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.

Clinical Review Report: Insulin Degludec (Tresiba): (Novo Nordisk Canada Inc): Indication: For once-daily treatment of adults with diabetes mellitus to improve glycemic control [Internet]. Ottawa (ON): Canadian Agency for Drugs and Technologies in Health; 2017 Dec.

Cover of Clinical Review Report: Insulin Degludec (Tresiba)

Clinical Review Report: Insulin Degludec (Tresiba): (Novo Nordisk Canada Inc): Indication: For once-daily treatment of adults with diabetes mellitus to improve glycemic control [Internet].

Show details

Results

Findings From the Literature

A total of 20 studies were identified from the literature for inclusion in the systematic review (Figure 1). The included studies are summarized in Table 11 and described in the Included Studies section. A list of excluded studies is presented in Appendix 3.

Figure 1. Flow Diagram for Inclusion and Exclusion of Studies.

Figure 1

Flow Diagram for Inclusion and Exclusion of Studies.

Table 11. Details of Included Studies — DEVOTE, SWITCH-1, and SWITCH-2.

Table 11

Details of Included Studies — DEVOTE, SWITCH-1, and SWITCH-2.

Included Studies

Description of Studies

Fifteen manufacturer-sponsored randomized controlled trials (RCTs) plus five extensions were included in this systematic review, four of which were double blinded and the remainder open label. The studies featured different populations, T1DM and T2DM. Another four RCTs were excluded because either their small sample size or their short duration suggested they were unlikely to add any additional information beyond what was obtained in the studies summarized in the report.

DEVOTE was a double-blind RCT that compared IDeg with IGlar, both in a basal-bolus regimen, in a population of 7,637 patients with T2DM and cardiovascular disease. The primary outcome was the time to first major adverse cardiovascular event (MACE), testing the noninferiority of IDeg to IGlar, with a margin for noninferiority of 1.3 for the upper limit of the 95% confidence interval (CI) for the hazard ratio. DEVOTE was an event-driven study, targeting 633 MACEs before study completion, and it lasted a mean of 24 months. Confirmatory secondary end points, which all tested the superiority of IDeg to IGlar, included the number of confirmed hypoglycemic episodes and the occurrence of at least one hypoglycemic episode within a participant. The study had a two-week screening period, as well as a 30-day follow-up period at the end of study. DEVOTE is by far the largest of the studies included in this review.

The SWITCH studies, SWITCH-1 (T1DM) and SWITCH-2 (T2DM), employed a crossover design, with patients randomized to start on either IDeg or IGlar and then cross over to the other intervention after 32 weeks of therapy, resulting in a total treatment period of 64 weeks. The primary outcome of the SWITCH studies was the proportion of participants with severe or blood glucose–confirmed symptomatic hypoglycemic episodes during the maintenance period, that is, after 16 weeks of treatment. SWITCH-1 tested the noninferiority of IDeg to IGlar, with noninferiority confirmed if the upper bound of the 95% CI for the rate ratio was ≤ 1.10. SWITCH-2 tested the superiority of IDeg to IGlar. In each study, before testing of the primary outcome could proceed, noninferiority had to be confirmed for the secondary supportive end point of change from baseline in glycated hemoglobin (A1C) after 32 weeks of therapy. The margin for noninferiority was 0.4%, the same margin used in the BEGIN trials, described below. Each of the SWITCH studies had a two-week screening period and a one-week follow-up.

All of the BEGIN trials that compared IDeg with another basal insulin (described in more detail below) were noninferiority trials that tested the noninferiority of IDeg to a comparator for the primary outcome of change from baseline in A1C. Most of the trials had confirmatory secondary outcomes that compared the superiority of IDeg against the comparator. The extension trials, where they occurred, focused on the long-term safety of IDeg; therefore, there were no efficacy outcomes assessed. In the extensions, all patients continued in their originally randomized groups.

Type 1 Diabetes Mellitus

Studies 3583 (BBT1 [basal-bolus TD1M] Long, 52-week treatment period), 3770 (Flex T1, 26 weeks), and 3585 (BBT1, 26 weeks) were all open label, and all had extensions where patients continued on their originally randomized treatments. Study 3585 had IDet as a comparator, while the other two studies compared IDeg with IGlar. In Flex T1, the objective of the study was to compare a flexible dosing regimen of IDeg with a regular IDeg regimen or to IGlar. The studies had one-week screening and one-week follow-up. The primary outcome of Studies 3770, 3583, and 3585 was the change from baseline to end of treatment in A1C. All of these studies tested the noninferiority of IDeg to a comparator, with a margin for noninferiority of 0.4% for the change from baseline in A1C. All but Study 3770 had confirmatory secondary end points that tested the superiority of IDeg to their respective comparators.

Type 2 Diabetes Mellitus

Insulin-Naive

Five open-label RCTs and one double-blind RCT enrolled patients with T2DM who were insulin-naive. All participants were receiving OADs. The comparator in four of the studies was IGlar, while in Study 3580 (BEGIN Early) the comparator was sitagliptin and in Study 3944, the only double-blind RCT, the comparator was placebo. Study 3672 (BEGIN Low Volume) compared the more concentrated formulation, IDeg 200 U/mL, with IGlar, while the other studies used the standard 100 U/mL concentration. Five studies were 26 weeks in duration, while Study 3579 (BEGIN Once Long) was a 52-week study. All studies except Study 3944 had a one-week screening period and at least a one-week follow-up, while Study 3944 also had a 15-week run-in where participants were initiated on liraglutide, which they would all continue once the double-blind period started. One trial (Study 3579) had an extension while the others did not. The primary outcome in all studies was the change in A1C from baseline to end of study. Studies with IGlar as a comparator tested the noninferiority of IDeg to IGlar for the primary end point, while Studies 3580 and 3944 tested the superiority of IDeg for the primary outcome. Confirmatory secondary outcomes, which all tested the superiority of IDeg to a comparator, included the number of treatmentemergent severe or minor hypoglycemic episodes, change from baseline in fasting plasma glucose (FPG), within-patient variability as measured by coefficient of variation in self-measured FPG, and responders without hypoglycemic episodes (A1C < 7.0% at end of trial and no severe or minor hypoglycemic episodes during the last 12 weeks of treatment including only patients exposed for at least 12 weeks). The confirmatory secondary outcomes in Study 3580 were change from baseline in FPG, frequency of responders (A1C < 7.0% at end of trial), and frequency of responders without hypoglycemic episodes (A1C < 7.0% at end of trial and no severe or minor hypoglycemic episodes during the last 12 weeks of treatment).

Basal Insulin Only (± OADs)

Two open-label RCTs (Studies 3668 and 3943) were included with this population. In Study 3668, all participants received metformin, with or without a dipeptidyl peptidase-4 inhibitor, and IDeg was compared with IGlar over a 26-week treatment course. The trial had a one-week screening period and at least one week of follow-up. The primary outcome was change from baseline in A1C to the study end point at 26 weeks; the study tested the noninferiority of IDeg to IGlar using the same noninferiority margin as the other BEGIN trials, 0.4%. Secondary end points, none of which appeared to be confirmatory, included the number of confirmed hypoglycemic episodes, change from baseline in FPG, within-patient variability in pre-breakfast, self-measured plasma glucose, and responders without hypoglycemic episodes (A1C < 7.0% at end of trial and no confirmed episodes during the last 12 weeks of treatment, including only patients exposed for at least 12 weeks). Study 3943 was a noninferiority, open-label RCT with a crossover design featuring two treatment periods of 16 weeks each where IDeg was compared with IGlar. Participants were all on metformin plus or minus an additional OAD. The trial had a one-week screening period and a 16-week run-in where participants discontinued their OAD (other than metformin) and were initiated on a regimen of IGlar, as well as a one-week follow-up. The purpose of the run-in was to establish which participants required a “high” dose of IGlar (> 81 units), as this was the population of interest for the study. The primary outcome again tested noninferiority for the change from baseline in A1C. Secondary outcomes were A1C responders, change from baseline in FPG, self-measured plasma glucose, and patient-reported outcomes; however, none of these appeared to be confirmatory.

