Summary Basis of Decision for Zelboraf ™

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
ZelborafTM

Vemurafenib, 240 mg, Tablet, Oral

Hoffmann-La Roche Ltd.

Submission control no: 148693

Date issued: 2012-05-30

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:

ZelborafTM

Manufacturer/sponsor:

Hoffmann-La Roche Ltd.

Medicinal ingredient:

Vemurafenib

International non-proprietary Name:

Vemurafenib

Strength:

240 mg

Dosage form:

Tablet

Route of administration:

Oral

Drug identification number(DIN):

  • 02380242

Therapeutic Classification:

Protein kinase inhibitor

Non-medicinal ingredients:

Colloidal anhydrous silica, croscarmellose sodium, hydroxypropyl cellulose, hydroxypropyl methylcellulose acetate succinate, magnesium stearate, iron oxide red, macrogol 3350, polyvinyl alcohol, talc, and titanium dioxide

Submission type and control no:

New Drug Submission, Control Number: 148693

Date of Submission:

2011-07-18

Date of authorization:

2012-02-15

™Trademark of F. Hoffmann-La Roche AG, used under license

2 Notice of decision

On February 15, 2012, Health Canada issued a Notice of Compliance to Hoffmann-La Roche Limited for the drug product, Zelboraf.

Zelboraf contains the medicinal ingredient vemurafenib which is a kinase inhibitor that slows down or stops the growth of melanoma tumour cells expressing mutated BRAF V600 proteins. BRAF is mutated in approximately half of patients with advanced melanomas.

Zelboraf is indicated as a monotherapy for the treatment of BRAF V600 mutation-positive unresectable or metastatic melanoma. A validated test is required to identify BRAF V600 mutation status.

The market authorization was based on quality, non-clinical, and clinical information submitted. The efficacy and safety of Zelboraf in patients with melanoma were evaluated in treatment naïve patients and patients who failed at least one prior systemic therapy.

  • BRAF V600 mutations were identified in the Phase III and Phase II studies by the Health Canada approved cobas® 4800 BRAF V600 Mutation Test.
  • Clinical data supporting the effectiveness of Zelboraf in patients with BRAF mutations other than V600E are limited.
  • Effectiveness of Zelboraf in patients with prior therapy, including dacarbazine and interleukin-2 (IL-2) treatments, was based on objective response rate data from a single-arm Phase II study.

Treatment Naïve Patients: In an open-label, multicentre, international, randomized Phase III study in previously untreated patients with BRAF V600 mutation-positive unresectable or metastatic melanoma, 675 patients were randomized 1:1 to treatment with Zelboraf (960 mg twice daily) or dacarbazine (1,000 mg/ every 3 weeks). Treatment was continued until time of disease progression, unacceptable toxicity and/or consent withdrawal. Statistically significant and clinically meaningful improvements were observed in overall survival and progression-free survival. Overall survival was longer with Zelboraf compared to dacarbazine with a hazard ratio of 0.37 which represents a 63% decrease in the hazard of death with Zelboraf compared to dacarbazine. Progression-free survival by investigator assessment was longer with Zelboraf compared to dacarbazine with a hazard ratio for progression or death of 0.26 which represents a 74% decrease in the hazard of progression or death for Zelboraf compared to dacarbazine.

Patients who Failed at least One Prior Systemic Therapy: A Phase II single-arm, multicentre, multinational study was conducted in 132 metastatic melanoma patients with BRAF V600 mutation-positive tumours. The primary endpoint of confirmed best overall response rate (BORR) by independent review committee assessment was 52%. Patients with prior IL-2 or dacarbazine therapy had a BORR of 48% and 60%, respectively. The median time to response was 1.4 months with 75% of responses occurring by month 1.6 of treatment. The median duration of response was 6.5 months.

Zelboraf (240 mg, vemurafenib) is presented in tablet form. The recommended dose of Zelboraf is 960 mg (four 240 mg tablets) twice daily. It is recommended that treatment with Zelboraf continue until disease progression or the development of unacceptable toxicity. Dosing guidelines are available in the Product Monograph. Zelboraf should be prescribed and supervised by a qualified physician experienced in the use of anti-cancer agents.

