Summary Basis of Decision for Mozobil ™

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
MozobilTM

Plerixafor, 20 mg/mL, Solution, Subcutaneous

Genzyme Canada Inc.

Submission control no: 142638

Date issued: 2012-04-26

Foreword

Health Canada's Summary Basis of Decision (SBD) documents outline the scientific and regulatory considerations that factor into Health Canada regulatory decisions related to drugs and medical devices. SBDs are written in technical language for stakeholders interested in product-specific Health Canada decisions, and are a direct reflection of observations detailed within the evaluation reports. As such, SBDs are intended to complement and not duplicate information provided within the Product Monograph.

Readers are encouraged to consult the 'Reader's Guide to the Summary Basis of Decision - Drugs' to assist with interpretation of terms and acronyms referred to herein. In addition, a brief overview of the drug submission review process is provided in the Fact Sheet entitled 'How Drugs are Reviewed in Canada'. This Fact Sheet describes the factors considered by Health Canada during the review and authorization process of a drug submission. Readers should also consult the 'Summary Basis of Decision Initiative - Frequently Asked Questions' document.

The SBD reflects the information available to Health Canada regulators at the time a decision has been rendered. Subsequent submissions reviewed for additional uses will not be captured under Phase I of the SBD implementation strategy. For up-to-date information on a particular product, readers should refer to the most recent Product Monograph for a product. Health Canada provides information related to post-market warnings or advisories as a result of adverse events (AE).

For further information on a particular product, readers may also access websites of other regulatory jurisdictions. The information received in support of a Canadian drug submission may not be identical to that received by other jurisdictions.

Other Policies and Guidance

Readers should consult the Health Canada website for other drug policies and guidance documents. In particular, readers may wish to refer to the 'Management of Drug Submissions Guidance'.

1 Product and submission information

Brand name:

MozobilTM

Manufacturer/sponsor:

Genzyme Canada Inc.

Medicinal ingredient:

Plerixafor

International non-proprietary Name:

Plerixafor

Strength:

20 mg/mL

Dosage form:

Solution

Route of administration:

Subcutaneous

Drug identification number(DIN):

  • 02377225

Therapeutic Classification:

Haematopoietic agent

Non-medicinal ingredients:

Sodium chloride, hydrochloric acid and sodium hydroxide for pH adjustment, and Water for Injection

Submission type and control no:

New Drug Submission, Control Number: 142638

Date of Submission:

2010-12-21

Date of authorization:

2011-12-08
MOZOBILTM is a trademark of Genzyme Corporation.
2 Notice of decision

On December 8, 2011, Health Canada issued a Notice of Compliance to Genzyme Canada Inc. for the drug product, Mozobil.

Mozobil contains the medicinal ingredient plerixafor which is a haematopoietic agent. Within the bone marrow, blood stem cells are “anchored” by binding of a receptor found on the stem cell surface with a protein located in the bone marrow microenvironment. Plerixafor blocks the receptor on the surface of blood stem cells which interferes with their “anchoring” in the bone marrow. Plerixafor improves the release of stem cells into the blood stream, allowing them to be collected by an apheresis machine, and then subsequently frozen and stored until transplantation.

Mozobil is indicated in combination with granulocyte-colony stimulating factor (G-CSF) to mobilize haematopoietic stem cells (HSCs) to the peripheral blood for collection and subsequent autologous transplantation in patients with non-Hodgkin’s lymphoma (NHL) and multiple myeloma (MM). Some patients with NHL and MM are able to meet minimal and target HSC collection criteria with G-CSF alone. Mozobil should only be administered under the supervision of a qualified health professional who is experienced in oncology and/or haematology, and in the management of cancer patients undergoing mobilization of haematopoietic stem cells to the peripheral blood.

The market authorization was based on quality, non-clinical, and clinical information submitted. The efficacy and safety of Mozobil in conjunction with G-CSF in NHL patients and MM patients were evaluated in two, multicentre, randomized, double-blind, parallel group, placebo-controlled studies (Studies 1 and 2). In Study 1, 59% of NHL patients who were mobilized with Mozobil and G-CSF collected ≥5 X 106 CD34+ cells/kg from the peripheral blood in four or fewer apheresis sessions, compared with 20% of patients who were mobilized with placebo and G-CSF. In Study 2, 72% of MM patients who were mobilized with Mozobil and G-CSF collected ≥6 X 106 CD34+ cells/kg from the peripheral blood in two or fewer apheresis sessions, compared with 34% of patients who were mobilized with placebo and G-CSF. In both studies, other CD34+ cell mobilization outcomes showed similar findings.

Mozobil (20 mg/mL plerixafor) is presented as a solution. Treatment with Mozobil begins after the patient has received G-CSF once daily for four days. The recommended dose of Mozobil is 0.24 mg/kg body weight by subcutaneous injection. Dosing guidelines are available in the Product Monograph.

Mozobil is contraindicated for patients who are hypersensitive to this drug or to any ingredient in the formulation or component of the container. Mozobil 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 Mozobil 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 Mozobil 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

Plerixafor, the medicinal ingredient of Mozobil, is a haematopoietic agent. Plerixafor is a bicyclam moiety that binds with high affinity to the human CXCR4 receptor and disrupts interactions with its cognate ligand SDF 1-alpha (CXCL12). Interruption of the CXCR4/SDF 1-alpha interaction results in mobilization of CD34+ haematopoietic stem cells (HSCs) to the peripheral blood where they can be collected for HSC transplantation.

Manufacturing Process and Process Controls

Plerixafor is manufactured via a multi-step synthesis. Each step of the manufacturing process is considered to be controlled within acceptable limits:

  • The sponsor has provided information on the quality and controls for all materials used in the manufacture of the drug substance.
  • The drug substance specifications were found to be satisfactory. Impurity limits meet International Conference on Harmonisation (ICH) requirements.
  • The processing steps have been evaluated and the appropriate ranges for process parameters have been established.
Characterization

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

Impurities and degradation products arising from manufacturing and/or storage were reported and characterized. These products were found to be within ICH-established limits and/or were qualified from toxicological studies and therefore, are considered to be 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 plerixafor.

The drug substance packaging is considered acceptable.

