Skip to main content
Erschienen in: Diabetologia 12/2016

Open Access 15.09.2016 | Short Communication

Twelve week liraglutide or sitagliptin does not affect hepatic fat in type 2 diabetes: a randomised placebo-controlled trial

verfasst von: Mark M. Smits, Lennart Tonneijck, Marcel H. A. Muskiet, Mark H. H. Kramer, Petra J. W. Pouwels, Indra C. Pieters-van den Bos, Trynke Hoekstra, Michaela Diamant, Daniël H. van Raalte, Djuna L. Cahen

Erschienen in: Diabetologia | Ausgabe 12/2016

download
DOWNLOAD
print
DRUCKEN
insite
SUCHEN

Abstract

Aims/hypothesis

Glucagon-like peptide (GLP)-1-based therapies have been suggested to improve hepatic steatosis. We assessed the effects of the GLP-1 receptor agonist liraglutide and the dipeptidyl peptidase (DPP)-4 inhibitor sitagliptin on hepatic steatosis and fibrosis in patients with type 2 diabetes.

Methods

In this 12 week, parallel, randomised, placebo-controlled trial, performed at the VU University Medical Center between July 2013 and August 2015, 52 overweight patients with type 2 diabetes treated with metformin and/or sulphonylurea agent ([mean ± SD] age 62.7 ± 6.9 years, HbA1c 7.3 ± 0.7% or 56 ± 1 mmol/mol) were allocated to once daily liraglutide 1.8 mg (n = 17), sitagliptin 100 mg (n = 18) or matching placebos (n = 17) by computer generated numbers. Both participants and researchers were blinded to group assignment. Hepatic fat content was measured using proton magnetic resonance spectroscopy (1H-MRS). Hepatic fibrosis was estimated using three validated formulae.

Results

One patient dropped out in the sitagliptin group owing to dizziness, but no serious adverse events occurred. At week 12, no between-group differences in hepatic steatosis were found. Liraglutide reduced steatosis by 10% (20.9 ± 3.4% to 18.8 ± 3.3%), sitagliptin reduced steatosis by 12.1% (23.9 ± 3.0% to 21.0 ± 2.7%) and placebo lessened it by 9.5% (18.7 ± 2.7% to 16.9 ± 2.7%). Neither drug affected hepatic fibrosis scores compared with placebo.

Conclusions/interpretation

Twelve-week liraglutide or sitagliptin treatment does not reduce hepatic steatosis or fibrosis in type 2 diabetes.

Trial registration

ClinicalTrials.gov NCT01744236

Funding

Funded by the European Community’s Seventh Framework Programme (FP7/2007-2013) under grant agreement no. 282521 – the SAFEGUARD project.
Begleitmaterial
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1007/​s00125-016-4100-7) contains peer-reviewed but unedited supplementary material, which is available to authorised users.
Professor Michaela Diamant, whose experience and expertise were crucial for the design of this study, died before publication of this work.
Abkürzungen
1H-MRS
Hydrogen magnetic resonance spectroscopy
γGT
Gamma glutamyl transferase
ALP
Alkaline phosphatase
ALT
Alanine aminotransferase
APRI
Aspartate aminotransferase to platelet ratio index
AST
Aspartate aminotransferase
DPP-4
Dipeptidyl peptidase-4
FIB-4
Fibrosis-4
GLP-1
Glucagon-like peptide-1
L-FABP
Liver fatty acid binding protein
NAFLD
Non-alcoholic fatty liver disease
NASH
Non-alcoholic steatohepatitis
NFS
NAFLD fibrosis score
VOI
Volume of interest