Basal-Bolus Insulin (± OADs)

One noninferiority open-label RCT (Study 3582) was included with this population. Participants were receiving metformin, plus or minus pioglitazone, and IDeg was compared with IGlar. Participants were on a regimen that combined these basal insulins with insulin aspart. The primary outcome was the change in A1C from baseline to the study end point at 52 weeks. Confirmatory secondary end points included change from baseline in FPG after 52 weeks, frequency of responders (A1C < 7.0% at end of trial), and frequency of responders without hypoglycemic episodes (A1C < 7.0% at end of trial and no severe or minor hypoglycemic episodes during the last 12 weeks of treatment).

Populations

Inclusion and Exclusion Criteria

Participants in DEVOTE had T2DM, with an A1C of 7% or more (or below 7% if receiving current insulin therapy of at least 20 units daily). Participants were currently treated with one or more oral or injectable antidiabetes drugs. They had to be at least 50 years old and have evidence of cardiovascular disease or chronic kidney disease (Table 11).

In the trials in T1DM, participants had to have been treated on a basal-bolus regimen for at least 12 months, with an A1C of 10% or less and a BMI of 35 kg/m2 or less. For a high-level summary of these trials, see Table 12; for detailed summaries, see Table 39 and Table 40.

Table 12. Type 1 Diabetes Mellitus and Type 2 Diabetes Mellitus Trial Details.

Table 12

Type 1 Diabetes Mellitus and Type 2 Diabetes Mellitus Trial Details.

In the T2DM trials in insulin-naive patients, participants had to have had T2DM for at least six months, an A1C of between 7.0% or 7.5% and 10%, and a maximum BMI of 40 kg/m2 to 45 kg/m2. All were receiving OADs for at least three months before randomization in a regimen that typically featured metformin with or without another OAD. See Table 12 for a high-level summary of study designs; for detailed summaries, see Table 41, Table 42, Table 43, Table 44, and Table 45.

Participants in Study 3668 (basal only) had to have had T2DM for at least six months and be on OAD monotherapy, insulin monotherapy, or a combination of the two. The only allowed OADs were metformin, insulin secretagogues, or pioglitazone. Participants on OAD alone had to have an A1C between 7% and 11%. Those on combination basal insulin and OAD or basal insulin monotherapy were to be between 7% and 10%. In the other basal-only study, participants were to have an A1C of at least 7.5% and be on metformin with or without another OAD. For a high-level summary of these two studies, see Table 12; for detailed summaries, see Table 46 and Table 47.

In the basal-bolus study, participants were to have hadT2DM for at least six months and could be on any insulin regimen, with or without OADs, for at least three months before randomization. Their A1C had to be between 7.5% and 11%, and their maximum BMI was to be 40 kg/m2. For a high-level summary, see Table 12; for a detailed summary, see Table 48.

Baseline Characteristics

Trials with populations with T1DM tended to feature younger participants (early to mid-40s) versus studies that focused on T2DM (mid-50s to mid-60s), which aligns with the onset and progression of the disease subtypes. Among T2DM studies, the oldest populations were in DEVOTE (65 years of age) and SWITCH-2 (61 years of age) (Table 13, Table 14).

Table 13. Summary of Baseline Characteristics — DEVOTE.

Table 13

Summary of Baseline Characteristics — DEVOTE.

Table 14. Summary of Baseline Characteristics — SWITCH-1 and SWITCH-2.

Table 14

Summary of Baseline Characteristics — SWITCH-1 and SWITCH-2.

Across all studies, the majority of participants were male, and most were Caucasian, with the exception of Studies 3585 and 3586, where almost all participants were Asian (non-Indian), and Study 3587, where about two-thirds were Asian (non-Indian).

In the T1DM studies, between 14% and 29% of participants had diabetes complications at baseline. In DEVOTE, 86% of participants had established cardiovascular or chronic kidney disease, while in the other T2DM studies, the proportion of participants with diabetes complications varied widely at baseline, from a low of around 10% in Studies 3579 and 3580 to a high of about 40% in Study 3586.

Baseline characteristics were generally balanced between groups within studies. The most common baseline parameter to differ between groups was gender, with the largest difference between groups found in Study 3668, where 59% of participants in the IDeg-Flex (IDeg flexible dosing regimen) group and 48% of participants in the IGlar group were male.

Table 15. Summary of Baseline Characteristics — Type 1 Diabetes Mellitus (Studies 3770, 3583, and 3585).

Table 15

Summary of Baseline Characteristics — Type 1 Diabetes Mellitus (Studies 3770, 3583, and 3585).

Table 16. Summary of Baseline Characteristics — Type 2 Diabetes Mellitus, Insulin-Naive (Studies 3579, 3580, 3586, and 3672).

Table 16

Summary of Baseline Characteristics — Type 2 Diabetes Mellitus, Insulin-Naive (Studies 3579, 3580, 3586, and 3672).

Table 17. Summary of Baseline Characteristics — Type 2 Diabetes Mellitus, Insulin-Naive (Studies 3587 and 3944).

Table 17

Summary of Baseline Characteristics — Type 2 Diabetes Mellitus, Insulin-Naive (Studies 3587 and 3944).

Table 18. Summary of Baseline Characteristics — Type 2 Diabetes Mellitus, Basal Insulin (Studies 3668 and 3943).

Table 18

Summary of Baseline Characteristics — Type 2 Diabetes Mellitus, Basal Insulin (Studies 3668 and 3943).

Table 19. Summary of Baseline Characteristics — Type 2 Diabetes Mellitus, Basal-Bolus (Study 3582).

Table 19

Summary of Baseline Characteristics — Type 2 Diabetes Mellitus, Basal-Bolus (Study 3582).

Interventions

The studies generally employed a treat-to-target strategy for insulin dosing. For example, in SWITCH-1, participants’ plasma glucose was titrated to a self-measured plasma glucose of 4.0 mmol/L to 5.0 mmol/L. A dose reduction was to be implemented if one or more of the pre-breakfast glucose values was < 4.0 mmol/L. Bolus insulin (insulin aspart) was titrated individually based either on carbohydrate counting or by using a sliding scale based on the lowest of three pre-meal or bedtime glucose values. Participants were typically given an algorithm they used to adjust their insulin regimens throughout the trial.

The majority of studies randomized participants in a 1:1 manner; however, Studies 3585, 3586, and 3587 randomized participants in a 2:1 manner (IDeg to comparator), and Studies 3579, 3583, and 3582 randomized 3:1. Two studies, 3770 and 3668, included an IDeg-Flex regimen in addition to the standard IDeg daily regimen, where participants’ injections were to be given in a rotating schedule with eight-hour to 40-hour intervals between doses. These studies randomized participants 1:1:1. If stratification was reported or performed, the most common variable was by region (Studies 3587, 3944, 3586, 3585). Study 3580 stratified by use of pioglitazone at screening, Study 3582 by prior insulin regimen, and Study 3668 by prior treatment (insulin, OAD, or both). The SWITCH studies and DEVOTE did not report whether stratification occurred.