Stability

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

3.1.2 Drug Product

Description and Composition

Mozobil (plerixafor) is supplied as a sterile, preservative-free, clear, colourless to pale yellow, pH neutral, isotonic solution in a single-use 2.0 mL clear glass (Type I) vial, sealed with a rubber stopper and an aluminum seal with a plastic flip-off cap.

Each 2.0 mL vial is filled to deliver 1.2 mL of 20 mg/mL solution, containing 24.0 mg of plerixafor. Each 1.2 mL of the solution contains 5.9 mg sodium chloride in sterile Water for Injection adjusted to a pH of 6.0 to 7.5 with hydrochloric acid and sodium hydroxide, if required.

All non-medicinal ingredients (excipients) found in the drug product are acceptable for use in drugs according to the Food and Drug Regulations.

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

Mozobil is tested to verify that its identity, appearance, assay, pH, fill volume, particulate matter, sterility, osmolality, degradation products, drug-related impurities, and bacterial endotoxins are within acceptance criteria. The test specifications and analytical methods are considered acceptable; the shelf-life and release limits, for individual and total degradation products, are within acceptable limits.

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 submitted, the proposed 36-month shelf-life at 15-30°C for Mozobil is considered acceptable when packaged in the proposed container closure system.

3.1.3 Facilities and Equipment

The design, operations, and controls of the facilities and equipment that are involved in the production of Mozobil are considered suitable for the activities and products manufactured.

3.1.4 Adventitious Agents Safety Evaluation

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

3.1.5 Conclusion

The Chemistry and Manufacturing information submitted for Mozobil 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

A number of studies were carried out by the sponsor to determine the specificity of plerixafor in binding CXCR4 (fusin) and competing with its ligand, stromal derived factor 1-alpha (SDF 1-alpha or CXCL12). There are many chemokine receptors with overlapping functions and binding partners so it was not possible to exclude binding of plerixafor to all related receptors. 125I-ligand binding studies showed that plerixafor binds to CXCR4 but failed to interact with all other chemokine receptors studied in this assay. Functional studies demonstrated that plerixafor could only inhibit calcium influx (mediated by SDF 1-alpha) into CXCR4 expressing cells but not in cells expressing other chemokine receptors. This led to the initial observations that plerixafor had a high degree of specificity for CXCR4 and was a potent inhibitor of SDF 1-alpha function.

The ability of plerixafor to mobilize CD34+ stem cells in vivo was assessed in dogs. Two- to 4-fold increases in CD34+ peripheral blood levels were observed at 6-8 hours after a single 4 mg/kg subcutaneous dose. In dogs, the CD34+ cells mobilized successfully engrafted in a canine model of autologous stem cell transplantation. A second study in dogs demonstrated that stem cells mobilized by plerixafor could be used in allogeneic transplantations.

There exists the possibility that plerixafor also mobilizes tumour cells into the peripheral blood that could contaminate the apheresis product. No studies were carried out in animals to address this and the potential impact of this possibility on the long term efficacy of plerixafor has not been fully established.

The synergy between granulocyte-colony stimulating factor (G-CSF) and plerixafor in mobilizing progenitor CD34+ cells from the bone marrow to the blood was determined in mice. Plerixafor was shown to be a mobilizer of haematopoietic progenitor cells (HPC) in mice when administered subcutaneously (SC). Furthermore plerixafor was able to mobilize HPC in mice which are poorly mobilized by G-CSF. Plerixafor was shown to augment G-CSF-induced mobilisation in DBA/2, C57Bl6 and C3H/HEJ mice. The increase in colony forming units-granulocyte macrophage (CFU-GM) mobilization with G-CSF plus plerixafor over G-CSF alone was 5.5, 15.4, and 6.4 fold respectively. Long-term engraftment in bone marrow by plerixafor and G-CSF mobilized haematopoietic stem cells (HSC) was demonstrated following a primary transplant in congenic mice, and the self renewing properties of the mobilized HSC was demonstrated by a further secondary transplant.

The safety pharmacology program included in vitro and in vivo assays. Plerixafor at 5 μg/mL had moderate or strong binding affinity for a number of different receptors predominantly located on pre-synaptic nerve endings in the central and/or peripheral nervous system. There was no effect of plerixafor on the human Ether-a-go-go-Related Gene (hERG) potassium channel. Effects on the cardiovascular system were observed in anesthetized rats and conscious dogs at higher intravenous (IV) doses of plerixafor. These effects included a dramatic fall in mean arterial blood pressure, heart rate and myocardial contractility followed by death 2 to 5 minutes after the end of the infusion in rats and increased heart rate in dogs. There were no effects on electrocardiogram (ECG) tracings in dogs. Effects of SC administration of plerixafor on the cardiovascular system included decreases in mean arterial blood pressure, heart rate, and cardiac output in rats. In addition, a visual inspection of the ECG tracing showed that the P wave became flat, negative or undetectable in rats.

Functional observational battery testing was performed in mice and rats after a single SC administration. The effects observed included central nervous system (CNS) depressant-like signs when not stimulated and CNS stimulant-like effects when handled. The effects were noted shortly after dosing, but at 24 hours post-dose, animals were essentially normal at these doses.

3.2.2 Pharmacokinetics

Absorption

Following single SC doses of radiolabelled plerixafor in rats, peak plasma concentrations of radioactivity occurred approximately 1 hour post injection. At the beginning, the plasma concentrations decreased rapidly with a mean half-life of distribution of 1.1 ± 0.1 hours and then slowly with a mean half-life of terminal elimination of 57 ± 9 hours. The absorption was estimated to be approximately 85% based on comparisons to previous IV injections of the drug. The pharmacokinetic (PK) parameters following SC administration were consistent across mice, rats, and dogs where the half-life (first elimination phase) was 0.75, 0.9-1.16, and 1.58 hours, respectively.

The time to reach maximum plasma concentration occurred at 0.5 hours after single and multiple SC doses in rats. The multiple dose study showed that the systemic exposure [area under the curve (AUC)] of plerixafor increased over time up to 7 days and that the tissues retained a significant amount of the drug up to 168 hours following the last dose.