Zelboraf is contraindicated for patients who are hypersensitive to Zelboraf or to any ingredient in the formulation. Zelboraf 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 Zelboraf are described in the Product Monograph.

Serious safety issues identified in the clinical studies included QTc interval prolongation and second malignancies. Also, Zelboraf was not studied in patients with severe hepatic impairment. These issues have been addressed through appropriate labelling in the Product Monograph.

Priority Review status was granted for the evaluation of Zelboraf as the new drug submission reported substantial evidence of clinical effectiveness that Zelboraf provided an improved benefit/risk profile over existing therapies for a serious, life-threatening disease that is not adequately managed by a drug marketed in Canada.

Based on the Health Canada review of data on quality, safety, and efficacy, Health Canada considers that the benefit/risk profile of Zelboraf as monotherapy is favourable for the treatment of BRAF V600 mutation-positive unresectable or metastatic melanoma. A validated test is required to identify BRAF V600 mutation status.

3 Scientific and Regulatory Basis for Decision

3.1 Quality Basis for Decision

3.1.1 Drug Substance (Medicinal Ingredient)

General Information

Vemurafenib, the medicinal ingredient of Zelboraf, is an orally bioavailable protein kinase inhibitor that slows or stops the growth of advanced melanomas that express BRAF V600 mutant serine-threonine kinases. Oncogenic mutations of the BRAF kinase, predominantly V600E, have been observed in approximately 50% of advanced melanomas.

Manufacturing Process and Process Controls

Vemurafenib 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 were 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.
Characterization

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

The sponsor has provided a summary of all drug-related impurities. The proposed limits are considered adequately qualified [that is (i.e.) within ICH limits and/or qualified from toxicological studies]. Control of the impurities and degradation products is therefore considered acceptable.

Control of Drug Substance

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

The proposed packaging components are considered acceptable.

Stability

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

3.1.2 Drug Product

Description and Composition

Zelboraf 240 mg film-coated tablets are oval, biconvex, pinkish-white to orange-white film-coated tablets with "VEM" engraved on one side. Each Zelboraf film-coated tablet contains 240 mg vemurafenib. Non-medicinal ingredients include: colloidal anhydrous silica; croscarmellose sodium; hydroxypropyl cellulose; hydroxypropyl methylcellulose acetate succinate; and magnesium stearate. The film-coating mixture includes: iron oxide red; macrogol 3350; polyvinyl alcohol; talc; and titanium dioxide.

Zelboraf tablets are available in aluminium blister packs containing 56 film-coated tablets (8 tablets per blister card and 7 blister cards per carton).

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 vemurafenib 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 pharmaceutical development are considered acceptable upon review.

Manufacturing Process and Process Controls

The manufacturing process uses conventional manufacturing techniques, namely: dry granulation; compression; and film-coating.

The validated process is capable of consistently generating product that meets release specifications.

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

Control of Drug Product

Zelboraf is tested to verify that its identity, appearance, content uniformity, assay, physical form, dissolution, moisture content, levels of degradation products, drug-related impurities, and microbiological 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.

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

Stability

Based on the real-time, long-term, accelerated stability data submitted, the proposed 24-month shelf-life is considered acceptable when Zelboraf is stored at 15-30°C in the original package, protected from moisture.

The compatibility of the drug product with the container closure system was demonstrated through 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 facility and equipment that are involved in the production of Zelboraf are considered suitable for the activities and products manufactured.

3.1.4 Adventitious Agents Safety Evaluation

Not applicable. The excipients used in the drug product formulation are not from animal or human origin.

3.1.5 Conclusion

The Chemistry and Manufacturing information submitted for Zelboraf has demonstrated that the drug substance and drug product can be consistently manufactured to meet the approved specifications. Proper development and validation studies were conducted, and adequate controls are in place for the commercial processes.

