, and clinical information submitted.

The efficacy of Saphris™ in the treatment of schizophrenia was evaluated in three fixed-dose and one flexible-dose, short-term (6 weeks), randomized, double-blind, placebo and active-controlled studies of patients who met the Diagnostic and Statistical Manual of Mental Disorders, 4th Edition (DSM-IV) criteria for schizophrenia and who were having an acute exacerbation of their schizophrenic illness. Of these four studies, two studies showed that Saphris™ [5 mg twice daily (BID)] was statistically superior to placebo on the Positive and Negative Syndrome Scale (PANSS) total score. In the third study, Saphris™ could not be distinguished from placebo; however the active control was superior to placebo. In the fourth study, neither Saphris™ nor the active control was superior to placebo. Yet, in a longer-term, open-label, randomized, placebo-controlled, double-blind, clinical study, Saphris™ was statistically significantly more effective than placebo in prolonging the time to relapse or impending relapse. Based on these five studies, there was sufficient evidence to support the efficacy of Saphris™ in the treatment of schizophrenia.

The efficacy of Saphris™ as monotherapy for the treatment of manic or mixed episodes associated with bipolar I disorder was evaluated in two similarly designed 3-week, randomized, double-blind, placebo-controlled, and active-controlled studies involving adult patients who met DSM-IV criteria for bipolar I disorder, with an acute manic or mixed episode, with or without psychotic features. Saphris™ demonstrated superior efficacy to placebo in the reduction of manic symptoms over 3 weeks. When the patients on active treatment extended their treatment for an additional 9 weeks, continuation of the effect of Saphris™ during the additional 9 weeks was also shown.

The efficacy of Saphris™ as co-therapy with lithium or divalproex sodium for the treatment of manic or mixed episodes associated with bipolar I disorder was demonstrated in a 12-week, randomized, placebo-controlled study. The use of Saphris™ as co-therapy with lithium or divalproex sodium resulted in superior efficacy compared to lithium or valproate monotherapy at Week 3 up to Week 12 post-randomization in the reduction of manic symptoms.

In the clinical studies, Saphris™ has been administered to more than 3,150 patients with schizophrenia or manic or mixed episodes associated with bipolar I disorder. The safety profile of Saphris™ was generally similar to other second-generation antipsychotic drugs that are dopamine 2 and 5-HT2A receptor blockers. A Serious Warnings and Precautions box is provided in the Product Monograph to warn against increased mortality in elderly patients with dementia. Analyses of thirteen placebo-controlled studies with various atypical antipsychotics (duration of 10 weeks) in elderly patients with dementia showed a mean 1.6-fold increase in death rate in the drug-related patients. Although the causes of death were varied, most of the deaths appeared to be either cardiovascular (e.g., heart failure, sudden death) or infectious (e.g., pneumonia) in nature.

Asenapine has anesthetic properties. Oral hypoesthesia and oral paresthesia may occur directly after administration and usually resolve within 1 hour.

Hypersensitivity reactions, including anaphylaxis and angioedema, have been observed in patients treated with asenapine. Therefore, Saphris™ is contraindicated in patients with a known hypersensitivity to this drug or to any ingredient in the formulation. In several cases, these reactions occurred after the first dose. These hypersensitivity reactions included: anaphylaxis; angioedema; hypotension; tachycardia; swollen tongue; dyspnea; wheezing; and rash. Patients should be informed of the signs and symptoms of a serious allergic reaction (e.g., difficulty breathing; itching; swelling of the face, tongue, throat; or feeling lightheaded, et cetera). Patients should be instructed to seek immediate emergency assistance if they develop any of these signs and symptoms.

Saphris™ (5 mg and 10 mg asenapine, as asenapine maleate) is presented as sublingual tablets. For the treatment of schizophrenia, the recommended starting and target dose of Saphris™ is 5 mg given twice daily. For the treatment of bipolar disorder, the recommended starting and target dose range of Saphris™ is 5 mg to 10 mg BID. To allow sublingual absorption, patients should be instructed to not eat or drink for 10 minutes after administration of Saphris™. Dosing guidelines are available in the Product Monograph. Saphris™ 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 Saphris™ are described in the Product Monograph.

Based on the Health Canada review of data on quality, safety, and efficacy, Health Canada considers that the benefit/risk profile of Saphris™ is favourable for the indications stated above.

3 Scientific and Regulatory Basis for Decision

3.1 Quality Basis for Decision

3.1.1 Drug Substance (Medicinal Ingredient)
General Information

Asenapine, as asenapine maleate, is the medicinal ingredient of Saphris™. Asenapine is a new antipsychotic agent with a high affinity for dopamine D2, serotonin-2, α-1 adrenergic and histamine-1 receptors, and no appreciable affinity for muscarinic cholinergic receptors.

Manufacturing Process and Process Controls

The drug substance is synthetically derived.

In-process controls performed during manufacture were reviewed and are considered acceptable. The specifications for the starting materials used in manufacturing the drug substance are also considered satisfactory.

Characterization

The route of synthesis of asenapine maleate supports the chemical structure assigned. Confirmation of the chemical structure was provided by elemental analysis and spectroscopic analysis.

Impurities and degradation products arising from manufacturing and/or storage were reported and characterized. These products were found to be within International Conference on Harmonisation (ICH) established limits and/or were qualified from batch analysis and toxicological studies, and therefore are considered acceptable.

