Vous êtes sur la page 1sur 9

Received: 19 June 2019 Revised: 31 October 2019 Accepted: 4 November 2019

DOI: 10.1111/dom.13911

ORIGINAL ARTICLE

Liraglutide reduces hyperglycaemia and body weight in


overweight, dysregulated insulin-pump-treated patients with
type 1 diabetes: The Lira Pump trial—a randomized, double-
blinded, placebo-controlled trial

Thomas F. Dejgaard MD1,2* | Signe Schmidt MD1,2,3 | Christian S. Frandsen MD2 |


Dorte Vistisen MSc1 | Sten Madsbad MD2 | Henrik U. Andersen MD1 |
Kirsten Nørgaard MD1,2

1
Steno Diabetes Center Copenhagen,
University of Copenhagen, Denmark Abstract
2
Department of Endocrinology, Copenhagen Aim: To investigate the efficacy of adding the glucagon-like peptide-1 receptor ago-
University Hospital Hvidovre, Hvidovre,
nist liraglutide to continuous subcutaneous insulin infusion (CSII) in overweight or
Denmark
3
Danish Diabetes Academy, Odense, Denmark obese persons with type 1 diabetes and non-optimal glycaemic control.
Materials and methods: A 26-week, randomized, double-blind, placebo-controlled
Correspondence
Thomas F. Dejgaard, Steno Diabetes Center trial including 44 overweight or obese adults with type 1 diabetes randomized 1:1 to
Copenhagen, Niels Steensens Vej 2-4, DK- liraglutide 1.8 mg once daily (QD) or placebo added to CSII treatment. The primary
2820 Gentofte, Denmark,
Email: thomas.fremming.dejgaard@regionh.dk endpoint was change in haemoglobin A1c (HbA1c). Secondary endpoints included
change in insulin dose, CSII settings, glycaemic variability, body weight and patient-
Funding information
Novo Nordisk, Grant/Award Number: reported outcome measures. Finally, adverse effects including hypoglycaemic events
Unrestricted grant; Copenhagen University were registered.
Hospital; European Association for the Study
of Diabetes; American Diabetes Association Results: HbA1c was reduced by 5 mmol/mol (0.5%) from a baseline of 66 mmol/mol
(8.2%) in patients treated with liraglutide compared with a non-significant change of
Peer Review
The peer review history for this article is +2.3 mmol/mol (0.2%) from a baseline of 66 mmol/mol (8.1%) in patients treated
available at https://publons.com/publon/10. with placebo (between-group difference 7 mmol/mol [0.7%], P < 0.001). Liraglutide
1111/dom.13911.
reduced total insulin dose by 8 units/day or 16% of total insulin dose (P = 0.008).
Mean body weight was reduced by 6.3 kg (P < 0.001) compared with placebo. Con-
comitantly, time spent in glycaemic target range 4–10 mmol/L (71–180 mg/dL)
increased while the risk of hypoglycaemia did not differ between groups at the end
of treatment.
Conclusion: Liraglutide treatment reduced HbA1c, total daily insulin dose and body
weight without increasing the risk of hypoglycaemia in CSII-treated patients with
type 1 diabetes and insufficient glycaemic control. Liraglutide may be considered a
potential add-on therapy to insulin in this subgroup of patients.

1 | INTRODUCTION
*Parts of this study were presented in abstract form at the American Diabetes Association
(ADA) 77th Scientific Sessions, San Diego, 9-13 June 2017 and at the 53rd Annual Meeting
Intensified insulin treatment is associated with hypoglycaemia and
of the European Association for the Study of Diabetes (EASD), Lisbon, 11-15
September 2017. substantial weight gain. Currently, more than 50% of persons with

Diabetes Obes Metab. 2019;1–9. wileyonlinelibrary.com/journal/dom © 2019 John Wiley & Sons Ltd 1
2 DEJGAARD ET AL.

