Summary Basis of Decision for Champix

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
Champix

Varenicline tartrate, 0.5 mg and 1.0 mg, Tablet, Oral

Pfizer Canada Inc.

Submission control no: 104007

Date issued: 2008-01-30

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:

Champix

Manufacturer/sponsor:

Pfizer Canada Inc.

Medicinal ingredient:

Varenicline tartrate

International non-proprietary Name:

Varenicline tartrate

Strength:

0.5 mg and 1.0 mg

Dosage form:

Tablet

Route of administration:

Oral

Drug identification number(DIN):

  • 02291177 0.5 mg/tablet
  • 02291185 1.0 mg/tablet

Therapeutic Classification:

Smoking-cessation aid

Non-medicinal ingredients:

Tablet: Microcrystalline cellulose, anhydrous dibasic calcium phosphate, croscarmellose sodium, colloidal silicon dioxide, and magnesium stearate.
Film-coating: Hypromellose, titanium dioxide, polyethylene glycol and triacetin. The 1.0 mg tablet also contains FD&C Blue #2/ I ndigo Carmine Aluminum Lake as a colouring agent.

Submission type and control no:

New Drug Submission, Control No. 104007

Date of Submission:

2006-02-03

Date of authorization:

2007-01-24

*™ Pfizer Products Inc.
Pfizer Canada Inc., licensee

2 Notice of decision

On January 24, 2007, Health Canada issued a Notice of Compliance to Pfizer Canada Inc. for the drug product Champix.

Champix contains the medicinal ingredient varenicline tartrate which is classified as a smoking-cessation aid.

Champix is indicated for smoking-cessation treatment in adults in conjunction with smoking-cessation counselling. The proposed mechanism of action is the binding of varenicline to the α4β2  subtype of the nicotinic acetylcholine receptor (the subtype believed to be associated with nicotine addiction). As a partial agonist, varenicline activates the receptor to produce a partial physiological response as compared to nicotine, while simultaneously preventing nicotine binding. How this receptor activity translates to the smoking cessation effect in humans is not known.

The market authorization was based on submitted data from quality (chemistry and manufacturing) studies, as well as data from non-clinical and clinical studies. The efficacy and safety of Champix in smoking-cessation was demonstrated in five double-blind, placebo-controlled clinical trials in which a total of 4190 chronic cigarette smokers (≥ 10  cigarettes per day) received varenicline. In all of the studies, statistical superiority in abstinence from smoking was demonstrated in patients treated with Champix compared to placebo. Results from the studies demonstrated that Champix was safe to use and was well tolerated when used as directed in the Product Monograph.

Champix (0.5 mg and 1.0 mg varenicline tartrate) is presented in tablet form. To optimize the success of the therapy, patients should be titrated up to the maximum recommended dose of 1.0 mg twice daily, using the 1-week titration schedule found in the Product Monograph. Patients who cannot tolerate the adverse effects of Champix may have the dose lowered. Dosing guidelines are available in the Product Monograph.

Champix is contraindicated for patients who are hypersensitive to varenicline or to any ingredient in the formulation or component of the container . Champix 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 Champix 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 Champix is favourable for smoking-cessation treatment in adults in conjunction with smoking-cessation counselling.

3 Scientific and Regulatory Basis for Decision

3.1 Quality Basis for Decision

3.1.1 Drug Substance (Medicinal Ingredient)

General Information

Varenicline (as varenicline tartrate), the medicinal ingredient of Champix, is a smoking-cessation aid. The proposed mechanism of action is the binding of varenicline to the α4β2 nicotinic acetylcholine receptor subtype, a receptor believed to be associated with nicotine addiction. Varenicline stimulates the receptor like a weaker version of nicotine, while simultaneously preventing nicotine from binding.

Manufacturing Process and Process Controls

Varenicline tartrate is manufactured via synthesis. 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

Characterization studies were performed to provide assurance that varenicline tartrate consistently exhibits the desired characteristic structure. The commercial synthesis has been demonstrated to consistently produce the desired polymorph. Results from process validation studies also indicate that the methods used during processing adequately control the levels of product and process-related impurities. The impurities that were reported and characterized were found to be within established limits.

Control of Drug Substance

Copies of the analytical methods and, where appropriate, validation reports are considered satisfactory for all analytical procedures used for release and stability testing of varenicline tartrate.

The specifications are considered acceptable for the drug substance. Data from the batch analyses were reviewed and were within the proposed acceptance criteria.

The drug substance packaging is considered to be acceptable.

Stability

Based upon the long-term and accelerated stability study data submitted, the proposed shelf-life, storage, and shipping conditions for the drug substance were supported and considered to be satisfactory.

