Summary Basis of Decision for Xalkori ™

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
XalkoriTM

Crizotinib, 200 mg and 250 mg, Capsule, Oral

Pfizer Canada Inc.

Submission control no: 145155

Date issued: 2012-07-26

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:

XalkoriTM

Manufacturer/sponsor:

Pfizer Canada Inc.

Medicinal ingredient:

Crizotinib

International non-proprietary Name:

Crizotinib

Strength:

200 mg and 250 mg

Dosage form:

Capsule

Route of administration:

Oral

Drug identification number(DIN):

  • 02384256 - 200 mg
  • 02384264 - 250 mg

Therapeutic Classification:

Anaplastic Lymphoma Kinase (ALK) Tyrosine Kinase Inhibitor

Non-medicinal ingredients:

Colloidal silicon dioxide, microcrystalline cellulose, anyhydrous dibasic calcium phosphate, sodium starch glycolate, and magnesium stearate.
Pink opaque hard gelatin capsule shell: gelatin, titanium dioxide, and red iron oxide.
White opaque hard capsule shell: gelatin and titanium dioxide.
Printing ink: shellac, propylene glycol, strong ammonia solution, potassium hydroxide, and black iron oxide.

Submission type and control no:

New Drug Submission, Control Number: 145155

Date of Submission:

2011-06-08

Date of authorization:

2012-04-25
2 Notice of decision

On April 25, 2012, Health Canada issued a Notice of Compliance under the Notice of Compliance with Conditions (NOC/c) Guidance to Pfizer Canada Inc., for the drug product Xalkori. The product was authorized under the NOC/c Guidance on the basis of the promising nature of the clinical evidence, and the need for confirmatory studies to verify the clinical benefit. Patients should be advised of the fact that the market authorization was issued with conditions.

Xalkori contains the medicinal ingredient crizotinib which is an anaplastic lymphoma kinase (ALK) tyrosine kinase inhibitor.

Xalkori is indicated as monotherapy for use in patients with ALK-positive advanced (not amenable to curative therapy) or metastatic non-small cell lung cancer (NSCLC). Prior to receiving therapy with Xalkori, patients must be tested and confirmed ALK-positive advanced or metastatic NSCLC using a validated ALK assay. This assessment should be performed by laboratories with demonstrated proficiency in the specific technology being utilized. The clinical benefit of Xalkori in patients with ALK-negative NSCLC has not been established; therefore, Xalkori is not recommended for these patients.

Xalkori inhibits the ALK receptor tyrosine kinase (RTK) and its oncogenic variants (such as ALK fusion events and selected ALK mutations), as well as the Hepatocyte Growth Factor (HGFR/c-Met) RTK. Through this inhibitory action, Xalkori blocks important growth and survival pathways, which may shrink or slow the growth of tumours in patients with ALK-positive NSCLC.

The market authorization was based on quality, non-clinical, and clinical information submitted. The efficacy of Xalkori as monotherapy to treat patients with ALK-positive advanced NSCLC with or without brain metastases has been assessed in two ongoing, open-label, single-arm, multicentre studies (Study A8081001 - 116 patients and Study A8081005 - 136 patients). All patients received prior systemic therapy, with the exception of 16 patients (13%) in Study A8081001 who had no prior systemic treatment for locally advanced or metastatic disease. Each patient received 250 mg of Xalkori taken orally twice per day. The primary efficacy endpoint for both studies was Objective Response Rate (ORR) according to Response Evaluation Criteria in Solid Tumors (RECIST Version 1.0). The ORR was 61% in Study A8081001 and 51% in Study A8081005. Notably, more than 90% of patients in these studies showed some degree oftarget lesion shrinkage. To date there are no data demonstrating overall survival or progression-free survival benefit in this patient population. Although this preliminary data is based on single-arm studies with no control arm, the ORR demonstrates promising evidence of a potential benefit of Xalkori in this patient population. The lack of effective systemic therapies for patients with this rare disease (ALK-positive NSCLC) constitutes an unmet medical need which Xalkori appears to fulfil. Market authorization was based on the ORR data from these two ongoing single-arm studies. Confirmation of clinical benefit will be assessed in two ongoing Phase III randomized control studies in previously treated and treatment-naïve patients.

Xalkori (200 mg and 250 mg, crizotinib) is presented in capsule form. The recommended dose schedule for Xalkori is 250 mg taken orally twice daily with or without food. Treatment should be continued as long as the patient is deriving clinical benefit from therapy. Capsules should be swallowed whole. The concurrent use of strong cytochrome P450 (CYP)3A inhibitors such as ketoconazole or strong CYP3A inducers such as rifampin should be avoided.

Xalkori is contraindicated for patients with congenital long QT syndrome or with persistent Fridericia-corrected electrocardiogram interval (QTcF) of 500 msec or greater. The use of Xalkori is also contraindicated in patients with a known hypersensitivity to the active substance, crizotinib, or to any ingredient in the formulation or component of the container. Xalkori 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. Dosing guidelines are available in the Product Monograph. Detailed conditions for the use of Xalkori are described in the Product Monograph.

Three major safety issues were identified in this submission:

  1. QT interval prolongation and bradychardia;
  2. hepatotoxicity, including fatal outcomes; and
  3. pneumonitis, including fatal outcomes.

Also, Xalkori has not been studied in patients with hepatic or severe renal impairment. These issues have been addressed through appropriate labelling (a Serious Warnings and Precautions box) 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 Xalkori is favourable for use as monotherapy in patients with ALK-positive advanced (not amenable to curative therapy) or metastatic NSCLC. As part of the market authorization, Health Canada has required a Risk Management Plan to monitor whether the benefits continue to outweigh any risk.

3 Scientific and Regulatory Basis for Decision

The Advance Consideration Notice of Compliance with Conditions (NOC/c) New Drug Submission (NDS) for Xalkori was originally submitted on June 8, 2011 in order to obtain marketing authorization for the treatment of patients with anaplastic lymphoma kinase (ALK)-positive advanced non-small cell lung cancer (NSCLC).

Subsequent review led to the decision to issue the sponsor market authorization under the NOC/c Guidance, in recognition of the promising nature of the clinical evidence, and the need for confirmatory studies to verify the clinical benefit. After analyzing the submitted data, Health Canada recommended a more specific indication for Xalkori than that originally proposed by the sponsor. The approved indication for Xalkori is for monotherapy for use in patients with ALK-positive advanced (not amenable to curative therapy) or metastatic NSCLC. Using a validated ALK assay, assessment for ALK-positive advanced or metastatic NSCLC should be performed by laboratories with demonstrated proficiency in the specific technology being utilized. Improper assay performance can lead to unreliable test results. ALK gene rearrangements were identified in Study A8081001 by clinical trial assays performed by local laboratories and in Study A8081005 by a Health Canada-approved Vysis ALK break-apart fluorescence in situ hybridization (FISH) assay.

