U.S. flag

An official website of the United States government

NCBI Bookshelf. A service of the National Library of Medicine, National Institutes of Health.

Dupilumab (Dupixent): CADTH Reimbursement Review: Therapeutic area: Asthma [Internet]. Ottawa (ON): Canadian Agency for Drugs and Technologies in Health; 2023 Apr.

Cover of Dupilumab (Dupixent)

Dupilumab (Dupixent): CADTH Reimbursement Review: Therapeutic area: Asthma [Internet].

Show details

Clinical Review

Executive Summary

An overview of the submission details for the drug under review is provided in Table 1.

Table 1. Submitted for Review.

Table 1

Submitted for Review.

Introduction

Asthma is a chronic respiratory disorder characterized by reversible airway obstruction.1 In Canada, it is estimated that 14% and 19% of children aged 5 years to 9 years and 10 years to 14 years suffer from asthma, respectively.2 According to the clinical experts consulted for this CADTH review, asthma has several diverse phenotypes, 1 of which is primarily driven by type 2 inflammation, presenting with an allergic or atopic profile and/or eosinophilic asthma.

The management of asthma is traditionally carried out using “reliever” medication for the acute relief of exacerbations, combined with controllers used on a regular or chronic basis, in an effort to prevent the onset of exacerbations.1 Treatment of patients in Canada follows an asthma management continuum, with inhaled corticosteroids (ICSs) as the backbone of maintenance anti-inflammatory therapy, and other medications added on as necessary.3 Monoclonal antibodies are the newest entrants into the asthma treatment paradigm, such as immunoglobulin E (IgE) inhibitors, interleukin (IL)-5 inhibitors, and IL-4 and IL-13 inhibitors. None of the monoclonal antibodies are intended to be used first line, and are reserved for those patients whose asthma is not well controlled despite optimized controller medications.

Dupilumab is an IL-4 and IL-13 inhibitor, indicated as add-on maintenance treatment in patients aged 6 years and older with severe asthma with a type 2 or eosinophilic phenotype or with oral corticosteroid (OCS)-dependent asthma. Dupilumab was previously reviewed by CADTH for the indication of severe asthma with a type 2/eosinophilic phenotype or OCS-dependent asthma in patients aged 12 years and older and received a positive recommendation in June 2021.4 Dupilumab is administered by subcutaneous injection in pediatric patients aged 6 years to 11 years at the dose of 100 mg every 2 weeks or 300 mg every 4 weeks for patients with a body weight from 15 kg to less than 30 kg, 200 mg every 2 weeks or 300 mg every 4 weeks for patients with a body weight from 30 kg to less than 60 kg, and 200 mg every 2 weeks for patients with a body weight of 60 kg or more. Dupilumab also received a Health Canada indication for atopic dermatitis and for chronic rhinosinusitis with nasal polyposis.5 It was also previously reviewed by CADTH for the atopic dermatitis indication in patients older than 12 years of age and received a positive recommendation in April 2020.6

The sponsor has requested that dupilumab be reimbursed for patients aged 6 years to younger than 12 years with severe asthma with the type 2 or eosinophilic phenotype characterized by the following: symptoms that are not controlled despite optimal treatment, defined by the daily use of a medium or high-dose ICS plus 1 controller medication or high-dose ICS alone; eosinophils (EOS) of 150 cells/μL or greater, fractional exhaled nitric oxide (FeNO) of 20 parts per billion (ppb) or greater, or allergy-driven asthma; uncontrolled asthma having at least 1 severe exacerbation, defined by having experienced 1 or more hospitalizations or emergency care visits or treatment with a systemic corticosteroid (SCS) (oral or parenteral) in the past year; and a baseline assessment of asthma symptom control using a validated asthma control questionnaire (ACQ) must be completed before initiation of dupilumab treatment.

The objective of this report was to perform a systematic review of the beneficial and harmful effects of dupilumab for the add-on maintenance treatment of severe asthma with a type 2 or eosinophilic phenotype or OCS-dependent asthma in patients aged 6 years to younger than 12 years.

Stakeholder Perspectives

The information in this section is a summary of input provided by the patient groups who responded to CADTH’s call for patient input and from the clinical experts consulted by CADTH for the purpose of this review.

Patient Input

Input from patients was provided by Asthma Canada, based on a survey conducted between February and March 2022, clinical practice guidelines, the product monograph, non-for-profit organization websites, and research papers. More than 100 patients (92%) and caregivers (8%) across all provinces responded to survey with 4 patients having had experience with dupilumab. In addition, the Lung Health Foundation submitted patients’ input based on a survey conducted between January 2021 and June 2022 from 27 patients with asthma and 2 caregivers, all living in Ontario.

Even with currently available treatments, 1 in 4 respondents to the Asthma Canada survey indicated that they have poor symptom control. Approximately 60% of respondents worry about or have a fear of exacerbations, 47% of respondents are concerned about potential hospital admissions, and 47% of survey participants are concerned with missing school or work. The survey findings highlighted challenges for children with asthma, including difficulties in inhaler use techniques, difficulties with making and keeping friends due to fatigue and less energy, activity limitations, inability to attend and concentrate at school, and sleep disturbances. Patients, parents, and caregivers noted several barriers to accessing health care providers (e.g., respirologists, specialized asthma clinics) including travel time and cost, missed school or work, and the financial burden of prescription refills. The Lung Health Foundation input from patients indicated common symptoms of asthma, such as shortness of breath (74.2%), fatigue (67.7%), cough (51.6%), as well as difficulties in activities of daily living such as climbing stairs (43.4%), housework (40.0%), and physical activities (40.0%). Some of the negative impacts of asthma that were highlighted by the patients included: night or early morning waking due to breathing problems (34.5%), emotional well-being (37.9%), and being short-tempered or impatient with others (31.0%).

Patients and caregivers identified the following expectations for new treatment for children with severe asthma: increasing lung function, making management of symptoms easier, reducing exacerbations, and reducing reliance on OCSs. Moreover, children with asthma and their parents expect to see improved day-to-day activities affecting quality of life (school attendance, sleep, energy, participation in activities), less health care visits including those to the emergency department (ED), less anxiety and panic for potential exacerbations, less time off work, and decreased financial hardships. Respondents indicated they would like to minimize side effects of medication; however, they are willing to tolerate certain side effects to improve management of asthma. Decreasing frequency and easing the administration of medication was an additional priority reported by the participants. Finally, it was noted that children on dupilumab cannot be vaccinated with live vaccines, which can pose challenges for children who are not fully immunized.

Clinician Input

Input From Clinical Experts Consulted by CADTH

According to the clinical experts consulted by CADTH for this review, the needs of the majority of patients with asthma are met with current standard therapies; however, a subset of patients remains poorly controlled despite maximized pharmacological treatment and nonpharmacological interventions such as inhaler education and improved medication adherence.

In Canada, pediatric patients with uncontrolled moderate-to-severe type 2 inflammatory asthma have access to treatment with biologics such as anti-IgE, anti-IL5, or anti-IL4/IL13 monoclonal antibodies. Clinical experts reported that the patients who would most likely benefit from dupilumab treatment include individuals with moderate-to-severe asthma who have not achieved optimal asthma control despite conventional therapy (i.e., high-dose ICS with add-on therapy [long-acting beta agonist {LABA} and/or leukotriene receptor antagonist {LTRA}] and requiring ongoing or multiple courses of SCSs) and presenting with a clear inflammatory phenotype, as assessed by peripheral blood eosinophil levels.

According to the clinical experts consulted by CADTH, relevant outcomes to assess treatment response in children include improvements in pulmonary function testing (improvement or stabilization of forced expiratory volume in 1 second [FEV1], elimination of airflow reversibility to bronchodilator), decreases in acute asthma exacerbations, improvements in symptom control, and improvements in health-related quality of life (HRQoL). The clinical experts believed that the primary factor in deciding whether to discontinue dupilumab treatment would be a lack of improvement in asthma control outcomes over many months. Moreover, treatment with dupilumab should be discontinued in case of serious adverse events (SAEs) (e.g., serious immune or allergic reactions, serious dermatological reactions, malignancy, and ophthalmologic adverse events [AEs]). Initiation of the drug should be limited to pediatric respirologists or allergy specialists with significant pediatric asthma experience.

Clinician Group Input

Input was received from 6 clinicians, on behalf of the Canadian Thoracic Society. There were no contrary views reported between the clinician group and the clinical experts consulted for this review. The clinician group indicated that children with severe asthma have limited treatment options compared to the adult population. In addition, there is a lack of effective add-on therapies in younger children with severe asthma with nontype 2 inflammation involving neutrophilic inflammation and recurrent exacerbations caused by viral respiratory infections. According to the clinician group, key outcomes in asthma management include prevention of asthma exacerbations, maximization of quality of life, symptom prevention, and maximization of exercise tolerance. Members of the Canadian Thoracic Society agreed that the use of dupilumab should be restricted to patients aged 6 years to 11 years with type 2 inflammation, moderate-to-severe asthma not adequately controlled on medium-dose ICS plus LABA (or other second controller) or high-dose ICS (or OCS), and who experienced a severe exacerbation in the past year. The Canadian Thoracic Society clinicians suggested that dupilumab should be discontinued if a lack of clinically meaningful positive outcomes over an expected time frame is observed, as well as in case of safety concerns. Assessment of pediatric patients’ eligibility for biologic asthma therapy should be limited to asthma specialists (e.g., respirologists, allergists, pediatricians with a focus on childhood asthma), according to the clinician group input.

Drug Program Input

Input was obtained from the drug programs that participate in the CADTH reimbursement review process. The following were identified as key factors that could potentially impact the implementation of a CADTH recommendation for dupilumab.

  • Relevant comparators
  • Considerations for initiation of therapy
  • Considerations for continuation or renewal of therapy
  • Considerations for discontinuation of therapy
  • Considerations for prescribing of therapy
  • System and economic issues

Clinical Evidence

Pivotal Studies and Protocol Selected Studies

Description of Studies

The VOYAGE study is a multinational, multicentre, randomized, double-blind, placebo-controlled study that compared dupilumab to placebo in patients aged 6 years to younger than 12 years with asthma who were already receiving standard of care. Four hundred and 8 patients with persistent asthma were randomized in 2:1 ratio to 1 dose of dupilumab (100 mg or 200 mg) every 2 weeks or placebo every 2 weeks, over a treatment course of 52 weeks. The primary outcome was the annualized rate of severe exacerbations, while the key secondary outcome was pulmonary function measurement (i.e., change from baseline in prebronchodilator percent predicted FEV1 at week 12). There were 2 main efficacy populations assessed in the trial: type 2 inflammatory asthma phenotype population, characterized by a baseline blood eosinophil count of 150 cells/µL or greater or baseline FeNO of 20 ppb or greater, and baseline blood eosinophils of 300 cells/µL or greater.

The median age of patients included in the VOYAGE trial was 9 years (range = 6 to 11). Across both efficacy populations, the majority of patients were male (range = 64.4% to 69%), White (range = 86.3% to 89.5%), and had a body weight of greater than 30 kg (range = ||||||||||||||||||||). Greater than 60% of patients in the VOYAGE study had experienced 1 or 2 severe asthma exacerbations in the past year. At baseline, FEV1 reversibility was slightly higher in the dupilumab versus placebo group, with a mean (standard deviation [SD]) of 21.5% (21.37) versus 15.81% (16.40) in the type 2 asthma population, and with a mean (SD) of 22.9% (23.23) versus 16.2 (15.8) in the population with baseline eosinophils of 300 cells/µL or greater. Regarding ICS dosing, greater than 40% of patients were on high dosing (dupilumab versus placebo: 43.2% versus 43.9% in the type 2 asthma population and 42.3% versus 48.8% in the baseline blood eosinophils ≥ 300 cells/µL population) and greater than 50% of patients were receiving medium ICS dosing (dupilumab versus placebo: 55.5% versus 56.1% in the type 2 population and 56.0% versus 51.2% in the baseline blood eosinophils ≥ 300 cells/µL population).

Efficacy Results
Mortality

In the VOYAGE trial, there were no deaths reported across the dupilumab and placebo groups.

Acute Asthma Exacerbation

The adjusted annualized rates of severe asthma exacerbations over 52 weeks in the type 2 inflammatory asthma population were 0.305 (95% CI, 0.223 to 0.416) with dupilumab and 0.748 (95% CI, 0.542 to 1.034) with placebo, for a relative risk (RR) of 0.407 (95% CI, 0.274 to 0.605; P < 0.0001) and a risk difference of ||||||||||||||||||||||||||||||. In the population with a baseline eosinophil count of at least 300 cells/µL at baseline, the adjusted rates of exacerbations were 0.235 (95% CI, 0.16 to 0.345) in the dupilumab and 0.665 (95% CI, 0.467 to 0.949) in the placebo group (RR = 0.353; 95% CI, 0.222 to 0.562; P < 0.0001; risk difference = |||||||||||||||||||||||||||||||||||||||| (Table 2).

RRs for the dupilumab versus placebo comparison of severe exacerbation events associated with ED visits or hospitalizations were |||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||| for the populations with type 2 asthma and baseline blood eosinophils of 300 cells/µL or greater, respectively. Rates associated with hospitalizations only were not estimable due to |||||||||||||||||||||||||||||| events experienced by the patients during the trial.

Asthma Symptoms

Symptoms were assessed using the 7-item ACQ (ACQ-7). At week 24, ACQ-7 scores decreased (improved) in both the dupilumab and placebo groups. The least squares (LS) mean (standard error [SE]) was –1.33 (0.05) in the dupilumab group and –1.00 (0.07) in the placebo group, for a LS mean difference versus placebo of –0.33 (95% CI, –0.50 to –0.16; P = 0.0001) in the type 2 population. In the population with a baseline blood eosinophil count of 300 cells/µL or greater, the LS mean (SE) change from baseline to week 24 was –1.34 (0.06) with dupilumab and –0.88 (0.09) with placebo, for a difference between groups of –0.46 (95% CI, –0.66 to –0.26; P < 0.0001).

Results were maintained during the trial period (to week 52) across both efficacy populations.

Reduction in Use of OCSs

The proportions of patients experiencing treatment with SCS during the trial was higher in the placebo compared to dupilumab arm (dupilumab versus placebo: 24.2% versus 40.4% within the type 2 inflammatory asthma phenotype and 22.3% versus 41.7% within the baseline blood eosinophils ≥ 300 cells/µL population). Adjusted RRs in annualized SCS courses, for the comparison of dupilumab to placebo, were 0.407 (95% CI, 0.272 to 0.609) and 0.340 (95% CI%, 0.212 to 0.545), within the type 2 inflammatory asthma phenotype and population with baseline eosinophils of 300 cells/µL or greater, respectively.

Pulmonary Function

The percent predicted prebronchodilator FEV1 at week 12 increased in both the dupilumab and placebo groups in the type 2 inflammatory asthma phenotype population, with an LS mean difference between groups of 5.21% (95% CI, 2.14 to 8.27%; ||||||||||). Similarly, the LS mean difference at week 12 between the dupilumab and placebo group of 5.32% (95% CI, 1.76 to 8.88%, ||||||||||), in the population with a baseline blood eosinophil count of 300 cells/µL or greater, was reported. In both primary efficacy populations, LS mean changes in the percent predicted prebronchodilator FEV1 were sustained through week 52.

Reduction in Dose of ICSs

The VOYAGE study protocol allowed a permanent increase in background medications after 2 or more severe asthma exacerbations. During the treatment period of the trial, ||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||, across all efficacy populations assessed.

Health-Related Quality of Life–Paediatric Asthma Quality of Life Questionnaire

In type 2 inflammatory asthma phenotype population of the VOYAGE study, Standardised Paediatric Asthma Quality of Life Questionnaire PAQLQ(S) scores increased (improved) from baseline to week 52, with an LS mean difference between dupilumab and placebo groups of 0.34 (95% CI, 0.16 to 0.52, ||||||||||). In the population with at least 300 cells/µL baseline blood eosinophils values, similar differences at week 52 between groups were observed (LS mean = 0.33; 95% CI, 0.12 to 0.53; ||||||||||).

Reduction in Use of Rescue Medication

An overall decrease in number of puffs of reliever medications across the 24-hour period was observed in both treatment arms (LS mean differences between dupilumab and placebo groups at week 52 were |||||||||||||||||||||||||||||||||||||||| and |||||||||||||||||||||||||||||||||||||||| for the type 2 and baseline blood eosinophils ≥ 300 cells/µL populations, respectively).

Harms Results

In the VOYAGE trial, AEs occurred in 83% and 79.9% of patients in the dupilumab and placebo groups, respectively. The most common AEs in the dupilumab versus placebo groups were: nasopharyngitis (18.5% versus 21.6%), viral upper respiratory tract infection (12.2% versus 9.7%), pharyngitis (8.9% versus 10.4%), bronchitis (6.3% versus 10.4%), allergic rhinitis (5.9% versus 11.9%), injection site erythema (12.9% versus 9.7%), and injection site edema (10.3% versus 5.2%). SAEs were reported by 4.8% of patients receiving dupilumab and 4.5% of patients receiving placebo, the majority of which were asthma (dupilumab versus placebo: 1.5% versus 0%) and eosinophilia (dupilumab versus placebo: 0.7% versus 0%). Discontinuation due to an AE occurred in 1.8% versus 1.5% of patients of the VOYAGE study, in the dupilumab versus placebo groups, respectively.

Regarding notable harms, injection site reactions were the most commonly reported, by 17.7% and 13.4% of patients in the dupilumab and placebo groups, respectively. Hypersensitivity and anaphylactic reactions occurred in |||||||||||||||||||||||||||||| placebo patients and 0 dupilumab patients versus 1.5% placebo patients, respectively. In terms of infections, severe cases occurred in |||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||| of patients in the placebo group. Parasitic infections were reported only among patients in the dupilumab group (2.6%). Eosinophilia was more frequently occurring in dupilumab arm compared to placebo (6.6% versus 0.7%, respectively). More patients in the placebo group experienced conjunctivitis compared to dupilumab group (dupilumab versus placebo: 2.6% versus 6.7% for conjunctivitis [narrow] and 3.0% versus 7.5% for conjunctivitis [broad]).

Table 2. Summary of Key Results From Pivotal and Protocol Selected Study.

Table 2

Summary of Key Results From Pivotal and Protocol Selected Study.

Critical Appraisal

The VOYAGE trial is a multinational, multicentre, randomized, double-blind, placebo-controlled study. The study used a matching placebo-controlled design, and patients and investigators were blinded to the study treatment assignment, but not the dosing of the injections. Potential for unblinding might have also occurred because of higher frequencies of injection site reactions and eosinophilic reactions in the dupilumab arm compared to the placebo arm. Multiplicity adjustments were implemented adequately for the analysis of severe exacerbation events during the 52-week treatment period, change from baseline in prebronchodilator percent predicted FEV1 at week 12, and change in the ACQ-7–Interviewer Administered (ACQ-7-IA) at week 24. Baseline characteristics were largely balanced between the groups of the study, except for FEV1 reversibility, which was slightly higher in the dupilumab compared to the placebo group. Clinical experts consulted by CADTH regarded the selection of specific time points for outcome assessment and their inclusion in the hierarchy (i.e., FEV1 at week 12 and ACQ-7 at week 24) as not optimal, noting that 52-week assessments would have been more clinically relevant. Many important outcomes such as HRQoL, exposure to OCSs, and ICS dose adjustments were not controlled for multiple comparisons. Even though treatment withdrawals were higher in the dupilumab group compared to placebo, proportions of individuals discontinuing study treatment due to an AE were balanced across the 2 study arms. The number of study withdrawals was generally low (< 6%) and appropriate sensitivity analyses were implemented to handle missing data for the primary and key secondary outcomes, suggesting limited impact on the validity of observed findings.

Dupilumab is indicated as add-on maintenance treatment in patients aged 6 years and older with severe asthma with a type 2 or eosinophilic phenotype or with OCS-dependent asthma. The current review focuses on the patient population of 6 years to 11 years of age, as dupilumab was previously reviewed by CADTH and received a positive recommendation for patients aged 12 years and older. Patients who were OCS-dependent were not included in the VOYAGE trial. The type 2 population was 1 of the main efficacy populations in the trial, defined as having a baseline blood eosinophil count of 150 cells/µL or greater or baseline FeNO of 20 ppb or greater, but the clinicians noted that FeNO assessments are not routinely performed in Canadian clinical practice, which represents an implementation limitation. Most of the VOYAGE trial population was White; hence, generalizability of study findings to people living in Canada may be limited in this regard. Even though background medications administered in the trial were considered reflective of treatments used in Canadian practice by the clinician experts, it was not clear whether inhaler technique was checked throughout the trial. Despite this, adherence to background therapy was high across both treatment groups and placebo responses were robust for many outcomes, suggesting that patients may have benefited from the close attention and monitoring they received in a clinical trial setting per the clinical experts consulted by CADTH. The VOYAGE trial compared dupilumab to placebo (added on to standard of care), which represents a limitation as comparative effectiveness and safety of dupilumab to other biologics approved for management of asthma in pediatric population is limited to available indirect comparisons.

Indirect Comparisons

Description of Studies

The sponsor submitted 1 indirect treatment comparison (ITC). No published ITCs were identified after a systematic search of the literature performed by CADTH. The sponsor-submitted ITC aimed to compare dupilumab to other biologics for the treatment of pediatric patients aged 6 to younger than 12 years with uncontrolled, moderate-to-severe asthma with a type 2 inflammatory phenotype. After a systematic literature review and a feasibility assessment, a total of |||||||||| connected via placebo as a common comparator were identified as eligible. A series of pairwise Bucher ITCs were performed on various outcomes (severe exacerbations, deterioration of asthma [post hoc analysis], asthma symptoms, rescue medication use, and HRQoL), comparing dupilumab (100 mg to 200 mg every 2 weeks) with the IgE inhibitor omalizumab (75 mg to 375 mg once or twice a month). Subgroup data were generated from the dupilumab trial population to match the allergic phenotype and inclusion criteria of omalizumab trials.

Efficacy Results

||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||

Harms Results

|||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||

Critical Appraisal

Several limitations of the sponsor-submitted ITC were noted. There was considerable heterogeneity in study characteristics, patient populations and outcomes assessed across the studies included in the network. Since the population of interest for the ITC was the type 2 inflammatory population, an assumption was made that the efficacy of the IgE inhibitor omalizumab would be maintained in these patients. Even though there is clinical overlap between severe allergic and eosinophilic asthma according to the clinical experts, the amount of population concordance and its impact on indirect estimates could not be determined as omalizumab trials were not designed to include an eosinophilic asthma population. In addition, it is unclear whether the placebo link for the ITC was sufficiently similar for making comparisons, since data from the VOYAGE trial suggested a robust placebo response on several outcomes assessed in the trial. In reference to the subgroup analysis, matching specific groups of patients with dupilumab to the omalizumab studies lead to considerable reductions in sample size. A limited number of studies as well as limited available data restricted the possibility to perform meta-regression and account for differences across trials. There were no direct comparisons between treatments; therefore, the assessment of consistency was not feasible. In summary, due to various methodological limitations, no robust conclusions can be drawn about the comparative clinical efficacy of dupilumab versus omalizumab in the treatment of patients aged 6 years to 11 years with uncontrolled moderate-to-severe asthma.

Other Relevant Evidence

Description of Studies

The LIBERTY ASTHMA EXCURSION (EXCURSION) study was an open-label, noncomparative longer-term extension study that enrolled patients who completed the VOYAGE trial. The primary objective of EXCURSION was to assess long-term safety and tolerability of dupilumab. All patients received open-label treatment with dupilumab during the period of 52 weeks. A total of 365 patients were enrolled in EXCURSION, of which 240 patients had been assigned to dupilumab treatment in the parent trial (dupilumab–dupilumab group) and 125 had been assigned to placebo treatment in the parent trial (placebo–dupilumab group). All patients in EXCURSION were receiving their background medication (ICS with or without a second controller) as well as reliever therapy, if necessary. As per the database lock of January 17, 2022, the median duration of study was ||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||| for the dupilumab–dupilumab and placebo–dupilumab groups, respectively.

Efficacy Results
Severe Asthma Exacerbations

As of January 17, 2022, a total of ||||||||||||||||| of patients with type 2 inflammatory asthma phenotype in the dupilumab–dupilumab and placebo–dupilumab groups, respectively, experienced a severe exacerbation event. When looking at the patients with eosinophils of 300 cells/µL or greater at baseline of the parent study, ||||||||||||||||| experienced an event in dupilumab–dupilumab and placebo–dupilumab groups, respectively. The unadjusted annualized rate of severe exacerbation was 0.118 and 0.124 for the dupilumab–dupilumab and placebo–dupilumab groups, respectively, in the type 2 inflammatory asthma phenotype population. Similarly, in the subgroup with baseline eosinophils of 300 cells/µL or greater, the unadjusted annualized severe exacerbation event rate was 0.120 and 0.119, for the dupilumab–dupilumab and placebo–dupilumab groups, respectively.

Pulmonary Function

At week 52, mean (SD) changes from baseline in percent predicted prebronchodilator FEV1 were |||||||||||||||||||||||||||||||||| for the dupilumab–dupilumab and placebo–dupilumab groups, respectively, in the type 2 inflammatory population, and ||||||||||||||||||||||||||||||||||||||||||||||||||| for the 2 groups, respectively, in the population with eosinophils of 300 cells/µL or greater.

Harms Results

Among patients who entered the EXCURSION study from the VOYAGE study, 61.3% of patients in the dupilumab–dupilumab and 68.0% of patients in the placebo–dupilumab groups reported at least 1 AE as per data cut-off of January 17, 2022. SAEs were experienced by 2.5% of patients in the dupilumab–dupilumab group and 0.8% in the placebo–dupilumab group. There were no deaths reported during the study period. AEs leading to discontinuation of the treatment were reported by 3 (1.3%) patients in the dupilumab–dupilumab group (pulmonary tuberculosis, ascariasis, and allergic conjunctivitis), and no patients in the placebo–dupilumab group.

In terms of notable harms, hypersensitivity was experienced |||||||||||||||||% of patients in the dupilumab–dupilumab and placebo–dupilumab groups, respectively, with ||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||| experiencing anaphylactic reaction. Other notable harms of interest reported during the long-term extension study period in the dupilumab–dupilumab versus placebo–dupilumab included: injection site reactions ||||||||||||||||||||||||||||||||||||||||||||||||||| conjunctivitis (4.2% versus 4.8%), eosinophilia (3.3% versus 8.0%), severe or serious infections (|||||||||||||||||), and parasitic infections (1.7% versus 1.6%).

Critical Appraisal

The EXCURSION trial provided additional data on the longer-term safety and efficacy of dupilumab relative to placebo. The validity of observed findings is limited due to the open-label and noncomparative study design. Statistical hypothesis testing was not part of the design. Furthermore, as the EXCURSION trial is a 1-year study, rare AEs might not be captured as of the data cut-off date. Given that the patients enrolled in the long term extension study were originally from the VOYAGE parent study, and the eligibility criteria remained the same, it is reasonable to expect that the same limitations to generalizability are relevant to EXCURSION.

Conclusions

One sponsor-submitted, multicentre, randomized, double-blind, phase III trial (VOYAGE), comparing add-on therapy with dupilumab to placebo in patients aged 6 years to younger than 12 years with persistent asthma demonstrated that dupilumab reduced the annualized rate of severe exacerbations and improved pulmonary function (FEV1) in patients whose asthma remains uncontrolled despite background therapy with medium to high doses of ICSs. There was supportive evidence on the overall treatment benefit on asthma-related symptoms, as measured by ACQ-7, but the differences between the dupilumab and placebo groups did not exceed the minimal important difference (MID). HRQoL analyses were not controlled for multiple comparisons in this randomized controlled trial (RCT); thus, it remains unclear what the effect of add-on dupilumab on patients’ HRQoL is compared to placebo. Likewise, observed reductions in OCS usage were not part of the statistical hierarchy, which precluded drawing conclusions about the effects of dupilumab on this outcome. With respect to harms, there were no obvious safety or tolerability issues associated with the use of dupilumab in children. A longer-term extension study did not identify any new safety issues. Findings from the sponsor-submitted ITC were inconclusive with respect to the efficacy and safety of dupilumab compared to omalizumab due to numerous methodological limitations.