3.2 Non-Clinical Basis for Decision

3.2.1 Pharmacodynamics

Pharmacological studies have demonstrated that vemurafenib is an inhibitor of BRAF and other members of the RAF family of kinases. In vitro, vemurafenib inhibits BRAF with mutations at position V600 in the low nanomolar range and at lower inhibitory concentrations than for wild-type BRAF. The concentrations of vemurafenib required to inhibit 50% of the enzyme activity (IC50) for a number of BRAF V600 mutant kinases were between 7 and 14 nM whereas the majority of other unrelated kinases screened were inhibited at much higher concentrations (IC50 values >2 μM.

In cell culture studies, vemurafenib inhibited the proliferation of human tumour cells at IC50 concentrations ranging between 0.04 and 25 μM. Most melanoma cell lines with BRAF V600 mutations had growth inhibition at IC50 values of <1 μM. In contrast, cells wild-type for BRAF or cells with BRAF mutations at sites other than V600 had IC50 values between 4 and 25 μM.

In vivo experiments showed that vemurafenib inhibited the growth of transplanted melanoma cells in athymic mice. All of these cell lines expressed BRAF V600E mutations. At high doses, mice achieved complete regressions of the tumours but at lower doses the tumours reappeared presumably due to acquired resistance to vemurafenib. Resistant clones were isolated in order to identify adaptive tumour responses that might occur in melanoma patients treated with this drug. At least a 100-fold greater concentration of vemurafenib was required to inhibit growth of the resistant clones compared to parental cells. A number of mechanisms have now been reported that may confer resistance of melamonas to vemurafenib.

In clinical studies, vemurafenib was found to stimulate the development of cutaneous squamous cell carcinomas (cuSCC) in approximately 24% of patients treated for melanoma. The potential mechanism by which vemurafenib contributes to the development of cuSCC was evaluated in vivo in a human epidermoid carcinoma cell line (A431) cuSCC xenograft mouse model. At vemurafenib doses of 25 and 75 mg/kg/day, there was dose-dependent growth of the xenograft tumours when compared to vehicle-treated control mice. Preliminary evidence suggests that vemurafenib causes a paradoxical increase in mitogen-activated protein kinase (MEK) activity (p-MEK) in tumours expressing wild-type BRAF. Subsequent genetic analysis of 35 cutaneous squamous cell carcinomas and keratoacanthom (cuSCC/KA) samples from patients treated with vemurafenib revealed that 60% (21/35) of the specimens harboured RAS isoform mutations, the most prevalent being HRAS Q61L. This information, coupled with the finding that cuSCC often develops within 7 to 8 weeks, suggests that there are pre-existing lesions whose growth is stimulated following vemurafenib administration.

Safety pharmacology studies were conducted to evaluate the potential effects of vemurafenib on the cardiovascular, respiratory and central nervous system. Vemurafenib inhibited the human Ether-à-go-go-Related Gene (hERG) channel expressed in human embryonic kidney cells with an IC50 of 1.24 μM which is much lower than the maximum plasma concentration (Cmax) of 90 μM observed in patients. This assay indicates that vemurafenib has the potential to cause QT prolongation.

Vemurafenib had no effect on conduction when tested in the Purkinje fibre assay. However, these results are confounded by the limited or highly variable solubility of vemurafenib under the conditions of this assay.

In an electrocardiogram (ECG) study in conscious dogs in which the exposure of vemurafenib was only half the human clinical exposure following a single oral administration, there was no effect on QT prolongation when corrected for heart rate. The low exposure levels achieved in dogs limit the interpretation of these QT results.

No adverse effects were observed in the central nervous system and respiratory system in the safety pharmacology studies conducted in rats.

3.2.2 Pharmacokinetics

Absorption

Pharmacokinetic (PK) studies conducted in rats and dogs demonstrated saturable absorption of vemurafenib following oral administration. In one study, two doses given 8 hours apart produced a 1.8-fold increase in systemic exposure based on area under the curve (AUC) values, in dogs as compared to a single, oral dose of 450 mg/kg, although the Cmax values were similar.

Distribution

Vemurafenib was highly bound (>99%) to plasma proteins, as determined in the in vitro studies with plasma obtained from rats, mice, dogs, monkeys, and humans.

