Summary Basis of Decision for Tezspire

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

Summary Basis of Decision (SBD) documents provide information related to the original authorization of a product. The SBD for Tezspire is located below.

Recent Activity for Tezspire

SBDs written for eligible drugs approved after September 1, 2012 will be updated to include post-authorization information. This information will be compiled in a Post-Authorization Activity Table (PAAT). The PAAT will include brief summaries of activities such as submissions for new uses of the product, and whether Health Canada's decisions were negative or positive. PAATs will be updated regularly with post-authorization activity throughout the product's life cycle.

Post-Authorization Activity Table (PAAT) for Tezspire

Updated:

2022-11-21

The following table describes post-authorization activity for Tezspire, a product which contains the medicinal ingredient tezepelumab. For more information on the type of information found in PAATs, please refer to the Frequently Asked Questions: Summary Basis of Decision (SBD) Project: Phase II and to the list of abbreviations that are found in PAATs.

For additional information about the drug submission process, refer to the Management of Drug Submissions and Applications Guidance.

Drug Identification Number (DIN):

  • DIN 02529548 - tezepelumab 110 mg/mL, solution, subcutaneous administration (single-use prefilled syringe)
  • DIN 02529556 - tezepelumab 110 mg/mL, solution, subcutaneous administration (single-use prefilled pen)

Post-Authorization Activity Table (PAAT)

Activity/submission type, control numberDate submittedDecision and dateSummary of activities
Drug product (DINs 02529548, 02529556) market notificationNot applicableDate of first sale:
2022-10-19

The manufacturer notified Health Canada of the date of first sale pursuant to C.01.014.3 of the Food and Drug Regulations.

NDS # 2561882021-08-27Issued NOC
2022-07-28

NOC issued for New Drug Submission.

Summary Basis of Decision (SBD) for Tezspire

Date SBD issued: 2022-11-21

The following information relates to the new drug submission for Tezspire.

Tezepelumab

Drug Identification Number (DIN):

  • DIN 02529548 - tezepelumab 110 mg/mL, solution, subcutaneous administration (single-use prefilled syringe)
  • DIN 02529556 - tezepelumab 110 mg/mL, solution, subcutaneous administration (single-use prefilled pen)

AstraZeneca Canada Inc.

New Drug Submission Control Number: 256188

On July 28, 2022, Health Canada issued a Notice of Compliance to AstraZeneca Canada Inc. for the drug product Tezspire.

The market authorization was based on quality (chemistry and manufacturing), non-clinical (pharmacology and toxicology), and clinical (pharmacology, safety, and efficacy) information submitted. Based on Health Canada’s review, the benefit‑risk profile of Tezspire is favourable for use as an add-on maintenance treatment in adults and adolescents aged 12 years and older with severe asthma.

1 What was approved?

Tezspire is a human monoclonal antibody directed against thymic stromal lymphopoietin. It was authorized for an add-on maintenance treatment in adults and adolescents aged 12 years and older with severe asthma. Tezspire is not indicated for relief of acute bronchospasm or status asthmaticus.

The safety and efficacy of Tezspire in children younger than 12 years of age have not been established.

Among the 665 patients with severe asthma who were treated with Tezspire in two clinical studies, 119 patients (18%) were 65 years of age and older. While the data are limited, no overall differences in safety or efficacy of Tezspire have been observed between patients 65 years of age and older and those younger that 65 years of age.

Tezspire (tezepelumab 110 mg/mL) is presented as a solution, supplied in a prefilled syringe or a prefilled pen. The non-medicinal ingredients include glacial acetic acid, L-proline, polysorbate 80, sodium hydroxide, and water for injection. Each single-use prefilled syringe or prefilled pen contains 210 mg tezepelumab in 1.91 mL deliverable volume.

The use of Tezspire is contraindicated in patients who are hypersensitive to this drug or to any ingredient in the formulation, including any non-medicinal ingredient, or component of the container.

The drug product was approved for use under the conditions stated in its Product Monograph taking into consideration the potential risks associated with its administration. The Tezspire Product Monograph is available through the Drug Product Database.

For more information about the rationale for Health Canada's decision, refer to the Clinical, Non-clinical, and Quality (Chemistry and Manufacturing) Basis for Decision sections.

