Summary Basis of Decision for Jakavi

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
Jakavi

Ruxolitinib, as ruxolitinib phosphate, 5 mg, 15 mg, and 20 mg, Tablet, Oral

Novartis Pharmaceuticals Canada Inc.

Submission control no: 151723

Date issued: 2012-11-06

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:

Jakavi

Manufacturer/sponsor:

Novartis Pharmaceuticals Canada Inc.

Medicinal ingredient:

Ruxolitinib, as ruxolitinib phosphate

International non-proprietary Name:

Ruxolitinib

Strength:

5 mg, 15 mg, and 20 mg

Dosage form:

Route of administration:

Oral

Drug identification number(DIN):

  • 02388006 - 5 mg
  • 02388014 - 15 mg
  • 02388022 - 20 mg

Therapeutic Classification:

Antineoplastic agent

Non-medicinal ingredients:

Microcrystalline cellulose, hydroxypropylcellulose, lactose monohydrate, magnesium stearate, colloidal silicon dioxide, sodium starch glycolate (Type A), povidone.

Submission type and control no:

New Drug Submission, Control Number: 151723

Date of Submission:

2011-11-29

Date of authorization:

2012-06-19
2 Notice of decision

On June 19, 2012, Health Canada issued a Notice of Compliance to Novartis Pharmaceuticals Canada Inc., for the drug product Jakavi.

Jakavi contains the medicinal ingredient ruxolitinib, an antineoplastic agent that functions as a Janus Associated Kinase 1 (JAK1) and JAK2 inhibitor. As a JAK inhibitor, ruxolitinib is the first drug in this class of agents to receive marketing authorization in Canada.

Jakavi is indicated for the treatment of splenomegaly and/or its associated symptoms in adult patients with primary myelofibrosis (also known as chronic idiopathic myelofibrosis), post-polycythaemia vera myelofibrosis or post-essential thrombocythaemia myelofibrosis.

Myelofibrosis is a disorder of the bone marrow in which the marrow is replaced by fibrous tissue. The abnormal marrow can no longer produce enough blood cells resulting in anaemia, bleeding problems, and a possible higher risk of infection. As a result, extramedullary haematopoiesis (the formation of blood cellular components outside of the bone) may occur which usually manifests as splenomegaly (an enlarged spleen). This disorder is associated with activation of the JAK2/STAT signaling pathway. Jakavi helps reduce the spleen size in patients with different forms of myelofibrosis by blocking the function of proteins called JAK1 and JAK2, thus relieving the symptoms associated with splenomegaly.

The market authorization was based on quality, non-clinical, and clinical information submitted. The clinical efficacy and safety of Jakavi were demonstrated in two pivotal Phase III studies (COMFORT-I and COMFORT-II)in patients with myelofibrosis (primary myelofibrosis, post-polycythaemia vera myelofibrosis or post-essential thrombocythaemia myelofibrosis) and one supporting Phase II study. The primary efficacy endpoint in the pivotal studies was the proportion of patients achieving a ≥35% reduction in spleen volume from baseline to Week 24 (COMFORT-I) or to Week 48 (COMFORT-II). A significantly larger proportion of patients in the Jakavi group (41.9% and 28.5%, respectively) achieved a ≥35% reduction in spleen volume from baseline compared with 0.7% in the placebo group at Week 24 in COMFORT-I, and compared with none (0%) in the best available therapy group at Week 48 in COMFORT-II. The Jakavi groups achieved the reductions in spleen volume regardless of the presence or absence of the JAK2V617F mutation or the disease subtype (primary myelofibrosis, post-polycythemia vera myelofibrosis, post-essential thrombocythemia myelofibrosis).

Jakavi (5 mg, 15 mg, and 20 mg ruxolitinib) is presented as tablets. The recommended starting dose is based on platelet count. A complete blood count and platelet count must be performed before initiating therapy, every 2 to 4 weeks until doses are stabilized, and then clinically as indicated. The recommended starting dose of Jakavi is 15 mg given orally twice daily for patients with a platelet count between 100,000 and 200,000/mm³ and 20 mg twice daily for patients with a platelet count >200,000/mm³. Prior to initiating treatment with Jakavi, the absolute neutrophil count of patients should be >1,000/µL. There is limited information to recommend a starting dose for patients with platelet counts between 50,000/mm³ and 100,000/mm³. The maximum recommended starting dose in these patients is 5 mg twice daily and the patients should be titrated cautiously. Dosing guidelines are available in the Product Monograph.

Jakavi is contraindicated for patients with a known hypersensitivity to ruxolitinib or to any ingredient in the formulation of Jakavi or component of the container. Jakavi 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 Jakavi are described in the Product Monograph.

Priority Review Status was granted for the evaluation of Jakavi as it appeared to provide a significant increase in efficacy and a significant decrease in risk such that the overall benefit/risk profile is improved 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 Jakavi is favourable for the treatment of patients with splenomegaly and/or its associated symptoms in adult patients with primary myelofibrosis (also known as chronic idiopathic myelofibrosis), post-polycythaemia vera myelofibrosis or post-essential thrombocythaemia myelofibrosis.

3 Scientific and Regulatory Basis for Decision

3.1 Quality Basis for Decision

3.1.1 Drug Substance (Medicinal Ingredient)

General Information

The medicinal ingredient of Jakavi, ruxolitinib (as ruxolitinib phosphate), is an antineoplastic agent that functions as a Janus Kinase 1 (JAK1) and JAK2 inhibitor. As a kinase inhibitor, ruxolitinib helps reduce the spleen size in patients with different forms of myelofibrosis by blocking the function of proteins called JAK1 and JAK2, thus relieving the symptoms associated with splenomegaly.