The clinical benefit of Xalkori in patients with anaplastic lymphoma kinase-(ALK) negative NSCLC has not been established; therefore Xalkori is not recommended for these patients.

Marketing authorization with conditions was based on a primary efficacy endpoint of objective response rate (ORR) as well as duration of response (DR) in clinical studies A8081001 and A8081005, based on investigator assessment using RECIST. There are no data available demonstrating improvement in survival with Xalkori.

An NOC/c for Xalkori was issued on April 25, 2012. The authorization is conditional upon further confirmation of clinical benefit.

3.1 Quality Basis for Decision

3.1.1 Drug Substance (Medicinal Ingredient)

General Information

Crizotinib, the medicinal ingredient of Xalkori is an ALK receptor tyrosine kinase (RTK) inhibitor.

The majority of lung cancers are NSCLC. In patients with ALK-positive NSCLC, a gene thought to normally be dormant, ALK, is fused with another gene. Expression of the resulting fusion gene leads to tumour growth. Only 3 to 5% of NSCLC patients are thought to have this ALK fusion event.

Xalkori is a selective small-molecule inhibitor of the ALK RTK and its oncogenic variants, as well as an inhibitor of the Hepatocyte Growth Factor (HGFR/c-Met) RTK. Through this inhibitory action, Xalkori blocks important growth and survival pathways, which may shrink or slow the growth of tumours.

Manufacturing Process and Process Controls

Crizotinib is manufactured via a multi-step 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 International Conference on Harmonisation (ICH) requirements.
  • The processing steps have been evaluated and the appropriate ranges for process parameters have been established.

The materials used in the manufacture of the drug substance are considered to be suitable and/or meet standards appropriate for their intended use.

Characterization

The structure of crizotinib has been adequately elucidated and the representative spectra have been provided. Physical and chemical properties have been described and are found to be satisfactory.

The sponsor has provided a summary of all drug-related impurities. All impurities were found to be within ICH established limits or were qualified from toxicological studies and therefore are considered acceptable.

Control of Drug Substance

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

The levels of product- and process-related impurities were adequately monitored throughout the manufacturing process. Results from process validation reports and in-process controls indicated that the impurities of the drug substance were adequately under control. The level of impurities reported for the drug substance was found to be within the established limits.

The drug substance packaging is considered acceptable.

Stability

Stability study results based on long-term and stress testing show that crizotinib is a stable compound when packaged as proposed over the proposed storage period.

3.1.2 Drug Product

Description and Composition

Xalkori (crizotinib) capsules are available in the following two dosage strengths:

  • 250 mg - containing 250 mg crizotinib supplied as a hard gelatine capsule, size 0, pink opaque/pink opaque, with "Pfizer" on the cap and "CRZ 250" on the body; and
  • 200 mg - containing 200 mg crizotinib supplied as a hard gelatine capsule, size 1, white opaque/pink opaque, with "Pfizer" on the cap and "CRZ 200" on the body.

The two strengths of the drug product are considered proportional according to the Health Canada guidance Bioequivalence of Proportional Formulations - Solid Oral Dosage Forms.

Xalkori contains the following non-medicinal ingredients: colloidal silicon dioxide; microcrystalline cellulose; anhydrous dibasic calcium phosphate; sodium starch glycolate; magnesium stearate; and hard gelatin capsule shells. The pink opaque capsule shell components contain gelatin, titanium dioxide, and red iron oxide. The white opaque capsule shell components contain gelatin and titanium dioxide. The printing ink contains shellac, propylene glycol, strong ammonia solution, potassium hydroxide, and black iron oxide.

Xalkori is supplied as bottles of 60 capsules, and as polyvinyl chloride (PVC)/aluminum foil blisters containing 60 capsules [6 cards of 10 (5 x 2) capsules].

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 crizotinib with the excipients is demonstrated by the stability data presented on the proposed commercial formulation.

Pharmaceutical Development

Pharmaceutical development data, including development of the container closure system, are considered acceptable. Data provided in this section include the composition of Xalkori, the rationale for choice of the formulation, the manufacturing process including packaging, information on batches used for in vitro studies for characterization, and discussion on the effect of formulation change on the safety and/or efficacy of Xalkori. Studies which justified the type and proposed concentration of excipients to be used in the drug product were also reviewed and are considered to be acceptable.

Manufacturing Process and Process Controls

The drug product is formulated, pre-blended, milled, blended, lubricated, dry granulated, encapsulated, and packaged using conventional pharmaceutical equipment and facilities.

All manufacturing equipment, in-process manufacturing steps and detailed operating parameters were adequately described in the submitted documentation and were found to be acceptable. The manufacturing process is considered to be adequately controlled within justified limits.

Control of Drug Product

Xalkori is tested to verify that its identity, appearance, assay, dissolution, weight, 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 the release limits, for individual and total degradation products, are within acceptable limits.

Validation results of the analytical method used for the determination of crizotinib and the drug-related impurities are considered acceptable.

Stability

Based on the real-time, long-term, and accelerated stability data submitted, the proposed 24-month shelf-life at 15-30°C for Xalkori is considered acceptable.

The compatibility of the drug product with the container closure system was demonstrated through compendial testing and 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 Xalkori are considered suitable for the activities and products manufactured.

All sites are compliant with Good Manufacturing Practices.

3.1.4 Adventitious Agents Safety Evaluation

Following investigations, Xalkori (crizotinib, 200 mg and 250 mg) capsules were found to be compliant with the Note for Guidance on Minimising the Risk of Transmitting Animal Spongiform Encephalopathy Agents via Human and Veterinary Medicinal Products [European Medicine Agency (EMA)/410/01, Revision 2]. The gelatin used to manufacture the hard gelatin capsules for these formulations is also produced in compliance with the above note.

Several packaging components may use small amounts of animal derived tallow, stearates or lubricants during manufacture. Processing conditions minimize the risk of bovine spongiform encephalopathy (BSE)/transmissible spongiform encephalopathy (TSE).

Certification letters attesting to these claims were provided by the sponsor.

3.1.5 Conclusion

The Chemistry and Manufacturing information submitted for Xalkori 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

The Canadian regulatory decision on the non-clinical pharmacology was based on a critical assessment of the Canadian application/submission. The foreign review completed by the United States Food and Drug Administration (FDA) (March 30, 2011) was used as an added reference in the assessment Xalkori.