Type 2 Diabetes Mellitus

Inclusion criteria specified adequate minimum doses of OADs to ensure that antidiabetes therapy was optimized before intervention and that inadequacy of glycemic control at baseline was not due to suboptimal dosing of OAD treatment. No washout period was applied. During the trials, patients continued on the pre-specified OADs at unchanged doses unless dose reduction was required for safety reasons.

Two studies, 3944 and 3943, had extensive run-in periods. In Study 3944, the 15-week run-in was used to initiate participants on liraglutide, which was to become the standard adjunctive therapy, added to IDeg and placebo, during the treatment phase. In Study 3943, the 16-week run-in was used to determine which participants needed a high (> 80 units) dose of IGlar to maintain glycemic control.

Outcomes

The primary outcome of DEVOTE was time from randomization to first occurrence of an event adjudication committee (EAC)–confirmed 3-component MACE: cardiovascular death, non-fatal myocardial infarction, or non-fatal stroke.

Confirmed hypoglycemic episodes consisted of episodes of severe hypoglycemia as well as minor hypoglycemic episodes with a confirmed plasma glucose value of < 3.1 mmol/L. Hypoglycemic episodes were defined as nocturnal if the time of onset was between 00:01 a.m. and 05:59 a.m., inclusive. Severe hypoglycemia was defined as an episode requiring the assistance of another person to actively administer carbohydrate or glucagon, or take other resuscitative actions.

Blood samples for A1C were analyzed using a Bio-Rad high performance liquid chromatography method at a central laboratory. A1C samples were collected at multiple visits in the main trial and extensions (where applicable). The assay method used was a National Glycohemoglobin Standardization Program–certified method. Blood samples for FPG were analyzed using a Roche enzymatic method at a central laboratory. FPG samples were collected at multiple visits in the main trials and extensions (where applicable). The patients were to attend these visits in a fasting state.

Within-patient variability as measured by coefficient of variation was to be derived from pre-breakfast plasma glucose values after 26 weeks of treatment. Logarithm-transformed self-measured plasma glucose values were to be analyzed as repeated measures in a linear mixed model with treatment, antidiabetes treatment at screening, sex, and region as fixed factors, age as a covariate, and patient as random factor. The model was to assume independent within-patient and between-patient errors with variances depending on treatment. Within-patient variability as measured by coefficient of variation for a treatment could be calculated from the corresponding residual variance. The CI for the coefficient of variation ratio between treatments was to be calculated using the delta method.

Changes in patients’ health-related quality of life (HRQoL) and treatment-related impacts of minor hypoglycemic episodes on patients’ daily function and well-being were evaluated using the Short Form (36) Health Survey, version 2.0 (SF-36v2) and Treatment-Related Impact Measure — Hypoglycemic Events (TRIM-HYPO) questionnaires, respectively. Responses for the SF-36v2 were measured on standardized scales from 1998 based on the US general population, with a mean of 50 and standard deviation of 10. Responses for the TRIM-HYPO were standardized to a scale of 0 to 100. In the SF-36v2 questionnaire, higher scores indicate a better HRQoL. In the TRIM-HYPO questionnaire, lower scores indicate better daily function and well-being for the patient.

SF-36 is a generic health assessment questionnaire that has been used in clinical trials to study the impact of chronic disease on HRQoL. SF-36 consists of 36 items representing eight dimensions: physical functioning, role physical, bodily pain, general health, vitality, social functioning, role emotional, and mental health. Item response options are presented on a 3-point to 6-point Likert-like scale. Each item is scored on a 0 to 100 range and item scores are averaged together to create the eight domain scores. SF-36 also provides two component summaries, the physical component summary and the mental component summary, which are created by aggregating the eight domains according to a scoring algorithm. On any of the scales, an increase in score indicates improvement in health status. Based on clinical anchor data, the SF-36 User’s Manual proposed the following minimal important differences for general use of the SF-36v2: a change of 2 to 4 points in each domain or 2 to 3 points in each component summary.26 No minimal clinically important difference (MCID) in patients with T1DM or T2DM was found in the literature.

Treatment-Related Impact Measure — Diabetes (TRIM-D) is a diabetes-specific questionnaire developed to assess the full impact of diabetes treatment on patients’ quality of life. This patient-reported outcome measure consists of 28 items encompassed in five domains: treatment burden (six items), daily life (five items), diabetes management (five items), psychological health (eight items), and compliance (four items). Response options are presented on a 5-point Likert-like scale. An increase in score indicates an improvement in health state. Domains can be scored individually or the measure can be scored as a total of these domains.27,28 No MCID has been determined for the TRIM-D.

TRIM-HYPO is a patient-reported outcome measure developed to measure the impact of non-severe hypoglycemic events on patients’ HRQoL arising from the use of insulin to treat both forms of diabetes (T1DM and T2DM). TRIM-HYPO is a self-reported questionnaire comprising 33 Likert-like scale items (scored 1 to 5) in five domains: daily functioning, emotional well-being, diabetes management, work productivity, and sleep disruption. Domains are scored individually. A total score is also calculated, using three of the five domains (daily functioning, emotional well-being, diabetes management), as work productivity and sleep disruption do not apply to all patients. Lower scores on the TRIM-HYPO indicate a better health state. Raw scores are obtained by aggregating scale items into their respective domain scales. A weighted score is then generated, based by the number of non-severe hypoglycemic occurrences in the past 30 days: the higher the number, the greater the impact on the weighted score. This weighting helps account for the difference in HRQoL of patients experiencing few events versus those experiencing many hypoglycemic events. A standard algorithm method transforms the weighted scores into a 0 to 100 score.29 No MCID has been determined for the TRIM-HYPO.

Statistical Analysis

All included studies carried out power calculations to determine sample size, and all studies randomized sufficient numbers of patients to ensure adequate power for assessing the primary end point.

DEVOTE

The primary end point (time from randomization to first occurrence of an EAC-confirmed, three-component MACE) was presented descriptively in a Kaplan–Meier plot and analyzed using a Cox proportional hazard regression with treatment (IDeg and IGlar) as a factor. The hazard ratio and the corresponding two-sided 95% CI were estimated. Noninferiority of IDeg to IGlar was considered confirmed if the upper limit of the two-sided 95% CI for the hazard ratio was below 1.3 or equivalent if the P value for the one-sided test of the null hypothesis, hazard ratio ≥ 1.3, against the alternative hypothesis, hazard ratio < 1.3, was < 2.5%. This is the margin recommended by the FDA for evaluating the cardiovascular safety of new antihyperglycemic drugs.30 Results for the full analysis set (FAS) population were also presented as for the per-protocol population.

Any EAC-confirmed MACE occurring after a patient’s first EAC-confirmed MACE did not contribute to the analysis (i.e., time to first event only). Where an EAC-confirmed cardiovascular death was linked by the EAC to an earlier myocardial infarction or stroke, the patient contributed to the analysis with time to the cardiovascular death. If a patient did not experience any EAC-confirmed MACE, the time was censored at the patient’s individual end-of-trial date.

Patients were allowed to go on and off randomized treatment during the trial (resulting in “on-treatment” and “off-treatment” periods). Sensitivity analyses were made using the same Cox regression model as the primary analysis. but including only EAC-confirmed MACEs occurring during an on-treatment period.