Introduction

Disease Background

Asthma is a chronic respiratory disorder characterized by reversible airway obstruction. Hallmarks of asthma include inflammation, bronchoconstriction, and airway remodelling, as well as hyperresponsive airways and mucous production.1 Symptoms of asthma include wheezing, dyspnea, chest tightness, sputum production, and coughing, and these symptoms can be exacerbated by exogenous influences such as allergens, upper respiratory tract infections, or environmental factors such as smoke or cold air.1 It is estimated that 2.4 million people living in Canada aged 12 years or older have asthma, or 12% of all children and 8% of adults.8 Moreover, prevalence data from the population of people living in Canada in 2011 and 2012 reported asthma estimates of 14% and 19% in children aged 5 years to 9 years and 10 years to 14 years, respectively.2

According to the clinical experts consulted for this CADTH review, asthma has several diverse phenotypes, 1 of which is primarily driven by type 2 inflammation, presenting with allergic or atopic profile and/or eosinophilic asthma. Eosinophils, among other functions, promote airway inflammation and contribute to airway hyperresponsiveness and remodelling,9,10 and eosinophilic asthma is characterized by increased peripheral blood eosinophil counts. Severe type 2 or eosinophilic asthma tends to be asthma that is poorly controlled despite optimized ICSs, plus add-on therapy with a LABA or LTRA.11

Standards of Therapy

Traditionally, the management of asthma is carried out using: medications for the acute relief of exacerbations (colloquially, “asthma attacks”), often referred to as “relievers” or “rescue medications”; and controllers, or maintenance drugs, which are used on a regular or chronic basis in an effort to prevent the onset of exacerbations.1

The Global Initiative for Asthma (GINA) guidelines support a stepwise approach for asthma treatment in children aged 6 years to younger than 12 years. Of note, detailed GINA recommendations for the management of children aged 6 years to younger than 12 years with severe asthma are currently under development and only the stepwise diagram for treatment is available in the available GINA guidance documents. In step 1, patients begin using a low-dose ICS whenever a reliever medication (short-acting beta agonist [SABA]) is used. As symptoms persist, step 2 involves a low-dose ICS with as-needed SABA, with an alternative treatment option of a LTRA. From here, patients may need to escalate to step 3, which includes a combination of low-dose ICS and LABA with as-needed reliever (SABA), or medium-dose ICS with as-needed reliever (SABA), or maintenance and reliever therapy with a very low dose of ICS plus formoterol. Step 4 includes stepping up to a medium-dose ICS and LABA combination or maintenance and reliever therapy of a low dose of ICS plus formoterol, with an alternative addition of tiotropium or LTRA and recommendation for expert advice in case the asthma is not well controlled. Finally, step 5 involves referral to a phenotypic assessment and the use of a higher-dose ICS and LABA combination, with additional possibility of add-on treatment (i.e., anti-IgE, anti-IL5/5R, anti-IL4R) in case asthma control is not achieved.1 Similarly, The Canadian Asthma Consensus Guidelines approach to therapy proposes SABA, or budesonide plus formoterol as needed for symptom control, and outlines low-dose, medium-dose, and high-dose ICSs for anti-inflammatory maintenance therapy, with add-on LTRA, LABA, and or tiotropium as necessary.3 In case the high-dose ICS with add-on therapy does not achieve control, treatment with OCSs and/or biologic therapy can be considered. According to the clinical experts consulted for this CADTH review, a very limited proportion of pediatric patients is on daily OCS therapy, but many patients may experience frequent or repeated short courses of OCSs. Monoclonal antibodies are the newest entrants into the asthma treatment paradigm for children aged 6 years to 12 years of age, such as an IgE inhibitor (omalizumab), IL-5 inhibitor (mepolizumab), and most recently IL-4 and IL-13 inhibitors (dupilumab). Nonpharmacologic therapies include asthma education, improvement of inhaler technique, allergen avoidance, and a written asthma action plan.1 The clinical experts consulted by CADTH stated that patients aged 6 years to younger than 12 years who are not well controlled on an ICS and another controller (e.g., GINA step 4) would receive a high-dose ICS plus a LABA (or alternative controller) when considering adding a biologic. The experts indicated that the goal would be to gain asthma control in a timely way and to lower the ICS dose when control is achieved. Aligned with previously mentioned guidelines, a Canadian Thoracic Society statement from 2017 provides guidance on the management of severe asthma.11

With respect to harms associated with pharmacologic therapies, chronic ICS use in children can have a number of concerning adverse effects, including growth retardation and adrenal suppression, particularly at high doses.12 The use of OCSs heightens the risk of harms such as fractures and decreased bone density, and their chronic use should be limited in children, according to the clinical experts.

According to the clinical experts consulted by CADTH, the goals of asthma therapy in children aged 6 years to 12 years are to achieve asthma symptom control, decrease future risk of worsening, prevent persistent airflow limitation, and decrease the frequency of exacerbations, which will improve HRQoL. Additionally, limiting complications and long-term adverse effects of current therapy is another important goal of asthma management in children.

Drug

Dupilumab is an IL-4 and IL-13 inhibitor.5 Both IL-4 and IL-13 are thought to play a role in inflammation and in the pathophysiology of asthma, and dupilumab is a monoclonal antibody that targets both. Dupilumab is administered by subcutaneous injection in pediatric patients aged 6 years to 11 years at the dose of 100 mg every 2 weeks or 300 mg every 4 weeks for patients with a body weight from 15 kg to less than 30 kg, 200 mg every 2 weeks or 300 mg every 4 weeks for patients with a body weight from 30 kg to less than 60 kg, and 200 mg every 2 weeks for patients with a body weight of 60 kg or more. Dupilumab is indicated as add-on maintenance treatment in patients aged 6 years and older with severe asthma with a type 2 or eosinophilic phenotype or with OCS-dependent asthma. Dupilumab is also indicated for atopic dermatitis in patients aged 6 years and older and for chronic rhinosinusitis with nasal polyposis in adults.5

Dupilumab was previously reviewed by CADTH for the indication of severe asthma with a type 2 or eosinophilic phenotype or OCS-dependent asthma in patients aged 12 years and older (June 2021; recommendation to reimburse with clinical criteria and/or conditions), and for the atopic dermatitis indication in patients older than 12 years (April 2020; recommendation to reimburse with clinical criteria and/or conditions).4,6

The sponsor has requested that dupilumab be reimbursed for patients aged 6 years to younger than 12 years with severe asthma with type 2 or eosinophilic phenotype characterized by the following: symptoms that are not controlled despite optimal treatment, defined by the daily use of a medium or high-dose ICS plus 1 controller medication or high-dose ICS alone; EOS of 150 cells/μL or greater or FeNO of 20 ppb or greater, or allergy-driven asthma; uncontrolled asthma having at least 1 severe exacerbation, defined by having experienced 1 or more hospitalization/emergency care visit or treatment with an SCS (oral or parenteral) in the past year; and a baseline assessment of asthma symptom control using a validated ACQ must be completed before initiation of dupilumab treatment. These criteria are in addition to the Health Canada indication.

Key characteristics of dupilumab and other biologics used for severe asthma are summarized in Table 3.

Table 3. Key Characteristics of Dupilumab, Mepolizumab, and Omalizumab.

Table 3

Key Characteristics of Dupilumab, Mepolizumab, and Omalizumab.

Stakeholder Perspectives

Patient Group Input

This section was prepared by CADTH staff based on the input provided by patient groups. The full original patient input(s) received by CADTH have been included in the stakeholder section at the end of this report.

Asthma Canada, the only national charity advocating for people with asthma and respiratory allergies, submitted patient group input based on a survey conducted between February and March, 2022, in addition to several other sources. More than 100 patients (92%) and caregivers (8%) across all provinces responded to the survey, including 4 patients having with experience with dupilumab. Another patient group, Lung Health Foundation, a registered charity that empowers people living with or caring for others with lung disease, submitted input based on a survey conducted between January 2021 and June 2022 from 27 patients with asthma and 2 caregivers, all living in Ontario.

One in 4 Asthma Canada survey participants indicated they have poor symptom control even with currently available treatments. Furthermore, many patients reported challenges in accessing the needed health providers, such as respirologists and specialized asthma clinics, to manage their health. When patients live in rural areas and must travel to get necessary care, children miss school and parents and caregivers miss work. The respondents said that managing care can require a significant amount of time and be a burden, which can be made worse with poor asthma control. Respondents emphasized that timely access to care becomes critical when patient must urgently travel to the ED for exacerbations that can be life-threatening and to restore airway function. Approximately 60% of respondents said that they worry about or have a fear of exacerbations; 47% respondents said they are concerned about potential hospital visits or admissions, which can be stressful; and 47% of survey participants said missed work and school days are concerning. In addition, some of the challenges that affect children with asthma identified by respondents were difficult techniques involved with inhaler use, other children not adequately understanding the impact of asthma on daily lives, making and keeping friends made difficult by fatigue and less energy, activity limitations that could be worsened by environmental triggers, inability to attend and concentrate at school, sleep disturbances, and significant time spent on educating friends, daycares, schools, and others about the seriousness of asthma. According to the patient group input, financial hardships associated with paying for asthma treatments are added stress. Lastly, the respondents to Asthma Canada survey said that drug shortages caused by limited supply have also impacted children living with asthma.

The Lung Health Foundation input highlighted that asthma symptoms such as shortness of breath (74.2%), fatigue (67.7%), and cough (51.6%), as well as difficulties in activities of daily living such as climbing stairs (43.4%), housework (40.0%), and physical activities (40.0%) are the challenging part of living with asthma. In addition, respondents to the Lung Health Foundation survey said that the negative impacts of asthma included waking up at night or in the early morning due to breathing problem (34.5%), emotional well-being (37.9%), and being short-tempered or inpatient with others (31.0%). They said they want treatments that improve symptom management, energy, and quality of life, as well as reduce exacerbations. In addition, the respondents indicated they wanted treatments with a reduced cost to the patient and caregiver.

Patients indicated that they want a new medication that can be another treatment option for children with severe asthma and their families to ease burden on patients, families, caregivers, and the health care system by increasing lung function (73%), making management of symptoms easier (61%), reducing exacerbations (56%) and reducing reliance on OCSs (56%). Also, children with asthma and their parents expect to see improved day-to-day activities affecting quality of life, such as attendance at school, sleep, energy, participation in activities, as well as less health care visits including those to the ED, less anxiety and panic (for potential exacerbations), less time off work, and less financial hardships. Respondents said they would like to minimize side effects of medication; however, they are willing to tolerate certain side effects to improve management of their asthma. One in 4 survey participants indicated that they need too many daily doses, therefore, a medication that can be taken less frequently and administered easily would be helpful for children with severe asthma.

Lastly, it was noted that children receiving dupilumab cannot be vaccinated with live vaccines (e.g., hepatitis, meningococcal, and HPV vaccines), which can be challenging for children who may not be fully immunized.

Clinician Input

Input From Clinical Experts Consulted by CADTH

All CADTH review teams include at least 1 clinical specialist with expertise regarding the diagnosis and management of the condition for which the drug is indicated. Clinical experts are a critical part of the review team and are involved in all phases of the review process (e.g., providing guidance on the development of the review protocol, assisting in the critical appraisal of clinical evidence, interpreting the clinical relevance of the results, and providing guidance on the potential place in therapy). The following input was provided by 2 clinical specialists with expertise in the diagnosis and management of asthma.

Unmet Needs

According to the clinical experts consulted by CADTH, the main goals of asthma therapy in children include achieving asthma symptom control, decreasing the risk and frequency of acute asthma exacerbations, and preventing sequelae of long-term uncontrolled asthma (i.e., fixed or only partially reversible airflow limitation). Additionally, limiting complications and long-term adverse effects of inhaled and OCSs among children was identified as important goal by the clinicians.

There are no treatments available that would cure asthma, but long-term control can be achieved for many patients, according to the experts. The clinical experts reported that the majority of patients achieve excellent control with existing medications and treatments, aligned with the Canadian Asthma Consensus Guidelines.3 The experts noted that increasing medication adherence, adopting self-management techniques and inhaler education are also important, yet challenging to adopt, factors in reaching asthma treatment goals. The clinical experts also stressed that a subset of patients will remain poorly controlled despite adoption of the best practices for asthma treatment.

Place in Therapy

The clinical experts reported that dupilumab would be utilized in treating children and youth with uncontrolled moderate-severe type 2 inflammatory asthma (i.e., individuals who remain uncontrolled on high-dose ICSs with add-on therapy [LABA and/or LTRA] and requiring ongoing or multiple courses of SCSs). Hence, they indicated that dupilumab would not be implemented as a first-line therapy, but would be a therapy used when conventional treatment has not achieved asthma control.

According to the clinical experts, biologic agents targeting IgE-mediated disease (e.g., the anti-IgE drug omalizumab) and decreasing eosinophilic inflammation (e.g., the anti-IL5 drug mepolizumab) have found limited use among pediatric patients in the Canadian setting so far. The clinicians noted that dupilumab therapy addresses a specific aspect of the inflammatory cascade (the IL-4 and IL-13 pathway) and highlighted its novelty in this regard.

Patient Population

The clinical experts reported that the target population for dupilumab would include individuals with moderate-to-severe asthma who have not achieved optimal asthma control despite conventional therapy and clearly present with an inflammatory phenotype. The type 2 inflammatory profile can be assessed by peripheral blood eosinophil levels in the Canadian context to determine initiation of therapy. The experts highlighted that the VOYAGE trial enrolled patients with type 2 inflammatory asthma (i.e., peripheral blood EOS > 150 cells/μL or FeNO > 20 ppb), and that the eosinophilic criteria might have implementation difficulties in certain laboratory facilities across Canada, which routinely measure rounded values of EOS (i.e., 100 cells/μL, 200 cells/μL, 300 cells/μL, and so forth). Furthermore, clinicians noted that FeNO assessments are not routinely performed in most Canadian centres.

Both clinical experts stated that misdiagnosis of asthma is common. Clinicians felt that 6 years of age represents a limit for reliable and reproducible pulmonary function measurement. Therefore, children older than 6 years can undergo pulmonary function and reversibility testing, which is required for confirming the diagnosis of asthma. According to the pediatric clinical experts, under-diagnosis of patients with moderate-to-severe asthma can sometimes be observed in the clinical practice.

Assessing Response to Treatment

The clinical experts reported that outcomes used clinically to assess response to treatment are typically targeting assessment of asthma control. Specifically, clinicians reported the following outcomes to be of relevance: improvements in pulmonary function testing, decreases in acute asthma exacerbations, improvements in symptom control, and improvements in quality of life scores. A validated measure such as the ACQ can be used to objectively assess improved control; however, 1 clinician reported that the instrument is mostly used for research purposes and is not used on a regular basis within the everyday clinical care. In addition, the experts reported that decreased health care resource utilization, improvement in ability to perform activities of daily living, and ability to participate in recreational activities can be considered supportive of a positive treatment effect of dupilumab. Regarding the frequency of disease assessments, the clinical experts reported that patients with moderate-to-severe asthma would be assessed in specialty asthma clinics approximately 2 to 4 times per year, depending on the asthma severity and clinic resources.

Discontinuing Treatment

According to the clinical experts, evidence of lack of impact on pulmonary function testing, asthma symptom control, acute exacerbations, and quality of life assessment need to be accounted for when considering discontinuation from therapy. Moreover, treatment with dupilumab should be discontinued the in case of SAEs (e.g., serious immune/allergic reactions, serious dermatological reactions, malignancy, and ophthalmologic AEs).

Prescribing Conditions

Clinical experts indicated that treatment with dupilumab should be initiated by a pediatric respirologist or allergy specialist with significant pediatric experience. For patients living in rural or remote areas, physician care could be provided in liaison with 1 of the previously mentioned specialists, while optimal access to care can be achieved though the implementation of telehealth practices.

Additional Considerations

One clinician reported that concomitant treatment of atopic dermatitis as an outcome should be considered, as atopic dermatitis may have significant adverse effects upon quality of life of patients.

Clinician Group Input

This section was prepared by CADTH staff based on the input provided by clinician groups. The full original clinician group input(s) received by CADTH have been included in the stakeholder section at the end of this report.

A total of 6 clinicians from the Canadian Thoracic Society (2 Canadian Thoracic Society Asthma Assembly Co-chairs and 4 pediatric respirologists from the Asthma Assembly Steering Committee) submitted clinician group input. The Canadian Thoracic Society is a national professional association for health care providers in respiratory care and research. The Canadian Thoracic Society promotes lung health and enhances the ability of health care providers through leadership, collaboration, research, learning, and advocacy, as well as providing the best respiratory practices in Canada. The Canadian Thoracic Society is also an accrediting body for specialist education and continuing professional development.

Unmet Needs

The clinician group stated that key goals of asthma control are to prevent asthma exacerbations, which can be life-threatening, maximize quality of life, prevent symptoms, and maximize exercise tolerance.

Secondary goals in asthma management include normalizing lung function, reducing airway inflammation, avoiding permanent airway remodelling, avoiding chronic use of OCSs, and using the lowest effective dose of ICSs to avoid growth suppression, adrenal suppression, and other significant side effects of corticosteroids in children.

The clinician group reported that in a relatively small subgroup of children with severe asthma, acceptable control requires high doses of ICSs and/or OCSs and add-on therapies, which puts them at a high risk of significant AEs. In some children, acceptable control cannot be achieved with these therapies and these patients experience frequent exacerbations that require OCSs. The clinician group indicated that chronic OCSs are not a viable treatment option given their side effect profile and highlighted that the recurrent use of OCSs is associated with a high risk of side effects.

According to the clinicians, children with severe asthma have limited treatment options compared to the adult population. Currently, the American Thoracic Society/European Respiratory Society,15 but not the Canadian Thoracic Society3 recommend tiotropium. However, tiotropium is not approved for those younger than 18 years in Canada and is not regularly used off-label in children aged 6 years to 11 years. The Canadian Thoracic Society and the American Thoracic Society/European Respiratory Society both recommend omalizumab for children aged 6 years to 11 years who meet the initiation criteria. However, the guidelines only include clinical trials published up to 2018. Lastly, mepolizumab is approved in Canada for children aged 6 years to 11 years with severe asthma but has not been widely used.

The clinician group identified other unmet needs. In younger children with severe asthma who have nontype 2 inflammation involving neutrophilic inflammation and recurrent severe exacerbations caused by viral acute respiratory tract infections, effective add-on therapies have not been identified when ICSs fail. Similarly, in a subgroup of older children with severe asthma who have neutrophil-triggered inflammation, effective add-on therapies are also needed.

Place in Therapy

The clinician group stated that dupilumab (an anti-IL4/IL-13 drug) targets different inflammatory pathways than omalizumab (an anti-IgE drug) and mepolizumab (an anti-IL5 drug); therefore, complements other available treatments. Following the Canadian Thoracic Society3 Severe Asthma Management Continuum, dupilumab would be considered as an add-on therapy in severe asthma when other biologics are indicated. Also, the group reported dupilumab could be helpful when asthma is not well controlled on a combination of a high-dose ICS, LABA, and LTRA, or when a child experiences significant side effects from these medications, or when these medications are contraindicated. The clinician group emphasized that since injectable medications can be less acceptable due to a higher burden on families and higher costs for the health care system, it is recommended to use biologics based on the child’s inflammatory profile and only after standard therapy has been adequately tried with good adherence.

Moreover, the clinician group suggested that when children with severe asthma concomitantly have atopic dermatitis, when children are allergic to or have had SAEs with mepolizumab, or when there is supply issue with mepolizumab, dupilumab can be a therapeutic option.

Patient Population

Based on input, patients aged 6 years to 11 years with type 2 inflammation, moderate-to-severe asthma not adequately controlled on a medium-dose ICS plus LABA (or other second controller) or high-dose ICS (or OCS) and who experienced a severe exacerbation in the past year are most likely to respond to dupilumab treatment. (Type 2 inflammation is defined as having a serum eosinophil count ≥ 150 cells/μL and FeNO ≥ 20 ppb or a serum eosinophil count ≥ 300 cells/μL.) The group added that those with an eosinophil count of greater than 300 cells/μL and a baseline FeNO of 25 ppb have shown to have the best response to dupilumab. However, the group said that there is still a good response with a lower-level eosinophil count as long as it is greater than 150 cells/μL.

According to the clinician group, the 2 markers of type 2 inflammation, namely an eosinophil count of greater than 150 cells/μL and FeNO elevation, are the only disease characteristics that would make differences in responses.

The clinician group indicated that the best-suited physicians to identify eligible patients are respirologists or allergists, who must confirm diagnosis based on symptoms and airway reversibility measured by spirometry (FEV1 change > 10%). If not under the care of a respirologist or allergist, the clinician group indicated that patients must be referred and assessed with spirometry, clinical exam, differential complete blood count (serum EOS), and FeNO, if available.

Assessing Response to Treatment

The clinician group said that clinically meaningful outcomes include reduced frequency and severity of symptoms, improved quality of life, and minimized side effects from existing maximal therapy (e.g., corticosteroids). Reducing daily medication burden and improving compliance are also noted as potential outcomes to measure response. Other outcomes, such as reduced inflammatory markers, improved lung function, and prevention of long-term airway remodelling are additional outcomes to determine response to treatment. The clinical measures, such as ACQs, FEV1, and exacerbation rates are routinely used in clinical practice and do not differ among specialists. However, according to the clinician group, the inflammatory markers may not be routinely used or accessible in clinical practice and may vary in their application.

Discontinuing Treatment

The clinician group said that a lack of clinically meaningful positive outcomes over an expected time frame, such as similar (or worse) rate of exacerbation, prebronchodilator FEV1, or patient symptom scores compared to pretreatment levels, is a reason to consider discontinuing treatment. Additionally, the group mentioned that safety concerns and patient’s choice could also play roles in making decision to discontinue treatment.

Prescribing Conditions

The clinician group cited the Canadian Thoracic Society Position Statement for the Recognition and Management of Severe Asthma11 for prescribing conditions. They said that “asthma specialists,” including but not limited to respirologists, allergists, and pediatricians with a focus on childhood asthma or who are specialists in asthma, general respirology, or allergy/immunology, and who have access to lung function tests and certified asthma or respiratory educators or nurse practitioners should assess a child for eligibility for biologic asthma therapy. According to the clinician group, an asthma specialist should diagnose, treat, and monitor for adherence and correct administration techniques.

The clinician group suggested the first 1 or more doses of dupilumab be administered by a health care practitioner in a hospital or medical setting. Following this, injection by a caregiver who has received proper training on correct administration can be undertaken in the community setting, such as in the home, if the health care practitioner determines it appropriate.

Drug Program Input

The drug programs provide input on each drug being reviewed through CADTH’s reimbursement review processes by identifying issues that may impact their ability to implement a recommendation. The implementation questions and corresponding responses from the clinical experts consulted by CADTH are summarized in Table 4.

Table 4. Summary of Drug Plan Input and Clinical Expert Response.

Table 4

Summary of Drug Plan Input and Clinical Expert Response.

Clinical Evidence

The clinical evidence included in the review of dupilumab is presented in 3 sections. The first section, the systematic review, includes pivotal studies provided in the sponsor’s submission to CADTH and Health Canada, as well as those studies that were selected according to an a priori protocol. The second section includes indirect evidence from the sponsor and indirect evidence selected from the literature that met the selection criteria specified in the review. The third section includes sponsor-submitted long-term extension studies and additional relevant studies that were considered to address important gaps in the evidence included in the systematic review.

Systematic Review (Pivotal and Protocol Selected Studies)

Objectives

To perform a systematic review of the beneficial and harmful effects of dupilumab (200 mg/1.14 mL [prefilled syringe)] and 300 mg/2 mL [prefilled syringe]) for the add-on maintenance treatment of severe asthma with a type 2 or eosinophilic phenotype or OCS-dependent asthma in patients aged 6 years to younger than 12 years.

Methods

Studies selected for inclusion in the systematic review included pivotal studies provided in the sponsor’s submission to CADTH and Health Canada, as well as those meeting the selection criteria presented in Table 5. Outcomes included in the CADTH review protocol reflect outcomes considered to be important to patients, clinicians, and drug plans.

Table 5. Inclusion Criteria for the Systematic Review.

Table 5

Inclusion Criteria for the Systematic Review.

The literature search for clinical studies was performed by an information specialist using a peer-reviewed search strategy according to the PRESS Peer Review of Electronic Search Strategies checklist.16

Published literature was identified by searching the following bibliographic databases: MEDLINE All (1946–) via Ovid and Embase (1974–) via Ovid. All Ovid searches were run simultaneously as a multifile search. Duplicates were removed using Ovid deduplication for multifile searches, followed by manual deduplication in Endnote. The search strategy comprised both controlled vocabulary, such as the National Library of Medicine’s MeSH (Medical Subject Headings), and keywords. The main search concepts were Dupixent (dupilumab) and asthma. Clinical trials registries were searched: the US National Institutes of Health’s clinicaltrials.gov, WHO’s International Clinical Trials Registry Platform search portal, Health Canada’s Clinical Trials Database, and the European Union Clinical Trials Register.

CADTH-developed search filters were applied to limit retrieval to RCTs or controlled clinical trials. Retrieval was not limited by publication date or by language. Conference abstracts were excluded from the search results. Refer to Appendix 1 for the detailed search strategies.

The initial search was completed on July 27, 2022. Regular alerts updated the search until the meeting of the CADTH Canadian Drug Expert Committee on November 23, 2022.

Grey literature (literature that is not commercially published) was identified by searching relevant websites from the Grey Matters: A Practical Tool For Searching Health-Related Grey Literature checklist.17 Included in this search were the websites of regulatory agencies (US FDA and European Medicines Agency). Google was used to search for additional internet-based materials. Refer to Appendix 1 for more information on the grey literature search strategy.

Two CADTH clinical reviewers independently selected studies for inclusion in the review based on titles and abstracts, according to the predetermined protocol. Full-text articles of all citations considered potentially relevant by at least 1 reviewer were acquired. Reviewers independently made the final selection of studies to be included in the review, and differences were resolved through discussion.

Findings From the Literature

One study was identified from the literature for inclusion in the systematic review (Figure 1). The included study is summarized in Table 6. A list of excluded studies is presented in Appendix 2.

Of the 320 citations identified, 4 were potentially relevant. There were no additional reports from the grey literature deemed relevant. In total 1 report is included in the systematic review section.

Figure 1

Flow Diagram for Inclusion and Exclusion of Studies.

Table 6. Details of Included Studies.

Table 6

Details of Included Studies.

Description of Studies

The VOYAGE RCT was designed with a primary objective to assess the efficacy of add-on dupilumab compared to placebo in children aged 6 year to younger than 12 years with uncontrolled persistent asthma who were already receiving standard of care. Secondary goals of the VOYAGE trial were to investigate safety and tolerability of dupilumab, evaluate patient-reported outcomes and HRQoL, and assess systemic exposure and association between treatment and pediatric immune responses to vaccines.

There were 2 primary efficacy populations in the trial: the type 2 inflammatory population, defined as either a baseline blood eosinophil count of 150 cells/µL or greater or baseline FeNO of 20 ppb or greater; and a population with a baseline blood eosinophil count of 300 cells/µL or greater.

The study randomized patients in a 2:1 ratio to receive a subcutaneous injection of dupilumab or placebo. Patients were randomized from 90 centres in 17 countries, including Canada. Patients and investigators were blinded to the study treatment assignment, but not to the dose of the injections. Dupilumab and placebo were provided in identically matched prefilled syringes, labelled with a treatment kit number generated by the sponsor.