In a quantitative whole body autoradiography study conducted in rats using radiolabelled vemurafenib, concentrations of radioactivity in tissues were relatively similar to blood concentrations at all time points, except for liver, kidney, adrenal cortex, lachrymal glands, lung, and alimentary canal tissues, which were generally higher than blood. Radioactivity was not detectable in the brain and spinal cord of rats suggesting vemurafenib does not cross the blood-brain barrier. Accumulation and retention of vemurafenib in melanin-containing tissues of the eye (uveal tract) or skin was not apparent.

Metabolism

The cytochrome P450 (CYP) isozyme, CYP3A4, was the primary enzyme responsible for the in vitro metabolism of vemurafenib. Eight in vitro minor metabolites were identified in liver microsomes/hepatocytes of humans, dogs, and rats. In plasma from rats, dogs, and humans, unchanged vemurafenib was the major drug component and two minor monohydroxylation metabolites were detected.

Excretion

In the rat studies with radiolabelled vemurafenib, the elimination of radioactivity was mostly in the faeces and only 0.19% was recovered in the urine at 96 hours post treatment.

In bile duct cannulated rats, most of the radioactivity collected over 24 hours was recovered in the bile (71%) and less in the faeces (13.4%) demonstrating that the major elimination route of drug-derived radioactivity was biliary excretion in this animal.

3.2.3 Toxicology

Single-Dose Toxicity

Single-dose toxicity studies were conducted in rats and dogs with a vemurafenib suspension in corn oil and the exposure was too low to induce acute toxicity. Due to the limited systemic exposures with the corn oil formulation, a new formulation was developed, called micro-precipitated bulk powder (MBP). This formulation provided significantly higher exposure than the previous formulation, and was used in the 13-week and 26-week repeat-dose toxicity studies, the prematurely terminated 39-week toxicity study, the in vivo micronucleus assay, and embryo-foetal developmental toxicity studies. This formulation was used subsequently for the remainder of the non-clinical and all clinical studies.

Repeat-Dose Toxicity

Repeat-dose toxicology studies identified the liver and bone marrow as target organs in the dog. Hepatocellular necrosis and degeneration were observed in the liver at exposures below the clinical exposure (based on AUC comparisons) in a 13-week dog study with twice-daily dosing. Focal bone marrow necrosis was noted in one dog in a prematurely terminated 39-week dog study with twice-daily dosing at exposures within the range of clinical exposures. In the 13-week dog study, increases in QT interval were reported in male dogs at the high dose after 24 days of dosing but these increases did not reach statistical significance and no other abnormalities in the other ECG measurements were observed.

Phototoxicity

Vemurafenib was phototoxic in vitro in cultured murine fibroblasts after Ultraviolet A irradiation.

Genotoxicity

Vemurafenib was not mutagenic in the in vitro assays (Ames test and chromosome aberration assay in human lymphocytes) nor in the in vivo rat bone marrow micronucleus test. The concentrations of vemurafenib used in these assays were relatively low, due to its low water solubility.

Carcinogenicity

Carcinogenicity studies were not conducted as they are not required to support marketing for therapeutics intended to treat patients with advanced cancer.

Reproductive and Developmental Toxicity

Embryo-foetal developmental studies were conducted in rats and rabbits; in rats at doses of 0, 10, 30 and 250 mg/kg/day during the gestation days of 6 through 17, and in rabbits at doses of 0, 30, 150, and 450 mg/kg/day during the gestation days of 7 through 20. Vemurafenib did not induce any effects on the embryo-foetal development of rats and rabbits. However, the exposure levels of pregnant animals were relatively low as compared to the human exposure levels in the clinical studies.

Vemurafenib crossed the placenta in rats. This exposure may cause foetal harm by interfering with BRAF function, which is essential for the developing embryo.

Fertility studies were not conducted as they are not required to support marketing for therapeutics intended to treat patients with advanced cancer.

3.2.4 Summary and Conclusion

The non-clinical pharmacology, safety pharmacology, pharmacokinetic, and toxicology studies have characterized the non-clinical profile of vemurafenib in sufficient detail to support the intended use of Zelboraf for the clinical indication. Based on the pharmacology studies, vemurafenib has the potential to selectively inhibit BRAF V600 mutants and act as an inhibitor of melanoma tumour growth expressing these BRAF V600 oncogenic driver mutations. No major safety concerns were identified that would predict unexpected toxicities in patients treated with vemurafenib.

