Summary Basis of Decision for Clolar ®

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
Clolar®

Clofarabine, 1 mg/mL, Solution, Intravenous

Genzyme Canada Inc.

Submission control no: 121874

Date issued: 2009-11-16

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:

Clolar®

Manufacturer/sponsor:

Genzyme Canada Inc.

Medicinal ingredient:

Clofarabine

International non-proprietary Name:

Clofarabine

Strength:

mg/mL

Dosage form:

Solution

Route of administration:

Intravenous

Drug identification number(DIN):

  • 02330407

Therapeutic Classification:

Antineoplastic agent

Non-medicinal ingredients:

Water for injection and sodium chloride

Submission type and control no:

New Drug Submission, Control Number: 121874

Date of Submission:

2008-05-08

Date of authorization:

2009-07-16
2 Notice of decision

On July 16, 2009, Health Canada issued a Notice of Compliance to Genzyme Canada Inc. for the drug product Clolar.

Clolar contains the medicinal ingredient clofarabine which is an antineoplastic agent.

Clolar is indicated for the treatment of paediatric patients 1 to 21 years old with relapsed or refractory acute lymphoblastic leukaemia (ALL) after at least two prior regimens. Clofarabine is a prodrug that exerts its activity through its active metabolites. The metabolite clofarabine triphosphate inhibits deoxyribonucleic acid (DNA) synthesis and induces apoptosis, while clofarabine monophosphate has been shown to inhibit DNA repair.

The market authorization was based on quality, non-clinical, and clinical information submitted. The pivotal study was a Phase II open-label, non-randomized, single-arm study of 61 paediatric patients with relapsed/refractory ALL, previously treated with a median of 3 prior induction regimens. Patients received a dose of 52 mg/m2 clofarabine over 2 hours for 5 consecutive days repeated every 2 to 6 weeks for up to 12 cycles (median 2 cycles). The overall remission rate was the primary endpoint. Twelve of the 61 (20%) ALL patients achieved complete remission (CR) or complete remission without platelet recovery (CRp). Of 35 patients who were refractory to their immediately preceding induction regimen, 6 (17.1%) achieved a CR or a CRp. Of 18 patients who had at least one prior haematopoietic stem cell transplant, 5 (27.8%) achieved a CR or CRp.

The use of Clolar is based on the induction of complete responses. Approval was based on objective response rates. No survival advantage was demonstrated and palliative benefit was not evaluated. The safety profile of clofarabine is considered to be acceptable and predictable in patients with relapsed/refractory ALL.

Clolar (1 mg/mL, clofarabine) is presented as a solution. The paediatric (ages 1-21) dose of 52 mg/m2 of body surface area is administered by intravenous infusion over 2 hours daily for 5 consecutive days. Detailed instructions and dosing guidelines are available in the Product Monograph.

Clolar is contraindicated for patients who have the following conditions:

  • Hypersensitivity to clofarabine or any of the excipients;
  • Symptomatic central nervous system involvement;
  • History of serious heart, liver, kidney or pancreas disease;
  • Severe hepatic impairment defined as elevated transaminases (aspartate aminotransferase and/or alanine aminotransferase) >5x the upper limit of normal (ULN) and/or elevated bilirubin >3x ULN, or severe renal insufficiency (defined as creatinine clearance <30 mL/min).

Clolar 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 Clolar are described in the Product Monograph.

Based on the Health Canada review of data on quality, safety, and efficacy, Health Canada considers that the benefit/risk profile of Clolar is favourable for the treatment of paediatric patients 1 to 21 years old with relapsed or refractory ALL after at least two prior regimens.

3 Scientific and Regulatory Basis for Decision

3.1 Quality Basis for Decision

3.1.1 Drug Substance (Medicinal Ingredient)

General Information

Clofarabine, the medicinal ingredient of Clolar, is an antineoplastic agent. Clofarabine is a prodrug and exerts its activity through its active metabolites. The metabolite clofarabine triphosphate inhibits DNA synthesis via: (1) reduction of deoxynucleotide triphosphate pools by inhibiting ribonucleotide reductase, (2) by competitive inhibition of DNA polymerase alpha, and (3) by terminating DNA chain elongation. In addition, the triphosphate induces apoptosis through the disruption of mitochondrial membrane integrity with the release of cytochrome C and other pro-apoptotic factors. Clofarabine monophosphate has been shown to inhibit DNA repair through incorporation into the DNA chain during the repair process.