Introduction

Non-alcoholic fatty liver disease (NAFLD) is the most common chronic liver condition in the developed world, affecting over 30% of the population [1]. Its prevalence is particularly high among patients with type 2 diabetes mellitus (>70%), probably because of insulin resistance as a common denominator. NAFLD may progress to non-alcoholic steatohepatitis (NASH), cirrhosis and hepatocellular carcinoma, and is an independent risk factor for cardiovascular disease [1]. As such, it is associated with increased liver-related and all-cause mortality.
Lifestyle modifications, including dietary changes and weight loss, are the mainstay of NAFLD management, yet most patients with type 2 diabetes do not achieve or maintain their goals [1]. Moreover, although several pharmacological substances have been explored with promising results, including pioglitazone (reductions of up to approximately 50%), vitamin E and obeticholic acid [1], no drug is currently licensed for NAFLD. With an expected increase in prevalence of type 2 diabetes, NAFLD and consequent health risks, the search for effective and safe therapeutic strategies is ongoing.
Glucagon-like peptide (GLP)-1 receptor agonists and dipeptidyl peptidase (DPP)-4 inhibitors have been postulated as treatment options for NAFLD because of their positive effects on glycaemic control, body weight, insulin resistance, lipid metabolism and inflammation [2]. Encouraged by several experimental and open-label uncontrolled clinical studies [2], the current placebo-controlled randomised trial assessed the effects of the GLP-1 receptor agonist liraglutide and the DPP-4 inhibitor sitagliptin on spectroscopy-measured hepatic steatosis in patients with type 2 diabetes.

Methods

Patients were evaluated in a 12-week, single-centre, randomised, placebo-controlled, double-blind, double-dummy, three-armed, parallel-group intervention trial. The study was approved by the local ethics review board, registered at ClinicalTrials.gov (NCT01744236) and conducted in accordance with the Declaration of Helsinki and the International Conference on Harmonization of Good Clinical Practice. All participants provided written informed consent before participation. This study was part of a larger trial performed between July 2013 and August 2015 at the VU University Medical Center, Amsterdam, the Netherlands, whose study objectives and protocol have previously been published [3]. Here, the effects of GLP-1-based therapies on the secondary endpoints of hepatic steatosis (measured using proton-magnetic resonance spectroscopy [1H-MRS]), hepatic fibrosis (assessed by fibrosis formulae) and hepatic function (from measurements of serum albumin and bilirubin) are reported.
Study population
Patients with type 2 diabetes were eligible if they were aged between 35 and 75 years (and women were postmenopausal), had an HbA1c level of 6.5–9.0% (48–75 mmol/mol), been treated with a stable dose of metformin and/or sulfonylurea derivatives for ≥3 months and had a BMI of 25–40 kg/m2. Relevant exclusion criteria were the use of GLP-1-based therapies or insulin, a history of hepatic or pancreatic disease, inability to undergo MRI scanning and alcohol intake >3 units/day [3].
Intervention
After inclusion, a 4-week run-in period and baseline testing, patients were randomised by the trial pharmacist using computer-generated numbers (allocation 1:1:1, block size six) to receive the GLP-1 receptor agonist liraglutide 1.8 mg (Novo Nordisk A/S, Bagsvaerd, Denmark), the DPP-4 inhibitor sitagliptin 100 mg (Merck, Kenilworth, NJ, USA) or matching placebos, taken once daily in the evening. Endpoint measurements were repeated after 12 weeks of treatment.
Endpoint measurements
Hepatic fat content was measured using proton 1H-MRS on a 1.5 T whole-body MRI scanner (Magnetom Avanto; Siemens Medical Solutions, Erlangen, Germany), with patients in the supine position and the body-array coil positioned at the upper abdominal region. Coronal and transverse structural T2-weighted images were used to localise a volume of interest (VOI). An 8 cm3 VOI (2 × 2 × 2 cm3) was selected at up to three locations in the liver (right superior, right inferior and left anterior), avoiding major blood vessels and bile ducts, and with sufficient distance from the liver edges. Using point-resolved spectroscopy sequences (echo time 30 ms, retention time 2000 ms, during free breathing, no gating), single voxel spectra were recorded. Eight acquisitions were obtained per VOI, and stored separately. User-independent spectral quantification was performed with LCModel (version 6.1; available from http://​s-provencher.​com).
Fat content was expressed as the percentage of the area under the methyl (0.9 ppm) and methylene (1.3 ppm) peaks, relative to the area under the water (4.65 ppm) peak. For each VOI, individual acquisitions were reviewed in order to discard occasional poor-quality spectra (e.g. due to motion artefacts). Individual spectra were combined to obtain the fat percentage for each VOI. The mean fat content of the available VOIs was used. Variation was assessed in 11 patients who underwent an additional baseline 1H-MRS session within 3 weeks as part of a different study [3]. The within-VOI variation was 6.7%, the between-VOI variation 12.2% and the variation in liver fat (combination of three VOIs; day-to-day variance) 11.7%.
Hepatic fibrosis was estimated at baseline and 12 weeks using validated formulae: the NAFLD fibrosis score (NFS), Fibrosis-4 (FIB-4) score and aspartate aminotransferase to platelet ratio index (APRI) [4]. Fasting blood samples were drawn for measurement of glucose (gluco quant-hexokinase method), HbA1c (HPLC), insulin (immunometric method), (cholestatic) liver enzymes (enzymatic assessment), albumin, total and conjugated bilirubin (colorimetric measurement), thrombocyte count (laser light scattering) and liver fatty acid binding protein (L-FABP; using sandwich ELISA).
Sample size, data management and statistical analysis
Sample size calculations have previously been published [3]. With an expected reduction in the percentage of steatosis of approximately 50% [5], a total of 13 patients per treatment arm were needed (parallel-group design, α = 0.05, power [1 − β] 80%). To test treatment effects vs placebo, multivariable regression analyses were performed on the per-protocol population, using SPSS 22 (IBM SPSS, Chicago, IL, USA). Treatment with liraglutide or sitagliptin was added as a dummy variable. To correct for baseline differences, pretreatment values were included in the model. Moreover, proportions of patients with any improvement in steatosis in each treatment group were compared using the χ 2 test. Analyses were repeated in the subgroup with NAFLD (>5.56% steatosis) at baseline. A two-sided p ≤ 0.05 was considered to be statistically significant.

