Summary Basis of Decision for Exjade

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
Exjade

Deferasirox, 125 mg, 250 mg, 500 mg, Tablet for suspension, Oral

Novartis Pharmaceuticals Canada Inc.

Submission control no: 099621

Date issued: 2007-11-27

Health Products and Food Branch

Our mission is to help the people of Canada maintain and improve their health.
Health Canada

HPFB's Mandate is to take an integrated approach to managing the health-related risks and benefits of health products and food by:

  • minimizing health risk factors to Canadians while maximizing the safety provided by the regulatory system for health products and food; and,
  • promoting conditions that enable Canadians to make healthy choices and providing information so that they can make informed decisions about their health.

Health Products and Food Branch

Également disponible en français sous le titre : Sommaire des motifs de décision (SMD), PrEXJADE, Déférasirox 125 mg, 250 mg, 500 mg, comprimés, Novartis Pharmaceuticals Canada Inc. No de contrôle de la présentation 099621

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:

Exjade

Manufacturer/sponsor:

Novartis Pharmaceuticals Canada Inc.

Medicinal ingredient:

Deferasirox

International non-proprietary Name:

Deferasirox

Strength:

125 mg, 250 mg, 500 mg

Dosage form:

Tablet for suspension

Route of administration:

Oral

Drug identification number(DIN):

  • 125 mg/tablet 02287420
  • 250 mg/tablet 02287439
  • 500 mg/tablet 02287447

Therapeutic Classification:

Iron-chelating agent

Non-medicinal ingredients:

Lactose monohydrate, crospovidone, povidone, sodium lauryl sulphate, microcrystalline cellulose, silicon dioxide and magnesium stearate.

Submission type and control no:

New Drug Submission, Control No. 099621

Date of Submission:

2005-06-24

Date of authorization:

2006-10-18

EXJADE is a registered trademark

2 Notice of decision

On October 18, 2006, Health Canada issued a Notice of Compliance with Conditions (NOC/c) to Novartis Pharmaceuticals Canada Inc. for the drug product Exjade. The product was authorized under the NOC/c Policy on the basis of the promising nature of the clinical evidence and the need for confirmatory studies to verify the clinical benefit. Patients should be advised of the fact that the market authorization was issued with conditions.

Exjade contains the medicinal ingredient deferasirox which is an iron chelating agent.

Exjade is indicated for the management of chronic iron overload in patients with transfusion-dependent anemias aged 6 years or older. Exjade is also indicated in the management of chronic iron overload in patients with transfusion-dependent anemias aged two to five who cannot be adequately treated with deferoxamine. In transfusion-dependent anemias, iron delivered to the patient in red blood cells of repeated transfusions accumulates in the body. Exjade has a high affinity for iron, removes the excess iron from the body, and reduces the risk of organ damage caused by iron overload.

Priority review status was granted based on the serious and fatal consequences of untreated transfusional iron overload and the potential of Exjade to provide significant clinical benefit over the existing therapy, deferoxamine. 

The market authorization was based on submitted data from quality (chemistry and manufacturing) studies, as well as data from preclinical and clinical studies. The clinical program enrolled more than 1000 patients worldwide, but there are limited data on pediatric patients aged 2 to 5 (52 patients) and lack of long-term data in adult patients.

Efficacy and safety of Exjade were investigated in a one-year, open-label, randomized Phase III head-to-head trial versus deferoxamine in patients with β thalassemia and transfusional hemosiderosis and in a one-year, open-label  uncontrolled Phase II study in patients with chronic anemias and transfusional hemosiderosis who were unable to be treated with deferoxamine. In addition, a randomized, controlled Phase II safety and tolerability study was conducted in sickle cell disease patients with chronic iron overload from repeated blood transfusions. The two efficacy studies did not meet the criteria for a primary efficacy endpoint, but post-hoc subgroup analyses showed that patients with liver iron content (LIC) 7 mg Fe/g dry weight (dw) who were treated with Exjade 20 to 30 mg/kg per day had a significant reduction in LIC from baseline that was no different from deferoxamine. Dose dependent effects in serum ferritin and in the ratio of iron excretion to iron intake were also observed following doses of Exjade 5 to 30 mg/kg per day.

Exjade (125 mg, 250 mg, or 500 mg, deferasirox) is presented as dispersible tablets for oral suspension. The recommended initial daily dose of Exjade is 10, 20, or 30 mg/kg/day body weight, depending on the patient's transfusion rate and the goal of treatment. Therapy with Exjade should be initiated and maintained by physicians experienced in the treatment of chronic iron overload due to blood transfusions. Dosing guidelines are available in the Product Monograph.

Exjade is contraindicated in patients with hypersensitivity to the medicinal ingredient, deferasirox, or to any of the excipients. Exjade 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 Exjade are described in the Product Monograph.