Manufacturing Process and Process Controls

Ruxolitinib phosphate 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.
Characterization

Detailed characterization studies were performed to provide assurance that ruxolitinib phosphate consistently exhibits the desired characteristic structure. Physical and chemical properties have been described and are considered satisfactory.

Impurities and degradation products arising from manufacturing and/or storage were reported and characterized. 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 ruxolitinib phosphate.

The drug substance packaging is considered acceptable.

Stability

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

3.1.2 Drug Product

Description and Composition

Jakavi (ruxolitinib tablets) is available in three strengths. Each tablet contains 5 mg, 15 mg, or 20 mg ruxolitinib free base (as ruxolitinib phosphate).

Jakavi 5 mg tablets are round, white to almost-white tablets with "NVR" debossed on one side and "L5" debossed on the other side.

Jakavi 15 mg tablets are ovaloid curved, white to almost-white tablets with "NVR" debossed on one side and "L15" debossed on the other side.

Jakavi 20 mg tablets are elongated curved, white to almost-white tablets with "NVR" debossed on one side and "L20" debossed on the other side.

The non-medicinal ingredients include hydroxypropylcellulose, lactose monohydrate, magnesium stearate, microcrystalline cellulose, colloidal silicon dioxide, sodium starch glycolate (Type A), and povidone.

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

Jakavi 5 mg, 15 mg, and 20 mg tablets are supplied in highly dense polyethylene (HDPE) bottles with child resistant closures (60 tablets) and in blister packaging (4 x 14 tablets).

Pharmaceutical Development

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

Manufacturing Process and Process Controls

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

Control of Drug Product

Jakavi is tested to verify that its identity, appearance, content uniformity, assay, dissolution, particle size, mean mass, moisture content, and levels of degradation products 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.

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

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

Stability

The real-time, long-term, accelerated stability data proposed the shelf-life of 24 months when the tablets are packaged in HDPE bottles and 12 months when packaged in blister packaging when both are stored between 15-30°C.

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 facilities and equipment that are involved in the production of Jakavi are considered suitable for the activities and products manufactured.

All sites are compliant with Good Manufacturing Practices.

3.1.4 Adventitious Agents Safety Evaluation

The excipient, lactose monohydrate, is sourced from bovine milk that is fit for human consumption and is unlikely to present any risk of transmissible spongiform encephalopathy (TSE) contamination.

Magnesium stearate is of vegetable origin.

3.1.5 Conclusion

The Chemistry and Manufacturing information submitted for Jakavi 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

Myelofibrosis is a myeloproliferative neoplasm (MPN) known to be associated with dysregulated JAK1 and JAK2 signalling. JAK signalling involves recruitment and activation of STATs (signal transducers and activators of transcription). The dysregulation of the JAK-STAT pathway has been associated with several cancers and increased proliferation and survival of malignant cells.

In vitro data demonstrated that ruxolitinib is an inhibitor of JAK1 [with a half-maximal inhibition concentration (IC50) of 3.3 nM] and JAK2 (IC50 2.8 nM). Ruxolitinib inhibited cell proliferation (IC50 141 nM) and phosphorylation of the transcription factor STAT3 (IC50 125 nM), in a cytokine dependent, JAK wild-type INA-6 multiple myeloma cell line. Inhibition of JAK-STAT signalling and cell proliferation was also observed in Ba/F3 cells rendered cytokine-independent by expressing the JAK2V617F mutated protein (IC50 80-320 nM).

In vivo, ruxolitinib was examined in models relevant to MPN. Treatment of mice with ruxolitinib resulted in a dose-dependent suppression of phosphorylated STAT3 and tumour growth in a cytokine-dependent, JAK wild type INA-6 multiple myeloma xenograft model. In a JAK2V617F driven Ba/F3-EpoR xenograft mouse model, ruxolitinib treatment suppressed splenomegaly, decreased mutant allele burden [33% decrease, probability (p) <0.01], decreased circulating inflammatory cytokines (tumour necrosis factor-alpha andinterleukin-6) and normalized aberrant JAK/STAT signalling as indicated by phosphorylated STAT3 levels from splenic lysates. Three weeks of ruxolitinib treatment led to significantly improved survival (>90%) versus vehicle-treated animals (<10%) in the Ba/F3-EpoR-JAK2V617F xenograft model.

Ruxolitinib was evaluated in a core battery of central nervous system (CNS), respiratory, and cardiovascular safety pharmacology studies including an in vitro human Ether-à-go-go Related Gene (hERG) channel assay. Ruxolitinib did not demonstrate strong inhibition of hERG (IC50 132 μM) in human cells. In vivo, the safety pharmacology studies of ruxolitinib in rats and dogs yielded adverse effects in respiratory, CNS, and cardiovascular function at exposure multiples greater than those observed in human studies. These effects included lower respiratory rates, higher tidal volumes, lower minute volumes, lower body temperature, lower activity, lower systolic and diastolic pressure, increased heart rate, and decreased mean and pulse arterial pressure. However, these findings have not been observed in the repeat-dose toxicity studies or recapitulated in the clinical studies.

3.2.2 Pharmacokinetics

Absorption

Ruxolitinib was rapidly absorbed in mice, rats, rabbits, and dogs.