Results

Of the 52 patients who were randomised, one patient in the sitagliptin group withdrew from the study because of adverse effects (dizziness and daytime urinary frequency). Fifty-one patients completed the 12 week study (17 per treatment arm) (see electronic supplementary material [ESM] Fig. 1). The baseline characteristics were similar between the groups (Table 1). Liraglutide and sitagliptin reduced fasting glucose (−1.6 ± 0.5 mmol/l and −1.8 ± 0.5 mmol/l, respectively; both p < 0.001) and HbA1c (−1.3 ± 0.2% [−14 ± 2 mmol/mol] and −0.9 ± 0.2% [−10 ± 2 mmol/mol], respectively; both p < 0.001), compared with placebo. Liraglutide tended to reduce the participant’s weight (−1.9 ± 1.0 kg; p = 0.06), whereas sitagliptin was weight-neutral (−0.6 ± 1.0 kg; p = 0.56).
Table 1
Baseline characteristics and treatment-induced effects
 
Liraglutide (n = 17)
Sitagliptin (n = 17)
Placebo (n = 17)
Parameter
Baseline
Follow-up
Placebo-corrected mean difference
Baseline
Follow-up
Placebo-corrected mean difference
Baseline
Follow-up
Participants’ characteristics
  Age (years)
60.8 ± 1.8
  
61.5 ± 1.7
  
65.8 ± 1.4
 
  Male sex (n (%))
12 (70.6)
  
14 (82.4)
  
13 (76.5)
 
  T2DM duration (years)
7.9 ± 1.2
  
8.5 ± 1.5
  
8.2 ± 1.2
 
  Metformin (n (%))
17 (100%)
  
16 (94.1%)
  
15 (88.2%)
 
  Sulfonylurea (n (%))
6 (35.3%)
  
9 (52.9%)
  
8 (47.1%)
 