2 Why was Tezspire approved?

Health Canada considers that the benefit-risk profile of Tezspire is favourable for use as an add-on maintenance treatment in adults and adolescents aged 12 years and older with severe asthma. Tezspire is not indicated for relief of acute bronchospasm or status asthmaticus.

Asthma is a heterogeneous disease, usually characterized by chronic airway inflammation. It is defined by the history of respiratory symptoms, such as wheeze, shortness of breath, chest tightness, and cough that vary over time and in intensity, together with variable expiratory flow limitations. Asthma may also be characterized by exacerbations, which are episodes of worsening symptoms and lung function. Severe asthma is defined as asthma that requires treatment with high-dose inhaled corticosteroids plus a second controller (e.g., long-acting beta-agonists) and/or oral corticosteroids to prevent it from becoming uncontrolled or which remains uncontrolled despite this therapy.

More than 3.8 million Canadians are affected by asthma. Asthma attacks result in over 70,000 emergency room visits and account for approximately 250 deaths in Canada each year. Severe asthma is estimated to affect between 150,000 and 250,000 Canadians.

Currently authorized biologics that are indicated for the treatment of subgroups of patients with severe asthma include the anti-immunoglobulin E (anti-IgE) monoclonal antibody omalizumab (Xolair), the anti-interleukin-5 (anti-IL-5) monoclonal antibodies, mepolizumab (Nucala) and reslizumab (Cinqair), the anti-IL-5 receptor monoclonal antibody benralizumab (Fasenra), and the anti-IL-4 and anti-IL-13 monoclonal antibody dupilumab (Dupixent).

Tezepelumab, the medicinal ingredient in Tezspire, is a novel human monoclonal antibody that binds to human thymic stromal lymphopoietin (TSLP) with high affinity and prevents its interaction with the heterodimeric TSLP receptor. Human TSLP is an epithelial cytokine that occupies an upstream position in the asthma inflammatory cascade. Blocking TSLP with tezepelumab reduces biomarkers and cytokines associated with inflammation, including blood eosinophils, airway submucosal eosinophils, IgE, fractional exhaled nitric oxide (FeNO), IL-5, and IL-13. However, the primary pharmacodynamic effect of tezepelumab in asthma remains to be definitively established.

Tezspire has been shown to be efficacious as an add-on maintenance treatment in adults and adolescents aged 12 years and older with severe asthma. The market authorization was primarily based on efficacy and safety data derived from three core studies: two asthma exacerbation studies (the pivotal study, NAVIGATOR, and the dose-ranging study, PATHWAY) and one oral corticosteroid-reduction study, SOURCE.

The pivotal study was a Phase III, randomized, double-blind, placebo-controlled study, with a 52-week treatment period. It included 1,059 patients (82 adolescents and 977 adults) with inadequately controlled severe asthma. Patients were randomized to receive Tezspire 210 mg (528 patients) or placebo (531 patients) administered subcutaneously every four weeks. The dosing regimen of Tezspire was chosen based on results obtained in the Phase IIb, dose-ranging study (PATHWAY), which evaluated three different dose levels of Tezspire.

Treatment with Tezspire resulted in a clinically meaningful and statistically significant reduction in the primary efficacy endpoint of annualized asthma exacerbation rate (AAER) over 52 weeks compared with placebo (AAER rate ratio of 0.44; 95% confidence interval [CI]: 0.37, 0.53; p < 0.001). The reduction in AAER over 52 weeks was observed in Tezspire-treated patients across clinically relevant subgroups, including patients with low blood eosinophil counts. Additionally, treatment with Tezspire led to clinically meaningful and statistically significant improvements in key secondary endpoints, including lung function (as measured by forced expiratory volume in the first second [FEV1]; the difference from placebo in the mean change from baseline in FEV1 was 0.13 L [95% CI: 0.08, 0.18]). Clinically meaningful improvements from baseline that were statistically significant versus placebo were also found in two clinically relevant patient-reported outcomes: asthma-related quality of life, as assessed by the Asthma Quality of Life Questionnaire (Standardized) for 12 years and older, AQLQ(S)+12; and asthma control, as assessed by the Asthma Control Questionnaire, ACQ-6.