Control of Drug Substance

The drug substance specifications and analytical methods used for quality control of asenapine maleate are considered acceptable.

Batch analysis results were reviewed and all results comply with the specifications and demonstrate consistent quality of the batches produced.

The drug substance packaging is considered acceptable.

Stability

Based on the long-term, real-time, and accelerated stability data submitted, the proposed retest period for the drug substance were supported and are considered satisfactory.

3.1.2 Drug Product
Description and Composition

Each Saphris™ tablet contains 5 mg or 10 mg asenapine, as asenapine maleate. The non-medicinal ingredients include gelatin and mannitol.

The 5 mg and 10 mg tablets are round, white to off-white, fast dissolving, sublingual tablets. The 5 mg tablet is debossed with "5" on one side, and the 10 mg tablet is debossed with "10" on one side.

Both strengths are packaged in peelable aluminum blister strips in cartons of 20, 60, or 100 sublingual tablets per carton. Not all pack sizes may be marketed.

All non-medicinal ingredients (excipients) found in the drug product are acceptable for use in drugs according to the Food and Drug Regulations. The compatibility of asenapine maleate with the excipients was demonstrated by the stability data presented on the proposed commercial formulation.

Pharmaceutical Development

Changes to the manufacturing process and formulation made throughout the pharmaceutical development are considered acceptable upon review.

Manufacturing Process and Process Controls

The method of manufacturing is considered acceptable and the process is considered adequately controlled within justified limits.

Control of Drug Product

Saphris™ is tested to verify that its identity, appearance, content uniformity, assay, disintegration time, water content, and levels of degradation products and drug-related impurities are within acceptance criteria. The test specifications and analytical methods are considered acceptable; the shelf-life and release limits, for individual and total degradation products, are within acceptable limits.

The validation process is considered to be complete.

Data from final batch analyses were reviewed and are considered to be acceptable according to the specifications of the drug product.

Although impurities and degradation products arising from manufacturing and/or storage were reported and characterized, these were found to be within ICH-established limits and/or were qualified from batch analysis and therefore, are considered acceptable.

Stability

Based on the real-time, long-term, and accelerated stability data submitted, the proposed 36-month shelf-life at 2-30°C for Saphris™ is considered acceptable when the tablets are stored in their original package.

The compatibility of the drug product with the container closure system was demonstrated through the stability studies. The container closure system met all validation test acceptance criteria.

3.1.3 Facilities and Equipment

The design, operations, and controls of the facilities and equipment that are involved in the production of Saphris™ are considered suitable for the activities and products manufactured.

All sites are compliant with Good Manufacturing Practices.

3.1.4 Adventitious Agents Safety Evaluation

The non-medicinal ingredient, gelatin, is of animal origin. A letter of attestation confirming that the material is not from a bovine spongiform encephalopathy (BSE)/ transmissible spongiform encephalopathy (TSE) affected country/area has been provided for this product indicating that it is considered to be safe for human use.

3.1.5 Conclusion

The Chemistry and Manufacturing information submitted for Saphris™ has demonstrated that the drug substance and drug product can be consistently manufactured to meet the approved specifications. 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 pharmacodynamic properties of asenapine showed similarities and differences when compared to other atypical antipsychotic drugs. Compared to established antipsychotic drugs, asenapine was generally more potent at binding to serotonin 5-HT1A, 5-HT2A, 5-HT2C, dopamine D1, D2, D3, and α1-, α2-adrenergic receptors in rat brain. Asenapine also had a high binding affinity for H1 receptors. In contrast, asenapine had very little binding affinity for muscarinic cholinergic receptors. Based on its binding properties and dose, asenapine is likely to show differential occupancy across serotonin, noradrenaline, dopamine, and histamine receptor subtypes. In particular, at doses producing occupancy at the D2 receptor, asenapine would be expected to have a preferentially stronger effect on the serotonergic receptor pathways. Further supportive evidence for potent and differential modulation of serotonin, noradrenaline and dopamine receptors was provided by studies examining the effects of chronic asenapine administration on the cortical and subcortical binding density of these receptors in rat brain.

Consistent with its 5-HT2A, D2, and α2 antagonist properties, asenapine increased D2, and α2 receptor levels while decreasing 5-HT2A receptor binding. Furthermore, asenapine produced distinct molecular changes such as increased levels of D1 receptor density in the medial prefrontal cortex, increased 5-HT1A receptors in the CA1 region of the hippocampus and had the ability to increase AMPA receptor density in the CA1 and CA3 sub-regions of the hippocampus. Asenapine did not significantly affect 5-HT2C receptor binding density at the doses tested despite high affinity and antagonist activity at this receptor.

The two enantiomers of asenapine generally showed similar pharmacological properties to asenapine in vitro and in vivo and would therefore be expected to contribute to the activity of asenapine. Contribution of pharmacological activity from the metabolites of asenapine is considered to be unlikely at the therapeutic dose range of 5 to 10 mg BID.

The results from the human Ether-à-go-go Related Gene (hERG) assay suggest a low risk for QT prolongation. The main haemodynamic effects of asenapine were a decrease in arterial blood pressure, probably as a result of α1-adrenoceptor blocking activity, and orthostatic hypotension, which were less apparent after sublingual administration compared to oral administration.