Hepatic fat content
   1H-MRS (%)
20.9 ± 3.4
18.8 ± 3.3
−0.04 ± 1.86 (p = 0.98)
23.9 ± 3.0
21.0 ± 2.7
0.05 ± 1.87 (p = 0.98)
18.7 ± 2.7
16.9 ± 2.7
Hepatic fibrosis formulae
  NFS
0.24 ± 0.18
0.10 ± 0.19
−0.25 ± 0.14 (p = 0.08)
0.05 ± 0.27
0.04 ± 0.24
−0.07 ± 0.13 (p = 0.61)
0.07 ± 0.22
0.15 ± 0.21
  FIB-4
1.3 ± 0.1
1.1 ± 0.1
−0.13 ± 0.08 (p = 0.12)
1.2 ± 0.1
1.2 ± 0.1
−0.00 ± 0.08 (p = 0.99)
1.2 ± 0.1
1.2 ± 0.1
  APRI
0.3 ± 0.03
0.3 ± 0.02
−0.01 ± 0.02 (p = 0.79)
0.3 ± 0.02
0.3 ± 0.02
0.03 ± 0.02 (p = 0.19)
0.3 ± 0.02
0.3 ± 0.03
Anthropometrics
  Weight (kg)
103.2 ± 3.2
101.0 ± 3.1
−1.89 ± 0.99 (p = 0.06)
98.5 ± 4.4
97.7 ± 4.5
−0.57 ± 0.97 (p = 0.56)
95.8 ± 2.4
95.7 ± 2.5
  BMI (kg/m2)
32.8 ± 1.0
32.2 ± 1.0
−0.51 ± 0.33 (p = 0.13)
31.4 ± 1.1
31.1 ± 1.1
−0.20 ± 0.35 (p = 0.54)
30.6 ± 0.7
30.5 ± 0.8
Laboratory tests
  Glucose (mmol/l)
8.3 ± 0.3
7.3 ± 0.4
−1.61 ± 0.47 (p = 0.001)*
7.9 ± 0.2
7.0 ± 0.2
−1.82 ± 0.49 (p < 0.001)*
8.9 ± .5
9.4 ± 0.5
  Insulin (pmol/l)
78.8 ± 9.7
113.7 ± 16.2
52.6 ± 11.4 (p < 0.001)*
103.6 ± 15.3
113.3 ± 18.4
28.0 ± 11.2 (p = 0.016)*
106.4 ± 17.4
87.9 ± 13.4
  HbA1c (%)
7.4 ± 0.2
6.7 ± 0.2
−1.26 ± 0.21 (p < 0.001)*
7.1 ± 0.1
6.7 ± 0.1
−0.91 ± 0.22 (p < 0.001)*
7.5 ± 0.2
8.0 ± 0.3
  HbA1c (mmol/mol)
57 ± 2
49 ± 2
−14 ± 2 (p < 0.001)*
54 ± 1
49 ± 1
−10 ± 2 (p < 0.001)*
58 ± 2
64 ± 4
  AST (U/l)
24.2 ± 1.9
22.4 ± 1.4
0.09 ± 1.82 (p = 0.96)
22.8 ± 1.4
23.5 ± 2.1
2.16 ± 1.79 (p = 0.23)
22.2 ± 1.8
21.6 ± 1.7
  ALT (U/l)
28.9 ± 2.9
29.4 ± 3.2
3.38 ± 2.59 (p = 0.19)
28.9 ± 3.1
30.0 ± 3.7
4.07 ± 2.59 (p = 0.12)
32.0 ± 5.2
28.5 ± 3.9
  γGT (U/l)
68.2 ± 3.8
67.6 ± 2.9
1.63 ± 2.50 (p = 0.52)
67.1 ± 4.5
64.1 ± 4.7
−0.92 ± 2.49 (p = 0.71)
62.6 ± 2.9
61.4 ± 2.5
  ALP (U/l)
31.4 ± 3.0
28.2 ± 2.8
−2.77 ± 3.51 (p = 0.43)
45.0 ± 5.9
42.2 ± 5.4
−0.05 ± 3.47 (p = 0.99)
43.2 ± 10.3
40.8 ± 9.1
  Albumin (g/l)
37.0 ± 0.4
35.8 ± 0.5
−1.59 ± 0.56 (p = 0.007)*
36.4 ± 0.6
35.8 ± 0.5
−1.22 ± 0.56 (p = 0.036)*
37.0 ± 0.6
37.4 ± 0.5
  Total bilirubin (μmol/l)
10.4 ± 1.2
7.6 ± 0.9
−1.02 ± 0.88 (p = 0.25)
9.3 ± 1.4
8.0 ± 1.3
0.14 ± 0.86 (p = 0.87)
7.4 ± 0.6
6.6 ± 0.5
  Conjugated bilirubin (μmol/l)
4.0 ± 0.4
3.2 ± 0.3
−0.48 ± 0.33 (p = 0.15)
3.7 ± 0.4
3.3 ± 0.4
−0.07 ± 0.32 (p = 0.83)
3.0 ± 0.2
2.9 ± 0.2
  L-FABP (ng/ml)
9.4 ± 1.2
8.3 ± 1.4
1.33 ± 1.57 (p = 0.40)
10.1 ± 2.5
8.9 ± 1.2
1.82 ± 1.58 (p = 0.26)
8.4 ± 1.1
6.4 ± 0.8
Data are presented as mean ± SEM for continuous data
p values represent differences between treatment and placebo, corrected for baseline values. *Statistically significant at p < 0.05
FLI, fatty liver index; T2DM, type 2 diabetes
Twelve-week treatment with liraglutide or sitagliptin did not affect hepatic steatosis (−10% and −12.1% from baseline, respectively), no different from placebo (−9.5%; p = 0.98 for both) (Table 1, Fig. 1). In addition, the proportion of patients with any reduction in hepatic fat content was similar in the three groups: liraglutide 60.0%, sitagliptin 62.5%, and placebo 68.8%.
Neither liraglutide nor sitagliptin affected NFS, FIB-4 or APRI compared with placebo (all p > 0.05) (Table 1). No treatment-induced differences occurred in plasma aspartate aminotransferase (AST), alanine aminotransferase (ALT), gamma glutamyl transferase (γGT), alkaline phosphatase (ALP) or L-FABP concentrations (Table 1). Compared with placebo, liraglutide and sitagliptin reduced plasma albumin levels (−1.4 ± 0.6 g/l, p = 0.03 and −1.7 ± 0.6 g/l, p = 0.01, respectively), without affecting total or conjugated bilirubin (p > 0.05 for both).
As shown in Fig. 1, baseline hepatic fat content (Fig. 1a) did not determine treatment effects . Moreover, a subanalysis of patients with NAFLD at baseline (liraglutide n = 15, sitagliptin n = 16, placebo n = 15) did not yield different results (data not shown).