Manufacturing Process and Process Controls

Clofarabine 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

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

The sponsor has provided a summary of all drug-related impurities. The proposed limits are considered adequately qualified.

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 clofarabine.

Batch analysis results were reviewed and all results comply with the specifications and demonstrate consistent quality of the batches produced.

The drug substance packaging is considered acceptable.

Stability

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

3.1.2 Drug Product

Description and Composition

Clolar (clofarabine, 1 mg/mL) is a clear and practically colourless solution for IV infusion supplied in a 20-mL single-use vial, fitted with a rubber stopper and sealed with an aluminum crimp and flip-off cap. The 20-mL vial contains 20 mg of clofarabine formulated in 20 mL of unbuffered normal saline [comprised of Water for Injection, United States Pharmacopeia (USP), and Sodium Chloride USP]. Clolar is packaged in single-pack and four-pack cartons. Not all package sizes may be marketed.

All non-medicinal ingredients (excipients) found in the drug product are acceptable for use in drugs according to the Food and Drug Regulations. The excipients are widely used in formulation of parenteral products.

Pharmaceutical Development

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

Manufacturing Process and Process Controls

The manufacturing process uses conventional manufacturing techniques normally used for solutions for injection. The manufacturing process is considered to be adequately controlled within justified limits.

Control of Drug Product

Clolar is tested to verify that its identity, appearance, assay, pH, particulate matter, extractable volume, sterility, osmolality, and levels of impurities/degradation products are within acceptance criteria as per the Health Canada/ICH requirements.

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

Stability

Based on the real-time long-term and accelerated stability data, the proposed 24-month shelf-life for Clolar is considered acceptable when the product is packaged in clear Glass Type I vial with West 4416/50 siliconized stopper and polypropylene flip off cap when stored at a controlled room temperature (25°C) with excursions between 15-30°C.

3.1.3 Facilities and Equipment

All of the proposed manufacturing sites comply with the requirements of Division 2 of the Food and Drug Regulations. The sites are rated Good Manufacturing Practices (GMP) compliant for the manufacturing activities.

3.1.4 Adventitious Agents Safety Evaluation

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

3.1.5 Conclusion

The Chemistry and Manufacturing information submitted for Clolar 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 results from the primary pharmacodynamic studies support the in vitro cytotoxicity of clofarabine in rapidly dividing proliferating and quiescent cancer cell-types with a reduction of tumour growth in immunocompromised animal models. The characterization of the mechanism of action is described and essentially delineates clofarabine as a prodrug for the pharmacologically active mono- and tri-phosphorylated metabolites. The non-clinical pharmacology efficacy results are qualitatively supportive of the rationale for developing the drug but there are no results that can be used to guide the quantitative relationship and scaling to potential clinical dosing regimen.

A battery of safety pharmacology studies was conducted. Deaths occurred at high IV dose levels of clofarabine. The signs at the high-dose levels included tremor, sedation, motor signs, hypothermia and transient bronchodilation. The cardiovascular safety evaluation was limited to one study in rats, and no measurements of electrocardiogram (ECG) intervals were determined [for example, (e.g.) QT intervals] in animals administered clofarabine. There were also no non-clinical in vivo studies to address the potential effects of circulating clofarabine on QT intervals (or on ECGs in general). Clofarabine was negative in the in vitro hERG assay. Thus, while the negative hERG assay is supportive of a lack of effect of clofarabine on QT intervals, the customary battery of non-clinical studies/evaluations to address this issue were not performed.

3.2.2 Pharmacokinetics

The pharmacokinetics (PK) of clofarabine have been studied in vivo in mice, rats, and dogs. No non-clinical PK data in animals are available regarding the phosphorylated metabolites of clofarabine. The metabolism of clofarabine was evaluated in vitro and in vivo.

Absorption

In an IV and oral PK study of clofarabine in mice, the increases in drug exposure were dose proportional with rapid elimination and high oral bioavailability. Oral bioavailability in rats and mice exceeded 50% in both species.

Distribution

The volume of distribution was high in rats, mice, and dogs supporting wide distribution in all species tested.

After 20 minutes of a single IV dose in mice, the highest concentrations of the drug were found in the liver, kidney, pancreas, and spleen; and relatively high levels were detected in the lung, myocardium, and skin. Clofarabine was found in the brain but at much lower concentrations than in the highly perfused tissues and organs.