In a respiratory study, asenapine caused a transient central depressor effect (increase in tidal volume, expired volume, enhanced pause) 20 minutes after a 5 mg/kg oral dose in rats.

Evaluation of gastrointestinal (GI) effects revealed GI ulceration after a single oral asenapine dose of 1 or 10 mg/kg in fasted rats, but not after 5 repeated doses in non-fasted rats. In guinea pigs, asenapine increased the frequency and decreased the amplitude of intestinal contractions after 1 or 5 mg/kg were administered intravenously. In monkeys previously sensitized to neuroleptics, asenapine caused extrapyramidal side effects (dystonia and bradykinesia), and decreased locomotor activity and reactivity to environmental stimuli at a dose range less than the 10 mg BID human therapeutic sublingual dose.

3.2.2 Pharmacokinetics
Absorption

Asenapine was well absorbed from all sites of administration, although systemic exposure after oral administration was relatively low due to extensive first-pass metabolism. Exposure in non-clinical species was dose-dependent and generally dose-proportional, comparable in males and females, and did not change with repeated dosing.

Distribution

After oral and sublingual administration, asenapine was widely distributed throughout the body and readily penetrated the central nervous system (CNS). Asenapine and its metabolites partitioned into melanin-containing tissues (skin and eyes) but no long-term retention was found.

Plasma/serum protein binding was high in all species tested including humans (≥96%). Asenapine was, at best, a weak substrate of the human P-gp transporter.

Asenapine drug-related radioactivity crossed the placental barrier of rats and rabbits. In the rat, asenapine drug-related radioactivity was measured in the milk of lactating dams. Therefore, it is recommended that women receiving asenapine should not breast feed.

Metabolism

Asenapine was extensively metabolized via oxidative and conjugative pathways. N-desmethyl asenapine was the main Phase 1 metabolite in all of the species and asenapine N-oxide was prominent in animals (mice, rats, rabbits, and dogs), but not humans.

The cytochrome P450 (CYP) 1A2 enzyme was the most important CYP enzyme in the metabolism of asenapine, with contributions from CYP3A4 and CYP2D6.

In vitro, asenapine did not inhibit most human CYP enzymes, but did inhibit CYP2D6 and CYP1A2. Asenapine may interact with drugs that are significantly metabolized by CYP2D6. Inhibition of CYP1A2 occurred at a higher inhibitory concentration (IC50 ) and is unlikely to result in an interaction.

Excretion

Asenapine and its metabolites were excreted in the urine and faeces; the percentages varied among species.

3.2.3 Toxicology

The non-clinical toxicology program conducted for asenapine was considered appropriate overall. In vivo toxicity was evaluated following oral and/or parenteral (intravenous or subcutaneous) administration of asenapine.

Single-Dose Toxicity

Following oral (rats and dogs) or intravenous (rats) administration of single high doses, asenapine had a moderate order of acute toxicity referable to the effects on the CNS.

Repeat-Dose Toxicity

Many of the effects observed in the toxicology studies reflect the pharmacologic activities that affect the dopaminergic and serotonergic mechanisms.

Morbidity/mortality following single-dose or repeated-dose administration of relatively high-dose levels was referable to effects on the CNS. While CNS effects in animals can be reasonably considered to represent an extension of the pharmacologic activity, it should be noted that clinical signs referable to the CNS, albeit mild, were present in animals (mice, rats, and dogs) at low-dose levels that resulted in asenapine systemic exposure values (AUC) lower than that anticipated in human patients taking asenapine 10 mg BID. Furthermore, prolactin-mediated effects on mammary glands and oestrus cycle disturbances were observed. Changes to the histopathology of the female reproductive organs (ovary, uterus, and vagina) and mammary gland that indicate a discrepancy or disturbance in oestrus cycle-related morphology can be related to potential effects on prolactin.

However, there are a number of effects in the toxicology studies that are not explained by the pharmacologic activity, for example (e.g.), increased levels of liver enzymes and liver weight; histopathologic changes in the liver; increased levels of urea and creatinine; thyroid follicular hypertrophy; and treatment-related tachycardia. Generally, the mild effects on the liver, kidney, and thyroid occurred at relatively high-dose levels. However, effects of asenapine on the reproductive organs and mammary glands in females, the adrenals, and the effects on the electrocardiograms in dogs, occurred in animals at lower systemic exposures than anticipated in patients taking 10 mg BID. The effects noted in the long-term toxicology studies in rats and dogs were not always reversed following a 6-week treatment-free recovery period.

In a juvenile toxicity study in rats, treatment-related hyperactivity was still present during a 4-week period following cessation of asenapine administration. In a similar study, adult rats also showed hyperactivity immediately after cessation of dosing, with a gradual decrease in activity level over 8 weeks. Apparently similar effects on activity in juvenile and adult rats have been described with haloperidol.

Genotoxicity

Asenapine is not considered genotoxic based on the bacterial mutagenicity assays and in vitro and in vivo mammalian studies.