Distribution

In studies conducted with radiolabelled plerixafor in rats, the highest levels of radioactivity were detected in the kidney, injection site, liver, cartilage, and spleen (red pulp). Whole-body autoradiography images showed that plerixafor could cross the blood-brain barrier (limited distribution) which could explain CNS-related effects of the drug in animals and humans.

Metabolism

Plerixafor undergoes very little, if any, metabolism by cytochrome P450 (CYP) enzymes. The study medication remained relatively unchanged following incubation with microsomes from mouse, rat, dog, or human. In addition, plerixafor did not inhibit metabolism of substrates for CYP3A4, CYP2D6, CYP2B6, CYP2C8, CYP2A6, CYP2E1, CYP2C9, CYP2C10 or CYP1A2 when compared to known positive control inhibitors. Additional experiments demonstrated that plerixafor also did not induce the expression of CYP2B6 or CYP1A2 messenger ribonucleic acid (mRNA).

Excretion

Plerixafor was excreted mainly via the kidneys. Approximately two-thirds of a single radioactive dose of plerixafor was excreted into the urine by 144 and 168 hours post administration. The majority was excreted in the first 24 hours compared to more gradual excretion in the faeces which accounts for less than 10% of the administered dose.

3.2.3 Toxicology

The overall program of study types for the acute and repeat-dose general toxicology, genetic and reproductive toxicology, and local irritancy studies performed for plerixafor was appropriate to support the proposed therapeutic route and duration of administration in humans. Compliant with regulatory guidelines for drugs intended for dosing clinically for up to 2 weeks, the duration of repeat-dose toxicity studies was 4 weeks in rodents and non-rodents. Should the duration of clinical dosing increase the requirements for longer duration dosing supporting non-clinical safety studies will have to be revisited.

Single-Dose Toxicity

The acute toxicity was generally similar in rats and mice. A single IV dose of 5 or 8 mg/kg was lethal and 2 mg/kg resulted in transient sedation. After SC administration, doses of ≥40 mg/kg and ≥14 mg/kg were lethal in rats and mice, respectively. In pigs, SC doses of 1, 2, 4, 6, 8 and 12 mg/kg were administered. A dose of 12 mg/kg resulted in rapid death (20 minutes post dose - cause not determined), while at 8 mg/kg the pig was observed to be in lateral recumbency between 1-3 hours and normal thereafter. Doses of 6 mg/kg and below were well tolerated. Clinical signs at these doses included lateral recumbency in mice, rats, and pig; and sedation, ventral or lateral recumbency, dyspnoea, uncoordinated movement, spasms, and/or ruffled fur in mice and rats.

Repeat-Dose Toxicity

At non-lethal doses, daily SC doses with plerixafor was associated with gastrointestinal (GI) clinical signs in dogs (diarrhoea, emesis, increased defaecation) and neurological signs in dogs and rats (sedation, tremors, spasms, twitching, recumbency and ataxia and mydriasis). There were some minor decreases in body weight gain and food consumption. Increases in white blood cell counts (predominantly neutrophils), and decreases in serum magnesium and increases in urinary calcium and/or magnesium were noted in both rats and dogs. There were increases in extramedullary haematopoiesis in the liver of rats and dogs, and the spleen in rats only. The extramedullary haematopoiesis was considered related to the mechanism of action. In addition, there was lymphoid atrophy in the thymus of rats at high doses. Injection site reactions (subcutaneous haemorrhage and inflammation) were noted at higher doses in rats.

One study assessed the effects of plerixafor on bone in rats. At dose levels of 11.4 mg/kg and 15.2 mg/kg, there were dose-dependent reductions in bone mass, volume, and radiodensity but no effect on the longitudinal growth. Histological examination did not reveal any abnormalities in the growth plate, primary or secondary spongiosa, or cortical bone. There were no effects on bone histopathology in any study and it was suggested that the effects on bone mass, volume, and radiodensity were related to mild reductions in food consumption and slight decreases in weight gain in this study. However plerixafor chelates calcium and an effect of plerixafor-induced calcium loss in this process cannot be ruled out.

In summary, the no adverse effect dose levels (NOAELs) in the pivotal 4-week studies were 0.6 -1.2 mg/kg/day in rats and 0.25-0.30 mg/kg/day in dogs. These NOAELs are in the same order of magnitude as the clinical dose (0.24 mg/kg). Exposures (AUCs) at these doses were 0.1 to 5 times the clinical exposure. At multiples of clinical doses, there were effects related to extended pharmacology (increases in white blood cell counts, increased extracellular haematopoiesis in liver and spleen) and effects on electrolytes (decreased serum concentrations and/or increased urinary excretion of calcium and magnesium). Diarrhoea and increases in heart rate were noted in dogs at ≥1 mg/kg. There were no effects on ECG intervals (when corrected for heart rate) or morphology. In rats, the maximum tolerated dose (MTD) was 7.6-24 mg/kg/day and in dogs the MTD was 4-8 mg/kg/day. At MTD, exposures (AUCs) were 7 to 18 times the clinical exposure. The nature of these findings and margins to MTD are sufficient to support the exposures at the human maximal recommended dose and dosing regimen.

In summary, the no adverse effect dose levels (NOAELs) in the pivotal 4-week studies were 0.6 -1.2 mg/kg/day in rats and 0.25-0.30 mg/kg/day in dogs. These NOAELs are in the same order of magnitude as the clinical dose (0.24 mg/kg). Exposures (AUCs) at these doses were 0.1 to 5 times the clinical exposure. At multiples of clinical doses, there were effects related to extended pharmacology (increases in white blood cell counts, increased extracellular haematopoiesis in liver and spleen) and effects on electrolytes (decreased serum concentrations and/or increased urinary excretion of calcium and magnesium). Diarrhoea and increases in heart rate were noted in dogs at ≥1 mg/kg. There were no effects on ECG intervals (when corrected for heart rate) or morphology. In rats, the maximum tolerated dose (MTD) was 7.6-24 mg/kg/day and in dogs the MTD was 4-8 mg/kg/day. At MTD, exposures (AUCs) were 7 to 18 times the clinical exposure. The nature of these findings and margins to MTD are sufficient to support the exposures at the human maximal recommended dose and dosing regimen.