Metabolism

In vivo metabolism of clofarabine was characterized in the rat. The most abundant metabolite was 6-ketoclofarabine, formed via oxidative deamination, a reaction that is not catalyzed by the cytochrome P450 (CYP) system. The lack of CYP metabolism of clofarabine in humans, rats, and dogs was supported by a number of in vitro studies. Clofarabine is not a CYP substrate, does not inhibit CYP-catalyzed reactions, and no CYP-mediated metabolites were detected in isolated primary cultures of hepatocytes. The active phosphorylated metabolites have only been detected in vivo at very low concentrations and were not formed in the isolated hepatocyte preparations. There is no non-clinical information regarding the correlation between plasma clofarabine and the cellular concentrations of the active metabolites.

Excretion

In rats, clofarabine was primarily excreted in the urine. Approximately 76% of the administered dose was excreted in the urine as unchanged clofarabine and 21.7% of the administered dose was excreted as metabolites in the urine and faeces. The renal clearance of clofarabine was 240 mL/hour which greatly exceeds the calculated glomerular filtration rate of unbound clofarabine in the rat consistent with both filtration and active secretion from blood to urine.

3.2.3 Toxicology

Single-Dose Toxicity

There were no dedicated single-dose toxicity studies in this drug submission. In a dose-finding study, a single IV dose of clofarabine at 50 mg/kg (300 mg/m2) was lethal to one of three rats and at 100 mg/kg (600 mg/m2) all three male rats died.

Repeat-Dose Toxicity

Repeat-dose studies in rats and dogs, and to some extent in mice, characterized the toxicity profile of clofarabine. The majority of treatment-related effects in rats and mice reflected adverse effects on tissues with rapidly dividing cells (e.g., gastrointestinal tract, bone marrow and blood cells, lymphoid tissues, and testes) as anticipated, based on the pharmacologic activity of clofarabine. In addition, heart, skeletal muscle, liver, and kidney were target organs in rats. Dose-limiting toxicity in the repeat-dose studies reflected mainly gastrointestinal toxicity in mice and dogs and cardiac toxicity in rats. Survivability following repeat-dose IV administration in dogs was feasible only at dose levels (and associated systemic exposures) that were considerably lower than those employed in the rat studies. The IV administration of daily dose levels and systemic exposures lower than those anticipated in human patients produced adverse effects on tissues with rapidly proliferating cells in rats ands dogs. Adverse effects on other target organs were also noted in rats. Definitive evaluation of potential effects of clofarabine on QT interval and ECGs in general will have to be solely derived from the results of clinical studies.

Genotoxicity

Consistent with its mechanism of action, clofarabine is genotoxic. Clofarabine tested negative in an in vitro bacterial mutagenicity study but was clastogenic in a mammalian in vitro assay and was positive in vivo in the rat micronucleus study.

Carcinogenicity

Carcinogenicity studies have not been conducted.

Reproductive and Developmental Toxicity

No fertility studies were conducted. However, effects on fertility would be anticipated based on pathologic effects on testes and ovaries in the general animal toxicology studies. Clofarabine was teratogenic in embryo-foetal development studies conducted in pregnant rats and rabbits. Thus, clofarabine administration would pose a clear developmental risk to the conceptus and foetus. Appropriate warnings have been included in the Product Monograph.

3.2.4 Conclusion

Data derived from the non-clinical studies adequately address the requirements for a new active substance non-clinical evaluation. Overall, the pharamacology and toxicology studies support the use of Clolar for the proposed indication.

3.3 Clinical basis for decision

3.3.1 Pharmacodynamics

Inhibition of DNA synthesis in leukaemia blast cells was measured in 4 patients who received doses of 11.25, 15, 40, or 52 mg/m2 clofarabine. Generally, DNA synthesis rapidly declined, however a 10-50% recovery was observed 24-hours later. The recovery of DNA synthesis appeared to be related to the clofarabine dose administered. At the maximum tolerated dose (52 mg/m2), the inhibition of DNA synthesis was maintained throughout therapy. In another study, administration of 52 mg/m2 clofarabine resulted in a significant, rapid reduction of peripheral leukaemia cells in 94% (31/33) of paediatric ALL patients who had measurable absolute blast counts.