type 1 diabetes in the industrialized part of the world are overweight, insulin sensitivity factors and insulin-to-carbohydrate ratios were
and interventions to prevent weight gain are strongly warranted.1,2 increased 15% at initiation of the investigational medicinal product
Compared with multiple daily injections (MDI) subcutaneous insu- (IMP). The basal rates, insulin sensitivity factors and insulin-to-
lin infusion (CSII) therapy improves glycaemic control and reduces the carbohydrate ratios were subsequently increased or decreased if nec-
incidence of severe hypoglycaemia, most pronounced in persons with essary during the trial in accordance with international guidelines.4 The
frequent hypoglycaemia and insufficient glycaemic control.3 The use randomization list was provided by the manufacturer Novo Nordisk
of CSII is recommended by international guidelines as a therapeutic A/S and the study personnel who did the randomization had no further
option for patients with type 1 diabetes who do not achieve target involvement in the trial.
glycaemic control on treatment with MDI.4 The T1D Exchange Clinic To evaluate endogenous insulin production, a meal test was per-
Registry reports that >55% of US adults with type 1 diabetes are cur- formed at randomization. C-peptide (Cobas 6000; Roche Diagnostics
rently treated with CSII and the number is increasing.5,6 However, GmbH, Mannheim, Germany) and plasma glucose (Cobas 6000; Roche
more than two-thirds of adults with type 1 diabetes do not achieve Diagnostics GmbH) were measured at time −10, 0, 30, 60, 90 and
the recommended glycaemic target [haemoglobin A1c (HbA1c) 120 min following ingestion of a standardized liquid mixed meal
<53 mmol/mol (7.0%)] despite improved insulin-delivery systems and (Boost Original; Nestle HealthCare Nutrition, NJ, USA).
insulin analogues with optimized pharmacokinetic characteristics com- After randomization, one telephone contact (week 1) and three
bined with improved methods for self-glucose monitoring.4,5 clinic visits were scheduled (weeks 3, 13 and 26).
Insulin treatment has several shortcomings and does not address During weeks 0, 3, 13 and 26, participants wore a blinded contin-
important abnormalities in type 1 diabetes. First, the gastric emptying rate uous glucose monitor (CGM) (Ipro2; Medtronic, Minneapolis, MN,
may be altered depending on the preprandial blood glucose level resulting USA) for six consecutive days.
in rapid and large postprandial glucose excursions.7 Second, the absence The trial was approved by the Scientific-Ethical Committee of the
of adequate portal insulin levels results in enhanced fasting and postpran- Capital Region of Denmark (H-3-2014-094), the Danish Medicines Author-
dial glucagon levels contributing to hyperglycaemia.8 Third, intensified ity (EudraCT: 2014–002285-76), and the Data Protection Agency. Further-
treatment with MDI or CSII is associated with weight gain. Glucagon-like more, the trial was registered with ClinicalTrials.gov (NCT02351232) and
peptide-1 receptor agonists (GLP-1RAs) could potentially address these conducted under the surveillance and guidance of the Good Clinical Prac-
shortcomings because of their pleiotropic effects of inducing satiety, tice unit, University of Copenhagen, Copenhagen, Denmark.
inhibiting glucagon secretion and delaying gastric emptying rate.
In patients with MDI-treated type 1 diabetes the efficacy has only
been modest with A1c reductions of about 2–3 mmol/mol (0.2%–0.3%) 2.2 | Trial participants
compared with placebo, but clinically significant weight loss of 4–6 kg
and reduced insulin requirements of 5% have been reported.9-13 Patients eligible for inclusion were adults with an age between 18 and
We investigated whether adding liraglutide 1.8 mg once daily 70 years and clinically diagnosed with type 1 diabetes ≥12 months
(QD) to CSII treatment could improve glycaemic control and reduce before the screening visit. Patients had to be treated with CSII, in
weight in overweight patients with type 1 diabetes. non-optimal glycaemic control with an HbA1c >58 mmol/mol (7.5%)
and use carbohydrate counting and the insulin pump bolus calculator
for all meals. No upper limit of HbA1c was specified. Finally, partici-
2 | MATERIALS AND METHODS pants had to be overweight or obese with a body mass index (BMI)
>25 kg/m2.
2.1 | Trial design Key exclusion criteria included the use of medicine affecting glu-
cose metabolism (other than insulin) during the study period or within
The trial was a 26-week randomized, double-blinded, placebo- 30 days before the screening; impaired renal or liver function (esti-
controlled trial performed at the Steno Diabetes Center Copenhagen, mated glomerular filtration rate <60 mL/min/1.73 m2 or alanine ami-
Gentofte, Denmark and Copenhagen University Hospital Hvidovre, notransferase >2.5 the upper limit of the reference interval,
Hvidovre, Denmark. After written informed consent was obtained and respectively), history of acute or chronic pancreatitis and people with
the screening visit (week −1) performed, eligible patients were ran- personal or family history of medullary carcinoma of the thyroid or
domized (week 0) 1:1 to receive liraglutide 1.8 mg (Novo Nordisk A/S, multiple endocrine neoplasia syndrome type 2. People with a history
Maalev, Denmark) or a corresponding volume of placebo (saline injec- of severe hypoglycaemia, hypoglycaemia unawareness or people with
tion) QD by a placebo pen that was indistinguishable from the a history of ketoacidosis were not excluded.
liraglutide pen. The treatment was initiated with an injection of 0.6 mg
liraglutide/0.1 mL placebo QD, increased to 1.2 mg liraglutide/0.2 mL
placebo QD after 1 week and increased to 1.8 mg/0.3 mL placebo 2.3 | Outcome measures
after another week. Intervals between the increments could be
extended at the discretion of the investigator. To reduce the risk of The primary objective was to study the efficacy of liraglutide 1.8 mg
hypoglycaemia the basal insulin rate was reduced by 10% and the QD added to CSII treatment on glycaemic control measured by the
DEJGAARD ET AL. 3

change in HbA1c from baseline to end of treatment at week 26. Sec- TABLE 1 Baseline characteristics
ondary objectives included the efficacy of liraglutide from baseline to Liraglutide
Placebo
week 26 on total insulin dose, including basal and bolus insulin, insulin 1.8 mg
pump settings, glycaemic variability and percentage of time spent in
(N = 22) (N = 22)
hypoglycaemia [blood glucose <4 mmol/L (71 mg/dL)], target range
Age (years) 50 ± 14 43 ± 12
[blood glucose 4–10 mmol/L (71–180 mg/dL)] and hyperglycaemia
Female sex 68% 68%
[>10 mmol/L (180 mg/dL)] evaluated by six consecutive days of
Duration of type 1 diabetes (years) 21 (15; 34) 20 (15; 35)
blinded CGM, lipids and body weight. Change in treatment satisfac-
tion [Diabetes Treatment Satisfaction Questionnaire (DTSQ)] and CSII treatment (years) 5 (3; 7) 6 (5; 9)

symptoms of depression [Hospital Anxiety and Depression Scale HbA1c (%) 8.2 ± 0.5 8.1 ± 0.5

(HADS)] were evaluated. HbA1c (mmol/mol) 66 ± 6 66 ± 6


Key safety assessments included adverse events (AEs), frequency Stimulated C-peptide (pmol/L) 9 (7; 18) 9 (6; 12)
of mild and severe hypoglycaemia as defined by the American Diabe- Stimulated C-peptide <30 pmol/L, n (%) 19 (86) 21 (95)
tes Association,4 heart rate, systolic and diastolic blood pressure and CSII total daily insulin dose (units) 48 ± 15 54 ± 19
laboratory safety variables, including amylase and lipase. To evaluate Total basal dose (units) 25 ± 9 28 ± 7
the effect of liraglutide on kidney function by serum creatinine and Total bolus dose (units) 23 ± 9 26 ± 14
urine albumin-to-creatinine ratio (UACR), first morning voids of three
Body weight (kg) 85 ± 10 88 ± 14
consecutive days before randomization, visit 3 (week 13) and end-of- 2
BMI (kg/m ) 30 ± 2 29 ± 3
treatment (week 26) were collected.
BMI 27–30 kg/m2, n (%) 9 (41) 10 (45)
BMI >30 kg/m2, n (%) 10 (45) 8 (36)
DTSQs 32 (31; 34) 31 (28; 34)
2.4 | Statistical analyses
Blood pressure (mmHg)