The randomized treatment kit number list was generated centrally using interactive voice response system or interactive web response system. Randomization was stratified by ICS dose level (medium or high) at screening, blood eosinophil count (< 300 cells/µL and ≥ 300 cells/µL) at screening, and region (Latin America, Eastern Europe, and Western countries).

The study consisted of 3 periods (Figure 2):

  • Screening (4 ± 1 weeks) to collect baseline data on asthma control and assure eligibility criteria
  • Randomized double-blind treatment (up to 52 weeks)
  • Posttreatment period (12 weeks)

Eligible patients who completed the randomized treatment period of the study were offered the opportunity to participate in the 1-year long-term extension study, EXCURISON.20 Patients who enrolled in the extension study were excluded from the posttreatment period of the trial.

The first patient was randomized on April 21, 2017, and the last patient completed the study on August 26, 2020. According to the VOYAGE trial protocol, the database lock was planned based on the time when all randomized patients completed the week 52 visit or discontinued from the study before week 52.

The figure is a flow chart that describes the various stages of the VOYAGE study from screening (4 weeks duration) to randomized treatment phase (52 weeks duration) to the posttreatment follow-up (12 weeks duration).

Figure 2

Study Design of the VOYAGE Trial.

Populations

Inclusion and Exclusion Criteria

VOYAGE included patients with asthma aged 6 years to younger than 12 years who were on existing background therapy of medium-dose ICSs in combination with a second controller (i.e., LABA, LTRA, long-acting muscarinic antagonist [LAMA], or methylxanthines) or high-dose ICSs alone or in combination with a second controller for at least 3 months and on a stable dose of at least 1 month before the study. Patients had to have a prebronchodilator FEV1 of 95% or less of predicted normal or postbronchodilator reversibility of at least 10% or greater, showed evidence of uncontrolled asthma during the screening period and had either been treated with an SCS or been hospitalized or visited an ED for worsening asthma within the past year.

Patients were excluded if their body weight was less than 16 kg, if they had other chronic lung diseases or history of life-threatening asthma (e.g., requiring intubation), and if they had a history of any malignancies or current comorbidities that may interfere with the drug under study. Moreover, nonadequate adherence with background therapy during the screening period (< 80% of total number of prescribed doses) was also an exclusion criterion in the trial.

Baseline Characteristics

In the VOYAGE trial, median age of patients was 9 years (range = 6 to 11) across type 2 inflammatory asthma phenotype and population with baseline blood EOS of 300 cells/µL or greater. Most patients were male (range = 64.4% to 69% across the study groups in the 2 efficacy populations), White (range = 86.3% to 89.5% across the study groups in the 2 efficacy populations) and weighed more than 30 kg (range = 66.7% to 68.4% across the study groups in the 2 efficacy populations). Within the type 2 inflammatory asthma phenotype population, a greater proportion of individuals had blood eosinophilic counts of 300 cells/µL or greater (74.2% in the dupilumab and 73.7% in the placebo group). Although slight imbalances were observed across the groups in terms of number severe exacerbations (1, 2, 3, or ≥ 4) in the year before the study enrolment, the average number of exacerbations (mean [SD]) reported were 2.61 (2.58) in the dupilumab and 2.18 (1.55) in the placebo groups for the type 2 asthma population, and 2.78 (2.90) in the dupilumab and 2.37 (1.71) in the placebo groups for the population with baseline blood EOS of 300 cells/µL or greater. FEV1 reversibility was larger in the dupilumab than placebo group, with a mean (SD) of 21.5% (21.37) versus 15.81 (16.4) in the type 2 asthma population, and with a mean (SD) of 22.9% (23.23) versus 16.2 (15.8) in the population with baseline blood EOS of 300 cells/µL or greater. Regarding the ICS dosage, approximately 43% of patients were on a high-dose ICS and 55% were on a medium-dose ICS within the type 2 asthma population. In the population with baseline blood EOS of 300 cells/µL or greater, approximately 45% of patients were using high doses of an ICS (dupilumab versus placebo: 42.3% versus 48.8%), and approximately 53% of patients were using medium doses of an ICS (dupilumab versus placebo: 56.0% versus 51.2%). Three patients in the dupilumab group were on a low-dose ICS, which was a protocol violation (Table 7).

Baseline characteristics of other efficacy populations (intention-to-treat [ITT] and baseline EOS ≥ 150 cells/µL populations) are presented in the Appendix 3.

Table 7. Summary of Baseline Characteristics.

Table 7

Summary of Baseline Characteristics.

Interventions

In the VOYAGE trial, the dupilumab 100 mg dose was administered subcutaneously in a 0.67 mL syringe (for patients with a body weight ≤ 30 kg) and the 200 mg dose was administered in a 1.14 mL syringe (for patients with a body weight > 30 kg), once every 2 weeks. To maintain blinding, the 100 mg dose was matched to a 0.67 mL placebo and the 200 mg dose was matched to a 1.14 mL placebo delivered once every 2 weeks.

After week 12, parents, caregivers, and legal guardians of patients were allowed to perform home administration of dupilumab if they elected to do so and if they followed a training demonstration and completed administration under close supervision by the investigator or delegate at not less than 3 visits.

Temporary treatment discontinuation could be considered by the investigator because of AEs, infections or infestations that do not respond to medical treatment, and laboratory abnormalities. Re-initiation of treatment was conducted after close monitoring, once clinical consideration that the AEs occurrence was not related to the treatment under study was complete and if the eligibility criteria for the study were still met.

Permanent discontinuation from the treatment and the study may have occurred on patients’ or patients’ legal representative request or based upon clinical investigators’ decision. Moreover, patients’ study withdrawal occurred for the following additional reasons: specific request from the sponsor, in case of protocol deviation, pregnancy, occurrence of anaphylactic reactions or systemic allergic reactions related to the treatment under study, malignancy diagnosis, opportunistic infections, levels of serum alanine transaminase greater than 3 times the upper limit of normal, and total bilirubin greater than 2 times the upper limit of normal.

After permanent treatment discontinuation, patients were encouraged to complete the remaining study visits according to the visit schedule until the end of study or up to recovery or stabilization of any AEs.

Background medications, including ICSs and second controller and reliever medications were administered during the trial and their usage was recorded. Patients supplied their controller medication.

Prior to study entry, patients needed to be on a stable dose of a medium-dose ICS with a second controller medication, high-dose ICS alone, or high-dose ICS for at least 3 months with at least 1 month of stable dose before the first visit. Dosing levels considered as medium- or high-dose ICS in children aged 6 years to younger than 12 years were aligned with the GINA guidelines, 2015 version.18 Only 1 second controller medication (LABA, LTRA, LAMA, or methylxanthines) for combined use with medium- or high-dose ICS was permitted. Use of a reliever medication other than albuterol/salbutamol or levalbuterol/levosalbutamol was discouraged.

During the treatment period, patients were on their baseline dose regimen of the controller medication(s) used during screening. Patients who experienced a deterioration of asthma during the trial were allowed to have an up to 4-fold increase in their ICS dose temporarily, for a maximum of 10 days (recorded as a loss of asthma control event). At that point, treatment could have been changed to SCSs (severe exacerbation event) or reverted to the original ICS dose depending on the progression of symptoms. Permanent adjustments to the controller medication dosing (i.e., step up in medium- to high-dose ICS or addition of second controller for patients on high-dose ICS monotherapy) were allowed only if the patient experienced 2 or more severe exacerbations events at any time during the trial. SCSs were allowed at any time in case of clinical symptoms of severe asthma exacerbation event, per the judgment of the study investigator.

During the posttreatment period, patients not continuing with the long-term open-label extension study, were treated with the controller medication regimen and dose used during the randomized treatment period. Adjustments were possible based on patients’ status and clinical judgment of the investigator.

Outcomes

A list of efficacy end points identified in the CADTH review protocol that were assessed in the clinical trials included in this review is provided in Table 8. These end points are further summarized in the following. A detailed discussion and critical appraisal of the outcome measures is provided in Appendix 4.

Table 8. Summary of Outcomes of Interest Identified in the CADTH Review Protocol.

Table 8

Summary of Outcomes of Interest Identified in the CADTH Review Protocol.

Severe Exacerbation Events

The annualized rate of severe asthma exacerbations during the 52-week period was the primary outcome of the VOYAGE trial. Severe exacerbations were defined as a deterioration in asthma that required the use of SCSs for at least 3 days or resulted in hospitalizations or ED visits requiring SCSs. Two events were considered as different if the interval between their start dates was at least 28 days. The reasons behind any exacerbation event (e.g., infections including viral and bacterial, allergen exposure, exercise, and others) were recorded in the electronic case report form. Study investigators were responsible for maintaining accurate and timely electronic case report forms, which were initially designed by the sponsor.

Patients who permanently discontinued the study medication were asked and encouraged to continue with all the remaining study visits and their additional off-treatment severe exacerbation events up to week 52 were recorded. Patients who discontinued treatment were not eligible for the 1-year long-term extension study.

Pulmonary Function

Spirometry was performed following American Thoracic Society/European Respiratory Society 2005 guidelines.21 The key secondary outcome was change from baseline in prebronchodilator percent predicted FEV1 at week 12, while the measurements at other time points (weeks 2, 4, 8, 24, 36, and 52) were considered secondary end points. Spirometry was to be performed preferably during morning hours and at the same time every day, using the same spirometer and standardized techniques.

Measurements of morning and evening peak expiratory flow (PEF) were recorded through daily assessments and collection using an electronic diary and PEF metre by caregiver(s) of the patients. Both morning and evening PEF measurements were performed before intake of any reliever medications. Three values were collected and the highest 1 was used for evaluation.

Electronic diaries were also used to record the number of inhalations of relievers used for symptom relief in a day, number of inhalations of background medication, and record OCS use for an exacerbation event.

ACQ-7-IA and 5-item ACQ–Interviewer Administered

The ACQ is a questionnaire designed to assess the degree of asthma control with questions on symptoms (completed by the patient and/or their caregiver), including activity limitation, nocturnal waking, shortness of breath, wheezing, and symptoms on waking. Moreover, the average number of daily doses of rescue inhaler used, as well as spirometry measured as FEV1 (completed by clinic staff, including prebronchodilator use, percent and percent predicted use). The ACQ-7 has 7 items with each measured on a 7-point scale scored from 0 (totally controlled) to 6 (severely uncontrolled). Clinical staff score the percent predicted FEV1 on a 7-point scale based on the spirometry result.22-24 The ACQ-7 score is computed as the unweighted mean of the responses to the 7 questions. Changes of at least 0.5 are considered to be clinically meaningful.24,25 ACQ assessments were conducted every 2 weeks up to the week 12, and then every 4 weeks until end of study visit of the VOYAGE trial.

The 5-item ACQ–Interviewer Administered (ACQ-5-IA) scores were deduced from the responses to the first 5 questions of ACQ-7-IA and were applied to patients aged 6 years to younger than 12 years in the VOYAGE trial.

PAQLQ(S)–Interviewer Administered

The PAQLQ(S) disease-specific instrument comprises 23 items across 3 domains: symptoms (10 items), activity limitation (5 items), and emotional function (8 items), which are graded on a 7-point Likert scale (1 = maximum impairment, 7 = no impairment).A standardized format of the instrument was used in the trial, which included generic activity questions (physical activity, activities with animals, and activities with friends and family). It was applied to patients who were aged 7 years and older in the VOYAGE trial. A global score ranges from 1 to 7, with higher score representing better HRQoL.26-28 The MID of approximately 0.5 was identified for the overall quality of life score.27 Assessments during VOYAGE trial were conducted every 12 weeks until end of study visit.

Paediatric Asthma Caregiver's Quality of Life Questionnaire

The PACQLQ is a self-administered 13-item questionnaire capturing the impact of child’s asthma on quality of life of the parent(s), caregiver(s), and legal guardian(s). The items are organized in 2 domains (4 items concern activity limitations and 9 concern emotional function). Responses to the individual items are given on a 7-point Likert scale where 1 (all of the time/very very worried or concerned) represents severe impairment and 7 (none of the time/not worried or concerned) represents no impairment. In the VOYAGE trial, the instrument was administered among parent(s), caregiver(s), and legal guardian(s) of children aged 7 years to younger than 12 years of age. Both domain and overall scores range from 1 to 7, with higher score representing better HRQoL.27,29 The estimated MID identified through the CADTH literature search is 0.5 for the overall score.27 Assessments during VOYAGE trial were conducted every 12 weeks until end of study visit.

Pediatric Rhinoconjunctivitis Quality of Life Questionnaire–Interviewer Administered in Patients With Comorbid Allergic Rhinitis

Pediatric Rhinoconjunctivitis Quality of Life Questionnaire (PRQLQ)–Interviewer Administered is a disease-specific instrument developed to measure HRQoL in children aged 6 years to younger than 12 years diagnosed with seasonal allergic rhinoconjunctivitis or hay fever. Twenty-three items of the instrument are organized into 5 domains (nose symptoms, eye symptoms, practical problems, activity limitation, and other symptoms), which are assessed on a 7-point Likert scale (0 = no impairment, 6 = maximum impairment). Both domain and overall scores range from 0 to 6, with higher score representing worse HRQoL.30,31 The CADTH literature search could not identify studies reporting MID values for this instrument. Assessments during VOYAGE trial were conducted every 12 weeks until end of study visit.

EQ-5D-Youth

The EQ-5D-Youth (EQ-5D-Y) is a generic preference-based HRQoL measure that contains a descriptive system comprising of 5 dimensions using child-friendly wording (mobility, looking after myself, doing usual activities, having pain or discomfort, feeling worried, sad or unhappy), each of which has 3 levels (no problems, some problems, a lot of problems). The next component of the instrument consists of a visual analogue scale (VAS) on which the respondent rates their perceived health from 0 to 100, with respective anchors of “worst imaginable health state” and “best imaginable health state.”32,33 Patients enrolled in the VOYAGE trial, who could read, were encouraged to complete the questionnaire by themselves.

The instrument produces 3 types of data for each respondent, a profile indicating the extent of problems on each of the 5 dimensions represented by a 5-digit descriptor, a population preference-weighted health index score based on the descriptive system, and a self-reported assessment of health status based on the VAS. The index score is arrived at by applying a multiattribute utility function to the descriptive system. Scores of 0 represent the health state “dead” and 1 represents “perfect health.” Lower scores on 5-digit health status and higher scores on index and VAS represent better HRQoL.32,33 No MID specific to asthma in children was identified during the CADTH literature review. Assessments during VOYAGE trial were conducted on weeks 2, 24, and 52.

Harms

For each patient, SAEs and AEs of special interest were monitored and documented from the initial visit until the end of the study or rollover to the extension study. AEs that were ongoing at database lock were also captured. An independent data monitoring committee was reviewing the safety data on a periodic basis throughout the course of the trial.

Statistical Analysis

Primary Outcome of the Study
Power Calculation

The sample size of this study was based on the primary outcome (i.e., annualized rate of severe exacerbations over 52 weeks of treatment) for the following populations: baseline blood EOS of 300 cells/µL or greater, baseline blood EOS of 150 cells/µL or greater, and patients with type 2 inflammatory phenotype (baseline blood EOS ≥ 150 cells/µL or baseline FeNO ≥ 20 ppb).

The following assumptions were made: the number of severe exacerbations follows a negative binomial distribution, a randomization ratio of 2:1 was used, and a linear discontinuation rate of 20% at 1 year (i.e., average exposure duration for patients of 0.9 year). Moreover, the assumed RR reductions were based on the results in the phase III study EFC13579 (QUEST) for dupilumab in adolescent and adult patients with asthma.34

For patients with baseline EOS of 300 cells/µL or greater, assuming a placebo annualized severe exacerbation rate of 0.8 and a dispersion parameter of 1.5, with approximately 255 patients randomized (170 for dupilumab and 85 for matching placebo group), there was 96% power to detect a RR reduction of 60% (i.e., annualized rate of 0.32 for the dupilumab group) at a 2-sided significance of 5%.

For patients with baseline EOS of 150 cells/µL or greater, assuming a placebo annualized severe exacerbation rate of 0.7 and a dispersion parameter of 1.5, with approximately 327 patients randomized (218 for dupilumab and 109 for matching placebo group), there was 93% power to detect a RR reduction of 54% (i.e., annualized rate of 0.322 for the dupilumab group) at a 2-sided significance of 5%.

For patients with type 2 inflammatory phenotype, assuming a placebo annualized severe exacerbation rate of 0.7 and a dispersion parameter of 1.5, with approximately 345 patients randomized (230 for dupilumab and 115 for matching placebo group), there was 94% power to detect a RR reduction 54% (i.e., annualized rate of 0.322 for the dupilumab group) at a 2-sided significance of 5%.

According to the sponsor, approximately 402 patients in the overall population (268 for dupilumab and 134 for placebo) needed to be randomized to achieve target sample sizes, with an assumption that 86%, 81%, and 64% of patients have a type 2 inflammatory phenotype, baseline blood EOS of 150 cells/µL or greater, and baseline blood EOS of 300 cells/µL or greater, respectively.

Multiplicity Considerations

In VOYAGE, a hierarchical testing strategy was implemented to test for superiority of dupilumab over placebo in the primary (annualized rate of severe exacerbation events during the 52-week placebo-controlled treatment period), key secondary (change from baseline in prebronchodilator percent predicted FEV1 at week 12), and secondary outcomes (change in ACQ-7-IA at week 24, change from baseline in FeNO at week 12), while controlling the overall type I error rate at 0.05 (2-sided). The sponsor included 2 distinct sequential testing procedures, based on approved indication in adults and adolescents.

  • For the US and US reference countries: testing hierarchy started with the population with a baseline blood EOS of 300 cells/µL or greater (Appendix 3; Table 40)
  • For European Union and European Union reference countries: testing hierarchy started with the population with a type 2 inflammatory phenotype (Appendix 3; Table 41)
Statistical Test or Model

The analysis of the primary end point estimated annualized exacerbation rates over 52 weeks for treatment groups as well as the RR, 95% CIs, and P values between treatment and placebo arms, and was performed using a negative binomial model. Details of the statistical model, adjustments, and populations analyzed are provided in Table 9. In the primary analysis approach, off-treatment measurements of patients who prematurely discontinued treatment were included in the analysis since patients who permanently discontinued study medication were encouraged to complete remaining study visits. For patients discontinuing the study before week 52, analyses were censored at the time of study discontinuation, and all observed severe exacerbation events up to the last contact date were included in the analysis. No imputations were performed for the unobserved events after study discontinuation and up to week 52.

An assessment of time to first severe asthma exacerbation event was conducted via Cox regression model that yielded hazard ratio estimates (dupilumab versus placebo) along with its 95% CIs. Kaplan-Meier curves were used to derive the proportion of patients with a severe asthma exacerbation event at various time points in each treatment group (Table 9).

Preplanned sensitivity analyses were conducted on the primary outcome in the VOYAGE trial for the type 2 inflammatory phenotype and baseline blood EOS of 300 cells/µL or greater populations (Refer to Table 9).

Key Secondary Outcome

The change from baseline in prebronchodilator percent predicted FEV1 at week 12 was compared to placebo using a mixed-effect model with repeated measures (MMRM), and LS mean of each treatment group as well as difference of LS means between treatment and placebo arms, 95% CIs, and P values were reported. Details of the statistical model, adjustments, and populations analyzed is provided in Table 9.

The primary analytical approach included additional off-treatment percent predicted FEV1 values measured up to week 12 for patients discontinuing the treatment before week 12, who were encouraged to complete all remaining study visits. Data from patients who withdrew from the study before week 12 were considered as missing after study discontinuation and no imputation was performed in the primary analysis.

Preplanned sensitivity analyses were conducted on the key secondary outcome in the VOYAGE trial for the type 2 inflammatory phenotype and baseline blood EOS of 300 cells/µL or greater populations (Refer to Table 9).

Subgroup Analyses

Preplanned subgroup analyses were conducted on the primary and key secondary outcome in the VOYAGE trial for the following subgroups identified in the CADTH review protocol:

  • baseline eosinophil counts ( < 300 cells/µL, ≥ 300 cells/µL; < 150 cells/µL, ≥ 150 cells/µL; < 150 cells/µL, ≥ 150 to < 300 cells/µL, ≥ 300 to < 500 cells/µL, ≥ 500 cells/µL) (performed in the ITT population)
  • ICS dose at baseline (medium or high) (performed in the type 2 inflammatory asthma phenotype population and baseline blood EOS ≥ 0.3 Giga/L population)
  • Number of exacerbations in the past year (≤ 1, 2, > 2) (performed in the type 2 inflammatory asthma phenotype population and baseline blood EOS ≥ 0.3 Giga/L population)
  • Atopic medical condition (yes, no) and comorbid atopic dermatitis (yes, no) (performed in the type 2 inflammatory asthma phenotype population and baseline blood EOS ≥ 0.3 Giga/L population)

For the primary outcome, treatment by subgroup interaction and its P value was derived from a negative binomial model. Similarly, treatment by subgroup interaction at week 12 and its P value was derived from an MMRM for the key secondary outcome of interest. RRs and risk differences for the primary outcome, and LS mean differences for the key secondary outcome comparing dupilumab versus placebo for the subgroups were reported. In both cases, evidence of quantitative treatment by subgroup interaction with nominal P < 0.05 for any subgroup would follow-up with an assessment of qualitative interaction via Gail-Simon test.

In VOYAGE, the specific subgroup of patients with eosinophil counts of 150 cells/μL or greater, with eosinophil counts of 300 cells/μL or greater, and with high ICS dosing at baseline were included as part of the statistical hierarchy. All other subgroups were not adjusted for multiplicity.

Other Secondary Outcomes

Analyses of change from baseline in other secondary end points with a continuous nature was derived through a similar MMRM (Table 9).

In addition to the MMRM analyses, a responder analysis was performed for the ACQ-7, 5-item ACQ (ACQ-5), and Asthma Quality of Life Questionnaire total scores at weeks 12, 24, 36, and 52. Specifically, a logistic regression model was used to obtain the odds ratio (OR) of being a responder comparing dupilumab and placebo group along with the corresponding 95% CI and P value (Table 9).

Analysis Populations

In the VOYAGE trial, there were 2 primary efficacy populations, for which all efficacy outcomes were analyzed:

  • Type 2 inflammatory asthma phenotype (randomized patients with baseline blood eosinophil count ≥ 150 cells/µL or baseline FeNO ≥ 20 ppb)
  • Baseline blood EOS count of 300 cells/µL or greater (randomized patients with baseline blood EOS count ≥ 0.3 Giga/L)

There were additional efficacy populations, for which primary and selected secondary end points were analyzed in a multiplicity-controlled manner, including patients with baseline blood eosinophil count of 150 cells/µL or greater.

Safety analyses were conducted according to the treatment patients actually received and were based on the safety population (i.e., all patients receiving at least 1 dose or part of a dose of the trial treatment).

Table 9. Statistical Analysis of Efficacy End Points.

Table 9

Statistical Analysis of Efficacy End Points.

Protocol Deviations

The sponsor identified major protocol deviations that may have a potential adverse impact on data integrity, patients’ rights, or safety. These were reported, within the type 2 inflammatory asthma phenotype population, in 5 (2.1%) and 3 (2.6%) patients in the dupilumab and placebo group, respectively. Within the population with a baseline blood EOS count of 300 cells/µL or greater, these were reported in 4 (2.3%) and 2 (2.4%) patients in the dupilumab and placebo group, respectively. The types of protocol deviations included missing assessments of FeNO or blood EOS at baseline, low adherence to background and study treatments, and use of prohibited concomitant medications.

Randomization and Dosing Irregularities

In the 2 primary efficacy populations, randomization and drug allocation irregularities were reported in 19 (8.1%) in the dupilumab and 8 (7.0%) patients in the placebo group (type 2 inflammatory asthma phenotype population) and in 12 (6.9%) in the dupilumab and 6 (7.1%) patients in the placebo group (baseline blood EOS ≥ 300 cells/µL population). The majority of errors were related to the ICS dose level stratum classification reported by the investigators, which occurred in both directions (i.e., medium erroneously classified as high and vice versa). Specifically, misclassification according to the ICS dose levels occurred in 18 (7.6%) and 7 (6.1%) of type 2 population patients in the dupilumab and placebo group, respectively, and in 11 (6.3%) and 5 (6.0%) patients in the population with baseline EOS of 300 cells/µL or greater in the dupilumab and placebo group, respectively. The sponsor reported that the analyses factoring ICS dose levels were conducted according to the actual calculated total daily dose of all medications containing ICSs and not on the investigators’ reported classification.

Breaking of the Blind

In the VOYAGE trial, breaking of the blind for regulatory purposes occurred due to serious adverse reactions suspected to be related to treatment (2 patients in the dupilumab group: 1 reported pneumonia and 1 patient had eosinophilia, headache, and blurred vision; 1 patient had lymphadenitis viral in the placebo group)

Local breaking of the blind by the investigators occurred only in the dupilumab group, among 4 patients from Brazil (discontinuation due to yellow fever vaccination) and 1 patient from the US (reporting AE of eosinophilia, headache, and blurred vision).

The sponsor specified that patient withdrawal from treatment would occur only in the case of breaking of the blind occurring at a local level, not a study level.

Amendments to the Protocol

The original protocol, dated August 04, 2016, was modified per 3 global amendments (Amendment 1: March 10, 2017; Amendment 2: June 18, 2018; Amendment 3: October 1, 2019) and 1 local amendment for Brazil (February 2, 2018).

Notably, Amendment 3 included following modifications:

  • Changes to the study primary efficacy analysis population (from an overall uncontrolled persistent asthma population to the population with baseline eosinophil count ≥ 0.3 Giga/L or with the type 2 inflammatory asthma phenotype)
  • Changes to the sample size
  • Specification of different hierarchy orders used for US and US reference countries and European Union and European Union reference countries
  • Removal of the limit in enrolling patients according to background therapy with medium-dose ICS or blood eosinophil count level
  • Defining the planned database lock
  • Classification of FeNO as a secondary end point instead of an exploratory end point

Additional changes to the Study Analysis plan were made as per regulatory agency request on August 14, 2020, to provide a detailed elaboration of the primary estimate and to provide further analyses to evaluate whether baseline FeNO values can independently predict treatment effect. Moreover, addition of outcome measures (SCS exposure, loss of asthma control), COVID-19 relevant analysis, and adjustments to subgroup analyses (removal of elevated IgE [yes/no] and age subgroup analyses; addition of treatment exposure-adjusted analysis for key AE end points) were implemented.

Post hoc analyses (i.e., implemented after database lock) was added to evaluate effects of dupilumab on some end points of interest. These included annualized rate of the subtype of severe asthma exacerbation resulting in hospitalization or ED visits resulting in hospitalization (for the type 2 inflammatory asthma phenotype and with baseline blood EOS ≥ 300 cells/µL populations), responder analyses for ACQ-7-IA (based on minimal clinically important difference cut-off points of 0.75,1, and 1.5 as a responder definition), and biomarker analyses (interaction of biomarkers of type 2 inflammation, baseline EOS and FeNO levels) on the effect of dupilumab on the primary end point and key secondary end point based on the ITT population.

Results

Patient Disposition

A total of ||||||||| patients were screened for the study. The screening failure rate was ||||||||| and the major reason for screening out of the study was failure to meet inclusion criteria described in the Table 6 (|||||||||), consisting of lack of appropriate background therapy, exceeding the specified maximum prebronchodilator FEV1, reversibility in FEV1 that was lower than specified minimum, or lack of evidence of uncontrolled asthma.

Out of ||||||||| screened patients, 408 underwent randomizations (ITT population). In the type 2 inflammatory phenotype and population with baseline blood EOS of 300 cells/µL or greater, there were a total of 350 (236 in dupilumab and 114 in the placebo arm) and 259 patients (175 in dupilumab and 84 in the placebo arm), respectively (Table 10). Three patients in the dupilumab group were randomized, but not treated (due to erroneous randomization despite ineligibility of 2 patients and withdrawal from the study before treatment by 1 patient).