3.1.2 Drug Product

Description and Composition

Champix immediate release, film-coated tablets are presented in two strengths containing 0.5 mg or 1.0 mg of varenicline (as varenicline tartrate) per tablet. Both strengths are capsular shaped but differ in colour and debossing.

  • The 0.5 mg tablet is a white to off-white, film-coated tablet debossed with "Pfizer" on one side and "CHX 0.5" on the other side.
  • The 1.0 mg tablet is a light blue film-coated tablet debossed with "Pfizer" on one side and "CHX 1.0" on the other side.

The non-medicinal ingredients present in Champix are microcrystalline cellulose, anhydrous dibasic calcium phosphate, croscarmellose sodium, colloidal silicon dioxide, and magnesium stearate. The film-coating contains hypromellose, titanium dioxide, polyethylene glycol and triacetin. The 1.0 mg tablet also contains FD&C Blue #2/ I ndigo Carmine Aluminum Lake as a colouring agent.

Champix tablets are packaged in high-density polyethylene bottles with induction seal lined closures and in blisters with aluminum foil backing.

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

Pharmaceutical Development

Changes to the manufacturing process and formulation made throughout the 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

Champix tablets are tested and released against the approved specification to ensure the product meets all predefined quality standards. The test specifications and analytical methods are considered acceptable.

Copies of the analytical methods and, where appropriate, validation reports are considered satisfactory for all analytical procedures used for release and stability testing of Champix.

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

Stability

Based on the stability data provided, the proposed shelf-life at 15-30°C for Champix is considered acceptable.

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

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 Adventitious Agents Safety Evaluation

N/A

3.1.5 Conclusion

The Chemistry and Manufacturing information submitted for Champix 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

A conventional non-clinical testing program in support of Champix followed ICH and FDA guidelines. Studies included pharmacodynamic (PD), pharmacokinetic (PK), and toxicologic studies in mice, rats, ferrets, rabbits, dogs, and monkeys. All pivotal studies were Good Laboratory Practice (GLP) compliant.

3.2.1 Pharmacodynamics

In vitro receptor binding studies demonstrated that of the four nicotinic receptor sub-types tested (α4β2, αβ4, α7, or α1β1γδ) varenicline specifically binds with high affinity to the human and rat α4β2 neuronal nicotinic acetylcholine receptor sub-type. In vitro functional studies showed that varenicline acts as a partial agonist, at the α4β2 subtype. It also acts as a partial agonist at the α3β4 nicotinic subtypes, but as a full agonist at the α7 nicotinic subtype (central pre-synaptic receptor), for which varenicline has very low binding affinity. The α4β2 subtype is believed to mediate the behaviorally reinforcing effects of nicotine. Nicotine is known to cause complex CNS behavioural, neuromuscular, endocrine, renal, metabolic and cardiovascular effects in humans, due to variety of peripheral and central receptor subtypes. It is not possible to know the extent to which varenicline is similar in effects, but varenicline did not have significant interactions with the peripheral α3β4 or α1β1γδ nicotinic subtypes.

Varenicline was shown to bind moderately to serotonergic 5HT3 receptor, known to be associated with nausea/vomiting and changes to bowel motility.

No significant interaction was shown with 56 other neurotransmitter receptors, ion channels, and neurotransmitter uptake sites.

Functional activity in vitro was demonstrated in oocytes and human embryonic kidney (HEK) cells expressing α4β2 neuronal nicotinic receptors in which maximal currents obtained were less than half those for nicotine, consistent with a partial agonist action. In both in vitro and in vivo models of mesolimbic dopamine function, activation by varenicline was about 50% of that induced by nicotine, consistent with a partial agonist mechanism of action.

As expected, when varenicline was substituted for nicotine in drug-discrimination studies, there was a dose-dependent generalization of the nicotine response, with the rats responding to the maximum dose of 1 mg/kg varenicline as if it were nicotine. In self-administration studies, varenicline reduced nicotine intake and the rate at which nicotine was self-administered by rats. Varenicline was also less reinforcing than nicotine, since rats worked harder for nicotine than for varenicline. Furthermore, varenicline (unlike nicotine, as determined from the literature) did not produce detectable effects in a rat model of withdrawal.

The behavioural and urine electrolyte effects seen in the safety pharmacology studies were mild and generally at high dose/exposures and therefore not suggestive of significant risk for humans. In vitro and in vivo non-clinical experiments demonstrated that varenicline performs as expected for a partial agonist of the α4β2 nicotinic receptor subtype, and the data are consistent with utility as a smoking-cessation treatment. 

3.2.2 Pharmacokinetics

Overall, varenicline is characterized by high absorption, negligible first-pass effect, moderate distribution across many tissues, moderate half-life and extensive renal excretion.