Genotoxicity

Plerixafor was not mutagenic in the bacterial Ames test and not clastogenic in the chromosome aberration test in V79 Chinese hamster cells in the presence or absence of rat liver S9-mix. Subcutaneously administered plerixafor up to 25 mg/kg did not induce bone marrow toxicity and did not induce micronucleus formation.

Carcinogenicity

Carcinogenicity studies with plerixafor were not conducted. The duration of dosing and indication obviate the need for carcinogenicity studies.

Reproductive and Developmental Toxicity

In rats and rabbits, embryofoetal toxicities and maternal toxicities were seen. Plerixafor caused dose-dependent embryotoxicity (increased resorption and post implantation loss in rats and rabbits, decreased foetal weight in rats, and decreased litter size in rabbits), as well as foetal toxicity (external, visceral and head malformations and/or variations in rats, and external malformation in rabbits). The effects are probably mechanism based, as the CXC chemokine ligand 12 [CXCL12; stromal cell-derived factor 1 (SDF-1)] signals through its cognate receptor CXCR4 and plays an essential role not only in haematopoiesis, but also in organogenesis. Maternal toxicities in the rat included deficits in corrected gestation weight gain and decreased food consumption and in the rabbit included mortality, body weight loss, decreased food consumption and clinical signs.

Local Tolerance

In dedicated local tolerance studies, the clinical formulation was slightly irritating in rabbits. In the repeat-dose studies, histopathologic changes in animals at the SC injection site at NOAEL doses (at or slightly higher than clinical doses) were similar to vehicle controls.

3.2.4 Summary and Conclusion

The non-clinical pharmacology and toxicology studies for this drug submission are considered suitable. The non-clinical pharmacodynamic data support an important role for plerixafor in mobilizing CD34+ cells by antagonizing CXCR4 and SDF 1-alpha interactions. The overall programme and outcome of acute and repeat-dose general toxicology, genetic and reproductive toxicology, and local irritancy studies performed for plerixafor was appropriate to support the proposed therapeutic route and duration of administration in humans.

3.3 Clinical basis for decision

3.3.1 Pharmacodynamics

The effects of plerixafor on CD34+ mobilization was determined in healthy subjects and oncology patients.

In healthy subjects, the administration of a single dose of plerixafor (0.04 to 0.24 mg/kg) with no G-CSF led to dose-proportional increases from baseline of peripheral blood (PB) CD34+ cell counts, while the response to the 0.32 mg/kg dose was similar to that obtained with the 0.16 mg/kg dose. The pharmacodynamic response to plerixafor 0.24 mg/kg (no G-CSF) in healthy subjects occurred 6 to 10 hours after dosing. The median peak fold-increase was 15.8 over baseline.

Following 4 days of pre-treatment with G-CSF in healthy volunteers, the administration of plerixafor (0.24 mg/kg) in conjunction with G-CSF produced a sustained elevation in the PB CD34+ cell counts from 4 to 18 hours after plerixafor administration, with a peak response between 10 to 14 hours. The administration of a lower dose of plerixafor (0.16 mg/kg) in conjunction with G-CSF produced higher peak PB CD34+ cell counts compared to the treatment with either plerixafor (0.16 mg/kg) alone or G-CSF alone. These results showed a 3.8-fold increase, versus a 3.2-fold increase, versus a 1.2-fold increase, respectively, over the baseline response achieved on Day 4 with G-CSF alone.

In combination with G-CSF, patients with multiple myeloma (MM) in general, had higher responses than patients with non-Hodgkin’s lymphoma (NHL) in mobilizing CD34+ cells. In the NHL group, patients with higher baseline concentrations of PB CD34+ cell counts had better responses than those with lower baseline PB CD34+ cell counts. Plerixafor increased the PB CD34+ count by 3- to 6-fold over the pre-plerixafor dose level after the first injection, which was similar to the 3- to 4-fold increase observed in healthy subjects.

Cumulatively, pharmacodynamic studies showed that in healthy subjects, the plerixafor dose of 0.24 mg/kg elicited a higher and later peak response compared with the 0.16 mg/kg dose. The increase in PB CD34+ cells with plerixafor following 4 days of pre-treatment with G-CSF was higher than with plerixafor or G-CSF alone. When added to a dosing regimen of G-CSF in healthy subjects, 0.16 mg/kg and 0.24 mg/kg plerixafor had similar magnitudes of fold-increases in PB CD34+ cells. In patients with MM and NHL, the 0.24 mg/kg dose with G-CSF elicited a greater response (greater fold-increase in apheresis yields) than the 0.16 mg/kg dose with G-CSF. Based upon the above data and given the difference in response rates of patients with MM and NHL, the recommended dose of Mozobil (plerixafor injection) is 0.24 mg/kg body weight by SC injection.

3.3.2 Pharmacokinetics

Absorption

Plerixafor was rapidly absorbed following SC injection. Maximum plasma concentrations occurred in approximately 30-60 minutes.

Distribution

The apparent volume of distribution of plerixafor in humans is 0.3 L/kg, suggesting that plerixafor is largely confined to, but not limited to, the extravascular fluid space.

Plerixafor was moderately bound to human plasma proteins; up to 58%.

Metabolism

Plerixafor was not metabolized in vitro using human liver microsomes or human primary hepatocytes and did not exhibit inhibitory activity in vitro towards the major drug metabolizing CYP450 enzymes (1A2, 2A6, 2B6, 2C8, 2C9, 2C19, 2D6, 2E1, and 3A4/5). In the in vitro studies with human hepatocytes, plerixafor did not induce CYP1A2, CYP2B6, and CYP3A4 enzymes. These findings suggest that plerixafor has a low potential for involvement in P450-dependent drug-drug interactions.

Excretion

Renal excretion was the primary route of elimination. Following a 0.24 mg/kg dose in healthy volunteers with normal renal function, approximately 70% of the dose was excreted in the urine as the parent drug during the first 24 hours following administration. The median terminal half-life in plasma was 4.6 hours.

Special Populations and Conditions
Renal Impairment

Renal impairment did not affect the maximum plasma levels of plerixafor (Cmax) but did increase exposure due to impaired clearance in patients with varying degrees of renal impairment.

