Placebo-controlled randomised trial with liraglutide on magnetic resonance endpoints in individuals with type 2 diabetes: a pre-specified secondary study on ectopic fat accumulation

Maurice B Bizino, Ingrid M Jazet, Paul de Heer, Huub J van Eyk, Ilona A Dekkers, Patrick C N Rensen, Elisabeth H M Paiman, Hildebrandus J Lamb, Johannes W Smit, Maurice B Bizino, Ingrid M Jazet, Paul de Heer, Huub J van Eyk, Ilona A Dekkers, Patrick C N Rensen, Elisabeth H M Paiman, Hildebrandus J Lamb, Johannes W Smit

Abstract

Aims/hypothesis: The aim of this work was to assess the effect of liraglutide on ectopic fat accumulation in individuals with type 2 diabetes mellitus.

Methods: This study is a pre-specified subanalysis of the MAGNetic resonance Assessment of VICTOza efficacy in the Regression of cardiovascular dysfunction In type 2 diAbetes mellitus (MAGNA VICTORIA) study, with primary endpoints being the effects of liraglutide on left ventricular diastolic and systolic function. The MAGNA VICTORIA study was a single-centre, parallel-group trial in 50 individuals with type 2 diabetes mellitus (BMI >25 kg/m2) who were randomly assigned (1:1, stratified for sex and insulin use) to receive liraglutide 1.8 mg once daily or placebo for 26 weeks, added to standard care. Participants, study personnel and outcome assessors were blinded to treatment allocation. The secondary endpoints of visceral adipose tissue (VAT), abdominal subcutaneous adipose tissue (SAT) and epicardial fat were measured with MRI. Hepatic triacylglycerol content (HTGC) and myocardial triacylglycerol content (MTGC) were quantified with proton MR spectroscopy. Between-group differences (change from baseline) were tested for significance using ANCOVA. Mean differences with 95% CIs were reported.

Results: The trial was completed in 2016. Twenty-four participants were randomised to receive liraglutide and 26 to receive placebo. One patient in the liraglutide group withdrew consent before having received the study drug and was not included in the intention-to-treat analysis. Liraglutide (n = 23) vs placebo (n = 26) significantly reduced body weight (liraglutide 98.4 ± 13.8 kg to 94.3 ± 14.9 kg; placebo 94.5 ± 13.1 kg to 93.9 ± 13.2 kg; estimated treatment effect -4.5 [95% CI -6.4, -2.6] kg). HbA1c declined in both groups without a significant treatment effect of liraglutide vs placebo (liraglutide 66.7 ± 11.5 mmol/mol to 55.0 ± 13.2 mmol/mol [8.4 ± 1.1% to 7.3 ± 1.2%]; placebo 64.7 ± 10.2 mmol/mol to 56.9 ± 6.9 mmol/mol [8.2 ± 1.0% to 7.5 ± 0.7%]; estimated treatment effect -2.9 [95% CI -8.1, 2.3] mmol/mol or -0.3 [95% CI -0.8, 0.2]%). VAT did not change significantly between groups (liraglutide 207 ± 87 cm2 to 203 ± 88 cm2; placebo 204 ± 63 cm2 to 200 ± 55 cm2; estimated treatment effect -7 [95% CI -24, 10] cm2), while SAT was reduced by a significantly greater extent with liraglutide than with placebo (liraglutide 361 ± 142 cm2 to 339 ± 131 cm2; placebo 329 ± 107 cm2 to 333 ± 125 cm2; estimated treatment effect -29 [95% CI -51, -8] cm2). Epicardial fat did not change significantly between groups (liraglutide 8.9 ± 4.3 cm2 to 9.1 ± 4.7 cm2; placebo 9.6 ± 4.1 cm2 to 9.6 ± 4.6 cm2; estimated treatment effect 0.2 [95% CI -1.5, 1.8] cm2). Change in HTGC was not different between groups (liraglutide 18.1 ± 11.2% to 12.0 ± 7.7%; placebo 18.4 ± 9.4% to 14.7 ± 10.0%; estimated treatment effect -2.1 [95% CI -5.3, 1.0]%). MTGC was not different after treatment with liraglutide (1.5 ± 0.6% to 1.2 ± 0.6%) vs placebo (1.3 ± 0.5% to 1.2 ± 0.6%), with an estimated treatment effect of -0.1 (95% CI -0.4, 0.2)%. There were no adjudicated serious adverse events.

Conclusions/interpretation: Compared with placebo, liraglutide-treated participants lost significantly more body weight. Liraglutide primarily reduced subcutaneous fat but not visceral, hepatic, myocardial or epicardial fat. Future larger studies are needed to confirm the results of this secondary endpoint study.

Trial registration: ClinicalTrials.gov NCT01761318.