3.2.1 Pharmacodynamics

Primary Pharmacodynamics

The properties of crizotinib have been studied extensively in a variety of in vitro and in vivo model systems to determine the potency for inhibition of ALK or c-Met/HGFR RTK activity, kinase selectivity, antitumour efficacy, pharmacokinetic (PK)/pharmacodynamic (PD) relationships, non-clinical toxicological profile, and mechanism of action.

Crizotinib was found to be a potent inhibitor of ALK and c-Met kinases. This translates into promising dose-dependent in vivo efficacy against tumours harbouring oncogenic ALK fusion events or dysregulated c-Met. The antitumor efficacy of crizotinib was dose-dependent and correlated with PD inhibition of phosphorylation of ALK fusion proteins in tumours in vivo. Specifically, 50% ALK inhibition is required to significantly inhibit tumour growth by more than 50%.

Secondary Pharmacodynamics

Crizotinib displayed functional antagonism of the 5-HT4e and 5-HT7 serotonin receptors as well as the alpha-1a adrenergic receptors. Further testing identified crizotinib as an inhibitor of dopamine and serotonin uptake. Chromatography data demonstrated the presence of crizotinib in the central nervous system, indicating the potential for an array of neurological disorders. The safety of crizotinib was supported, however, in acute-dose safety pharmacology neurological assessments in rats.

Safety Pharmacology

Several safety pharmacology studies assessed respiratory and cardiovascular safety following administration of crizotinib. The results support the safety of crizotinib in acute pulmonary assessments but suggest the strong potential for adverse cardiovascular effects post-crizotinib treatment. This could correlate with clinical observations of QT prolongation, bradycardia, and cardiac arrest. Despite these safety issues, with appropriate labelling and monitoring, the benefit risk profile continues to support the use of crizotinib for the treatment of advanced ALK-positive NSCLC.

3.2.2 Pharmacokinetics

The PK profile of crizotinib was evaluated in rats, dogs, and monkeys following intravenous (IV) and oral administration.

Absorption

Following a single oral crizotinib dose, time to peak plasma concentration (Tmax) ranged from 1 to 6 hours. Oral bioavailability of crizotinib was 26-63% in rats, 38-66% in dogs, and 44% in monkeys.

Distribution

Following a single IV administration, the apparent volume of distribution (Vss) values greatly exceeded the volume of total body water of the respective species, indicating extensive distribution of crizotinib into body tissues from plasma.

Following a single oral administration of [14C]-labeled crizotinib in rats, crizotinib-derived radioactivity was detected in most tissues and organs, with the highest concentrations observed in liver, uveal tract, adrenal gland, small intestine, and pituitary gland.

Crizotinib is a substrate and an inhibitor of P-glycoprotein (P-gp) in vitro. Hepatic transporters did not appear to play a significant role in crizotinib hepatic uptake in vitro, indicating that crizotinib entered hepatocytes via passive diffusion. Crizotinib demonstrated moderate to high plasma protein binding, with mean unbound fractions of 0.036, 0.057, 0.070, 0.043, 0.072, and 0.093 for mice, rats, rabbits, dogs, monkeys, and humans, respectively.

Following a single oral dose, crizotinib was detected in rat cerebrospinal fluid (CSF) at a CSF/plasma ratio similar to the unbound fraction in plasma, suggesting that crizotinib may cross the blood/brain barrier. The data were approached with caution and considered preliminary, as a non-validated high-performance liquid chromatography/mass spectrometry (HPLC/MS) was used to detect and quantify crizotinib in CSF in this experiment. In the definitive distribution study in rats, crizotinib concentration in brain and spinal cord was below the limit of quantification (45 ng equivalents [14C] crizotinib/g tissue).

Metabolism

In human hepatocytes and liver S9 fractions, the major metabolic pathways were oxidation and hydroxylation of the piperidine ring of crizotinib. Oxidation of the piperidine ring of crizotinib also appeared to be the major metabolic pathway in rats and dogs, although differences were observed regarding the relative amount of various metabolites across species and by genders. In female rats, direct sulfate conjugation of crizotinib was a major metabolic pathway, accounting for 18% of circulating radioactivity versus 3% in male rats.

In vitro studies demonstrated that cytochrome P450 (CYP) 3A4/5 were the major enzymes involved in the metabolic clearance of crizotinib and in the formation of key metabolites.

Elimination

Following a single IV crizotinib dose, the apparent terminal elimination half-life (t1/2) ranged from 2.3 to 9.6 hours in rats, 12 to 17 hours in dogs, and 6.5 to 7.9 hours in monkeys. In a whole body autoradiography study in rats, delayed elimination was observed in a number of tissues and organs: tissues with the longest t1/2 (range: 576 to 118 hours) were eye, epididymis, testis, pigmented skin, kidney cortex, and brown fat.

The primary route of elimination in animals was the faeces. After a single oral dose of [14C]-labeled crizotinib, >74% of the administered radioactive dose was eliminated in the faeces and <3% was recovered in the urine in rats and dogs. In rats, 38% and 62% of the administered dose was recovered in bile in males and females, respectively.

3.2.3 Toxicology

Single-Dose Toxicity

Single-dose toxicity studies were not formally conducted. Single doses of crizotinib were administered to both rats and dogs by oral gavage in various studies in safety-pharmacology and dose-escalation studies. Single doses of up to 25 mg/kg and up to 500 mg/kg were well tolerated in dogs and rats respectively, incurring minimal toxicity.

Repeat-Dose Toxicity

The chronic toxicity profile of crizotinib was evaluated in a series of studies conducted in rats and dogs. Crizotinib targeted several organ systems at approximately one- to five-times the human clinical exposure. Target organ systems of toxicity included: gastrointestinal (emesis and diarrhoea); hepatic (elevated transaminases without histological correlates); pulmonary (histiocytosis); cardiovascular (QT prolongation); reproductive (ovarian follicle necrosis and spermatocyte degenerations); and haematopoietic systems (bone marrow hypocellularity including myleloid and erythroid). These adverse effects correlate with clinical experience for crizotinib. Following a two-month recovery period, gastrointestinal, haematopoietic, hepatic and reproductive observations in animals were reversible.

Other crizotinib-related findings observed in rats included decreased body weight and food consumption, an effect on retinal function, salivary glands, and actively growing long bones. Phospholipidosis in multiple organs was of unclear clinical relevance and was observed at significantly higher exposures (1.6- to 2.9-fold) in male rats (1-month and 3-month repeat-dose studies) with a similar trend observed in a 7-day dog study. In rats, full reversibility of phospholipidosis after a 2-month recovery period was observed in all tissues except the prostate and mesenteric lymph node (kidney not evaluated), where foamy macrophages were noted with decreased incidence, severity and/or distribution.