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

3 Scientific and Regulatory Basis for Decision

3.1 Quality Basis for Decision

3.1.1 Drug Substance (Medicinal Ingredient)

General Information

Deferasirox, the medicinal ingredient of Exjade, is an iron chelating agent. In transfusion-dependent anemias, iron delivered to the patient in red blood cells of repeated transfusions accumulates in the body. Deferasirox has a high affinity for iron, removes the excess iron from the body, and reduces the risk of organ damage caused by iron overload.

Manufacturing Process and Process Controls

Deferasirox is synthetically derived. Materials used in the manufacture of deferasirox are considered to be suitable and/or meet standards appropriate for their intended use. The manufacturing process is considered to be adequately controlled within justified limits.

Characterization

The elucidation of the structure of deferasirox is considered to be acceptable. The batches manufactured were shown to consistently yield the same desired polymorphic form and particle distribution.

Impurities and degradation products arising from manufacturing and/or storage were reported and characterized. The proposed limits were considered satisfactorily qualified (e.g., within recommended ICH limits). Control of impurities in the drug substance is therefore considered to be acceptable.

Levels of residual solvents detected in the drug substance have been reported and are well within acceptance limits.

Control of Drug Substance

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

All batches are within the proposed specification limits. The deferasirox specification is considered to be adequately justified.

The deferasirox packaging is considered to be acceptable.

Stability

Based upon the long-term and accelerated stability study data submitted, the proposed re-test period, storage, and shipping conditions for deferasirox were supported and considered to be satisfactory.

3.1.2 Drug Product

Description and Composition

Exjade dispersible tablets are available in three strengths of the medicinal ingredient, deferasirox: 125 mg, 250 mg and 500 mg. The tablets are off-white, round, and flat with a bevelled edge, imprinted with "NVR" on one side and with "J" followed by the strength of the tablet on the other side.

The non-medicinal ingredients (excipients) are: lactose monohydrate, crospovidone, povidone, sodium lauryl sulphate, microcrystalline cellulose, silicon dioxide and magnesium stearate. All excipients found in the drug product are acceptable for use in drugs by the Canadian Food and Drug Regulations.

Exjade is packaged in blisters with aluminum lidding foil. Each package has 28 dispersible tablets.

Pharmaceutical Development

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

Manufacturing Process and Process Controls

The manufacturing process involves the following operations: pre-blending, granulation, drying, screening, blending, and compression. The method of manufacturing is considered acceptable and the process is considered adequately controlled within justified limits.

Control of Drug Product

Exjade is tested to verify that its identity, appearance, assay, mass, fineness of dispersion, disintegration, dissolution, content uniformity, and levels of degradation products and microbiological impurities are within acceptance criteria. The test specifications and analytical methods are considered acceptable.

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

Batch analysis results are within the proposed specification limits for all lots tested.

Stability

The stability results provided were within specification limits at all times and no trends or failures were noted in the data. Based upon the long-term and accelerated stability study data submitted, the proposed 36-month shelf-life at 15-30°C for the drug product is considered acceptable.

3.1.3 Facilities and Equipment

The design, operations and controls of the facility and equipment that are involved in the production are considered suitable for the activities and products manufactured. All of the proposed manufacturing sites comply with the requirements of Division 2 of the Food and Drug Regulations and have been rated Good Manufacturing Practices (GMP) compliant.

3.1.4 Adventitious Agents Safety Evaluation

Lactose monohydrate is the only excipient of animal origin. A letter of attestation confirming that lactose monohydrate is not from a BSE/TSE-affected country/area has been provided for this product indicating that it is considered safe for human use.

3.1.5 Conclusion

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

3.2 Non-Clinical Basis for Decision

3.2.1 Pharmacodynamics

The affinity and selectivity of deferasirox for iron were assessed in a number of in vivo and in vitro studies. Deferasirox was shown to have a high affinity for ferric iron, and a low affinity for ferrous iron. At deferasirox concentrations up to 80 µmol/L, a concentration range typically seen in plasma of patients receiving deferasirox at doses ≥ 20 mg/kg, the efficacy of deferasirox was approximately 40% and similar to that of deferoxamine (50%), a drug currently available for this/similar indication.

Excreted iron and reduction of organ iron levels were used as direct measures of pharmacodynamic effects. Iron excretion measurements were done in two relevant models, the non-iron loaded, bile duct cannulated rat and iron-loaded marmoset monkey. In rats, the excretion of iron in bile began rapidly (within three hours after oral administration of deferasirox). In rats and monkeys, iron excretion was largely dose proportional and protracted, indicating dose-dependent pharmacodynamic effects for up to 24 hours. In both animals, the bulk of the iron was excreted in the bile, while urinary excretion was consistently low (<15%). The efficiency of the chelator, expressed as the amount of iron excreted as a percentage of the theoretical iron binding capacity of the dose, was around 18% and 29% for rats and monkeys, respectively. For comparison, with deferoxamine given subcutaneously and deferiprone given orally, efficiencies in the range of 2-4% were achieved in both rats and monkeys.