Absorption

Drug exposure levels determined by the PK parameters Cmax and AUC in single and multiple dose studies were dose-dependent in all species (mice, rats, ferrets, rabbits, dogs, and monkeys) with no significant gender differences.

Distribution

Plasma protein binding was low in all species tested with percent-bound values of 18%, 45%, 19%, 41%, and 20% in mice, rats, dogs, monkeys, and humans, respectively. In rats, oral administration of radiolabelled varenicline revealed drug-related material distributed throughout all tissues and organs in the body, except for lens and vitreous humor of the eye, with maximal radioactivity concentrations achieved at the first sampling point (1 hour post-dosing) in the gastrointestinal tract, and in other eye tissues. Varenicline demonstrated an increased but reversible affinity for melanin-containing tissues with drug-related material present in the eye and skin at 168 hours post-dose. Radiolabelled concentrations in pigmented skin were much higher (up to 18 fold) than in non-pigmented skin and showed accumulation from days 1 to 3 of dosing. In the reproductive toxicology studies, varenicline was detected in fetal rat and rabbit serum and was also detected in the serum of nursing offspring of varenicline-dosed rats.

Metabolism

A total of 13 metabolites of varenicline were identified in the circulation and urine of all laboratory animals and healthy human subjects tested, and all were products of oxidation and conjugation pathways. In mice, rats, monkeys, and humans the drug-related material in circulation and excreta was primarily unchanged drug (75-93%), indicating that metabolism is not a primary route of varenicline clearance in these species. No metabolite was present in excreta at more than 4.6% of the dose in any species. All metabolites observed in humans were observed in one or more species tested. In rabbits, two metabolites were present in higher concentrations in circulation than varenicline.

Excretion

Varenicline excretion in mice, rats, monkeys, and humans was mainly via the urine. In vitro data suggest that the renal excretion of varenicline occurs by both passive filtration and an active transport process (renal transporter OCT-2). The percentages of total dose recovered in the urine were 83%, 68%, 75%, and 87%, respectively; while the amounts in feces were 11%, 22%, 6.5%, and 0.9%, respectively.

Drug Interactions

Drugs cleared by, or which affect, cytochrome P450 enzymes

In vitro studies demonstrated that varenicline does not inhibit cytochrome P450 enzymes. The P450 enzymes tested for inhibition were: 1A2, 2A6, 2B6, 2C8, 2C9, 2C19, 2D6, 2E1, and 3A4/5. Also, in human hepatocytes in vitro, varenicline did not induce the activity of cytochrome P450 enzymes 1A2 and 3A4. Therefore, varenicline is unlikely to alter the pharmacokinetics of compounds that are primarily metabolised by cytochrome P450 enzymes. Furthermore, since metabolism of varenicline represents less than 10% of its clearance, drugs known to affect the cytochrome P450 system are unlikely to alter the pharmacokinetics of varenicline and therefore a dose adjustment of Champix should not be required for these types of drugs.

Drugs cleared by, or which affect, renal secretion

In vitro studies demonstrated that varenicline does not inhibit human renal transport proteins at therapeutic concentrations. Therefore, drugs that are cleared by renal secretion (e.g., metformin) are unlikely to be affected by varenicline.

In vitro studies demonstrated the active renal secretion of varenicline is mediated by the human organic cation transporter, OCT2. Although varenicline is a substrate and weak inhibitor of the human organic cation transporter (hOCT-2), and cimetidine and probenacid (as hOCT-2 inhibitors) reduce the clearance of varenicline, the drug interaction effects between these drugs were modest and occurred at much higher drug concentrations than achieved in humans. In cases of severe renal impairment, it is possible that the increase in systemic exposure to varenicline with an inhibitor of OCT2 such as cimetidine, might be clinically meaningful. This is reflected in the Product Monograph.

3.2.3 Toxicology

Single-Dose Toxicity

Acute oral studies were conducted in rat (30, 100, 200, 300 mg/kg), and monkey (3 mg/kg) and intravenous studies in monkeys (0.08 - 0.3 mg/kg).

Rats that were administered oral (PO ) doses of varenicline at ≥ 200 mg/kg (approximately 235-300 fold above the expected human exposure) were uncoordinated and developed convulsions and tremors with decreased body weight and loose stools. There was one death at 300 mg/kg. Effects observed were generally rapid in onset (with 2.5 hours post- dosing) and transient (reversed by the end of the 14-day observation period. The single-dose No Observed Adverse Effect Level (NOAEL) was ≤ 100 mg/kg corresponding to an exposure approximately 100-120 fold above the expected human exposure.