3.3 Clinical basis for decision

3.3.1 Pharmacodynamics

Clinical studies on the mechanism of action of vemurafenib were not submitted for review (see 3.2.1 Pharmacodynamics for the non-clinical studies). Dose-escalation studies were carried out in patients in order to establish an appropriate dose of 960 mg twice daily (BID) for a total daily dose of 1,920 mg (8 tablets total) for the Phase II and Phase III studies. This is considered the maximum tolerated daily dose.

3.3.2 Pharmacokinetics

A population PK analysis using pooled data from 458 patients estimated the median of the steady-state Cmax, Cmin and AUC0-12hours to be 62 μg/mL, 59 μg/mL and 734 μg*h/mL, respectively. A one-compartment open model with first-order absorption and first-order elimination adequately describes the vemurafenib concentration-time profile in the population PK analysis.

Absorption

Using the to-be-marketed 240 mg tablets, vemurafenib demonstrated linear pharmacokinetics between 240 and the recommended daily dose of 960 mg BID. Steady-state levels of vemurafenib were achieved at Day 22 in most patients (range: 15 to 29 days). The bioavailability of vemurafenib has not been established and the effect of food on absorption of vemurafenib is not currently known.

Vemurafenib at 960 mg BID (using the 240 mg tablets) was absorbed with a median time to reach maximum plasma concentration of approximately 4 hours. Vemurafenib exhibited marked accumulation after repeat dosing at 960 mg BID with high inter-patient variability.

Distribution

In patients with metastatic melanoma , the apparent volume of distribution for vemurafenib was estimated to be 91 L (with 64.8% inter-patient variability). Vemurafenib was highly bound to human plasma proteins in vitro (>99%).

Metabolism

In a human mass balance study using radiolabelled vemurafenib, vemurafenib underwent limited metabolism by the liver. Metabolites were on average <6% of the plasma radioactivity. In faeces, the parent compound and three primary metabolites accounted for 54.6% and 13.5% of the total starting radioactivity, respectively, when collected up to 96 hours.

Excretion

Results from the mass balance study showed that the majority of the radioactivity was excreted in the faeces (94%) and very little (<1%) was found in the urine.

Drug Interaction Studies

Vemurafenib may increase the plasma exposure of drugs predominantly metabolized by CYP1A2 and CYP2D6 and decrease the plasma exposure of drugs predominantly metabolized by CYP3A4. Dose reductions for medications predominantly metabolized via CYP1A2 and CYP2D6 should be considered based on their therapeutic windows before concomitantly treating with vemurafenib. When a single dose of warfarin was co-administered after repeat dosing with vemurafenib for 15 days, some patients exhibited increased warfarin exposure (mean 20%). Additional studies demonstrated that vemurafenib inhibited CYP2C9 in vitro. Therefore caution should be exercised when vemurafenib is co-administered with warfarin (a CYP2C9 substrate) in patients.

In vitro studies suggested that vemurafenib was only metabolized to a small extent by human hepatocytes and this was mainly due to CYP3A4-mediated oxidation. Studies in humans to determine the effects of either a strong CYP3A4 inhibitor or inducer on vemurafenib pharmacokinetics have not been conducted.

Special Populations

The kidneys account for less than 1% of excreted vemurafenib (mass balance study) and renal impairment is not expected to affect exposure to vemurafenib. Studies in rats suggest that the major route of elimination is biliary excretion and therefore hepatic impairment may increase systemic exposure to the drug. Based on population pharmacokinetic analysis, no adjustment to the starting dose is needed for patients with pre-existing mild and moderate hepatic impairment. The safety and efficacy of Zelboraf in patients with severe hepatic impairment have not been established. Limited pharmacokinetic data suggest that patients with severe hepatic impairment may have higher systemic concentrations of vemurafenib, and this could result in more frequent exposure-related adverse events including QTc prolongation. Therefore, Zelboraf should be used with caution in patients with severe hepatic impairment as outlined in the Product Monograph.