The sample size was determined to detect a minimum difference in Systolic 129 ± 18 127 ± 15

HbA1c of 0.3% (3 mmol/mol) between liraglutide 1.8 mg and placebo Diastolic 80 ± 9 82 ± 9


after 26 weeks of treatment with a power of 80%, a 5% significance Heart rate (b.p.m.) 70 ± 13 72 ± 12
level and a standard deviation of 0.3% (3 mmol/mol) based on data UACR (mg/g) 3 (2; 5) 3 (2; 7)
obtained from an observational study with GLP-1RA added to CSII
Abbreviations: BMI, body mass index; CSII, continuous subcutaneous
treatment in persons with type 1 diabetes.14 To allow for a 22% drop- insulin infusion; DTSQ, Diabetes Treatment Satisfaction Questionnaire;
out rate, 44 participants were included in the study, 22 in each arm. HbA1c, haemoglobin A1c; UACR, urine albumin-to-creatinine ratio.
Baseline characteristics are presented as means ± standard devia-
tions or as medians with interquartile range for characteristics with a
skewed distribution (Table 1). values for secondary endpoints were adjusted for multiple compari-
During follow-up, the primary outcome HbA1c was measured at sons using Benjamin and Hochberg. After adjustment, the found asso-
two visits (13 and 26 weeks) after randomization. Secondary out- ciations remained statistically significant.
comes were additionally measured at week 3 and insulin doses were Statistical analyses were conducted in R version 3.3.3 (The R
further assessed at weeks 1, 2, 6.5 and 19.5 after randomization. For Foundation for Statistical Computing, http://www.R-project.org) and
the analysis of change in total insulin per body weight, weight at SAS version 9.4 (SAS Institute, Cary, NC, USA).
weeks 1, 2, 6.5 and 19.5 was interpolated from weight at randomiza-
tion and weeks 3, 13 and 26, respectively.
In the repeated measurement analysis, changes in primary out- 3 | RE SU LT S
come HbA1c and secondary outcomes over the intervention period
were modelled by linear mixed-effects models with an individual- From February 17, 2015 to April 4, 2016, 171 patients with CSII-
specific random intercept to account for the correlation of repeated treated type 1 diabetes were assessed, of whom 54 were screened
measurements within participants. Treatment groups and follow-up and 44 persons were randomized for the study and exposed to the
visits (weeks 0, 1, 2, 3, 6.5, 13, 19.5 and 26) were used as fixed effects trial product (Figure S1; see Supporting Information). We identified no
in the analyses. We tested for a difference in change between the differences in baseline characteristics in relation to gender, age, diabe-
treatment groups at the follow-up visits. tes duration, HbA1c and BMI between individuals who did or did not
All analyses were performed as an intention-to-treat analysis agree to participate (data not shown). During the 26 weeks of inter-
(i.e. including all available measurements for the participants). Out- vention, all persons assigned to the placebo completed the study;
comes with a skewed distribution were log-transformed before ana- however, two people assigned to receive liraglutide discontinued the
lyses to fulfil the assumption of normally distributed model residuals. trial due to AEs not related to the trial product (i.e. breast cancer and
Statistical significance was inferred at a two-tailed P < 0.05. All P- multiple sclerosis; 3 and 20 weeks after randomization, respectively).
4 DEJGAARD ET AL.