Discussion

In the current study, we did not observe a beneficial effect of 12 week liraglutide or sitagliptin treatment on hepatic steatosis or fibrosis. Although high-quality human studies were lacking when the present study was designed, several randomised controlled trials have in the meantime been performed and published. In line with our findings, Tang et al demonstrated no effect of 12 week treatment with liraglutide on MRI-measured liver fat [6]. Moreover, 24 week treatment with sitagliptin did not affect liver fat in patients with NAFLD when compared with placebo [7]. In contrast, a 48 week double-blind, placebo-controlled trial with liraglutide demonstrated an improvement in steatosis and a histological resolution of NASH in patients with and without type 2 diabetes [8]. Also, 26 week treatment with exenatide reduced hepatic fat by 24% [9]. A 6 month, double-blind, placebo-controlled, randomised controlled trial reported a 27% reduction in hepatic fat with vildagliptin that was unrelated to changes in body weight [10]. Finally, an open-label trial comparing sitagliptin with glimepiride found no change in intrahepatic fat content after a 12 week treatment, but reported a 15% decrease after 24 weeks [11]. These contradictory outcomes may result from differences in treatment duration, study population and the method used to assess hepatic fat.
Albeit only as statistical trend, liraglutide decreased body weight within the expected range [12]. In recent studies with liraglutide [8, 13], weight loss reached a nadir after 24 weeks, indicating that further weight loss could have been observed with a longer duration of the current trial. Since weight loss per se reduces hepatic steatosis [14], a longer trial might have yielded different results.
Unexpectedly, we observed a modest decrease in plasma albumin levels in the liraglutide and sitagliptin groups, for which we cannot provide an explanation. Inhibition of hepatic synthetic function seems unlikely, as bilirubin levels remained unchanged and an increased risk of bleeding due to GLP-1 based therapies has never been reported. In addition, other potential causes, such as inflammation or renal loss of albumin, were not observed.
The limitations of the current study were untriggered magnetic resonance spectroscopy measurements and manual VOI positioning. To diminish the effect of small variations before and after intervention, we averaged over three positions in the liver. Proton-density fat fraction MRI is an alternative and widely available option [15]. Although the sample size was small, it is unlikely that a larger trial would have yielded different results given the minimal between-group differences.
In conclusion, 12 week treatment with liraglutide or sitagliptin did not improve hepatic steatosis or fibrosis in overweight patients with type 2 diabetes. Further, longer term studies are needed to assess the potential of these agents as treatment strategy for NAFLD.

