Summary Basis of Decision for Arzerra ™

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
ArzerraTM

Ofatumumab, 20 mg/mL, Solution, Intravenous

GlaxoSmithKline Inc.

Submission control no: 128188

Date issued: 2012-11-02

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:

ArzerraTM

Manufacturer/sponsor:

GlaxoSmithKline Inc.

Medicinal ingredient:

Ofatumumab

International non-proprietary Name:

Strength:

20 mg/mL

Dosage form:

Solution

Route of administration:

Intravenous

Drug identification number(DIN):

  • 02381559 - 100 mg/5 mL
  • 02381567 - 1,000 mg/50 mL

Therapeutic Classification:

Non-medicinal ingredients:

Submission type and control no:

New Drug Submission Control Number: 128188

Date of Submission:

2011-01-21

Date of authorization:

2012-03-09
ARZERRA is used under license by GlaxoSmithKline Inc.
2 Notice of decision

On March 9, 2012 Health Canada issued a Notice of Compliance with conditions under the Notice of Compliance with Conditions (NOC/c) Policy to GlaxoSmithKline Inc., for the drug product, Arzerra. 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.

Arzerra contains the medicinal ingredient ofatumumab, which is an antineoplastic agent. Ofatumumab is a recombinant human monoclonal antibody (IgG1κ) that binds specifically to the CD20 molecule expressed on B lymphocytes. Binding of Arzerra to the CD20 molecule induces tumour cell death.

Arzerra is indicated for the treatment of patients with chronic lymphocytic leukaemia (CLL) refractory to fludarabine and alemtuzumab. The efficacy of Arzerra is based on the demonstration of durable objective responses. No data demonstrate an improvement in disease related symptoms or increased survival with Arzerra.

The market authorization was based on quality, non-clinical, and clinical information submitted. The efficacy of Arzerra (ofatumumab) was studied in a Phase III, single-arm, open-label, multicentre study of 154 patients with relapsed or refractory CLL. Efficacy was evaluated in a planned interim analysis that included data from 59 patients with CLL, refractory to fludarabine and alemtuzumab, who were administered Arzerra as monotherapy. The primary efficacy endpoint was objective response rate (ORR) determined using National Cancer Institute-Working Group (NCI-WG) 1996 guidelines. The study design consisted of patients receiving 12 infusions of Arzerra over a period of 24 weeks. The study results demonstrated Arzerra was efficacious with an ORR of 42.4% [99% Confidence Interval (CI): 26.2, 59.8].

The safety of Arzerra was evaluated in 250 patients with relapsed or refractory CLL in two open-label, single-arm studies. In these studies Arzerra was administered at 2,000 mg beginning with the second dose for 11 doses in Study 1 [number (n) = 223)] or 3 doses in Study 2 (n = 27). The most common adverse reactions in Study 1 were cough, pyrexia, anaemia, neutropenia, diarrhoea, fatigue, dyspnoea, pneumonia, chills, nausea, bronchitis, peripheral oedema, back pain and upper respiratory tract infection. The most common serious adverse reactions were pneumonia and neutropenia. Infections were the most common adverse reactions leading to drug discontinuation in Study 1.

Arzerra (20 mg/mL, ofatumumab) is presented as a sterile solution in two vial sizes, 100 mg/5 mL in a 10 mL vial and 1,000 mg/50 mL in a 60 mL vial. Prior to intravenous (IV) administration, Arzerra must be diluted into an infusion bag containing 0.9% sodium chloride for infusion. In addition, patients should be pre-medicated 30 minutes to 2 hours prior to each dose with oral acetaminophen 1,000 mg (or equivalent), oral or intravenous antihistamine (cetirizine 10 mg or equivalent), and intravenous corticosteroid (prednisolone 100 mg or equivalent). The recommended dose of Arzerra is 300 mg for the first infusion and 2,000 mg for all subsequent infusions. The infusion schedule is 8 consecutive weekly infusions, followed 4 weeks later by 4 consecutive monthly [that is (i.e.) every 4 weeks] infusions. While on Arzerra therapy, complete blood counts and platelet counts should be monitored at regular intervals, and more frequently in patients who develop cytopenias. Dosing guidelines are available in the Product Monograph.

Arzerra is contraindicated in patients who are hypersensitive to ofatumumab or to any ingredient in the formulation or components of the container. Arzerra is also contraindicated in patients who have or have had progressive multifocal leukoencephalopathy (PML). Arzerra 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 Arzerra 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 Arzerra is favourable for the indication stated above.

3 Scientific and Regulatory Basis for Decision

GlaxoSmithKline Inc. submitted a New Drug Submission (NDS) on January 21, 2011 to seek approval for the marketing of Arzerra, indicated for the treatment of patients with chronic lymphocytic leukaemia (CLL) refractory to fludarabine and alemtuzumab. The efficacy of Arzerra is based on the demonstration of durable objective responses. No data demonstrate an improvement in disease related symptoms or increased survival with Arzerra therapy. The product was authorized under the Notice of Compliance with Conditions (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.

3.1 Quality Basis for Decision

3.1.1 Drug Substance (Medicinal Ingredient)

General Information

Ofatumumab, the medicinal ingredient of Arzerra, is an antineoplastic agent. It is a human monoclonal antibody (IgG1κ) used to treat CLL. Chronic lymphocytic leukaemia is a cancer of the blood, characterized by a gradual increase in the number of white blood cells (called lymphocytes), in the blood and bone marrow. Chronic lymphocytic leukaemia can also affect other organs in your body. Arzerra acts by binding to the lymphocytes causing them to die.

Manufacturing Process and Process Controls

Ofatumumab is produced in a recombinant murine cell line using standard mammalian cell cultivation and purification technologies. The manufacture of ofatumumab consists of a series of steps which include cell culture, product harvest, purification, virus inactivation, removal, and formulation.

The consistency of the manufacturing process is ensured through defined production procedures, critical quality tests, in-process limits, and ofatumumab certificate of analysis specifications. Process validation data demonstrate that the manufacturing processes operate in a consistent manner, yielding product of acceptable quality. Microbial control is maintained throughout the manufacturing process by testing for bioburden as well as for bacterial endotoxins. In-process controls performed during the manufacture were reviewed and are considered acceptable. The specifications for the raw materials used in manufacturing the drug substance are also considered satisfactory.

Characterization

Detailed characterization studies were performed to provide assurance that ofatumumab consistently exhibits the desired characteristic structure and biological activity.

The drug substance manufacturing process has been optimized and scaled up during development. The process changes introduced at each generation of the process were adequately described and comparatively addressed. Lot release, stability, and characterization data have also been used to support the comparability assessment.