While the small subgroup of 82 adolescents (41 of whom received Tezspire) was not powered to demonstrate statistical significance for the primary endpoint, it showed a decrease in the AAER over 52 weeks compared to placebo (AAER rate ratio of 0.70 [95% CI: 0.34, 1.46]). Lung function results in this subgroup also favoured Tezspire.

The Phase III oral corticosteroid-reduction study (SOURCE) enrolled 150 patients (18 years of age and older) who required treatment with daily oral corticosteroids (7.5 mg to 30 mg per day) in addition to regular use of high-dose inhaled corticosteroids and a long-acting beta-agonist with or without additional asthma controller(s). The study failed to meet its primary endpoint, the percent reduction from baseline in daily oral corticosteroid dose at Week 48 while not losing asthma control. While the primary endpoint was not met, secondary endpoints, including the AAER, showed improvements with Tezspire compared with placebo despite patients reducing their maintenance oral corticosteroid dose. Subgroup analyses for patients on maintenance oral corticosteroids in NAVIGATOR further support the efficacy trends noted in SOURCE. Based on these results, a limitation of use of Tezspire in patients on maintenance oral corticosteroids was not included in the Tezspire Product Monograph.

The safety evaluation of Tezspire was primarily based on pooled data from the studies NAVIGATOR and PATHWAY, including 665 patients with severe uncontrolled asthma treated with Tezspire for a period of up to 52 weeks. The overall incidence of adverse events was similar between Tezspire and placebo treatment groups. The most commonly reported adverse events were nasopharyngitis, upper respiratory tract infection, headache, and asthma. Fewer subjects in the Tezspire group experienced serious adverse events compared to the placebo group.

Adverse events of special interest included infections, serious hypersensitivity reactions, and malignancy. There were no safety signals or imbalances between groups relating to these safety concerns in the clinical programme. Notably, data submitted from an extension trial showed an imbalance in serious cardiac adverse events associated with Tezspire treatment; however, there does not appear to be a causal link between these occurrences and Tezspire.

A Risk Management Plan (RMP) for Tezspire was submitted by AstraZeneca Canada Inc. to Health Canada. The RMP is designed to describe known and potential safety issues, to present the monitoring scheme and when needed, to describe measures that will be put in place to minimize risks associated with the product. Upon review, the RMP was considered to be acceptable.

The submitted inner and outer labels, package insert, and Patient Medication Information section of the Tezspire Product Monograph meet the necessary regulatory labelling, plain language, and design element requirements.

The sponsor submitted a brand name assessment that included testing for look-alike sound-alike attributes. Upon review, the proposed name Tezspire was accepted.

Overall, there were no safety concerns that outweighed the efficacy benefits provided by Tezspire as an add-on maintenance treatment in adults and adolescents aged 12 years and older with severe asthma. The potential risks are considered mitigated through appropriate labelling in the Tezspire Product Monograph and pharmacovigilance activities.

This New Drug Submission complies with the requirements of sections C.08.002 and C.08.005.1 and therefore Health Canada has issued the Notice of Compliance pursuant to section C.08.004 of the Food and Drug Regulations. For more information, refer to the Clinical, Non-clinical, and Quality (Chemistry and Manufacturing) Basis for Decision sections.

3 What steps led to the approval of Tezspire?

The review of the clinical and quality components of the New Drug Submission (NDS) for Tezspire was based on a critical assessment of the data package submitted to Health Canada. The review completed by the United States Food and Drug Administration (FDA) and the sponsor’s correspondence with the FDA or the European Medicines Agency (EMA) were used as added references, as per Method 3 described in the Draft Guidance Document: The Use of Foreign Reviews by Health Canada. Additionally, the review of the non-clinical component of the NDS for Tezspire was based on a critical assessment of the review conducted by the FDA, in accordance with Method 1 described in the aforementioned guidance document.

The Canadian regulatory decision regarding the Tezspire NDS was made independently based on the Canadian review.

For additional information about the drug submission process, refer to the Management of Drug Submissions and Applications Guidance.