Four sensitivity analyses were performed, covering two types of censoring mechanisms: strict censoring (censor at time of first EAC-confirmed MACE if occurring during an off-treatment period) and censoring where the first EAC-confirmed MACE occurring during an off-treatment period was ignored. These censoring mechanisms were applied to two types of on-treatment definition:

  • on-treatment: EAC-confirmed MACEs occurring on randomized treatment
  • on-treatment + 30 days: EAC-confirmed MACEs occurring on randomized treatment plus up to 30 days of a subsequent off-treatment period.

Provided that noninferiority for the primary end point was confirmed, the number of EAC-confirmed severe hypoglycemic episodes was analyzed using a negative binomial regression model with log-link function and the logarithm of the observation time as offset. The model included treatment (IDeg versus IGlar) as a fixed factor, and was fitted using the FAS. Superiority was considered confirmed if the upper limit of the two-sided 95% CI for the rate ratio was below 1.0, or equivalent if the P value for the one-sided test of the null hypothesis, rate ratio ≥ 1.0, against the alternative hypothesis, rate ratio < 1.0, was less than 2.5%.

Several subgroup analyses were reported for the primary outcome, of which one was relevant to our protocol: cardiovascular risk group (patients with established cardiovascular disease or chronic kidney disease versus patients with risk factors for cardiovascular disease).

Multiplicity

The primary and secondary confirmatory end points were tested in a predefined hierarchical order to control the overall type I error. In this testing sequence, it was necessary to fulfill the test criteria (i.e., to reject the corresponding null hypothesis) in order to go to the next step. If the corresponding null hypothesis was not rejected, the testing was stopped, and no further hypotheses were tested.

  • Step 1: Noninferiority of IDeg versus IGlar for the primary end point
  • Step 2: Superiority of IDeg versus IGlar for the number of EAC-confirmed severe hypoglycemic episodes
  • Step 3: Superiority of IDeg versus IGlar for the occurrence of at least one EAC-confirmed severe hypoglycemic episode in a patient.

Because the statistical tests and results of the interim analysis did not affect the continuation of the trial or the statistical tests and results of the full trial data (as stated in the statistical analysis plan for the interim analysis), there was no need to adjust the alpha level for the statistical tests of the full trial data.

Missing Data

In DEVOTE, a tipping-point analysis was made to address the impact of missing information for patients not completing the trial. Events were added for all patients randomized to IDeg not having an EAC-confirmed MACE in the primary analysis who were non-completers (i.e., 66 patients) and lost to follow-up (i.e., four patients).

As these patients had observation periods of different lengths, the order in which they were added to the analysis could potentially have an impact. Hence, patients were sorted based on the duration of their observation times using the following approaches:

  • forward imputation of events, in which events were imputed for patients with the shortest observation period first and the longest observation last
  • backward imputation, where events were imputed for patients with the longest observation period first and the shortest observation last
  • iImputation using median time to event in reference group for patients with non-informative censoring and observation time less than median time in reference.

Finally, the tipping point was established by adding first EAC-confirmed MACEs to the IDeg group until the tipping point (i.e., upper limit of the one-sided 95% CI for hazard ratio > 1.3) was reached. Each added EAC-confirmed MACE was assumed to have an onset date on the day following the patient’s end-of-trial date.

SWITCH (Studies 3995 and 3998)

Analyses of all end points were based on the FAS. Efficacy end points and patient-reported outcomes were summarized using the FAS.

Multiplicity

In both SWITCH studies, before testing the primary end point, the secondary supportive efficacy end point (“Change from baseline in A1C after 32 weeks of treatment”) was tested for noninferiority as a prerequisite for testing the primary end point. The analysis was made for each treatment period separately. Analysis was based on a mixed model for repeated measurement; treatment, sex, region, pre-trial insulin treatment regimen, visit, and dosing time were fixed effects, and age and baseline A1C were covariates.

Noninferiority was considered confirmed if the upper bound of the two-sided 95% CI for A1C was equal to or below 0.40% or if the P value for the one-sided test of the null hypothesis (treatment difference > 0.40%) against the alternative hypothesis (treatment difference ≤ 0.40%) was less than 2.5% (IDeg once daily + insulin aspart [IAsp] minus IGlar once daily + IAsp).

Upon confirmation of noninferiority for both treatment periods, the primary end point was tested for noninferiority in SWITCH-1 and for superiority in SWITCH-2. The number of treatment-emergent severe or blood glucose–confirmed symptomatic hypoglycemic episodes during the maintenance period was analyzed using a Poisson model with patient as a random effect; treatment, period, sequence, and dosing time as fixed effects; and time exposure to trial drug in each counting period for hypoglycemic episodes as an offset.

Noninferiority was considered confirmed if the 95% CI for the rate ratio (IDeg followed by IGlar) was ≤ 1.10 or if the P value for the one-sided test of the null hypothesis (rate ratio > 1.10) against the alternative hypothesis (rate ratio ≤ 1.10) was less than 2.5%, where rate ratio is the estimated rate ratio of IDeg followed by IGlar. If noninferiority was confirmed, the superiority of IDeg followed by IGlar was investigated outside of the test hierarchy. Superiority was considered confirmed if the upper bound of the two-sided 95% CI was < 1.00.

Two confirmatory secondary end points were tested provided that superiority was confirmed for the primary end point. The confirmatory secondary end points are given below together with the direction of the test.

The following safety end points were assessed in the maintenance period (i.e., after 16 weeks of treatment) and in each treatment period (weeks 16 to 32 and weeks 48 to 64):

  • number of treatment-emergent severe or blood glucose–confirmed symptomatic nocturnal hypoglycemic episodes
  • proportion of patients with one or more severe hypoglycemic episodes.

The number of treatment-emergent severe or blood glucose–confirmed symptomatic nocturnal hypoglycemic episodes during the maintenance period was tested using the same model and sensitivity analyses as for the primary end point. In SWITCH-1, this was a noninferiority analysis, as for the primary outcome, and in SWITCH-2, this was a superiority analysis, as for the primary outcome.

The proportion of patients with one or more severe hypoglycemic episodes in the maintenance period was tested for superiority in both SWITCH studies using McNemar’s test, in which the proportion of patients with severe hypoglycemic episodes treated with IDeg was tested against the proportion of patients with severe hypoglycemic episodes treated with IGlar and not treated with IDeg.

Missing Data

Patients who withdrew or dropped out of the trial were explored with the purpose of investigating whether, in particular, the population that dropped out before the first maintenance period was different from the population exposed in the first maintenance period, and whether there were any differences in dropout between the two treatments. This analysis was added to the statistical analysis plan.

In the primary analysis, patients who were not exposed in the second maintenance period contributed to the estimation of the treatment difference. This implies that these patients were assumed to behave like patients who were exposed in both maintenance periods; that is, a “missing completely at random” assumption. To investigate how this assumption influenced the primary results, a sensitivity analysis was added that included only patients who were exposed in both maintenance periods. This analysis follows the randomization principle in that the same patients were analyzed on both treatments. The treatment estimate from this analysis is an unbiased estimate in the subset of patients who were exposed to the maintenance period for both treatments, under the assumption that missing data for patients who drop out in the second maintenance period are missing at random. Since data from patients who were exposed only in the first maintenance period were excluded, the pragmatic effectiveness principle is violated.