A population pharmacokinetic analysis simulated the effect of creatinine clearance (CrCl), as determined by the Cockcroft-Gault formula, on the plasma clearance of plerixafor. These results support a dose reduction to 0.16 mg/kg in patients with a CrCl of 20-50 mL/min to reduce the increased exposure in these patients comparable to patients with a CrCl >50 mL/min receiving a 0.24 mg/kg dose of Mozobil. There are insufficient clinical data to make dosing recommendations for patients with a creatinine clearance <20 mL/min or for patients on dialysis. Tissue accumulation of plerixafor in patients with renal impairment was not studied.

Hepatic Impairment

Studies in patients with hepatic impairment have not been conducted. Plerixafor is not metabolized by the liver, and there is no recommendation for dose adjustment.

Paediatrics

The safety and efficacy of Mozobil in paediatric patients has not been established in controlled clinical studies; approximately 20 patients 8-18 years of age have been treated in compassionate use programs. As certain types of NHLs occur in children, a paediatric study is being conducted.

Pregnant Women

Use of Mozobil is not recommended during pregnancy due to its mechanism of action and results of reproductive toxicology studies. Because CXCR4 plays an essential role in foetal development and plerixafor is a selective antagonist of CXCR4, Mozobil is suggested to cause congenital malformations when administered during pregnancy. Studies in animals have shown teratogenicity. There are no adequate and well-controlled studies in pregnant women using Mozobil. Women of childbearing potential are advised to use effective contraception during treatment.

3.3.3 Clinical Efficacy

Data from a total of 345 adult oncology patients [165 NHL, 158 MM, and 22 Hodgkin’s lymphoma (HL)] treated with G-CSF in combination with Mozobil in two Phase II studies and two Phase III studies [Study 1 (NHL) and Study 2 (MM)] were used to demonstrate the efficacy of Mozobil.

The two pivotal Phase III studies were multi-centre, randomized, double-blind, parallel group, placebo-controlled studies. On the evening of Day 4 of daily morning doses of G-CSF 10 μg/kg, the first dose of assigned study treatment, either Mozobil 0.24 mg/kg or placebo was administered. On Day 5, patients received a morning dose of G-CSF 10 μg/kg and underwent apheresis approximately 10 to 11 hours after the first dose of study treatment (within 60 minutes after administration of G-CSF). Patients continued to receive an evening dose of study treatment followed the next day by a morning dose of G-CSF and apheresis for up to a maximum of 4 aphereses or until the target collection of CD34+ haematopoietic stem cells (HSCs) was achieved.

In both studies, patients who failed to collect ≥0.8 times (x) 106 CD34+ cells/kg after 2 days of apheresis or at least 2 x 106 CD34+ cells/kg in 4 or fewer days of apheresis had the option of entering an open-label rescue procedure. After a minimum 7-day rest period, they received another 4-day course of G-CSF followed by a course of Mozobil (0.24 mg/kg) in combination with G-CSF for repeat mobilization.

Following the last apheresis, patients underwent a rest period, then pre-transplant ablative chemotherapy followed by autologous transplantation within 5 weeks after the last apheresis. Transplantation was performed according to standard of care at each study centre.

Patients received G-CSF (5 μg/kg once daily) beginning on the fifth or sixth day after transplantation and continuing until the absolute neutrophil count (ANC) was ≥0.5 x 109/L for 3 days or ≥1.0 x 109/L for 1 day. Platelet (PLT) engraftment was defined as a PLT count ≥20 x 109/L without transfusion for the preceding 7 days.

Graft durability was assessed at 100 days, 6 months, and 12 months post-transplantation.

The primary objective of Study 1 was to determine if NHL patients mobilized with G-CSF plus Mozobil 0.24 mg/kg were more likely to achieve a target number of ≥5 x 106 CD34+ cells/kg in 4 or fewer days of apheresis than NHL patients mobilized with G-CSF plus placebo. The primary objective of Study 2 was to determine if MM patients mobilized with G-CSF plus Mozobil 0.24 mg/kg were more likely to achieve a target number of ≥6 x 106 CD34+ cells/kg in 2 or fewer days of apheresis than MM patients mobilized with G-CSF plus placebo.

Secondary efficacy objectives common to both studies were to compare the two treatment arms with respect to the number of patients who achieved a minimum of 2 x 106 CD34+ cells/kg (minimum number required for transplantation) in 4 or fewer days of apheresis, the number of days of apheresis required to reach target cell numbers, the time to engraftment of polymorphonuclear leukocytes (PMNs) and PLTs, and the durability of the graft at pre-specified times post-transplantation. A secondary objective unique to Study 2 was to compare the two treatment arms with respect to the number of MM patients who achieved the target number of cells in 4 or fewer apheresis days.

G-CSF plus Mozobil was a superior mobilizing regimen compared to G-CSF plus placebo in NHL and MM, as demonstrated in the two Phase III studies. There was achievement of statistically significant target HSC collections (primary efficacy endpoint) as follows: ≥5 x 106 CD34+ cells/kg in 4 or fewer days of apheresis in NHL patients [59.3% versus (vs.) 19.6%, G-CSF plus Mozobil vs. G-CSF plus placebo, respectively, (p <0.001)]; and ≥6 x 106 CD34+ cells/kg in 2 or fewer days of apheresis in MM patients [71.6% vs.  34.4%, G-CSF plus Mozobil vs. G-CSF plus placebo, respectively (p <0.001)]. An additional secondary efficacy endpoint in MM patients demonstrated a statistically significant achievement of target HSC collection in 4 or fewer days of apheresis [75.7% vs. 51.3%, G-CSF plus Mozobil vs. G-CSF plus placebo, respectively (p <0.001)].

Additional benefits were seen in the results of the secondary efficacy endpoints. There was a statistically significant achievement of the minimum HSC required for autologous transplantation (≥2 x 106 CD34+ cells/kg) in 4 or fewer days of apheresis in both NHL patients [86.7% vs. 47.3%, G-CSF plus Mozobil vs. G-CSF plus placebo, respectively (p <0.001)] and MM patients [95.3% vs. 88.3%, G-CSF plus Mozobil vs. G-CSF plus placebo, respectively (p = 0.031)]. Target HSC collections were all achieved in fewer days with G-CSF plus Mozobil compared with G-CSF plus placebo (median number of days to reach target HSC collection was 3 days for the plerixafor group and not evaluable for the placebo group in Study 1 and 1 day for the plerixafor group compared with 4 days for the placebo group in Study 2). For transplanted patients, time to neutrophil engraftment (10-11 days) and platelet engraftment (18-20 days) were similar across the treatment groups. Based on an adjusted analysis which used laboratory measurements and clinical criteria to assess graft durability, results were similar in both treatment groups. For transplanted patients, the frequency of graft failure was low in the Phase III studies; three events in Mozobil-treated NHL patients and none in MM patients. None of these graft failures were considered by the investigator as related to Mozobil.