Control of Drug Substance

The drug substance specifications and analytical methods used for quality control of ofatumumab are considered acceptable.

The levels of product- and process-related impurities were adequately monitored throughout the manufacturing process. Results from process validation reports and in-process controls indicated that the impurities of the drug substance were adequately controlled. The level of impurities reported for the drug substance was found to be within the established limits.

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

Stability

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

3.1.2 Drug Product

Description and Composition

Arzerra is a sterile, clear to opalescent, colourless, preservative-free liquid concentrate (20 mg/mL) which requires dilution prior to intravenous administration. Arzerra is presented in a single-use glass vial with a latex-free rubber stopper and an aluminum overseal. Arzerra is supplied in two vial sizes, 100 mg/5 mL in a 10 mL vial and 1,000 mg/50 mL in a 60 mL vial. Arzerra 100 mg/5 mL is packaged in a carton containing 3 single-use vials with 2 filter sets. Arzerra 1,000 mg/50 mL is packaged in a carton containing 1 single-use vial with 2 filter sets.

Each Arzerra single-use vial contains either 100 mg ofatumumab in 5 mL of solution or 1,000 mg ofatumumab in 50 mL of solution along with the following non-medicinal ingredients: arginine; sodium acetate; sodium chloride; polysorbate 80; edetate disodium; hydrochloric acid; and water for injection.

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 formulation made throughout the pharmaceutical development are considered acceptable upon review.

Manufacturing Process and Process Controls

The drug product manufacturing process consists of diluting the drug substance into the dilution buffer followed by sterile filtration, and aseptic filling into sterile glass vials.

All manufacturing equipment, in-process manufacturing steps and detailed operating parameters were adequately described in the submitted documentation and are found to be acceptable. The manufacturing process is considered to be adequately controlled within justified limits.

The drug product manufacturing process has been adequately validated and is capable of consistently generating product that meets release specifications.

Control of Drug Product

Each lot of Arzerra drug product is tested for appearance, content, identity, potency, purity, and impurities. Established test specifications and validated analytical test methods are considered acceptable.

Through Health Canada's lot release testing and evaluation program, consecutively manufactured final product lots were tested, evaluated, and found to meet the specifications of the drug product and demonstrate consistency in manufacturing.

Stability

Based on the long-term, accelerated, and stress stability data submitted, the proposed 24-month shelf-life at 2-8°C for Arzerra is considered acceptable when protected from light.

The compatibility of the drug product with the container closure system was demonstrated through compendial testing and stability studies. The container closure system met all validation test acceptance criteria.

3.1.3 Facilities and Equipment

An on-site evaluation (OSE) of the facility involved in the manufacture and testing of the drug substance (ofatumumab) was not warranted since the facility was recently evaluated in good standing.

3.1.4 Adventitious Agents Safety Evaluation

The ofatumumab manufacturing process incorporates adequate control measures to prevent contamination and maintain microbial control. Pre-harvest culture fluid from each lot is tested to ensure freedom from adventitious microorganisms (bioburden, mycoplasma, and viruses). Steps from the purification process designed to remove and inactivate viruses are adequately validated.

Raw materials of animal and recombinant origin used in the manufacturing process are adequately tested to ensure freedom from adventitious agents. The excipients used in the drug product formulation are not from animal or human origin.

3.1.5 Conclusion

Chemistry and Manufacturing information submitted for Arzerra 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

Primary pharmacodynamic studies demonstrated that ofatumumab bound specifically to transfected CD20-NSO and CD20-SKBR3 cells and competed with rituximab on the transfected CD20-NSO cells, but did not bind to non-transfected cells. It was determined that binding of ofatumumab and rituximab to CD20 on B-cell lines saturated at similar levels. Binding of ofatumumab to human B-cells occurred with an effective concentration (EC50) value of 287 ng/mL, and dissociation from CD20 was slow with a dissociation rate constant (Koff) value of 6.4×10-5 seconds. This corresponds to a dissociation half-life of three hours, which is twice as long as the dissociation half-life for rituximab.

The mutating residues 159, 170 or 172 of human CD20 did not influence binding of ofatumumab. These residues, however, are critical for CD20 recognition by rituximab and all other known CD20 monoclonal antibodies. The residues involved for ofatumumab were identified as N163, N166 and possibly N159.

Low numbers of T-helper epitopes were found on ofatumumab which could induce an anti-antibody response in humans, whereas rituximab contained a relatively high number of epitopes. Ofatumumab is therefore expected to be less immunogenic in the Caucasian population compared to rituximab.

Flow cytometry and immunochemistry assays revealed that ofatumumab bound specifically to CD20+ B-cells in rhesus and cynomolgus monkeys, with comparable apparent affinity. There was no cross-reactivity of ofatumumab in dog, pig, rabbit, mouse, or rat splenic tissue. Hence, rhesus and cynomolgus monkeys were considered to be the most appropriate species for use in the non-clinical safety studies. It was also shown that transfectoma-derived ofatumumab is similar to hybridoma-derived ofatumumab and was therefore considered suitable for use in the toxicity studies. Binding of ofatumumab to CD20 molecules on the membrane led to the clustering of CD20 molecules in lipid rafts, which may be important in inducing complement-dependent cytotoxicity (CDC) in human B-cells.

Induction of CDC by ofatumumab in primary tumour B-cells and various tumour B-cell lines involved the binding of complement factors C1q, C3 and C4c. The slower rate of dissociation from cell surface CD20 was reflected in the capacity to induce efficient CDC over a significantly longer period. Ofatumumab induced lysis in more than 80% of the cells for all of the five B-cell lines tested, whereas rituximab only induced more than 80% cell lysis for two of these cell lines, that is (i.e) u-DHL-4 and Daudi cells. Rituximab induced approximately 50% cell lysis of DOHH cells and less than 20% of ARH77 and Raji cell lines. Cell lysis was dependent on the concentration of normal human serum (NHS). Addition of ofatumumab caused maximal cell lysis of Daudi cells at the highest NHS and antibody concentration. Experiments conducted with CEM T-cells demonstrated that ofatumumab was able to kill cells with very low CD20 expression.

Ofatumumab was capable of inducing lysis of all primary tumour B-cells tested (i.e., B-cell CLL, hairy cell leukaemia cells, B-cell acute lymphoblastic leukaemia, follicular lymphoma cells, primary mantle cell lymphoma cells) in vitro and was more effective in performance when compared to rituximab. Ofatumumab-mediated antibody-dependent cell-mediated cytotoxicity (ADCC) was similar to rituximab-mediated ADCC, and was evident at lower antibody binding levels than CDC.