In monkeys at a dose of 3 mg/kg (approximately 2-4 fold above the expected human exposure) the findings were also in the GI (emesis), and central nervous system (tremours), along with decreased activity, and decreased food consumption. This dose was also associated with decreased heart rate and QT interval, and increased PRQ interval and P wave width on the electrocardiogram (ECG). All of the findings were reversed by the next day. The NOAEL was 0.2 mg/kg corresponding to an exposure of approximately 1.3 fold above the expected human exposure. ECG alterations were not seen in any of the subsequent multiple repeat-dose monkey studies or the cardiac safety pharmacology study, therefore it would be reasonable to suggest based on this data that there is not significant cardiac risk in humans.

Multiple-Dose Toxicity

Eleven oral, repeat-dose studies were completed in rats and monkeys. Rat studies included 6-week, 3- and 6-month studies. Monkey studies included 6-week, 3- and 9-month studies.

The multiple-dose NOAEL in rats was 10 mg/kg/day in the 3- and 6-month studies, which corresponds to approximately 40-65 fold above the maximum recommended human exposure.

Decreased in food consumption were seen in rats at doses of 10 mg/kg, likely due to decreases in gastric motility. Rats dosed with varenicline at 30 mg/kg (approximately 75-140 fold above the expected human exposure) had clinical signs related to the nervous system and gastrointestinal tract: decreased food consumption, decreased body weight, loose stool, dehydration, microscopic and macroscopic evidence of intestinal dilatation (ileum, jejunum, cecum, colon). These changes likely reflect the pharmacologic effects of varenicline on gastric emptying and intestinal motility in the rats. Decreases in body temperature were also seen, with body temperature perturbation a well known effect of nicotine.

Slight increases in alanine aminotransferase (ALT), alkaline phosphatase (ALP), and total bilirubin, suggestive of hepatobilary alteration without microscopic hepatic findings, were reported in rats dosed with varenicline at ≥ 10 mg/kg. The significance of the reported increases in absolute liver weights in females at ≥ 10 mg/kg is uncertain as similar increases were not observed in previous studies. Hepatic changes occurred at the highest dose of the 10-day and 6-week studies (i.e., 100 mg/kg/day and 30 mg/kg/day, respectively), but were not seen in the 6-month study. These changes included: small increases in ALT, ALP, aspartate amino transferase (AST)), and/or total bilirubin, and in some cases, microscopic evidence of hepatocellular necrosis. The changes are noted in the Product Monograph.

The multiple-dose NOAEL in monkeys was 0.2 mg/kg/day in the 3- and 6-months studies, which corresponds to approximately 3 fold above the maximum recommended human exposure.

Monkeys dosed with varenicline at 1.2 mg/kg (approximately 10-12 fold above the maximum recommended human exposure) were all sacrificed prematurely due to the severity of decreased food consumption, decreased body weight, and severe/prolonged dehydration. One female monkey at this dose was found dead of megacolon, secondary to colonic torsion and ischemic necrosis. Megacolon is an uncommon spontaneous finding in monkeys. This finding was likely secondary to the gastrointestinal dysfunction (loose stools) that was evident prior to and during treatment, although a drug treatment effect can not be excluded.

Slight increases in fibrinogen in male and female monkeys were observed at 1.2 mg/kg. Increased fibrin was also previously observed in monkeys at 1.2 mg/kg (0.6 BID) with concurrent increases in neutrophils and monocytes. This is suggestive of inflammation, but is of uncertain significance. Decreases in body temperature were also seen at this dose, and at the lower dose of 0.6 mg/kg/day.

Genotoxicity

Varenicline was non-genotoxic in a battery of in vitro and in vivo genetic toxicity tests.

Carcinogenicity

Two pivotal 2-year studies investigated the carcinogenic potential of varenicline, one in mice and one in rats. The studies were GLP compliant.

The mice studies showed no evidence of carcinogenicity associated with the oral administration of varenicline tartrate. Mice were administered doses up to 20 mg/kg. Exposure at 20 mg/kg was approximately 120 fold above the expected maximum human exposure.

The incidence of neoplasms associated with varenicline was increased in the rat studies, however the results were not statistically significant. Rats were administered doses of 1, 5, and 15 mg/kg. One male at 5 mg/kg had a benign hibernoma (tumor of brown fat in the mediastinum) and 2 males at 15 mg/kg had malignant hibernoma which was the cause of death in both animals. Exposure at 1, 5, and 15 mg/kg represented an exposure of approximately 15, 30, and 50 fold, respectively, above the expected maximum human exposure.

Hibernoma is a rare neoplasm and a review of a few cases in control animals from published reports was discussed. Discussions included the physiologically relevant differences between rodent and human brown fat. It was proposed that because of the physiological differences between human and rodent brown fat, the human risk is theoretical and probably non-existent. Varenicline is not genotoxic. Also, there was no abnormal histopathological evidence of brown fat proliferation at doses of 30 mg/kg in the 3- and 6-month rat studies. Hibernomas were not observed in female rats or male and female mice. The weight of evidence suggests that the risk is extremely low but the relevance to humans is uncertain. The presence of the benign and malignant hibernomas is acknowledged as treatment-related in the Product Monograph.