Ruxolitinib is extensively metabolized, highly soluble (pH 1.0-8.0), and permeable with a range of bioavailability (22-105%) depending on the species.

Distribution

Ruxolitinib was rapidly and widely distributed in rats with maximal serum levels detected 0.5-2.0 hours post-oral dosing. The highest concentrations were found in the gastrointestinal system, urinary bladder, liver, renal cortex, aorta, adrenal glands, skin, and kidneys. Ruxolitinib was highly bound to blood plasma of the tested species.

Ruxolitinib and its metabolites crossed the blood brain barrier (<10% of plasma concentrations) and placental barrier of rats. Ruxolitinib and its metabolites transferred substantially into the milk of lactating rats.

Metabolism

The metabolic profile of ruxolitinib was examined in vitro by incubating ruxolitinib with human liver microsomes and hepatocytes. The prominent in vitro metabolic pathway in human liver microsomes was oxidation. The major enzyme responsible for the metabolism of ruxolitinib was cytochrome P450 (CYP) 3A4.

Ruxolitinib and its M18 metabolite were found not to inhibit CYP1A2, CYP2C9, CYP2C19, CYP2D6, CYP3A4, CYP2B6 and CYP2C8. Ruxolitinib did not induce CYP3A4, CYP1A2 and CYP2B6 at relevant clinical concentrations. Ruxolitinib was not a substrate or an inhibitor for P-glycoprotein nor an inhibitor of the organic anion transporters (OAT) OATP1B1 and OATP1B3. However, ruxolitinib may inhibit OAT1 and OAT3, and the organic cation transporter (OCT) OCT1 and OCT2. The metabolite M18 was found not to be an inhibitor for any of the transporters investigated.

Excretion

The excretion profiles were similar in male and female rats. Following a single oral dose of radiolabelled ruxolitinib, 45.4-52%, 36.9-39.5%, and 11.9-20.1% of the dose was recovered in the urine, bile, and faeces, respectively. The excretion was rapid with approximately 100% of the dose recovered by 24 hours post-dose.

In male and female dogs, excretion in the urine accounted for 34.4% and 36.3% of the dose, respectively, and excretion in the faeces accounted for 54.8% and 57.9% of the dose, respectively. Excretion was essentially complete with a total overall recovery in male and female dogs of 94.0% and 96.1% of the dose, respectively, by 96 hours post-dose.

3.2.3 Toxicology

Single-Dose Toxicity

Ruxolitinib was well-tolerated following single oral doses of up to 100 mg/kg in rats and 40 mg/kg in dogs. At these dose levels, mild lethargy and emesis were observed in rats and dogs, respectively.

Repeat-Dose Toxicity

The repeat, oral dose studies included studies of up to 4-weeks in mice, 6-months in rats, and 12-months in dogs. The target organs associated with the pharmacological action of ruxolitinib were bone marrow, peripheral blood, and lymphoid tissues. Specific findings included decreases in lymphocytes, eosinophils, reticulocytes, red blood cells, haemoglobin, and haematocrit as well as hypocellularity of the bone marrow and lymphoid organs (spleen, thymus, lymph nodes). Dogs (6- and 12-month study) developed bacterial, parasitic and viral infections that were generally associated with immunosuppression. Other findings included gastrointestinal inflammation [4-week dog study; at an exposure approximately 5-fold the exposure at the maximum recommended human dose (MRHD) based on area-under-the-curve (AUC)], prostatic atrophy (6-month dog study; at an exposure approximately 1.9-fold the exposure at the MRHD based on AUC), heart fibrosis (13-week female rat study; at an exposure approximately 9.5-fold the exposure at the MRHD based on AUC), adrenal cortical atrophy (6 month rat study; at an exposure approximately 0.14-fold the exposure at the MRHD based on AUC), hyperplasia of non-glandular stomach (4 week mouse study; at an exposure approximately 6.5-fold the exposure at the MRHD based on AUC), increases of alkaline phosphatase and gamma-glutamyl transpeptidase (13-week female rat study; at an exposure approximately 9.6-fold the exposure at the MRHD based on AUC), and decreases in phosphorous and calcium levels (dogs ≥5 mg/kg/day; at an exposure approximately 1.6-fold the exposure at the MRHD based on AUC).

Genotoxicity

Ruxolitinib did not test positive for mutagenicity in a bacterial mutagenicity assay (Ames test) or clastogenicity in an in vitro chromosomal aberration assay or in the in vivo rat bone marrow micronucleus assay. Therefore, ruxolitinib is considered non-genotoxic.

Carcinogenicity

In a 6-month carcinogenicity study, no significant increase in neoplastic lesions was observed in mice at systemic exposures (AUC) that exceeded (8-fold) those observed in clinical studies. Non-neoplastic intranasal inflammation was observed in the treated mouse model at an exposure approximately 8-fold the exposure at the MRHD based on AUC. Ruxolitinib was considered non-carcinogenic in the mouse model.

Reproductive and Developmental Toxicity

Ruxolitinib was not teratogenic but was associated with maternal toxicity, embryo lethality (increases in post-implantation loss resulting in decreased litter sizes) and foetal toxicity (decreased foetus weights) in rats and rabbits. No effects were noted on reproductive performance or fertility. In a pre- and post-natal development study, there were no adverse findings for fertility indices and maternal and embryo-foetal survival, growth, and developmental parameters. All of these observations occurred at exposures significantly less than those observed in the clinical populations (at an exposure approximately 0.07 to 0.34-fold the MRHD based on AUC).