Submission Milestones: Tezspire

Submission MilestoneDate
Pre-submission meetings2021-06-09
Pre-submission meetings2021-06-18
New Drug Submission filed2021-08-27
Screening
Screening Acceptance Letter issued2021-10-04
Review
Quality evaluation completed2022-06-30
Review of Risk Management Plan completed2022-07-08
Non-clinical evaluation completed2022-07-22
Clinical/medical evaluation completed2022-07-22
Labelling review completed2022-07-26
Biostatistics evaluation completed2022-07-28
Notice of Compliance issued by Director General, Biologic and Radiopharmaceutical Drugs Directorate2022-07-28

4 What follow-up measures will the company take?

Requirements for post-market commitments are outlined in the Food and Drugs Act and Regulations.

6 What other information is available about drugs?

Up-to-date information on drug products can be found at the following links:

7 What was the scientific rationale for Health Canada's decision?
7.1 Clinical basis for decision

Clinical Pharmacology

Tezepelumab, the medicinal ingredient in Tezspire, is a human monoclonal antibody directed against human thymic stromal lymphopoietin (TSLP).

Pharmacodynamic data of tezepelumab in asthma patients were obtained from a Phase II study, CASCADE, a Phase IIb dose-ranging study, PATHWAY, and the pivotal Phase III study, NAVIGATOR (the pivotal study is described in the Clinical Efficacy section). These data showed that the administration of tezepelumab reduced inflammatory mediators such as blood eosinophils, immunoglobulin E (IgE), fractional exhaled nitric oxide (FeNO), interleukin-5 (IL-5), and interleukin-13 (IL-13). However, the primary pharmacodynamic effect of tezepelumab in asthma remains to be definitively established.

Pharmacokinetic data of tezepelumab were derived from six Phase I studies, the PATHWAY study, the NAVIGATOR study, and the Phase III study PATH-HOME.

In single-ascending-dose and multiple-ascending-dose studies, healthy adult subjects received up to 420 mg of tezepelumab administered subcutaneously and up to three doses of 700 mg administered intravenously every four weeks. Exposure to tezepelumab was found to be dose proportional following subcutaneous administration over a dose range of 2.1 mg to 420 mg. After repeated subcutaneous administration every four weeks, serum tezepelumab concentrations approached steady state by 12 weeks, with an estimated mean accumulation ratio of 1.86. The estimated absolute bioavailability was 77%.

A Phase I comparative bioavailability study (PATH-BRIDGE) demonstrated that tezepelumab exposures following single-dose subcutaneous administration via the to-be-marketed prefilled syringe and autoinjector presentations or via the vial presentation used in the NAVIGATOR study were within the bioequivalence margins specified in the relevant Health Canada’s guidance document. Consistent exposures were also observed between at-home or in-clinic administration of tezepelumab via either the prefilled syringe or autoinjector in the PATH-HOME study.

The population pharmacokinetic model submitted by the sponsor was a two-compartment model with first-order absorption and elimination. The model was appropriately parameterized and provided accurate fits to the observed data. For the recommended dosing regimen of 210 mg tezepelumab administered subcutaneously every four weeks to an individual with a body weight of 70 kg, the model-predicted mean (standard deviation) exposure values at steady state were 40.9 (14) µg/mL for the maximum serum concentration (Cmax) and 901 (328) µg*day/mL for the area under the concentration-time curve (AUC). The estimated half-life of tezepelumab was approximately 26 days, which was consistent with observed data.

The recommended dosing regimen of 210 mg tezepelumab administered subcutaneously every four weeks was also supported by two exposure-response models derived from pharmacokinetic data collected in PATHWAY and NAVIGATOR, respectively. The models show that the predicted tezepelumab exposures for this dosing regimen lie on the plateau phase of the exposure-response curve.

In the population pharmacokinetic analysis, body weight was the most influential covariate for tezepelumab exposure, as higher body weight was shown to be associated with lower exposure. An increase in exposure was also observed in adolescent patients compared with adults; body weight was the main driver of this difference. However, the model-predicted exposures of tezepelumab in an adolescent weighing 30 kg were consistent with the exposures observed in patients treated with the dosing regimen of 280 mg of tezepelumab administered subcutaneously every two weeks in the PATHWAY study. This dosing regimen was well tolerated and no dose-dependent safety signals were observed. Accordingly, no dose adjustment of tezepelumab is required in terms of body weight.