Treatment discontinuations were generally below ||||||||| in each group across both efficacy populations, and were higher in the dupilumab group, compared to placebo group (dupilumab versus placebo: |||||||||||||||||| in the type 2 population; |||||||||||||||||| in the baseline EOS ≥ 300 cells/µL population). The most common reason for discontinuation was “other,” occurring in ||||||||||||||||||||||||||| of dupilumab versus placebo patients in both efficacy populations, respectively. The sponsor reported that |||||||||||||||||||||||||||||||||||| due to “other” reasons was related to the safety issues. Specifically, in the dupilumab group (ITT population), discontinuation due to “other reasons” included: |||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||| Discontinuation for “other reasons” in the placebo group (ITT population) was due to |||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||. Percentage of patients who withdrew due to AEs was similar in dupilumab and placebo arms, across all efficacy populations. Study withdrawals were reported in ||||||||||||||||||||||||||| of type 2 patients of the dupilumab and placebo group, and in ||||||||||||||||||||||||||| of patients with baseline EOS of 300 cells/µL or greater in the dupilumab and placebo groups, respectively.

Overall, more patients in the placebo group, compared to dupilumab group, continued to the long-term extension study (dupilumab versus placebo: ||||||||||||||||||||||||||| in the type 2 population; ||||||||||||||||||||||||||| in the baseline EOS ≥ 300 cells/µL population).

Disposition of study patients in the ITT and baseline blood EOS of 150 cells/µL or greater populations is presented in the Appendix 3.

Table 10. Patient Disposition (VOYAGE Trial) .

Table 10

Patient Disposition (VOYAGE Trial) .

Exposure to Study Treatments

Study duration for the dupilumab and placebo groups of the VOYAGE trial was similar, with mean values of ||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||| for dupilumab and placebo arm, respectively, in the type 2 inflammatory asthma population and mean values of |||||||||||||||||||||||||||||||||||||||||||||||||||||| for dupilumab and placebo arm, respectively, in the population with baseline blood EOS of 300 cells/µL or greater. Median duration of the study treatment was the same for both groups (365 days) in both efficacy populations.

Regarding the adherence to study treatment, an average of |||||||||||||||||||||||||||||||||||| of adherence to injections was observed among the patients in dupilumab and placebo groups in the type 2 inflammatory asthma population, respectively. For the population with baseline blood EOS of 300 cells/µL or greater, mean adherence to injections was ||||||||||||||||||||||||||||||||||||||||||||| in the dupilumab and placebo arms, respectively.

Mean percentage of days in which patients were compliant with all background controller medications was ||||||||||||||||||||||||||| in the dupilumab group and ||||||||||||||||||||||||||| in the placebo group (for the type 2 population) and ||||||||||||||||||||||||||| in the dupilumab group and ||||||||||||||||||||||||||| in the placebo group (baseline blood EOS ≥ 300 cells/µL population).

Efficacy

Only those efficacy outcomes and analyses of subgroups identified in the review protocol are reported in the following. Refer to Appendix 3 for detailed efficacy data.

Mortality

In the VOYAGE trial, there were no deaths reported across the dupilumab and placebo groups.

Asthma Exacerbations

The adjusted rate over 52 weeks in the type 2 inflammatory asthma population was 0.31 (95% CI, 0.22 to 0.42) with dupilumab and 0.75 (95% CI, 0.54 to 1.03) with placebo, for a RR of 0.41 (95% CI, 0.27 to 0.61; P < 0.0001) and a risk difference of –0.44 (95% CI, –0.68 to –0.20) (Table 11). In the patients with an EOS count of at least 300 cells/µL at baseline, the adjusted rate of severe asthma exacerbations across 52 weeks was 0.24 (95% CI, 0.16 to 0.35) in the dupilumab group and 0.67 (95% CI, 0.47 to 0.95) in the placebo group (RR = 0.35; 95% CI, 0.22 to 0.56; P < 0.0001; risk difference: –0.43; 95% CI, –0.66 to –0.20).

Table 11. Outcome: Severe Asthma Exacerbations During the 52-Week Randomized Treatment Period (Type 2 Inflammatory Asthma Phenotype Population; Population With Baseline Blood Eosinophils of 300 cells/µL or Greater).

Table 11

Outcome: Severe Asthma Exacerbations During the 52-Week Randomized Treatment Period (Type 2 Inflammatory Asthma Phenotype Population; Population With Baseline Blood Eosinophils of 300 cells/µL or Greater).

Various sensitivity analyses were conducted to account for missing data, and results were consistent with that of the primary analysis, (Appendix 3).

RRs for the comparison of severe exacerbations associated with an ED visit or hospitalization between dupilumab and placebo were ||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||, while the risk differences were ||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||| for the populations with type 2 asthma and baseline blood EOS of 300 cells/µL or greater, respectively. Adjusted rates of severe exacerbations associated with a hospitalization were not estimable due to ||||||||| events reported in the placebo group.

Subgroup analyses for the primary outcome were performed for several subgroups relevant to the CADTH protocol (baseline eosinophil levels, baseline ICS dose, number of previous asthma exacerbations, atopic medical history) (Table 12, Table 13, Table 14, and Table 15). In the subgroup of patients with baseline blood EOS of 150 cells/µL or greater, RRs between dupilumab and placebo were 0.390 (95% CI, 0.261 to 0.583 to |||||||||||||) and the risk difference ||||||||||||||||||||||||||||||||||||, with a P value for the interaction of ||||||||||||||||||. Other subgroup analyses did not yield statistically significant interaction.

Results of the primary end point analyses in the ITT and baseline blood EOS of 150 cells/µL or greater populations were aligned with those presented in the type 2 and baseline blood EOS of 300 cells/µL or greater populations (Table 12, Appendix 3).

Table 12. Subgroup Analysis Results From VOYAGE Trial for the Primary Outcome by Baseline Blood Eosinophil Count (Adjusted Annualized Rate of Severe Exacerbation Events During the 52-Week Treatment; ITT population) .

Table 12

Subgroup Analysis Results From VOYAGE Trial for the Primary Outcome by Baseline Blood Eosinophil Count (Adjusted Annualized Rate of Severe Exacerbation Events During the 52-Week Treatment; ITT population) .

Table 13. Redacted.

Table 13

Redacted.

Table 14. Redacted.

Table 14

Redacted.

Table 15. Redacted.

Table 15

Redacted.

Asthma Symptoms

ACQ-7 scores decreased (improved) from baseline to week 24 in both the dupilumab and placebo groups, for an LS difference versus placebo of –0.33 points (95% CI, –0.50 to –0.16; |||||||||||| in the type 2 population and of –0.46 points (95% CI, –0.66 to –0.26; |||||||||||| in the baseline blood eosinophil count of 300 cells/µL or greater population. In the type 2 inflammatory asthma population, there were |||||||||||| of dupilumab patients and |||||||||||| of placebo patients who were responders at week 24, for an OR of ||||||||||||||||||||||||||||||||||||||||||||||||. In the baseline eosinophil count of 300 cells/µL or greater population, there were |||||||||||| responder patients in the dupilumab and |||||||||||| responder patients in the placebo arm at week 24, with an OR of |||||||||||||||||||||||||||||||||||||||||||||||| (Table 16).

Results were maintained during the trial period (to week 52) |||||||||||||||||||||||||||||||||||||||||||||||| (Table 16, Figure 3, and Figure 4). Appendix 3 contains findings from the ACQ-7 assessments conducted across additional efficacy populations of the VOYAGE trial (ITT and baseline blood EOS ≥ 150 cells/µL populations), as well as results obtained from the ACQ-5 assessments in the type 2 inflammatory asthma phenotype and baseline eosinophil count of 300 cells/µL or greater populations.

Table 16. Symptoms According to the ACQ-7 (Weeks 24 and 52) .

Table 16

Symptoms According to the ACQ-7 (Weeks 24 and 52) .

The figure presents mean change from baseline ACQ-7-IA at each time point, for the type 2 inflammatory asthma phenotype population. Least squares mean change from baseline is depicted on the y-axis and week is depicted on the x-axis. The solid red curve represents the placebo group and the dotted blue line represent the dupilumab group. The vertical bars represent the standard errors.

Figure 3

LS Mean Change From Baseline in ACQ-7-IA Over Time (MMRM Including Measurements Up to Week 52): Type 2 Inflammatory Asthma Phenotype Population.

Reduction in Use of OCSs

Total SCSs usage during the treatment period was captured in the VOYAGE trial. Overall, more patients in the placebo arm (40.4% and 41.7%) compared to patients in the dupilumab arm (24.2% and 22.3%) received treatment with SCS during the trial within the type 2 inflammatory asthma phenotype and baseline blood EOS of 300 cells/µL or greater populations, respectively (Table 17).

The adjusted RR in annualized number of SCS courses, comparing dupilumab to placebo, was 0.407 (95% CI, 0.272 to 0.609) for the type 2 inflammatory asthma phenotype population and 0.340 (95% CI, 0.212 to 0.545) for the baseline blood EOS of 300 cells/µL or greater population.

The figure presents mean change from baseline of ACQ-7-IA at each time point, for the population with a baseline blood eosinophils of 300 cells/µL or greater. Least squares mean change from baseline is depicted on the y-axis and week is depicted on the x-axis. The solid red curve represents the placebo group and the dotted blue line represent the dupilumab group. The vertical bars represent the standard errors.

Figure 4

Least Squares Mean Change From Baseline in ACQ-7-IA Over Time (MMRM Including Measurements Up to Week 52): Population With Baseline Blood Eosinophils of 300 cells/µL or Greater.

Table 17. Total SCS Usage During the Treatment Period.

Table 17

Total SCS Usage During the Treatment Period.

Pulmonary Function

For the key secondary outcome (change from baseline in percent predicted prebronchodilator FEV1 at week 12) within the type 2 inflammatory asthma phenotype population, LS mean difference between the groups was 5.21% (95% CI, 2.14 to 8.27%; P = 0.0009). Similarly, in the baseline blood eosinophil count of 300 cells/µL or greater population, at week 12 the LS mean difference between groups was 5.32% (95% CI, 1.76 to 8.88%; P = 0.0036). In both primary efficacy populations, LS mean changes in the percent predicted prebronchodilator FEV1 were sustained through week 52 (Table 18).

Findings from other multiplicity-controlled analyses for the ITT and baseline blood EOS of 150 cells/µL or greater populations were similar to those observed in the 2 primary efficacy populations (Appendix 3).

A series of sensitivity analyses were conducted to account for missing data as well as the effects of SCS exposure, showing consistent and similar findings to the primary analysis (Appendix 3).

Subgroup analyses for change from baseline in percent predicted prebronchodilator FEV1 at week 12 in patients with baseline blood EOS of 300 cells/µL or greater and 150 cells/µL or greater were generally consistent with the primary analyses. Other subgroup analyses reported were not adjusted for multiplicity (Table 19, Table 20, Table 21, and Table 22).

In both the type 2 inflammatory asthma phenotype and baseline blood eosinophil count of 300 cells/µL or greater populations, there was an improvement in morning PEF scores at week 12 in both treatment groups, with the LS mean difference versus placebo of 14.02 L/min (95% CI, 4.92 to 23.12; P = 0.0026) and 14.48 L/min (95% CI, 3.49 to 25.47; P = 0.01) for the 2 populations, respectively (Table 18).

Changes at week 12 of evening PEF values were reported among the 2 populations, with LS mean differences versus placebo of 17.97 L/min (95% CI, 8.98 to 26.95; P = 0.0001) and 19.10 L/min (95% CI, 8.36 to 29.83; P = 0.0005) for the 2 populations (Table 18).

Table 18. Pulmonary Function Measurements (Percent Predicted Prebronchodilator FEV1, a.m. and p.m. PEF) .

Table 18

Pulmonary Function Measurements (Percent Predicted Prebronchodilator FEV1, a.m. and p.m. PEF) .

Table 19. Subgroup Analysis Results From VOYAGE Trial for the Key Secondary Outcome by Baseline Blood Eosinophil Count (Change From Baseline in Prebronchodilator Percent Predicted FEV1 at Week 12; ITT population) .

Table 19

Subgroup Analysis Results From VOYAGE Trial for the Key Secondary Outcome by Baseline Blood Eosinophil Count (Change From Baseline in Prebronchodilator Percent Predicted FEV1 at Week 12; ITT population) .

Table 20. Subgroup Analysis Results From VOYAGE Trial for the Key Secondary Outcome by Baseline ICS Dose (Change From Baseline in Prebronchodilator Percent Predicted FEV1 at Week 12) .

Table 20

Subgroup Analysis Results From VOYAGE Trial for the Key Secondary Outcome by Baseline ICS Dose (Change From Baseline in Prebronchodilator Percent Predicted FEV1 at Week 12) .

Table 21. Redacted.

Table 21

Redacted.

Reduction in Dose of ICSs

The VOYAGE study protocol allowed a permanent increase in background medications after 2 or more severe asthma exacerbations. During the treatment period of the trial, ||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||, across all efficacy populations assessed.

Health-Related Quality of Life

PAQLQ(S)–Interviewer Administered was assessed in the VOYAGE trial among children aged 7 years to younger than 12 years at randomization. LS mean values of PAQLQ(S) scores showed increases (improvements) from baseline to week 24 in type 2 inflammatory asthma phenotype population, with a difference between groups of 0.19 points (95% CI, –0.03 to 0.40, |||||||||||||||) (Table 23). In the baseline blood EOS of 300 cells/µL or greater population, the LS mean difference between groups at week 24 was 0.30 points (95% CI, 0.06 to 0.54; ||||||||||).

PAQLQ(S) responders at week 24 were also reported, defined as a change from baseline of 0.5 or greater, and in type 2 inflammatory asthma population there were |||||||||| of dupilumab patients and |||||||||| of placebo patients who were responders (OR = |||||||||| |||||||||| |||||||||| ||||||||||. In the baseline blood EOS of 300 cells/µL or greater population, there were 72.8% of dupilumab responder patients and 63% placebo responder patients (OR = 1.84; 95% CI, 0.92 to 3.65).

Improvements in the PAQLQ(S) scores were maintained through the week 52 of the VOYAGE trial, across both efficacy populations (Figure 5 and Figure 6).

Findings from the PAQLQ(S) assessments conducted across additional efficacy populations of the VOYAGE trial (ITT and baseline blood EOS ≥ 150 cells/µL populations) are reported in the Appendix 3.

Table 22. Redacted.

Table 22

Redacted.

The EQ-5D-Y VAS also showed improved HRQoL from baseline in both analysis populations in favour of dupilumab at week 24 (Table 24).

Impact of children’s (≥ 7 years and < 12 years old) asthma on the caregivers’ quality of life was assessed with PACQLQ. Increases from baseline to week 24 in the PACQLQ global score compared to placebo were observed in both the type 2 inflammatory asthma phenotype population (LS mean difference between groups = 0.25; 95% CI, 0.00 to 0.50; P = 0.0531) and baseline blood EOS of 300 cells/µL or greater population (LS mean difference between groups = 0.34; 95% CI, 0.01 to 0.67; P = 0.0445). At week 52, observed PACQLQ LS mean differences for comparison of dupilumab versus placebo were 0.47 (95% CI, 0.22 to 0.72; P = 0.0003) and 0.50 (95% CI, 0.21 to 0.79; P = 0.0007), in the 2 populations, respectively (Table 24).

Table 23. PAQLQ(S)-IA Global Score.

Table 23

PAQLQ(S)-IA Global Score.

This figure has been redacted.

Figure 5

Redacted.

This figure has been redacted.

Figure 6

Redacted.

Table 24. Redacted.

Table 24

Redacted.

Reduction in Use of Rescue Medication

Regarding the use of reliever medications in the VOYAGE trial, a decrease in number of puffs in 24-hour period was observed in both treatment arms. The LS mean difference versus placebo for the number of puffs of reliever medication in a 24-hour period at week 24 was |||||||||| |||||||||| ||||||||||||||| |||||||||| ||||| and |||||||||| |||||||||| ||||||||||||||| |||||||||| ||||| for the 2 populations, respectively. At week 52, |||||||||| |||||||||| ||||||||||||||| |||||||||| ||||||||||||||| |||||||||| ||||| (Table 25).

Table 25. Redacted.

Table 25

Redacted.

Improvements in Symptoms of Atopic Dermatitis and Rhinosinusitis

In patients with comorbid allergic rhinitis, PRQLQ–Interviewer Administered global scores at weeks 24 were improved in favour of dupilumab (LS mean difference between groups = |||||||||| |||||||||| ||||||||||||||| |||||||||| |||||, in the type 2 phenotype population. Improvements in allergic rhinitis related quality of life were also observed in the baseline blood EOS of 300 cells/µL or greater population (LS mean difference between groups = |||||||||| |||||||||| ||||||||||||||| |||||||||| ||||||||||||||| |||||||||| |||||) (Table 24).

Harms

Only those harms identified in the review protocol are reported in the following. Refer to Table 26 for detailed harms data.

Adverse Events

AEs in the dupilumab versus placebo groups occurred in 83% versus 79.9% of patients in the safety population of the VOYAGE trial, respectively (Table 26). The most common AEs for dupilumab versus placebo were nasopharyngitis (18.5% versus 21.6%), viral upper respiratory tract infection (12.2% versus 9.7%), pharyngitis (8.9% versus 10.4%), bronchitis (6.3% versus 10.4%), allergic rhinitis (5.9% versus 11.9%), injection site erythema (12.9% versus 9.7%) and injection site edema (10.3% versus 5.2%).

Serious Adverse Events

SAEs occurred in 4.8% patients receiving dupilumab and 4.5% of patients receiving placebo, most common of which were asthma (dupilumab versus placebo: 1.5% versus 0%) and eosinophilia (dupilumab versus placebo: 0.7% versus 0%).

Withdrawals Due to AEs

AE resulting in discontinuation of study drug occurred in 1.8% versus 1.5% of patients of the VOYAGE study, in the dupilumab versus placebo groups, respectively.

Notable Harms

Injection site reactions were the most commonly reported notable harms, with 17.7% patients versus 13.4% of patients in the dupilumab versus placebo groups, respectively. Hypersensitivity and anaphylactic reactions occurred in 1.8% of dupilumab patients versus 3.7% of placebo patients and 0.0% of dupilumab patients versus 1.5% of placebo patients, respectively. Severe infections occurred in |||||||||| of patients in the dupilumab and |||||||||| of patients in the placebo group. Parasitic infections were reported among 7 patients (2.6%) in the dupilumab group, and no patients in the placebo arm. There were no opportunistic infections reported in the trial. Eosinophilia was more frequently occurring in dupilumab arm compared to placebo (6.6% versus 0.7%, respectively). Conjunctivitis, classified using a narrow custom Medical Dictionary for Regulatory Activities (MedDRA) Query search, occurred in 2.6% of the dupilumab group versus 6.7% of the placebo group. Similarly, conjunctivitis classified using a broad MedDRA search, occurred in 3.0% of the dupilumab group versus 7.5% of the placebo group.

Table 26. Summary of Harms (Safety Population) .

Table 26

Summary of Harms (Safety Population) .

Critical Appraisal

Internal Validity

The VOYAGE trial is a multinational, multicentre, randomized, double-blind, placebo-controlled study. Treatment allocation was appropriately performed through a central interactive voice and web response systems. However, the randomization ratio of 2:1 was implemented in the trial, and no justification was provided from the sponsor. Furthermore, the study randomization was stratified by ICS dose level (medium or high) at screening, patients’ blood eosinophil count (< 300 cells/µL and ≥ 300 cells/µL populations) at screening, and region (Latin America, Eastern Europe, and Western countries).

The study was double-blind, and both the patients and investigators were blinded to the study treatment. Additional steps to maintain blinding were undertaken by using a matching placebo in the trial (i.e., 2 different volumes of injection solutions, corresponding to the 2 different doses of dupilumab were used; these were each matched to corresponding volumes of placebo injection). However, this meant that patients and investigators were aware of the dose or volume of the injections administered in the trial. Even though dosing awareness might have introduced the possibility of bias, the fact that both patients and investigators were blinded to the type of treatment would have mitigated this bias. Furthermore, patients in the dupilumab group experienced more injection site reactions and eosinophilic reactions compared to placebo, which might have created possibilities for study participants or investigators to anticipate the AEs of active treatment, therefore, introducing unblinding in the trial. The impact of such unblinding (i.e., patients believing they were assigned to the treatment group rather than placebo) might have introduced bias in assessing patient-reported and HRQoL outcomes of the trial. During the VOYAGE trial, breaking of the blind leading to study discontinuation occurred in a limited number of patients receiving dupilumab. These irregularities were reported at the local level among 4 patients from Brazil (discontinued due to yellow fever vaccination) and 1 patient from US (reported AE of eosinophilia, headache, and blurred vision, leading to study discontinuation).

The sponsor controlled for multiplicity using a hierarchical testing procedure, which was applied for the primary outcome (annualized rate of severe exacerbation events during the 52-week placebo-controlled treatment period), key secondary outcome (change from baseline in prebronchodilator percent predicted FEV1 at week 12) and secondary outcomes (change in ACQ-7-IA at week 24, change from baseline in FeNO at week 12). Each hypothesis was formally tested only if the preceding 1 was significant at the 2-sided 5% level. There were 2 distinct sequential testing procedures that incorporated the same end points, but in a different order, based on the approved indication in adults and adolescents in the US and European Union. For European Union and European Union reference countries, the hierarchy procedure started with the type 2 inflammatory asthma phenotype population, and for the US and US reference countries, the hierarchy procedure started with the population with baseline blood eosinophil count of 0.3 Giga/L or greater. Of note, statistical significance was reached across all multiplicity-controlled end points in both hierarchical testing procedures.

Sample size and power calculations were based on the primary outcome for the populations with a type 2 inflammatory phenotype, baseline blood EOS of 300 cell/μL or greater, and of 150 cells/μL or greater. When calculating sample size and power of the trial, the sponsor made assumptions regarding expected RR reductions in the pediatric population, based on data previously reported from trials on individuals aged older than 12 years.34 Considering that there is a paucity of data in pediatric populations, this approach was deemed reasonable for a priori estimation of the sample size.

Regarding baseline characteristics, there were slight imbalances across the groups in terms of severe exacerbations (1, 2, 3, or ≥ 4) in the year before the study enrolment, but the median number exacerbations for the dupilumab and placebo groups was the same (median = 2). Moreover, FEV1 reversibility was slightly higher in the dupilumab compared to placebo group, which might have introduced bias in the study findings either for or against the study drug. Bias for the active treatment group might have occurred due to the fact that the VOYAGE trial included patients with uncontrolled asthma, who were more likely to respond to a new therapy and show a difference versus placebo. On the other hand, clinical experts reported that higher baseline reversibility in the dupilumab group suggested that that this group may be more uncontrolled than the placebo, making it more difficult for the active treatment group to reach a beneficial effect, compared to placebo (bias against the drug). Nevertheless, other baseline and demographic characteristics were largely balanced within the 2 study arms, suggesting that randomization was successfully implemented. Screening failure was reported in 35.3% of the initial number of patients, most commonly occurring because patients failed to meet prespecified inclusion criteria of the trial. However, clinicians consulted during this CADTH review reported that higher levels of screening out are very common in asthma biologic trials, noting that virally triggered asthma presentation, commonly reported in pediatric population, might have accounted for these screening failures.

Withdrawals from the study were generally low in the VOYAGE study, but higher proportions were observed in the dupilumab group (dupilumab versus placebo for the type 2 population: ||||||||||||||||||||; dupilumab versus placebo for the baseline EOS ≥ 300 cells/µL population: ||||||||||||||||||||). Similarly, discontinuation from the treatment occurred in approximately 8% of individuals receiving dupilumab, compared to approximately 3.5% of patients receiving placebo across the 2 efficacy populations. According to the data presented in the study report, proportions of individuals who discontinued study treatment due to an AE were balanced across the 2 study arms (approximately 2% for both study arms in the type 2 population and approximately 1% for both study arms in the baseline EOS ≥ 300 cells/µL population), suggesting lower possibility of attrition bias. In an attempt to account for missing data for the primary and key secondary outcomes, the sponsor performed a variety of sensitivity analyses, which are appropriate for scenarios when data are not missing at random. Finding of the sensitivity analyses were aligned with the primary findings. Considering all the above, CADTH deemed that the impact of missing data on the estimated treatment effects was limited.

Annualized rate of severe exacerbation events during the 52-week placebo-controlled treatment period and change from baseline in prebronchodilator percent predicted FEV1 at week 12 were the primary and the key secondary outcomes included in the statistical hierarchy of the VOYAGE trial, respectively. Both outcomes were considered clinically relevant for patients with severe asthma, according to the clinician experts consulted and patient input provided. The definition of the primary outcome (i.e., deterioration in asthma requiring SCSs for at least 3 days or resulting in hospitalizations or ED visits requiring SCSs) as well as the interval between which 2 events were considered as separate (28 days) were regarded as appropriate, according to the clinical experts. Assessment and recording of severe exacerbations were conducted by a treating physician through the electronic case reports. Clinical experts reported that measuring percent predicted values of prebronchodilator FEV1, instead of absolute values, was appropriate for the pediatric population under study. However, the clinicians stated the 12-week assessment of FEV1 was not optimal for assessing the treatment effects of dupilumab versus placebo on pulmonary function because it would not be a sufficient duration to assess the seasonal and other factors that affect this outcome. The experts regarded the 52-week assessment of FEV1 as more clinically relevant.

In the primary analyses for the primary and key secondary outcomes, off-treatment measurements of patients prematurely discontinuing treatment were included in the models, while data of patients after study discontinuations were censored. A series of sensitivity analyses were conducted by the sponsor in an attempt to evaluate the robustness of the primary findings. Specifically, on-treatment analyses to assess treatment effects if patients adhere to the study treatment as directed and missing data imputations (PMM-MI, PMM placebo-based and tipping point analyses) were conducted for both outcomes, while censoring methods according to the SCS usage were performed only for the key secondary outcome. Considering that the multiple imputation methods applied in the VOYAGE trial are applicable to situations when data are not missing at random, CADTH evaluation deemed the methodology applied as appropriate. Findings from the sensitivity analyses were consistent with the primary analyses across the 2 main efficacy populations of the trial.

The VOYAGE trial prespecified a series of subgroup analyses for the primary and the key secondary outcome, aligned with the subgroups of interest identified in the CADTH protocol (baseline eosinophil levels, baseline ICS dose, number of previous asthma exacerbations, atopic medical history). However, only subgroups of patients with baseline blood EOS of 300 cells/µL or greater, baseline blood EOS greater than150 cells/µL and patients receiving high doses of ICS were adjusted for multiple statistical comparisons, as they were part of the hierarchical testing procedure. Statistical tests of interaction were performed to test whether treatment effects differed among subgroups. However, the analyses for other subgroups of interest were not adjusted for multiplicity, and results from these analyses were treated as supportive of the overall benefit of dupilumab but no decisive conclusion can be drawn.

Another secondary outcome included in the hierarchical testing of the VOYAGE trial covered asthma symptoms, measured by a well-established and validated questionnaire (ACQ-7). The MID of 0.5 was established, which is reviewed in detail in Appendix 4.24,25 Although not part of the statistical hierarchy, data regarding the responders’ analysis for the ACQ-7 was reported in the trial, with a response being defined using the 0.5 MID.

Of note, multiplicity adjustments were not conducted for other secondary and exploratory end points of interest, including annualized rates of severe asthma exacerbations resulting in hospitalization or ED visits and resulting in hospitalization only, HRQoL (PAQLQ, PACQLQ, EQ-5D-Y, PRQLQ), pulmonary function (PEF), SCS exposure, reliever medication use, and ICS dose adjustment; therefore, the results for these outcomes will be considered as supportive of the treatment effect but no definitive conclusions can be made.