Cell death via induction of apoptosis was not a major mechanism for ofatumumab. Binding of ofatumumab to CD20 and induction of CDC and ADCC were reduced after cholesterol depletion of freshly isolated B-cells, which could be overcome by increasing the antibody concentration to levels which saturate CD20 binding sites.

There was a positive correlation between the galactose content of ofatumumab and its ability to induce CDC. However, the difference in potency was small (i.e., less than two-fold) and lay within the limits of specification of the ofatumumab potency assay set at 70 to 130%. The maximal change in galactose content of ofatumumab did not influence the in vivo therapeutic efficacy of ofatumumab in the B-cell tumour of severe combined immunodeficiency (SCID) mouse model. The difference in potency reflected a reduction in binding of the complement factor C1q, and subsequent C3 and C4 deposition.

Deglycosylation of ofatumumab did not affect binding to CD20, however, it resulted in reduced C1q binding, C4 and C3 deposition, CDC induction and bonding of the antibody to Fcγ receptor IIIa.

Cells opsonized with ofatumumab underwent high levels of complement activation and CDC in C1q-depleted serum supplemented with low concentrations of C1q. Under comparable conditions, rituximab-opsonized cells bound less C1q. In addition, even when higher concentrations of C1q were used to achieve comparable C1q binding to rituxomab-opsonized cells, less complement activation and CDC were observed.

Ofatumumab effectively reduced the growth of human lymphoma xenografts in SCID mice. Effectiveness was dependent on time of administration after tumour inoculation. Treatment in the first week after tumour induction was more effective than later treatment. There was no difference in efficacy between different dose levels used, indicating that maximum efficacy was already reached at the lowest dose of 0.5 mg/kg body weight (bw). Hence, increasing dose levels above the point of target saturation is not expected to increase the therapeutic effect.

Secondary Pharmacodynamics

A single secondary pharmacodynamic study was conducted, which demonstrated that ofatumumab depleted B-cells in rheumatoid arthritis synovial tissue which had been subcutaneously implanted in SCID mice.

Overall, the pharmacodynamic data indicates that specific lysis of CD20+ B-cells by ofatumumab leads to a profound and prolonged depletion of B-cells in vivo. Ofatumumab has the potential to be an efficacious treatment for CLL in humans.

3.2.2 Pharmacokinetics

Pharmacokinetic studies were included within toxicology studies and are fully described under section 3.2.3 Toxicology.

3.2.3 Toxicology

All in vivo toxicology studies were conducted in cynomolgus monkeys as ofatumumab does not bind to the B-cell CD20 antigen of lower species.

Acute Toxicity

Single dose, acute toxicity studies were not performed with ofatumumab. In general, single-dose studies do not provide meaningful information in respect to monoclonal antibodies given acute effects tend to be restricted to exaggerated pharmacology that is better characterized in repeat-dose studies.

Repeat-Dose Toxicity

Four week and seven month repeat-dose toxicity studies were conducted at doses of 20 or 100 mg/kg bw. After one month of ofatumumab treatment, B-cell depletion was observed in the peripheral blood and lymph nodes at both dose levels. This observation correlated histopathologically with minimal to moderate atrophy of germinal centre or follicular B-cells in lymphoid organs [for example (e.g.) mandibular and mesenteric lymph nodes, Peyer's patch, and spleen]. In both studies, during the six-month no treatment recovery period, B-cell levels gradually returned to baseline levels, however, the length of time required for this reversal to occur appeared to be dose-dependent, with the higher dose animals recovering later than lower dose animals. It was also noted during the treatment and recovery period, that the majority of animals who were administered ofatumumab at either dose level developed haemolytic anaemia. Results obtained from the direct Coomb's test leads to suggest that the monkeys developed antibodies to ofatumumab, which through formation of immune complexes and complement activation enabled binding to the red cell surface, most likely resulting in the development of haemolytic anaemia.

Evaluation of humoral immune function in monkeys immunized with keyhole limpet haemocyanin (KLH) followed by continuous intravenous infusion of ofatumumab revealed inhibition of specific KLH antibodies in the 100 mg/kg bw group, which was only partially reversed by the end of the six month recovery period. This inability to recover from complete depletion of the B-cell populations could indicate a chronic immunosuppression which could lead to increased susceptibility to infections. There were no treatment-related effects noted for cellular immune function at any dose level.

Results of a cycled repeat-dose toxicity study conducted at doses of 20 and 100 mg/kg bw also resulted in depletion of B-cells with corresponding histopathology, and slightly lower red blood cell indices. Full recovery was noted after the four month recovery period. Treatment resulted in inhibition of the secondary humoral immune response. In addition, scabbing/bruising on the scrotum was observed for two males in the 100 mg/kg bw group; one of these males also had enlarged inguinal lymph nodes and scabbing and swelling of the prepuce.

Five mortalities occurred in the animals tested during the toxicology studies. Two of these deaths were due to haemolytic anaemia, possibly resulting from the formation of antibodies capable of binding to the red cell surface. Two more deaths were associated with gastro-intestinal disease resulting from bacterial infection, which was determined to be campylobacter jejuni for one of these animals. Since these infections are usually self-limiting, treatment with ofatumumab may have exacerbated the disease progression through immunosuppression. The review of evidence for these four mortalities indicates these events were secondarily related to the administration of ofatumumab. The cause of the remaining death could not be determined with certainty, and therefore a potential primary relationship to ofatumumab administration could not be ruled out.

Genotoxicity and Carcinogenicity

Studies to assess the potential for genotoxicity and carcinogenicity of ofatumumab were not conducted. Ofatumumab is not expected to interact directly with deoxyribonucleic acid (DNA) or other chromosomal material, therefore the use of standard genotoxicity studies does not apply. In addition, ofatumumab is not pharmacologically active in rodents therefore standard carcinogenicity bioassays are unsuitable. Additionally, no proliferative and/or pre-neoplastic changes were reported in any of the monkeys in the six month repeat-dose toxicology study. Furthermore, there is no scientific evidence to suggest that depletion of B-cells by any other anti-CD20 therapeutics leads to cancer. It is also important to note that populations of natural killer (NK) cells, which are central to immune surveillance, were not altered in the repeat-dose toxicity studies.