Tezepelumab exhibited low immunogenicity across the clinical studies. In the pivotal study (NAVIGATOR), 26 out of 527 subjects (4.9%) tested positive for anti-drug antibodies (ADAs), with treatment-emergent ADAs occurring in 10 out of 522 subjects (1.9%). Only one subject had neutralizing ADAs. There was no apparent effect of ADAs on the pharmacokinetics or the safety profile of tezepelumab.

The clinical pharmacology data support the use of Tezspire for the recommended indication.

For further details, please refer to the Tezspire Product Monograph, approved by Health Canada and available through the Drug Product Database.

Clinical Efficacy

The clinical efficacy of Tezspire as an add-on maintenance treatment in adults and adolescents aged 12 years and older with severe asthma is primarily supported by data derived from three core studies: two asthma exacerbation studies (the pivotal study, NAVIGATOR, and the dose-ranging study, PATHWAY) and one oral corticosteroid-reduction study, SOURCE.

The pivotal study was a Phase III, randomized, double-blind, placebo-controlled study, with a 52-week treatment period. It included 1,059 patients (82 adolescents and 977 adults) with inadequately controlled severe asthma. Patients had to have experienced two or more asthma exacerbations requiring oral or systemic corticosteroid treatment, or resulting in hospitalization in the 12 months prior to enrollment, despite receiving regular treatment with medium- or high-dose inhaled corticosteroids and at least one additional asthma control therapy with or without oral corticosteroids. Background asthma therapy was continued throughout the duration of the study. Notably, NAVIGATOR enrolled patients with a broad range of baseline blood eosinophil counts, total serum IgE levels, FeNO levels, and patients with allergic or non-allergic status, as determined by a positive serum IgE result specific to any perennial aeroallergen. Patients were randomized to receive Tezspire 210 mg (528 patients) or placebo (531 patients) administered subcutaneously every four weeks. The dosing regimen of Tezspire was chosen based on results obtained in the Phase IIb, dose-ranging study (PATHWAY), which evaluated three different dose levels of Tezspire.

The primary efficacy endpoint of the pivotal study was the annualized asthma exacerbation rate (AAER) over 52 weeks. An asthma exacerbation was defined as a worsening of asthma that led to any of the following: a temporary bolus/burst of systemic corticosteroids (or a temporary increase in a stable background dose of oral corticosteroids for at least 3 consecutive days); an emergency room or urgent care visit (that lasts less than 24 hours) due to asthma that required systemic corticosteroids; or an in-patient hospitalization (for at least 24 hours) due to asthma.

Treatment with Tezspire resulted in a clinically meaningful and statistically significant reduction of 56% in the AAER over 52 weeks compared with placebo (AAER rate ratio of 0.44; 95% confidence interval [CI]: 0.37, 0.53; p < 0.001). The reduction in AAER over 52 weeks was observed in Tezspire-treated patients across clinically relevant subgroups, including patients with low blood eosinophil counts. In Tezspire-treated patients who had blood eosinophil counts lower than 150 eosinophils/µL at baseline, the AAER over 52 weeks was reduced by 39% relative to placebo.

Compared to placebo, treatment with Tezspire resulted in benefits across multiple endpoints related to exacerbations, including a reduction of 79% in the rate of asthma exacerbations requiring hospitalizations and emergency room visits over 52 weeks, and a reduction of 85% in the rate of asthma exacerbations requiring hospitalization over 52 weeks.

Additionally, treatment with Tezspire led to clinically meaningful and statistically significant improvements in key secondary endpoints, including lung function (as measured by forced expiratory volume in the first second [FEV1]; the difference from placebo in the mean change from baseline in FEV1 was 0.13 L [95% CI: 0.08, 0.18]). Clinically meaningful improvements from baseline that were statistically significant versus placebo were also found in two clinically relevant patient-reported outcomes: asthma-related quality of life, as assessed by the Asthma Quality of Life Questionnaire (Standardized) for 12 years and older, AQLQ(S)+12; and asthma control, as assessed by the Asthma Control Questionnaire, ACQ‑6.

While the small subgroup of 82 adolescents (41 of whom received Tezspire) was not powered to demonstrate statistical significance for the primary endpoint, it showed a decrease of 30% in the AAER over 52 weeks compared to placebo (AAER rate ratio of 0.70 [95% CI: 0.34, 1.46]). Lung function results in this subgroup also favoured Tezspire.