Phototoxicity

Ruxolitinib absorbs light in the range of 290 to 700 nm, with a peak at 310 nm. In studies performed on guinea pigs, ruxolitinib did not show any photoallergic or phototoxic potential when applied either topically or dermally at concentrations ≤1.5%. Repeated daily topical administration of ruxolitinib with or without simulated sunlight in hairless mice for a period of 13 weeks did not result in adverse findings. However, no phototoxicity, photoallergy or irritancy studies have been performed via the oral route of administration.

3.2.4 Summary and Conclusion

The non-clinical studies for this drug submission are considered acceptable. The non-clinical pharmacology and toxicology program for Jakavi (ruxolitinib) demonstrated that the compound is relatively safe in reference to the approved indication. Adequate statements are in place in the Product Monograph to address the identified safety concerns. The non-clinical data submitted in this drug submission does not raise substantial issues that would preclude the market authorization of Jakavifor the treatment of splenomegaly and/or its associated symptoms in adult patients with primary myelofibrosis (also known as chronic idiopathic myelofibrosis), post-polycythaemia vera myelofibrosis or post-essential thrombocythaemia myelofibrosis.

3.3 Clinical basis for decision

The Canadian regulatory decision on the clinical pharmacology, and clinical efficacy and safety studies was based on a critical assessment of the Canadian application/submission. Under the provisions of the Draft Guidance Document: The Use of Foreign Reviews by Health Canada, Method 3 was used whereby, during the review of the current submission, the foreign reviews completed by the United States Food and Drug Administration (FDA) and the European Union's centralized procedure European Medicines Agency (EMA) were used as an added reference in the assessment of Jakavi.

3.3.1 Pharmacodynamics

The STAT3 transcription factor is directly phosphorylated by JAKs in response to cytokine stimulation and therefore can be used as a pharmacodynamics (PD) marker for JAK inhibition. Following fasting, oral, multiple-dose administration, ruxolitinib demonstrated dose- and time-dependent inhibition of cytokine-induced STAT3 phosphorylated (p) with maximal inhibition occurring 1 to 2 hours after administration for all doses, coincident with the maximum plasma concentration (Cmax). Maximal inhibition of STAT3p ranged from approximately 40% at the lowest dose (5 mg) to greater than 90% inhibition at the highest dose (200 mg). Levels of STAT3p returned to control levels by 24 hours in all treatments examined. The cytokine-induced STAT3p was inhibited by ruxolitinib with an IC50 of 224 nM.

In a double-blind, placebo-controlled, crossover electrocardiogram study in 49 healthy volunteers, there was no indication of a QTc prolonging effect of ruxolitinib at single doses of 25 mg and 200 mg.

3.3.2 Pharmacokinetics

Absorption

Ruxolitinib is a Class 1 molecule under the Biopharmaceutical Classification System, with high permeability, high solubility and rapid dissolution characteristics. The drug was absorbed rapidly with a peak plasma concentration approximately 1 hour after oral administration. The plasma concentrations declined in a monophasic or biphasic fashion. The absorption of ruxolitinib was 95% or greater. The mean Cmax and total exposure (AUC) were dose-proportional and linear pharmacokinetics (PK) were demonstrated over a single dose range of 5 mg to 200 mg. It was demonstrated that ruxolitinib does not accumulate and that it has time-independent PK.

The food effect was evaluated in healthy volunteers. The PK results demonstrated that there should be no clinically significant differences when ruxolitinib is administered under fed or fasted states in patients.

Distribution

Ruxolitinib was highly bound to human serum albumin with an unbound fraction of 3.3%. The volume of distribution at steady-state was 53-65 L in myelofibrosis patients.

Metabolism

CYP3A4 is the major enzyme responsible for the metabolism of ruxolitinib. Ruxolitinib is metabolized mostly by oxidation and the two major metabolites were identified as M18 and M16. The metabolites do retain pharmacological activity. The parent compound was the predominant entity in circulation representing approximately 60% of the drug-related material in circulation.

Excretion

The recovered parent compound was 74% from urine and 22% from faeces, thus urine is the main route of excretion. Less than 1% was unchanged drug, in concordance to the observation that ruxolitinib is eliminated almost completely by oxidative metabolism with a terminal half-life of approximately 3 hours.

Drug-Drug Interaction Studies

The PK parameters (Cmax, AUC, and clearance) were strongly influenced when ruxolitinib was administered concomitantly with ketoconazole, a strong CYP3A4 inhibitor, therefore dose adjustments were recommended. There were no significant PK changes observed when ruxolitinib was co-administered with erythromycin, a moderate CYP3A4 inhibitor, therefore dose adjustments were not recommended with the co-administration of moderate to mild CYP3A4 inhibitors.

The PK effects of the co-administration of ruxolitinib with rifampicin, a strong CYP3A4 inducer, were minor and the PD parameters did not change. Therefore, no dose alterations were recommended when strong CYP3A4 inducers are co-administered.

Special Populations

A hepatic impairment study showed that a single 25 mg dose of ruxolitinib administered in patients with mild, moderate, and severe hepatic impairment, increased the AUC 1.87, 1.28, and 1.65 times, respectively. The terminal elimination half-life (t½) was 1.6, 1.5, and 1.8-fold prolonged for the mild, moderate, and severe hepatically impaired patients, respectively. The clearance (CL/F) was approximately 2-fold decreased for the three groups. It was recommended that patients with mild, moderate, or severe hepatic impairment receive a lower dose of ruxolitinib based on their platelet count.