Dose-limiting toxicity resulting in mortality occurred in rats (250 mg/kg/day in a 90-day study; 500 mg/kg/day in a 7-day study; and 2,000 mg/kg/day in a 2-day study); and dogs (up to 40 mg/kg/day in a 7-day dose escalation study).

Carcinogenicity

Carcinogenicity studies have not been performed.

Genotoxicity

Crizotinib demonstrated genotoxicity in a human lymphocyte chromosome aberration assay (in vitro) and a rat bone marrow micronucleus assay (in vivo). A positive kinetochore assay suggests an aneugenic mechanism. A no-effect level for aneugenicity was identified at 100 mg/kg/day (approximately four-fold the human clinical exposure).

Mutagenicity

Crizotinib is not considered a mutagen based on negative results in bacterial reverse mutation assays.

Phototoxicity

In an in vitro 3T3 fibroblast Neutral Red Uptake assay, crizotinib demonstrated a photo-irritation factor (PIF) of 3.4 suggesting it has phototoxic potential. Accordingly, the Product Monograph recommends that patients minimize their exposure to sunlight and other ultraviolet (UV) light-emitting sources.

Reproductive and Developmental Toxicity

Crizotinib was not teratogenic in pregnant rats or rabbits. Reduced foetal body weights were considered adverse effects in the rat and rabbit at 200 and 60 mg/kg/day, respectively (approximately two-fold human clinical exposure).

3.2.4 Summary and Conclusion

Overall, the non-clinical pharmacology and toxicology data submitted support the use of Xalkori (crizotinib) for the proposed indication. Crizotinib has demonstrated active potency in biochemical and cellular assays against ALK and c-MET. In vivo, crizotinib achieved sufficient oral exposure, was widely distributed, was metabolized by CYP3A4/5, and was primarily eliminated through the liver. Strong anti-tumour activity was exhibited in crizotinib-treated mice bearing xenografts that expressed EML4-ALK fusions and dysregulated c-Met. Efficacy was dose-dependent and correlated with PD inhibition of ALK fusion variants.

Crizotinib demonstrated an acceptable toxicity profile in rats and dogs that corresponds with clinical observations. The toxicology studies suggest multiple target organs or systems of toxicity. These issues have been addressed through appropriate labelling in the current Product Monograph. In summary, in vitro and in vivo data suggest that crizotinib is well tolerated, safe and selectively but potently inhibits ALK fusion proteins resulting in anti-tumour efficacy. The non-clinical results obtained to date reasonably predict the clinical benefit of single-agent Xalkori (crizotinib) in patients with ALK-positive advanced NSCLC. Clinical monitoring will clarify the safety profile of Xalkori in the clinical setting.

3.3 Clinical basis for decision

The Canadian regulatory decision on the clinical pharmacology, efficacy, and safety of Xalkori was based on a critical assessment of the Canadian application/submission. The foreign reviews completed by the United States FDA (March 30, 2011) were used as an added reference in the assessment Xalkori.

In parallel with the review of this submission, the Medical Device Bureau, Health Canada assessed and provided a positive regulatory decision of a companion diagnostic kit to identify patients with ALK-positive disease. This is reflected in the indication wording within the Product Monograph.

The clinical efficacy claim for Xalkori as monotherapy to treat patients with ALK-positive advanced NSCLC with or without brain metastases is supported based on a preliminary report of data from the pivotal expanded Phase I Study A8081001 (herein referred to as Study A1001) and supportive Phase II Study A8081005 (herein referred to as Study A1005). Safety data were also included from a supportive Phase III Study A8081007 (herein referred to as A1007). This study is ongoing. Studies A1001 and A1005 also provided pharmacodynamic, pharmacokinetic, and safety data.

Studies A1001 and A1005 are ongoing, open-label, single-arm, multicentre studies with 116 and 136 enrolled patients, respectively. Prior to enrolling in Study A1001, patients were required to have ALK-positive tumours identified using a number of local clinical trial assays. The median duration of treatment for patients in Study A1001 was 32 weeks. Patients in Study A1005 were required to have received at least one prior treatment regimen and harbour ALK-positive tumours prior to entering the study. ALK-positive NSCLC was identified using a Health Canada-approved Vysis ALK break-apart fluorescence in situ hybridization (FISH) assay. The median duration of treatment for Study A1005 was 22 weeks. For both studies, all patients received prior systemic therapy, with the exception of 16 patients (13%) in Study A1001 who had no prior systemic treatment for locally advanced or metastatic disease. Each patient received the recommended dose of 250 mg Xalkori taken orally twice per day.

Study A1007 is an ongoing Phase III, open-label, two-arm, randomized, multicentre, multinational study comparing crizotinib to standard of care chemotherapy (permetrexed or docetaxel) in patients with previously treated ALK-positive advanced NSCLS. In this study ALK-positive NSCLC patients were identified using a validated break-apart FISH assay kit. At the time of this review, 71 patients with ALK-positive NSCLC had received crizotinib treatment.

3.3.1 Pharmacodynamics

At the recommended therapeutic dose of 250 mg twice daily, crizotinib had a pronounced negative chronotropic effect in patients with ALK-positive NSCLC. In Study A1001, there was a clinically and statistically significant mean heart-rate reduction at steady-state of -10 to -14 beats per minute (bpm). In Study A1005, the mean heart rate reduction at steady-state was -15 to -16 bpm. A mechanistic explanation for this effect could be the antagonistic effects of the drug on L-type calcium channels. A negative chronotropic effect raises concerns about bradycardia and bradyarrhythmias.

At a dose of 250 mg twice daily, crizotinib caused QTc interval prolongation according to the Fridericia cube root heart rate correction. In Study A1005, the magnitude of the estimated effect was mean 10.3 msec at 6 h post-dosing on Day 22. QTc prolongation with crizotinib is consistent with its activity as an inhibitor of human Ether-à-go-go Related Gene (hERG) channels. QTc prolongation creates an electrophysiological environment that favours the occurrence of Torsade de Pointes, which can progress to ventricular fibrillation and sudden cardiac death. Concerns regarding Torsade de Pointes are increased when QTc prolongation occurs in association with a reduction in heart rate. Crizotinib also appeared to cause some prolongation of the QRS duration, with a mean 2.5 msec increase from baseline at 6 h post-dosing on Day 22 in Study A1005.