Reproductive and Developmental Toxicity

The standard battery of reproductive toxicity studies were not considered necessary for ofatumumab, due to the nature of CLL and the given patient population age group. The reproductive toxicology package was limited to a single enhanced embryo-foetal development study. The embryo-foetal developmental study was conducted in pregnant cynomolgus monkeys who were administered ofatumumab at dose levels of 20 and 100 mg/kg bw. The treatment results obtained showed no overt maternal toxicity, developmental toxicity or teratogenicity. Yet, administration of ofatumumab did show complete depletion of B-lymphocytes in the peripheral blood of the maternal animals at both dose levels. At the time of caesarean section, lymphocyte numbers had still not yet recovered. In addition, the absolute number of B-lymphocytes in the foetal blood, and the percentage of B-lymphocytes in foetal spleen tissue were decreased. Although these findings reflect the pharmacological activity of ofatumumab, depleted B-cells could alter immune responses of the dam and foetuses.

In the repeat-dose toxicology studies, there were no specific histopathological changes observed in the reproductive tissues (epididymus, testis and ovaries). Additionally, the CD20 antigen is not expressed in tissues associated with fertility, and binding to reproductive tissues was not observed in a human tissue cross-reactivity study.

3.2.4 Summary and Conclusion

The non-clinical toxicology database was considered adequate to assess the safety profile of ofatumumab (Arzerra) and support its use in humans for the treatment of CCL refractory to fludarabine and alemtuzumab, provided adequate safety precautions are taken against the possible immunosuppressive effect of treatment. Patients should be monitored for cytokine release syndrome, and for the possible development of haemolytic anemia. In addition, patients should not be vaccinated until at least six months after cessation of treatment. Despite this, it is still possible that after six months, a robust antibody response to vaccination may not occur. There is also the possibility that certain patients may be unable to restore B-lymphocyte numbers to pre-treatment levels upon cessation of treatment. In addition, given that decreased B-cell counts were evident in foetuses at the time of caesarean section, and considering that ofatumumab has an inhibitory effect on the humoral immune response, Arzerra should not be administered to pregnant women.

3.3 Clinical basis for decision

3.3.1 Pharmacodynamics

Peripheral B-cell counts decreased after the first Arzerra infusion in patients with haematologic malignancies. In patients with refractory CLL, the median decrease in B-cell counts was 23% after the first infusion and 92% after the eighth infusion. Peripheral B-cell counts remained low in most patients throughout the remainder of Arzerra therapy administration and then gradually recovered (median decrease in B-cell counts was 68% below baseline three months after the end of Arzerra therapy). Significant B-cell depletion may continue for up to three to twelve months after the last Arzerra infusion.

There is also a potential for immunogenicity with therapeutic proteins such as Arzerra; however the formation of anti-Arzerra antibodies may be decreased because Arzerra is a human antibody that depletes B-cells.

In the pivotal clinical study (discussed in detail in section 3.3.3 Clinical Efficacy), no anti- Arzerra antibodies were detected in the 46 patients who had received at least eight infusions (33 of whom received all 12 infusions) and had sufficiently low circulating Arzerra concentrations to allow detection.

3.3.2 Pharmacokinetics

The population pharmacokinetic (PPK) analysis for Arzerra combined the data from the two studies specific for CLL (Hx-CD20-402, Hx-CD20-406) along with two other indications (Rheumatoid Arthritis, Follicular Lymphoma).

As is characteristic of most IgG monoclonal antibodies, the pharmacokinetic profile of Arzerra could be described by a two compartment model with a long half-life. However, the large mass of CD20+B-cells, typical of the CLL indication, created a relatively large target-mediated clearance (TMC), which could not be ignored. Although the authors managed to capture this TMC through mathematical constructions, they emphasized that the modelled total body B-cell mass and the corresponding TMC effect were only indirectly related to measurable peripheral B-cell counts. While CD5+CD19+B-cell counts showed a time course similar to the pharmacokinetic-derived target mass during the treatment period in CLL, the modelled total B-cell mass and TMC cannot be directly validated with the available clinical data. Like most pharmacokinetic analyses, the model parameters could not be equated with particular biological processes. Furthermore, it is the opinion of the authors of the final population pharmacokinetic/pharmacodynamics report (page 76):

While the pharmacokinetic-derived target B-cell mass seemed to be a useful mathematical component in this modelling, it would be an unreasonable extrapolation to suggest that this quantity was in any way correlated with effect of ofatumumab on disease.

While the model described above was the most complete in terms of describing the TMC, this model was not available for use to generate the estimated pharmacokinetic parameters presented in the submission. A simpler pharmacokinetic model was used to estimate pharmacokinetic parameters from the pivotal study (Hx-CD20-406). However, this model did not take into account the TMC and depended on data from pharmacokinetic profiles obtained in a previous study (Hx-CD20-402). This simpler model was considered to be a 'reduced' model in comparison to the first. Its two main assumptions 1) ofatumumab pharmacokinetic behaviour was similar for the populations studied in the two trials (i.e., the pharmacokinetic profiles were of similar shape with comparable interindividual differences) and 2) the expected change in the elimination rate with repeated dosing associated with B-cell depletion was assumed to be comparable after four weekly infusions and after eight weekly or eight weekly followed by four monthly infusions were considered to be too speculative for a valid assessment.

The only measured pharmacokinetic parameters available from the pivotal study (Hx-CD20-406) were maximum plasma concentration (Cmax), minimum plasma concentration (Cmin), and time to maximum plasma concentration (Tmax). Maximum plasma concentration values were 63, 1,482, and 881 mg/L after the first, eighth and twelfth infusions, respectively. Minimum plasma concentration values were 579, and 47 mg/L prior to the eighth and twelfth infusion, respectively. Time to Cmax (Tmax) was 7.4 h at the first infusion and 4.5 h at both the eighth and twelfth infusions.

The Phase I dose-escalation study (Hx-CD20-402) in CLL patients employed a dense sampling method allowing for the measurement and calculation of relevant pharmacokinetic parameters at the first and fourth infusions. Patients in the highest dose group in this study [number (n) = 27] received an initial infusion of 500 mg followed weekly by 3 infusions of 2,000 mg. The observed pharmacokinetic values at the first and fourth infusions respectively were 136 and 1,061 µg/mL for Cmax and 7,848 and 420,840 µg.h/mL, for area under the curve AUC0-∞. Clearance decreased from 63.7 mL/h at the first infusion to 8.5 mL/h at the fourth as did the volume of distribution (Vss), which was 3.24 L at the first infusion and 1.73 L at the eighth. The half-life increased from 1.3 days after the first infusion to 11.5 days after the fourth and final infusions.

These parameters are consistent with a relatively large proportion of the clearance process being directed through the specific binding with a large initial mass of CD20+B-cells. Once the source of this target-mediated clearance is depleted after several infusions the removal of ofatumumab shifts to non-specific clearance mechanisms which are not as rapid and are associated with a longer half-life.