The results reported in the Clinical Study Report of the VOYAGE trial were based on all the data collected up to the database lock on August 26, 2020. The study protocol underwent 3 global amendments and 1 local amendment for Brazil. Notable changes included the following: change to the study primary efficacy populations (from an overall uncontrolled persistent asthma population to the population with baseline eosinophil count ≥ 300 cells/µL or with the type 2 inflammatory asthma); changes to the sample size; specification of different testing hierarchies based on the US, European Union, and their reference countries; removal of the limitations for enrolling patients according to the background therapy, and EOS levels. These amendments were made before the database lock, suggesting that their ability to affect the end results or imply bias due to patient selection is limited.

Overall, no critical deviations from the protocol were reported during the VOYAGE trial. The sponsor identified a subset of major deviations, which had the potential adverse impact on integrity of the data, patients’ rights, or safety of the participants. These occurred in about 2% of patients across both the placebo and dupilumab arms in the 2 main efficacy populations and included: missing FeNO assessments at baseline, use of prohibited concomitant medications during the treatment period, less than 80% adherence with the treatment under study, and nonadherence with mandatory background therapy during the screening period. Considering that the number of patients with important deviations was low and balanced across the 2 study groups, CADTH reviewers deemed the risk of bias owing to protocol deviations to be low and to have negligible influence on comparative efficacy findings. Moreover, some randomization and dosing irregularities happened during the trial due to a difference in the ICS dose level as reported by the investigator at randomization in comparison to the actual calculated total daily dose of all medications containing ICSs, which was derived after converting to fluticasone dose equivalent according to GINA guidelines.1 The irregularities were fairly balanced between the study groups (7.6% and 6.1% of type 2 population patients in the dupilumab and placebo group, respectively; 6.3% and 6.0% of baseline EOS ≥ 300 cells/µL patients in the dupilumab and placebo group, respectively). To overcome this, the sponsor specified that the analyses factoring ICS dose levels were conducted according to the actual calculated total daily dose of all medications containing ICSs and not on the investigators’ reported classification.

External Validity

The VOYAGE study was a multinational, multicentre trial spanning across 90 sites in 17 countries, including Canada. The population requested for the reimbursement is narrower than the population included in the Health Canada–approved indication, as the OCS-dependant individuals were not taking part in the VOYAGE trial. According to the clinical experts, the number of pediatric patients on regular OCS therapy is limited, but many patients might experience frequent and repeated short course of OCS treatment to gain control of their symptoms or for exacerbations.

Overall, the clinical experts consulted by CADTH agreed that the inclusion and exclusion criteria of the trial were reflective for the severe asthma pediatric population. They also believed that the baseline and demographic characteristics of patients in the trial were largely consistent with the population that would be expected to use the drug in the real-world practice (i.e., poorly controlled individuals experiencing high FEV1 and high reversibility despite being on appropriate therapy). Of note, the majority of the study population included individuals who were White; hence, generalizability of study findings to people living in Canada may be limited in this regard. Moreover, 1 of the main efficacy populations in the VOYAGE trial required that the identification of eligible patients be performed based on FeNO levels. Clinical experts consulted for this review reported that FeNO assessments are not routinely performed in Canadian clinical practice, which limits the generalizability of the study population.

According to the experts, the dosing of dupilumab applied during the clinical trial was consistent with the dose that would be applied in the Canadian clinical practice per the Health Canada–approved indication. Background medications administered during the trial were considered appropriate by the clinical experts consulted and reflective of medications administered in the Canadian setting. The experts also noted that adherence to therapy is a major issue in the management of asthma. The study documentation did not report whether the inhaler technique was checked throughout the trial, which can be considered a limitation. However, adherence to background therapy was high in both groups across the 2 efficacy populations during the study, suggesting that patients might have benefited from the close attention and monitoring of the trial. This might have also explained the level of benefits observed in the placebo population, which was receiving only background medication.

The VOYAGE trial did not compare dupilumab to any of the other biologics approved for the management of asthma in pediatric population, such as IgE inhibitors or the IL-5 inhibitors. Comparison of dupilumab to placebo (added on to standard of care) may be appropriate for establishing efficacy; however, the lack of an active control represents a limitation when trying to assess the comparative effects of dupilumab, leaving indirect comparisons available to assess the relative efficacy and harms of dupilumab compared to other biologics.

Clinical experts consulted during this CADTH review highlighted that the primary and key secondary outcomes were relevant outcomes for assessing pediatric patients with asthma, notably exacerbation rate, pulmonary function (FEV1), and symptoms (ACQ-7). One of the clinical experts consulted for this CADTH review noted that ACQ is mostly applied for research purposes, but additionally commented that the tool can be regarded as appropriate and valid for assessing symptoms in the pediatric population.

Indirect Evidence

Objectives and Methods for the Summary of Indirect Evidence

Due to the lack of direct evidence comparing dupilumab with other existing therapies as an add-on maintenance treatment in patients aged 6 years to younger than 12 years with severe asthma with a type 2 or eosinophilic phenotype or with OCS-dependent asthma, the sponsor submitted 1 ITC analysis.35 In addition, CADTH’s focused literature search for ITCs dealing with asthma was run-in MEDLINE All (1946–) on July 26, 2022. No limits were applied to the search. No published ITCs were identified in the CADTH literature search. The objective of this section is to summarize and critically appraise the indirect evidence from the sponsor-submitted ITC. To align with the research protocol of this review, only information pertaining to the criteria outlined in Table 5 are presented in this section.

Description of Indirect Comparison

The sponsor submitted 1 ITC, which had the objective to identify, evaluate, and synthesize the empirical evidence on the clinical efficacy of dupilumab compared to other recommended biologics for the treatment of pediatric patients who were aged 6 years to younger than 12 years with uncontrolled, moderate-to-severe asthma with a type 2 inflammatory phenotype.

Methods of the Sponsor-Submitted ITC

Study Selection Methods
Systematic Literature Review

The systematic review was conducted in line with the PRISMA guidelines36,37 and Cochrane Handbook,38 and the search strategy was developed based on the PICOS-T criteria presented in Table 27 to identify relevant studies investigating the efficacy and safety of dupilumab against other existing treatments. The systematic literature search was conducted by screening electronic literature databases, spanning from 1998 (aligned with the earliest published omalizumab trial in pediatric asthma) to February 2021. The searches were limited to studies conducted in humans and published in English language. In addition, grey literature searches (January 2019 to February 2021) were conducted as well as snowball screening of the bibliographies of relevant systematic reviews identified across the electronic database. Methods for extracting data and performing quality assessment are presented in the Table 27.

The original searches identified 2,317 publications for further screening. Following screening and a feasibility assessment, a total of 3 trials were included in the ITCs (1 evaluating treatment with dupilumab7 and 2 trials evaluating treatment with omalizumab39,40).

Network Meta-Analysis Feasibility Assessment

To ensure that the assumptions inherent to ITCs were appropriate for any planned analyses, the clinical heterogeneity across all included studies was assessed. This was done by establishing a list of potential treatment effect modifiers (Table 28), which was developed by reviewing subgroup results across the included RCTs and consultations with medical experts to discuss selected subgroups as well as comparability of included trials and feasibility of the analyses.

Table 27. Study Selection Criteria and Methods for the Sponsor-Submitted ITC.

Table 27

Study Selection Criteria and Methods for the Sponsor-Submitted ITC.

Table 28. Characteristics of Potential Treatment Effect Modifiers.

Table 28

Characteristics of Potential Treatment Effect Modifiers.

ITC Analysis Methods

The ITC technical report noted that the clinical experts consulted for the feasibility assessment were split in their assessments of the clinical importance of the heterogeneity between the identified studies. Nonetheless, an “exploratory” Bucher ITC comparing dupilumab with omalizumab was conducted. The ITC analyses included a total of 3 trials connected with placebo as a common comparator (VOYAGE: dupilumab trial; IA05 and ICATA: omalizumab trials). Data from the VOYAGE and IA05 trials were used in the base case ITCs, while data from the ICATA study were included in separate analyses of certain outcomes, such as severe exacerbations, because of differences in key characteristics between the VOYAGE and ICATA trials that “would not allow for sufficient comparability” in the base case.35

The technical report specified that the type 2 inflammatory population from the VOYAGE study was selected as the primary population of interest for the ITCs, in line with the regulatory agencies’ approvals. Since the type 2 asthma population data were not available in the omalizumab trials, an assumption was made that the efficacy of omalizumab would be maintained in the population of interest.

ITC analyses were performed for the following outcomes: severe exacerbations (annualized rates); deterioration of asthma from the VOYAGE study (post hoc analysis) versus severe exacerbations from the IA05 study (annualized rates); morning asthma symptom score at 24 weeks; rescue medication use at 24 weeks; change in PAQLQ(S)-Interviewer Administer at 24 weeks; discontinuations due to AEs at 52 weeks.

Statistical Approach

Since most of the analyses included a single trial per comparison in the network, a fixed-effect model was applied to the Bucher ITCs. For the analysis of severe exacerbations data, which included additional data from the ICATA trial, random effects Bucher ITC was performed.

The P values of significance for the ITC effects were computed in 2-sided tests. The technical report states that due to the limited number of trials in the ITCs, indirectly estimated effects with a P value of 0.05 or less were classified as statistically favourable, a P value greater than 0.05 and 0.15 or greater, as numerically favourable and a P value greater than 0.15 as comparable.

Results of the Bucher ITCs were presented as a central estimate of the relative effect of interest (MID for a continuous outcome and OR or RR for a binary outcome) along with 95% CIs. Analyses were carried out using the metafor package in R 4.0.3.

In the Bucher ITC, statistical heterogeneity for a treatment effect between the same comparison (i.e., omalizumab versus placebo in the IA05 and ICATA trials when the latter study is included) was evaluated using the I2 statistic for the studies. A consistency assessment between direct and indirect sources of evidence was not applicable in the ITCs because there were no head-to-head comparisons between dupilumab and other comparators.

Subgroup Analysis

To account for differences in patient traits (i.e., omalizumab trials included patients with allergic phenotype, assessed via a positive skin prick test or a positive in vitro response to ≥ 1 perennial allergen) and biomarker or clinical characteristics (baseline EOS, percent predicted FEV1, and prior exacerbations), an additional set of analyses was conducted in the “omalizumab-eligible” subgroup with allergic phenotype and corresponding to the inclusion criteria of the IA05 study. The following criteria were used in a post hoc analysis of VOYAGE to better align type 2 inflammatory patients from the VOYAGE trial with those in the IA05 trial:

  • Baseline weight between 20 kg to 50 kg and serum IgE level of 30 IU/mL to 1,300 IU/mL and weight-IgE values combinations based on omalizumab dosing table
  • At least 1 positive perennial allergen-specific IgE (concentration ≥ 0.35 IU/mL) among the following allergens: Alternaria tenuis/alternata; Cladosporium herbarum/hormodendrum; Aspergillus Fumigatus; Cat Dander; D. Farinae; D. Pteronyssinus; Dog Dander; and German Cockroach

Of note, the VOYAGE trial did not include the performance of skin prick test, so the presence of 1 positive perennial allergen-specific IgE was used as a proxy for the allergic phenotype in the IA05 study.

Sensitivity Analyses

To account for differences in the definition of the outcome of interest (severe exacerbations) and increase the comparability between the 2 trials included in the ITC, an additional post hoc analyses of VOYAGE data were performed using an outcome labelled as deterioration of asthma (Table 29).

Table 29. Overview of Outcomes of Interest Assessed in the Eligible Trials and Included in the ITC.

Table 29

Overview of Outcomes of Interest Assessed in the Eligible Trials and Included in the ITC.

Results of ITC

Summary of Included Studies

The 3 double-blind, RCTs trials were VOYAGE (dupilumab 100 mg to 200 mg every 2 weeks), IA05 (omalizumab 75 to 375 mg every 1 month or every 2 months), and ICATA (omalizumab 150 mg to 375 mg every 2 weeks or every 4 weeks), connected via placebo as a common comparator. Characteristics of the study design and patient populations across the included studies are presented in Table 30 and Table 31.

Assessment of Risk of Bias of Included Trials

Methodological quality of all included studies was assessed using the Cochrane Risk of Bias Assessment Tool 1.0. The technical report for the ITC states that the assessed trials were rated as low risk of bias overall. Specifically, all studies showed no evidence of selection, performance, and attrition bias. Both omalizumab trials had unclear reporting, and the presence of detection bias was rated as unclear. Moreover, presence of reporting bias was also unclear in the IA05 study.

Table 30. Redacted.

Table 30

Redacted.

Table 31. Redacted.

Table 31

Redacted.

Results

The key findings of the ITC are presented in Table 32, both for the type 2 Inflammatory asthma phenotype as well as the omalizumab-eligible type 2 inflammatory asthma phenotype patient populations. A visual representation of the evidence network is provided in Figure 7.

This figure has been redacted.

Figure 7

Redacted.

Dupilumab Versus Omalizumab

For the type 2 population, there was an improvement in annualized rates of severe exacerbations (||||||||||||||||||||||||||||||||||||||||||||||||||), and an improvement in deterioration of asthma for the comparison of dupilumab versus omalizumab (||||||||||||||||||||||||||||||||||||||||) (Table 32). No statistically significant between-group differences were found in terms of changes from baseline in morning asthma score, rescue medication use, and PAQLQ(S)–Interviewer Administered as well as withdrawals due to AEs.

Subgroup Analyses

In the omalizumab-eligible type 2 inflammatory subgroup, no significant differences were observed for the comparison of dupilumab with omalizumab in any of the outcomes assessed.

Sensitivity Analyses

Overall, the results of the sensitivity analyses that included data from additional omalizumab trial (ICATA) were aligned with the primary analyses of reduction in severe exacerbation events.

Table 32. Redacted.

Table 32

Redacted.

Critical Appraisal of ITC

Regarding the systematic literature review, the technical report adopted standard methods for conducting and reporting of reviews, aligned with Cochrane and PRISMA guidelines.36,38 Methods of the systematic review included defining the research question according to the PICOS-T criteria, searching through multiple database sources with comprehensive literature searches, adequate screening processes (double reviewers), data extraction using single reviewers and quality control by an additional reviewer, and quality appraisal of included studies.

A feasibility assessment for the ITC was performed and the rationale for exclusion of initially eligible studies in the ITC was provided. The technical report identified several patient characteristics for which there was “strong evidence of effect modification” and the report also suggested that there were limitations regarding reporting of characteristics in trials thereby complicating the assessment of heterogeneity. Based on this and input from 2 clinical experts (who were reported as disagreeing on the degree of the clinical importance of the identified effect modifiers), the technical report states that the ITC was exploratory given the potential limitations related to comparability of the trials. It is not clear what an exploratory ITC is or the intended objective for informing a reimbursement review, especially when the drug of interest—dupilumab—was compared with only 1 relevant comparator for which the included trials indeed appear to be heterogeneous.

||||||||||||||||||| were incorporated in the ITC base case and 1 additional study included in the separate/sensitivity analysis of severe exacerbations. The population of interest was the type 2 inflammatory population; however, data on this population were not available in the omalizumab trials, and an assumption was made that the efficacy of omalizumab would be maintained in the population of interest. It has been acknowledged in previous CADTH reviews and with input from clinicians that severe allergic asthma may also have an eosinophilic component (i.e., there is an overlapping patient population of patients with severe asthma who would be eligible for omalizumab as well as IL inhibitors). Therefore, there is validity to the assumption used in the ITC, but it is unclear how well the subset of patients from the omalizumab studies included in the ITC reflect this overlap population given the omalizumab trials were not designed specifically to include and identify these patients. It could not be determined what impact this had on the indirect estimates between dupilumab and omalizumab.

To improve the similarities between the trials in the ITC, a post hoc subgroup of patients with type 2 inflammatory asthma from the VOYAGE trial who had characteristics that may make them eligible for omalizumab was identified. This subgroup was identified largely based on IgE levels, which is an important marker of allergic asthma. However, it could not be determined how well this subpopulation matched the omalizumab trials population in part because the latter did not report baseline IgE levels. As well, the use of subgroup data resulted in a reduced sample size when compared to the original trial population (i.e., it included approximately 50% of the VOYAGE patient population), limiting the certainty of the results due to smaller evidence base as well as the generalizability of findings relative to the original trial population.

The Bucher indirect method assumes similar relative effectiveness of treatments across all trials, requiring that the included studies involve sufficiently similar populations to be compared. As noted, in this ITC, there were variations related to the study design, patient populations, and outcomes assessed across the studies included. The omalizumab and dupilumab studies varied in study region, length of the run-in periods, and treatment duration. Moreover, the fixed steroid dosing requirement differed among the trials. In contrast to the dupilumab trial, omalizumab trials required fixed steroid dosing for a limited period of 24 weeks, which influenced the outcome assessment in the ITC analysis. Of note, patients included in the omalizumab trials had asthma with an allergic phenotype, while the dupilumab trial did not apply this limitation in the patient population. Moreover, there were differences observed between groups at baseline in the racial distribution, baseline ICS dosing, baseline EOS levels, number of previous exacerbations as well as percent predicated FEV1 values, all of which may be prognostic or affect modifying factors. Additionally, as noted from the VOYAGE trial, patients randomized to placebo also saw improvements from baseline on several outcomes, including asthma control and lung function (i.e., an observable placebo response likely owing to improved administration of standard of care treatments). Therefore, it is not clear whether the placebo link for the ITC was sufficiently similar for making comparisons. Thus, the available data suggest the presence of residual heterogeneity and that key assumption of homogeneity was violated for the ITC analysis. According to the clinical experts consulted by CADTH, the differences in effect modifiers were considered clinically important, impacting the validity of the presented ITC analysis.

The ITC covered many of the relevant clinical outcomes for patients with severe asthma, identified in the CADTH review protocol (asthma exacerbations, HRQoL, symptoms, rescue medication usage, and AEs). However, there were variations in terms of the outcome definitions and outcome estimation time points across trials. To account for differences in severe exacerbation definition, a new outcome, called deterioration of asthma, was identified via post hoc analyses. Clinical experts consulted by CADTH reported that deterioration of asthma as an outcome could be considered comparable across the trials, but also noted that deterioration is already included in the definition of exacerbation and ACQ. Of note, a validated definition for this outcome was not identified. Given the fixed steroid dosing requirement for the omalizumab trials and variations in time points assessed across the studies, the ITC analyses were limited to 24 weeks for the majority of outcomes. Harms data were assessed at 52 weeks, a period which included ICS dose reduction in the omalizumab trial, which limits the interpretability of these findings. Finally, the majority of the analyses included single trial available per comparison in the network, and fixed effects Bucher ITCs were adequately applied to these assessments (except for the sensitivity analysis of severe exacerbations that included 2 omalizumab trials). However, the approach for considering diverse P value cut-offs for ITC effects as comparable, numerically, or statistically favourable in the technical report were not supported by literature and were deemed inappropriate by CADTH reviewers. Also, limited data availability restricted the possibility to perform a meta-regression and account for differences across trials, such as variation in the time points or study and patient characteristics. There were no direct comparisons between treatments, therefore, the assessment of consistency was not feasible.

Due to the limitations described above, the findings of the ITCs can be regarded as highly uncertain, and no conclusions can be drawn on the comparative efficacy of dupilumab compared to omalizumab in patients with uncontrolled moderate-to-severe asthma.

Other Relevant Evidence

This section includes an additional study that could address important gaps in the evidence, such as a short follow-up in the pivotal study.

Long-Term Extension Studies

One sponsor-submitted long-term extension study, EXCURSION, has been summarized to provide evidence regarding dupilumab as add-on therapy to ICSs with or without another controller for the treatment of moderate-to-severe uncontrolled asthma in children aged 6 years to younger than 12 years.20 This report presents the results from the ongoing EXCURSION study as of the data cut-off date of August 18, 2020, and from a second interim analysis with a database lock date of January 17, 2022.42

Methods

The EXCURSION study (N = 365) is a multicentre (involving 17 countries including Canada), open-label, noncomparative study evaluating dupilumab given subcutaneously to pediatric patients aged 6 years to younger than 12 years with asthma who participated in the VOYAGE study. All patients enrolled in EXCURSION received open-label treatment with dupilumab for a period of 52 weeks. After completion of the 1-year treatment period, all patients were to continue into the 12-week posttreatment period. This study was designed to evaluate the long-term safety and tolerability of dupilumab, as well as its long-term efficacy.

Populations

To be eligible for enrolment into the EXCURSION study, pediatric patients with asthma must have completed the parent study, the VOYAGE trial. The placebo–dupilumab group is defined as the patients who were in the placebo group of the parent study and then exposed to dupilumab in the long term study. The dupilumab–dupilumab group is defined as the patients who were in the dupilumab group of the parent study and exposed to dupilumab in the long term study.

The baseline characteristics were reported from the time of enrolment in the parent study, VOYAGE. For a full list of baseline characteristics of the VOYAGE study, refer to Table 7. At week 0 of the EXCURSION study, age, weight, height, body mass index, ICS dose level, serum total IgE, blood eosinophil count, and FEV1 (pre- and postbronchodilator, percent predicted, and reversibility [%]) were re-assessed; however, these are not reported in this section.

Interventions

The dosing regimens in the EXCURSION study are the same as the ones evaluated in the VOYAGE trial.

Patients whose weight changed from 30 kg or less to greater than 30 kg during the study switched their dosing regimen from 100 mg every 2 weeks or 300 mg every 4 weeks to 200 mg every 2 weeks and maintained the same dosing regimen regardless of further weight changes.

All patients in EXCURSION were receiving ICSs with or without a second controller (e.g., LABA, LTRA, LAMA, or methylxanthines) as background maintenance therapy. Also, patients were allowed to use albuterol/salbutamol or levalbuterol/levosalbutamol as a reliever therapy as needed during the study.

All patients were required to remain on stable dosing regimens of their background medications. During deterioration, the ICS dose could be increased or switched to SCS (for a severe exacerbation) temporarily as indicated by the physician and/or the sponsor. When the exacerbation resolved, patients could change back to the original ICS dose or modified ICS dose depending on their control of asthma symptoms.

Outcomes

The primary objective of the EXCURSION study was to assess the long-term safety and tolerability of dupilumab. The efficacy outcome definitions in the EXCURSION study are the same as those in the VOYAGE study.

Statistical Analysis

The results from the EXCURSION study were presented as descriptive summary statistics with observed data only. No model-based imputation for missing data was performed. Also, statistical adjustment including multiplicity adjustment was not conducted. The results were presented by treatment categories according to the actual treatment group in the parent study VOYAGE (i.e., the dupilumab–dupilumab and the placebo–dupilumab groups).

The full analysis set contained all data observed in the study without excluding any data or censoring and was used for the planned analyses. For efficacy outcomes, results are presented by 2 main subgroups, namely patients with type 2 inflammatory asthma phenotype at baseline of the parent study (blood eosinophil ≥ 0.15 Giga/L or FeNO ≥ 20 ppb) and patients with blood eosinophil of 300 cells/µL or greater at baseline of the parent study.

The term “baseline” refers to the baseline of the parent study VOYAGE, while the term “week 0” refers to the beginning of the EXCURSION study (at enrolment, before the first dupilumab administration). The full analysis set includes data collected from all enrolled patients regardless of the dosing schedules they had been assigned to in the parent trial, VOYAGE.

Patient Disposition

Patient disposition in the EXCURSION study as of both interim analyses is summarized in Table 33. A total of 365 patients (ITT population from the VOYAGE study, ||||||||||||||||||| from dupilumab group, ||||||||||||||||||| from placebo group) were rolled over from the parent study and received open-label dupilumab for an additional 52 weeks in the EXCURSION study. Of |||||||||||||||||||||||||||||||||||||| previously received dupilumab and ||||||||||||||||||| previously received placebo in the VOYAGE study.

At the cut-off date of August 18, 2020, |||||||||||| of the dupilumab–dupilumab group and |||||||||||| of the placebo–dupilumab group completed the study treatment, respectively. It was noted that more patients in the dupilumab–dupilumab group discontinued treatment compared to the placebo–dupilumab group (||||||||||||) due to AEs, poor treatment adherence, voluntary withdrawal, and other reasons A total of |||||| additional patients (|||||| in dupilumab–dupilumab group and |||||| in placebo–dupilumab group) discontinued study after completion of treatment period due to AEs, poor treatment adherence, or other reasons (|||||| in dupilumab–dupilumab group versus |||||| in placebo–dupilumab group).

As of the database lock date of January 17, 2022, almost all patients either completed treatment (95% in the dupilumab–dupilumab group and 97.6% in the placebo–dupilumab group) or the study period (95% and 94.4%, respectively). Compared to the previous data cut-off date, 1 more patient discontinued treatment due to an AE in the dupilumab–dupilumab group and 1 more patient discontinued study for other reason in the placebo–dupilumab group. Overall, 12 (5.0%) and 3 (2.4%) patients prematurely discontinued treatment in dupilumab–dupilumab and placebo–dupilumab groups, respectively. The number (%) of patients who did not complete the end of study visit were 12 (5%) and 7 (5.6%) in the dupilumab–dupilumab and placebo–dupilumab arms, respectively (Table 32).

Table 33. Patient Disposition (EXCURSION) .

Table 33

Patient Disposition (EXCURSION) .

Exposure to Study Treatments

Exposure to study treatments is summarized in ||||||||||||||||||||||||.

At the cut-off date of August 18, 2020, the median duration of the study was 372 days (range = 1 to 488) and 375 days (range = 2 to 518 days) for the dupilumab–dupilumab and placebo–dupilumab groups, respectively. The total cumulative study duration was 217 patient-years and 116 patient-years for the dupilumab–dupilumab and placebo–dupilumab groups, respectively, reflecting a 2:1 randomization scheme from the parent trial.

As of the January 17, 2022, data cut-off date, the median duration of study was |||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||| for the dupilumab–dupilumab and placebo–dupilumab groups, respectively. The cumulative study duration |||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||| for the dupilumab–dupilumab and placebo–dupilumab groups, respectively. All enrolled patients received ICSs with or without a second controller in addition to dupilumab. In general, all patients had high treatment adherence with nearly everyone having achieved equal or more than |||||||||||||||||||||||||||||||||||||||||||||||| in the dupilumab–dupilumab group and placebo–dupilumab groups, respectively, in the August 18, 2020, dataset; |||||||||||||||||||||||||||||||||||||||||||||||| respectively, in the January 17, 2022, dataset).

Table 34. Redacted.

Table 34

Redacted.

Efficacy

All efficacy end points were secondary end points in the EXCURSION study.

Severe Asthma Exacerbation

Exacerbation data are summarized in ||||||||||||||||||||||||.

When analyzed as a subgroup, patients with type 2 inflammatory asthma phenotype at baseline of the parent study |||||||||||||||||||||||| experienced an event in the dupilumab–dupilumab and placebo–dupilumab groups, respectively. When analyzed as a subgroup, patients with EOS of 300 cells/µL or greater at baseline of the parent study |||||||||||||||||||||||| experienced an event in the dupilumab–dupilumab and placebo–dupilumab groups, respectively. The unadjusted annualized rate of severe exacerbation was |||||||||||||||||||||||| for the dupilumab–dupilumab and placebo–dupilumab groups, respectively |||||||||||||||||||||||| in a type 2 inflammatory asthma phenotype subgroup; |||||||||||||||||||||||| in an eosinophil ≥ 300 cells/µL subgroup).

As of the January 17, 2022, data cut-off, for both the type 2 inflammatory asthma and EOS of 300 cells/µL or greater at baseline subgroups, there was |||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||| whereas the numbers |||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||| compared to the previous dataset. Compared to the previous dataset, in a type 2 inflammatory asthma phenotype subgroup, the unadjusted annualized severe exacerbation event rate decreased from ||||||||||||||| to 0.118 and from ||||||||||||| to 0.124 for the dupilumab–dupilumab and placebo–dupilumab groups, respectively. Similarly, in the subgroup with eosinophil of 300 cells/µL or greater, the unadjusted annualized severe exacerbation event rate decreased from |||||||||||| to 0.120 and from |||||||||||| to 0.119, for the dupilumab–dupilumab and placebo–dupilumab groups, respectively. There were |||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||| who experienced a severe exacerbation requiring hospitalization or ED visit during the treatment period.