Acknowledgements

The authors are grateful to the study participants, whose time and effort were critical to the success of our research protocol.

Funding

The research leading to these results has received funding from the European Community’s Seventh Framework Programme (FP7/2007-2013) under grant agreement no. 282521 – the SAFEGUARD project. Novo Nordisk A/S kindly provided the liraglutide and liraglutide placebo. The funders had no role in the study design, the collection, analysis and interpretation of the data, the writing of the report or the decision to submit the article for publication.

Duality of interest statement

Through MD and MHHK, the VU University Medical Center received research grants from AstraZeneca, Boehringer Ingelheim, Novo Nordisk and Sanofi. All other authors declare that there is no duality of interest associated with their contribution to this manuscript.

Contribution statement

MMS developed the study protocol, performed the measurements and analyses, and drafted the manuscript. LT performed measurements and made a substantial contribution to data interpretation and revision of the manuscript. MD developed the study protocol and was involved in the discussion. TH contributed to data analysis and interpretation, and critically revised the manuscript. MHAM, ICP-vdB, PJWP, MHHK, DHvR and DLC contributed to data interpretation and revised the manuscript. MMS and DHR had full access to all of the data and can take responsibility for the integrity of the data and the accuracy of the data analysis. All authors have approved the final version of this manuscript.
Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

e.Med Allgemeinmedizin

Kombi-Abonnement

Mit e.Med Allgemeinmedizin erhalten Sie Zugang zu allen CME-Fortbildungen und Premium-Inhalten der allgemeinmedizinischen Zeitschriften, inklusive einer gedruckten Allgemeinmedizin-Zeitschrift Ihrer Wahl.