Long-term studies performed in iron-loaded rats and non iron-loaded monkeys demonstrated effective removal of iron from the main iron storage organ, the liver. Iron was also mobilized from the reticuloendothelial (RE) system. The iron-deferasirox complexes which are either released into the blood or formed in the blood, were taken up by the liver and directed to biliary excretion. This explains the predominantly faecal excretion and the limited urinary excretion of iron in animals and man.

3.2.2 Pharmacokinetics

Absorption

The extent of oral absorption and bioavailability of deferasirox was investigated after intravenous and oral administration of 14C-labelled deferasirox in mice, rats and marmoset monkeys and with non-radiolabelled deferasirox in dogs. Human data from the clinical studies were provided for comparison. Rapid absorption of deferasirox was observed in the single oral dose studies in mice, rats, dogs, monkeys, and humans.

Distribution

Deferasirox displayed a low volume of distribution. Although distributed throughout the body, deferasirox was mainly present intravascularly. Substantial levels were found in organs of the GI tract and excretory organs. Deferasirox and the iron complex were extensively (98-99%) bound to plasma proteins for all species tested. Most of the deferasirox was confined to the blood circulation and organs that are well perfused such as heart, liver, lung, kidney, and intestine. In lactating rats, deferasirox was also transferred into the milk and consisted primarily of unchanged deferasirox.

Metabolism

Deferasirox was extensively metabolized by the liver. Oxidative metabolism of deferasirox was deemed important in rats and monkeys, but low in humans. Glucuronidation was predominant in rodents, monkeys and humans. UGT1A1 and UGT1A3 were the predominant isoenzymes in the glucuronidation of deferasirox. The common systemically available metabolite across species was the acyl-glucuronide (M3). Drug-drug interaction potential by way of competition for UGTs is unlikely given the very high plasma protein binding of deferasirox. Significant inhibition of CYP isozymes is also not expected.

Excretion

In all investigated species, the excretion of deferasirox and its metabolites occurred predominantly via the biliary/faecal route. There was evidence of enterohepatic recirculation of deferasirox and its metabolites.

3.2.3 Toxicology

The toxicology studies provided a reasonably complete toxicology data package, which is required for routine non-clinical drug evaluation. Most of the studies were Good Laboratories Practices (GLP) compliant and were conducted by qualified personnel. However, the toxicity of an iron chelator is difficult to assess. It is difficult to induce iron overload by dietary iron supplementation in animals to a level that is attainable by parenteral iron administration which occurs in the target patient population.

Toxicity of deferasirox was investigated following parenteral iron loading in the rat and marmoset. Both mechanistic studies were non-GLP compliant and used only a single, high dose. Therefore, the highest exposure level at which there are no significant increases in adverse events (NOAELs) could not be established. It is worth noting that some of the systemic toxicity of deferasirox was either reduced or absent in animals on the iron-supplemented diets, with the exception of toxicity to the kidney, eye, and lymphoid tissue which occurred independent of iron supplementation. The short-comings in the iron-loaded models (i.e., dietary iron supplemented animals) used in the pre-clinical toxicology testing of deferasirox may be addressed by outcomes in the scheduled clinical studies.

Acute Toxicity

Acute oral gavage and intravenous toxicity studies were performed in normal mice and rats. In general deferasirox was of low to moderate acute toxicity in the rodents tested.  The intravenous LD50 valueswere 150 mg/kg in the mouse and >75mg/kg in the rat. The oral LD50 values were >500mg/kg in the mouse and >1000mg/kg in the rat. Clinical signs were observed only in the mouse, and included ataxia, dehydration, dyspnoea. Acute oral studies in rodents indicated low to moderate toxicity, while the acute intravenous toxicity ranged between 50-150 mg/kg. Clinical signs were observed only in the mouse, and included ataxia, dehydration, dyspnoea, and decreased locomotor activity.

Long-Term Toxicity

In the repeat dosing studies, the major target organs of toxicity were the kidney, haematopoietic tissue, bile duct, gall bladder, gastrointestinal tract, and adrenal glands in both rodents and monkeys, as well as the lymphoid tissue, eye, and heart in rodents. Kidney and ocular toxicity occurred in both iron-supplemented and non-supplemented animals. Unlike other target organ toxicities, ocular toxicity was not diminished by iron supplementation.

Antibody response and lymphoid tissue B-cell population were suppressed by deferasirox at the 60 mg/kg/day dose in non-iron supplemented adult rats.

The highest NOAEL in non-iron supplemented adult animals was 10 mg/kg/day, based on myocardial degeneration at 30 mg/kg/day in a 4-week study in rats. However, myocardial effects were not observed in iron-supplemented rats administered deferasirox orally at doses up to 180 mg/kg/day for 26 weeks, nor in iron-supplemented or non-supplemented monkeys administered deferasirox up to a dose level of 80 mg/kg/day for 39-weeks.