Table 35. Redacted.

Table 35

Redacted.

Change in Percent Predicted FEV1

Data for change in the percent predicted FEV1 are presented in Table 35.

As of the August 18, 2022, data cut-off, at week 52, improvement in percent predicted prebronchodilator FEV1 was |||||||||||||||||||||||||||||||||||||||||||||||||| for the dupilumab–dupilumab and placebo–dupilumab groups, respectively, in the type 2 inflammatory asthma subgroup. In the same dataset, at week 52, improvement in percent predicted prebronchodilator FEV1 was ||||||||||||||||||||||||| and ||||||||||||||||||||||||| for the dupilumab–dupilumab and placebo–dupilumab groups, respectively, in the baseline blood EOS of 300 cells/µL or greater subgroup. Of note, the total number of patients contributing to the analysis reduced to almost half (n = 111 versus n = 58 for the dupilumab–dupilumab and placebo–dupilumab groups, respectively) compared to the number of enrolled patients (n = 240 versus n = 125) at week 52 in data from the August 18, 2020, cut-off .

The second interim analysis based on the updated dataset (as of January 17, 2022) included more patients (n = ||||||||||||||||||||||||| for the dupilumab–dupilumab and placebo–dupilumab groups, respectively) than previous analysis. Additional data showed sustained lung function improvement in the type 2 inflammatory asthma subgroup as shown by the FEV1 increase from baseline by |||||||||||||||||||||||||||||||||||||||||||||||||| in the dupilumab–dupilumab and placebo–dupilumab groups, respectively, at week 52. Similarly, in subgroup with baseline blood EOS of 300 cells/µL or greater, improvement in FEV1 was sustained at week 52 as shown by increase of |||||||||||||||||||||||||||||||||||||||||||||||||| in the dupilumab–dupilumab and placebo–dupilumab groups, respectively.

Table 36. Change From Baseline in Percent Predicted Prebronchodilator FEV1 Over Time (EXCURSION) .

Table 36

Change From Baseline in Percent Predicted Prebronchodilator FEV1 Over Time (EXCURSION) .

Harms

Only those harms identified in the review protocol outlined in Table 5 are reported below. Refer to Table 36 for detailed harms data.

Adverse Events

In the initial analysis with data from the August 18, 2020, cut-off, the most frequently reported (≥ 5%) treatment-emergent AEs were nasopharyngitis (7.5% and 8.8% for the dupilumab–dupilumab and placebo–dupilumab groups, respectively), pharyngitis (5.0% and 8.8% for the dupilumab–dupilumab and placebo–dupilumab groups, respectively), and upper respiratory tract infection (6.7% and 3.2%, for the dupilumab–dupilumab and placebo–dupilumab groups, respectively). As of the January 17, 2022, data cut-off, the number of treatment-emergent AEs reported slightly increased for both groups (8.8% and 9.6% for nasopharyngitis, 6.3% and 9.6% for pharyngitis, and 7.9% and 4.0% for upper respiratory tract infections for the dupilumab–dupilumab and placebo–dupilumab groups, respectively).

Serious Adverse Events

As of the data cut-off date of August 18, 2020, there were 4 (1.7%) and 1 (0.8%) patient(s) from the dupilumab–dupilumab and placebo–dupilumab groups, respectively, who experienced SAEs during the EXCURSION study period. ||||||||||||||||||||||||||||||||||||||||||||||||||. As of the January 17, 2022, data cut-off, 2 more cases of SAEs were reported in the dupilumab–dupilumab group (for a total of 6 patients or 2.5%); whereas the number remained the same for the placebo–dupilumab group compared to the previous data cut-off.

Withdrawals Due to AEs

Based on data from the cut-off date of August 18, 2020, there were 2 (0.8%) patients in the dupilumab–dupilumab group who discontinued treatment due to AEs (pulmonary tuberculosis and allergic conjunctivitis). As of January 17, 2022, 1 more patient discontinued treatment due to ascariasis and a total of 3 (1.3%) patients discontinued treatment. No patient in the placebo–dupilumab group discontinued treatment due to an AE.

Mortality

No deaths were reported during the EXCURSION study period.

Notable Harms

As of the August 18, 2020, data cut-off, a total of |||||||||||||||||||||||||||||||||||||||||||||||||| patient(s) in the dupilumab–dupilumab and placebo–dupilumab groups, respectively, experienced hypersensitivity (medically reviewed) reactions with ||||||||||||||||||||||||| patients in the dupilumab–dupilumab group and none in the placebo–dupilumab group experiencing anaphylactic reaction. According to the study report, |||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||| in intensity and none led to treatment interruption or discontinuation.

As of the August 18, 2020, data cut-off, severe or serious infection was reported in |||||||||||| patients in the dupilumab–dupilumab group (complicated appendicitis, pulmonary tuberculosis, and upper respiratory tract infection) and |||||||||||| patient in the placebo–dupilumab group (pneumonia). One (0.4%) patient from the dupilumab–dupilumab group and none in the placebo–dupilumab group experienced an opportunistic infection (pulmonary tuberculosis). |||||||||||||||||||||||||||||||||||| patient(s) in the dupilumab–dupilumab and placebo–dupilumab groups, respectively, reported to have had parasitic infections during the study period.

As of the August 18, 2020, data cut-off, injection site reaction was reported more frequently in the placebo–dupilumab group than the dupilumab–dupilumab group (||||||||||||||||||||||||).

As of the August 18, 2020, data cut-off, conjunctivitis occurred in |||||||||||||||||||||||||||||||||||| of patients in the dupilumab–dupilumab and placebo–dupilumab groups, respectively. Based on the Clinical Study Report, ||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||||

Lastly, eosinophilia was reported in |||||||||||||||||||||||||||||||||||| patients in the dupilumab–dupilumab and placebo–dupilumab groups, respectively as of the August 18, 2020, data cut-off. According to the study report, all cases of eosinophilia were asymptomatic and a transient increase in blood eosinophil counts that showed a gradual decrease over time. For some eosinophilia episodes, corrective treatment was administered.

As of January 17, 2022, the number of patients who experienced hypersensitivity (medically reviewed), parasitic infections, injection site reaction, conjunctivitis (broad), and eosinophilia increased by 1 to 3 patients per category from those of the earlier cut-off date, August 18, 2020. For a detailed description, refer to Table 37.

Table 37. Summary of Harms–FAS (EXCURSION) .

Table 37

Summary of Harms–FAS (EXCURSION) .

Critical Appraisal
Internal Validity

The EXCURSION study was an open-label, noncomparative, longer-term extension study that evaluated the safety of dupilumab as a primary objective. A lack of blinding could have had impact on patient report of AEs. However, most AEs reported in the EXCURSION study could be assessed by signs, thereby reducing the potential biases. Furthermore, as the EXCURSION study is a 1-year study, rare AEs could not be captured as of the data cut-off date. Lastly, as efficacy outcomes were secondary objectives and given this is a noncomparative trial, no definitive conclusion about long-term comparative efficacy of dupilumab as add-on therapy to standard of care can be drawn based on the data from the EXCURSION study. Of note, 300 mg dosing of dupilumab was not assessed in the parent study, VOYAGE, which represents an additional limitation for the assessment of safety and efficacy of treatment of this dosage regimen.

External Validity

As patients were rolled over from the VOYAGE study, the same generalizability issues and limitations hold for the EXCURSION study. In the EXCURSION study, although most dupilumab injections were administered by a health care provider, home administration of 300 mg every 4 week dosing following first administration onsite was allowed with a protocol amendment (n = 18). The home administration of dupilumab was not regarded as a generalizability concern according to the clinical experts consulted, as this approach may be implemented in the Canadian real-world practice setting.

Discussion

Summary of Available Evidence

Dupilumab has been resubmitted to CADTH to expand the reimbursement for severe asthma to include patients aged 6 years to younger than 12 years. The sponsor provided 1 multinational, multicentre, double-blind RCT, VOYAGE, that was included in the systematic review section. The VOYAGE trial was conducted in 408 patients aged 6 years to younger than 12 years with persistent uncontrolled asthma, despite standard treatment use. The VOYAGE study compared dupilumab to placebo (both added to standard of care) administered over 52 weeks. The primary outcome was annualized rate of severe exacerbations. There were 2 efficacy populations in the trial that informed the reimbursement request: the type 2 inflammatory asthma phenotype population, characterized by a baseline blood eosinophil count of 150 cells/µL or greater or baseline FeNO of 20 ppb or greater, and a population with baseline blood EOS of 300 cells/µL or greater.

Additional evidence was available from an ITC submitted by the sponsor, comparing the efficacy of dupilumab to another biologic treatment (the IgE inhibitor omalizumab), and a longer-term extension study, EXCURSION (N = 365), evaluating the safety and tolerability as well as efficacy of dupilumab for an additional 52 weeks of treatment after the VOYAGE trial.

Interpretation of Results

Efficacy

Dupilumab was previously reviewed by CADTH for the indication of severe asthma with a type 2 or eosinophilic phenotype or OCS-dependent asthma in patients aged 12 years and older and received a positive recommendation in June 2021. On March 25, 2022, the Health Canada indication was revised and now dupilumab has a Health Canada indication for add-on maintenance treatment in patients aged 6 years and older with severe asthma with a type 2 or eosinophilic phenotype or OCS-dependent asthma. It is important to consider that the VOYAGE trial did not include patients who were OCS-dependant, therefore the treatment effect in OCS-dependant individuals aged 6 years to younger than 12 years remains unclear. However, the clinical experts consulted by CADTH for this review noted that very few pediatric patients undergo daily or alternate daily OCS treatment, but many may experience frequent short courses of treatment with OCSs.

Efficacy in the VOYAGE trial was assessed in 2 main populations, defined as the type 2 inflammatory asthma phenotype population (baseline blood eosinophil count ≥ 150 cells/µL or baseline FeNO ≥ 20 ppb) and a population with a baseline blood eosinophil count of 300 cells/µL or greater, due to differences in approved label indications across the US and European Union reference countries. Additional efficacy populations that were multiplicity controlled were reported, including ITT, baseline blood eosinophil count of 150 cells/µL or greater, and baseline FeNO of 20 ppb or greater. Certain implementation issues were highlighted by the clinical experts, mainly the applicability of utilizing FeNO measurements to identify eligible patients in the Canadian settings. Even though this restriction was suggested by the sponsor in their reimbursement request, the experts reported that FeNO assessments are not routinely performed in clinical practice in Canada.

Both clinical experts and patient groups consulted during the CADTH review highlighted that reducing asthma exacerbations is considered an important treatment goal for patients with asthma. In the VOYAGE trial, the number of severe asthma exacerbations over 52 weeks was lower in the dupilumab group versus placebo. The observed between-group effects were both statistically significant and clinically meaningful, favouring patients randomized to the dupilumab group compared to placebo. Clinical experts noted that the effects of asthma treatments may be driven by external and environmental factors that can vary within 1 year and even between years, such as viral infection, air pollution, and allergen exposure. Hence, treatment effects in asthma studies with limited time intervals may be biased by these factors, and longer-term studies are needed for assessment of effectiveness. The VOYAGE trial assessed exacerbation rates at 52 weeks which is appropriate given the aforementioned variation in asthma over time; however, asthma is a chronic condition and longer follow-up periods are needed to be able to extrapolate the benefits in the longer-term. The sponsor provided additional evidence from an interim analysis of a longer-term extension study (EXCURSION), which followed the VOYAGE trial population for up to an additional year after the parent study. Even though the EXCURSION study showed continued low severe asthma exacerbation event rates, these findings are limited by the lack of formal hypothesis testing in this open-label, noncomparative trial.

The CADTH systematic review protocol prespecified acute asthma exacerbations leading to hospitalization and/or to ED visits as outcomes of interest for this review. There was a limited number of these events experienced by the patients during the VOYAGE trial, thus limiting the interpretability of the observed results. Moreover, these analyses were introduced after the database lock occurred in the trial, suggesting that no conclusions can be made on the efficacy of dupilumab in preventing hospitalizations and ED visits.

Change from baseline in prebronchodilator percent predicted FEV1 at week 12 was the key secondary outcome included in the statistical testing hierarchy of the VOYAGE trial. Despite being the key secondary efficacy outcome in the pivotal study, prebronchodilator percent predicted FEV1 was rated with a lower ranking of importance by the clinical experts, as listed in the CADTH protocol, and was labelled as a secondary goal of asthma management, according to the clinical group input. However, this outcome was regarded as important by the patient groups as more than two-thirds of respondents from the patient survey submitted to CADTH reported an increase in lung function to be an important expectation for new asthma medication. At week 12, the differences in mean change from baseline for the key secondary end point of percent predicted prebronchodilator FEV1 were statistically significant. The clinical experts reported that the magnitude of change from baseline between groups at week 12 in percent predicted prebronchodilator FEV1 was modest yet acceptable. Furthermore, the experts reported that the 12-week assessment of FEV1 was not optimal as this duration might be insufficient to evaluate seasonal and other factors affecting the outcome; hence, the experts placed more clinical relevance on the 52-week assessment. While no definitive conclusions can be drawn because multiplicity considerations were not accounted for, the results of pulmonary function analyses across the other time points are supportive of a treatment benefit with dupilumab versus placebo.

Subgroup analyses was performed for the primary and key secondary outcome of the VOYAGE trial, based on the following factors of interest for this CADTH review: baseline eosinophil levels, baseline ICS dose, number of previous asthma exacerbations, and atopic medical history. Subgroups of patients with baseline blood EOS of 300 cells/µL or greater, baseline blood EOS of greater than 150 cells/µL, and patients receiving high doses of ICSs were part of the hierarchical testing procedure and, thus, adjusted for multiplicity. The findings from the baseline eosinophil count subgroup analyses are indicative of greater treatment benefit, in terms of annualized exacerbation rates, among patients with higher eosinophil counts. This is aligned with the input received from clinical experts, who expected an almost linear relationship to be observed between baseline blood eosinophil count and exacerbation frequency. It is also supportive of the dupilumab-indicated patient population as well as suggested reimbursement criteria to use dupilumab in patients with baseline EOS of 150 cells/µL or greater, a requirement that is also consistent with the GINA definition of type 2 inflammatory asthma.1 Still, clinical experts expressed some implementation concerns and reported that different provinces across Canada may report lab values with diverse acuity (i.e., some labs only report round numbers of 100 cells/μL, 200 cells/μL, 300 cells/μL, and so forth). In terms of the analyses for other subgroups, findings were considered only supportive of the overall treatment effect with dupilumab by CADTH as there were no multiplicity adjustments performed for these analyses.

Reduction in asthma symptoms represents an important outcome to patients and clinicians. In the VOYAGE trial, asthma symptoms were captured through a validated questionnaire (ACQ-7), with an established MID of 0.5, and the analyses at the week 24 time point were incorporated in the hierarchical testing procedures. Mean changes from baseline in the ACQ-7 in both groups exceeded the MID of 0.5 points; however, differences in ACQ scores between dupilumab and placebo were less than 0.5 at week 24. Responder analyses showed that approximately 80% of patients receiving dupilumab patients were considered responders, exceeding the established MID, across both the type 2 and baseline eosinophil of 300 cells/µL or greater populations. However, it should be noted that slightly more than 60% of the patients in the placebo group were also considered responders. Despite the lack of multiplicity adjustments for the responder analyses, findings can be considered supportive of improved asthma control with dupilumab treatment compared to placebo. According to the clinical experts, robust placebo responses observed in the trial indicate that getting patients who have uncontrolled asthma to adhere to their background medication is very important when treating asthma. Indeed, adherence to background therapy was high in both groups during the VOYAGE study (approximately 80%), which suggests that patients might have benefited from the close attention and monitoring of the trial. As a result, clinicians stressed that patients who are well managed should not be eligible for dupilumab treatment and highlighted the importance of going through an asthma clinic or subspecialist or specialist care when initiating treatment with dupilumab.

Asthma has a substantial impact on HRQoL, according to the patient input received by CADTH. HRQoL, assessed through the validated, disease-specific PAQLQ instrument among children aged 7 years to younger than 12 years, was a secondary outcome of the VOYAGE trial. The within-group mean changes from baseline exceeded the MID of 0.5 regardless of treatment group or subpopulation; however, the differences in PAQLQ global scores between dupilumab and placebo were less than 0.5. Other HRQoL measures (PACQLQ, EQ-5D-Y, and PRQLQ) were prespecified as exploratory outcomes in the VOYAGE study. The clinical experts consulted agreed with the CADTH reviewers that the clinical significance of the comparative estimates of effect on HRQoL are uncertain. As well, the HRQoL analyses presented in the trial were without adjustments for multiplicity; thus, the results presented were considered supportive by CADTH, yet it remains unclear what the impact of add-on dupilumab on patients’ HRQoL is, compared to placebo.

Additional important treatment goals of asthma, according to the clinical experts and the patients, included reducing OCS usage and limiting complications of the existing maximal therapy. The VOYAGE study findings showed that more than 40% of patients in the placebo arm and more than 20% of patients in the dupilumab arm received at least 1 course of SCSs. The decreases in SCS burden observed between the 2 groups were regarded as clinically significant, with possible impact on linear growth in patients, according to the clinical experts. Regarding the ICS usage, permanent increases in dosing were captured during the trial only after 2 or more severe asthma exacerbations, as specified in the study protocol. Even though none of the patients had their background medications increased during trial, clinical experts reported assessment concerns and noted that there should have been earlier escalation of therapy or at least reassessment for need for escalation before the 2 events occurring. In addition to these concerns, it is important to note that OCS usage was reported under additional efficacy analyses and the ICS was considered an exploratory outcome in the pivotal trial. As such, they were absent from the statistical testing hierarchy. This resulted in an inability to draw conclusions about the efficacy of dupilumab to decrease the OCS use and reduce the burden of existing ICS therapy; thus, an evidence gap remains regarding treatment effect on these outcomes of importance in the clinical setting.

Currently, there are no head-to-head trials that have evaluated the efficacy of dupilumab with relevant biologics of interest for patients with severe uncontrolled asthma. The sponsor submitted a Bucher ITC analysis that compared the efficacy of dupilumab to omalizumab in patients who are aged 6 years to 11 years of age with uncontrolled moderate-to-severe asthma. Limitations of the ITC include the heterogeneity of included studies in terms of study characteristics, patient populations, and outcomes assessed. The exploration of between study differences and potential biases was further limited by the small number of trials included in the ITC. Considering that prognostic factors and effect modifiers are likely imbalanced between treatment groups, the results of the ITC are subject to an unknown amount and direction of bias. Thus, the findings of the ITC are highly uncertain. According to the clinical experts, presented data are limited and the results do not support superiority of dupilumab, compared to omalizumab, in patients aged 6 years to younger than 12 years with severe uncontrolled asthma. Evidence on comparative efficacy of dupilumab to the other Health Canada–approved biologic drug for patients 6 and older (mepolizumab) was not identified.

Harms

Dupilumab appeared to have an acceptable safety profile, based on the small number of patients who discontinued therapy due to an AE in the VOYAGE study. The proportions of patients experiencing SAEs during the VOYAGE trial were also balanced between the dupilumab and placebo arms. In terms of notable harms, there seemed to be no indication of an increased risk of hypersensitivity reactions with dupilumab versus placebo. Even though parasitic infections are common with monoclonal antibodies, due to the creation of an immunosuppressive environment that decreases the host’s resistance to infections,5 only 2.6% of dupilumab patients had a parasitic infection, compared to none experienced in the placebo group. The incidence of conjunctivitis was low across both studied groups, which was unexpected according to the clinical experts. Clinical experts reported that the higher incidence of injection site reactions and eosinophilia reported in the dupilumab compared to the placebo group are aligned with the data from the adult and adolescent population.

Longer-term follow-up data from the EXCURSION study did not reveal any new safety issues from those observed in the parent trial VOYAGE; however, these findings are limited considering the open-label and noncomparative design.

The sponsor-submitted ITC reported on safety outcomes, mainly withdrawals due to AEs. There were no significant differences observed between dupilumab and omalizumab at 52 weeks, but the interpretation of these findings is limited due to numerous methodological limitations as well as the fact that the period of assessment for harm outcomes included an ICS dose reduction period from the omalizumab trial. As a result, no definitive conclusions can be drawn regarding the comparative safety of dupilumab relative to omalizumab.

Conclusions

One sponsor-submitted, multicentre, randomized, double-blind, phase III trial (VOYAGE), comparing add-on therapy with dupilumab to placebo in patients aged 6 years to younger than 12 years with persistent asthma demonstrated that dupilumab reduced the annualized rate of severe exacerbations and improved pulmonary function (FEV1) in patients whose asthma remains uncontrolled despite background therapy with medium to high doses of ICSs. There was supportive evidence on the overall treatment benefit on asthma-related symptoms, as measured by ACQ-7, but the differences between the dupilumab and placebo groups did not exceed the MID. HRQoL analyses were not controlled for multiple comparisons in this RCT; thus, the impact of add-on dupilumab on patients’ HRQoL compared to placebo remains. Likewise, observed reductions in OCS usage were not part of the statistical hierarchy, which precluded drawing conclusions about the effects of dupilumab on this outcome. With respect to harms, there were no obvious safety or tolerability issues associated with the use of dupilumab in children. A longer-term extension study did not identify any new safety issues. Findings from the sponsor-submitted ITC were inconclusive with respect to the efficacy and safety of dupilumab compared to omalizumab due to numerous methodological limitations.

Abbreviations

ACQ

Asthma Control Questionnaire

ACQ-5

5-item Asthma Control Questionnaire

ACQ-7

7-item Asthma Control Questionnaire

ACQ-5-IA

5-item Asthma Control Questionnaire–Interviewer Administered

ACQ-7-IA

7-item Asthma Control Questionnaire–Interviewer Administered

AE

adverse event

CI

confidence interval

ED

emergency department

EOS

eosinophils

EQ-5D-Y

EQ-5D-Youth

FeNO

fractional exhaled nitric oxide

FEV1

forced expiratory volume in 1 second

GINA

Global Initiative for Asthma

HRQoL

health-related quality of life

ICS

inhaled corticosteroid

IgE

immunoglobulin E

IL

interleukin

ITC

indirect treatment comparison

ITT

intention to treat

LABA

long-acting beta agonist

LAMA

long-acting muscarinic antagonist

LS

least squares

LTRA

leukotriene receptor antagonist

MID

minimal important difference

MMRM

mixed-effect model with repeated measures

OCS

oral corticosteroid

OR

odds ratio

PACQLQ

Paediatric Asthma Caregiver’s Quality of Life Questionnaire

PAQLQ(S)

Standardised Paediatric Asthma Quality of Life Questionnaire

PEF

peak expiratory flow

Ppb

parts per billion

PRQLQ

Pediatric Rhinoconjunctivitis Quality of Life Questionnaire

RCT

randomized controlled trial

RR

relative risk

SABA

short-acting beta agonist

SAE

serious adverse events

SCS

systemic corticosteroid

SD

standard deviation

SE

standard error

VAS

visual analogue scale

Appendix 1. Literature Search Strategy

Clinical Literature Search

Overview

Interface: Ovid

Databases:

  • MEDLINE All (1946-present)
  • Embase (1974-present)
  • Note: Subject headings and search fields have been customized for each database. Duplicates between databases were removed in Ovid.

Date of search: July 27, 2022

Alerts: Bi-weekly search updates until project completion

Search filters applied: Randomized controlled trials or controlled clinical trials.

Limits:

  • Publication date limit: none
  • Language limit: none
  • Conference abstracts: excluded.
Table 38. Syntax Guide.

Table 38

Syntax Guide.

Multidatabase Strategy

  1. (dupixent* or dupilumab* or SAR-231893 or SAR231893 or REGN-668 or REGN668 or 420K487FSG).ti,ab,kf,ot,hw,nm,rn.
  2. exp Asthma/ or Bronchial Spasm/
  3. (asthma* or antiasthma* or wheez*).ti,ab,kf.
  4. (bronchospas* or bronchiospas* or (bronch* adj2 spas*)).ti,ab,kf.
  5. (lung adj5 allerg*).ti,ab,kf.
  6. or/2-5
  7. 1 and 6
  8. 7 use medall
  9. *dupilumab/
  10. (dupixent* or dupilumab* or SAR-231893 or SAR231893 or REGN-668 or REGN668).ti,ab,kf,dq.
  11. or/9-10
  12. exp Asthma/ or exp bronchospasm/
  13. (asthma* or antiasthma* or wheez*).ti,ab,kf,dq.
  14. (bronchospas* or bronchiospas* or (bronch* adj2 spas*)).ti,ab,kf,dq.
  15. (lung adj5 allerg*).ti,ab,kf,dq.
  16. or/12-15
  17. 11 and 16
  18. 17 use oemezd
  19. 18 not (conference abstract or conference review).pt.
  20. 8 or 19
  21. (Randomized Controlled Trial or Controlled Clinical Trial or Pragmatic Clinical Trial or Equivalence Trial or Clinical Trial, Phase III).pt.
  22. Randomized Controlled Trial/
  23. exp Randomized Controlled Trials as Topic/
  24. "Randomized Controlled Trial (topic)"/
  25. Controlled Clinical Trial/
  26. exp Controlled Clinical Trials as Topic/
  27. "Controlled Clinical Trial (topic)"/
  28. Randomization/
  29. Random Allocation/
  30. Double-Blind Method/
  31. Double Blind Procedure/
  32. Double-Blind Studies/
  33. Single-Blind Method/
  34. Single Blind Procedure/
  35. Single-Blind Studies/
  36. Placebos/
  37. Placebo/
  38. Control Groups/
  39. Control Group/
  40. (random* or sham or placebo*).ti,ab,hw,kf.
  41. ((singl* or doubl*) adj (blind* or dumm* or mask*)).ti,ab,hw,kf.
  42. ((tripl* or trebl*) adj (blind* or dumm* or mask*)).ti,ab,hw,kf.
  43. (control* adj3 (study or studies or trial* or group*)).ti,ab,kf.
  44. (Nonrandom* or non random* or non-random* or quasi-random* or quasirandom*).ti,ab,hw,kf.
  45. allocated.ti,ab,hw.
  46. ((open label or open-label) adj5 (study or studies or trial*)).ti,ab,hw,kf.
  47. ((equivalence or superiority or non-inferiority or noninferiority) adj3 (study or studies or trial*)).ti,ab,hw,kf.
  48. (pragmatic study or pragmatic studies).ti,ab,hw,kf.
  49. ((pragmatic or practical) adj3 trial*).ti,ab,hw,kf.
  50. ((quasiexperimental or quasi-experimental) adj3 (study or studies or trial*)).ti,ab,hw,kf.
  51. (phase adj3 (III or "3") adj3 (study or studies or trial*)).ti,hw,kf.
  52. or/21-51
  53. 20 and 52
  54. remove duplicates from 53

Clinical Trials Registries

ClinicalTrials.gov

Produced by the US National Library of Medicine. Targeted search used to capture registered clinical trials.

[Search -- Studies with results | dupixent OR dupilumab OR SAR-231893 OR SAR231893 OR REGN-668 OR REGN668 | asthma]

WHO ICTRP

International Clinical Trials Registry Platform, produced by the World Health Organization. Targeted search used to capture registered clinical trials.

[Search terms -- (dupixent OR dupilumab OR SAR-231893 OR SAR231893 OR REGN-668 OR REGN668) AND asthma]

Health Canada’s Clinical Trials Database

Produced by Health Canada. Targeted search used to capture registered clinical trials.