Some patients with NHL and MM met minimal and target HSC collection criteria with G-CSF alone, thus avoiding any additional toxicity that might result from the addition of Mozobil. In Study 1 (NHL), 47.3% of G-CSF plus placebo patients achieved minimum CD34+ stem cell count in 4 or fewer days of apheresis and 19.6% of G-CSF plus placebo patients achieved target CD34+ stem cell count in 4 or fewer days of apheresis, while in Study 2 (MM), 88.3% of G-CSF plus placebo patients achieved minimum CD34+ stem cell count in 4 or fewer days of apheresis and 55.9% of G-CSF plus placebo patients achieved target CD34+ stem cell count in 4 or fewer days of apheresis.

Overall survival (OS) or other clinical benefit endpoints were not used to demonstrate efficacy in the Phase III studies. Information required for progression- or relapse-free survival analyses was not captured. As an exploratory endpoint, OS data were collected and summarized for patients who went to transplantation. The OS data at 12 months post-transplantation were similar in both treatment groups in both pivotal studies.

Patients with NHL received considerable doses of prior cytotoxic chemotherapy which may have negatively impacted HSC mobilisation and collection. In Study 1, 52 NHL patients in the G-CSF plus placebo group entered into the rescue procedure with Mozobil and G-CSF. Of these patients, 60% (31 out of 52) mobilized ≥2 x106/kg CD34+ cells and had successful engraftment. In Study 2, 7 MM patients in the G-CSF plus placebo group entered the rescue procedure, all of whom mobilized ≥2 x106/kg CD34+ cells and had successful engraftment.

There were insufficient data to support use of G-CSF plus Mozobil with rituximab in NHL or use of G-CSF plus Mozobil in the indication of tandem transplantation in MM.

Patients with HL were not included in pivotal Study 1. Efficacy results from a supportive uncontrolled open-label Phase II study are promising but limited and did not support inclusion of HL in the indication.

Mozobil was not studied in controlled clinical studies in patients with severe renal impairment or in patients on dialysis. A total of 60 patients with an estimated creatinine clearance (CrCl) 51-80 mL/min, 11 patients with CrCl 31-50 mL/min, and none with CrCl ≤30 mL/min were enrolled in the pivotal studies and received at least one dose of Mozobil (0.24 mg/kg body weight subcutaneously).

Mozobil dose and treatment of patients weighing more than 175% of ideal body weight was not investigated. Based on increasing exposure with increasing body weight, the Mozobil dose should not exceed 40 mg/day for patients with a CrCl >50 mL/min and 27 mg/day for patients with a CrCl 20-50 mL/min. The Sponsor has initiated a fixed dose (20 mg) vs. weight based dose study in lower weight NHL patients to investigate the effects of weight on pharmacokinetics and pharmacodynamics. This study has been restricted to patients weighing 70 kg or less.

3.3.4 Clinical Safety

The clinical safety evaluation of Mozobil was based on safety data obtained from 25 clinical studies including a thorough QT/QTc study, as well as the North American Compassionate Use Programme. Data from over 1,400 patients who received at least 1 dose of Mozobil in the clinical development programme were used to establish the safety profile. Safety data for G-CSF plus Mozobil were obtained from two Phase III studies (301 patients) and 10 uncontrolled studies (242 patients). Patients were primarily treated with Mozobil at daily doses of 0.24 mg/kg by SC injection. Median exposure to Mozobil in these studies was 2 days (range 1 to 7 days).

The adverse reactions reported in oncology patients who received Mozobil in the controlled Phase III studies and uncontrolled studies, including a Phase II study of Mozobil as monotherapy for HSC mobilization, were similar. No notable differences in the incidence of adverse reactions were observed for oncology patients by disease, age, or sex.

In both Phase III studies, the majority of adverse events (AEs) were reported during mobilization and apheresis and were mild to moderate in severity. The AEs that occurred during mobilization and apheresis in ≥5% of the pooled Phase III results from patients who received Mozobil, regardless of causality and more frequent with Mozobil than placebo, were as follows: diarrhea; nausea; vomiting; flatulence; injection site reactions; fatigue; arthralgia; headache; dizziness; and insomnia. The incidence of anxiety during HSC mobilization and apheresis was 5.3% vs. 4.5%, Mozobil vs. placebo, respectively. Paresthesia was considered an AE related to study treatment in 7.0% of patients in the Mozobil group and 5.1% of patients in the placebo group.

Hypokalaemia and hypomagnesaemia were reported as treatment-related AEs more frequently with Mozobil than with placebo during mobilization and apheresis in the pooled Phase III data. The administration of G-CSF plus Mozobil caused leukocytosis. Thrombocytopaenia was also observed in patients receiving Mozobil.

The effect of Mozobil on spleen size in patients has not been specifically evaluated in clinical studies. The possibility that Mozobil in conjunction with G-CSF can cause splenic enlargement cannot be excluded.

The AE reporting beyond mobilization and apheresis was limited by protocol. The majority of serious adverse events (SAEs) occurred after the mobilization and apheresis period, and were considered unrelated to study treatment. In the pooled Phase III data, the incidence of bactaeremia, lung infections and febrile neutropaenia was higher in the Mozobil group than in the placebo group; however, the majority of these events followed myeloablative chemotherapy and most were considered by the investigator to be unrelated to Mozobil administration. The incidence, cause, and timing of deaths, as well as the incidence of study or treatment discontinuations due to AEs were similar in both treatment groups (Mozobil and placebo). The majority of deaths occurred post-engraftment.