A renal impairment study showed no major differences in the PK or PD of ruxolitinib when administered in patients with varying degree of renal impairment (mild, moderate, or severe). However, the 8 active metabolites contributed to PK parameter changes evaluated in patients with renal impairment. These PK changes translated into an increased PD combined effect (parent + metabolites) of 117%, 123%, 134%, 153%, 212% and 192% for normal, mild, moderate, severe, end-stage renal disease (ESRD) with dosing after dialysis, and ESRD with dosing before dialysis, respectively. Based on the observed changes of the PK and PD due to ruxolitinib's active metabolites, the final recommendations propose dose reductions in patients with moderate and severe renal impairment. Even though there was a limited data of ESRD patients on dialysis, the results showed significant PK and PD changes, thus the final recommendations were to alter the administration schedule of ruxolitinib.

3.3.3 Clinical Efficacy

The efficacy and safety findings were derived from two pivotal Phase III studies and one supporting Phase II study. COMFORT-I [number of patients (n) = 309] was a randomized, double-blind, placebo-controlled Phase III study in patients with Primary Myelofibrosis (MF), Post-Polycythaemia Vera Myelofibrosis (PPV-MF) or Post-Essential Thrombocythaemia-Myelofibrosis (PET-MF). COMFORT-II (n = 219) was a randomized, open-label, efficacy and safety Phase III study of JAKAVI tablets compared to best available therapy (BAT) in patients with PMF, PPV-MF or PET-MF.

The starting dose of Jakavi was based on platelet count. Patients with a platelet count between 100,000 and 200,000/mm³ were started on Jakavi 15 mg twice daily (BID) and patients with a platelet count >200,000/mm³ were started on Jakavi 20 mg BID. A standardized dosing paradigm was used to determine dose adjustments for safety and efficacy so that each patient was titrated to their most appropriate dose. Doses were not to exceed 25 mg BID. The recommended dose titration has been fully justified by the results of the pivotal studies and by one supporting Phase II study.

The comparator groups were relevant. In COMFORT-I, a placebo was used as the comparator while in COMFORT-II, BAT was used as the comparator. Although, none of the drugs used in the BAT group were approved for use in the treatment of MF in Canada, the results of COMFORT-II are supportive of the other pivotal study which compared Jakavi to placebo.

Primary Endpoints

The primary efficacy endpoint in the pivotal studies was the proportion of patients achieving ≥35% reduction in spleen volume from baseline to Week 24 (COMFORT-I) or to Week 48 (COMFORT-II) as measured by Magnetic Resonance Imaging (MRI) or Computerized Axial Tomography (CAT).

Both pivotal studies met their primary endpoint. A statistically significant higher proportion of patients achieved ≥35% reduction in spleen volume with Jakavi compared to controls at Week 24 [COMFORT-I; 41.9% versus (vs.) 0.7%] or at Week 48 (COMFORT-II; 28.5% vs. 0%).

The response rates were lower in male patients compared to female patients, suggesting that the efficacy of ruxolitinib could be lower in male patients. However, a higher starting dose in male patients is not recommended because it may increase the risk of developing Jakavi-associated toxicities. Any potential difference in efficacy in male patients can be managed by the titrating approach while preserving these patients from excessive toxicity.

The response rates were smaller in patients with a starting dose of 15 mg compared to patients with a starting dose of 20 mg. However, the administration of a 20 mg starting dose in patients with a platelet count of 100,000/mm³ to 200,000/mm³ is not recommended since it may result in an unacceptable risk-benefit profile in these patients.

Secondary Endpoints

Secondary endpoints in COMFORT-I included the duration of maintenance of a ≥35% reduction from baseline in spleen volume, the proportion of patients who had ≥50% reduction in total symptom score from baseline to Week 24 as measured by the modified Myelofibrosis Symptom Assessment Form (MFSAF) v2.0 diary, the change in total symptom score from baseline to Week 24 as measured by the modified MFSAF v2.0 diary, and overall survival (OS).

In COMFORT-II, the secondary endpoints were the proportion of patients achieving a ≥35% reduction of spleen volume measured by MRI or CAT from baseline to Week 24 and the duration of maintenance of a ≥35% reduction from baseline in responding patients.

In COMFORT-I, a significantly larger proportion of patients in the Jakavi group achieved a ≥50% improvement from baseline in the Week 24 total symptom score compared with the placebo group [45.9% and 5.3%, respectively, probability (p) <0.0001 using the Chi-Squared test], as measured by the modified MFSAF v2.0 diary.

COMFORT-II also met its key secondary endpoint. A statistically significant higher proportion of patients treated with Jakavi achieved ≥35% reduction in spleen volume comparing baseline to Week 24 (32% vs. 0% in Jakavi group and BAT group respectively).

No statistical difference in OS between the groups was observed in either pivotal study. An additional OS analysis was performed in COMFORT-I, which included the data from an additional follow-up period. Although the results from the follow-up analysis showed an OS improvement in the Jakavi group, there was no adjustment for repeated testing and it was not pre-specified in the protocol. These results are considered as exploratory.

In COMFORT-II, the median duration of maintenance of spleen volume reduction in Jakavi-treated patients was 48 weeks. The median duration of response in COMFORT-I was not reached as the majority of patients were still responding at the data cutoff.