[Search terms -- (dupixent OR dupilumab) AND asthma]

EU Clinical Trials Register

European Union Clinical Trials Register, produced by the European Union. Targeted search used to capture registered clinical trials.

[Search terms -- (dupixent OR dupilumab OR SAR-231893 OR SAR231893 OR REGN-668 OR REGN668) AND asthma]

Grey Literature

Search dates: July 14, 2022 – July 20, 2022

Keywords: [dupixent OR dupilumab OR SAR-231893 OR SAR231893 OR REGN-668 OR REGN668 | asthma]

Limits: Publication years: 2017-present for guidelines; none for other sections

Relevant websites from the following sections of the CADTH grey literature checklist Grey Matters: A Practical Tool for Searching Health-Related Grey Literature were searched:

  • Health Technology Assessment Agencies
  • Health Economics
  • Clinical Practice Guidelines
  • Drug and Device Regulatory Approvals
  • Advisories and Warnings
  • Drug Class Reviews
  • Clinical Trials Registries
  • Databases (free)
  • Internet Search.

Appendix 2. Excluded Studies

Note that this appendix of tables has not been copy-edited.

Table 39. Excluded Studies.

Table 39

Excluded Studies.

Appendix 3. Detailed Outcome Data

Note that this appendix of tables has not been copy-edited.

Table 40. Hierarchical Testing Order for US and US Reference Countries in the VOYAGE Trial.

Table 40

Hierarchical Testing Order for US and US Reference Countries in the VOYAGE Trial.

Table 41. Hierarchical Testing Order for EU and EU Reference Countries in the VOYAGE Trial.

Table 41

Hierarchical Testing Order for EU and EU Reference Countries in the VOYAGE Trial.

Table 42. Patient Disposition (ITT and Baseline Blood Eosinophils ≥ 150 cells/µL Populations) .

Table 42

Patient Disposition (ITT and Baseline Blood Eosinophils ≥ 150 cells/µL Populations) .

Table 43. Summary of Sensitivity Analyses for Adjusted Annualized Severe Exacerbation Event Rate (Type 2 Inflammatory Asthma Phenotype and Baseline Blood Eosinophils ≥ 300 cells/µL Populations) .

Table 43

Summary of Sensitivity Analyses for Adjusted Annualized Severe Exacerbation Event Rate (Type 2 Inflammatory Asthma Phenotype and Baseline Blood Eosinophils ≥ 300 cells/µL Populations) .

Table 44. Severe Asthma Exacerbations (ITT and Baseline Blood Eosinophils ≥ 150 cells/µL Populations) .

Table 44

Severe Asthma Exacerbations (ITT and Baseline Blood Eosinophils ≥ 150 cells/µL Populations) .

Table 45. Symptoms ACQ-7 (Weeks 24 and 52; ITT and Baseline Blood Eosinophils ≥ 150 cells/µL Populations) .

Table 45

Symptoms ACQ-7 (Weeks 24 and 52; ITT and Baseline Blood Eosinophils ≥ 150 cells/µL Populations) .

Table 46. Symptoms ACQ-5 (Type 2 Inflammatory Asthma Phenotype Population and Baseline Blood Eosinophils ≥ 300 cells/µL Populations) .

Table 46

Symptoms ACQ-5 (Type 2 Inflammatory Asthma Phenotype Population and Baseline Blood Eosinophils ≥ 300 cells/µL Populations) .

This figure has been redacted.

Figure 8

Redacted.

This figure has been redacted.

Figure 9

Redacted.

Table 47. Pulmonary Function Measurements (Percent Predicted Prebronchodilator FEV1; ITT and Baseline Blood Eosinophils ≥ 150 cells/µL Populations) .

Table 47

Pulmonary Function Measurements (Percent Predicted Prebronchodilator FEV1; ITT and Baseline Blood Eosinophils ≥ 150 cells/µL Populations) .

Table 48. Summary of Sensitivity Analyses for Change From Baseline in Prebronchodilator % Predicted FEV1 at Week 12 (Type 2 Inflammatory Asthma Phenotype and Baseline Blood Eosinophils ≥ 300 cells/µL Populations) .

Table 48

Summary of Sensitivity Analyses for Change From Baseline in Prebronchodilator % Predicted FEV1 at Week 12 (Type 2 Inflammatory Asthma Phenotype and Baseline Blood Eosinophils ≥ 300 cells/µL Populations) .

Table 49. Redacted.

Table 49

Redacted.

Appendix 4. Description and Appraisal of Outcome Measures

Note that this appendix has not been copy-edited.

Aim

To describe the following patient-reported outcome measures and review their measurement properties (validity, reliability, responsiveness to change, and MID):

  • ACQ-IA (ACQ-7 and ACQ-5) for children aged 6 to less than 12 years
  • PAQLQ(S)-IA for patients older than 7 and younger than 12 years
  • PRQLQ-IA for children aged 6 to less than 12 years, with history of allergic rhinitis
  • PACQLQ for caregivers of patients older than 7 years
  • EQ-5D-Y

Findings

Table 50. Summary of Outcome Measures and Their Measurement Properties.

Table 50

Summary of Outcome Measures and Their Measurement Properties.

Asthma Control Questionnaire

The complete ACQ, also termed the ACQ-7, was developed to evaluate asthma control in adult patients with asthma.22 The ACQ is 1 of the most commonly used instruments measuring asthma control.22,23 The questionnaire comprises 7 questions, the responses of which are scored on a 7-point scale (0 = no impairment; 6 = maximum impairment).24 Questions regarding 6 aspects of the patient’s previous week’s experiences are answered by the patient and include questions on activity limitation, nocturnal waking, shortness of breath, wheezing, symptoms on waking, and the average number of daily doses of short-acting beta2-agonist used.23 In addition, the seventh item includes calculations performed by clinical staff with regard to prebronchodilator FEV1% predicted on a similar 7-point scale.22,23 The ACQ score is calculated as the mean of the 7 questions (as all questions are equally weighted), with scores ranging from 0 (well-controlled) to 6 (extremely poorly uncontrolled).22,23,51 The ACQ is used extensively in clinical trials to measure clinically meaningful change in asthma control.22

The ACQ also exists in abbreviated versions: the ACQ-5 focusing only on the symptoms (omitting bronchodilator use and FEV1) and the ACQ-6 scoring symptoms and bronchodilator use (only omitting FEV1).

During development of the ACQ, it was found that the self-administered version was easily and accurately understood by children aged ≥ 11 years and children < 6 years of age had difficulty understanding the concept of “during the last week.” Therefore, an interviewer-administered version of the ACQ was devised for children aged 6 – 10 years. ACQ should not be used in children < 6 years of age.24 In the IA version, there are instructions to the interviewer on how to ensure that children understand the 7-point scale and the concept of “during the last week.” For example, if the child does not understand the primary wording in the ACQ, such as ‘how limited were you in your activities,’ the interviewer uses the secondary wording, such as ‘how bothered were you in the things you do every day.’ Interviewers were asked to help only when children have difficulties understanding questions. It is recommended that in children < 10 years, the questionnaire should be administered by a trained health professional.24 The IA version of ACQ and an online self-administered version of ACQ are strongly correlated (Pearson r = 0.79), however, are not completely interchangeable. For example, a study showed that better control of asthma is detected when measured with ACQ-IA than by online self-assessment ACQ.52

Validity

Content validity of ACQ has been ensured by having involved pediatric asthma clinicians during development phase. They selected the items in ACQ that are considered the most important to determine asthma control.46

Juniper et al.24 tested measurement properties of the ACQ-7 in 35 children (aged 6 – 16 years) with a wide range of asthma severity and had current symptoms of asthma (ACQ score > 0.5) in England. The cross-sectional construct validity has been demonstrated by strong Pearson correlation coefficients (r > 0.547) between established measures (Mini Pediatric Asthma Quality of Life Questionnaire [MiniPAQLQ], Royal College of Physicians’ “3 questions” [RCP], Asthma Control Diary [ACD], PACQLQ) and ACQs (ACQ-7 = -0.83, 0.52, 0.77, -0.63; ACQ-5 = -0.84, 0.57, 0.71, -0.56, respectively). Longitudinal construct validity has also been shown by similarly strong Pearson correlation coefficients (r > 0.547) between established health status measures,53 (changes in MiniPAQLQ, RCP, ACD, PACQLQ) and changes in ACQ scores over 1-4 weeks (ACQ-7 = -0.89, 0.81, 0.83, -0.49; ACQ-5 = -0.93, 0.81, 0.79, -0.84, respectively). Multiple measures were used since there is no gold standard for measuring asthma control in children. These correlations were close to a priori predications about the degrees of correlations expected of the ACQ if it truly measures change in asthma control. Strong correlations demonstrated that established instruments and ACQ measure a similar concept. However, the author of the study noted that asthma control and quality of life are 2 distinct components of clinical asthma assessment, therefore, some degree of discrepancies are expected between ACQ and the HRQoL measurement instruments.54

The study by Juniper, et al.24 has several limitations in that it tested ACQ in 35 pediatric patients from a single centre with a UK version that had alternative wording to younger children and was administered by a trained interviewers. To overcome some of the limitations, Nguyen, et al.25 tested measurement properties of ACQ in 305 pediatric patients (aged 6 – 17 years old) with inadequately controlled asthma on ICS that were enrolled in a clinical trial using the North American English version of ACQ. They confirmed the moderate to strong construct validity of ACQ by measuring Pearson correlation coefficients with Asthma Control Test (ACT for ages 12 to 17; r = -0.73) or childhood ACT (cACT for ages 6 to 11; r = -0.57), the Asthma Symptom Utility Index (ASUI; for ages 6 to 11, r = -0.74; for ages 12 to 17, r = -0.74), and the PAQLQ (for ages 6 to 11, r = -0.71; for ages 12 to 17 r = -0.66). Furthermore, ACQ differentiated patients who had an episode of poor asthma control from those who did not (for ages 6 to 11, mean difference [SD] = 0.46 [0.01]; P < 0.0001; for ages 12 to 17, mean difference [SD] = 0.30 [0.02]; P < 0.0001) in all aspects such as decrease in PEF rate, increase in rescue medication use, urgent care (ED, hospital, clinic or doctor visit), and SCS use. However, the group did not find such correlation between exhaled NO (n = 146) or expired breath condensate pH (n = 239) and ACQ scores.25

Reliability

Juniper, et al.24 categorized 2 groups of children, e.g., a group of children who remained clinically stable and another group who experienced change in their asthma control between clinic visits (weeks 0-1 and 1-4) based on the Clinician’s Global Rating of Change score. Pearson correlation coefficients between ACQ changes and Global Rating of Change Questionnaire over a period of 1 – 4 weeks were considered to be strong (r > 0.5).47 Test-retest reliability was determined from the stable group by estimating intraclass correlation coefficient (ICC estimated as the within-patient SD and related to the total SD). The group made a priori predications about the level of correlation expected if the ACQ truly measures asthma control. In 19 children who remained stable, there was good evidence for reliability as ICC for the ACQ-7 was 0.79 and ACQ-5 was 0.67.

Even though the concordance between the ACQ-7 and ACQ-5 was high (ICC = 0.93), there was statistical difference between the ACQ-7 and ACQ-5 scores (mean ± SD = 0.12 ± 0.26; P = 0.010) with the greater change in the ACQ-5 than ACQ-7 in scores between baseline and 4 weeks (mean ± SD = 0.17 ± 0.34; P = 0.006).24 Therefore, the 2 versions of ACQ should not be used interchangeable.24

Another group25 found that Cronbach alpha for the ACQ to be 0.74 (n = 305), 0.75 for ages 6 to 11 group (n = 164) and 0.72 for ages 12 to 17 group (n = 141), all of which are considered acceptable for group-level internal consistency (alpha > 0.7).55 Of note, a value of alpha > 0.9 is considered necessary for an individual-level comparison. The ICC for ACQ scores between 2 consecutive visits (total 8 visits) among stable patients who reported no episode of poor asthma control for that period ranged between 0.42 to 0.82 for the overall group, 0.34 to 0.95 for the 6 to 11 age group and 0.22 to 0.74 for the 12 to 17 age group.25 The reliability for ACQ in the overall group and younger patient group was moderate to strong,47 whereas that for older children varied from weak to strong depending on a period of visit measured.

Responsiveness to Change

Responsiveness was measured by detecting within-patient change in the unstable group of children using a paired t-test and between-group changes (stable vs. unstable) using an unpaired t-test. The ACQ-7 and the ACQ-5 both were able to detect changes in asthma control in unstable patients between clinic visits 4 weeks apart (P = 0.026 and 0.007, respectively) and were able to distinguish between stable and unstable patients during the same period (P = 0.072 and 0.027, respectively), with ACQ-5 showing statistically better responsiveness than ACQ-7.24

Similarly, Nguyen, et al.25 demonstrated responsiveness of ACQ to changes in asthma control. Mean changes in ACQ scores between consecutive visits in continuously well-controlled (n = 196 over 547 days) and continuously poorly controlled patients (n = 148 over 337 days) were -0.02 (95% CI = -0.05, 0.01) and 0.06 (95% CI = 0.00, 0.12), respectively. For those who had worsening control (n = 155 over 177 days) and improved control (n = 165 over 200 days), mean changes in ACQ scores were 0.26 (95% CI = 0.11, 0.41) and -0.33 (95% CI = -0.44, -0.22), respectively. Also, mean changes in ACQ scores differed significantly across groups of patients categorized based on their health status for the overall group (P < 0.0001), 6 to 11 age group (P < 0.001), and 12 to 17 age group (P = 0.01).

MID

With the Global Rating of Change method (n = 11), Juniper, et al. estimated the MID for ACQ-7 to be 0.52 (SD = 0.45) and for ACQ-5 to be 0.65 (SD = not reported). The estimated MID was confirmed with the geometric mean regression method (n = 31), which gave a similar result with 0.50 (SEM = 0.05).24 The sponsor used an MID estimate of 0.5 based on the same reference as identified by CADTH literature search.24

Nguyen, et al.25 estimated MID to be 0.375 and 0.382 based on the distribution-based method using 0.5*SD and SEM for the overall group. For those aged 6 to 11 years, MID was estimated to be 0.40 and for those between the ages of 12 and 17 years old, estimated MID was 0.35 using both 0.5*SD and SEM. The group also estimated MIDs based on various anchors. Using the mean ACQ scores among patients experiencing a specified clinical event compared to those who did not as anchor, Nguyen, et al. estimated MID to be 0.4-0.5 for the combined age group, 0.5-0.6 for the 6 to 11 age group, and 0.3-0.4 for the 12 to 17 age group. Anchoring the ACQ against the ACT that uses MID of 3 points, an estimated MID for ACQ was 0.42 for the 12 to 17 age group, and against a 3-point change in the cACT, estimated MID was 0.33 for children ages 6 to 11. Lastly, using the triangulation method,56 the estimated MID was 0.40 for the overall group.

Standardized PAQLQ

The original PAQLQ was developed in McMaster University to measure the problems that children with asthma experience in their daily lives.27 The PAQLQ is the only disease-specific quality of life measurement tool to be specifically developed for use in children with asthma and referred to as the gold standard.28 The PAQLQ has 23 items grouped into 3 domains: symptoms (10 items), emotional aspects (8 items), and activity limitation (5 items) that ask about the most troublesome functional problems to children with asthma.26,28 In the activity domain, there are 3 questions that are patient specific, meaning the child is asked to choose 3 activities that they are most likely to undertake at the time they complete the questionnaire.26,28 These 3 activities remain constant at each follow-up visit. Although the individualized questionnaire enhances content validity by incorporating activities that are most relevant to children, the main disadvantage is that it is time-consuming to complete.28 Also, children tend to change their favourite activities.26 In the PAQLQ(S), the 3 patient-specific questions have been replaced by generic activity questions (physical activity, activities with animals, activities with friends and family) that are the same for all children. These 3 generic activity questions incorporate the activities that were most frequently chosen by children when the original PAQLQ was used. For some studies, such as in long-term clinical trials and patient monitoring, the generic activities become more appropriate since they are easier and more convenient to administer and compare across samples.28 PAQLQ(S) addresses this need, however, one or more of the generic activities may be irrelevant giving rise to a risk of a ceiling effect. The PAQLQ(S) is more appropriate for use in large-scale research projects, which aim to measure quality of life for children with asthma at a group level. The PAQLQ(S) is shorter, quicker to complete and more cost-effective for use in large cohort studies, such as clinical trials for long-term monitoring. The PAQLQ(S) provides standardized responses, which can be coded and analyzed relatively easily to provide broad comparisons across different populations.28

The PAQLQ(S) is a disease-specific, quality of life questionnaire available in both interviewer and self-administered formats, designed for children aged 7 to 17 years of age.26 Questions in each domain are scored on a Likert scale of 1 – 7 (1 = maximum impairment, 7 = no impairment) and are equally weighted to be combined to create mean scores for each domain as well as overall quality of life.28 Therefore, both the domain and overall scores range from 1 – 7, with higher score representing better HRQoL. The recall period is the past 1 week.27

Validity

Wing, et al.28 tested measurement properties in 42 children with asthma (mean age = 11.3 years of old, SD = 2.7; ranges 7 – 17) receiving standard asthma care in UK. All data from Wing, et al. study was generated from interviewer-led questionnaires and none from the self-administered versions. Criterion validity was ensured by comparing PAQLQ(S) overall and domain scores to those of the PAQLQ, which had been developed based on guidelines for a dozen validated disease-specific quality of life instruments and input from patients, parents and health professionals.27 There was no statistically significant difference in scores between the original PAQLQ and the PAQLQ(S), apart from the “activities” subscale (P < 0.03). Both cross-sectional (which demonstrates that the observed differences between children truly reflect differences in asthma-specific quality of life) and longitudinal construct validity (which demonstrates that observed changes in scores over a period of time correlate in a predictable manner with changes in other measures of health status) were evaluated by the Pearson’s correlation coefficients with the original PAQLQ. The cross-sectional construct validity measured at week 9 against ACQ, % predicted PEF, ACD, Health Utilities Index (HUI), and PACQLQ showed Pearson’s coefficients of -0.77 (P < 0.001), 0.25 (P < 0.05), -0.68 (P < 0.001), -0.40 (P < 0.01), and 0.51 (P < 0.001), respectively, for overall scores (data for domain scores are not shown). The longitudinal construct validity measured by Pearson coefficients between weeks 5 and 9 for ACQ, PEF, ACD, HUI, and PACQLQ were -0.79 (P < 0.001), 0.41 (P < 0.01), -0.45 (P < 0.001), -0.46 (P < 0.001), and 0.43 (P < 0.01), respectively for overall scores (data for domain scores are not shown).28

Reliability

The concordance between the original PAQLQ and PAQLQ(S) was measured by ICC (Pearson correlation coeifficient) with a paired Student’s independent t-test and was 0.974, 0.669, 0.955, and 0.863 for overall, activity, symptom, and emotion scores, respectively (P value < 0.001). Internal consistency estimated by Cronbach alpha values at baseline for overall, activity, symptom, and emotion scores were 0.96, 0.90, 0.90, and 0.94, respectively. Of note, reliability in activity domain was higher for PAQLQ(S) compared to PAQLQ probably due to generic questions included to standardize responses. These measures indicate that the correlation with PAQLQ is strong (r > 0.547) and PAQLQ(S) has an acceptable internal consistency (alpha > 0.755), all exceeding alpha = 0.90 set for individual monitoring. Test-retest reliability assessed in patients with stable asthma between weeks 1 – 5 and 5 – 9 (n = 16) was also strong (ICC [95% CI] = 0.92 [0.82, 0.97], 0.81 [0.63, 0.92], 0.89 [0.77, 0.96], and 0.92 [0.84, 0.97] for overall, activity, symptom, and emotion scores, respectively). These results exceed 0.70, which is commonly accepted minimal standard for reliability coefficients.28 However, Wing, et al. expected ICC for test-retest stability greater than 0.85 based on ICCs reported in previous studies.

Responsiveness to Change

The PAQLQ(S) was able to detect the changes in patients who either improved or deteriorated by a score > 0.5 between weeks 1 – 5 and 5 – 9 (paired Student’s t-test, all P < 0.0001) and differences between stable and unstable groups for each of the measures (unpaired Student’s t-test, overall differences all < 0.0003).28

MID

Juniper, et al.27 used anchor-based method to estimate an MID in children with asthma. Briefly, the group took the mean difference in PAQLQ score in patients who score -3, -2, +2, or +3 on the global rating of change as MIDs. According to this method, MID estimates for overall quality life, symptom, activity, and emotion domains were 0.42 ± 0.55 (n = 29), 0.54 ± 0.64 (n = 0.29), 0.70 ± 1.17 (n = 26), and 0.28 ± 0.67 (n = 17), respectively. The sponsor took 0.5 as an MID estimate for PAQLQ(S)-IA based on the same reference.27

Pediatric Rhinoconjunctivitis Quality of Life Questionnaire

The PRQLQ is a disease-specific, HRQoL instrument developed in children 6 to 12 years of age diagnosed with seasonal allergic rhinoconjunctivitis or hay fever who had experienced troublesome symptoms in the previous month.57 The PRQLQ was developed in Southern Ontario to measure the functional problems (physical, emotional, and social) that are most troublesome to children with SAR. The questionnaire should be administered by a trained interviewer. Parents should not be present during the interview31 The PRQLQ consists of 23 items grouped in 5 domains (nose symptoms, eye symptoms, practical problems, activity limitation and other symptoms). Children are asked to recall how they have been during the previous week and respond to each question on a 7-point Likert scale. Responses are given by children with two 7-point scales (a green card and a blue card), where 0 represents no impairment (not bothered/none of the time) and 6 represents maximum impairment (extremely bothered/all of the time). All items are weighted equally. Each domain score and overall score is the mean of the items in each domain and overall questionnaire, respectively. The overall score is estimated from the mean score of all items, thus ensuring that greater weight is given to the domains with the larger number of items. Both the domain and overall scores range from 0 to 6, with higher score representing worse HRQoL. All the words used in the PRQLQ are child-friendly language; however, the recall time (1 week) can be difficult for younger children to understand. Therefore, the PRQLQ should not be used in children under 6 years old.31

Validity

Content validity has been ensured by incorporating several measures during development period: 1) item pools from the Rhinoconjunctivitis Quality of Life Questionnaire, the Adolescent Rhinoconjunctivitis Quality of Life Questionnaire, and the PAQLQ; 2) discussion with children 6 to 12 years old with allergic rhinoconjunctivitis; 3) discussion with clinicians; and 4) a review of the pediatric HRQoL literature.30

Juniper, et al.30 conducted validation study in 75 children (a mean age of 9.8 years, SD = 1.9) who had troublesome allergic rhinoconjunctivitis that required additional medication during the fall pollen season in the US. They assessed longitudinal construct validity by correlating within-patient changes in quality of life scores with within-patient changes in diary symptom scores and global rating of change. The a priori prediction was that if the PRQLQ truly could measure rhinoconjunctivitis-specific quality of life, the correlations should be similar to those observed in adults with rhinoconjunctivitis. Actual and predicated correlations between change in the PRQLQ and diary symptom scores were close across all domains of the PRQLQ and the different symptoms of the diary. For example, stuffy nose (r = 0.58) and mean nose symptoms (r = 0.67) in symptom diary were highly correlated with nose symptom in the PRQLQ (r = 0.55 – 0.7 a priori prediction).

The group also assessed cross-sectional construct validity by correlating quality of life scores with daily symptom diary scores. Again, a priori prediction was that the correlations should be close to those observed in adults. Correlations between the PRQLQ and diary symptoms were very similar to those observed in adults and adolescents with rhinoconjunctivitis, with the highest correlations observed between the nasal domain of the PRQLQ and diary nasal symptoms (r = 0.47 actual vs. r = 0.45 – 0.6 for a priori prediction) and between the eye domain of the PRQLQ and eye symptom scores in the diary (r = 0.59 actual vs. r = 0.45 – 0.6 for a prior prediction). The poorest correlations were found between other symptom domain of the PRQLQ and the diary stuffy nose scores (r = 0.06 actual vs. r < 0.25 a priori prediction).30

Reliability

Juniper, et al.30 measured reliability in 13 children whose rhinoconjunctivitis remained stable between clinic visits at weeks 1 and 3 using ICC. The result shows that ICC was 0.93 for overall quality of life and 0.57, 0.91, 0.79, 0.88, and 0.87 for nose symptoms, eye symptoms, practical problems, other symptoms, and activity limitations, respectively. High ICCs indicate that within-patient variance was consistent in general, except for nose symptoms that showed lower ICC possibly as a result of a low between-patient variance.

Responsiveness to Change

Responsiveness was measured in 61 children who experienced a change in their rhinoconjunctivitis between weeks 1 and 3.30 Using a paired t-test, within-patient changes in children who changed (n = 61) between visits were detected (change in mean score ranges from 0.37 to 0.80). No such changes were found in children (n = 13) who had stable rhinoconjunctivitis (change in mean score ranges from -0.22 to 0.12). Using an unpaired t-test to examine the ability of the instrument to distinguish between patients whose quality of life changed from the beginning to end of the study period and those whose quality of life remained unchanged for the same period was tested. The PRQLQ was able to detect the difference on in overall quality of life (P = 0.005), not each domain (P value ranges from 0.061 to 0.087).

MID

An estimate for MID has not been identified through literature search. The sponsor has taken 0.5 to be an MID for the VOYAGE study based on their literature search.48 However, CADTH could not verify the MID estimate referenced by the sponsor.

Paediatric Asthma Caregiver’s Quality of Life Questionnaire

The PACQLQ is a self-administered questionnaire designed to measure the problems that are most troublesome to the parents (primary caregivers) of children with asthma.29 The questionnaire asks the ways in which child’s asthma has interfered with caregiver’s normal daily activities and how this has made them feel. The PACQLQ was developed and validated using the same methodology as other questionnaires such as PAQLQ(S) or PRQLQ.27

The problems experienced by the parents of younger children (< 7 years) may not be the same as those experienced by the parents of older children and adolescents. For example, by 7 years old, most children are at school and participating in activities without parental involvement and these children are often responsible for taking their own asthma medications. In contrast, children under 7 years may not understand their asthma and how to manage it (e.g., running until severe shortness of breath) and parents worry that they may not detect deterioration in symptoms. Due to these differences, the PACQLQ should only be used in the parents of children 7-17 years. It is likely to be inaccurate in assessing the impact of younger children’s asthma on their parents.29

There are 13 questions in 2 domains (4 items concern activity limitations and 9 concern emotional function). Parents recall the impact that their child’s asthma has had during the previous week.27 Responses to each item in the PACQLQ are given on a 7-point Likert scale where 1 (all of the time/very worried or concerned) represents severe impairment and 7 (none of the time/not worried or concerned) represents no impairment.27 Each item is weighted equally. Domain and overall quality of life scores are calculated as the mean scores; therefore, both range from 1 to 7, with higher score representing better HRQoL. The recall period is the previous 1 week.29

Validity

Content validity has been ensured during development of PACQLQ. Briefly, a pool of items was generated from interviews with parents of children with asthma, a literature review, and discussion with health professionals. The items that were identified most frequently and rated most bothersome by caregivers were selected for the final questionnaire.27

Juniper, et al.27 assessed measurement properties of PACQLQ in 52 children with current symptoms of asthma and their primary caregivers. The children were between 7 and 17 years and represented a wide range of asthma severity. The primary caregiver (age ranged from 30 to 63 years; mean 40.94, SD = 5.6 years) was usually a parent and lived with the child at least 75% of the time. The a priori predictions for both longitudinal and cross-sectional validity was that correlations would be observed if investigators can truly measure quality of life in caregivers. The longitudinal construct validity was measured by correlating within-patient changes in the caregiver’s quality of life scores over a 4-week period with within-patient changes in caregiver burden of illness (generic questionnaire), child’s asthma severity (FEV1% predicted, PEF, beta agonist use, and so forth), global ratings of change, and perception of change in child’s asthma survey. The highest correlations were observed between the PACQLQ emotional function and caregiver global ratings of change – emotions (r = 0.55 vs. r > 0.50 a priori prediction) or caregiver perception of change in child’s’ asthma survey (r = 0.52 vs. r = 0.20 – 0.35 a priori prediction). The poorest correlations were between the change in caregiver burden of illness – mastery and the PACQLQ emotional function (r = -0.0007 vs. r = 0.20 – 0.35 a priori prediction) or activity limitation (r = 0.02 vs. r = no a priori prediction made). The cross-sectional construct validity was measured by correlating caregiver quality of life scores at each clinic visit with the measures of the child’s asthma severity and with generic caregiver quality of life scores. The highest correlations were observed between the PACQLQ activity limitation and caregiver burden of illness – overall (r= -0.65), family/social (r = -0.62 vs. r = 0.20 – 0.35 a priori prediction), and personal strain (r = -0.70). The poorest correlations were observed between the caregiver burden of illness – mastery and the PACQLQ emotional function (r = 0.28 vs. r = 0.20 – 0.35 a priori prediction) and activity limitation (r = 0.26 vs. no a priori prediction). Similarly, such poor correlations were observed between child’s’ asthma severity – beta agonist use and the PACQLQ emotional function (r = -0.22 vs. r = 0.20 – 0.35 a priori prediction) or activity limitation (r = -0.28).27

As predicted, the correlation between the PACQLQ and change in child’s peak flow rates or asthma control was weak, with FEV1 even lower than predicted. In general, correlations between the PACQLQ and the global ratings of change or parental rating of change in the children’s severity of asthma were moderate to high as predicted, with the Impact on Family Scale substantially higher than predicted. These results suggest that problems associated with caring for a child with asthma is not entirely the same as general problems of looking after a sick child.27

Reliability

Reliability was measured in 44 caregivers who were stable between consecutive clinic visits.27 The ICCs for overall quality of life, emotional function, and activity limitation were 0.85, 0.80, and 0.84, respectively. The PACQLQ showed a high degree of reliability with a low within-patient variance (within-patient SD = 0.31 – 0.39) and ability to detect differences in quality of life between caregivers (between-patient SD = 0.71 – 0.88).