3.3.3 Clinical Efficacy

The efficacy of Arzerra was evaluated primarily in one Phase II, single-arm, multicentre study conducted in 154 patients with relapsed or refractory CLL. Within this study, efficacy was assessed based on an interim analysis that included data from 59 patients with CLL, refractory to fludarabine and alemtuzumab.

The study population comprised of three CLL groups which were classified by an independent review committee (IRC). Patients who were refractory to both fludarabine and alemtuzumab were classified as the double refractory (DR) group. Patients who were fludarabine-refractory and considered inappropriate for alemtuzumab due to bulky lymphadenopathy were classified as the bulky fludarabine refractory (BFR) group. Patients, who did not meet either the DR or BFR classification criteria, were grouped in the 'Other' category. Patients who met the criteria for both double refractory and bulky fludarabine refractory were assigned to the DR group.

Patients enrolled in the study received a maximum of 12 infusions of Arzerra over a period of 24 weeks. Dosing consisted of 8 weekly infusions of Arzerra from week 0 to week 7, followed by 1 infusion of Arzerra every 4 weeks from week 12 through week 24. The first infusion was 300 mg Arzerra. All subsequent infusions were 2,000 mg Arzerra.

Patients who completed the treatment period entered a subsequent follow-up period where disease status was evaluated every 3 months. Hereafter, end-of-study procedures were completed and the extended follow-up period was initiated. In the extended follow-up period, patients were monitored every 3 months for survival and malignant B-cell values. Monitoring continued until the B-cell values reached baseline level or above, alternative CLL therapy was initiated, or until Month 48. Patients were also allowed to enter the extended follow-up period following withdrawal from treatment- or follow-up period.

The primary efficacy endpoint was objective response rate (ORR). The objective response was measured over a 24-week period beginning with the first administration of Arzerra. All clinical assessments were conducted by site investigators, while response evaluations and classification of patients were conducted centrally by an IRC that followed NCI-WG 1996-guidelines. Patients were classified as either responders or non-responders as follows: patients achieving complete remission (CR) or partial remission (PR) were classified as responders, while patients whose best response was stable disease (SD) or progressive disease (PD) were classified as non-responders. Responses were required to be maintained for a minimum of two months, in accordance to NCI-WG 1996 guidelines.

Secondary efficacy endpoints also assessed were as follows:

  • Duration of objective response - time from initial response (first visit where response was observed) to disease progression or death as assessed by the IRC.
  • Progression free survival (PFS) - time from treatment allocation (Week 0) until disease progression or death.
  • Time to next CLL therapy - time from treatment allocation (Week 0) until the time of first administration of CLL treatment other than Arzerra.
  • Overall Survival (OS) - time from treatment allocation (Week 0) until death.

It was determined by Health Canada that to a large extent these secondary endpoints could not be adequately characterized through a single-arm non-randomized study. Therefore, other than that of 'duration of objective response', these secondary endpoints were considered as adding little to the overall evaluation of the efficacy and safety of Arzerra.

Other secondary endpoints evaluated included: malignant B-cells in peripheral blood, reduction in tumour size; resolution of lymphadenopathy; resolution of constitutional symptoms; resolution of splenomegaly and hepatomegaly (organomegaly); improvement in performance status [assessed through Eastern Cooperative Oncology Group (ECOG) scale]; improvement in haemoglobin; improvement in thrombocytopaenia; improvement in neutropaenia; and blood product transfusions. The composite endpoint of ORR which takes into account of each of the other endpoints listed above was determined to be appropriate.

Based on advice obtained from clinical experts, it was estimated that the ORR for best supportive care in this patient populations was 15% and that an ORR of 30% for Arzerra therapy would be a clinically important improvement. Knowing, based on previous scientific literature, that ORR with salvage chemotherapy was established between 20-25%, and 0% with monoclonal antibodies, it was suggested by the sponsor that observing a 30% overall response rate which excludes a 15% overall response rate at the 1% significance level would indicate meaningful efficacy in this refractory population.

When discussing efficacy results below, highlights of results should be viewed in context of the following important observations made by Health Canada. The primary efficacy endpoint of the pivotal study was ORR, which was measured over a 24 week period. Thereafter, each patient's data was assessed by the IRC. The IRC conducted their assessment based on the review of each patient's case report form (CRF), which had been completed by each respective clinical investigator. Review of radiographical images in assisting the IRC's assessment was not performed in the majority of cases. Therefore, the IRC's review was based solely on investigator's measurements and observations. Based on these observations, it could be considered that the IRC assessment was a confirmation of the results obtained by the algorithm applied to the clinical investigator assessments.

Also during the review, the IRC was instructed to following NCI-WG 1996 guidelines; however, it appears the IRC did not strictly adhere to these guidelines given the IRC indicated they "may have determined that transient changes were clinically insignificant and compatible with continued response instead of progressive disease". Furthermore, the IRC considered a response to begin on the date at which it was first evaluated and to end on the date on which an assessment of progressive disease was first recorded. This approach in methodology overestimates the length of time a patient is considered a responder, given that progression does not begin on the day it is evaluated, but rather at an earlier point in time.

Additionally, an examination of the responses assigned to each visit by IRC members suggests that differences existed in the way that IRC members applied the NCI-WG 1996 guidelines. Because different IRC readers were assigned to determine the responses of each patient, it is not possible to determine what the inter-reader variability may be, and whether it is considered acceptable or not.

Finally, some patients were assigned an overall assessment of response, when clear evidence of non-response was recorded on the submitted CRF. This oversight persisted after the sponsor requested two rounds of re-consensus readings due to a number of patients having responses of less than the required two months. Therefore, given investigator bias was not reduced through the IRC's independent review, and because the investigator calculated ORR in accordance with NCI-WG 1996 guidelines (i.e. requiring response to be present for a minimum of two months), Health Canada chose to focus on the investigators' assessment of the ORR for evaluation purposes.

The sensitivity analyses applied in determining the ORR were carried out using two methods. First, the investigator based responses were used to calculate the ORR. Second, an algorithm developed by the sponsor was used to determine an ORR value. The investigators' response rate for the DR group was 42% (compared to the IRC response rate of 58%). The concordance rate (71 %) between these two assessments was considered to be low. In more detail, the IRC and investigator assessments agreed on 21 responders and 21 non-responders. Yet, there was disagreement on the status of 17 patients. If the IRC had relied on independent measurements made from radiographic images, a discordance rate of this size may have been expected; however, the IRC assessment was based entirely on measurements performed by the investigators. In this case, it would be expected that the two assessments should have a higher concordance given the response is defined in the NCI-WG 1996 guidelines. As previously discussed above, one of the major differences in the investigator versus the IRC assessment is that the IRC panel could, based on expert judgement, classify a patient as a responder if they met the guidelines for a response in only one evaluation, which goes against the NCI-WG 1996 guidelines, which specify that a response must be observed for at least 2 months (a period that covers two evaluations). On the other hand, the investigator assessment would not consider a patient as a responder unless the guidelines were met for at least two consecutive evaluations spanning over an 8 week period. It is Health Canada's opinion that 56 days (8 weeks) is the minimum number of days which could reasonably fulfill the criteria of a response, as per NCI-WG 1996 guidelines.