The mean steady-state exposure in females was approximately 14% higher than in males. Although there is no need to adjust the dose based on gender it remains unclear whether this exposure difference is related to gender or a body size effect.

3.3.3 Clinical Efficacy

The efficacy of Zelboraf (vemurafenib) in melanoma was evaluated in a Phase III comparative clinical study of 675 patients and a Phase II single-arm clinical study of 132 patients. Prior to study enrolment, tumour specimens from all patients were tested for the presence of BRAF V600 mutations by the cobas® 4800 BRAF V600 Mutation Test. Approximately 50% of the patients with melanoma screened for clinical studies had BRAF V600 mutation-positive tumours. The test was designed for, and has a high specificity for, detecting V600E. It is less sensitive for detecting other V600 mutations including the second most common mutation, V600K. Clinical data supporting use of Zelboraf in patients with BRAF mutations other than V600E are limited. Non-clinical and limited clinical data suggest that vemurafenib will not be effective in patients whose tumours are not positive for BRAF V600 mutations, and for this reason the efficacy and safety of Zelboraf have not been evaluated in this patient population.

Phase III Study in Treatment Naïve Patients

In an open-label, multicentre, international, randomized Phase III study in previously untreated patients with BRAF V600 mutation-positive unresectable or metastatic melanoma, 337 patients were randomized to treatment with Zelboraf (960 mg BID) and 338 patients were randomized to dacarbazine (1,000 mg/ every 3 weeks). Fifteen percent of patients (48) randomized to dacarbazine did not receive any study medication. This selective drop-out, however, was considered unlikely to impact the efficacy analyses. Thirty-two of the 48 patients had follow-up data for survival and subsequent anti-cancer therapy was reported for 9 patients (18.8%). Consistent with the intent-to-treat (ITT) principle, all available data for such patients were included in the ITT analyses, including any data reported after use of subsequent anti-cancer therapies. In addition, assessment of patients who received at least one course of study medication indicated that the measured baseline characteristics remained well balanced between the two study arms.

Treatment was continued until time of disease progression, unacceptable toxicity and/or consent withdrawal. The co-primary efficacy endpoints of the study were overall survival (OS) and progression-free survival (PFS). Key secondary efficacy endpoints included confirmed best overall response rate (BORR) and response duration.

A statistically significant and clinically meaningful improvement in OS was found for patients treated with Zelboraf with a hazard ratio of 0.37 [95% confidence interval (CI): 0.26, 0.55] which represents a 63% decrease in the hazard of death with Zelboraf compared to dacarbazine. Progression-free survival by investigator assessment was longer with Zelboraf compared to dacarbazine with a hazard ratio for PFS of 0.26 (95% CI: 0.20, 0.33) which represents a 74% decrease in the hazard of progression or death for Zelboraf compared to dacarbazine. The benefit in OS was maintained at the 90-day safety update analysis.

The secondary endpoint BORR (complete response plus partial response) as assessed by the investigator, was significantly improved (probability <0.0001) in the Zelboraf arm (48.4%) (95% CI: 41.6%, 55.2%) compared to the dacarbazine arm (5.5%) (95% CI: 2.8%, 9.3%). Among the 106 Zelboraf patients with a confirmed response, the median time to response was 1.4 months (range: 1.0 to 5.5). Among the 12 dacarbazine patients with a confirmed response, the median time to response was 2.7 months (range: 1.6 to 5.8).

Quality of life was assessed using the Functional Assessment of Cancer Therapy-Melanoma v.4 (FACT-M) questionnaire. Analyses of FACT-M and its subscales suggested that there was no difference in quality of life measured over time on study treatment in patients treated with Zelboraf compared with patients treated with dacarbazine. The proportion of patients with improvement in the physician's assessment of performance status was higher in the patients treated with Zelboraf (63.4%) (95% CI: 57%, 69%) than in the patients treated with dacarbazine (20.2%) (95% CI: 15%, 26%).