Anhänge

Electronic supplementary material

Below is the link to the electronic supplementary material.
Literatur
1.
Zurück zum Zitat Ahmed A, Wong RJ, Harrison SA (2015) Nonalcoholic fatty liver disease review: diagnosis, treatment, and outcomes. Clin Gastroenterol Hepatol 13:2062–2070CrossRefPubMed Ahmed A, Wong RJ, Harrison SA (2015) Nonalcoholic fatty liver disease review: diagnosis, treatment, and outcomes. Clin Gastroenterol Hepatol 13:2062–2070CrossRefPubMed
2.
Zurück zum Zitat Smits MM, van Raalte DH, Tonneijck L et al (2016) GLP-1 based therapies: clinical implications for gastroenterologists. Gut 65:702–711CrossRefPubMed Smits MM, van Raalte DH, Tonneijck L et al (2016) GLP-1 based therapies: clinical implications for gastroenterologists. Gut 65:702–711CrossRefPubMed
3.
Zurück zum Zitat Smits MM, Tonneijck L, Muskiet MHA et al (2015) Cardiovascular, renal and gastrointestinal effects of incretin-based therapies: an acute and 12-week randomised, double-blind, placebo-controlled, mechanistic intervention trial in type 2 diabetes. BMJ Open 5:e009579CrossRefPubMedPubMedCentral Smits MM, Tonneijck L, Muskiet MHA et al (2015) Cardiovascular, renal and gastrointestinal effects of incretin-based therapies: an acute and 12-week randomised, double-blind, placebo-controlled, mechanistic intervention trial in type 2 diabetes. BMJ Open 5:e009579CrossRefPubMedPubMedCentral
4.
Zurück zum Zitat Kim D, Kim WR, Kim HJ, Therneau TM (2013) Association between noninvasive fibrosis markers and mortality among adults with nonalcoholic fatty liver disease in the United States. Hepatology 57:1357–1365CrossRefPubMedPubMedCentral Kim D, Kim WR, Kim HJ, Therneau TM (2013) Association between noninvasive fibrosis markers and mortality among adults with nonalcoholic fatty liver disease in the United States. Hepatology 57:1357–1365CrossRefPubMedPubMedCentral
5.
Zurück zum Zitat Cuthbertson DJ, Irwin A, Gardner CJ et al (2012) Improved glycaemia correlates with liver fat reduction in obese, type 2 diabetes, patients given glucagon-like peptide-1 (GLP-1) receptor agonists. PLoS One 7:e50117CrossRefPubMedPubMedCentral Cuthbertson DJ, Irwin A, Gardner CJ et al (2012) Improved glycaemia correlates with liver fat reduction in obese, type 2 diabetes, patients given glucagon-like peptide-1 (GLP-1) receptor agonists. PLoS One 7:e50117CrossRefPubMedPubMedCentral
6.
Zurück zum Zitat Tang A, Rabasa-Lhoret R, Castel H et al (2015) Effects of insulin glargine and liraglutide therapy on liver fat as measured by magnetic resonance in patients with type 2 diabetes: a randomized trial. Diabetes Care 38:1339–1346CrossRefPubMed Tang A, Rabasa-Lhoret R, Castel H et al (2015) Effects of insulin glargine and liraglutide therapy on liver fat as measured by magnetic resonance in patients with type 2 diabetes: a randomized trial. Diabetes Care 38:1339–1346CrossRefPubMed
7.
Zurück zum Zitat Cui J, Philo L, Nguyen P et al (2016) Sitagliptin vs placebo in the treatment of nonalcoholic fatty liver disease: a randomized controlled trial. J Hepatol 65:369–376CrossRefPubMed Cui J, Philo L, Nguyen P et al (2016) Sitagliptin vs placebo in the treatment of nonalcoholic fatty liver disease: a randomized controlled trial. J Hepatol 65:369–376CrossRefPubMed
8.
Zurück zum Zitat Armstrong MJ, Gaunt P, Aithal GP et al (2016) Liraglutide safety and efficacy in patients with non-alcoholic steatohepatitis (LEAN): a multicentre, double-blind, randomised, placebo-controlled phase 2 study. Lancet 387:679–690CrossRefPubMed Armstrong MJ, Gaunt P, Aithal GP et al (2016) Liraglutide safety and efficacy in patients with non-alcoholic steatohepatitis (LEAN): a multicentre, double-blind, randomised, placebo-controlled phase 2 study. Lancet 387:679–690CrossRefPubMed
9.
Zurück zum Zitat Dutour A, Abdesselam I, Ancel P et al (2016) Exenatide decreases liver fat content and epicardial adipose tissue in patients with obesity and type 2 diabetes: a prospective randomised clinical trial using magnetic resonance imaging and spectroscopy. Diabetes Obes Metab. doi:10.1111/dom.12680 PubMed Dutour A, Abdesselam I, Ancel P et al (2016) Exenatide decreases liver fat content and epicardial adipose tissue in patients with obesity and type 2 diabetes: a prospective randomised clinical trial using magnetic resonance imaging and spectroscopy. Diabetes Obes Metab. doi:10.​1111/​dom.​12680 PubMed
10.
11.
Zurück zum Zitat Kato H, Nagai Y, Ohta A et al (2015) Effect of sitagliptin on intrahepatic lipid content and body fat in patients with type 2 diabetes. Diabetes Res Clin Pract 109:199–205CrossRefPubMed Kato H, Nagai Y, Ohta A et al (2015) Effect of sitagliptin on intrahepatic lipid content and body fat in patients with type 2 diabetes. Diabetes Res Clin Pract 109:199–205CrossRefPubMed
12.
Zurück zum Zitat Potts JE, Gray LJ, Brady EM et al (2015) The effect of glucagon-like peptide 1 receptor agonists on weight loss in type 2 diabetes: a systematic review and mixed treatment comparison meta-analysis. PLoS One 10:e0126769CrossRefPubMedPubMedCentral Potts JE, Gray LJ, Brady EM et al (2015) The effect of glucagon-like peptide 1 receptor agonists on weight loss in type 2 diabetes: a systematic review and mixed treatment comparison meta-analysis. PLoS One 10:e0126769CrossRefPubMedPubMedCentral
13.
Zurück zum Zitat Marso SP, Daniels GH, Brown-Frandsen K et al (2016) Liraglutide and cardiovascular outcomes in type 2 diabetes. N Engl J Med 375:311–322CrossRefPubMed Marso SP, Daniels GH, Brown-Frandsen K et al (2016) Liraglutide and cardiovascular outcomes in type 2 diabetes. N Engl J Med 375:311–322CrossRefPubMed
14.
Zurück zum Zitat Patel NS, Doycheva I, Peterson MR et al (2015) Effect of weight loss on magnetic resonance imaging estimation of liver fat and volume in patients with nonalcoholic steatohepatitis. Clin Gastroenterol Hepatol 13:561–568.e1CrossRefPubMed Patel NS, Doycheva I, Peterson MR et al (2015) Effect of weight loss on magnetic resonance imaging estimation of liver fat and volume in patients with nonalcoholic steatohepatitis. Clin Gastroenterol Hepatol 13:561–568.e1CrossRefPubMed
15.
Metadaten
Titel
Twelve week liraglutide or sitagliptin does not affect hepatic fat in type 2 diabetes: a randomised placebo-controlled trial
verfasst von
Mark M. Smits
Lennart Tonneijck
Marcel H. A. Muskiet
Mark H. H. Kramer
Petra J. W. Pouwels
Indra C. Pieters-van den Bos
Trynke Hoekstra
Michaela Diamant
Daniël H. van Raalte
Djuna L. Cahen
Publikationsdatum
15.09.2016
Verlag
Springer Berlin Heidelberg
Erschienen in
Diabetologia / Ausgabe 12/2016
Print ISSN: 0012-186X
Elektronische ISSN: 1432-0428
DOI
https://doi.org/10.1007/s00125-016-4100-7