BEGIN Trials (Studies 3770 [Plus Extension], 3585 [Plus Extension], 3583 [Plus Extension], 3579, 3580, 3672, 3586, 3587, 3944, 3668, 3943, and 3582 [Plus Extension])

A1C was analyzed centrally using a National Glycohemoglobin Standardization Program–certified method. The primary objective in all the therapeutic confirmatory trials was to confirm the efficacy of IDeg with respect to glycemic control as measured by change in A1C from baseline to end of trial between IDeg and an active comparator. The primary end point was analyzed using an analysis of variance method with treatment, antidiabetes therapy at screening, sex, and region as fixed factors, and age and baseline A1C as covariates. In the three-arm trials, the primary analysis was IDeg-Flex versus the comparator IGlar. It is not clear how multiplicity was adjusted for when the IDeg-Flex group was compared with the IDeg group.

All efficacy analyses, as well as analyses of hypoglycemia and body weight, were based on the FAS and followed the intention-to-treat principle, with patients contributing to the evaluation “as randomized.” Unless otherwise specified, missing values (including intermittent missing values) were imputed using the last observation carried forward (LOCF) method as recommended for its transparency in the FDA guidance. Both baseline and post-baseline values were used for LOCF, in line with the intention-to-treat principle. As adherence to the intention-to-treat principle could bias the results toward null (i.e., no difference between treatments), the noninferiority assessments for A1C were also confirmed using a per-protocol analysis set, which included only patients treated for at least 12 weeks with a valid post-baseline A1C assessment. In addition, a post hoc analysis was made including only patients who completed the trial.

With the exception of Studies 3580 and 3944, all trials were noninferiority trials, and efficacy was considered confirmed if the upper bound of the two-sided 95% CI for the estimated treatment difference for A1C (IDeg versus comparator) was ≤ 0.4%. This limit — which, according to the manufacturer, is in agreement with the FDA guidance on diabetes — has been used in previous submissions for other insulin products (NovoRapid/NovoLog, NovoMix, and Levemir). Study 3580 tested the superiority of IDeg to sitagliptin, and Study 3944 tested the superiority of IDeg to placebo.

There were five extensions among the BEGIN trials. All three studies in T1DM had extensions, as did T2DM Studies 3579 (insulin-naive) and 3582 (basal-bolus). In all cases, the primary objective of the extensions was to assess safety and tolerability; therefore, they focused on outcomes such as hypoglycemia, adverse events, body weight, and insulin dose, which were all considered to be primary end points. The extensions did not have a primary efficacy variable. The data from the core studies and extensions were to be combined and analyzed as one trial using the original baseline values from core trials. This combined data set was to be the basis for derivation, analyses, and presentation of end points. An additional analysis set, the extension trial set, was reported for efficacy analyses.

Data from study site 109 in Study 3582 was excluded from the analysis due to concerns about the quality of the data after an audit.

Multiplicity

The overall type I error rate was controlled using a hierarchical testing procedure. Hence, if noninferiority was confirmed for the primary end point (superiority in Studies 3580 and 3944), the therapeutic confirmatory trials (except for Studies 3668 and 3770) aimed at demonstrating superiority for a number of confirmatory secondary end points, ordered on the basis of their clinical relevance within the respective treatment regimens and populations investigated. Consequently, superiority could be confirmed only for end points where all previous hypotheses had been confirmed, and the term “superior” is used solely if statistical superiority was confirmed based on hierarchical testing. If superiority was not confirmed, the result was considered to have the same level of evidence as the remaining, non-confirmatory end points.

Missing Data
Type 1 Diabetes Mellitus

Missing values were imputed by LOCF for all end points. To assess the sensitivity of the LOCF method on the conclusion from the analysis of the primary A1C analysis, two sensitivity analyses were performed. The repeated measurement model addressed whether the inclusion of the A1C values at all scheduled visits in the model would provide different results versus the simpler approach in the primary analysis, where only baseline information and the last A1C measurement were included.

All observed A1C measurements available post-randomization at scheduled measurement times were also to be analyzed in a linear mixed model using an unstructured residual covariance matrix (if possible). This approach relies on the assumption that data are missing at random according to the taxonomy defined by Rubin. The results were to be compared against the results of the LOCF method for dealing with missing data. Any marked difference concerning treatment differences between the missing-at-random approach and the LOCF approach was to be commented upon in the clinical trial report

The per-protocol analysis addressed whether the LOCF from patients withdrawing early in the trial (before the A1C measurement had stabilized) or patients randomized in error (not necessarily fulfilling all inclusion and exclusion criteria) influenced the conclusion. The conclusions from these analyses were very similar and resulted in the same conclusion as the primary analysis.

Type 2 Diabetes Mellitus

For studies in T2DM, missing data were accounted for in a similar manner to the studies in T1DM. In Study 3582, an additional sensitivity analysis was performed that addressed the impact of the patients from site 109 who were removed from the FAS.

Analysis Populations

DEVOTE

The following analysis sets were defined in the protocol:

  • FAS: All randomized patients. The statistical evaluation of the FAS followed the intention-to-treat principle. Patients were to contribute to the evaluation “as randomized.”
  • Per-protocol analysis set: This included all patients who had been continuously on the investigational medicinal product the first three months after randomization, as well as those who had an EAC-confirmed MACE within the first three months and took at least one dose of the investigational medicinal product before the event.

No safety analysis set was defined because safety was analyzed using the FAS. All analyses were based on the FAS population, with the exception of one sensitivity analysis of the primary end point that used the per-protocol population.

SWITCH

The following analysis sets were defined in accordance with the ICH E9 guidance:

  • FAS: All randomized patients. In exceptional cases, patients from the FAS could be eliminated. In such cases, the elimination was to be justified and documented. The statistical evaluation of the FAS would follow the intention-to-treat principle and patients would contribute to the evaluation “as randomized.”
  • Safety analysis set: All patients receiving at least one dose of the investigational product or its comparator. Patients in the safety set would contribute to the evaluation “as treated.”
  • Completer analysis set: All patients who complete both treatment periods. If a patient withdrew during follow-up after the second treatment period, the patient was considered a completer.

Type 1 Diabetes Mellitus (Studies 3585, 3583, and 3770) and Type 2 Diabetes Mellitus (Studies 3579, 3580, 3586, 3672, 3944, 3587, 3668, and 3943)

The following analysis sets were defined in accordance with the ICH-E9 guidance (International Conference on Harmonization, Guideline on Statistical Principles for Clinical Trials, E9):

  • FAS: All randomized patients. In exceptional cases, patients from the FAS could be eliminated. In such cases, the elimination was to be justified and documented. The statistical evaluation of the FAS was to follow the intention-to-treat principle, and patients were to contribute to the evaluation “as randomized.”
  • Per-protocol analysis set: Patients without any major protocol violations that may have affected the primary end point. Moreover, patients must have been exposed to the investigational product or its comparator for more than 12 weeks and must have had a valid assessment necessary for deriving the primary end point. Patients in the per-protocol set were to contribute to the evaluation “as treated.”
  • Safety analysis set: All patients who received at least one dose of the investigational product or its comparator. Patients in the safety set were to contribute to the evaluation “as treated.”