In the Phase III studies, examination of all clinical cardiovascular adverse events did not identify any rhythm-related cardiac safety signals attributable to Mozobil treatment in the populations studied. However, in a randomized, double-blind, placebo-controlled crossover study in healthy subjects, Mozobil was associated with an asymptomatic shortening of the PR interval and therefore, caution is recommended in patients with pre-excitation syndromes. Additional cardiovascular events, such as decreases in blood pressure, vasovagal reactions, and myocardial infarctions were also included in the Warnings and Precautions section of the Product Monograph. An increased incidence of hypotension occurred during mobilization and apheresis in the Phase III studies for patients in the G-CSF plus Mozobil group compared to the G-CSF plus placebo group. In the Mozobil oncology and non-oncology clinical studies, 0.8% of the subjects experienced vasovagal reactions (the majority within 1 hour of Mozobil administration). In the clinical studies, 0.9% of oncology patients in the G-CSF plus Mozobil group experienced myocardial infarctions (MIs) after mobilization as compared to 0.3% of oncology patients in the G-CSF plus placebo group after mobilization. All MIs occurred at least 14 days after the last Mozobil administration and in patients with known cardiac risk factors for MI and/or previous exposure to cardiotoxic chemotherapy.

In the Mozobil oncology clinical studies, 5/752 (0.7%) of patients experienced mild or moderate systemic reactions within approximately 30 minutes after Mozobil administration. Events included one or more of the following: urticaria [(n) = 2]; periorbital swelling (n = 2); dyspnoea (n = 1); or hypoxia (n = 1). Symptoms generally responded to treatments or resolved spontaneously.

In the Phase III studies, the incidence of psychiatric disorders during mobilization and apheresis was 14.8% in the G-CSF plus Mozobil group compared to 10.2% in the G-CSF plus placebo group. Insomnia and anxiety were the most common events.

G-CSF mobilization is recognized to have the potential to mobilize tumour cells from the marrow; however, the effect of reinfusion of tumour cells with the apheresis product has not been well-studied and only limited data are available. It is also known that chemotherapy, with or without growth factors, results in the mobilization of tumour cells in patients with haematologic malignancies resulting in tumour cell contamination of the graft. When Mozobil and G-CSF have been administered to patients with acute myelogenous leukemia and plasma cell leukemia, some patients experienced an increase in the number of circulating leukemia cells. Therefore, Mozobil should not be used for HSC mobilization and harvest in patients with leukemia.

There exists a potential for tumour cell mobilization in patients with MM or NHL receiving treatment with Mozobil for the purpose of stem cell mobilization. Furthermore, Mozobil and G-CSF mobilize HSC via different mechanisms; therefore, they might theoretically result in a broader potential for mobilization of tumour cells when used in combination. Tumor cell mobilization by Mozobil has not been thoroughly studied. Nonclinical studies are lacking. Based on laboratory investigations conducted in clinical studies of patients with NHL and MM, an increase in mobilization of tumour cells above that which occurs with G-CSF mobilization alone has not been observed with Mozobil, although limitations in assays utilized and patient numbers examined are important to acknowledge. The severity and nature of the risk associated with potential enhancement of tumour cell mobilization with Mozobil in combination with G-CSF when used for stem cell mobilization in patients with MM or NHL remains uncertain at present. However, by excluding patients with plasma cell and other leukemias from Mozobil stem cell mobilization, the risk and safety concern is considered to be acceptable.

3.4 Benefit/Risk Assessment and Recommendation

3.4.1 Benefit/Risk Assessment

Based on review of the submission, the overall benefit/risk assessment is positive and supports a recommendation of approval of Mozobil for use in combination with G-CSF to mobilize HSCs to the peripheral blood for collection and subsequent autologous transplantation in patients with NHL and MM.

In pivotal clinical studies supporting the use of Mozobil in adult patients, the addition of Mozobil to a G-CSF mobilization regimen significantly enhanced CD34+ cell mobilization; decreased the required number of apheresis days; increased the proportion achieving a target number of cells for transplant compared to G-CSF alone; and resulted in prompt and durable engraftment of polymorphonuclear cells and platelets. Graft durability results were similar in both treatment groups. For transplanted patients, the frequency of graft failure was low and none of these graft failures were considered by the investigator as related to Mozobil.

Overall survival or other clinical benefit endpoints were not used to demonstrate efficacy in Phase III studies. As an exploratory endpoint, overall survival data collected and summarized for patients who went to transplantation were similar in both treatment groups in both pivotal studies at 12 months post-transplantation.

It is considered important to note that some patients with NHL and MM met minimal and target HSC collection criteria with G-CSF alone. NHL patients received considerable doses of cytotoxic chemotherapy which may have negatively impacted HSC mobilization and collection. Achievement of minimal and target collection with G-CSF alone occurred more frequently for MM patients than for NHL patients. In Study 1 (NHL), 47.3% of G-CSF plus placebo patients achieved minimum CD34+ stem cell count in 4 or fewer days of apheresis and 19.6% of G-CSF plus placebo patients achieved target CD34+ stem cell count in 4 or fewer days of apheresis, while in Study 2 (MM), 88.3% of G-CSF plus placebo patients achieved minimum CD34+ stem cell count in 4 or fewer days of apheresis and 55.9% of G-CSF plus placebo patients achieved target CD34+ stem cell count in 4 or fewer days of apheresis. However, treating only those patients who do not achieve sufficient mobilization in response to G-CSF alone is not considered an optimal approach. Prognostic indicators [for example, pre-apheresis peripheral blood (PB) CD34+ cell count] are not sufficient to identify many of those patients who ultimately fail to mobilize or mobilize poorly. A “just-in-time” approach in which pre-apheresis PB CD34+ cell count is used to make real-time mobilization regimen decisions is likely not practical at all Canadian centres. G-CSF mobilization has a window of maximal mobilization of CD34+ cells which peaks around days 4-5 of treatment, and by delaying optimal therapy, the opportunity to collect enough cells could be missed and the patient might have to either undergo a second mobilization procedure or additional apheresis procedures with the potential for significant risks.

For the purpose of HSC mobilization, Mozobil can cause mobilization of leukemic cells and subsequent contamination of the apheresis product. Therefore, Mozobil should not be used for HSC mobilization and harvest in patients with leukemia.