In iron and non-iron supplemented juvenile rodents, the eye and the lymphoid tissue toxicity were the most sensitive organs with toxicity noted at 20 mg/kg/day for the eye and 30 mg/kg/day for lymphoid tissues, and the respective NOAELs were 10 mg/kg/day and 15 mg/kg/day.

Carcinogenicity

The oral carcinogenic potential of deferasirox was conducted in a 2-year study in iron-supplemented rats and a 26-week study in mice fed on standard or iron-supplemented diets. Deferasirox appeared to be non-carcinogenic in rats treated for 104 weeks at dose levels up to 60 mg/kg/day.

No major toxicological effects were reported at all dose levels (up to 60 mg/kg/day) in the 104 week study in rats, but this is surprising as ocular toxicity was noted at 30 mg/kg in the 26-week study.

Deferasirox appeared to be non-carcinogenic in mice (p53 heterozygous and wild-type) treated at doses up to 200 or 300 mg/kg/day for 26 weeks. Unfortunately, the wild-type mice were terminated at 26-weeks instead of the standard 18-month exposure period. Since transgenic p53 mice may not exhibit the same tissue predilection for tumours as the wild mice, and carcinogenicity induced by non-genotoxic mechanisms usually requires longer term exposure, the full carcinogenic potential of deferasirox in the mouse can not be ascertained.

Mutagenicity

The genotoxic potential of deferasirox was equivocal. Deferasirox was not genotoxic in the Ames assays, the chromosome aberration tests with human lymphocytes and in the ex vivo micronucleus test in rat liver cells. Positive results were observed in two in vitro micronucleus tests, and at a high dose (500 mg/kg) in an in vivo bone marrowmicronucleus test in non-iron supplemented rats, which may have been a result of altered hematopoiesis due to iron chelation. However, a negative response was observed at the same high dose when the rats were given iron supplementation.

Reproductive Toxicity

Deferasirox was not teratogenic in the rabbit and rat, and did not affect fertility and reproduction in adult rats. Nephropathy and cataracts were observed in rat pups at 30 mg/kg/day. These effects occurred at lower doses in the juvenile animals than in the dams.

3.2.4 Conclusion

Deferasirox has been shown to be an effective iron chelator. The animal data confirms the high affinity and selectivity of deferasirox for iron, as well as the efficient absorption and excretion of the iron-complex. The assessment of toxicity was difficult as the animal models were not able to portray the complex pathology of transfusional hemosiderosis in humans. The short-comings with the iron loading models, i.e., animals with iron supplemented diets, may be addressed by outcomes in the scheduled clinical studies.

Close clinical monitoring for potential toxicity involving the kidney, eye, heart, gastrointestinal tract, gall bladder/bile duct, and the immune system is recommended based on the non-clinical toxicity profile. Decreased tissue iron due to the pharmacological action of deferasirox was observed in all species and was generally accompanied by changes in hematologic parameters consisting mainly of anemia, with alteration of red cell morphology in some cases. Effects on erythron in the toxicology animals is likely the result of depletion of iron in the animals with normal tissue iron levels. Some of the toxic effects of deferasirox in animals appeared to occur predominantly under normal (non iron-supplemented) body iron status.

3.3 Clinical basis for decision

3.3.1 Pharmacodynamics

A pharmacodynamic study evaluated the dose response of overall iron balance, i.e., the difference between total iron intake (iron in consumed diet and medication) and iron excreted in urine and feces, following multiple oral doses of deferasirox. Negative iron balance was achieved at all three doses of active drug, and averaged approximately 0.119 mg/kg/day at the 10 mg/kg dose, 0.329 mg/kg/day at the 20 mg/kg dose, and 0.445 mg/kg/day at the 40 mg/kg dose. For each active dose, the fraction of iron excreted in urine was on average approximately 2-5% of the total dose.

The studies have confirmed that deferasirox is an active and effective iron-chelating compound in humans. There appears to be a dose-dependent effect of this compound, at least in the range of 20-30 mg/kg/day. Higher doses appear more effective than lower ones, and more so in patients with more severe iron overload at baseline. It is clear that the dosing of any iron-chelating agent needs to be adjusted based on the patient's response, since excessive chelation may lead to morbid events, while underchelation will result in progression of hemosiderosis.

3.3.2 Pharmacokinetics

Absorption

Following the administration of a single oral dose of 2.5 to 80 mg/kg of deferasirox, the maximum plasma concentration and exposure of deferasirox increased approximately proportionally with the dose, but with substantial variability. The mean half-life of the drug was between 11 and 19 hours.

The mean percentage of deferasirox chelated to iron ranged from 15 to 31%. The pharmacokinetic profile of deferasirox and its iron complex appears compatible with a once a day oral administration.