Reproductive and Developmental Toxicity

Five studies designed to examine the reproductive toxicity of varenicline were conducted in rats (Segments I, II, and III) and rabbits (Segment II).

Rats in the Segment I studies were administered doses of 0.3, 3, and 15 mg/kg. There were no effects on fertility and the male and female reproductive parameters, and no effects on early embryonic development. Therefore, the fertility and early embryonic NOAEL was 15 mg/kg corresponding to an exposure approximately 37-43 fold above the expected human exposure. The NOAEL for the F0 males and females was 3 mg/kg, approximately 16 fold above the expected human exposure.

Segment II studies were conducted in rats and rabbits. Rabbits that were administered doses of 30 mg/kg demonstrated significant reductions in fetal and placental weights. The rabbit F0 maternal toxicity and F1 fetotoxicity NOAEL were both 10 mg/kg, corresponding to an exposure approximately 15-28 fold above the expected human exposure. In rats and rabbits, the maternal and fetal serum varenicline concentrations increased as the doses increased. Fetal concentrations were higher than the maternal concentrations confirming that the fetus was exposed in utero following administration of varenicline to the dam. The Product Monograph acknowledges this fetotoxicity and recommends that pregnant women not use varenicline.

Segment III studies were completed in rats dosed at 0, 0.3, 3, and 15 mg/kg and there was no effect on the reproductive parameters. The NOAEL for F0 maternal reproductive toxicity is 15 mg/kg/day, corresponding to an exposure approximately 39 fold above the expected human exposure. Body weights for the F1 male and female offspring in the 15 mg/kg/day group were reduced and some alterations were noted in functional development. A treatment-related increase in the mean amplitude of the auditory startle response (ASR) was noted in the 15 mg/kg/day group males and considered to be treatment-related. The rat studies also noted a reduction in fertility of the F1 offspring in the 15 mg/kg/day group. The Product Monograph acknowledges these post natal developmental findings and recommends that pregnant and nursing women not use varenicline.

Serum-drug concentrations of the F0 females and F1 pups indicate that pups are exposed to varenicline through milk after oral administration of varenicline to dams. The Product Monograph indicates nursing women should discontinue nursing or discontinue taking varenicline.

Local Toxicity

Varenicline was classified as a mild ocular irritant and a mild skin irritant in rabbits. However, no deaths or evidence of systemic toxicity following a single 24-hour semi-occluded dermal application to intact skin at a dose level of 2000 mg/kg in rats were observed. Varenicline was non-sensitizing following dermal challenge in guinea pigs. Oral administration of varenicline at 100 mg/kg/day for three days did not cause phototoxicity in rats.

3.2.4 Summary and Conclusion

Overall, the pharmacology and toxicology studies support the use of Champix for the proposed indication. Treatment-related clinical signs and alterations on food consumption and body weight in all animal species which could be anticipated based on the pharmacology of varenicline generally occurred at dose levels and corresponding systemic exposures above the anticipated exposure in humans. Treatment-related pathologic findings in the gastrointestinal tract of rats occurred at doses resulting in exposure at significant levels above the anticipated human exposure. The carcinogenicity studies in mice did not indicate carcinogenic potential. However, the occurrence of hibernomas in rats is discussed in the Product Monograph as the relevance to humans is uncertain. Adverse effects were observed in the animal reproduction studies. The Product Monograph indicates that Champix is not recommended in pregnant women and recommends discontinuation of the drug or nursing in nursing women.

3.3 Clinical basis for decision

3.3.1 Pharmacodynamics

Varenicline, the medicinal ingredient of Champix, binds to α4β2 nicotinic receptors, the neuronal nicotinic receptors believed to be most associated with nicotine addiction. It is thought that the nicotinic α4β2 receptor mediates dopamine release in the nucleus accumbens and therefore, is involved in the motivational effects of smoking.

Both nicotine and varenicline reversibly bind to the same receptor binding site. Although varenicline is not nicotine, the drug acts like nicotine at the α4β2 nicotinic receptor, albeit a less effective version. Varenicline is a "partial agonist", i.e., binds more strongly than nicotine to the α4β2 nicotinic receptor, but activates it less effectively, thus acting both to block nicotine from binding and to partially stimulate the receptor. Affinities for three other nicotinic sub-types were 500-20,000 times less.