The electrocardiogram (ECG) data from studies A1001 and A1005 were read using an automated reading method. Automated ECG reading methods are considered to be potentially unreliable when performing measurements of low amplitude waveforms, such as T waves and P waves. Erroneous automated readings are of particular concern in a patient population in which many subjects may have abnormal ECG waveforms because of the underlying disease state, co-morbidities, and concomitant medications. It is therefore necessary to consider the possibility that the analyses provided in these studies under-estimated the magnitude of the QTc or PR prolongation produced by crizotinib.

Two unexplained deaths occurred for which an arrhythmic cause cannot be excluded.

Both outlier analyses and analyses of central tendency raise concerns about an association of crizotinib with decreased serum calcium levels. Hypocalcaemia is a risk factor for Torsade de Pointes during treatment with a QTc prolonging drug.

Crizotinib decreases systolic and diastolic blood pressure, effects that seem likely to be related to its blockade of L-type calcium channels and alpha-1 adrenergic receptors. This PD effect raises concerns about orthostatic hypotension and syncope in patients with low blood pressure at baseline. Patients may need adjustment of concomitant antihypertensive therapies when receiving crizotinib.

Many tyrosine kinase inhibitors are associated with cardiomyopathy and decreased ventricular performance which can result in congestive heart failure. The administration of crizotinib 250 mg twice daily was associated with adverse events (AEs) such as edema, dyspnoea, cough, and wheezing that raise concerns about cardiotoxicity. An alternative explanation for the respiratory events would be pulmonary toxicity, as crizotinib has been associated with pneumonitis. In subsequent clinical trials, prospective echocardiography or multi-gated acquisition scans (MUGAs) should be performed for monitoring of ejection fraction.

Finally, treatment-emergent thrombotic events occurred in Studies A1001 and A1005 for which a causal relationship to crizotinib should be considered.

3.3.2 Pharmacokinetics

The single-dose PKs of Xalkori (crizotinib) were evaluated in healthy subjects and in patients with advanced tumours. The multiple-dose PKs of crizotinib were evaluated in patients with advanced solid tumours.

A single-dose, four-period crossover comparative bioavailability study (Study A8081011) in healthy subjects demonstrated that the Xalkori (crizotinib) commercial image 250 mg capsules met the recommended standards for bioequivalence in comparison to the clinical study formulations (immediate release tablets and powder in capsule formulations). In addition, the data from this study support the proposed labelling of the effect of food on crizotinib PKs. A high-fat meal decreased the rate and extent of absorption of crizotinib from the Xalkori capsules by approximately 14% in comparison to fasting conditions.

The maximum tolerated dose was 250 mg twice daily based on dose-limiting toxicities. The lowest crizotinib plasma concentrations (trough concentrations) achieved at steady-state following the 250 mg twice-daily dose exceeded the target efficacious concentrations predicted for ALK inhibition (111 ng/mL total drug).

Absorption

Following repeated 250 mg twice-daily dosing (recommended dose) in patients with advanced solid tumours, crizotinib plasma concentration reached steady-state within 15 days. In general, the systemic exposure appeared to increase in a greater than dose-proportional manner over the dose range of 200 to 300 mg twice daily.

Distribution

Crizotinib showed extensive distribution into tissues from the plasma. Binding of crizotinib to human plasma proteins in vitro was 91% and appeared to be independent of drug concentrations.

Metabolism

In vitro studies demonstrated that CYP3A4 and CYP3A5 were the major enzymes involved in the metabolic clearance of crizotinib. The primary metabolic pathways in humans were oxidation and O-dealkylation. In vitro studies in human microsomes demonstrated that crizotinib was a time-dependent inhibitor of CYP3A.

Following a single dose of 250 mg [14C]-labeled crizotinib in healthy volunteers, crizotinib accounted for 33% of the total recovered radioactivity in pooled plasma; crizotinib lactam metabolite was the predominant metabolite in plasma and accounted for 10% of the total recovered radioactivity.

Crizotinib lactam was approximately 2.5- and 7.7-fold less potent than crizotinib in inhibiting ALK and c-Met tyrosine kinases, respectively, in vitro. The O-desalkyl crizotinib and O-desalkyl crizotinib lactam were inactive against ALK and c-Met.

Elimination

Following a single 250 mg oral dose in patients with advanced solid tumours, the terminal half-life of crizotinib was 42 hours and the mean apparent clearance was 100 L/hr. At steady state, the clearance appeared to be lower (65 L/h) possibly due to auto-inhibition of CYP3A by crizotinib following repeated dosing.

Following the administration of a single 250 mg radiolabeled crizotinib dose to healthy subjects, 63% and 22% of the administered dose was recovered in faeces and urine, respectively. Unchanged crizotinib represented approximately 53% and 2.3% of the administered dose in faeces and urine, respectively.

Drug-Drug Interactions
CYP3A Inhibitors

Crizotinib is predominantly metabolized by CYP3A. Co-administration of a single 150 mg oral dose of crizotinib in the presence of ketoconazole (200 mg twice daily), a strong CYP3A4 inhibitor, resulted in a 3.2-fold increase in crizotinib systemic exposure compared to that when crizotinib was administered alone. The Product Monograph recommends avoiding concurrent use of strong CYP3A inhibitors. Caution should be exercised when moderate CYP3A inhibitors are co-administered.

CYP3A Inducers

Co-administration of a single 250 mg crizotinib dose with rifampin, a strong CYP3A inducer, resulted in an 82% decrease in crizotinib exposure compared to the administration of crizotinib alone. The Product Monograph recommends avoiding the concurrent use of strong CYP3A inducers.

CYP3A Substrates

Crizotinib is a moderate inhibitor of CYP3A and may increase plasma concentrations of co-administered CYP3A substrates. The Product Monograph recommends that caution be exercised in administering crizotinib in combination with drugs that are predominantly metabolized by CYP3A, particularly those that have narrow therapeutic indices and that are associated with life-threatening arrhythmias.

P-glycoprotein (P-gp) Substrates

Crizotinib is an inhibitor of P-gp in vitro. Consequently, crizotinib may increase plasma concentrations of co-administered drugs that are substrates of P-gp.This is indicated in the Product Monograph.

Gastric Protectant Medications

The aqueous solubility of crizotinib is pH dependent, with high pH (less acidic) resulting in lower solubility. A formal drug-interaction study with an antacid has not been conducted. The use of antacids was permitted during crizotinib treatment in clinical trials.

Other Drug-Drug Interactions

The concomitant use of Xalkori with QT interval-prolonging drugs or with drugs that can disrupt electrolyte levels should be avoided to the extent possible. For further information, please consult the Product Monograph.