Baseline characteristics were similar between groups (Table 1). significantly more in persons treated with liraglutide compared with
placebo (P = 0.005; Table 2). No differences between groups were
found in relation to the perceived frequency of hypoglycaemia or
3.1 | Efficacy hyperglycaemia during the intervention. No significant treatment dif-
ferences between liraglutide and placebo were found in the DTSQs
After 26 weeks of treatment the HbA1c level decreased from and the HADS questionnaires.
66 mmol/mol (8.2%) to 61 mmol/mol (7.7%) (P < 0.001) for persons In a post hoc analysis we adjusted the change in HbA1c for the
treated with liraglutide while no significant change was found with change in bodyweight, BMI and the total insulin dose, but no signifi-
placebo 66 mmol/mol (8.1%) to 68 mmol/mol (8.3%) (P = 0.058). Thus, cant changes compared with the unadjusted data were found.
the difference between groups at end of treatment was 7 mmol/mol
(0.7%) (P < 0.001). The significant difference between groups was
already present after 13 weeks of treatment (Table 2 and Figure 1). At 3.2 | Safety
end-of-treatment, 20% versus 5% achieved an HbA1c <53 mmol/mol
(7%) in the liraglutide versus placebo group, respectively. The number of events of documented self-monitored blood glucose
At baseline, the absolute time spent in hypoglycaemia, target range <3.9 mmol/L (70 mg/dL) or <3.0 mmol/L (54 mg/dL) were 1736 and
and hyperglycaemia evaluated by blinded CGM did not differ between 739 in total over the 26-week follow-up period with no difference in
groups. However, after 26 weeks of treatment persons treated with the rate of events between the treatment groups (<3.9: liraglutide
liraglutide had more time in target range compared with placebo, 57% 83 events per person-year and placebo 74 events per person-year,
versus 45% (P = 0.044). This difference was a consequence of less time P = 0.695; <3.0: liraglutide 30 and placebo 37, P = 0.494).
spent in hyperglycaemia for persons treated with liraglutide (Figure S2; Similarly, when evaluating the percentage of time spent in
see Supporting Information). Furthermore, glycaemic variability esti- hypoglycaemia level 1 [glucose value <4.0 mmol/L (71 mg/dL)]
mated by the standard deviation of mean glucose significantly and hypoglycaemia level 2 [glucose value <3.0 mmol/L (54 mg/dL)],
decreased in persons treated with liraglutide compared with placebo hypoglycaemia during three periods of six consecutive days of blinded
after 13 weeks of treatment; however, at 26 weeks no significant dif- CGM, no difference was found between liraglutide and placebo
ference was found between groups. The coefficient of variation of glu- (6% vs. 6% of time spent in level 1 and 1% vs. 2% of time in level
cose did not change at any time point in the groups (Table 2). 2, respectively).
The total daily insulin dose decreased in persons treated with Ambulatory systolic blood pressure decreased 7 mmHg shortly
liraglutide compared with placebo (P = 0.008). This difference was after initiating liraglutide treatment, while no significant change was
mainly due to a difference between groups of 7 IU/day of bolus insu- found in persons treated with placebo, resulting in a difference
lin after 13 weeks and 8 IU/day at end of treatment. When adjusted between groups of 10 mmHg. However, after 26 weeks of treatment
for body weight no significant change was found in either the total this effect was diminished, and no difference was found between
bolus or the total basal insulin doses in the groups during the study groups (P = 0.340). Concomitantly with the initial drop in systolic
(Table 2). Concomitantly, the total daily carbohydrate intake after blood pressure, ambulatory heart rate increased significantly, with
13 weeks decreased by 32 g/day in patients treated with liraglutide 11 beats per min (b.p.m.) compared with 5 b.p.m. in persons treated
compared with placebo (P = 0.031). However, at end of study this dif- with placebo. At the end of treatment, a difference between groups
ference decreased to 20 g/day and no difference was found between of 8 b.p.m. was found (P = 0.008). Ambulatory diastolic blood pressure
groups (P = 0.188; Table S1, see Supporting Information). measurements did not change at any time point in the two groups.
The insulin sensitivity factors and the insulin-to-carbohydrate In relation to the effect of liraglutide treatment on kidney func-
ratios between 06:00 and 11:00 h, between 11:00 and 16:00 h and tion, no significant effect on serum-creatinine or UACR was found
between 16:00 and 24:00 h (weighted means) did not change within during the 26 weeks of treatment.
or between groups after 26 weeks of treatment. Mean lipase levels were approximately 20 U/L at randomization
No differences within or between groups were found in total cho- and increased with liraglutide treatment to a difference between
lesterol, triglycerides, low-density lipoprotein and high-density lipo- groups of 32 U/L, P = 0.023. Similarly, the mean amylase levels were
protein at any time point. initially 14 U/L with an increase in the group treated with liraglutide,
Compared with placebo, liraglutide led to a significant reduction resulting in a difference between groups of 22 units/L (P = 0.019). No
in body weight from baseline to week 26 (Figure 1C). Already after cases of pancreatitis were reported.
13 weeks of treatment a reduction of 5.2 kg was found in persons The overall rate of AEs was more common in persons treated
treated with liraglutide (P < 0.001), increasing to −6.8 kg after with liraglutide compared with placebo (90% vs. 59%). Most of the
26 weeks (P < 0.001) compared with 0.4 kg (P = 0.491) for placebo, AEs were mild in severity and, in general, liraglutide treatment was
with a difference between groups of −6.3 kg (P < 0.001) (Table 2). well tolerated. Gastrointestinal AEs were reported more frequently in
Liraglutide treatment positively affected patient-reported out- persons treated with liraglutide compared with placebo (nausea 64%
come measures. The mean change in treatment satisfaction scores vs. 32%, diarrhoea 18% vs. 9% and vomiting 18% vs. 5%). Although
assessed with the DTSQ version c increased in both groups; however, gastrointestinal AEs were transient, five patients who received
TABLE 2 Estimated changes from baseline in primary and secondary outcomes by treatment group

13 weeks 26 weeks
Liraglutide Placebo Liraglutide Liraglutide Placebo Liraglutide vs. placebo
DEJGAARD ET AL.