Drug accumulation occurred in all age groups after multiple dosing of deferasirox. Total drug exposure (AUC) was 60% higher after 2 weeks of treatment compared to Day 1 (90% CI for the ratio: 1.38, 1.85). After 4 weeks of deferasirox treatment, the AUC0-24 had doubled compared to Day 1 (90% CI for the ratio: 1.72, 2.31), showing a 25% further increase in the last 2 weeks (90% CI for the ratio: 1.08, 1.45).

A food effect on bioavailability was reported. Food increased the fraction available to the systemic circulation.

Distribution

Deferasirox was extensively bound (>98%) to plasma proteins. In blood, only 5% of the radiolabelled deferasirox was associated with red blood cells.

Metabolism

The main metabolic pathway of deferasirox was glucuronidation and the primary enzyme responsible was UGT1A1. Based on molecular mass and comparison with reference material, M3 was identified as the acyl glucuronide metabolite of deferasirox. Studies with radiolabelled deferasirox showed that M3 exposure accounted for 3.3% of the total radioactivity in plasma.

Oxidative metabolism by CYP enzymes was a minor elimination process, therefore any inhibition or induction of those enzymes by co-medications is not expected to significantly affect the pharmacokinetics of deferasirox.

Excretion

Deferasirox was primarily eliminated in the faeces (84% of the dose) with enterohepatic recirculation likely but not proven. The predominant radioactive compound excreted in the faeces was unchanged deferasirox. Renal excretion amounted to only 8% of the dose. In the collection period of 7 days, the mean total recovery amounted to 91%, with low inter-individual variability.

Drug Interactions

Potential for drug-drug interactions are considered to be minimal although this conclusion is based more on theoretical reasoning based on the extremely high protein binding of deferasirox and of its iron complex rather than on definitive studies.

Special Populations

The pharmacokinetics (PK) of deferasirox in children were significantly different from that in adults. In children under 6 years of age, blood levels of deferasirox were half that obtained in adults at equivalent doses, for reasons that remain to be elucidated. There were no studies in patients with significant renal or hepatic impairment although transaminase levels of approximately five times the upper limit of normal did not seem to affect the PK of deferasirox. The sponsor is currently conducting a single dose (20 mg/kg per day) PK study of deferasirox in patients with hepatic impairment and the results will be submitted when available.

3.3.3 Clinical Efficacy

The efficacy of Exjade was investigated primarily in two studies, Study 107 and Study 108.

Study 107 was a one-year, multi-centre, open-label, randomized Phase III active comparator control study that compared Exjade (deferasirox) to deferoxamine in patients ≥ 2 years of age with β thalassemia and transfusional hemosiderosis. The starting doses were based on LIC (liver iron concentration) at baseline [2-3, >3-7, >7-14 and >14 mg Fe/g dry weight (dw)]. Patients treated with deferasirox (n=296) received starting doses of 5, 10, 20 or 30 mg/kg orally once daily and patients treated with deferoxamine (n=290) received subcutaneous starting doses of 20 to 60 mg/kg for at least 5 days per week. The ratio of deferasirox to deferoxamine doses for the two lower LIC categories was disproportionately low (1:4) compared to the two upper LIC categories (1:2). The primary efficacy endpoint, was defined as a reduction in LIC of ≥ 3 mg Fe/g dw for baseline values ≥ 10 mg Fe/g dw, reduction of baseline values between 7 and <10 to <7 mg Fe/g dw, or maintenance or reduction for baseline values <7 mg Fe/g dw.

Non-inferiority of Exjade (deferasirox) to deferoxamine in terms of success rates based on changes in LIC was not shown in the overall study population due to the disproportionately low dosing of deferasirox relative to deferoxamine in patients with baseline LIC <7 mg Fe/g dw. Non-inferiority was demonstrated in patients with LIC 7 mg Fe/g dw in which doses of deferasirox and deferoxamine were used as specified in the protocol. On average, 20 mg/kg deferasirox in regularly transfused adult and pediatric β-thalassemia patients was able to maintain iron balance whereas a negative iron balance was induced at 30 mg/kg. Doses of 5 and 10 mg/kg deferasirox on average did not maintain iron balance in this regularly transfused population. Dose-dependent responses in all age groups were consistently observed for the secondary efficacy parameters including absolute changes in LIC, serum ferritin and iron balance, and also for liver pathology. Serum ferritin trends over time closely reflected changes in LIC supporting the use of repeated measurements of this parameter to monitor iron accumulation and removal during chelation therapy.

Study 108 was a one year, multi-centre, open-label, non-comparative, Phase II study of Exjade (deferasirox) in patients ≥ 2 years of age with chronic anemias and transfusional hemosiderosis unable to be treated with deferoxamine. Patients (n=184) received 5, 10, 20, or 30 mg/kg per day of deferasirox based on baseline LIC. The primary efficacy endpoint was to demonstrate a success rate significantly greater than 50% with deferasirox.