A study was conducted to determine whether or not the inhibition of DNA synthesis is dose-dependent. At a dose of 22.5 mg/m2/day, a 60% decrease in DNA synthesis was observed after 3-4 days treatment followed by a partial recovery period before the second dose of clofarabine. At 30 mg/m2/day, the inhibition of DNA synthesis was almost complete but not sustainable. Only at 55 mg/m2/day, was the inhibition of DNA synthesis completely sustainable.

3.3.2 Pharmacokinetics

Absorption

The PK of clofarabine were studied in 22 ALL paediatric patients who received daily administration of 52 mg/m2 clofarabine over a period of 2 hours by IV infusion for 5 consecutive days. Maximum plasma clofarabine concentrations were generally achieved by the end of the infusion, ranging from 141-838 ng/mL on Day 1 and 214-1390 ng/mL on Day 5. Plasma concentrations appeared to decline in a biphasic manner. Little accumulation was seen on Day 5 compared to Day 1.

The maximum plasma concentration (Cmax) was inversely proportional to the patient's body weight. Longer infusion times should be considered for smaller children with a body weight <20 kg.

Distribution

Clofarabine was only 47% bound to human plasma proteins, predominantly to albumin. Clofarabine showed high tissue distribution. Volume of distribution at steady-state was estimated to be 122 L/m2 in patients with ALL. Minimum accumulation was seen with once-daily dosing in the clinical studies.

Metabolism

Clofarabine is a prodrug and exerts its activity through its active metabolites. Clofarabine is transported into target cells by both diffusion and facilitated diffusion. Intracellular clofarabine is mono-phosphorylated by deoxycytidine kinase and then serially phosphorylated by other kinases to form clofarabine triphosphate, the active moiety. The metabolic pathways for clofarabine triphosphate remain unknown.

In vitro studies in isolated human hepatocytes showed that clofarabine does not induce hepatic enzymes nor does clofarabine inhibit CYP. In isolated human hepatocytes, clofarabine showed very limited metabolism suggesting that CYP inhibitors and inducers are unlikely to affect the metabolism of clofarabine.

Excretion

The major route of clofarabine elimination is by renal excretion of unchanged drug. An average of 58% of the dose was excreted within 24 hours after a single-dose administration in paediatric patients with ALL. The mean renal clearance was 15.1 L/hour which, after correction for protein binding, was much greater than the glomerular filtration rate in humans. Hence, clofarabine showed evidence of filtration and tubular secretion as kidney elimination mechanisms.

3.3.3 Clinical Efficacy

The clinical efficacy of Clolar was evaluated in two Phase II, open-label, non-randomized, single-arm studies in paediatric patients who experienced multiple relapses or were refractory to front-line therapy.

The pivotal, Phase II study (Study CLO-212) was conducted in 61 paediatric patients (≤21 year of age at initial diagnosis) who were previously treated with a median of 3 prior induction regimens. Patients received a dose of 52 mg/m2 Clolar over 2 hours for 5 consecutive days repeated every 2 to 6 weeks for up to 12 cycles (median 2 cycles). Approximately one-third of the patients received at least 1 prior transplantation, and 57% were refractory to the last therapeutic regimen. The primary endpoint was the overall remission rate determined by the sum of the number of patients with either a CR or CRp. Secondary endpoints included overall survival, and duration of remission. Twelve of 61 ALL patients achieved CR or CRp. The overall remission rate was 20%. Of those who were refractory to their most recent prior regimen, the overall remission rate was 17%. Responses were observed in patients with both T-cell and B-cell lineage ALL, and in patients who had a prior haematopoietic stem cell transplant (HSCT). A total of 31% patients achieved at least a partial response. Ten of the 61 ALL patients proceeded to HSCT, with no apparent increase in transplantation-related toxicity. The median survival was 12.9 weeks in all patients. The median overall survival was 66.6 weeks in patients who achieved a CR or CRp.