vs. placebo
Change P Change P Change Pdiff Change P Change P Change P–diff
HbA1c (mmol/mol) −5.2 (−7.7; −2.7) <0.001 1.7 (−0.8; 4.1) 0.182 −6.9 (−10.4; −3.4) <0.001 −5.0 (−7.5; −2.4) <0.001 2.3 (−0.1; 4.7) 0.058 −7.3 (−10.8; −3.8) <0.001
HbA1c (% point) −0.5 (−0.7; −0.2) <0.001 0.2 (−0.1; 0.4) 0.182 −0.6 (−0.9; −0.3) <0.001 −0.5 (−0.7; −0.2) <0.001 0.2 (0.0; 0.4) 0.058 −0.7 (−1.0; −0.3) <0.001
Weight (kg) −5.2 (−6.5; −3.9) <0.001 0.8 (−0.5; 2.0) 0.244 −5.9 (−7.8; −4.1) <0.001 −6.8 (−8.1; −5.4) <0.001 −0.4 (−1.7; 0.8) 0.491 −6.3 (−8.2; −4.5) <0.001
Total daily insulin dose −4.4 (−8.5; −0.3) 0.037 2.2 (−1.7; 6.1) 0.273 −6.6 (−12.2; −0.9) 0.024 −4.9 (−9.1; −0.7) 0.021 2.8 (−1.1; 6.7) 0.164 −7.7 (−13.5; −2) 0.008
(IU/day)
Basal insulin (IU/day) −1.2 (−2.7; 0.2) 0.098 −0.4 (−1.8; 1.0) 0.551 −0.8 (−2.9; 1.2) 0.434 −1.8 (−3.3; −0.3) 0.017 −0.5 (−1.9; 0.9) 0.473 −1.3 (−3.4; 0.8) 0.214
Daily basal insulin −0.7 (−2; 0.7) 0.331 0.5 (−0.8; 1.8) 0.454 −1.1 (−3.0; 0.7) 0.223 −0.9 (−2.3; 0.4) 0.164 0.7 (−0.5; 1.9) 0.259 −1.7 (−3.5; 0.2) 0.074
06:00–24:00 h (IU/day)
Nightly basal insulin −0.1 (−0.5; 0.3) 0.587 −0.1 (−0.5; 0.3) 0.546 0.0 (−0.5; 0.5) 0.976 −0.1 (−0.5; 0.3) 0.579 −0.1 (−0.5; 0.2) 0.411 0.0 (−0.5; 0.6) 0.882
00:00–06:00 h (IU/day)
Bolus insulin (IU/day) −3.4 (−7.1; 0.3) 0.074 2.6 (−0.9; 6.2) 0.15 −6.0 (−11.1; −0.9) 0.022 −3.1 (−6.8; 0.7) 0.113 3.3 (−0.3; 6.9) 0.069 −6.4 (−11.6; −1.2) 0.016
CGM high (% point) −10.2 (−17.2; −3.3) 0.004 1.7 (−5.2; 8.6) 0.627 −12.0 (−21.8; −2.2) 0.017 −7.2 (−14.6; 0.1) 0.053 2.7 (−4.1; 9.5) 0.441 −9.9 (−19.9; 0.1) 0.052
CGM target (% point) 10.1 (3.8; 16.5) 0.002 −3.5 (−9.8; 2.8) 0.277 13.6 (4.7; 22.6) 0.003 4.6 (−2.1; 11.3) 0.178 −4.8 (−11.0; 1.5) 0.133 9.4 (0.2; 18.5) 0.044
CGM low (% point) 0.1 (−2.2; 2.4) 0.935 1.8 (−0.5; 4.1) 0.131 −1.7 (−5.0; 1.6) 0.313 2.5 (0.1; 5.0) 0.044 2.1 (−0.2; 4.4) 0.077 0.5 (−2.9; 3.8) 0.787
CGM mean glucose −0.8 (−1.5; −0.2) 0.013 0.2 (−0.4; 0.9) 0.518 −1.1 (−2.0; −0.1) 0.027 −0.7 (−1.4; 0.0) 0.064 0.3 (−0.4; 0.9) 0.394 −0.9 (−1.9; 0.0) 0.053
CGM SD −0.5 (−0.9; −0.2) 0.003 0.3 (−0.1; 0.6) 0.124 −0.8 (−1.3; −0.3) 0.001 −0.2 (−0.6; 0.2) 0.285 0.2 (−0.2; 0.5) 0.337 −0.4 (−0.9; 0.1) 0.151
CGM CV (% point) −2.8 (−6.5; 0.8) 0.131 1.7 (−2.0; 5.4) 0.365 −4.6 (−9.8; 0.7) 0.087 0.0 (−3.9; 3.9) 0.983 0.8 (−2.8; 4.4) 0.664 −0.8 (−6.2; 4.5) 0.756
Systolic blood pressure −7.2 (−13.3; −1.2) 0.019 2.4 (−3.5; 8.3) 0.422 −9.7 (−18.1; −1.2) 0.025 −6.6 (−12.7; −0.4) 0.036 −2.5 (−8.3; 3.3) 0.403 −4.1 (−12.6; 4.3) 0.34
(mmHg)
Diastolic blood pressure −1.4 (−3.8; 1.1) 0.286 1.2 (−1.2; 3.6) 0.319 −2.6 (−6.0; 0.9) 0.144 −0.7 (−3.2; 1.8) 0.585 0.2 (−2.2; 2.6) 0.866 −0.9 (−4.4; 2.6) 0.608
(mmHg)
Heart rate (beats/min) 10.9 (6.7; 15.1) <0.001 4.6 (0.4; 8.7) 0.031 6.3 (0.4; 12.3) 0.036 9.0 (4.7; 13.3) <0.001 1.1 (−3.0; 5.1) 0.607 8.0 (2.0; 13.9) 0.008
DTSQs sumscore 0.0 (−1.8; 1.8) 0.988 −1.1 (−2.9; 0.6) 0.195 1.1 (−1.4; 3.6) 0.375 0.0 (−1.8; 1.9) 0.968 −1.4 (−3.2; 0.3) 0.105 1.5 (−1.1; 4.0) 0.253
DTSQs hyperglycaemia −1.0 (−1.6; −0.4) <0.001 0.2 (−0.4; 0.8) 0.445 −1.3 (−2.1; −0.4) 0.004 −0.8 (−1.4; −0.2) 0.01 0.0 (−0.6; 0.6) 0.999 −0.8 (−1.7; 0.0) 0.062
question 2
DTSQs hypoglycaemia −0.2 (−0.8; 0.5) 0.58 −0.5 (−1.2; 0.1) 0.095 0.3 (−0.5; 1.2) 0.447 0.4 (−0.2; 1.1) 0.198 0.0 (−0.6; 0.6) 0.979 0.4 (−0.5; 1.3) 0.36
question 3
DTSQc sumscore 13.4 (9.1; 19.7) <0.001 6.0 (4.0; 9.0) <0.001 2.2 (1.3; 3.9) 0.005
DTSQc hyperglycaemia 1.6 (1.2; 2.1) 0.002 1.3 (1.0; 1.6) 0.015 1.2 (0.9; 1.8) 0.225
(question 2)
DTSQc hypoglycaemia 1.1 (0.9; 1.4) 0.233 1.2 (0.9; 1.4) 0.156 1.0 (0.7; 1.3) 0.916
(question 3)

Abbreviations: CGM, continuous glucose monitor; DTSQ, Diabetes Treatment Satisfaction Questionnaire; HbA1c, haemoglobin A1c.
5

CGM high glucose value >180 mg/dL (>10 mmol/L). CGM target glucose value between 71 and 180 mg/dL (4.0–10.0 mmol/L). CGM low glucose value <71 mg/dL(<4 mmol/L).
6 DEJGAARD ET AL.