Patient Disposition

The proportion of participants withdrawing varied greatly between studies, with the highest withdrawal rates seen in the SWITCH studies, ranging between 18% and 23% between groups, and the lowest in DEVOTE, around 2% (Table 20). Proportions of withdrawals above 20% were also seen in studies 3579 (22%) and 3580 (24%), although no differences in the proportion of withdrawals were evident between groups (Table 22). The largest difference in proportion of withdrawals was in Study 3944, with the IDeg + liraglutide group having a much lower proportion than the placebo + liraglutide group (8% versus 24%, respectively) (Table 23). In Study 3770, there was a numerically higher proportion of withdrawals in both IDeg groups versus IGlar (16% in each IDeg group versus 7% in IGlar) (Table 21).

Table 20. Patient Disposition — DEVOTE, SWITCH-1, and SWITCH-2.

Table 20

Patient Disposition — DEVOTE, SWITCH-1, and SWITCH-2.

Table 22. Patient Disposition — Type 2 Diabetes Mellitus, Insulin-Naive (Studies 3579, 3643, 3586, 3672, and 3580).

Table 22

Patient Disposition — Type 2 Diabetes Mellitus, Insulin-Naive (Studies 3579, 3643, 3586, 3672, and 3580).

Table 23. Patient Disposition — Type 2 Diabetes Mellitus, Insulin-Naive (Studies 3587 and 3944).

Table 23

Patient Disposition — Type 2 Diabetes Mellitus, Insulin-Naive (Studies 3587 and 3944).

Table 21. Patient Disposition — Type 1 Diabetes Mellitus (Studies 3770, 3583, and 3585 + Extensions).

Table 21

Patient Disposition — Type 1 Diabetes Mellitus (Studies 3770, 3583, and 3585 + Extensions).

Table 24. Patient Disposition — Type 2 Diabetes Mellitus, Basal Insulin (Studies 3668 and 3943).

Table 24

Patient Disposition — Type 2 Diabetes Mellitus, Basal Insulin (Studies 3668 and 3943).

Table 25. Patient Disposition — Type 2 Diabetes Mellitus, Basal-Bolus (Studies 3582 and 3667).

Table 25

Patient Disposition — Type 2 Diabetes Mellitus, Basal-Bolus (Studies 3582 and 3667).

Table 26. Exposure to Study Treatments — Mean.

Table 26

Exposure to Study Treatments — Mean.

Exposure was generally similar between groups among studies, with the largest difference in Study 3944, which is also the study with the largest difference in withdrawal rates between groups.

Table 27. Basal Insulin Dose.

Table 27

Basal Insulin Dose.

Critical Appraisal

Internal Validity

All studies used an appropriate method of randomization using an interactive voice/Web response system with appropriate allocation concealment. Patient characteristics were generally well balanced in most studies, although in Study 3668 the baseline characteristics were not well balanced with respect to gender. With respect to blinding, only DEVOTE, SWITCH-1, and SWITCH-2, as well as Study 3944, were double blinded, and the insulins were provided in “visually identical” vials. All other studies were open labelled and therefore subject to bias, particularly for subjective outcomes such as HRQoL, which could be influenced by patient knowledge of their assigned treatment. A blinded EAC was employed in DEVOTE, and an interim analysis was prepared and submitted to the FDA.

The BEGIN studies that included an insulin comparator used a noninferiority design for testing the primary outcome (change from baseline to end of treatment in A1C), and all used the same margin for noninferiority of a change in A1C of 0.4%. The manufacturer provided a rationale for the choice of this noninferiority margin, and this rationale appeared to be reasonable. This margin is also suggested by the FDA, which considers an A1C reduction of > 0.3% to be clinically meaningful; therefore, a difference in A1C of 0.3 to 0.4% between treatments could be considered clinically significant.36 Noninferiority was also tested for the primary outcome in DEVOTE; again, the rationale of the margin for noninferiority was described and the margin appeared reasonable. The SWITCH studies employed a noninferiority design in a hierarchy in order to determine testing of the primary outcome. In both studies, noninferiority for change from baseline in A1C, a secondary end point, had to be met before the primary end point and subsequent secondary end points in the hierarchy were tested.

A hierarchical testing procedure was employed to account for type I error in all studies that included confirmatory secondary end points, and the hierarchy was adhered to. The studies that did not include confirmatory secondary end points were Studies 3770, 3668, 3943, and 3944.

In the SWITCH-1 study, a per-protocol sensitivity analysis of the primary end point, a test of noninferiority, does not appear to have been conducted, as a per-protocol population was not defined in the study. The use of a per-protocol population is a recommended approach for noninferiority trials; thus, differences between the groups could have been masked, particularly given the high withdrawal rates in these studies.

With the exception of DEVOTE, no studies conducted a true intention-to-treat analysis; however, the small number of participants excluded from the main analysis is unlikely to have biased results.

The proportion of participants withdrawing varied greatly between studies, with the highest withdrawal rates seen in the SWITCH studies, ranging between 18% and 23% between groups. The direction of bias is confounded by the crossover design; however, given the high proportion of withdrawals, it is likely that the results were affected in some way, as the composition of the original randomized population would have been altered significantly throughout the trial. Proportions of withdrawals above 20% were also seen in studies 3579 (22%) and 3580 (24%), although generally no differences in proportion of withdrawals were evident between groups. The largest difference in proportion of withdrawals was in Study 3944, with the IDeg + liraglutide group having a much lower proportion than the placebo + liraglutide group (8% versus 24%, respectively). In Study 3770, there was a numerically higher proportion of withdrawals in both IDeg groups versus IGlar (16% in each, versus 7%). A high proportion of withdrawals may understate important outcomes such as hypoglycemic events, for example; and a higher proportion of withdrawals in one group versus another may bias results in favour of the group with more withdrawals, as they have less exposure to risk of hypoglycemia. Additionally, extensive withdrawals are a concern, given the noninferiority study designs employed across the studies.

The included studies typically accounted for missing data using an LOCF approach. Sensitivity analyses were also performed and appeared to support the results of the primary analysis. The LOCF approach can introduce bias into the results, and the risk of bias would be expected to increase with higher proportions of withdrawals and when there are differential withdrawals between groups within studies; both of these phenomena were seen among the included studies. The fact that the sensitivity analyses supported the conclusions of the LOCF results does allay some concern about the use of this approach for the imputation of missing data; however, a major assumption in the sensitivity analysis is that the data were missing at random, which is rarely the case and could also bias the results. The DEVOTE study, which was an event-driven study, employed a tipping-point analysis to account for missing data due to early withdrawals.

All studies employed a treat-to-target design with respect to dosing; therefore, differences in A1C would not be expected. This approach is recommended by the FDA for assessing differences in safety, tolerability, and clinical utility when insulin dosing and efficacy are maximized. However, these studies have limited utility for evaluating treatment efficacy.

External Validity

There were numerous clinical trials included in this review, with representation across the globe. Across all the included studies, there was a relatively low proportion of Indigenous participants (< 1% across the studies). The consistent majority of participants were Caucasian, with the exception of studies conducted in Asia. The lack of representation of Indigenous populations is a generalizability issue for Canada, given the relatively high proportion of Indigenous peoples diagnosed with diabetes mellitus. The clinical expert also noted that participants had a relatively long duration of disease at baseline, but did not note any other generalizability issues.

The trials largely focused on IGlar as a comparator, with IDet a comparator in only one of the studies. These are the two most commonly used intermediate-acting and long-acting insulins; however, NPH is still a popular option in some patients due to its lower cost. Thus, at least one trial comparing IDeg with NPH may have provided some additional useful insight into the comparative efficacy and harms of IDeg.