A total of 19 patients out of 220 whose tumours were retrospectively analysed by sequencing were reported to have BRAF V600K mutation-positive melanoma in this Phase III study. Although limited by the low number of patients, compared to V600E patients, efficacy analyses suggested a benefit of Zelboraf in patients with the V600K mutation. Four out of 10 V600K patients treated with Zelboraf responded to treatment compared to zero out of 9 patients in the dacarbazine arm. Furthermore, the treatment benefit of Zelboraf on OS and PFS in the V600K subpopulation was observed by the hazard ratios of 0.27 (95% CI: 0.05, 1.51) and 0.09 (95% CI: 0.02, 0.45), respectively.

Phase II Study in Patients who Failed at least One Prior Systemic Therapy

A Phase II single-arm, multicentre, multinational study was conducted in 132 metastatic melanoma patients with BRAF V600 mutation-positive tumours. More patients received prior therapy with IL-2 (39%) than with dacarbazine treatment (23%) in this study. The median duration of follow-up was 6.9 months (range 0.6 to 11.3).

The endpoint of confirmed best overall response rate (BORR) as assessed by an independent review committee was 52% (95% CI: 43%, 61%). Patients with prior IL-2 or dacarbazine therapy had a BORR of 48% (95% CI: 34%, 62%) or 60% (95% CI: 41%, 77%), respectively. The median time to response was 1.4 months with 75% of responses occurring by month 1.6 of treatment. The median duration of response was 6.5 months.

Nine of the 132 patients had BRAF V600K mutations confirmed by Sanger sequencing. Amongst these patients, 3 had a partial response, 3 had stable disease, 2 had progressive disease and 1 was not evaluable.

The Product Monograph clearly states that the effectiveness of Zelboraf in patients with prior therapy, including dacarbazine and IL-2 treatments, is based on the objective response rate data from a single-arm Phase II study. In the absence of additional effective therapies and unprecedented response rates of significant duration in these patients, Health Canada granted market authorization appropriate for patients with BRAF V600 mutations who failed prior therapies based on this single-arm study.

3.3.4 Clinical Safety

The adverse drug reactions were reported from the two clinical studies described in section 3.3.3 Clinical Efficacy. The median duration of treatment in the Phase III and Phase II studies were 4.2 months and 5.7 months, respectively.

The most common adverse events reported with the use of Zelboraf were arthralgia; rash; alopecia; fatigue; photosensitivity reaction; nausea; pruritus; and skin papilloma. Other very common events (>10%) included: sunburn; depression; decreased appetite; headaches; fever; arthritis; back pain; hand-foot syndrome; alopecia; and peripheral oedema. Common events (>1% and <10%) included: dehydration; loss of consciousness/syncope; facial paralysis; joint swelling; muscle weakness; and skin lesions. Less common but serious events included QTc prolongation, ophthalmologic reactions (including uveitis, iritis and retinal vein occlusion); serious hypersensitivity reactions (including anaphylactic shock, Stevens-Johnson syndrome, toxic epidermal necrolysis and hypotension); and grade 3 and grade 4 liver enzyme abnormalities.

Females experienced approximately twice as many clinically significant events (either > Grade 3 or serious adverse events) of arthralgia, photosensitivity reactions and rash compared to males in this study. In the clinical studies, elderly patients (>65 years of age) experienced more adverse events including cutaneous squamous cell carcinoma (cuSCC); decreased appetite; and cardiac disorders. Tumour lysis syndrome was also observed in the clinical studies and has been included in the Warnings and Precautions section of the Product Monograph.

Second malignancies are of significant concern with Zelboraf. While Zelboraf is effective in treating metastatic melanoma in the BRAF V600-mutation positive population, there is a paradoxical increase in other malignancies, primarily the less invasive cuSCC but also in potentially more aggressive melanomas and non-cutaneous SCCs. Cutaneous SCCs (24% across all studies) and new primary melanomas (2% in the Phase III study) have been reported in the submitted clinical studies, and two cases of non-cutaneous SCCs of the head and neck (tonsils and tongue) have recently been reported in ongoing studies.