F I G U R E 1 Estimated levels
(±SEM) by treatment group
during follow-up. A, Haemoglobin
A1c. B, Total daily insulin dose. C,
Body weight. Stars indicate level
of significance test of difference
in baseline corrected change
between treatment groups:
***P <0.001 and *P <0.05

liraglutide had their dose temporarily reduced to 1.2 mg QD shortly treated with liraglutide had been exposed to a mean of 1.7 mg/day,
after escalation of the dose to 1.8 mg QD (range 6–34 days). One including the planned period for up-escalation.
patient who received liraglutide was unable to tolerate 1.8 mg QD Three severe AEs occurred (two in the liraglutide group vs. one in
and completed the trial on 1.2 mg QD. At end-of-treatment, patients the placebo group), including one case of severe hypoglycaemia in a
DEJGAARD ET AL. 7

patient who received placebo. No events of ketoacidosis were HbA1c compared with placebo when adding liraglutide to MDI.9-13
reported. Our population may have obtained a greater benefit of liraglutide due
A summary of AEs is reported in Table 3. to a more advanced insulin regimen at baseline. Furthermore, in the
present study, a small but insignificant increase in HbA1c of 2 mmol/
mol (0.2%) in the placebo group may be of importance for the results
4 | DISCUSSION when estimating the effect of liraglutide on HbA1c as a mean differ-
ence between treatment groups. In the previous studies evaluating
We demonstrated that the GLP-1RA liraglutide 1.8 mg QD as an add- liraglutide treatment in type 1 diabetes, the HbA1c reduction
on to CSII treatment in overweight or obese patients with type 1 dia- obtained with liraglutide has been in the range of 3–8 mmol/mol
betes and non-optimal glycaemic control reduced the HbA1c levels (0.3%–0.8%), which was similar to our findings, but a concomitant
without an increase in events of hypoglycaemia or time spent in reduction in HbA1c in the placebo arm has minimized the effect of
hypoglycaemia. Further, the total daily insulin dose and body weight liraglutide between groups.9-13
were significantly reduced. Liraglutide treatment positively affected In line with previous studies, treatment with a GLP-1RA reduced
patient-reported outcome measures compared with placebo. body weight progressively during the 26 weeks.9-13 The majority of
None of the patients included in the present study had reached patients included in this study were overweight at baseline, which
the recommended goals for glycaemic control with an HbA1c of increased the chance of obtaining a significant reduction in body
approximately 66 mmol/mol (8.1%–8.2%) when randomized. When weight. We obtained a total reduction of 8% of body weight in per-
adding liraglutide 1.8 mg QD to CSII therapy in this population, we sons treated with liraglutide, which is likely to be of clinical
demonstrated a clinically significant improvement in HbA1c of importance,15 as weight loss has been reported to improve quality of
−7.3 mmol/mol (−0.7%) when compared with placebo. This is in con- life and insulin sensitivity, and reduce cardiovascular risk factors in
trast to the majority of previous randomized studies reporting only people with type 1 diabetes.16-18
minor treatment effects of 2–3 mmol/mol (0.2%–0.3%) reduction in Even though patients were included with non-optimal glycaemic
control and significantly improved HbA1c during the study, the total
daily insulin was reduced by 17% when adding liraglutide. This differ-
TABLE 3 Adverse events
ence between groups disappeared when adjusting for body weight,
Liraglutide Placebo though the decrease in insulin may not only contribute to a reduction
1.8 mg (N = 22),
in hypoglycaemic events, but also have importance for the improve-
(N = 22), n (%) n (%)
ments in body weight. Despite a reduction of 10% in basal insulin
Patients with one or more adverse eventa 20 (91) 13 (59)
dose and an increase in insulin sensitivity factors and insulin-to-
≥1 adverse events*
carbohydrate ratios a significant reduction was seen in insulin dose
Nausea 16 (73) 9 (41)
within the first 2 weeks after randomization. This reduction in insulin
Decreased appetite 5 (23) 1 (5)
dose could be the result of the effect on gastric emptying and sup-
Diarrhoea 4 (18) 2 (9)
pression of postprandial glucagon in the early phase, independent of
Vomiting 3 (14) 0 (0)
the reduction in body weight, as the decline in body weight was more
Dizziness 3 (14) 0 (0) protracted. Furthermore, the reduction in both insulin dose and
Constipation 2 (9) 3 (14) bodyweight may be a consequence of the appetite-reducing effect of
Dyspepsia 1 (5) 1 (5) GLP-1 inducing a lower food volume and carbohydrate intake.19 The
Stomach ache 1 (5) 1 (5) direct effect of GLP-1RA on insulin resistance in peripheral tissue is
Fatigue 1 (5) 0 (0) limited and, if present, may be a result of reduction in adipose tissue
Headache 1 (5) 3 (14) in humans.19 Despite a reduction in body weight of 8%, no changes in
Allergic reaction not related to IMP 1 (5) 0 (0) CSII settings for insulin sensitivity factors were found. Concomitantly,