The overall success criteria based on changes in LIC were not reached in the combined disease groups due to a relatively high drop-out rate due to the underlying disease and inappropriately low dosing of deferasirox in patients with baseline LIC <7 mg Fe/g dw. These success criteria were met in the combined disease groups in patients with LIC ≥ 7 mg Fe/g dw in which doses of 20 and 30 mg/kg deferasirox were employed. On average, 20 mg/kg deferasirox in regularly transfused adult and pediatric patients was able to maintain iron balance in both disease groups whereas a negative iron balance was induced at 30 mg/kg. Doses of 5 and 10 mg/kg, on average, did not maintain iron balance in this regularly transfused population. However, in patients with myelodysplastic syndrome (MDS), who had a lower transfusional iron intake, this dose response relationship was shifted such that 10 mg/kg deferasirox was able to maintain iron balance and marked negative iron balance was induced at 20 and 30 mg/kg. The analyses of iron balance indicate that the frequency and amount of blood transfusions, and hence the quantity of transfusional iron administered, has a direct impact on the dose of deferasirox needed to maintain or induce negative iron balance.

Both efficacy studies, Study 107 and Study 108, did not meet the criteria for their primary efficacy endpoints, but post-hoc subgroup analyses showed that patients with LIC ≥ 7 mg Fe/g  dw who were treated with 20 to 30 mg/kg per day Exjade had a significant reduction in LIC from baseline that was no different from deferoxamine. Dose dependent effects in serum ferritin and in the ratio of iron excretion to iron intake were also observed following doses of 5 to 30 mg/kg per day Exjade. The studies did not establish the efficacy of Exjade with moderate iron overload, i.e. LIC <7 Fe/g dw. Additional studies of Exjade at 20-30 mg/kg per day in these populations have been scheduled.

3.3.4 Clinical Safety

The safety of Exjade was investigated in several Phase II clinical studies (105, 106, 108, 109) and one Phase III study (107). Evidence regarding therapeutic benefit of deferasirox was based mainly on Study 107 which comprised of β-thalassemia patients aged ≥ 2 years, Study 108 which comprised of β-thalassemia patients and patients with other rare anemias aged ≥ 2 years [including elderly patients with myelodysplastic syndrome (MDS)], and Study 109 which comprised of patients with sickle cell disease aged ≥ 2years. Studies 107 and 108 (described in section 3.3.3 Efficacy) contributed 480 patients exposed to Exjade and Studies 105, 106, and 109 contributed another 220 patients treated with Exjade.

Deferasirox had a favourable safety profile with a low rate of discontinuation due to adverse events (AEs). The most frequently reported AEs most likely related to Exjade included transient, manageable, mild to moderate gastrointestinal (GI) complaints (abdominal pain, nausea, vomiting and diarrhea) and skin rash. Major adverse drug reactions that often required dose reductions involved renal, hepatobiliary, skin rash and ocular effects.

In Study 107, Exjade appeared to be associated with a higher incidence of GI adverse effects compared to deferoxamine. Effects on vision and hearing appeared to be comparable. In a number of cases, serious adverse reactions, such as cardiac rhythm disturbances and retinal degeneration, were not reported or discussed because they were deemed to be not drug related. The causality assessments for these serious adverse reactions were not explicitly explained in the study report. In approximately one third of the patients treated with Exjade , mild non-progressive increases in serum creatinine (generally within the normal range) were reported. Some of the increases were transient and in less than 10% of the patients precautionary dose reductions were performed. Four patients discontinued due to transaminase elevations. Probable drug-induced hepatotoxicity with a marked increase in transaminase levels occurred in at least one or possibly two patients. There were no episodes of drug-induced neutropenia, agranulocytosis or thrombocytopenia in either treatment group. There was no evidence of consistent relevant effects on any of the cardiac safety parameters measured. The results of vital signs and ECG monitoring were unremarkable. The higher incidence of renal effects, including increased serum creatinine and proteinuria, and hepatic transaminases, should be further characterized in adequate post-marketing studies.

Study 108 was a difficult study due to the number of various diseases with a wide spectrum of transfusion requirements and morbidity. In particular, the MDS group had significant withdrawal due to the complications of the underlying disease. This was true for all patients in the rare disease group. Additional long-term studies with MDS patients are required to better define the safety profile. The β-thalassemia population did not contribute any new information regarding Exjade Similar to Study 107, the major adverse finding (laboratory finding) was a non-progressive increase in serum creatinine in patients treated with Exjade which on average measured approximately 10 μmol/L.  Dose reductions were performed in 32 of the 73 patients (43.8%) who had >33% creatinine increases from baseline. Of the 32 patients who underwent dose reductions, only 8 patients returned to normal baseline serum creatinine levels. 