The oral corticosteroid-reduction study, SOURCE, aimed to evaluate the effect of Tezspire 210 mg administered subcutaneously every four weeks versus placebo on reducing the maintenance oral corticosteroid dose in adult patients with severe, oral corticosteroid-dependent asthma. This was a Phase III, randomized, double-blind, placebo-controlled study with a 48-week treatment period. The study enrolled 150 asthma patients (18 years of age and older) who required treatment with daily oral corticosteroids (7.5 mg to 30 mg per day) in addition to regular use of high-dose inhaled corticosteroids and a long-acting beta-agonist with or without additional asthma controller(s). Of the 150 patients, 74 received Tezspire.

The primary endpoint was the categorized percent reduction from baseline in daily oral corticosteroid dose at Week 48 while maintaining asthma control. The categories of percent change from baseline in daily oral corticosteroid dose were ≥90% reduction, ≥75% to <90% reduction, ≥50% to <75% reduction, >0% to <50% reduction, and no change or no reduction in the daily oral corticosteroid dose. Treatment with Tezspire did not demonstrate a statistically significant reduction in maintenance oral corticosteroid dose compared with placebo (cumulative odds ratio [OR] = 1.28; 95% CI: 0.69, 2.35). While the primary endpoint was not met, secondary endpoints, including the AAER, showed improvements with Tezspire relative to placebo despite patients reducing their maintenance oral corticosteroid dose. Subgroup analyses for patients on maintenance oral corticosteroids in NAVIGATOR further support the efficacy trends noted in SOURCE. Based on these results, a limitation of use of Tezspire in patients on maintenance oral corticosteroids was not included in the Tezspire Product Monograph.

Indication

The New Drug Submission for Tezspire was filed by the sponsor with the following indication, which was subsequently approved by Health Canada:

  • Tezspire (tezepelumab injection) is indicated as an add-on maintenance treatment in adults and adolescents aged 12 years and older with severe asthma.
  • Tezspire is not indicated for relief of acute bronchospasm or status asthmaticus.

For more information, refer to the Tezspire Product Monograph, approved by Health Canada and available through the Drug Product Database.

Clinical Safety

The safety evaluation of Tezspire was based on data derived from the pooled safety population (primary safety pool) from the pivotal study NAVIGATOR (described in the Clinical Efficacy section) and the supportive dose-ranging study PATHWAY. The primary safety pool included 665 asthma patients who received Tezspire 210 mg via subcutaneous administration every four weeks for up to one year, representing approximately 640 patient-years of exposure.

The overall incidence of subjects with adverse events in the on-treatment period was similar between the Tezspire and placebo groups (74.6% and 76.5% of subjects, respectively). These adverse events were mostly non-serious in nature and of mild or moderate intensity.

The incidence of adverse events considered causally related to the investigational product by the investigator was 9.0% in the Tezspire group and 8.1% in the placebo group. Permanent discontinuation due to adverse events (including serious adverse events) occurred in 2.0% of patients in the Tezspire group and 3.0% of patients in the placebo group. The four most commonly reported adverse events in the Tezspire group were nasopharyngitis (19.5%), upper respiratory tract infection (9.3%), headache (7.8%), and asthma (7.4%); in comparison, incidences of patients with these adverse events in the placebo group were 19.1%, 13.3%, 7.5%, and 15.7%, respectively.

The incidence of serious adverse events in the on-treatment period was 8.6% in the Tezspire-treated patients and 13.0% in the placebo-treated patients. Asthma was the most commonly reported serious adverse event in both treatment groups, with an incidence of 2.3% in the Tezspire group versus 6.9% in the placebo group. Apart from asthma, no serious adverse events by a preferred term (as per the Medical Dictionary for Regulatory Activities [MedDRA]) were reported in more than two patients in the Tezspire group. The incidence of serious adverse events considered causally related to the investigational product was 0.9% in the Tezspire group and 0.7% in the placebo group.