In addition to the investigator sensitivity analysis, the sponsor also carried out an algorithmic assessment of response. The sponsor's sensitivity analysis calculated that the proportion of responders in the DR group was 37% with a concordance to the IRC assessment of 80%. There was disagreement between these two assessments on the response of 12 patients, all of whom were classified as responders by the IRC assessment but as non-responders by the sponsor's algorithmic approach.

Significant variation in ORR was observed by the three different assessment approaches. However, the minimum ORR calculated, per the sponsor's algorithm, was 37% for the DR group. This level of response exceeded the pre-selected target of 30% with a 99% confidence bound that excludes an ORR of 15%. Therefore, the sponsor did meet the primary endpoint, ORR. The IRC assessment was the protocol specified assessment. However this assessment was blinded only to investigators' assessments and could not be considered a true independent assessment as it relied on physical measurements made by investigators' (e.g., lymph node, spleen, and liver measurements), which were not confirmed by radiographic imaging. Therefore, for the reasons discussed above, the investigator assessment of response was considered as the more appropriate measure of ORR as it managed to adhere to the guidelines specified in the study protocol, but still allowed for expert judgement to be applied to the by visit assessments of response.

The analyses of secondary endpoints, 'time to response' and 'duration of response' indicated that response onset was achieved quickly with a median time to onset of 1.8 months in responders. Time to onset was 2.1 months in the DR group. The duration of response for DR responders was 7.1 months by IRC assessment but 5.1 months by sensitivity analysis. The duration of response analysis indicates that the response to Arzerra is not maintained for a long period of time once treatment is complete; however, this is to be expected as Arzerra is a targeted therapy that cannot be expected to continue to reduce or maintain the number of malignant B-cells once treatment is withdrawn and circulating Arzerra is cleared.

In the pivotal study, no endpoint was specified to measure the patient's quality of life. However, an analysis of the individual components of the NCI-WG 1996 criteria for ORR in patients with B-cell CLL indicates that a large proportion of patients experienced resolution of constitutional symptoms that were present at baseline. Those who did not have symptoms at baseline remained symptom free for the duration of study. Furthermore, many patients experienced improvement or resolution in one or more of the following parameters: lymphadenopathy; splenomegaly; hepatomegaly; and haematological parameters.

3.3.4 Clinical Safety

The safety of Arzerra in relapsed/refractory CLL population was evaluated in one pivotal study previously described in section 3.3.3 Clinical Efficacy and in addition one Phase I/II dose-escalation study. For the pivotal study, while efficacy data was based on an interim analysis of 59 patients; the analysis of safety data was based on the fully enrolled population of 223 patients.

The assessment of the safety of Arzerra monotherapy was difficult due to the uncontrolled, unblinded design of the pivotal study. Given the single-arm design, it was impossible to discern whether causal relationships existed between observed adverse events (AEs) and exposure to Arzerra. It is possible that many AEs could be manifestations of the underlying illness that given these patients were heavily pre-treated and in many cases immunocompromised. Furthermore, the number of patients treated within the pivotal study was small, making the detection of rarer events difficult.

In the pivotal study, reporting of AEs occurred following the first dose administration (visit 2) until the final clinic visit at Month 24 (end of follow-up period). Only Serious Adverse Events (SAEs) were reported once a patient entered the extended follow-up period (Month 24-48); therefore, AEs are likely to have been under-reported during this period. Also, the study protocol allowed for patients who withdrew from the study during treatment to complete the Visit 21 (end of follow-up) assessments and immediately enter into the extended follow-up period. This permitted a number of patients to continue directly into the extended-follow up period after only a short period of time had passed since their final Arzerra administration. This design may have contributed to under-reporting of AEs that would have otherwise been reported had these patients continued in the treatment or follow-up period.

The most common adverse reactions (≥10%) during therapy with Arzerra were cough, pyrexia, anaemia, neutropaenia, diarrhoea, fatigue, dyspnoea, pneumonia, chills, nausea, bronchitis, peripheral oedema, back pain, and upper respiratory tract infection. The majority of patients (62%) had at least one adverse event (AE) ≥Grade 3 in severity, with the DR group showing a higher proportion of Grade 3 AEs than the BFR group (72% versus 52%). The 'Other' group had the highest number of ≥Grade 3 AEs at 76%, but this was based on a small sample size [number (n) = 17]. A total of 67% of patients had an AE considered to be related to Arzerra treatment; however, the AE frequency and patterns appeared to be typical for a monoclonal antibody treatment in a refractory CLL population.

Overall, 59% of patients experienced SAEs. A total of 17% of patients had SAEs that were considered to be drug-related. The most common drug-related SAEs were neutropaenia, pneumonia, and sepsis.

Of the 223 patients enrolled in the pivotal study, 138 died as of the safety analysis cut-off date. The most common cause of death was disease progression (27 patients) followed by septic complications (20 patients). All patients who experienced septic complications died (sepsis n = 10; neutropenic sepsis n = 5; escherichia sepsis n = 1; bacterial urosepsis n = 1; septic shock n = 3). Pneumonia was responsible for 15 (7%) deaths. One case of progressive multifocal leukoencephalopathy (PML), which was considered drug-related by the investigator, resulted in death. Cases of PML have been documented in CLL patients receiving either fludarabine or chlorambucil, and are most commonly observed in immunocompromised individuals such as human immunodeficiency virus (HIV) infected individuals. Therefore, it is unclear whether the manifestation of PML is due in this case to Arzerra exposure or due to the underlying disease. Progressive multifocal leukoencephalopathy has also been observed in patients receiving other anti-CD20 treatment. As such, a cautionary approach should be taken for any patient that exhibits neurological symptoms. Patients should be monitored for signs of PML and appropriate action should be taken if neurological symptoms manifest. The use of Arzerra is contraindicated in patients who have PML or have had PML. This is specified in the serious warnings and precautions section of the Product Monograph.