Carcinogenicity

In a 2-year carcinogenicity study in mice, there was no clear evidence of carcinogenicity in mice administered asenapine 0.5, 1.5, and 5 to 4 mg/kg (males) or 7.5 to 5 mg/kg (females) once daily, subcutaneously. A statistically higher incidence of microscopic lymphomas in 7.5/5 mg/kg females compared with vehicle controls was considered to represent an incidental finding unrelated to asenapine as there was no increase in lymphomas in treated female groups compared with concurrent untreated control female rats or with historical control data.

Asenapine administered subcutaneously to male and female rats at 0.3, 1.2, and 3 to 5 mg/kg once daily for approximately 2 years did not result in organ specific, systemic, or local tumorigenic potential.

Reproductive and Developmental Toxicity

Asenapine caused significant embryo-foetal toxicity, including pup loss, at doses with no safety margin relative to human exposure. Therefore it poses a significant risk to the embryo/foetus and should not be taken during pregnancy unless clearly necessary and only if the potential benefit outweighs the potential risk to the foetus. In a study conducted in juvenile rats, the potential for a developmental long-lasting behavioral effect was evident from increased activity that developed after cessation of the repeated daily dosing regimen, with evidence of recovery seen only in males, but not females, after 30 days of recovery, at all dose levels tested. In the rat, asenapine drug-related radioactivity was measured in the milk of lactating dams. Therefore it is recommended that women taking asenapine should not breast feed.

Local Tolerance

Asenapine-containing tablets administered sublingually at dose levels of 1.6 or 15 mg BID for 7 consecutive days to female dogs did not cause local irritation at the dosing site.

Antigenicity and Phototoxicity

Asenapine did not cause humoral or cellular antigenicity in guinea-pigs.

Asenapine was non-phototoxic in the in vitro 3T3 Neutral Red Uptake assay.

3.2.4 Summary and Conclusion

The non-clinical studies for this drug submission are considered suitable. Adequate statements are in place in the Product Monograph to address the identified safety concerns. In view of the intended use of Saphris™, there are no pharmacological/toxicological issues within the submission which preclude approval of the requested product indication. The non-clinical studies together with the clinical studies are considered sufficient to characterize the safety profile of Saphris™.

3.3 Clinical basis for decision

3.3.1 Pharmacodynamics

Asenapine has a high affinity for serotonin 5-HT1A, 5-HT1B, 5-HT2A, 5-HT2B, 5-HT2C, 5-HT5, 5-HT6, and 5-HT7 receptors (Ki values of 2.5, 2.7, 0.07, 0.18, 0.03, 1.6, 0.25, and 0.11 nM, respectively); dopamine D2A, D2B D3, D4, and D1 receptors (Ki values of 1.3, 1.4, 0.42, 1.1, and 1.4 nM, respectively); α1A, α2A, α2B and α2C-adrenergic receptors (Ki values of 1.2, 1.2, 0.33, and 1.2 nM, respectively); and histamine H1 receptors (Ki value 1.0 nM); and moderate affinity for H2 receptors (Ki value of 6.2 nM).

Asenapine had no appreciable affinity for muscarinic cholinergic receptors.

The in vitro assays showed that asenapine acts as an antagonist at most of the 5HT1 receptors. However, as with other antipsychotic drugs showing appreciable affinity for 5HT1A receptors, electrophysiological studies showed that asenapine is a partial agonist at this receptor.

3.3.2 Pharmacokinetics
Absorption

Following sublingual administration, asenapine was rapidly absorbed with peak plasma concentrations occurring within 0.5 to 1.5 hours. Up to a dose of 5 mg BID, the systemic exposure of asenapine increased proportionally to the dose. At higher doses [that means (i.e.) 5-10 mg BID] deviation from dose-proportionality was observed.

The absolute bioavailability of sublingual asenapine at 5 mg was estimated to be approximately 35%. Part of the asenapine dose administered via the sublingual route was swallowed, as the AUC of N-desmethylasenapine after sublingual administration of asenapine was considerably lower after treatment with charcoal, than without charcoal pre-treatment, while the AUC of asenapine was essentially unchanged. The absolute bioavailability of asenapine when swallowed was very low (<2% with an oral tablet formulation).

Intake of water 2 or 5 minutes after asenapine sublingual administration resulted in decreased systemic exposure. A high-fat meal immediately before asenapine dosing, as well as food given 4 hours after asenapine administration also reduced asenapine exposure.

Distribution

Asenapine was rapidly distributed with a large volume of distribution. It was highly bound (95%) to plasma proteins, including albumin and α1-acid glycoprotein.

Metabolism

The primary metabolic pathways for asenapine were direct glucuronidation by the protein UDP-glucuronosyltransferase 1-4 (UGT1A4) and oxidative metabolism by CYP isoenzymes (predominantly CYP1A2).

Metabolite profiling demonstrated at least 5 different peaks in plasma, at least 15 different peaks in urine, and at least 13 different peaks in faeces. The peaks in plasma were associated with asenapine, N-desmethylasenapine, asenapine N+-glucuronide, N-desmethylasenapine-N-carbamoylglucuronide and asenapine 11-O-sulfate. None of these metabolites are expected to provide a relevant contribution to the pharmacological activity of asenapine due to lower affinity for the receptors or the inability to cross the blood brain barrier. Asenapine N+-glucuronide and unchanged asenapine were the major drug-related entities in plasma samples.

Excretion

After administration of a single dose of radiolabelled asenapine, approximately 90% of the dose was recovered: approximately 50% in the urine; and 40% in the faeces.