Nicotine is known to cause complex central nervous system (CNS) behavioural, neuromuscular, endocrine, renal, metabolic and cardiovascular effects in humans, due to variety of peripheral and central receptor subtypes; it is not possible to know the extent to which varenicline is similar in effects.

The binding of varenicline to α4β2 nicotinic receptors was demonstrated in the non-clinical studies, and in human cells in a dish. While these data are consistent with the partial-agonist receptor activity of varenicline mediating its effect in preclinical models, there are at present no methods available to provide evidence that this mechanism is causing the known pharmacodynamic effects in humans (i.e., higher abstinence rates than for placebo, and reduction in urge to smoke and withdrawal symptoms compared to placebo). It is interesting to note that these PD effects in humans have also been achieved to some extent via medications which do not act as partial agonists at the α4β2 receptors.

Varenicline has also been shown to bind to 5HT3 serotonergic receptors, and stimulation of these is associated with the gastrointestinal effects (e.g., nausea, vomiting, slow bowel motility), consistent with the drug pharmacological profile of the drug.

3.3.2 Pharmacokinetics

Absorption

Orally administered varenicline was virtually completely absorbed and was highly available systemically. Varenicline exhibited linear pharmacokinetics (PK) following single or multiple dosing within the recommended dose range. The maximum plasma concentration following a single dose occurred at approximately three hours, and steady state was reached within approximately four days of repeat dosing.

Distribution

Plasma protein binding of varenicline was low. In healthy adults, the mean unbound fraction ranged from 87.9% to 93.5%. Similar protein binding results were obtained in subjects with varying degrees of renal impairment and in elderly subjects. Weight was shown to influence the volume of distribution, therefore the plasma concentration fluctuations will be affected by body weight.

Metabolism

Varenicline undergoes minimal metabolism. A clinical mass balance study showed that varenicline did not go through oxidative metabolism in the liver, but instead was primarily excreted as unchanged drug in the urine. Unchanged varenicline was the predominant drug-related entity in the circulation (90.8%) and urine (91.6%); less than 10% of the varenicline dose was excreted as metabolites. The minor routes of metabolism were N-carbomyl glucuronidation, N-formylation and conjugation with a hexose sugar.

Elimination

Varenicline was almost exclusively excreted in the urine, primarily through glomerular filtration, with an additional component of active secretion via the human organic cation transport system OCT2. The elimination half-life of varenicline was approximately 24 hours.

Special Populations

Results from a population PK analysis showed that renal function and body weight (affecting systematic clearance and volume of distribution, respectively) were the important factors leading to inter-individual variability in the PK of varenicline. Also, no clinically meaningful PK differences due to age, race, or gender were detected. Weight was shown to influence the volume of distribution, therefore the plasma concentration fluctuations will be affected by body weight. Clearance of varenicline was linearly related to glomerular filtration rate (GFR), and exposure was increased in patients with renal impairment. Consequently, a dose reduction to half the recommended dose is indicated for patients with severe (estimated GFR<30 mL/min) renal insufficiency.

Drug Interactions

Varenicline is not expected to alter the PK of co-administered compounds that are metabolized by the cytochrome P450 isoforms, CYP3A4 or CYP1A2. As well, drugs that inhibit/induce the P450 enzymes would not be expected to alter the systemic exposure to varenicline.

No clinically relevant drug interactions were observed when varenicline was
co-administered with the narrow therapeutic index drugs digoxin or warfarin.

Patients with severe renal impairment should avoid the concomitant use of varenicline with OCT2 inhibitors.

Abuse Liability

For smokers, the results were consistent with little motivation to abuse. For non-smokers, sensation of receiving an active drug was apparent; the sensation was perceived as different compared to amphetamine.

From the abuse liability data, there was little evidence of reinforcing effects, but the data were limited in terms of what was examined.

3.3.3 Clinical Efficacy

Five adequately-designed, double-blind, placebo-controlled studies demonstrated the efficacy of Champix (varenicline tartrate) as a smoking cessation aid, with doses of 0.5 mg BID (twice daily) and 1.0 mg BID identified as being efficacious. A total of 3282 chronic cigarette smokers received varenicline in these five trials. Doses >1.0 mg BID generally caused nausea and vomiting, to the extent that higher doses could not be adequately studied.

For four of the studies, the drug treatment period was 12 weeks, with patients able to remain in the study and be counted as a responder regardless of discontinuing drug treatment prior to the 12 weeks. In two of the 12-week studies the highest recommended dose of Champix, 1.0 mg BID, was tested against the comparator drug, bupropion (NOTE: This was in the absence of concomitant nicotine replacement therapy, as was also true for all other arms in these studies). After the 12 weeks of drug treatment, there was a subsequent 40 weeks of double-blind assessment of the patients, post drug-treatment. The primary endpoint was the Abstinence Responder Rate (the percentage of patients reporting 4 weeks continuous abstinence, from Week 9 to Week 12). Abstinence was defined as no smoking at all, not even a puff, and was measured with self-reports and exhaled carbon monoxide (CO) measurements (≤ 10 ppm was considered as supportive of reported abstinence). The key secondary endpoint was the continuous abstinence rate (CAR), defined as the percentage of patients who abstained from smoking from Week 9 to Week 52.