Additionally, the adverse event profile of Tezspire appeared similar across predefined subgroups, including subgroups defined by baseline blood eosinophil count, allergic status (determined by a perennial aeroallergen-specific IgE), baseline inhaled corticosteroid dose, baseline oral corticosteroid dose, age, gender, body mass index, race, country, and region. Based on data derived from a small subgroup of adolescents (only 41 patients aged 12 to 17 years received Tezspire in NAVIGATOR), the safety profile of Tezspire was similar to that observed in Tezspire-treated adults.

In patients with oral corticosteroid-dependent asthma in the SOURCE study (described in the Clinical Efficacy section), Tezspire was well tolerated and demonstrated an adverse event profile similar to that seen in the primary safety pool.

In a long-term extension study, DESTINATION, the safety profile of Tezspire was generally similar to that observed in the primary safety pool. However, there was a notable increased incidence of serious adverse events in the MedDRA System Organ Class (SOC) Cardiac disorders among Tezspire-treated patients relative to placebo (exposure-adjusted incidence rates [95% CI]: 1.30 [0.77, 2.06] versus 0.23 [0.03, 0.83], respectively). Notably, all patients had risk factors for cardiac disease and some patients also had a confounding medical history. There was no pattern in specific MedDRA preferred terms (i.e., adverse events were spread across the MedDRA SOC Cardiac disorders). Similarly, the data did not reveal any pattern in time to onset for the serious cardiac events, there were no imbalances in the overall (serious and non-serious) cardiac events between treatment arms, no causal relationship to Tezspire treatment as deemed by both investigators and a blinded adjudication committee, and no identified pathophysiologic link based upon literature, non-clinical data, and data from other clinical studies. A small increase was also noted in fatalities reported in this study for Tezspire-treated patients relative to placebo; there was no causal association for this finding, nor was it observed in other clinical studies.

Based on the clinical data available at this time, the safety profile of Tezspire was considered acceptable. Appropriate warnings and precautions are in place in the approved Tezspire Product Monograph to address the identified safety concerns.

For more information, refer to the Tezspire Product Monograph, approved by Health Canada and available through the Drug Product Database.

7.2 Non-Clinical Basis for Decision

Tezepelumab, the medicinal ingredient in Tezspire, is a human monoclonal antibody directed against human thymic stromal lymphopoietin (TSLP).

In in vitro primary pharmacodynamic studies, tezepelumab was shown to bind to human TSLP and inhibit its binding to the heterodimeric TSLP receptor complex.

Key toxicology studies were conducted in cynomolgus monkeys, as tezepelumab did not bind to mouse, rat, or rabbit TSLP.

In a 3-month repeat-dose toxicity study, conducted mainly in sexually immature animals, tezepelumab was administered subcutaneously (the intended route of administration in humans). A delay and/or reduction in the immune response to an antigen challenge was observed at the highest dose tested (300 mg/kg body weight once weekly). Since this was not accompanied by changes indicative of infection, it was considered a non-adverse effect.

In a 6-month repeat-dose toxicity study in sexually mature animals, tezepelumab was administered subcutaneously or intravenously. The intravenous administration of tezepelumab at a dose of 50 mg/kg body weight once weekly led to the formation of circulating immune complexes in one animal, resulting in adverse vascular changes and poor clinical condition that necessitated early euthanasia of the animal. A similar finding was not observed following subcutaneous administration of tezepelumab at the same or higher dose.

No adverse effects were observed up to the highest subcutaneous dose tested in the 3-month and 6-month repeat-dose toxicity studies. Exposures (as measured by the area under the concentration-time curve) at the no-observed-adverse-effect level of 300 mg/kg body weight once weekly (both studies) were at least 85-fold greater than the estimated human exposures in both adults and adolescents.

In an enhanced prenatal and postnatal development study, the maternal, embryo-fetal, and postnatal toxicity of tezepelumab was assessed following intravenous administration to pregnant cynomolgus monkeys. No adverse effects on maternal health, pregnancy outcome, embryo-fetal development, or infant growth and development up to 6.5 months of age were observed. Tezepelumab was detected in infant serum throughout the postnatal period, indicating transport across the placenta. Tezepelumab was also detected in milk at concentrations lower than 1% of the maternal serum concentrations.

A dedicated juvenile toxicity study of tezepelumab was not conducted. Results of the 3-month repeat-dose toxicity study, which included sexually immature animals, did not reveal safety concerns with respect to the clinical use of tezepelumab in adolescents. The lack of a dedicated juvenile toxicity study was justified in accordance with relevant International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use (ICH) guidelines.