When Mozobil is used in conjunction with G-CSF for HSC mobilization in patients with NHL or MM‚ tumour cells may be released from the marrow and subsequently collected in the leukapheresis product. Mozobil and G-CSF mobilize HSC via different mechanisms; therefore, they might theoretically result in a broader potential for mobilization of tumour cells when used in combination. CD34+ stem cells collected after Mozobil mobilization are genetically and phenotypically different from those collected after using G-CSF; the significance of these findings is not clear at this time. Based on laboratory investigations conducted in clinical studies of patients with NHL and MM, an increase in mobilization of tumour cells above that which occurs with G-CSF mobilization alone has not been observed with Mozobil, although limitations in assays utilized and patient numbers examined are important to acknowledge. The clinical relevance of Mozobil-mobilized tumour cells remains unclear at present as it is currently unclear in the scientific community whether the presence of tumour cells in the collected peripheral blood stem cell product is a marker of increased risk of relapse or whether detected tumour cells themselves are contributors to relapse and decreased survival in those patients who receive them. The Sponsor is following Phase III patients for up to 5 years for death and disease status, particularly relapse. At 12 months post transplantation, OS was similar in the two treatment arms. Additionally, adverse events associated with tumour cell mobilization will be monitored during routine postmarketing surveillance and reported expeditiously as required.

Use of Mozobil is not recommended during pregnancy due to its mechanism of action and results of reproductive toxicology studies. Because CXCR4 plays an essential role in foetal development and Mozobil is a selective antagonist of CXCR4, Mozobil is suggested to cause congenital malformations when administered during pregnancy. Studies in animals have shown teratogenicity. There are no adequate and well-controlled studies in pregnant women using Mozobil. Women of childbearing potential are advised to use effective contraception during treatment.

The safety and efficacy of Mozobil in paediatric patients have not been established in controlled clinical studies. As certain types of NHLs occur in children, a paediatric study is being conducted.

The primary route of elimination of Mozobil is renal excretion. No overall differences in effectiveness were observed between subjects 65 years of age and older and younger subjects with normal renal function or mild-moderate renal impairment. Nonetheless, in general, care should be taken in dose selection for elderly patients due to the greater frequency of decreased renal function with advanced age. Furthermore, renal impairment and renal failure are well known disease-related complications of multiple myeloma.

Mozobil was not studied in controlled clinical studies in patients with severe renal impairment or in patients on dialysis. In Phase III studies, despite small sample size, mobilization results for patients with mild or moderate impairment appeared to be consistent with those for patients with normal renal function. Adverse event reporting during mobilization and apheresis appeared to be consistent for patients with moderate impairment and with normal renal function. Dose reduction is recommended for patients with an estimated CrCl 20-50 mL/min although clinical data with this dose adjustment are limited. There are insufficient clinical data to make dosing recommendations for patients with CrCl <20 mL/min or on dialysis. A further dose reduction may be warranted in these subpopulations for safety reasons; renal impairment was associated with a prolongation of Mozobil half-life and increased exposure due to impaired clearance, and tissue accumulation of Mozobil in patients with renal impairment has not been studied. However, the potential impact on efficacy of dose reduction below 0.16 mg/kg/day is unknown. The effects of co-administration of Mozobil with other drugs that are renally eliminated or are known to affect renal function have not been evaluated in formal drug interaction studies.

Mozobil dose and treatment of patients weighing more than 175% of ideal body weight were not investigated. Based on increasing exposure with increasing body weight, the Mozobil dose should not exceed 40 mg/day for patients with a CrCL >50 mL/min and 27 mg/day for patients with a CrCL 20-50 mL/min. The Sponsor has initiated a fixed dose (20 mg) vs. weight based dose study in lower weight NHL patients to investigate the effects of weight on pharmacokinetics and pharmacodynamics. This study has been restricted to patients weighing 75 kg or less.

Appropriate precautions prior to driving or using machines is recommended because of the potential for dizziness, fatigue or vasovagal reactions while undergoing treatment with Mozobil. In a randomized, double-blind, placebo-controlled crossover study in healthy subjects, Mozobil was associated with an asymptomatic shortening of the PR interval and therefore caution is recommended in patients with pre-excitation syndromes. Additional cardiovascular events included in the Warnings and Precautions section of the Product Monograph were as follows: hypotension during mobilization and apheresis; vasovagal reactions, the majority with onset within one hour of subcutaneous administration of Mozobil; and myocardial infarctions, all occurring at least 14 days after last Mozobil dose and in patients with known cardiac risk factors for myocardial infarction and/or previous exposure to cardiotoxic chemotherapy. Appropriate precautions were recommended because of the potential for mild or moderate allergic systemic reactions within approximately 30 minutes of Mozobil administration. A potential for an increased incidence of psychiatric disorders, most commonly insomnia and anxiety, during mobilization and apheresis was also communicated.

Administration of Mozobil in conjunction with G-CSF increases circulating leukocytes as well as HSC populations. Thrombocytopaenia has been observed in patients receiving Mozobil. The possibility that Mozobil in conjunction with G-CSF can cause splenic enlargement cannot be excluded. In Phase III studies, hypokalaemia and hypomagnesaemia were reported as treatment related AEs more frequently with Mozobil than with placebo during mobilization and apheresis. Appropriate monitoring is recommended.

A risk management plan has been submitted and reviewed.

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 Mozobil in combination with G-CSF is favourable to mobilize HSCs to the peripheral blood for collection and subsequent autologous transplantation in patients with NHL and MM while recognizing that some patients with NHL and MM are able to meet minimal and target HSC collection criteria with G-CSF alone. The NDS complies with the requirements of sections C.08.002 and C.08.005.1 and therefore Health Canada has granted the Notice of Compliance pursuant to section C.08.004 of the Food and Drug Regulations.

4 Submission Milestones

Submission Milestones: MozobilTM

Submission MilestoneDate
Pre-submission meeting:2010-09-30
Submission filed:2010-12-21
Screening
Screening Acceptance Letter issued:2011-02-11
Review
Quality Evaluation complete:2011-12-05
Clinical Evaluation complete:2011-12-08
Electrocardiogram Review complete:2011-08-24
Labelling Review complete:2011-12-08
Notice of Compliance issued by Director General:2011-12-08