Weitere Artikel der Ausgabe 12/2016

Diabetologia 12/2016 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Die „Zehn Gebote“ des Endokarditis-Managements

30.04.2024 Endokarditis Leitlinie kompakt

Worauf kommt es beim Management von Personen mit infektiöser Endokarditis an? Eine Kardiologin und ein Kardiologe fassen die zehn wichtigsten Punkte der neuen ESC-Leitlinie zusammen.

Strenge Blutdruckeinstellung lohnt auch im Alter noch

30.04.2024 Arterielle Hypertonie Nachrichten

Ältere Frauen, die von chronischen Erkrankungen weitgehend verschont sind, haben offenbar die besten Chancen, ihren 90. Geburtstag zu erleben, wenn ihr systolischer Blutdruck < 130 mmHg liegt. Das scheint selbst für 80-Jährige noch zu gelten.

Gegen postoperative Darmmotilitätsstörung: Erster Kaffee schon im Aufwachraum

30.04.2024 DCK 2024 Kongressbericht

Nach einer Operation kann es zu einer vorübergehenden Störung der Darmmotilität kommen, ohne dass es eine mechanische Ursache gibt. Auf eine postoperative Darmmotilitätsstörung weist besonders hin, wenn Nahrung oral nicht toleriert wird.

Frauen bekommen seltener eine intensive Statintherapie

30.04.2024 Statine Nachrichten

Frauen in den Niederlanden erhalten bei vergleichbarem kardiovaskulärem Risiko seltener eine intensive Statintherapie als Männer. Ihre LDL-Zielwerte erreichen sie aber fast ähnlich oft.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.