Carcinogenicity and genotoxicity studies were also not conducted, which was considered acceptable and in accordance with relevant ICH guidelines.

The results of the non-clinical studies as well as the potential risks to humans have been included in the Tezspire Product Monograph. In view of the intended use of Tezspire, there are no pharmacological or toxicological issues within this submission which preclude authorization of the product.

For more information, refer to the Tezspire Product Monograph, approved by Health Canada and available through the Drug Product Database.

7.3 Quality Basis for Decision

Characterization of the Drug Substance

Tezepelumab is a human monoclonal antibody (immunoglobulin G2 lambda [IgG2λ]) directed against the cytokine human thymic stromal lymphopoietin (TSLP). The monoclonal antibody binds to TSLP with high affinity and prevents its interaction with the heterodimeric TSLP receptor.

Tezepelumab contains two heavy chains and two light chains covalently linked through disulphide bonds. The overall molecular weight of tezepelumab is approximately 147 kDa.

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

Results from process validation studies indicate that the processing steps adequately control the levels of product- and process-related impurities. The impurities that were reported and characterized were found to be within established limits.

Manufacturing Process of the Drug Substance and Drug Product and Process Controls

Tezepelumab is produced in a genetically engineered Chinese hamster ovary (CHO) cell line. The manufacturing process begins with the thaw of a vial from the working cell bank, followed by stages of cell culture expansion prior to inoculating a production bioreactor. After purification of the harvested cell culture fluid through a series of chromatography and viral clearance steps, the obtained drug substance is stored frozen.

The drug product manufacturing process consists of thawing and pooling of the drug substance, drug product formulation, bioburden reduction filtration, sterile filtration, and aseptic filling into glass syringes. The glass syringes are assembled into a prefilled syringe system with a needle guard and a finger flange, or into an autoinjector/pen system. Each single-use, prefilled syringe or pen contains a solution of 210 mg tezepelumab in 1.91 mL (110 mg/mL). The final drug product is stored at 2 °C to 8 °C.

Process validation was conducted with several consecutive drug substance and drug product batches manufactured at the scale intended for the commercial processes. The process parameters, process attributes, release testing results, and stability testing results met predefined criteria, acceptance limits, and specifications for all process validation lots. Any deviations during process validation were investigated and shown to have no impact on the validation of the process. Overall, the process performance data demonstrate that the manufacturing processes are well controlled and capable of consistently yielding a drug substance and drug product of appropriate quality.

Control of the Drug Substance and Drug Product

The release and stability specifications for the drug substance and drug product include assays for identity, biological activity, purity, impurities, and safety. All in-house analytical procedures are validated and in compliance with relevant International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use (ICH) guidelines.

Tezspire is a Schedule D (biologic) drug and is, therefore, subject to Health Canada's Lot Release Program before sale as per the Guidance for Sponsors: Lot Release Program for Schedule D (Biologic) Drugs.

Stability of the Drug Substance and Drug Product

Based on the stability data submitted, the proposed shelf life and storage conditions for the drug substance and drug product were adequately supported and are considered satisfactory.

The proposed shelf life of 36 months for Tezspire, when stored at 2 °C to 8 °C and protected from light, is considered acceptable. Tezspire must not be frozen or shaken. After removal from the refrigerator, the drug product must be used within 30 days or discarded.

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.

Facilities and Equipment

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

Based on risk assessment scores determined by Health Canada, on-site evaluations of the drug substance and drug product manufacturing facilities were not deemed necessary.

The sites involved in production are compliant with good manufacturing practices.

Adventitious Agents Safety Evaluation

The manufacturing process of Tezspire incorporates adequate control measures to ensure freedom from adventitious microorganisms (bacteria, fungi, mycoplasma, and viruses). Purification process steps designed to remove and inactivate any potential viral contaminants from the cell culture process are adequately validated.

Apart from the mammalian, CHO-derived tezepelumab production cell line, materials of animal or human origin are not used in the manufacturing process. The risk of contamination of the drug product with bovine spongiform encephalopathy and transmissible spongiform encephalopathy agents is considered negligible.

No excipients of animal or human origin are present in the formulation of Tezspire.