In all four 12-week studies, Champix was significantly superior to placebo in the primary outcome, and in the two studies with the comparator arm, Champix was significantly superior to the comparator, bupropion. The abstinence responder rates after 12 weeks of treatment for Champix 1.0 mg BID, Champix 0.5 mg BID, placebo, and bupropion were 44-50%, 45%, 12-18%, and approx. 30%, respectively.

Champix also showed superiority to placebo for the secondary endpoints.

The CARs at 52 weeks for Champix 1.0 mg BID and for placebo were approximately 23% and approximately 9%, respectively. Champix 0.5 mg BID was superior to placebo, with CARs of approximately 19% and approximately 4%, respectively. When compared to bupropion, Champix 1 mg BID was superior to bupropion in only one of the two studies.

Other secondary endpoints included the measurements of urge to smoke and withdrawal symptoms. Based on the responses to the Brief Questionnaire of Smoking Urges and the Minnesota Nicotine Withdrawal Scale, craving and urge to smoke were significantly reduced in patients randomized to Champix compared to those randomized to placebo, as were negative affect withdrawal symptoms (depressed mood; irritability, frustration, or anger; anxiety; difficulty concentrating).

Overall, all of the Champix doses tested in the pivotal trials were superior to placebo (CQR from week 9 through 12: 40 -51% responder rate and CAR from week 9 through 52: 19-23% responder rate). The 1.0 mg BID dose demonstrated a slight tendency towards greater efficacy, when compared to the 0.5 mg BID dose.

Limited knowledge was gained regarding the 0.5 mg BID dose as the one study which included the 0.5 mg BID dose was not designed to statistically compare the 1.0 mg BID dose to the 0.5 mg BID dose for abstinence rates. Instead, the study compared each dose to placebo. The abstinence responder rates for the two doses appeared to be similar, however statistical comparisons between the two doses, as well as a comparison of the 0.5 mg BID dose to bupropion would have been worth exploring.

The fifth study assessed the benefit of an additional 12 weeks of drug treatment on maintenance of abstinence. The first part of the study was a 12-week, open-label treatment period with Champix, 1.0 mg BID, followed by a double-blind treatment period, in which all abstinent subjects were randomized to 12 weeks on Champix 1.0 mg BID or 12 weeks on placebo. Double-blind monthly assessments of abstinence were carried out post drug-treatment for up to 52 weeks. The primary study endpoint was the CO-confirmed CAR from Week 13 to Week 24. The key secondary endpoint was the CAR for Week 13 to Week 52 .

In the fifth study, superiority to placebo was shown for both the primary and secondary endpoints where patients were treated for 24 weeks. The CAR from Week 13 through Week 24 was higher for patients continuing treatment with Champix (approx. 70%) compared to patients switched to placebo (approx. 50%). At Week 52, the CARs for Champix and placebo were approximately 44% and 37%, respectively. However, it is worth noting that concerns did arise with the CAR data from Week 13 through Week 52: (a) the appropriateness of a comparison between: an arm with drug removed versus an arm with drug continued; (b) the evidence of abrupt/accelerated relapse rates in both

arms, at the point where drug treatment is stopped, i.e., Week 12 for the placebo arm, and Week 24 for the drug arm; and (c) the potential for the complete loss of difference between drug versus placebo, with extra time.

3.3.4 Clinical Safety

The safety profile of Champix (varenicline tartrate) was investigated in a comprehensive program of clinical studies.Total clinical exposure (open-label and double-blind) at the therapeutic doses included approximately 2300 patients treated for at least 12 weeks, approximately 700 for 6 months, and approximately 100 for one year. The maximum dose administered was 1.0 mg BID due primarily to dose-limiting nausea and vomiting events. The safety program was considered acceptable.

The most commonly observed adverse events associated with Champix (>5% and twice the rate seen in placebo-treated patients) were nausea, abnormal dreams, constipation, flatulence, and vomiting.

The incidence of nausea, abnormal dreams, and constipation increased in a dose-dependent manner and were greater than placebo. The incidence of nausea was 30% in the 1.0 mg BID group, compared to 16% in the 0.5 mg BID group, and approximately 10% in the placebo group. Nausea generally started during the first week of treatment and the median duration was 10 days. Reports of nausea were similar to that reported with placebo by approximately Week 4, and in some cases, continued for months.