The other Phase II study (Study BIOV-111) was conducted in 71 paediatric patients (≤21 year of age at initial diagnosis) who failed to achieve primary response on conventional treatment regimens, or failed to achieve a response at first relapse. Patients were also eligible at second and subsequent relapse. Patients received a dose of 52 mg/m2 Clolar over 2 hours for 5 consecutive days, repeated every 21±7 days, for up to 12 cycles. The 65 patients who received one full 5-day course of Clolar were included in the primary efficacy analyses. The primary endpoint was the overall remission rate, same as the study above. Additional study endpoints included partial response, evaluation of the duration of remission, overall survival, the number of patients proceeding to HSCT, and the time to bone marrow transplantation. The overall remission rate was 23.1% and 31.3% in patients who completed 1 and 2 treatment cycles, respectively. A CR was observed in 3 (4.6%) patients, two of whom received a bone marrow or stem cell transplant following clofarabine treatment. Seven (9.9%) of the patients received a bone marrow transplant or peripheral blood stem cell transplant after the first dose of Clolar. Median survival among transplant recipients was 358 days. For three patients, the CR survival times were censored at 358, 941, and 954 days. Although the study was uncontrolled, the overall remission rate, transplant rate, overall survival, and survival among patients with CR indicate that Clolar treatment has a clinically important effect in this patient population.

3.3.4 Clinical Safety

The safety data for 115 paediatric patients, with relapsed or refractory ALL (n=70) or acute myeloid leukemia (AML, n=45), who received at least one infusion of Clolar at 52 mg/m2 were combined into an integrated database. The analysis of data identified groups of AEs that characterize the safety profile of Clolar in this population. Foremost were haematologic toxicities and infections resulting from the immunosuppressive effect of Clolar. Other significant groups of AEs observed for Clolar were found in the hepatobiliary, renal, and gastrointestinal organ systems. In addition, 11 events of tumour lysis syndrome, capillary leak syndrome or systemic inflammatory response syndrome were reported. The most common toxicities observed during exposure to Clolar were gastrointestinal system AEs (including vomiting, nausea, and diarrhoea), adverse haematologic effects (including anaemia, leukopaenia, thrombocytopenia, neutropaenia, and febrile neutropaenia), and infection.

The safety profile of Clolar is difficult to delineate considering the complicated medical history presented by most patients in the integrated safety database. Many factors could potentially affect the interpretation and assignment of causality for AEs including underlying infection, concomitant medications, progressive disease, and organ damage associated with prior high-dose chemotherapy and total body irradiation for transplant. Due to prolonged immuno- and myelosuppression from prior therapies, this population was susceptible to a wide range of illnesses, particularly fungal or bacterial infections. Most patients entered a study with at least one concurrent infection and the majority of patients experienced at least one infection during treatment with Clolar.

Haematologic toxicity was frequently observed during exposure to Clolar. Virtually all patients had pre-existing haematologic and chemistry abnormalities. These toxicities included the onset or worsening of anaemia, leukopaenia, thrombocytopenia, neutropaenia, and febrile neutropaenia. Febrile neutropaenia was the most common grade 3 AE considered related to the study drug. Fifty percent of the patients in the integrated safety database reported a serious adverse event (SAE) of febrile neutropaenia.

The majority (83%) of patients experienced at least one infection during treatment with Clolar. Post-baseline infections were generally grade 3 and included cellulitis, bacteraemia, Herpes simplex, Herpes zoster, catheter-related infection, clostridium colitis, and pneumonia. There was a smaller incidence of grade 4 infections (10/95 patients) observed in the safety population. These infections included bacterial sepsis, enterococcal infection, Escherichia sepsis, fungal infection, fungal sepsis, pneumonia fungal, pneumonia, septic shock, sepsis, Staphylococcal bacteraemia, Staphylococcal sepsis, systemic mycosis and varicella. Infections that were considered to be a contributing factor in the deaths of 8 patients were: sepsis, septic shock, and pneumoniae. However, only one grade 5 infection (septic shock) was considered related to Clolar.

The liver was known to be a target organ of toxicity based on earlier non-clinical and Phase I data. Approximately 25% of patients experienced hepatobiliary AEs during treatment with Clolar. The most frequent hepatobiliary AEs observed included jaundice and hepatomegaly. Three patients experienced hepatic AEs considered related to study drug resulting in discontinuation for one patient and the deaths of two patients.

Although few patients entered the study with elevated AST or ALT, the incidence of grade 3 or 4 AST or ALT following Clolar treatment ranged from 36% to 44%, respectively. A total of 13.2% of patients reported grade 3 or 4 total bilirubin post-baseline. Almost half of the patients overall experienced the onset or worsening of elevated total bilirubin during treatment with Clolar.