Development of infections was a common occurrence during treatment with Arzerra and subsequent follow-up; however, it must also be noted that the rate of infections in persons with advanced CLL is also high. Of the 223 patients enrolled, 163 (73%) patients reported 392 infections which were either bacterial, viral, or fungal in nature. A total of 43 (19%) patients experienced Grade 3 infections and 16 (7%) patients experienced Grade 4 infections. Of 223 patients, [22 (10%)] had fatal infections. The most frequent types of infections reported were of the respiratory tract, with lower respiratory tract infections being more common than upper. Respiratory tract infections (all pneumonia) were the most common cause of death by infection. Due to the large number of infections observed in the pivotal study and the number of cases of neutropaenia (discussed below) considered to be related to study drug exposures, it should be assumed that Arzerra may contribute to the acquisition of infections in the CLL population. Infection was the most common reason for drug discontinuation.

Infusion reactions were notably observed in both the pivotal and supportive study. These reactions included anaphylactic reactions, bronchospasm, cardiac events, chills/rigors, cough, cytokine release syndrome, diarrhoea, dyspnoea, fatigue, flushing, hypertension, hypotension, nausea, pain, pyrexia, rash, and urticaria. In the majority of cases, infusion reactions were generally mild in nature and did not require study withdrawal. Furthermore, none of the infusion reactions resulted in death. Pre-medication may help to attenuate infusion reactions, but reactions may still occur especially during the first two infusions.

Hematologic AEs were also commonly noted during Arzerra treatment. Neutropaenia occurred in 17% of patients and was considered by investigators to be related to treatment in 13% of patients. Three of the neutropaenia events were fatal. These included one neutropaenic sepsis, one Grade 5 neutropaenia, and one grade 5 febrile neutropaenia. Neutropaenia has been associated with other anti-CD20 monoclonal antibody therapies; however, it is unlikely that Arzerra is directly toxic to neutrophils since these cells do not express CD20. Nevertheless, Arzerra may act as a trigger for neutropaenia via other mechanisms. Overall, patients with baseline neutropaenia did not have a decrease in the median neutrophil counts; however, they also did not show any improvement in the median neutrophil count. Neutropaenia is common in heavily treated CLL patients. Therefore, it may be important to manage this complication in patients that present with baseline neutropaenia and those that develop neutropaenia to prevent serious infections from occurring.

Serious and fatal cardiovascular events were reported following administration of Arzerra. These events included, myocardial infarction, myocardial ischaemia, atrial fibrillation, perimyocarditis, ventricular systolic dysfunction with cardiac failure, hypotension leading to heart failure, tachyarrhythmia, and tachycardia. Fatal cardiac events (myocardial infarction, cardiac arrest, and cardiac failure) occurred in four patients during the treatment or follow-up periods of the pivotal study. One fatal case of myocardial infarction occurred within one day of the fourth infusion of Arzerra. Given these events and the proximity with which several occurred to study drug administration, patients with a history of cardiac disease should be monitored closely during and after infusions and resuscitative measures should be readily available. Arzerra should be discontinued in patients who experience serious or life-threatening cardiac arrhythmias.

Tumour lysis syndrome was observed in one patient within the pivotal study during administration of the first Arzerra infusion. The infusion was stopped and restarted later after the patient had recovered. Therefore, tumour lysis syndrome may occur in patients with high tumour burden when they are treated with a tumour lysing agent, such as Arzerra.

3.3.5 Additional Issues

Under the Notice of Compliance with Conditions (NOC/c) policy, additional data is required to confirm the clinical benefit and safety of Arzerra. The sponsor has agreed to submit the following:

  • Data from the completed pivotal study Hx-CD20-406, a single-arm international, multicentre study of HuMax-CD20, a fully human monoclonal anti-CD20 antibody, in patients with B-cell chronic lymphocytic leukaemia who have failed fludarabine and alemtuzumab.
  • Data from the completed study OMB11091, Phase III open-label, randomized, multicentre study of ofatumumab added to chlorambucil versus chlorambucil monotherapy in previously untreated patients with chronic lymphocytic leukaemia.
  • Data from the completed study OMB112855, a study of QTc intervals in patients who have been administered ofatumumab.
  • Conduct an assessment of QTc intervals as a sub-study in the clinical study OMB110911.
  • Conduct an assessment of anti-drug antibody (ADA) response to ofatumumab with a validated assay capable of sensitively detecting ADA responses in the presence of ofatumumab levels that are expected to be present at the time of patient sampling.
  • Submit to Health Canada any other analyses that have been designated as post-marketing commitments to other international authorization granting agencies.
  • Provide information in relation to communications and consultations with other agencies, when available.
  • Provide safety updates for all on-going Arzerra studies.
  • Provide reports of all serious adverse reactions that occur in Canada and all serious unexpected adverse reactions that occur outside of Canada within fifteen days to Health Canada.
  • Submit Periodic Safety Reports for Arzerra (ofatumumab) on a semi-annual basis until such time as conditions associated with the market authorization are removed.
  • Implement the Risk Management Plan (RMP) in Canada and provide any updates to the RMP when available. The Risk Management Plan is currently under review by Health Canada's Marketed Health Products Directorate (MHPD). The sponsor should comply with the recommendations of the MHPD in regards to the RMP.
  • Comply with the notification and reporting of specific issues of concern as outlined in Section 6.2.4 of the Guidance for Industry: Notice of Compliance with Conditions (NOC/c) and section 6Eiii of the NOC/c policy.
  • Provide a complete listing of ongoing additional clinical studies related to Arzerra, appended to the draft Letter of Undertaking, as per Section 4.5 of the Guidance for Industry: Notice of Compliance with Conditions (NOC/c).
  • Provide copies of any marketing authorisations or other regulatory actions for Arzerra from any other drug regulatory authority as per Section 4.6 of the Guidance for Industry: Notice of Compliance with Conditions (NOC/c).

3.4 Benefit/Risk Assessment and Recommendation

3.4.1 Benefit/Risk Assessment

Benefit

The treatment of patients with double refractory (to fludarabine/alemtuzumab) B-cell CLL was evaluated by applying the NCI-WG 1996 guidelines to determine the ORR, which takes into account both tumour load and haematopoietic functioning. In this population (n = 59), an ORR of 58% was determined by the protocol specified IRC evaluation and an ORR of 42% was determined from the investigator's clinical assessments. For reasons specified in the review, it is considered that the investigator's ORR is the more appropriate assessment for labelling purposes. In either case, the sponsor has shown that Arzerra, administered at the specified dosing regimen, resulted in ORRs that exceeded the pre-specified ORR of 30%. In addition, the lower 99% confidence bound exceeded the pre-specified target of 15% in both cases. Therefore, it is considered that the results obtained from the pivotal study demonstrated both clinical significance and statistical significance. Additionally, improvements of various endpoints that make up the ORR were observed in some patients that were not considered to be responders. Therefore, patients that do not achieve clinical response may also experience some benefit from Arzerra monotherapy.