Special Populations

Renal Impairment

The pharmacokinetics of asenapine and N-desmethylasenapine following a single dose of 5 mg asenapine were similar among subjects with varying degrees of renal impairment and subjects with normal renal function, indicating that dosage adjustment based upon the degree of renal impairment is not required.

Hepatic impairment

The pharmacokinetics of asenapine and its metabolites N-desmethylasenapine and asenapine N+-glucuronide following a single dose of 5 mg asenapine were similar among subjects with mild or moderate hepatic impairment (Child-Pugh Class A and B) and subjects with normal hepatic function, indicating that dosage adjustment is not required for these subjects. In subjects with severe hepatic impairment (Child-Pugh Class C) substantial increases in exposure were observed: 7-fold for asenapine; 3-fold for N-desmethylasenapine; and 2-fold for asenapine N+-glucuronide. Based on these observations,asenapine is not recommended for patients with severe hepatic impairment.

Adolescents

At the 5 mg BID dose level, asenapine pharmacokinetics in the adolescent population were similar to those observed in adults. In adolescents, the 10 mg BID dose did not result in higher exposure of asenapine compared to the 5 mg BID dose. This appears to result from subjects partially swallowing the 10 mg dose in the study.

Elderly

In a preliminary data set, exposure to asenapine was on average 30% higher in elderly patients (65-85 years of age) compared to younger adult patients. No dose adjustment is necessary.

Race

A comparative study in Caucasian and Japanese subjects did not detect an effect of race on asenapine pharmacokinetics.

Gender

No clinically significant differences in pharmacokinetics were observed between men and women.

Drug Interaction Studies

Asenapine is cleared primarily through direct glucuronidation and oxidative metabolism (predominantly by CYP1A2). The potential effects of inhibitors and an inducer of several of these enzyme pathways on asenapine pharmacokinetics were studied. With the exception of fluvoxamine (a strong CYP1A2 inhibitor), none of the drugs resulted in clinically relevant alterations in asenapine pharmacokinetics. A summary of the effects of these coadministered drugs is included in the Saphris™ Product Monograph.

3.3.3 Clinical Efficacy
Efficacy in Schizophrenia

The efficacy of Saphris™ in the treatment of schizophrenia was evaluated in five pivotal studies. Four studies (Studies 041004, 041021, 041022, 041023) were short-term (6 weeks), randomized, double-blind, placebo and active-controlled studies of patients who met DSM-IV criteria for schizophrenia and were having an acute exacerbation of their schizophrenic illness. Three of the placebo/active controlled studies were fixed-dose studies: a Phase II study with Saphris™ 5 mg BID (Study 041004); and two Phase III studies with Saphris™ 5 mg BID and 10 mg BID (Studies 041021 and 041023). Study 041022 was a flexible dose study (Saphris™ 5 mg - 10 mg BID). All four studies included an active control group (risperidone, olanzapine, or haloperidol) but were not powered to compare Saphris™ with the active comparator. The fifth pivotal study (Study A7501012) was a standard relapse prevention study with six months of open-label treatment with Saphris™ followed by randomization to placebo or Saphris™ for an additional six months. This study allowed flexible dosing of Saphris™ 5-10 mg BID.

Three out of four of the short-term 6-week studies were Phase III studies that included approximately 100 patients per treatment group. The fourth short-term study was a Phase II study (Study 041004) with approximately 60 patients per group. In the relapse prevention study, there were approximately 200 patients in each of the randomized treatment arms.

The primary efficacy rating scale was the Positive and Negative Syndrome Scale (PANSS). The primary efficacy analysis was a comparison of the least squares (LS) mean changes from baseline to endpoint (LOCF) in the PANSS total score for each Saphris™ treatment group versus (vs.) placebo. The primary and secondary efficacy endpoints included change from baseline on PANSS total score and each of the PANSS subscale scores (PANSS positive, negative and general psychopathology subscales), the PANSS Marder factor scores, and the Clinical Global Impression severity score (CGI-S).

Pivotal Study Results

Study 041004 was the placebo-controlled Phase II study which used Saphris™ at 5 mg BID, and included an active control (risperidone). Saphris™ was significantly more effective than placebo in reducing the symptoms of schizophrenia at Weeks 2 through 6 (endpoint), as measured by total PANSS score. Saphris™ was also significantly more effective than placebo in reducing PANSS subscale scores at Weeks 3 through 6. For Clinical Global Impression (Severity of Illness, and Global Improvement), both Saphris™ and risperidone were significantly better than placebo at endpoint, and showed similar magnitudes of change. Study 041004 was deemed a positive study in support of efficacy of Saphris™ in acute treatment of schizophrenia.

Study 041021 had two fixed-dose treatment arms of Saphris™ (5 mg BID and 10 mg BID), as well as an active control (olanzapine). Statistically significant results were seen only for olanzapine. Therefore, Study 041021 was deemed a negative study.

Study 041022 was a flexible-dose study (initial Saphris™ target dose was 10 mg BID with an option to reduce to 5 mg BID) with an active control (olanzapine). In this study, neither Saphris™ nor olanzapine was statistically superior to placebo. Study 041022 was deemed a failed study.