DEVOTE had the longest follow-up of all the included studies, with a mean exposure of approximately 24 months, and was powered to assess clinical outcomes in a T2DM population. However, there were no trials that similarly assessed diabetes complications in T1DM. Such trials would likely require a much longer follow-up than those included in this review. Other included trials focused on hypoglycemia as a primary outcome (SWITCH studies); the majority of included studies (BEGIN trials) focused on A1C, a widely used surrogate marker of disease in diabetes mellitus.

Two studies had extensive run-in periods, which can suggest enrichment of the study population. In Study 3943, the run-in period was to establish that the study population was one requiring high-dose insulin (participants were included only if they failed to reach target while on high-dose IGlar); this does seem appropriate, given the study objective. In Study 3944, participants were initiated on liraglutide in the 15-week run-in, which they then continued on in the study. The purpose of the study was to assess the combination of IDeg with liraglutide versus liraglutide alone (i.e., placebo plus liraglutide) in patients on metformin. A large proportion of participants were screened out during the run-in (in most cases due to failure to reach A1C targets); this was consistent with the planned 941 participants in the run-in versus 320 that were to be randomized. The study set a relatively narrow target for A1C (7.0% to 9.0%) for inclusion in the study. The rationale was that, as it was placebo-controlled, this narrow range would reduce the risk of intensified treatment being needed during the 26-week treatment phase.

HRQoL was consistently assessed in the included studies, but not as a confirmatory (i.e., high-priority) secondary outcome and not at all in the largest study, DEVOTE. Thus, HRQoL appears to have been given a lower priority in the included studies than would be expected based on the importance that patients place on this outcome.

Efficacy

Only those efficacy outcomes identified in the review protocol are reported below. See Appendix 5: Detailed Outcome Data for detailed efficacy data.

Morbidity

In DEVOTE, the primary composite cardiovascular outcome occurred in fewer IDeg than IGlar participants (8.5% versus 9.3% of participants), for a hazard ratio of 0.91 (95% CI, 0.78 to 1.06) (Table 32). The upper bound of the 95% CI was below 1.3, confirming noninferiority of IDeg relative to IGlar with respect to cardiovascular safety (P < 0.001). Other signs of morbidity where there was no difference between IDeg and IGlar were non-fatal myocardial infarction (hazard ratio 0.85; 95% CI, 0.68 to 1.06; P = 0.15), non-fatal stroke (hazard ratio 0.90; 95% CI, 0.65 to 1.23; P = 0.50), and unstable angina leading to hospitalization (hazard ratio 0.95; 95% CI, 0.68 to 1.31; P = 0.74).

Table 32. Key Efficacy Outcomes — DEVOTE.

Table 32

Key Efficacy Outcomes — DEVOTE.

Type 1 Diabetes Mellitus

There were few MACEs in any of the three studies in T1DM (Studies 3770, 3583, and 3585) and no clear differences between groups (Table 28).

Table 28. BEGIN Trials — Patients With an Adjudicated Major Adverse Cardiovascular Event.

Table 28

BEGIN Trials — Patients With an Adjudicated Major Adverse Cardiovascular Event.

Type 2 Diabetes Mellitus

There were few MACEs in most of the studies in T2DM and generally no differences between groups. One exception may have been Study 3643, the extension to Study 3579, where after 104 weeks, 3.8% of IDeg and 1.6% of IGlar-treated participants had a MACE (Table 28).

Health-Related Quality of Life

HRQoL was assessed using the SF-36 and TRIM (both TRIM-D and TRIM-HYPO) instruments.

HRQoL was not assessed in DEVOTE. In SWITCH-1 and SWITCH-2, there was no statistically significant difference between IDeg and IGlar in any of the SF-36 subscales. In SWITCH-2, the manufacturer noted that there was an improvement in TRIM-HYPO for IDeg versus IGlar; however, this outcome was not part of the confirmatory end points and no P values were reported (Table 33). There is no MCID for the TRIM-HYPO.

Table 33. Key Efficacy Outcomes — SWITCH-1 and SWITCH-2.

Table 33

Key Efficacy Outcomes — SWITCH-1 and SWITCH-2.

Type 1 Diabetes Mellitus

There were no consistent differences between IDeg and comparators in any of the subscales of the SF-36 or the TRIM-D instruments (Table 49, Table 50, and Table 51).

Type 2 Diabetes Mellitus

There were no consistent differences between IDeg and comparators in any of the subscales of the SF-36 or the TRIM-D instruments (Table 52, Table 53, Table 54, Table 55, Table 56, Table 57, and Table 58).

Glycated Hemoglobin

Type 1 Diabetes Mellitus

Change in A1C from baseline to end of treatment was the primary outcome of Studies 3770, 3583, and 3585. IDeg was noninferior to IGlar (Studies 3770 and 3583) and to IDet (Study 3585) in these studies. In Study 3583, the treatment difference between IDeg and IGlar was −0.01 (95% CI, −0.14 to 0.11), and in Study 3585, after 52 weeks, the treatment difference between IDeg and IDet was −0.09 (95% CI, −0.23 to 0.05). In Study 3770, only the flexible IDeg group was tested versus IGlar, and the treatment difference was 0.17 (95% CI, 0.04 to 0.30), which was judged to be noninferior, as the upper limit of the 95% CI for the estimated treatment difference was > 0 and ≤ 0.4% (Table 29).

Table 29. Primary Outcome — Change From Baseline in Glycated Hemoglobin (BEGIN Trials).

Table 29

Primary Outcome — Change From Baseline in Glycated Hemoglobin (BEGIN Trials).

Type 2 Diabetes Mellitus

Insulin-Naive

The primary outcome of Studies 3579, 3586, 3587, and 3672 was to test the noninferiority of IDeg to IGlar for the change from baseline to end of study in A1C. In Study 3579, the treatment difference after 52 weeks between IDeg and IGlar was 0.09 (95% CI, −0.04 to 0.22). In Study 3672, after 26 weeks, the treatment difference between IDeg and IGlar was 0.04 (95% CI, −0.11 to 0.19). In Study 3586 after 26 weeks, the treatment difference between IDeg and IGlar was 0.11 (95% CI, −0.03 to 0.24) (Table 29).

The primary outcome of Study 3580 was to test the superiority of IDeg to sitagliptin for the change from baseline to end of treatment (26 weeks) in A1C. IDeg was superior to sitagliptin, with a treatment difference of −0.43 (95% CI, −0.61 to −0.24; P < 0.001).

Study 3944 tested the superiority of IDeg + liraglutide to placebo + liraglutide. The combination of IDeg + liraglutide was found to be superior to placebo + liraglutide, with a treatment difference of −0.92 (95% CI, −1.10 to −0.75; P < 0.0001) (Table 29).

Insulin-Experienced (Basal Only)

Change from baseline to end of treatment (52 weeks) in A1C was the primary outcome of Study 3668. In Study 3668, after 52 weeks of therapy, IDeg was noninferior to IGlar, with a treatment difference after 52 weeks between IDeg and IGlar of 0.09 (95% CI, −0.04 to 0.22) (Table 29).

Insulin-Experienced (Basal-Bolus)

Change from baseline to end of treatment (26 weeks) in A1C was the primary outcome of Study 3582. In Study 3582, after 26 weeks of therapy, IDeg was noninferior to IGlar, with a treatment difference between IDeg and IGlar of 0.08 (95% CI, −0.05 to 0.21) (Table 29).