Important to note however, is the small, single-arm open label design of the pivotal study. These design characteristics did not allow for comparisons versus currently available treatment options. Nonetheless, there is currently no authorized treatment option available to patients classified as fludarabine/alemtuzumab refractory and these patients can only be treated by salvage therapy. Furthermore, due to the relative rarity of patients with this designation, it would be difficult to recruit the number of patients required for a large randomised double-blind study of Arzerra monotherapy versus salvage therapies. Lastly, salvage therapies may not be appropriate for a large proportion of these patients due to baseline characteristics (neutropaenia) that would prevent them from receiving many cytotoxics. Therefore, the study design has been considered acceptable to support the proposed indication, but it is also important to point out that the submitted data is based on an interim analysis of the pivotal study, which was on-going at the time of submission.

Risk

The design of the pivotal study was considered problematic as it did not allow for the identification of safety concerns for which the proposed patient population may be at increased risk. However, several concerning safety issues were identified in the review of the pivotal and supporting studies. Notably, there were high incidences of infusion reactions (despite pre-medication), and infections. Infusion reactions were generally mild and did not require study withdrawal in the majority of cases. Additionally, no infusion reactions resulted in death; however, several deaths did occur within close proximity to infusion. Although, these were not considered to be due to infusion reactions, it is important that patients be monitored carefully, particularly those with a history of cardiovascular and pulmonary diseases. Deaths related to infection were frequent in the pivotal study, occurring in 10% of patients during the treatment or follow-up phase. Neutropaenia, often considered to be related to treatment by local investigators, was also common. As such, it is very important to weigh the risks and benefits of treating patients with active infections or baseline neutropaenia, which can predispose patients to serious infections. While several issues were identified in the study, it is also recognized that these conditions are common complications of advanced B-cell CLL. Given the prognosis facing patients at this stage in their disease, the AE profile appears to be acceptable. Further study in the B-cell CLL population is recommended to determine where Arzerra increases the risk of SAEs.

Overall Assessment

Patients with advanced B-cell CLL who are refractory to fludarabine and alemtuzumab represent a group of patients with a poor prognosis for which no authorised therapy exists. Current, treatment strategies are limited to salvage therapy, which carries with it significant risks and has thus far, provided little benefit to patients. Therefore, there is a need for new, effective therapies for these patients. Treatment of this population with Arzerra resulted in an ORR of 42% as assessed by local investigator while improvements in many parameters of response were observed in patients that were not considered to be responders. These results are supported by a similarly high ORR observed in a supportive study. These responses were considered to be durable with a median duration of response, based on investigator's assessments, of 6.5 months. Although, a high number of infections, and death due to infection were observed, in addition to infusion reactions, and haematological disturbances, treating patients with Arzerra did not result in patients experiencing AEs, or SAEs that are not normally seen in patients with advanced, heavily pre-treated disease. Therefore, the overall benefit/risk assessment for Arzerra in double-refractory B-cell CLL patients 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 Arzerra is favourable for the treatment of patients with chronic lymphocytic leukaemia (CLL) refractory to fludarabine and alemtuzumab. The efficacy of Arzerra is based on the demonstration of durable objective responses. No data demonstrate an improvement in disease related symptoms or increased survival with Arzerra. This New Drug Submission (NDS) qualifies for authorization under the Notice of Compliance with Conditions (NOC/c) Policy. The New Drug Submission complies with the requirements of sections C.08.002 and C.08.005.1 and therefore Health Canada has granted the Notice of Compliance (NOC) pursuant to section C.08.004 of the Food and Drug Regulations.

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

  • Data from the completed pivotal study Hx-CD20-406, a single-arm international, multicentre study of HuMax-CD20, a fully human monoclonal anti-CD20 antibody, in patients with B-cell chronic lymphocytic leukaemia who have failed fludarabine and alemtuzumab.
  • Data from the completed study OMB110911, Phase III open-label, randomized, multicentre study of ofatumumab added to chlorambucil versus chlorambucil monotherapy in previously untreated patients with chronic lymphocytic leukaemia
  • Data from the completed study OMB112855, a study of QTc intervals in patients who have been administered ofatumumab.
  • Conduct an assessment of QTc intervals as a sub-study in the clinical study OMB110911.
  • Conduct an assessment of anti-drug antibody (ADA) response to ofatumumab with a validated assay capable of sensitively detecting ADA responses in the presence of ofatumumab levels that are expected to be present at the time of patient sampling.
  • Submit to Health Canada any other analyses that have been designated as post-marketing commitments to other international authorisation granting agencies.
  • Provide information in relation to communications and consultations with other agencies, when available.
  • Provide safety updates for all on-going Arzerra studies.
  • Provide reports of all serious adverse reactions that occur in Canada, and all serious unexpected adverse reactions that occur outside of Canada within 15 days to Health Canada.
  • Submit Periodic Safety Reports for Arzerra (ofatumumab) on a semi-annual basis until such time as conditions associated with the market authorisation are removed.
  • Implement the Risk Management Plan (RMP) in Canada and provide any updates to the RMP when available. The Risk Management Plan is currently under review by Health Canada's Marketed Health Products Directorate (MHPD). The sponsor should comply with the recommendations of the MHPD in regards to the RMP.
  • Comply with the notification and reporting of specific issues of concern as outlined in Section 6.2.4 of the Guidance for Industry: Notice of Compliance with Conditions (NOC/c) and section 6Eiii of the NOC/c policy.
  • Provide a complete listing of ongoing additional clinical studies related to Arzerra, appended to the draft Letter of Undertaking, as per Section 4.5 of the Guidance for Industry: Notice of Compliance with Conditions(NOC/c).
  • Provide copies of any marketing authorisations or other regulatory actions for Arzerra from any other drug regulatory authority as per Section 4.6 of the Guidance for Industry: Notice of Compliance with Conditions (NOC/c).

4 Submission Milestones

Submission Milestones: ArzerraTM

Submission MilestoneDate
Pre-submission meeting:2010-08-26
Submission Filed:2011-01-21
Screening
Screening Acceptance Letter issued:2011-03-14
Review
On-Site Evaluation:
Quality Evaluation complete:2012-03-08
Clinical Evaluation complete:2012-03-07
Labelling Review complete:2012-03-07
Notice of Compliance with Conditions (NOC/c) Qualifying Notice (NOC/c-QN) issued2012-01-06
Response filed:2012-02-06
Notice of Compliance (NOC) issued by Director General under the NOC/c Policy:2012-03-09