Inflammation in joint 1 (5) 0 (0) only a minor non-significant effect on insulin-to-carbohydrate ratios
was found during the intervention period. This may support the evi-
Pneumonia 1 (5) 0 (0)
dence that the effect of liraglutide on insulin resistance is sparse,
Urinary tract infection 0 (0) 2 (9)
though a direct measure of insulin resistance was not performed.
Epididymitis 0 (0) 1 (5)
Generally, liraglutide 1.8 mg was well tolerated. The most com-
Angina pectoris 1 (5) 0 (0)
mon AEs were gastrointestinal, primarily nausea. In most patients the
≥1 Serious adverse events
gastrointestinal side effects were transient, but five patients had to
Multiple scleroses 1 (5) 0 (0) reduce the liraglutide dose to 1.2 mg QD temporarily and one patient
Breast cancer 1 (5) 0 (0) completed the study on 1.2 mg QD. Notably, patients in the present
Severe hypoglycaemia 0 (0) 1 (5) study were overweight or obese. From previous studies, including
Abbreviations: IMP, investigational medicinal product. lean patients with type 1 diabetes, we have experienced that the
a
Events of hypoglycaemia are described separately. 1.2 mg QD of liraglutide is a more optimal dose than 1.8 mg QD in
8 DEJGAARD ET AL.

relation to minimizing gastrointestinal events in non-obese patients, CONFLIC T OF INT ER E ST


without compromising the effect on glycaemic control.11,20 Despite T.F.D. has received research support from Novo Nordisk and
more AEs in the liraglutide-treated patients, the improvement in treat- AstraZeneca; and has received lecture fees from Novo Nordisk,
ment satisfaction score increased more with liraglutide compared with AstraZeneca and Boehringer-Ingelheim. S.S. has served on an advisory
placebo. panel for Roche Diagnostics and has consulted for Unomedical.
As reported in previous trials investigating liraglutide, levels of C.S.F. has received research support and lecture fees from Novo
plasma amylase and lipase increased during the 6 months of treat- Nordisk. D.V. is a shareholder in Novo Nordisk. S.M. has served on
ment. In trials with longer duration, levels seemed to increase over the advisory panels for Novartis Pharma, Novo Nordisk, Merck Sharp &
first 6 months of treatment and then remain stable. The increases Dome, Sanofi-Aventis, AstraZeneca, Johnson & Johnson, Roche Diag-
seem to be dose-independent. Importantly, the increases are not pre- nostics, Mankind, Boehringer-Ingelheim, Zealand, Eli Lilly and Intarcia
12,21,22
dictive for events of subsequent acute pancreatitis. Therapeutics; and has received lecture fees from Novo Nordisk,
Compared with the ADJUNCT trials, where an increased number Merck Sharp & Dome, Astra-Zeneca, Johnson and Johnson, Roche,
of symptomatic or confirmed events of hypoglycaemia were reported Shering-Ploug, Sanofi-Aventis, Novartis Pharma, Eli Lilly and Bristol-
for patients treated with liraglutide 1.2 and 1.8 mg QD, we did not Meyer Squibb. H.U.A. is a shareholder of Novo Nordisk, has served on
find any difference between groups. This may be due to a less- advisory panels for Abbott, Novo Nordisk and AstraZeneca and has
intensive treat-to-target regimen with less contacts during the trial received lecture fees from Nordic Infucare. K.N. is a shareholder of
combined with a higher level of total insulin dose used in the Novo Nordisk; has received research support from Novo Nordisk,
ADJUNCT trials or the fact that this study was not powered for evalu- Roche Diagnostics and Zealand Pharma; has received lecture fees
9,12
ating events of hypoglycaemia. from Medtronic, Roche Diagnostics, Rubin Medical, Sanofi, Zealand
The strength of the present study is the double-blinded, placebo- Pharma, Novo Nordisk and Bayer; and has served on advisory panels
controlled design only including patients treated with CSII. We chose for Medtronic, Abbott and Novo Nordisk.
liraglutide 1.8 mg, the highest dose approved for the treatment of
type 2 diabetes, to bring about the largest effect on both glycaemic AUTHOR CONTRIBU TIONS
control and weight loss. The dropout rate was low. T.F.D., S.S. and C.S.F. contributed to the study design, data collection
A limitation to the present study is the relatively small number of and analysis. S.M., H.U.A. and K.N contributed to the study design
participants. In addition, the duration of 26 weeks does not provide and data collection. D.V. contributed to the data analysis. All authors
data on long-term efficacy and safety of the treatment and may not were involved in the writing of the manuscript and approved the final
have impact in a lifelong disease unless compliance is optimized. version of the manuscript. T.F.D. is the guarantor of this work and, as
In conclusion we demonstrated that adding liraglutide 1.8 mg QD such, had full access to all the data in the study and takes responsibil-
to CSII treatment in a population of overweight or obese persons with ity for the integrity of the data and the accuracy of the data analysis.
type 1 diabetes and insufficient glycaemic control induced a clinically
relevant improvement in HbA1c and increased the time spent in nor-
OR CID
moglycaemia with less need of insulin and no elevated risk of
Thomas F. Dejgaard https://orcid.org/0000-0002-0097-7052
hypoglycaemia compared with placebo. Furthermore, body weight
Signe Schmidt https://orcid.org/0000-0002-6968-6675
was significantly reduced. Liraglutide was well tolerated with transient
gastrointestinal AEs that were described during treatment with GLP-
RE FE RE NCE S
1RAs in patients with type 1 as well as type 2 diabetes.
1. Diabetes Control and Complications Trial Research Group,
The clinical use of GLP-1RAs may be limited in the general popu- Nathan DM, Genuth S, Lachin J, et al. The effect of intensive treat-
lation of persons with type 1 diabetes; however, our results with an ment of diabetes on the development and progression of long-term
attractive benefit–risk profile suggest that liraglutide may be a promis- complications in insulin-dependent diabetes mellitus. The Diabetes
Control and Complications Trial Research Group. N Engl J Med. 1993;
ing adjunct treatment in a subpopulation of obese patients treated
329:977-986.
with CSII and non-optimal glycaemic control. Nevertheless, specific 2. Chillaron JJ, Flores Le-Roux JA, Benaiges D, Pedro-Botet J. Type
clinical outcomes are still needed to predict the treatment effect in 1 diabetes, metabolic syndrome and cardiovascular risk. Metabolism.
this group of patients. 2014;63:181-187.
3. Pickup JC. Insulin-pump therapy for type 1 diabetes mellitus. N Engl J
Med. 2012;366:1616-1624.
ACKNOWLEDGMENTS 4. American Diabetes Association. 6. Glycemic targets: Standards of
The authors thank the staff at the Clinical Research Unit, Steno Diabe- Medical Care in Diabetes—2018. Diabetes Care. 2018;41(Suppl. 1):
tes Center Copenhagen and the Department of Endocrinology, S55-S64.
5. Foster NC, Beck RW, Miller KM, et al. State of type 1 diabetes man-
Copenhagen University Hospital Hvidovre for assistance with collect-
agement and outcomes from the T1D exchange in 2016-2018. Diabe-
ing data and laboratory work. In particular, we are grateful to study tes Technol Ther. 2019;21:66-72.
nurse Annette Hougaard and Merete Meldgaard Andersen for an unri- 6. Miller KM, Foster NC, Beck RW, et al. Current state of type 1 diabetes
valled commitment to the study. Finally, we thank the patients for treatment in the US: updated data from the T1D Exchange clinic reg-
their participation in this trial. istry. Diabetes Care. 2015;38:971-978.
DEJGAARD ET AL. 9