Study 041023 compared Saphris™ 5 mg BID, Saphris™ 10 mg BID, haloperidol 4 mg BID, and placebo (approximately 110 patients per group). Statistically significant differences were observed in the primary efficacy analysis (baseline to endpoint in the PANSS total score) with patients treated with Saphris™ 5 mg BID and haloperidol 4 mg BID, compared to patients treated with placebo. The results from the Saphris™ 10 mg BID group were not significant. However, in the observed case and repeated measure analyses, the Saphris™ 10 mg BID group also reached statistically significance compared to placebo. This was also seen on some secondary efficacy endpoints (e.g., PANSS positive subscale score, and PANSS response rates). Overall, Study 041023 was deemed positive. However, the study highlighted that Saphris™ 5 mg BID was as, if not more, efficacious than the higher dose.

Based on the four short-term acute treatment studies, there was one negative study, one failed study, and two studies supporting Saphris™ efficacy at the 5 mg BID dose, with equivocal findings at the 10 mg BID dose.

Study A7501012 was a relapse prevention study. While this study design is not considered to support a relapse prevention indication, it is considered of value for the assessment of drug efficacy. In this study, for those patients who were able to tolerate and remain on Saphris™ for a 26-week open-label period prior to randomization to placebo or ongoing Saphris™, Saphris™ was shown to be significantly superior to placebo at controlling symptoms of schizophrenia. While this study allowed flexible dosing (Saphris™ 5-10 mg BID), this study supports the efficacy of Saphris™ 10 mg BID as the majority of patients were receiving this dose compared to the 5 mg  BID dose (80% vs. 20% respectively).

On the basis of all 5 pivotal studies, there is sufficient evidence to support the efficacy of Saphris™ in the treatment of schizophrenia. However, there was no evidence to suggest that a 10 mg BID dose was superior to a 5 mg BID dose. Thus, the recommended dose should be 5 mg BID. (Early Phase II studies failed to demonstrate efficacy at doses lower than 5 mg BID).

Efficacy in Manic or Mixed Episodes Associated with Bipolar I Disorder

The efficacy of Saphris™ in the treatment of manic or mixed episodes associated with bipolar I disorder was assessed in two similarly designed 3-week, Phase III studies (Studies A7501004, A7501005). The evaluation also included longer-term studies; a 9-week extension study (A7501006), and a 12-week co-therapy study (A7501008). Of these four studies, three were considered pivotal. Study A7501006 was considered non-pivotal.

Studies A7501004 and A7501005 were 3-week, randomized, double-blind, placebo-controlled and active-controlled (olanzapine), flexible-dose studies (Saphris™ 10 mg BID with an option to decrease to 5 mg BID) of identical design, in patients with acute manic or mixed episodes associated with bipolar I disorder. Patients were randomized in a 2:2:1 ratio for treatment with Saphris™, olanzapine, or placebo. Each of these two monotherapy studies randomized approximately 100 patients to placebo and 200 patients to Saphris™.

Study A7501006 was a 9-week extension study of patients who completed Studies A7501004 and A7501005 and who were on active treatment. The patients [number (n) = 397] could continue on the same active treatment [Saphris™ 5-10 mg BID or olanzapine 5-20 mg once daily (QD)] for an additional 9 weeks. The primary objective of Study A7501006 was to demonstrate non-inferiority acute efficacy of Saphris™ compared with olanzapine for up to 12 weeks. However, the design was not adequate to assess non-inferiority of two active treatments as the study only continued patients on the same drug that they had a good experience with in the previous studies, keeping them on the same active drug without further randomization. The study was not regarded as a pivotal study. However, the study did demonstrate the continuation of effect of Saphris™ during the additional 9 weeks.

Study A7501008 was a 12-week study conducted in patients who had an acute manic or mixed episode associated with bipolar I disorder, and who had been receiving lithium or valproate for at least 2 weeks prior to randomization. Patients were randomized (1:1) to Saphris™ or placebo, with lithium and/or valproate continued for the duration of the study. Treatment with Saphris™ started at 5 mg BID and thereafter the dose was flexible Saphris™ 5-10 mg BID. This combination study with lithium and/or valproate randomized approximately 106 patients to each group (i.e., Saphris™ or placebo).

The primary rating instrument used for assessing manic symptoms in the efficacy studies was the Young Mania Rating Scale (YMRS). Patients were also assessed on the Clinical Global Impression - Bipolar (CGI-BP) scale. The primary efficacy endpoint was the change from baseline to Day 21 on the YMRS total score based on LOCF analysis. For Study A7501006, the primary efficacy endpoint was as above, but was determined at Day 84 (12 weeks). Study A7501008 used the same primary efficacy endpoint at Day 21, and measurements continued up to Day 84.

Pivotal Study Results

Study A7501004 was conducted in the United States of America, Eastern Europe, India, and a few sites in south-east Asia. The study showed a positive outcome for Saphris™ versus placebo [probability (p) = 0.007], but the study was confounded by a significant treatment-by-centre interaction. Two large sites in India showed differences on YMRS of more than 25 points between Saphris™ and placebo. While these two sites constituted only 3% of the study population, their exclusion not only eliminated the significant treatment-by-site interaction, but also resulted in a non-significant study outcome (p = 0.1412).