The median time to ≥35% reduction in spleen volume was 12.29 weeks in COMFORT-II, which may have been overestimated due to the lack of spleen assessment before 12 weeks.

The clinical studies contain no evidence that Jakavi can modify the course of myelofibrosis. The goal of therapy with Jakavi appears to be the control of some of the MF symptoms, namely splenomegaly.

There is no evidence demonstrating the clinical benefit of the Jakavi treatment in asymptomatic myelofibrosis patients (i.e., without splenomegaly or constitutional symptoms).

In conclusion, the data from the pivotal studies demonstrated that Jakavi can significantly reduce splenomegaly in patients with PMF, PPV-MF or PET-MF, including patients with massively enlarged spleens. Jakavi was able to halt the progression of splenomegaly in almost all patients. Patients also had significant clinical benefit in the reduction of important symptoms of MF as assessed by the modified MFSAF v2.0 diary. However, it is unclear whether the reduction of the symptoms of MF is due to a reduction of the splenomegaly or to a direct effect of Jakavi on those symptoms. Consequently, it is currently unclear whether MF patients (symptomatic or asymptomatic) without splenomegaly may benefit from the Jakavi therapy.

The proposed clinical population in the indication was too broad since it included all patients with MF regardless of the presence of splenomegaly, and the efficacy results did not support the sponsor's proposed indication. The targeted population should be defined by the symptoms to be treated, in occurrence splenomegaly and its associated symptoms. Therefore, the indication should specify that Jakavi is for the treatment of splenomegaly and/or its associated symptoms in patients with PMF, PPV-MF or PET-MF.

3.3.4 Clinical Safety

The safety profile of Jakavi in patients with myelofibrosis was derived from 589 patients treated in two pivotal Phase III studies and one Phase II supporting study.

In the two pivotal studies COMFORT-I and COMFORT-II, 301 patients had a median duration of exposure to Jakavi of 10.8 months (range 2 weeks to 19.5 months). The majority of patients (68.4%) were treated for at least 9 months. Of the 301 patients, 111 (36.9%) had a baseline platelet count between 100,000/mm³ and 200,000/mm³, and 190 (63.1%) had a baseline platelet count >200,000/mm³.

Study results suggested that individual dose titration within the 10 to 25 mg range offered the best balance of efficacy and safety for an individual. The dose that was ultimately tolerated appeared to be mainly influenced by the baseline platelet count. There is no experience with the administration of Jakavi to patients with platelet counts <100,000/mm³ at the start of treatment. Grade 3 and 4 thrombocytopenia can usually be avoided by dose interruption or reduction.

In the Jakavi group, the most common adverse drug reactions (ADRs) were related to myelosuppression, namely thrombocytopaenia and anaemia. This may be due to the disruption of the JAK/STAT pathway by Jakavi, which is an important signalling pathway in haematopoiesis. Anemia and thrombocytopaenia were dose-related effects.

The most frequently reported AE leading to dose interruption or discontinuation of study medication was thrombocytopaenia. The most frequent AE leading to dosage reduction was thrombocytopaenia/decreased platelet count. In both pivotal studies, dose reductions due to anaemia occurred in 5.3% of Jakavi-treated patients.

Other ADRs reported with a higher frequency in the Jakavi group compared to control groups (in both pivotal studies) were: neutropaenia; ecchymosis; pyrexia; headache; weight increased; haematoma; dizziness; urinary tract infection; hypercholesterolemia; herpes zoster; palpitations; bradycardia/sinus bradycardia; angina pectoris/angina unstable; and flatulence.

Haematoma and haemorrhage are adverse events of interest due to the mechanism of action of the drug. Although the proportion of patients with at least one bleeding event was higher in the subgroup of patients who received Jakavi and acetylsalicylic acid compared to patients who received acetylsalicylic acid in the control groups, there is insufficient evidence to suggest that concomitant use of aspirin increases the risk of bleeding events in Jakavi patients. In fact, data shows that there was a lower risk of bleeding events in patients who received Jakavi and acetylsalicylic acid compared to patients who received Jakavi and no anticoagulant (33.7% vs. 51.0%). Bleeding events accounted for nine deaths (1.1%) in the Jakavi group (subdural haematoma, retroperitoneal haemorrhage, 2 gastrointestinal haemorrhage, 2 cerebral haemorrhage, 3 other haemorrhage) and 2 deaths (0.7%) in the placebo group (gastrointestinal haemorrhage and subdural haematoma). Therefore, there is insufficient evidence to support a boxed warning or the addition into the Adverse Reactions section of information related to an association between fatal haemorrhage and the concomitant use of Jakavi and acetylsalicylic acid.

The proportion of patients who had a serious adverse event (SAE) was higher among patients in the placebo group (35.1%) compared with the Jakavi group (27.7%) in COMFORT-I. In COMFORT-II, SAEs were reported for a similar proportion of patients in the two groups: 30.1% and 28.8% of patients in the Jakavi group; and the BAT group, respectively.

Increased levels of alanine transaminase (ALT) and aspartate aminotransferase (AST) were more frequent in the Jakavi group compared to the control groups. Most cases of hepatotoxicity in the Jakavi group were of mild severity and appeared to be manageable. Severe drug-induced liver injury does not seem to be associated with Jakavi.