Drug-Food Interactions

A high-fat meal reduced crizotinib exposure by approximately 14% when a 250 mg dose was given to healthy volunteers. The Product Monograph recommends that crizotinib be administered with or without food.

Foods and herbal products that may modify CYP3A activity should be avoided. These include grapefruit, pomegranate, Seville oranges, and St. John's wort.

Special Populations and Conditions
Hepatic Impairment

Xalkori has not been studied in patients with hepatic impairment. The clinical studies conducted excluded patients with alanine aminotransferase (ALT) or aspartate aminotransferase (AST) levels greater than 2.5-times the upper limit of normal (ULN) or, if due to underlying malignancy, greater than 5.0-times the ULN, or with total bilirubin greater than 1.5-times the ULN.

Renal Impairment

Xalkori has not been studied in patients with severe renal impairment. No starting dose adjustment is recommended for patients with mild or moderate renal impairment, although data are limited. Clinical data are not sufficient to determine if there is the potential need for starting dose adjustment in patients with severe renal impairment. No data are available for patients with end-stage renal disease.

Ethnicity

After 250 mg twice daily dosing, steady-state crizotinib maximum concentration (Cmax) and the area under the curve for the dosing interval (AUCt) in Asian patients were 1.57- and 1.50-fold those seen in non-Asian patients, respectively. There was a higher incidence of Grade 3 or 4 adverse events (AEs) in non-Asians (17%) than Asians (10%).

The identified risks and precautions related to PK findings have been properly labelled in the PM to facilitate the safe use of the drug.

3.3.3 Clinical Efficacy

The efficacy of Xalkori as monotherapy to treat patients with ALK-positive advanced NSCLC with or without brain metastases has been assessed in the ongoing studies A1001 and A1005. For a description of these studies please refer to the beginning of section 3.3 Clinical Basis for Decision.

In both studies combined, 255 previously treated or untreated ALK-positive advanced or metastatic NSCLC patients received the recommended dose of 250 mg crizotinib twice daily. The primary efficacy endpoint for both studies was Objective Response Rate (ORR) according to Response Evaluation Criteria in Solid Tumors (RECIST) Version 1.0 in Study A1001 and RECIST Version 1.1 in Study A1005. The ORR was 61% in Study A1001 (with two complete responses) and 51% in Study A1005 (with one complete response). These values, although mostly comprised of partial responses, were substantial. Notably, more than 90% of patients in these studies showed some degree of target lesion shrinkage.

The secondary endpoints for both studies included Duration of Response (DR), Time to Tumour Response (TTR), Disease Control Rate (DCR), Progression-Free Survival (PFS), and Overall Survival (OS). The median time to response in both studies was between 6 and 7.7 weeks with a median duration of response of 42 to 48 weeks. There were 31 patients in study A1001 who had stable disease for a DCR at 8 weeks of 79%. Fifty-five percent (55%) of objective tumour responses in this study were achieved during the first 8 weeks of treatment. There were 45 patients in study A1005 who had stable disease for a DCR at 6 weeks of 85%. Seventy-nine percent (79%) of objective tumour responses in this study were achieved during the first 8 weeks of treatment. To date, there are no data demonstrating an OS or PFS benefit in this patient population.

Although these preliminary data are based on single-arm studies with no control arm, the ORR is substantial and may in fact underestimate the true value which is expected from randomized controlled study data. The indication for Xalkori is granted on the basis of the ORR data from the ongoing studies A1001 and A1005. Confirmation of clinical benefit will be assessed based on the two Phase III randomized controlled studies (Study A1007 and A1014). The final Clinical Study Reports (CSRs) from Study A1007 and A1014 are the confirmatory studies listed as post-approval commitments. In addition, the sponsor will also provide the final complete CSRs from Study A1001 and A1005 to Health Canada.

3.3.4 Clinical Safety

The clinical safety of Xalkori was evaluated based on the data provided from the studies described at the beginning of section 3.3 Clinical Basis for Decision.

The major AEs reported included gastrointestinal and visual disturbance predominantly mild to moderate in severity. Serious adverse events (SAEs) included elevated transaminase levels, QT prolongation, interstitial lung disease, neutropaenia, and lymphopaenia. These SAEs were typically manageable through dose modification. Nevertheless, deaths did occur including those from pneumonitis, interstitial lung disease, cardiac arrest, and hepatic failure. Hy's Law cases were reported in patients treated with Xalkori. The proportion of patients with AEs differed slightly with age with more AEs noted for patients over the age of 65. As well, more AEs were reported in females versus (vs.) males and in Asian vs. non-Asian patients. In general, results to date suggest that crizotinib 250 mg, twice daily, is generally well tolerated and has a manageable safety and laboratory profile.

Although the number of patients in the safety database is adequate, the proportion of patients with ALK-positive NSCLC is small. In addition, the safety update for Study A1001 did not provide information on Grade 1-4 AEs in these patients, but only provided information on SAEs and deaths. Data from patients who were not ALK-positive and who received Xalkori are useful in helping to further evaluate the safety profile of Xalkori. Of note is that many of these patients, including those in the Dose Escalation Cohort and those with Xalkori-refractory tumours, had a short duration of exposure to Xalkori. Data with regard to these patients may not sufficiently reflect the AEs associated with crizotinib. In addition, the absence of randomized controlled trials in this submission makes it difficult to distinguish AEs due to disease progression and those due to the use of Xalkori. Two Phase III randomized studies (Study A1007 and Study A1014) are expected to allow for a comparison of safety and efficacy between Xalkori-treated and comparator-drug treated patient groups. Health Canada has requested the final CSRs for these Phase III studies as post-approval commitments.

Three major safety issues were identified in this submission:

  • QT interval prolongation and bradycardia;
  • Hepatotoxicity, including fatal outcomes; and
  • Pneumonitis, including fatal outcomes.

These issues have been addressed through appropriate labelling (a Serious Warnings and Precautions box) in the Product Monograph. Other important safety risks reported in studies with Xalkori are managed through strict labelling. The labelling also highlights the importance of the requirement to utilize laboratories with demonstrated proficiency in using a validated diagnostic assay to assess ALK fusion, to avoid inappropriate treatment in ALK-negative patients for whom the benefit of Xalkori is not established. For more information about QT interval prolongation and bradycardia, see section 3.3.1 Pharmacodynamics.