Approximately 60% of the dataset was from the United States, 20% from Eastern Europe, and 20% from Asia. There were notable differences between regions in efficacy outcomes. In Asia, there was a marked and significant treatment effect for both active treatment groups (Saphris™ and olanzapine) compared to placebo, while in the much larger United States subgroup, or combined United States/Europe subgroup Saphris™ was no better than placebo, while olanzapine showed superiority over placebo. The treatment-by-centre interaction and the regional differences remain unexplained.

This study was considered equivocal due to the treatment-by-centre interaction on the primary outcome, the lack of any observable treatment superiority of Saphris™ over placebo in the United States/Europe (which contributed 80% of the study population), as well as the consistent superiority of olanzapine compared to placebo across efficacy outcomes, and the lack of such with Saphris™.

Study A7501005, in contrast to Study A7501004, showed a positive outcome for Saphris™ versus placebo (p<0.0001), in the absence of a significant treatment-by-centre interaction. While Study A7501005 did show regional differences similar to Study A7501004, the United States and combined United States/Europe subgroups in Study A7501005 showed significant treatment effects of Saphris™ over placebo, (although the effect was smaller than in the Asian data subset). This study is considered a positive study, in support of the efficacy of Saphris.

Studies A7501004 and A7501005 were identical in study protocols. When the data from these two studies were pooled together, Saphris™ showed statistical superiority over placebo on the primary endpoint not only for the entire study population, but also for the United States/Europe subgroup.

Study A7501008 showed a positive outcome for Saphris™ versus placebo at Week 3 and at Week 12. There was no substantial regional difference in treatment outcome patterns. This study supports the efficacy of Saphris™ in the treatment of mania, in the presence of ongoing treatment with valproate or lithium. However, it does not support a separate augmentation therapy indication for Saphris™. The study population was not clearly defined as “non-responder” or “partial responder” to valproate or lithium.

Given that two out of three of the pivotal studies showed positive results, and that the pooled results of the Europe and United States subpopulation from Studies A7501004 and A7501005 also showed superiority over placebo, it is considered that overall there is sufficient evidence to support efficacy of Saphris™ in the treatment of acute mania and mixed states associated with bipolar disorder, as acute monotherapy or co-therapy with lithium or divalproex sodium.

Saphris™ dosing recommendations with respect to bipolar disorder are somewhat problematic, as no fixed-dose studies were conducted. The monotherapy studies started patients on a 10 mg BID dose regime, while the combination therapy study started patients on a 5 mg BID dose regime of Saphris™. Both studies allowed for up and down titration during the study within the dose range of 5-10 mg BID. In the monotherapy studies, almost all patients remained on the initial 10 mg BID dose, while in the combination study approximately 55% of patients remained on 5 mg BID while approximately 45% had a dosage increase to 10 mg BID at some point during the study. These data are far from optimal with respect to making the most appropriate dosing recommendations for the use of Saphris™ in treatment of bipolar mania. Nevertheless, Saphris™, is reasonably well-tolerated at the 10 mg BID dose, and there is evidence of efficacy at this dose. Therefore, the wording in the Saphris™ Product Monograph recommends a dosage range within 5 mg to 10 mg BID, highlighting the limitation of the current supporting clinical data. Both the European Medicines Agency (EMA) and the United States Food and Drug Administration (FDA) have approved the use of Saphris™ in the treatment of acute mania based on the same clinical study data.

3.3.4 Clinical Safety

In the Phase II or Phase III studies, Saphris™ sublingual tablets were administered at doses of 5-10 mg BID to approximately 3,100 patients with schizophrenia or mania associated with bipolar I disorder. In these studies, approximately 1,700 patients were treated for at least 12 weeks, 1,300 patients for at least 6 months, and almost 800 patients for one year.

As of December 1, 2008, there were 19 deaths (0.5%) in all of the Saphris™ groups: 1 death (0.1%) in the placebo groups; 5 deaths (0.4%) in the olanzapine groups; and 1 death (0.9%) in the haloperidol groups. There were some deaths by suicide, and deaths from a variety of different medical conditions, with no obvious pattern. Furthermore, these clinical study fatalities did not reveal a specific association with Saphris™ treatment.

In the Phase II/III database the following serious adverse events of potential relevance to Saphris™ were observed: syncope (0.2%, n = 7); neuroleptic malignant syndrome (NMS) (0.1%, n = 3); hyponatraemia (0.1%, n = 3); and rhabdomyolysis (0.1%, n = 2). Syncope and NMS are observed with most antipsychotic medications. Hyponatraemia is associated with schizophrenia (i.e., secondary to polydipsia). Rhabdomyolysis was secondary to hyponatraemia and its sequalae or another primary cause.

Common adverse events (at least 2.0% and at least twice the incidence of placebo) associated with Saphris™ (5-10 mg BID) in the short-term placebo-controlled studies (6-week schizophrenia and 3-week bipolar mania) were:

  • Sedation (9.1% Saphris™, 4.4% placebo);
  • Somnolence (8.4% Saphris™, 2.3% placebo);
  • Akathisia (5.4% Saphris™, 2.4% placebo);
  • Oral hypoaesthesia (5.0% Saphris™, 0.7% placebo);
  • Increased weight (3.5% Saphris™, 0.4% placebo);
  • Dystonia (2.5% Saphris™, 0.6% placebo);
  • Increased appetite (2.5% Saphris™, 0.7% placebo);
  • Parkinsonism (2.4% Saphris™, 1.1% placebo).