Treatment with Jakavi was associated with statistically significant PR interval prolongation and decrease heart rate. Cardiac disorders were observed at a higher frequency in the Jakavi group compared to the control groups (COMFORT-I; 18.9% vs. 13.9% while in COMFORT-II; 19.9% vs. 11%). Bradycardia/sinus bradycardia, palpitations, and angina pectoris/angina unstable, occurred at a higher rate during treatment with Jakavi in both pivotal studies. The association of cardiac failure/cardiac failure congestive, hypertension/hypertensive crisis, and myocardial infarction/acute myocardial infarction with Jakavi is less clear because they were observed at a higher rate in only one of the pivotal studies.

In contrast to COMFORT-I and the QT interval study in healthy subjects, a statistically significant increase from baseline in the QTcF interval (QT corrected using Fridericia's formula) was observed in COMFORT-II at Week 4 and Week 24, but not at Week 12 and Week 48. QTcF interval prolongation in the 450-480 ms range was observed in 7 patients (4.8%) in the Jakavi group and 0 patients in the BAT group, prolongation between 480-500 ms was seen in 1 Jakavi-treated patient (0.7%) and 2 BAT-treated patients (2.7%). However, the actual effect of Jakavi on QTc interval is currently unclear because the submitted studies were not designed to thoroughly assess the effect of Jakavi on QTc interval.

There is no experience with the administration of Jakavi to patients with absolute neutrophil count (ANC) ≤1,000/µL. Resolution of decreased levels of neutrophils occurred without dose adjustment in the majority of patients. Few patients had neutrophil counts that required dose interruption per the protocol (ANC <500/μL).

Overall, infection rates and infections associated with immunosuppressive drugs in particular were not more frequent in the Jakavi groups compared to the control groups.

In COMFORT-I, more patients moved from transfusion independence to dependence in the Jakavi group, suggesting that Jakavi increases the likelihood of transfusion requirement. This finding could not be confirmed in COMFORT-II due to the large number of patients with missing transfusion status.

Following interruption or discontinuation of Jakavi, symptoms of myelofibrosis may return over a period of approximately 1 week. It has not been established whether abrupt discontinuation of Jakavi contributed to these events. This is appropriately captured in the Product Monograph.

Animal studies have shown that ruxolitinib is embryotoxic and foetotoxic (in rats and rabbits). The potential risk of teratogenicity for humans is unknown. The exposure margins in the non-clinical studies compared to the highest clinical dose were low and the results were therefore of limited relevance for humans. In addition, the presence of ruxolitinib in human semen cannot be ruled out. The level of ruxolitinib in semen that would present no increased risk to the embryos is currently unknown. Appropriate warnings and precautionary measures are in place in the Product Monograph to limit the exposure of the embryos to ruxolitinib.

3.4 Benefit/Risk Assessment and Recommendation

3.4.1 Benefit/Risk Assessment

Priority Review Status was granted for the evaluation of Jakavi as it appeared to provide a significant increase in efficacy and a significant decrease in risk such that the overall benefit/risk profile is improved over existing therapies for a serious, life-threatening disease that is not adequately managed by a drug marketed in Canada.

Myelofibrosis (MF) is a disorder of the bone marrow, in which the marrow is replaced by fibrous tissue. Consequently, the bone marrow is not able to make enough blood cells. Anaemia, bleeding problems, and a higher risk of infections may occur. As a result, extramedullary haematopoiesis may occur which is usually manifested as splenomegaly. Clinically, patients suffer from the consequences of massive splenomegaly including abdominal pain or discomfort and pain under the left costal margin; risk of vascular events (including thrombosis and haemorrhage); severe constitutional symptoms (fevers, night sweats, weight loss); a hypermetabolic state; cachexia and premature death. Some of these symptoms are directly or indirectly related to extramedullary haematopoiesis and others to proinflammatory cytokine excess. Causes of death for patients with MF include leukemic transformation, infections, bleeding, thrombosis, heart failure, liver failure, solid tumours, respiratory failure, and portal hypertension.

The efficacy findings were derived from two pivotal Phase III studies. The key efficacy findings were statistically and clinically significant. The data from the pivotal studies demonstrate that Jakavi can significantly reduce splenomegaly in patients with PMF, PPV-MF or PET-MF, including patients with massively enlarged spleens. Jakavi was able to halt the progression of splenomegaly in almost all of the patients. Significant clinical benefit was also derived from the reduction of important symptoms of MF as assessed by the modified MFSAF v2.0 diary. However, it is unclear whether the reduction of the symptoms of MF is due to a reduction of the splenomegaly or to a direct effect of Jakavi on those symptoms. Consequently, the benefit of the Jakavi treatment in MF patients without splenomegaly (symptomatic or asymptomatic) has not been demonstrated.

Subgroup analyses showed a consistent favourable effect of Jakavi on the reduction of spleen volume for most subgroups. However, the response rates of spleen volume reduction were lower in male patients compared to female patients. The response rates were also smaller in patients with a starting dose of 15 mg compared to subjects with a starting dose of 20 mg.

As a JAK inhibitor, Jakavi (ruxolitinib) is the first drug in this class of agents to receive market authorization in Canada. The clinical risk associated with this class of drug is currently derived from a limited set of clinical studies. The safety of Jakavi in the paediatric population has not been established. There is no experience with the administration of Jakavi to patients with ANC <1,000/µL and platelet count <100,000/mm³ at the start of treatment. To mitigate the risk of developing infections, the Product Monograph specifies that the Jakavi treatment should not be initiated in patients with an ANC ≤1,000/µL. The Product Monograph appropriately recommends monitoring complete blood counts before initiating therapy and during therapy. Grade 3 and 4 thrombocytopaenia can usually be avoided by dose interruption or reduction. The most frequently reported AE leading to dose interruption, discontinuation or dosage reduction was thrombocytopaenia. Dose reductions due to anaemia were also observed. Jakavi increases the likelihood of blood transfusion requirement.