Blood Glucose (Fasting)

Type 1 Diabetes Mellitus

Change in FPG was a confirmatory outcome in Studies 3585 and 3583, and as part of the statistical hierarchy, was not tested. In Study 3770, FPG was not part of a statistical hierarchy and also was not tested (Table 30).

Table 30. Key Secondary Outcome — Change in Fasting Plasma Glucose (BEGIN Trials).

Table 30

Key Secondary Outcome — Change in Fasting Plasma Glucose (BEGIN Trials).

Type 2 Diabetes Mellitus

Insulin-Naive

As part of the statistical hierarchy, the difference in change in FPG between IDeg and comparators was not tested, as testing had been halted by this time. In other studies where FPG was not part of the statistical hierarchy, P values were not reported (Table 30).

In Study 3580, IDeg was superior to sitagliptin for change from baseline in FPG, with a treatment difference of −2.17 mmol/L (95% CI, −2.59 to −1.74; P < 0.001). IDeg also elicited a statistically significant reduction in FPG versus placebo in Study 3944, with a treatment difference of −2.55 mmol/L (95% CI, −3.07 to −2.02; P < 0.0001) (Table 30).

Insulin-Experienced (Basal)

In Studies 3668 and 3943, change from baseline in FPG does not appear to have been part of the statistical hierarchy; therefore, no P values were reported (Table 30).

Insulin-Experienced (Basal-Bolus)

In Study 3582, IDeg was not shown to be superior to IGlar for change from baseline in FPG (Table 30).

Blood Glucose (Variability)

Blood glucose variability was reported in the T1DM trials, and was part of the statistical hierarchy in studies 3585 and 3583; however, testing had been halted by this time, so no P value was reported (Table 31).

Table 31. Key Secondary Outcome — Blood Glucose Variability (BEGIN Trials).

Table 31

Key Secondary Outcome — Blood Glucose Variability (BEGIN Trials).

In the T2DM studies, blood glucose variability was part of the statistical hierarchy, but testing had been halted once this end point was reached, or the outcome was not part of the statistical hierarchy, and no P values were reported.

Harms

Only those harms identified in the review protocol (see the Objectives and Methods section) are reported here. See Appendix 5 for detailed harms data.

Adverse Events

There were no clear differences in the overall proportion of participants with an adverse event in DEVOTE or in the SWITCH studies. The most common adverse event was nasopharyngitis (Table 37).

Table 37. Harms — DEVOTE, SWITCH-1, and SWITCH-2.

Table 37

Harms — DEVOTE, SWITCH-1, and SWITCH-2.

In the BEGIN trials, in Study 3770, there was a numerically lower proportion of IDeg (68%) participants who were on a flexible dosing regimen who experienced an adverse event versus those on a regular IDeg (76%) regimen or on IGlar (72%) (Table 59). In Study 3586, 59% of IDeg-treated and 65% of IGlar participants experienced an adverse event (Table 64). Otherwise, there were no clear differences in the proportion of IDeg versus IGlar participants experiencing an adverse event in Study 3583 (or extension) or Study 3579 or extension (Study 3643), or between IDeg and IDet in Study 3585, while in the extension (Study 3725), 82% of IDeg and 78% of IDet participants had experienced an adverse event by 104 weeks There was no clear difference in the proportion of participants with an adverse event with IDeg compared with sitagliptin in Study 3580.

Serious Adverse Events

There were no clear differences between IDeg and IGlar in the proportion of participants experiencing a serious adverse event in DEVOTE or in the SWITCH studies (Table 37).

In the BEGIN trials, there was no clear difference in the proportion of participants with a serious adverse event between IDeg and IGlar (Table 34). In Study 3725, the extension to Study 3585, 12% of IDeg versus 7% of IDet participants had a serious adverse event. There was no clear difference in proportion of participants with a serious adverse event with IDeg compared with sitagliptin in Study 3580 or with IDeg + liraglutide versus placebo + liraglutide in Study 3944.

Table 34. Harms — BEGIN Trials.

Table 34

Harms — BEGIN Trials.

Withdrawals Due to Adverse Events

There were no clear differences in the proportion of participants who withdrew due to an adverse event in DEVOTE or in the SWITCH studies (Table 37). In the BEGIN trials, there were numerically more IDeg participants who withdrew due to an adverse event compared with sitagliptin (4% versus 1%) (Table 34).

Notable Harms

DEVOTE

In DEVOTE, the number of EAC-confirmed severe hypoglycemic episodes and the occurrence of at least one EAC-confirmed severe hypoglycemic episode within a patient were confirmatory secondary end points. The risk of a severe hypoglycemic event was lower in the IDeg treatment group compared with the IGlar treatment group, and this difference was statistically significant, with a rate ratio of 0.73 (95% CI, 0.60 to 0.89; P < 0.001) (Table 37).

SWITCH-1 and SWITCH-2

Severe or blood glucose–confirmed hypoglycemic episodes during the maintenance period (16 weeks into treatment) was the primary outcome of both SWITCH trials. In both trials, there was a statistically significant reduction in these episodes for IDeg versus IGlar, with a treatment ratio of 0.89 (95% CI, 0.85 to 0.94; P < 0.0001) in SWITCH-1 and 0.70 (95% CI, 0.61 to 0.80) in SWITCH-2 (Table 37).

BEGIN trials

Confirmed hypoglycemic events, both overall and nocturnal events, were consistently reported across the BEGIN trials as confirmatory secondary end points. In two of three studies where it was tested in T1DM and in all four comparisons versus IGlar in T2DM, insulin-naive patients, IDeg was not shown to be superior to IGlar (five studies) or IDet (one study) (Table 35). The only study that reported a statistically significant decrease in the risk of hypoglycemia with IDeg versus comparator (IGlar) was Study 3582, with a treatment ratio of 0.82 (95% CI, 0.69 to 0.99; P = 0.018) (Table 35). Data were collected for this outcome in other studies but not as a confirmatory end point, and no P values were reported.

Table 35. Key Secondary Outcome — Confirmed Hypoglycemic Events (BEGIN Trials).

Table 35

Key Secondary Outcome — Confirmed Hypoglycemic Events (BEGIN Trials).

Confirmed hypoglycemic episodes consisted of episodes of severe hypoglycemia as well as minor hypoglycemic episodes with a confirmed plasma glucose value of < 3.1 mmol/L. Hypoglycemic episodes were defined as nocturnal if the time of onset was between 00:01 a.m. and 05:59 a.m., inclusive. Hypoglycemic episodes occurring during sleep in the extended time range from 10:01 p.m. to 07:59 a.m. were also analyzed.

Table 36. Key Secondary Outcome — Confirmed Nocturnal Hypoglycemic Events (BEGIN Trials).

Table 36

Key Secondary Outcome — Confirmed Nocturnal Hypoglycemic Events (BEGIN Trials).

Copyright © 2017 Canadian Agency for Drugs and Technologies in Health.

The copyright and other intellectual property rights in this document are owned by CADTH and its licensors. These rights are protected by the Canadian Copyright Act and other national and international laws and agreements. Users are permitted to make copies of this document for non-commercial purposes only, provided it is not modified when reproduced and appropriate credit is given to CADTH and its licensors.

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), a copy of which is available at http://creativecommons.org/licenses/by-nc-nd/4.0/

Bookshelf ID: NBK533969

Views

  • PubReader
  • Print View
  • Cite this Page
  • PDF version of this title (6.4M)

Other titles in this collection

Recent Activity

Your browsing activity is empty.

Activity recording is turned off.

Turn recording back on

See more...