Discontinuation rates for adverse events (AEs) in the 3-month studies were approx 12% for patients on Champix 1.0 mg BID compared to 10% for placebo. The most common AEs that resulted in treatment discontinuation were: nausea (3%), insomnia (1.2%), headache (0.6%), and abnormal dreams (0.3%).

Safety data related to the following organ systems or syndromes were specifically evaluated: cardiac and vascular systems, nervous system and psychiatric, gastrointestinal, eye, renal function, rash and pruritus. None of these were identified as a serious safety concern for varenicline.

Of the various serious adverse events (SAEs) reported, no notable signals or trends in pattern or occurrence were apparent. As per the FDA review of vanenicline, the calculated values for SAE incidence per Patient Exposure Year (i.e., corrected for time on drug) are as follows: varenicline 0.086, bupropion 0.131, and placebo 0.075. The top categories were cardiac disorders, neoplasms, infections, central nervous system (CNS) disorders, and gastrointestinal (GI) disorders. In most cases of cardiac SAEs, there was significant cardiac history.

Two deaths were reported within the 30-day post-treatment period. Both patients were treated with Champix, however neither death was considered as treatment-related. The first death was a suicide (a man with suicidal ideation that was not previously disclosed), and the other death was from lung cancer.

A one-year, double-blind study with Champix 1.0 mg BID (n=251) and placebo (n=126) provided a similar safety profile when compared to the studies of 12-weeks exposure, except for the following events which were seen to be increased relative to placebo, as compared to the drug versus placebo ratio seen in the 12-week data: nausea (40% vs. 8% placebo), the pooled terms of: abdominal discomfort (16% vs. 2% placebo), and increased blood pressure (11% vs. 6% placebo).

The full consequences of using Champix in patients with concomitant illness have not been established, andcaution should be exercised. The use of Champix has not been studied in psychiatric patients, patients with epilepsy, patients undergoing chemotherapy treatment, or in patients with GI disturbances such as irritable bowel. Precautionary statements for the populations that were not studied are provided in the Product Monograph. Specific to schizophrenia: cholinergic agents are known to induce psychosis; two cases of treatment-emergent psychosis were reported. No remarkable signal, but given that schizophrenia is known to be associated with smoking, a smoking-cessation agent that blocks nicotine from getting to one or more receptor subtypes may have consequences for such patients.

Champix should not be used with nicotine replacement therapy (NRT). Due to the partial agonist nicotinic activity of varenicline, it is not anticipated that co-administration with NRT would confer additional benefits compared with Champix alone, and may result in increased side effects. In a 12-day safety study, the incidence of nausea, headache, vomiting, dizziness, dyspepsia and fatigue were greater for the combination of varenicline and NRT than for NRT alone.

3.4 Benefit/Risk Assessment and Recommendation

3.4.1 Benefit/Risk Assessment

Champix has been shown to be an efficacious smoking-cessation aid. Patients treated with Champix for 12 weeks demonstrated statistical superiority in abstinence from smoking compared to placebo, with similar responder rates for 0.5 mg BID and 1.0 mg BID. When all outcome measures are considered, there is a slight tendency towards greater efficacy for the 1.0 mg BID dose, when compared to the 0.5 mg BID dose. Given the importance of minimizing lapses in abstinence for maximal public health impact, and the fact that patients can readily lower their dose if they have problems with adverse events, the maximum recommended dose of 1.0 mg BID is the recommended dose, with instructions for the dose to be lowered for patients that have intolerable AEs.

In the fixed-dose placebo-controlled studies, Champix-treated patients most commonly reported treatment-emergent GI (e.g., nausea, constipation, dyspepsia, vomiting) and CNS (e.g., insomnia, abnormal dreams) AEs. Nausea was the most frequently reported adverse event. Most patients became tolerant to nausea and did not withdraw from the drug. For patients with intolerable nausea, a dose reduction should be considered.

Based on the submitted data, the potential risks of Champix are acceptable, under the specified conditions of use. At this time, it is considered that safety issues involving Champix treatment can be managed through labelling. The sponsor will be providing ongoing safety updates and reports with respect to safety issues raised in the review. Following review of those data, appropriate Product Monograph labelling and/or action will be taken, as necessary.

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 Champix is favourable for smoking-cessation treatment in adults in conjunction with smoking-cessation counselling. 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: Champix

Submission MilestoneDate
Submission filed2006-02-03
Screening 1
Screening Acceptance Letter issued2006-03-31
Review 1
Biopharmaceutics Evaluation complete2006-10-30
Quality Evaluation complete2007-01-19
Clinical Evaluation complete2007-01-23
Labelling Review complete2007-01-18
NOC issued by Director General2007-01-24