Other ADRs reported with a higher frequency in the Jakavi group compared to control groups (in both pivotal studies) were: ecchymosis; pyrexia; headache; weight increased; haematoma; dizziness; urinary tract infection; hypercholesterolemia; herpes zoster; palpitations; bradycardia/sinus bradycardia; angina pectoris/angina unstable; and flatulence. Jakavi causes a decrease in heart rate and a prolongation of the PR interval. The effect of Jakavi on QTc interval is currently unclear. Adequate warnings and precautions are stated in the Product Monograph.

Severe drug-induced liver injury does not seem to be associated with Jakavi. In order to reduce the frequency and severity of AEs in patients with moderate and severe renal impairment or any degree of hepatic impairment, the Product Monograph recommends reducing the dose of Jakavi to 10 mg BID for these patients. Based on PK studies, renal and hepatic impairments significantly increased the exposure to ruxolitinib.

The exposure to ruxolitinib was substantially increased when Jakavi was co-administered with strong CYP3A4 inhibitors. The Product Monograph recommends reducing the dose of Jakavi when administered with strong CYP3A4 inhibitors.

Following interruption or discontinuation of Jakavi, symptoms of myelofibrosis may return over a period of approximately 1 week. It has not been established whether abrupt discontinuation of Jakavi contributed to these events. A warning regarding the potential for withdrawal effect when Jakavi is abruptly interrupted is captured in the Product Monograph, and a gradual tapering of Jakavi, when feasible, is recommended.

Animal studies have shown that ruxolitinib is embryotoxic and foetotoxic. The Product Monograph contains appropriate recommendations to limit the exposure of the embryos to ruxolitinib.

The risks associated with Jakavi for the recommended indication are appropriately managed by the Product Monograph labelling. No additional post-marketed commitments are required in order to recommend the marketing authorization of Jakavi. Nevertheless, the sponsor has confirmed that the results of the following studies will be provided to Health Canada when they become available:

  1. Report of the JUMP trial. An open-label, single-arm, multicentre, expanded access study intended to provide additional data on the safety and efficacy of ruxolitinib in patients with PMF, PPV-MF, or PET-MF who have either previously received treatment or have not received treatment.
  2. The reports that will be submitted to the FDA as part of the post-marketed commitments from the American sponsor. These commitment are to:
    • Provide safety findings related to the interval of drug discontinuation in at least 75 patients previously entered on INCB-351 to determine if specific cautions are appropriate to describe discontinuation strategies.
    • Provide safety findings related to the interval of drug discontinuation in at least 75 patients previously entered on INCB-352 to determine if specific cautions are appropriate to describe discontinuation strategies.
    • Collect and analyze safety information on myelosuppression for up to 144 weeks of therapy following randomization in the patients entered on INCB-351 who are continuing on therapy past 24 weeks.
    • Collect and analyze safety information on myelosuppression for up to 144 weeks of therapy following randomization in the patients entered on INCB-352 who are continuing on therapy past 48 weeks.
    • Provide longer-term efficacy and safety outcomes of current clinical trial INCB-351 to provide at least 3-year follow-up data.
    • Provide longer-term efficacy and safety outcomes of current clinical trial INCB-352 to provide at least 3-year follow-up data.
  3. The reports from the completed extensions of the Phase III pivotal studies COMFORT-I and COMFORT-II, as requested by Health Canada's Marketed Health Products Directorate (MHPD).
  4. The final report of the safety of ruxolitinib in patients with baseline platelet count <100,000/mm³, as requested by MHPD.

The sponsor has prepared an acceptable risk management plan. The revised labelling information accurately reflects the safety, efficacy, and quality assessments. Jakavi can significantly reduce splenomegaly and its associated symptoms in adult patients with PMF, PPV-MF or PET-MF. The adverse drug reactions are clinically manageable. The benefit-risk profile for the administration of Jakavi for the revised indication is highly favourable. The marketing authorization of Jakavi would allow MF patients to benefit from a drug that has the potential to improve the disease-related splenomegaly and its symptoms, for which there is currently no drugs approved in Canada.

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 Jakavi is favourable in the treatment of splenomegaly and/or its associated symptoms in adult patients with primary myelofibrosis (also known as chronic idiopathic myelofibrosis), post-polycythaemia vera myelofibrosis or post-essential thrombocythaemia myelofibrosis. The New Drug Submission complies with the requirements of sections C.08.002 and C.08.005.1 and therefore Health Canada has granted the Notice of Compliance pursuant to section C.08.004 of the Food and Drug Regulations.

4 Submission Milestones

Submission Milestones: Jakavi

Submission MilestoneDate
Pre-submission meeting:2011-07-14
Request for priority status
Filed:2011-08-11
Approval issued by Director of Bureau of Metabolism, Oncology and Reproductive Sciences:2011-09-29
Submission filed:2011-11-29
Screening
Screening Acceptance Letter issued:2011-12-22
Labelling Review complete:2012-06-15
Notice of Compliance issued by Director General:2012-06-19