The incidence of the common adverse events was similar for both bipolar and schizophrenia studies, except for a somewhat higher incidence of sedation, somnolence and dizziness in the bipolar studies.

In the short-term, schizophrenia studies where fixed doses of Saphris™ were studied, the following adverse events showed a dose-related trend:

  • Akathisia (2.4% placebo, 0.7% Saphris™ <5 mg BID, 4.0% Saphris™ 5 mg BID, 10.6% Saphris™ 10 mg BID);
  • Tremors (1.4% placebo, 1.7% Saphris™ <5 mg BID, 1.8% Saphris™ 5 mg BID, 4.3% Saphris™ 10 mg BID).

In the Phase II/III studies, the safety profile of Saphris™ was similar to other second-generation antipsychotic drugs that block dopamine 2 and 5HT2 receptors. The only unique adverse event associated with Saphris™ that was identified in the clinical study database was that of hypoesthesia. The pharmacological profile of Saphris™ does demonstrate some local anesthesia activity of the drug. This is only clinically apparent due to the route of administration i.e., sublingual (to avoid a very large first pass effect, which limits oral bioavailability).

In addition to clinical study safety data, there is a post-marketing safety database (as Saphris™ has been marketed for treatment of schizophrenia and bipolar disorder in the United States and Puerto Rico since October 2009, and most recently Germany since December 2010). The post-marketing data (estimated at 1,300 patient years of exposure based on sales) identified two other potential safety signals. As of the most recent periodic safety update (February 2011), there have been four cases of serious hypersensitivity reactions. All of these cases involved throat or tongue swelling. In reviewing the available details on these cases, it was not entirely clear what role numbness of the oropharynx (caused by the anesthetic properties of asenapine) and acute dystonic reactions of tongue and pharynx may have played in the symptoms experienced.

In addition to the above potential hypersensitivity reactions, there were also 4 potential cases of Stevens-Johnson syndrome (SJS). Based upon the available information, a causal relationship between Saphris™ and the onset of SJS cannot be determined. All 4 cases either contained confounding factors or contained information that does not fit well with a diagnosis of SJS (i.e., resolution while continuation of Saphris™ treatment). The Saphris™ Product Monograph contains warnings with respect to the possibility of hypersensitivity reactions associated with Saphris™.

3.4 Benefit/Risk Assessment and Recommendation

3.4.1 Benefit/Risk Assessment

The pivotal studies in the current submission are deemed adequate to support the efficacy of Saphris™ for acute indications in the treatment of schizophrenia and mixed or manic episodes associated with bipolar I disorder.

In the treatment of schizophrenia, Saphris™ (5 mg BID with equivocal findings of the 10 mg BID dose) was statistically superior to placebo on the PANSS total score in two of the four short-term studies, as well as in a 26-week longer-term study where Saphris™ was also statistically significantly more effective than placebo in prolonging the time to relapse or impending relapse.

In the treatment of acute mania, two out of three of the Saphris™ studies (one monotherapy and one with concurrent use of valproate or lithium) were positive in support of efficacy. The third study was deemed equivocal; however, when the results of this study were pooled together with the positive study of the same design, Saphris™ showed superiority over placebo. Therefore, there was sufficient evidence to support the efficacy of Saphris™ in the treatment of acute mania and mixed states associated with bipolar I disorder.

The safety profile of Saphris™ is similar to other second-generation antipsychotic drugs that block dopamine 2 and 5HT2 receptors. The risk of increased mortality in the treatment of elderly patients with dementia has been highlighted in a Serious Warnings and Precautions box in the Product Monograph. This risk is valid for all atypical antipsychotic drugs. Thirteen placebo-controlled studies with various atypical antipsychotics (duration of 10 weeks) showed a mean 1.6-fold increase in death rate in this patient population.

The only unique adverse event associated with Saphris™ that was identified in the clinical study database was that of hypoesthesia. Saphris™ has anesthetic properties, and oral hypoesthesia and oral paresthesia may occur directly after administration and usually resolve within 1 hour.

Hypersensitivity reactions, including rash, anaphylaxis and angioedema, have been observed in patients treated with Saphris™. In several cases, these reactions occurred after the first dose. Saphris™ is contraindicated in patients with a known hypersensitivity to this drug or to any ingredient in the formulation. The risk of hypersensitivity reactions is emphasized in the Warnings and Precautions section of the Saphris™ Product Monograph.

Overall, the data presented in this application demonstrated that Saphris™ has a favourable benefit to risk profile. Restrictions to manage risks associated with the identified safety concerns have been incorporated into the Saphris™ Product Monograph.

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 Saphris™ is favourable for the treatment of schizophrenia, and for the acute treatment of manic or mixed episodes associated with bipolar I disorder. Saphris™ may be used as acute monotherapy or co-therapy with lithium or divalproex sodium. 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: SaphrisTM

Submission MilestoneDate
Submission filed:2009-11-12
Pre-submission meeting:2009-12-22
Screening 1
Screening Acceptance Letter issued:2010-12-24
Review 1
Quality Evaluation complete:2011-03-31
Clinical Evaluation complete:2011-10-06
Labelling Review complete:2011-10-07
Notice of Compliance issued by Director General:2011-10-07