Study 109 assessed the safety of Exjade in patients with sickle cell disease. As in Study 107, patients received 5, 10, 20, or 30 mg/kg per day of Exjade or subcutaneous deferoxamine at doses of 20 to 60 mg/kg for five days per week based on baseline LIC. With a median exposure of six months, Exjade had an acceptable safety profile in patients ≥ 2 years old with sickle cell disease. The side effects of Exjade in patients with sickle cell disease were similar to that observed in patients with β-thalassemia. Mild, non-progressive increases in serum creatinine were observed in 25% of patients receiving Exjade. The most significant finding was that the renal toxicity was not more prevalent or serious in this group of patients with a high probability of renal pathology associated with their sickle cell disorders.

The safety profile in children less than 6 years of age was generally comparable to that seen in adolescents and adults with the exception of an increased incidence of upper respiratory tract symptoms, cough, rhinitis, fever, and diarrhea. This may be indicative of increased incidence of infection due to depression of the immune system, a finding supported by non-clinical data. The clinical program enrolled more than 1000 patients worldwide, but there are limited data on paediatric patients aged 2 to 5 (52 patients). A multi-national pediatric registry is being set up as a five-year observational study specifically to generate additional safety information in the 2- to 6-year age group of patients with transfusional hemosiderosis.

Additional studies are required to evaluate cardiac iron concentration and cardiac function, as well as clinical outcomes with patients treated with Exjade. The relationship between LIC and iron levels in the heart remains unclear but needs to be investigated as most inadequately chelated transfusion-dependent patients succumb to fatal cardiotoxicity.

Although no adverse effects were observed in a variety of anthropomorphic measurements or in indices of sexual development, the one-year studies are not sufficiently long in duration nor sufficiently powered to definitively conclude that Exjade is devoid of adverse effects in this respect. Overall, the relatively small numbers of patients investigated for each dose level and age group precluded the detection of adverse effects that occur with an incidence below 5%. Thus, more infrequently occurring potential adverse events, such as agranulocytosis and neutropenia, (toxicities associated with deferiprone, an oral iron chelating agent marketed in Europe) would not have been identified. Furthermore, studies longer than one year in duration are required to determine whether reduction in morbidity and mortality is similar to that achieved with deferoxamine patients. In recognizing the need for additional long-term studies, the sponsor has implemented four-year extensions to Studies 105, 106, 107, 108 and 109.

3.3.5 Issues Outstanding

Under the Notice of Compliance with Conditions (NOC/c) policy, the sponsor is required to submit the results of the following confirmatory studies:

  • Extension studies 105E2, 106E1, 107E1, 108E1 and 109E1 for a total of four years after the core trial.

  • A registry for children aged 2 to <6 years to enroll approximately 200 patients with a 5-year follow-up.

  • A single dose (20 mg/kg per day) pharmacokinetics study of Exjade in subjects with hepatic impairment.

  • A single-arm study in patients (adults and pediatrics) with congenital or acquired anemias and chronic iron overload to obtain additional data in patients with LIC <7 mg Fe/g dw treated with Exjade doses of 20 or 30 mg/kg per day.

  • A study of long-term follow-up (3 years) in 150 patients with myelodysplastic syndromes (MDS) receiving Exjade including patients with baseline serum creatinine values up to two times the upper limit of normal to evaluate safety and haematologic and clinical benefit of Exjade in these patients.

  • Studies of cardiac iron concentration, cardiac function, as well as clinical outcomes to explore the relationship amongst iron body burden as measured by LIC, cardiac iron, serum ferritin, and transfusion rates in patients treated with Exjade, using a validated methodology.

The sponsor has agreed to conduct additional post-market safety and mechanistic studies to further characterize the safety profile of Exjade and final reports will be provided upon completion. The sponsor will include monitoring and follow-up of adverse events from these studies as well as events noted in post-marketing data and in the major safety concerns identified. The sponsor has agreed to submit Periodic Safety Reports (PSURs) until the conditions have been fulfilled and removed from the NOC/c by Health Canada.

3.4 Benefit/Risk Assessment and Recommendation

3.4.1 Benefit/Risk Assessment

Exjade was granted priority review status based on the serious and fatal consequences of untreated transfusional iron overload and the potential of Exjade to provide significant clinical benefit over the existing therapy, deferoxamine. The greatest risk associated with deferoxamine is lack of compliance. The mode of administration of deferoxamine can be so burdensome that patients forgo treatment despite the knowledge that such behaviour will significantly affect their quality of life and considerably shorten their life span. In poorly compliant thalassemia patients, survival at age 25 is only 32%, whereas with good compliance almost all patients reach age 25. Deferoxamine is also difficult to administer in very young children who often must begin iron chelation therapy at 2 to3 years of age.