7. Phillips LK, Deane AM, Jones KL, Rayner CK, Horowitz M. Gastric type 1 diabetes on lipid levels and blood pressure: results from the
emptying and glycaemia in health and diabetes mellitus. Nat Rev DCCT. Diabetes Control and Complications Trial. JAMA. 1998;280:
Endocrinol. 2015;11:112-128. 140-146.
8. Greenbaum CJ, Prigeon RL, D'Alessio DA. Impaired beta-cell function, 18. Vitola BE, Deivanayagam S, Stein RI, et al. Weight loss reduces liver
incretin effect, and glucagon suppression in patients with type 1 dia- fat and improves hepatic and skeletal muscle insulin sensitivity in
betes who have normal fasting glucose. Diabetes. 2002;51:951-957. obese adolescents. Obesity (Silver Spring). 2009;17:1744-1748.
9. Ahren B, Hirsch IB, Pieber TR, et al. Efficacy and safety of liraglutide 19. Drucker DJ. Mechanisms of action and therapeutic application of
added to capped insulin treatment in subjects with type 1 diabetes: glucagon-like peptide-1. Cell Metab. 2018;27:740-756.
the ADJUNCT TWO Randomized Trial. Diabetes Care. 2016;39:1693- 20. Kielgast U, Krarup T, Holst JJ, Madsbad S. Four weeks of treatment
1701. with liraglutide reduces insulin dose without loss of glycemic control
10. Dejgaard TF, Frandsen CS, Hansen TS, et al. Efficacy and safety of in type 1 diabetic patients with and without residual beta-cell func-
liraglutide for overweight adult patients with type 1 diabetes and tion. Diabetes Care. 2011;34:1463-1468.
insufficient glycaemic control (Lira-1): a randomised, double-blind, 21. Steinberg WM, Buse JB, Ghorbani MLM, et al. Amylase, lipase, and
placebo-controlled trial. Lancet Diabetes Endocrinol. 2016;4:221-232. acute pancreatitis in people with type 2 diabetes treated with
11. Frandsen CS, Dejgaard TF, Holst JJ, Andersen HU, Thorsteinsson B, liraglutide: results from the LEADER randomized trial. Diabetes Care.
Madsbad S. Twelve-week treatment with liraglutide as add-on to 2017;40:966-972.
insulin in normal-weight patients with poorly controlled type 1 diabe- 22. Steinberg WM, Rosenstock J, Wadden TA, Donsmark M, Jensen CB,
tes: a randomized, placebo-controlled, double-blind parallel study. DeVries JH. Impact of liraglutide on amylase, lipase, and acute pan-
Diabetes Care. 2015;38:2250-2257. creatitis in participants with overweight/obesity and normoglycemia,
12. Mathieu C, Zinman B, Hemmingsson JU, et al. Efficacy and safety of prediabetes, or type 2 diabetes: secondary analyses of pooled data
liraglutide added to insulin treatment in type 1 diabetes: the from the SCALE Clinical Development Program. Diabetes Care. 2017;
ADJUNCT ONE treat-to-target randomized trial. Diabetes Care. 2016; 40:839-848.
39:1702-1710.
13. Kuhadiya ND, Dhindsa S, Ghanim H, et al. Addition of liraglutide to
insulin in patients with type 1 diabetes: a randomized placebo- SUPPORTING INF ORMATION
controlled clinical trial of 12 weeks. Diabetes Care. 2016;39:1027- Additional supporting information may be found online in the
1035.
Supporting Information section at the end of this article.
14. Kuhadiya ND, Malik R, Bellini NJ, et al. Liraglutide as additional treat-
ment to insulin in obese patients with type 1 diabetes mellitus. Endocr
Pract. 2013;19:963-967.
15. Conway B, Miller RG, Costacou T, et al. Temporal patterns in over- How to cite this article: Dejgaard TF, Schmidt S, Frandsen CS,
weight and obesity in Type 1 diabetes. Diabet Med. 2010;27: et al. Liraglutide reduces hyperglycaemia and body weight in
398-404. overweight, dysregulated insulin-pump-treated patients with
16. Hassan MK, Joshi AV, Madhavan SS, Amonkar MM. Obesity and
type 1 diabetes: The Lira Pump trial—a randomized, double-
health-related quality of life: a cross-sectional analysis of the US pop-
ulation. Int J Obes Relat Metab Disord. 2003;27:1227-1232. blinded, placebo-controlled trial. Diabetes Obes Metab. 2019;
17. Purnell JQ, Hokanson JE, Marcovina SM, Steffes MW, Cleary PA, 1–9. https://doi.org/10.1111/dom.13911
Brunzell JD. Effect of excessive weight gain with intensive therapy of

Vous aimerez peut-être aussi