No data exist in patients with hepatic or severe renal impairment. There is an ongoing multiple-dose study to determine the appropriate dose of Xalkori in patients with various levels of severity of hepatic impairment. The anticipated study completion date is July 2013 and the final CSR is expected by January 2014. Although not part of the post-approval commitments, the Sponsor has agreed to submit the final CSR to the Health Canada to evaluate the risk of Xalkori treatment in patients with hepatic impairment.

There is also an ongoing clinical trial to determine the appropriate dose of Xalkori in patients with severe renal impairment. The anticipated study completion date is April 2012 and the final CSR is expected by October 2012. Although not part of the post-approval commitments, the Sponsor agreed to submit the final CSR to the Bureau to evaluate the risk of Xalkori treatment in patients with severe renal impairment.

The sponsor has agreed to provide final CSRs to further evaluate the possible serious safety risks associated with the use of Xalkori. A list of these reports is found in section 3.3.5 Additional Issues.

3.3.5 Additional Issues

In keeping with the provisions outlined in the Notice of Compliance with Conditions (NOC/c) Guidance, the sponsor is required to submit the results of the following confirmatory studies:

  • Study A1007 is an ongoing Phase III, randomized, open-label study of the clinical efficacy and safety of crizotinib vs. standard of care (pemetrexed or docetaxel) in patients with advanced NSCLC harbouring a translocation or inversion event involving the ALK gene locus. The anticipated completion date of the study is December 2013. Submission of the final CSR is expected by June 2014.
  • Study A1014 is an ongoing Phase III, randomized, open-label study of the efficacy and safety of crizotinib vs. pemetrexed/cisplatin or pemetrexed/carboplatin in previously untreated patients with non-squamous carcinoma of the lung harbouring a translocation or inversion event involving the ALK gene locus. To evaluate the clinical benefit of crizotinib as a first-line therapy, the sponsor will submit the final CSR from this study. The anticipated completion date of the study is December 2015. Submission of the final CSR is expected by June 2016.

In addition, to evaluate the efficacy and safety of the use of crizotinib, the sponsor will submit the final exposure-response analysis for PFS, response rate, OS, and safety endpoints, utilizing data from each study. These data are important given that the efficacy endpoints, PFS, response rate, and OS are outcomes that Health Canada deems to be sufficient to grant a Notice of Compliance (NOC) for the proposed indication, along with a continued favourable benefit/risk profile for crizotinib.

The sponsor has also agreed to Health Canada's request to submit the following:

  • The final CSR from Study A1001 and Study A1005;
  • Final study reports to further evaluate the serious risk of drug interactions (including CYP2B and CYP2C enzymes, CYP3A inhibitors, and CYP3A inducers);
  • A final CSR to evaluate the serious risk of visual impairment;
  • A final CSR to evaluate the serious risk of QTc interval prolongation;
  • Pending results for echocardiography/MUGA. The high incidence of oedema and dyspnea raise concerns about decreased left ventricular ejection fraction (LVEF) and heart failure. These data will be provided in the final CSRs for Study A1001, A1005, and A1007. The sponsor should incorporate troponin and B-type natriuretic peptide as clinical laboratory biomarkers in subsequent clinical studies, in addition to MUGA or echocardiography monitoring for the measurement of ejection fraction;
  • A final CSR to evaluate the risk of crizotinib treatment in patients co-administered pH elevating agents;
  • A final CSR (as part of the CSR for Study A1001) to evaluate the efficacy and safety of the use of crizotinib in patients with ALK-negative tumours; and
  • As part of future Periodic Safety Update Reports (PSURs) for products authorized under the Notice of Compliance with Conditions Guidance (PSUR/Cs), data with regard to crizotinib resistance.

These items are not listed as post-approval commitments.

In addition, as part of the market authorization, Health Canada has requested a Risk Management Plan (RMP) to monitor whether the benefits continue to outweigh any risk. Health Canada has requested the following post-marketing information from the sponsor (and agreed to by the sponsor) which addresses the following issues raised by Health Canada and which are not included as post-approval commitments in the Letter of Undertaking. In future RMP submissions, the sponsor should address the following issues:

  • Information pertaining to the conception, specificity, sensitivity, and availability of companion tests;
  • A discussion as to what extent a companion test or other available tests could generate false negative/positive results and the strategy adopted to minimize the impact of these false negative/positive results;
  • The sponsor should provide an estimation of mean treatment duration;
  • Data from future clinical trials and/or post marketed surveillance activities should be collected and analysed to evaluate the effect of crizotinib on the elderly population;
  • The rationale supporting the decision to include vision disorders and QT prolongation as "potential" risks rather than "identified" risks associated with the use of crizotinib should be more explicitly developed;
  • The sponsor should update the RMP with more details from non-clinical studies related to cardiotoxicity and QTc prolongation in order to better appraise the cardiotoxicity associated with crizotinib. Those results should identify whether crizotinib's metabolites conserved their inhibitory effect on hERG and Ca2+ channels;
  • The sponsor should provide a more in depth discussion of the inhibitory effects of crizotinib on 5-HT4 receptors (along with the other 5-HT receptors) and on the dopamine active transporter (DAT) and its possible role in bradycardia observed in patients using crizotinib;
  • The sponsor should discuss the implication of the inhibitory effects of crizotinib on 5-HT4 receptors and DAT on the observed neuropathy associated with the use of crizotinib. The sponsor should also provide a more in depth discussion of the data ascertaining that crizotinib does not cross the blood-brain barrier;
  • Considering that several clinical studies are still ongoing and that long-term effects of crizotinib on patients are still unknown, Health Canada recommends that the sponsor submit PSURs annually for the next 3 years (which Health Canada will review) following the Notice of Compliance in order to monitor adverse events associated with the cardiac, respiratory and hepatic functions; and
  • Updates on the ongoing clinical studies can be summarized in the PSUR, or presented as a separate report if warranted.

All the identified and potential risks addressed in the RMP should be discussed separately in each PSUR report.

3.4 Benefit/Risk Assessment and Recommendation

3.4.1 Benefit/Risk Assessment

Xalkori was authorized under the NOC/c Guidance on the basis of the promising nature of the clinical evidence, and the need for confirmatory studies to verify the clinical benefit. Patients should be advised of the fact that the market authorization was issued with conditions. Xalkori is indicated as monotherapy for use in patients with ALK-postive advanced (not amenable to curative therapy) or metastatic NSCLC. Assessment for ALK-positive advanced or metastatic NSCLC should be performed by laboratories with demonstrated proficiency in the specific technology being utilized. Improper assay performance can lead to unreliable test results.