Zelboraf at the therapeutic dose of 960 mg BID was associated with QTc prolongation, decreased heart rate, and increased blood pressure in the Phase II and Phase III studies. Exposure-dependent QTc prolongation was observed. The magnitude of QTc prolongation ranged from a mean of 11-15 msec in the Phase II study and reached a maximum effect of 22.1 msec (90% CI: 14.2, 30.0) by Cycle 6 in the Phase III study. Elevations in blood pressure have been reported in association with Zelboraf. In the Phase III study, the mean change from baseline ranged from 4-10 mm Hg for systolic blood pressure and 0-8 mm Hg for diastolic blood pressure over the course of treatment.

Despite important safety concerns for Zelboraf and the limitations of the available safety data (limited patient population over a relatively short duration), the overall safety profile of Zelboraf is acceptable and manageable. The Health Canada-approved labelling will help ensure proper monitoring and mitigating, where possible, of some serious adverse events. A Serious Warnings and Precaution box has been added to the Product Monograph listing QTc prolongation and second malignancies as clinically significant adverse events. A statement that Zelboraf has not been studied in patients with severe hepatic impairment has also been included in the Serious Warnings and Precautions box. In the Warnings and Precautions section, it is stated that Zelboraf should not be used in patients with wild-type BRAF tumours or in patients where the BRAF mutational status is not known; and that the clinical data supporting use of Zelboraf in patients with BRAF mutations other than V600E are limited.

3.4 Benefit/Risk Assessment and Recommendation

3.4.1 Benefit/Risk Assessment

Priority Review status was granted for the evaluation of Zelboraf. The new drug submission reported substantial evidence of clinical effectiveness of Zelboraf over existing therapies for a serious, life-threatening disease that is not adequately managed by a drug marketed in Canada.

Zelboraf has shown overall survival (OS) and progression-free survival (PFS) benefits in untreated patients with melanoma harbouring BRAF V600 mutations compared to dacarbazine, the current standard of care, in a randomized phase III study. In addition, Zelboraf has shown significant activity (>50% response rates) in a single-arm Phase II study in patients with BRAF V600-mutation positive melanoma who have received prior therapies, predominantly temazolamide, dacarbazine or IL-2. However, clinical data supporting the effectiveness of Zelboraf in patients with BRAF mutations other than V600E are limited. Furthermore, non-clinical and limited clinical data suggest that Zelboraf will not be effective in the treatment of patients with melanoma that does not harbour a BRAF V600 mutation and therefore it is important that the mutation status of the tumor is identified with a validated test prior to treatment with Zelboraf. Finally, while treatment with Zelboraf provides an important clinical benefit for patients with metastatic melanoma expressing BRAF V600 mutations, many patients will develop resistance within the first year of treatment.

Risks identified with the use of Zelboraf include QTc prolongation, phototoxicity, liver function abnormalities, cuSCC growth, other new primary malignancies, ophthalmological changes, rash, joint pain and other common adverse events. These risks, including second malignancies (largely cuSCCs), QT prolongation, photosensitivity, and hepatotoxicity, are considered manageable.

Overall, the benefit-risk assessment for this patient population is acceptable, particularly for previously untreated patients with BRAF V600 mutation-positive melanoma tumours where Zelboraf provides an important overall survival advantage compared to dacarbazine. The approved labelling accurately reflects the safety and efficacy of Zelboraf. Proper monitoring of adverse events and dosing considerations together with the approved labelling will help maintain a favourable benefit-risk profile for this drug in the post-market setting.

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 Zelboraf is favourable as a monotherapy for the treatment of patients with BRAF V600 mutation-positive unresectable or metastatic melanoma. A validated test is required to identify BRAF V600 mutation status. 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.

4 Submission Milestones

Submission Milestones: ZelborafTM

Submission MilestoneDate
Pre-submission meeting:2011-05-19
Request for priority status
Filed:2011-06-14
Approval issued by the Director of the Bureau of Metabolism, Oncology, and Reproductive Sciences:2011-07-08
Submission Filed:2011-07-18
Screening
Screening Acceptance Letter issued:2011-08-19
Review
Quality Evaluation complete:2011-02-13
Clinical Evaluation complete:2011-02-15
Labelling Review complete:2012-02-14
Notice of Compliance issued by Director General:2011-02-15