Thus, the clinical benefit of Exjade is not necessarily related to enhanced efficacy or decreased toxicity compared to deferoxamine, but to enhanced compliance due to a once daily oral dosing regimen.

Evaluation of the reports has confirmed that Exjade is an active and effective iron-chelating compound in humans. Adequate evidence exists that show patients with LIC ≥ 7 mg Fe/g dw who were treated with 20 to 30 mg/kg Exjade per day had a significant reduction in LIC from baseline that was no different from deferoxamine. Dose dependent effects in serum ferritin and in the ratio of iron excretion to iron intake were also observed following doses of 5 to 30 mg/kg Exjade per day.

Treatment with deferasirox for up to a year, indicate that 20-30 mg/kg is well tolerated in patients with high baseline LIC and that 10 mg/kg is well tolerated in patients with low baseline LIC. Doses of 20-30 mg/kg are also well tolerated in patients whose LIC fell from >7 to <7 mg Fe/g dw during the studies. The tolerability of deferasirox was similar across different anemias studied. No differential safety results were identified that raise a material concern about a higher drug-related risk in a particular patient population with transfusion-dependent anemias, whether adult or paediatric.

Due to the fact that efficacy data were obtained in a limited number of pediatric patients and long term data were not available, Health Canada proposed a restricted indication for Exjade, as well as the requirement for confirmatory studies to further establish clinical benefit.The restricted indication stipulates that Exjade can only be used in children aged 2 to 5 years who cannot be adequately treated with deferoxamine. Furthermore, the indication restricts the use of Exjade to use only under supervision by specialists experienced in the treatment of chronic iron overload due to blood transfusions. The sponsor is committed to pursue the assessment of safety and efficacy of Exjade in this paediatric patient population by establishing a registry involving approximately 200 children worldwide with a five year follow-up. To address the issue of long-term safety, the sponsor has implemented extensions to generate up to five years of follow-up treatment for selected clinical studies and has agreed to conduct additional post-market studies to further characterize the safety profile.

The dosing and administration instructions have been revised based on current knowledge and clinical practice and are considered to be adequate. The risks and uncertainties regarding safety are appropriately labelled in the Product Monograph. The Product Monograph will be also revised accordingly, pending the results of further confirmatory and post-market safety studies being conducted by the sponsor as part of the approval conditions.  

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 Exjade is favourable for the management of chronic iron overload in patients with transfusion-dependent anemias aged 6 years or older, and for the management of chronic iron overload in patients with transfusion-dependent anemias aged 2 to 5 who cannot be adequately treated with deferoxamine.

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

Consistent with the NOC/c policy, the sponsor has agreed to submit the results of the following confirmatory studies:

  • Extension studies 105E2, 106E1, 107E1, 108E1 and 109E1 for a total of four years after the core trial.

  • A registry for children aged 2 to <6 years to enroll approximately 200 patients with a 5 year follow-up.

  • A single dose (20 mg/kg per day) pharmacokinetics study of Exjade in subjects with hepatic impairment.

  • A single arm study in patients (adults and pediatrics) with congenital or acquired anemias and chronic iron overload to obtain additional data in patients with LIC <7 mg Fe/g dw treated with Exjade doses of 20 or 30 mg/kg per day.

  • A study of long-term follow-up (3 years) in 150 patients with myelodysplastic syndromes (MDS) receiving Exjade including patients with baseline serum creatinine values up to two times the upper limit of normal to evaluate safety and hematologic and clinical benefit of Exjade in these patients.

  • Studies of cardiac iron concentration, cardiac function, as well as clinical outcomes to explore the relationship amongst iron body burden as measured by LIC, cardiac iron, serum ferritin, and transfusion rates in patients treated with Exjade, using a validated methodology.

The sponsor has agreed to conduct additional post-market safety and mechanistic studies to further characterize the safety profile of Exjade and final reports will be provided upon completion. The sponsor will include monitoring and follow-up of adverse events from these studies as well as events noted in post-marketing data and in the major safety concerns identified. The sponsor has agreed to submit Periodic Safety Reports until the conditions have been fulfilled and removed from the NOC/c by Health Canada.

4 Submission Milestones

Submission Milestones: Exjade

Submission MilestoneDate
Pre-submission meeting2005-04-06
Request for priority status
Filed2007-05-17
Approval issued by Bureau of Metabolism, Oncology and Reproductive Sciences2005-06-15
Submission filed2005-06-24
Screening 1
Screening Acceptance Letter issued2005-07-28
Review 1
Quality Evaluation complete2005-10-26
Clinical Evaluation complete2006-01-20
NON issued by Director General (safety, efficacy, issues)2006-01-23
NON Response filed2006-04-21
Screening 2
Screening Acceptance Letter issued2006-05-19
Review 2
NOC/c-QN issued2006-08-17
Response filed2006-09-16
Labelling Review complete2006-10-16
NOC/c issued by Director General2006-10-18