Adverse events in the renal system organ class (SOC) are significant because clofarabine is known to be eliminated by the kidney via glomerular filtration and tubular secretion. Approximately one third of the patients reported an AE in the renal and urinary system. Overall, a total of 53% (61/115) of the patients experienced the onset of or worsening of elevated creatinine. The most frequently reported renal AE was haematuria which may reflect the thrombocytopenia that was common in many of these patients. Nephrotoxic medications, tumour lysis, and tumour lysis with hyperuricaemia may contribute to renal toxicity.

A majority of the paediatric patients in the integrated database reported at least one AE in the gastrointestinal disorders SOC, and the AEs most frequently observed were vomiting and nausea. Other frequent gastrointestinal AEs were diarrhoea, abdominal pain and constipation. Most of these events were grade 1 or 2 in severity and rarely met the criteria for an SAE.

A capillary leak syndrome was reported as a side effect of Clolar in several patients. It may result from the release of cytokines and may also reflect either direct or indirect damage to endothelial cells. However, underlying medical conditions, (e.g., fungal infections, effusions, and cardiovascular abnormalities), toxicities from prior therapies, infections, or disease progression, may also cause a capillary leak-like syndrome. The signs and symptoms of cytokine release (e.g., tachypnaea, tachycardia, hypotension, pulmonary eodema) could progress to systemic inflammatory response syndrome (SIRS), capillary leak syndrome and multi-organ dysfunction.

Forty-nine percent of patients experienced at least one cardiac AE. Since some cardiotoxicity had been observed in the non-clinical rat studies, a paediatric cardiologist reviewed all of the echocardiograms (ECHOs). The cardiologist concluded that direct cardiotoxicity of clofarabine could not be completely ruled out, but observed that patients who experienced the majority of cardiac function changes were frequently ill from disease progression, sepsis, or other substantial adverse medical conditions when the ECHOs were performed.

Thirty-three of 115 patients (29%) reported an AE of hypotension. Five events (four Grade 3 and one Grade 4) were Clolar-related. The occurrence of treatment-emergent hypotension was usually associated with severe or systemic infections and pre-existing medical conditions.

Safety data from Study BIOV-111 were not included with the integrated dataset. The spectrum of AEs seen in this study was consistent with the known safety profile of Clolar, and did not give rise to any new concerns specific to the use of Clolar in a paediatric population. The safety profile of Clolar is also supported by the post-market safety profile of approximately 5000 patients since the drug was first marketed in the United States in December 2004. Considering the concordance of AE profiles across Study BIOV-111, the integrated safety summary, and in the post-marketing setting, Clolar demonstrated an acceptable safety profile in heavily pre-treated paediatric patients with poor prognosis.

3.4 Benefit/Risk Assessment and Recommendation

3.4.1 Benefit/Risk Assessment

Randomised trials demonstrating increased survival or other clinical benefit have not been conducted with Clolar. Paediatric Phase II clinical trials demonstrated that Clolar treatment was able to produce a beneficial response in some ALL patients when several other options have failed. Clolar was shown to facilitate transplant and enable survival up to nearly 3 years in a small proportion of these patients who have few remaining alternative options.

The most frequent AEs with Clolar are haematologic toxicities and infections. Other significant AEs for Clolar were observed in the hepatobiliary, renal, and gastrointestinal organ systems. The safety profile of Clolar is considered to be acceptable and predictable in this population of heavily pre-treated multiple relapsed or refractory generally immunocompromised patients with numerous significant concurrent conditions and concomitant medications. Considering the poor prognosis of these heavily pre-treated patients, the benefit-risk relationship for Clolar in the treatment of pediatric ALL is considered favourable.

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 Clolar is favourable for the treatment of paediatric patients 1 to 21 years old with relapsed or refractory ALL after at least two prior regimens. The New Drug Submission (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 (NOC) pursuant to section C.08.004 of the Food and Drug Regulations.

4 Submission Milestones

Submission Milestones: Clolar®

Submission MilestoneDate
Pre-submission meeting:2008-02-07
Submission filed:2008-05-08
Screening 1
Sreening Deficiency Notice issued:2008-06-26
Response filed:2008-08-06
Screening Acceptance Letter issued2008-09-19
Review
Quality Evaluation complete2009-07-16
Clinical Evaluation complete2009-05-01
Labelling Review complete2009-06-18
Notice of Compliance issued by Director General2009-07-16