Responsiveness to Change

Responsiveness was measured using a paired t-test as ability to detect important within-patient changes when caregivers changed over 4 weeks.27 Caregivers who changed (n = 23) showed the change in mean score per item as 0.72, 0.71, and 1.80 for overall quality of life, emotional function, and activity limitation, respectively (P < 0.001 for all), whereas those who were stable showed -0.03, -0.02, and -0.01, respectively. Also, using an unpaired t-test of the differences between the beginning and end of each period, the ability of the instrument to distinguish between patients who remained stable and those who changed was measured. Changes in those caregivers who changed over 4 weeks were significantly different from the changes in score in the caregivers who reported staying stable (P < 0.0003 for all). The questionnaire proved highly responsive to within-patient changes over time.

MID

Based on global rating of change as anchor, the MID for overall quality of life was estimated to be 0.50 (SD = 0.51; n = 16) with similar values for the emotional function domain (0.64, SD = 0.56; n = 18) and activity limitation domain (0.67, SD = 0.63; n = 9).27

EQ-5D-Y

In 2009, EuroQol group introduced the EQ-5D-Y that is a child-friendly version of EQ-5D and a more comprehensible instrument suitable for children and adolescents. EQ-5D-Y is available in more than 100 languages and in various modes of administration. The EQ-5D-Y is based on the EQ-5D-3L and essentially consists of 2 pages: the EQ-5D descriptive system and the EQ VAS.32 EQ-5D-Y is designed for self-completion by children and adolescents aged 8-15 years. Between the ages of 4 and 7 years, a proxy-completed version should be used. Proxy versions, for completion by a caregiver or someone who knows the person well, are used when children or adolescents are mentally or physically incapable of reporting on their HRQoL, for instance because of severe intellectual disability or mental health problems. Four proxy versions are currently available.32 For adolescents between the ages of 12 and 15 years, both EQ-5D-Y and adult version of EQ-5D can be used depending on study design. All the questions in descriptive questionnaire and VAS ask to describe a health status on the day of administration or “today.”33

The EQ-5D-Y descriptive system comprises 5 dimensions: 1) mobility, 2) self-care, 3) usual activities, 4) pain or discomfort, 5) anxiety/depression. Each dimension has 3 levels: no problems, some problems and a lot of problems. The younger patient is asked to indicate their health state by ticking the box next to the most appropriate statement in each of the 5 dimensions. Even though the EQ-5D-Y descriptive system comprises the same 5 dimensions as the EQ-5D-3L, it uses more appropriate, child-friendly wording. The most relevant differences with the adult EQ-5D-3L are32:

  • The ‘Mobility’ dimension header includes ‘(walking about)’ to facilitate understanding.
  • The title of the second dimension was changed from ‘Self-Care’ to ‘Looking After Myself’.
  • The ‘Usual Activities’ dimension became more child relevant: the new title, ‘Doing Usual Activities,’ is followed by ‘(for example, going to school, hobbies, sports, playing, doing things with family or friends).’
  • For the fifth dimension, ‘Anxiety/Depression’ was replaced with ‘Feeling Worried, Sad or Unhappy.’
  • The wording of the items representing the highest level of severity were changed in all dimensions, from ‘confined to bed’ to ‘a lot of problems walking about,’ in the first dimension, and from ‘being unable to’ to ‘having a lot of problems’ (with washing or dressing myself, or doing usual activities) in the second and third dimensions. In the pain/discomfort dimension, the upper (worst) level was changed from ‘I have extreme pain or discomfort’ in the adult 3L version to ‘I have a lot of pain or discomfort’ in the Y version; in the final dimension, the upper level was changed from ‘I am extremely anxious or depressed’ to ‘I am very worried, sad or unhappy.’
  • The wording of the first response level in the Looking after Myself dimension was also changed from ‘I have no problems with self-care’ to ‘I have no problems washing or dressing myself.’

Each dimension results in a 1-digit number and the digits for the 5 dimensions can be combined to form a 5-digit score that describes the younger patient’s health state. Lower number in 5-digit score indicates better HRQoL. A summary index value can be obtained based on societal preference weights for the health state. The weights or ‘utilities’ are often used to compute QALYs for health economic analyses. Health state index scores generally range from < 0 (negative values represent a health state worse than dead; 0 = dead) to 1 (the value of full health), with higher scores indicating higher health utility. The health state preferences often represent national or regional values and can therefore differ between countries/regions. The second part of the questionnaire consists of a VAS on which the respondent rates their perceived health from 0 (the worst imaginable health) to 100 (the best imaginable health).33 The EQ VAS records the younger patient’s self-rated health and can be used as a quantitative measure of health outcome that reflects the younger patient’s own judgment.32 Higher scores on index and VAS represent better HRQoL.

So far, EQ-5D-Y utility index value sets for children have been published for Slovenian and Spanish population. Similar to EQ-5D-3L, ceiling effect (60.3%), but not floor effect (0%) has been reported with EQ-5D-Y.49

Validity

Mayoral et al.49 assessed measurement properties of EQ-5D-Y administered through a smartphone app in 119 children with asthma (81 self-responded and 38 through proxy response) in Spain between 2018 and 2020. Children aged 8 – 11 years completed self-response version. For those under the age of 8 years, their parents filled out the proxy response version of EQ-5D-Y.

For convergent validity, a hypothesis for a moderate correlation between the Patient-Reported Outcomes Measurement Information System-Pediatric Asthma Impact (PROMIS-PAIS) and the EQ-5D-Y utility index (except for certain questions where a weak correlation is expected) was made considering their asthma-specific and generic aspects, respectively. For divergent validity, a priori hypothesis was a weak correlation between the EQ-5D-Y utility index and the ACQ (except for certain questions that were expected to be moderately correlated), since they differ on the construct being measured (HRQoL and disease control, respectively). Multitrait–multimethod matrix method confirmed the associations for the EQ-5D-Y utility index with the PROMIS-PAIS to be moderate47 (Spearman r = 0.38) and with ACQ to be weak47 (r = 0.28). With the EQ VAS, the correlation coefficients with both PROIMS-PAS and ACQ were weak47 (r = 0.16 and 0.26, respectively).

Another research group tested convergent validity of EQ-5D-Y with PAQLQ in Swedish children (aged 8 – 16 years) with asthma (n = 53 – 90).50 This group hypothesized that most of the domains of 2 questionnaires would be correlated since they do not capture the same aspects of HRQoL as they are constructed differently. The absolute Spearman’s rank correlations between 5 domains of EQ-5D-Y and 3 subdomain scores of PAQLQ ranged from 0.015 (weak) to 0.583 (strong).47 The highest correlations were found in ‘doing usual activities’ and ‘having pain or discomfort’ domains, whereas the lowest correlation was found in ‘looking after myself’ domain of EQ-5D-Y. Also, in general, EQ VAS was moderately to strongly47 correlated with symptoms (r = 0.435), activity limitations (r = 0.567), emotional functions (r = 0.411), and total score (r = 0.517) of PAQLQ. Patients who reported no problems on the EQ-5D-Y consistently reported fewer problems on the PAQLQ.50

Based on the known-groups approach, a priori hypothesis was made by Mayoral, et al.49 that patients with worse control asthma (according to ACQ: well-controlled, immediate, and not well controlled), asthmatic exacerbations last 6 months, higher frequency of SABA inhaler use during the previous 4 weeks, and second-hand smoke exposure would have worse HRQoL. Statistically significant differences were found in the EQ-5D-Y utility index and EQ VAS between groups defined by asthma control, reliever inhalers use, and second-hand smoke exposure, but not with asthmatic exacerbations (which could be attributed to attrition bias effect). Of note, EQ-5D-Y utility index and second-hand smoke exposure were moderately correlated (effect size [ES] = -0.45 [95% CI = -1.01 to 0.1]), but not statistically significant (P = 0.35). The absolute ES coefficients (mean change over SD of change) ranged from 0.38 to 0.75 indicating moderate to strong relationship between the 3 known groups and EQ-5D-Y utility index and EQ VAS.

Reliability

In the same study,49 Mayoral et al. showed test-retest reproducibility of EQ-5D-Y in pediatric patients with asthma who remained stable (n = 19 – 30) and completed the questionnaire twice 6 months apart. When subsample of patients was classified to be stable based on EQ VAS, the ICC was 0.81 with EQ-5D-Y utility index. When the stable patients were classified according to the ACQ, the ICC was 0.79 with EQ-5D-Y utility index and 0.70 with EQ VAS. Based on high ICC values obtained, it can be concluded that EQ-5D-Y reliably reproduces results over time.

Responsiveness to Change

Regarding responsiveness, Mayoral, et al.49 hypothesized that the EQ-5D-Y would be able to detect change over time, though with a lower sensitivity than the asthma-specific instrument PROMIS-PAIS. At 10 months follow-up, responsiveness of EQ-5D-Y utility index for worsening subsamples (n = 6 – 17) presented a degree of change (ES = 0.68 based on EQ VAS and ES = 0.78 based on ACQ), though without statistical significance (P = 0.12 and 0.50, respectively). With respect to EQ VAS, the ES was 1.15 in worsening subsample as measured by ACQ, however, the statistical significance was not detected (P = 0.67), either. In contrast, the PROMIS-PAIS demonstrated better responsiveness in worsening subsample of patients over the same period when patients were evaluated by EQ VAS (ES = 1.08, P = 0.07) or ACQ (ES = 1.28, P = 0.82), though still not be able to detect statistically significant differences.

Minimal Important Difference

The MID for EQ-5D-Y in pediatric patients with asthma has not been estimated based the literature search results.

References

1.
Global strategy for asthma management and prevention (2022 update). Fontana-on-Geneva Lake (WI): Global Initiative for Asthma; 2022: https://ginasthma​.org​/wp-content/uploads​/2022/07/GINA-Main-Report-2022-FINAL-22-07-01-WMS.pdf. Accessed 2022 Sep 08.
2.
Public Health Infobase. Asthma in Canada (2018-05-01). Data Blog. Ottawa (ON): Government of Canada; 2018: https:​//health-infobase​.canada.ca/datalab/asthma-blog.html. Accessed 2022 Sep 08.
3.
Yang CL, Hicks EA, Mitchell P, et al. Canadian Thoracic Society 2021 Guideline update: Diagnosis and management of asthma in preschoolers, children and adults. Can J Respir Crit Care Sleep Med. 2021;5(6):348-361.
4.
CADTH Drug Reimbursement Expert Review Committee final recommendation: dupilumab (Dupixent - Sanofi Genzyme). Can J Health Technol. 2021;1(6). https://www​.cadth.ca​/sites/default/files​/attachments/2021-06​/CADTH_reimbursement​_recommendation_dupilumab_​%28dupixent%29_1.pdf. Accessed 2022 Sep 17.
5.
Dupixent (dupilumab injection): solution for subcutaneous injection, 300 mg single-use syringe (300 mg/2 mL), 200 mg single-use syringe (200 mg/1.14 mL), and 100 mg single-use syringe (100 mg/0.67 mL); Dupixent (dupilumab injection): solution for subcutaneous injection, 300 mg single-use pen (300 mg/2 mL) and 200 mg single-use pen (200 mg/1.14 mL) [product monograph]. Laval (QC): Sanofi-aventis Canada Inc.; 2022 Mar 25.
6.
CADTH Drug Reimbursement Expert Review Committee final recommendation: dupilumab (Dupixent — Sanofi-aventis Canada Inc.). Ottawa (ON): CADTH; 2020 Apr 24: https://www​.cadth.ca​/sites/default/files​/cdr/complete/SR0636​%20Dupixent%20-%20CDEC​%20Final%20%20Recommendation​%20April%2024​%2C%202020%20for%20posting.pdf. Accessed 2022 Sep 17.
7.
Clinical Study Report: SAR231893-EFC14153. VOYAGE: A randomized, double-blind, placebo-controlled, parallel group study to evaluate the efficacy and safety of dupilumab in children 6 to <12 years of age with uncontrolled persistent asthma [internal sponsor's report]. Paris (FR): Sanofi Group; 2020 Dec 07.
8.
Asthma, 2014. Health Fact Sheets. Ottawa (ON): Statistics Canada; 2014: https://www150​.statcan​.gc.ca/n1/pub/82-625-x​/2015001/article/14179-eng.htm. Accessed 2022 Sep 08.
9.
Carr TF, Zeki AA, Kraft M. Eosinophilic and Noneosinophilic Asthma. Am J Respir Crit Care Med. 2018;197(1):22-37. [PMC free article: PMC5765385] [PubMed: 28910134]
10.
Wenzel S. Severe asthma phenotypes. In: Post TW, ed. UpToDate. Waltham (MA): UpToDate; 2021: http://www​.uptodate.com. Accessed 2022 Jul 18.
11.
FitzGerald JM, Lemiere C, Lougheed MD, et al. Recognition and management of severe asthma: A Canadian Thoracic Society position statement. Can J Respir Crit Care Sleep Med. 2017;1(4):199-221.
12.
Sawicki G, Haver K. Asthma in children younger than 12 years: managementof persistent asthma with controller therapies. In: Post TW, ed. UpToDate. Waltham (MA): UpToDate; 2021: http://www​.uptodate.com. Accessed 2022 Jul 18.
13.
Nucala (mepolizumab for injection): 100 mg/mL lyophilized powder for subcutaneous injection in single-use vial; Nucala (mepolizumab injection): 100 mg/mL solution for subcutaneous injection in single-use pre-filled autoinjector or safety syringe [product monograph]. Mississauga (ON): GlaxoSmithKline Inc.; 2021 Nov 05: https://pdf​.hres.ca/dpd_pm/00063467.PDF. Accessed 2022 Sep 08.
14.
Xolair (omalizumab): 150 mg vial, sterile powder for reconstitution for subcutaneous injection; Xolair (omalizumab): 75 mg and 150mg solution for subcutaneous injection in pre-filled syringe [product monograph]. Dorval (QC): Novartis Pharmaceuticals Canada Inc.; 2022 Aug 11: https://pdf​.hres.ca/dpd_pm/00066970.PDF. Accessed 2022 Sep 08.
15.
Holguin F, Cardet JC, Chung KF, et al. Management of severe asthma: a European Respiratory Society/American Thoracic Society guideline. Eur Respir J. 2020;55(1):1900588. [PubMed: 31558662]
16.
McGowan J, Sampson M, Salzwedel DM, Cogo E, Foerster V, Lefebvre C. PRESS Peer Review of Electronic Search Strategies: 2015 guideline statement. J Clin Epidemiol. 2016;75:40-46. [PubMed: 27005575]
17.
Grey matters: a practical tool for searching health-related grey literature. Ottawa (ON): CADTH; 2019: https://www​.cadth.ca/grey-matters. Accessed 2022 Jul 14.
18.
Pocket guide for asthma management and prevention (for adults and children older than 5 years): a pocket guide for physicians and nurses, updated 2015. Fontana-on-Geneva Lake (WI): Global Initiative for Asthma; 2015: https://ginasthma​.org​/wp-content/uploads​/2016/01/GINA_Pocket_2015.pdf. Accessed 2022 Sep 08.
19.
Bacharier LB, Maspero JF, Katelaris CH, et al. Dupilumab in Children with Uncontrolled Moderate-to-Severe Asthma. N Engl J Med. 2021;385(24):2230-2240. [PubMed: 34879449]
20.
Clinical Study Report: SAR231893 - LTS14424. EXCURSION: One-year study to evaluate the long-term safety and tolerability of dupilumab in pediatric patients with asthma who participated in a previous dupilumab asthma clinical study [internal sponsor's report]. Paris (FR): Sanofi Group; 2020 Dec 03.
21.
Miller MR, Hankinson J, Brusasco V, et al. Standardisation of spirometry. Eur Respir J. 2005;26(2):319-338. [PubMed: 16055882]
22.
Barnes PJ, Casale TB, Dahl R, Pavord ID, Wechsler ME. The Asthma Control Questionnaire as a clinical trial endpoint: past experience and recommendations for future use. Allergy. 2014;69(9):1119-1140. [PubMed: 25039248]
23.
Sastre J, Olaguibel J, Vega JM, Del Pozo V, Picado C, Lopez Viña A. Cut-off points for defining asthma control in three versions of the Asthma Control Questionnaire. J Asthma. 2010;47(8):865-870. [PubMed: 20846084]
24.
Juniper EF, Gruffydd-Jones K, Ward S, Svensson K. Asthma Control Questionnaire in children: validation, measurement properties, interpretation. Eur Respir J. 2010;36(6):1410-1416. [PubMed: 20530041]
25.
Nguyen JM, Holbrook JT, Wei CY, et al. Validation and psychometric properties of the Asthma Control Questionnaire among children. J Allergy Clin Immunol. 2014;133(1):91-97.e91-96. [PMC free article: PMC3874246] [PubMed: 23932458]
26.
Qoltech, Measurement of Health-Related Quality of Life & Asthma Control. Standardised Paediatric Asthma Quality of Life Questionnaire (PAQLQ(S)). https://www​.qoltech.co.uk/paqlq_s.html. Accessed 2022 Jul 28.
27.
Juniper EF, Guyatt GH, Feeny DH, Ferrie PJ, Griffith LE, Townsend M. Measuring quality of life in children with asthma. Qual Life Res. 1996;5(1):35-46. [PubMed: 8901365]
28.
Wing A, Upton J, Svensson K, Weller P, Fletcher M, Walker S. The standardized and mini versions of the PAQLQ are valid, reliable, and responsive measurement tools. J Clin Epidemiol. 2012;65(6):643-650. [PubMed: 22537468]
29.
American Thoracic Society Quality of Life Resource. Pediatric Asthma Caregiver's Quality of Life Questionnaire (PACQLQ). 1999; https://qol​.thoracic​.org/sections/instruments​/pt/pages/pacqlq.html. Accessed 2022 Aug 09.
30.
Juniper EF, Howland WC, Roberts NB, Thompson AK, King DR. Measuring quality of life in children with rhinoconjunctivitis. J Allergy Clin Immunol. 1998;101(2 Pt 1):163-170. [PubMed: 9500748]
31.
Qoltech, Measurement of Health-Related Quality of Life and Asthma Control. Paediatric Rhinoconjunctivitis Quality of Life Questionnaire (PRQLQ). http://www​.qoltech.co.uk/prqlq.html. Accessed 2022 Jul 27.
32.
EQ-5D, EuroQol Research Foundation. EQ-5D-Y (Youth) | About. 2021; https://euroqol​.org/eq-5d-instruments​/eq-5d-y-about/. Accessed 2022 Aug 05.
33.
EQ-5D-Y User Guide, version 2.0. Rotterdam (NL): EuroQol Research Foundation; 2020: https://euroqol​.org/publications​/user-guides/. Accessed 2022 Aug 05.
34.
Busse WW, Maspero JF, Rabe KF, et al. Liberty Asthma QUEST: Phase 3 Randomized, Double-Blind, Placebo-Controlled, Parallel-Group Study to Evaluate Dupilumab Efficacy/Safety in Patients with Uncontrolled, Moderate-to-Severe Asthma. Adv Ther. 2018;35(5):737-748. [PMC free article: PMC5960488] [PubMed: 29725983]
35.
Efficacy and safety of dupilumab in uncontrolled moderate-to-severe asthma in 6 to <12-year-old children: systematic literature review and indirect treatment comparison technical report, version 9 [internal sponsor's report]. In: Drug Reimbursement Review sponsor submission: Dupixent (dupilumab), 200 mg and 300 mg solution for subcutaneous injection. Mississauga (ON): Sanofi-aventis Canada Inc; 2022 Jul 14.
36.
Hutton B, Salanti G, Caldwell DM, et al. The PRISMA extension statement for reporting of systematic reviews incorporating network meta-analyses of health care interventions: checklist and explanations. Ann Intern Med. 2015;162(11):777-784. [PubMed: 26030634]
37.
Moher D, Liberati A, Tetzlaff J, Altman DG. Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. PLoS Med. 2009;6(7):e1000097. [PMC free article: PMC2707599] [PubMed: 19621072]
38.
Cochrane handbook for systematic reviews of interventions, version 5.1.0 (updated March 2011). In: Higgins JPT, Green S, eds. London (UK): The Cochrane Collaboration; 2011: https:​//handbook-5-1.cochrane.org/. Accessed 2022 Sep 08.
39.
Lanier B, Bridges T, Kulus M, Taylor AF, Berhane I, Vidaurre CF. Omalizumab for the treatment of exacerbations in children with inadequately controlled allergic (IgE-mediated) asthma. J Allergy Clin Immunol. 2009;124(6):1210-1216. [PubMed: 19910033]
40.
Szefler SJ, Casale TB, Haselkorn T, et al. Treatment Benefit with Omalizumab in Children by Indicators of Asthma Severity. J Allergy Clin Immunol Pract. 2020;8(8):2673-2680.e2673. [PubMed: 32298853]
41.
National Institute for Health and Care Excellence. Single technology appraisal and highly specialised technologies evaluation: user guide for company evidence submission template. (Process and methods PMG24) 2022; https://www​.nice.org​.uk/process/pmg24/chapter​/clinical-effectiveness#quality-assessment-of-the-relevant-clinical-effectiveness-evidence. Accessed 2022 Sep 08.
42.
Clinical Study Report: SAR231893 - LTS14424. Liberty Asthma Excursion: One year study to evaluate the long-term safety and tolerability of dupilumab in pediatric patients with asthma who participated in a previous dupilumab asthma clinical study [internal sponsor's report]. Chilly-Mazarin (FR): Sanofi-Aventis Research and Development; 2022 Apr 12.
43.
Canonica GW, Bourdin A, Peters AT, et al. Dupilumab Demonstrates Rapid Onset of Response Across Three Type 2 Inflammatory Diseases. J Allergy Clin Immunol Pract. 2022;10(6):1515-1526. [PubMed: 35259535]
44.
Gallagher A, Edwards M, Nair P, et al. Anti-interleukin-13 and anti-interleukin-4 agents versus placebo, anti-interleukin-5 or anti-immunoglobulin-E agents, for people with asthma. Cochrane Database Syst Rev. 2021;10:CD012929. [PMC free article: PMC8524317] [PubMed: 34664263]
45.
Miller MA, Davis KL. Dupilumab Efficacy in Patients With Uncontrolled, Moderate-to-Severe Allergic Asthma. Pediatrics. 2020;146(Supplement_4):S375-S375.
46.
Juniper EF, O'Byrne PM, Guyatt GH, Ferrie PJ, King DR. Development and validation of a questionnaire to measure asthma control. Eur Respir J. 1999;14(4):902-907. [PubMed: 10573240]
47.
Cohen J. A power primer. Psychol Bull. 1992;112(1):155-159. [PubMed: 19565683]
48.
Juniper EF, Guyatt GH, O'Byrne PM, Viveiros M. Aqueous beclomethasone diproprionate nasal spray: regular versus “as required” use in the treatment of seasonal allergic rhinitis. J Allergy Clin Immunol. 1990;86(3 Pt 1):380-386. [PubMed: 2212409]
49.
Mayoral K, Garin O, Lizano-Barrantes C, et al. Measurement properties of the EQ-5D-Y administered through a smartphone app in children with asthma: a longitudinal questionnaire study. Health Qual Life Outcomes. 2022;20(1):51. [PMC free article: PMC8959271] [PubMed: 35346225]
50.
Bergfors S, Astrom M, Burstrom K, Egmar AC. Measuring health-related quality of life with the EQ-5D-Y instrument in children and adolescents with asthma. Acta Paediatr. 2015;104(2):167-173. [PubMed: 25393977]
51.
Juniper EF, Chauhan A, Neville E, et al. Clinicians tend to overestimate improvements in asthma control: an unexpected observation. Prim Care Respir J. 2004;13(4):181-184. [PMC free article: PMC6750691] [PubMed: 16701667]
52.
Honkoop PJ, Loijmans RJ, Termeer EH, et al. Comparison between an online self-administered and an interviewer-administered version of the Asthma Control Questionnaire: a cross-sectional validation study. Prim Care Respir J. 2013;22(3):284-289. [PMC free article: PMC6442815] [PubMed: 23616053]
53.
Guyatt GH, Kirshner B, Jaeschke R. Measuring health status: what are the necessary measurement properties? J Clin Epidemiol. 1992;45(12):1341-1345. [PubMed: 1460470]
54.
Juniper EF, Wisniewski ME, Cox FM, Emmett AH, Nielsen KE, O'Byrne PM. Relationship between quality of life and clinical status in asthma: a factor analysis. Eur Respir J. 2004;23(2):287-291. [PubMed: 14979505]
55.
Reeve BB, Wyrwich KW, Wu AW, et al. ISOQOL recommends minimum standards for patient-reported outcome measures used in patient-centered outcomes and comparative effectiveness research. Qual Life Res. 2013;22(8):1889-1905. [PubMed: 23288613]
56.
Leidy NK, Wyrwich KW. Bridging the gap: using triangulation methodology to estimate minimal clinically important differences (MCIDs). COPD. 2005;2(1):157-165. [PubMed: 17136977]
57.
American Thoracic Society Quality of Life Resource. Pediatric Rhinoconjunctivitis Quality of Life Questionnaire (PRQLQ). https://qol​.thoracic​.org/sections/instruments​/pt/pages/prqlq.html. Accessed 2022 Jul 27.
Copyright Notice

Copyright © 2023 - Canadian Agency for Drugs and Technologies in Health. Except where otherwise noted, this work is distributed under the terms of a Creative Commons Attribution-NonCommercial- NoDerivatives 4.0 International licence (CC BY-NC-ND).

Bookshelf ID: NBK596600

Views

Related information

  • PMC
    PubMed Central citations
  • PubMed
    Links to PubMed

Recent Activity

Your browsing activity is empty.

Activity recording is turned off.

Turn recording back on

See more...
-