The market authorization with conditions was based on the primary efficacy endpoint of ORR as well as DR in clinical studies A1001 and A1005 based on investigator assessment using RECIST. There are no data demonstrating improvement in OS or PFS with Xalkori. It remains to be determined whether, based on long-term data from randomized Phase III studies A1007 and A1014, the endpoint measures ORR and DR translate into improvements of OS and PFS.

In Study A1001, only 13% of patients had no prior systemic therapy while the majority (87%) had prior therapy. In the ongoing study A1014, patients are administered Xalkori in the first-line setting. Therefore, based on the current single arm studies, although the benefit of Xalkori is demonstrated in patients with and without previous systemic therapies, the results of Study A1014 will provide more conclusive results.

Most of the NSCLC patients analyzed in studies A1001 and A1005 were ALK-positive with only five patients having ALK-negative status. Due to this small sample size, it was not possible to perform a subgroup analysis to determine whether Xalkori demonstrates a benefit in ALK-negative patients. The benefit of Xalkori in ALK-negative NSCLC patients has therefore not been established, so uncertainty with regard to this subgroup is evident. This is based on the assumption that the diagnostic assay to detect ALK fusion has a high sensitivity and specificity. For this reason, it is important to be aware of the false-positive rate associated with a validated assay to identify ALK fusion. The risk is that patients who are truly ALK-negative, yet incorrectly identified as ALK-positive based on the assay, may be unnecessarily treated with Xalkori and thus exposed to the adverse risks. The stipulation to use a diagnostic assay with a high sensitivity and specificity to ensure that only those patients for whom the benefit of Xalkori treatment has been established are treated has been included as part of the authorized indication.

The fact that these single-arm studies are comprised of small sample sizes and the lack of a comparator arm renders it difficult to determine which AEs are attributable to Xalkori and which are attributable to disease progression. Confirmation of clinical benefit and a more definitive Xalkori safety profile is expected from the results of two Phase III randomized controlled studies (Study A1007 and Study A1014). The final CSRs for these studies are the confirmatory studies listed as post-approval commitments. Study A1007 will be reported by June 2014 and Study A1014 will be reported by June 2016. The CSRs for each trial will include the final exposure-response analysis for PFS, response rate, OS and safety endpoints, utilizing data from each trial. The sponsor has also agreed to submit to Health Canada on an annual basis status reports on the progress of these ongoing confirmatory trials within 60 calendar days of the market authorization anniversary.

Based on current evidence provided in the expanded Phase I and Phase II studies, the important safety risks associated with Xalkori treatment (such as, pneumonitis/interstitial lung disease, hepatotoxicity, QT/PR interval prolongation, bradycardia, visual disorders, peripheral oedema, and neuropathy) are managed through strong labelling.

Reports of all serious adverse drug reactions (ADRs) in Canada and all serious unexpected ADRs outside of Canada will be forwarded within 15 days to Health Canada in accordance with the current Food and Drug Regulations (C.01.017) and guidance documents. PSUR/Cs will be prepared in accordance with ICH guidelines and submitted semi-annually to Health Canada until the conditions have been fulfilled and removed from the NOC/c by Health Canada. Included as a stand-alone document or contained within the PSUR/C will be the genetic analysis data from patients who have progressed on crizotinib.

In the event of specific issues of concern, the Sponsor will:

  1. notify Health Canada within 15 days when, on the basis of safety, efficacy or quality concerns related to Xalkori, an expert panel or advisory committee has been struck in a foreign jurisdiction to address an issue or when there has been significant regulatory action in another jurisdiction including a direction to issue warnings, a health advisory or the removal of a product from the market; and
  2. within 30 days of the notification in 1., or in a timeframe acceptable to Health Canada and the Sponsor, submit to Health Canada a written report on the specific issue that prompted the regulatory action in the foreign jurisdiction. To date, eight countries have received marketing authorization for Xalkori.

The benefits of Xalkori for use as monotherapy to treat patients with ALK-positive advanced or metastatic NSCLC were judged to outweigh the risks. The lack of effective systemic therapies for patients with this rare disease (advanced or metastatic ALK-positive NSCLC) constitutes an unmet medical need which Xalkori appears to fulfil. Although the preliminary efficacy values are based on single-arm studies with no control arm, the ORR is substantial and may in fact underestimate that true value which is expected from randomized controlled study data from studies A1007 and A1014. In order to monitor that the benefits of Xalkori continue to outweigh any risk, an RMP has been required that includes several post-approval recommendations. In addition, Health Canada has requested that future RMPs address the issues listed in section 3.3.5 Additional Issues.

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 Xalkori is favourable for use as monotherapy in patients with ALK-positive advanced (not amenable to curative therapy) or metastatic NSCLC. Using a validated ALK assay, assessment for ALK-positive advanced or metastatic NSCLC should be performed by laboratories with demonstrated proficiency in the specific technology being utilized. Improper assay performance can lead to unreliable test results. ALK gene rearrangements were identified in Study A1001 by clinical trial assays performed by local laboratories and in Study A1005 by a Health Canada-approved Vysis ALK break-apart FISH assay.

The clinical benefit of Xalkori in patients with ALK-negative NSCLC has not been established; therefore Xalkori is not recommended for these patients.

Marketing authorization with conditions was based on a primary efficacy endpoint of ORR as well as DR in clinical Studies A1001 and A1005, based on investigator assessment using RECIST and confirmed through an independent radiology review. There are no data available demonstrating improvement in survival with Xalkori.

This New Drug Submission (NDS) qualifies for authorization under the Notice of Compliance with Conditions (NOC/c) Guidance. The NDS 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.

In keeping with the provisions outlined in the Notice of Compliance with Conditions (NOC/c) Guidance, the sponsor has agreed to submit the results of the confirmatory studies listed as well as the additional data listed in section 3.3.5 Additional Issues.

4 Submission Milestones

Submission Milestones: XalkoriTM

Submission MilestoneDate
Pre-submission meeting:2011-02-11
Acceptance of Advance Consideration under the Notice of Compliance with Conditions (NOC/c) Guidance:2011-03-29
Submission filed:2011-06-08
Screening
Screening Deficiency Notice issued:2011-07-15
Response filed:2011-08-11
Screening Acceptance Letter issued:2011-08-30
Review
Biopharmaceutics Evaluation complete:2011-12-21
Quality Evaluation complete:2012-03-12
Clinical Evaluation complete:2012-04-24
Biostatistics Evaluation complete:2012-02-20 - 2012-02-22
Labelling Review complete:2012-04-24
NOC/c Qualifying Notice (NOC/c-QN) issued2012-03-15
Response filed:2012-03-23
Notice of Compliance (NOC) issued by Director General under the NOC/c Guidance:2012-04-25