Funding: This study was funded by Novo Nordisk A/S (Bagsvaerd, Denmark).

Keywords: Ectopic fat; Glucagon-like peptide-1 receptor agonist; Hepatic steatosis; Liraglutide; Myocardial steatosis; Non-alcoholic fatty liver disease; Type 2 diabetes mellitus.

Figures

Fig. 1
Fig. 1
Scatter plot of HbA1c vs HTGC difference (baseline – follow-up at 26 weeks) for all participants. Circles, placebo; squares, liraglutide. The unstandardised regression line (y = 2.57 + 0.28x) with 95% CI is shown

References

    1. van der Meer RW, Lamb HJ, Smit JW, de Roos A. MR imaging evaluation of cardiovascular risk in metabolic syndrome. Radiology. 2012;264(1):21–37. doi: 10.1148/radiol.12110772.
    1. Park HS, Lee K. Greater beneficial effects of visceral fat reduction compared with subcutaneous fat reduction on parameters of the metabolic syndrome: a study of weight reduction programmes in subjects with visceral and subcutaneous obesity. Diabet Med. 2005;22(3):266–272. doi: 10.1111/j.1464-5491.2004.01395.x.
    1. Levelt E, Mahmod M, Piechnik SK, et al. Relationship between left ventricular structural and metabolic remodeling in type 2 diabetes. Diabetes. 2016;65(1):44–52. doi: 10.2337/db15-0627.
    1. Chaston TB, Dixon JB. Factors associated with percent change in visceral versus subcutaneous abdominal fat during weight loss: findings from a systematic review. Int J Obes. 2008;32(4):619–628. doi: 10.1038/sj.ijo.0803761.
    1. Ben-Shlomo S, Zvibel I, Shnell M, et al. Glucagon-like peptide-1 reduces hepatic lipogenesis via activation of AMP-activated protein kinase. J Hepatol. 2011;54(6):1214–1223. doi: 10.1016/j.jhep.2010.09.032.
    1. Sharma S, Mells JE, Fu PP, Saxena NK, Anania FA. GLP-1 analogs reduce hepatocyte steatosis and improve survival by enhancing the unfolded protein response and promoting macroautophagy. PLoS One. 2011;6(9):e25269. doi: 10.1371/journal.pone.0025269.
    1. Parlevliet ET, Wang Y, Geerling JJ, et al. GLP-1 receptor activation inhibits VLDL production and reverses hepatic steatosis by decreasing hepatic lipogenesis in high-fat-fed APOE*3-Leiden mice. PLoS One. 2012;7(11):e49152. doi: 10.1371/journal.pone.0049152.
    1. Noyan-Ashraf MH, Shikatani EA, Schuiki I, et al. A glucagon-like peptide-1 analog reverses the molecular pathology and cardiac dysfunction of a mouse model of obesity. Circulation. 2013;127(1):74–85. doi: 10.1161/circulationaha.112.091215.
    1. Bi Y, Zhang B, Xu W, et al. Effects of exenatide, insulin, and pioglitazone on liver fat content and body fat distributions in drug-naive subjects with type 2 diabetes. Acta Diabetol. 2014;51(5):865–873. doi: 10.1007/s00592-014-0638-3.
    1. Jendle J, Nauck MA, Matthews DR, et al. Weight loss with liraglutide, a once-daily human glucagon-like peptide-1 analogue for type 2 diabetes treatment as monotherapy or added to metformin, is primarily as a result of a reduction in fat tissue. Diabetes Obes Metab. 2009;11(12):1163–1172. doi: 10.1111/j.1463-1326.2009.01158.x.
    1. Li CJ, Yu Q, Yu P, et al. Changes in liraglutide-induced body composition are related to modifications in plasma cardiac natriuretic peptides levels in obese type 2 diabetic patients. Cardiovasc Diabetol. 2014;13:36. doi: 10.1186/1475-2840-13-36.
    1. Santilli F, Simeone PG, Guagnano MT, et al. Effects of liraglutide on weight loss, fat distribution, and beta-cell function in obese subjects with prediabetes or early type 2 diabetes. Diabetes Care. 2017;40(11):1556–1564. doi: 10.2337/dc17-0589.
    1. Shi Li, Zhu Jing, Yang Ping, Tang Xiaoqiang, Yu Wenlong, Pan Changjie, Shen Moyu, Zhu Dalong, Cheng Jinluo, Ye Xinhua. Comparison of exenatide and acarbose on intra-abdominal fat content in patients with obesity and type-2 diabetes: A randomized controlled trial. Obesity Research & Clinical Practice. 2017;11(5):607–615. doi: 10.1016/j.orcp.2017.01.003.
    1. Suzuki D, Toyoda M, Kimura M, et al. Effects of liraglutide, a human glucagon-like peptide-1 analogue, on body weight, body fat area and body fat-related markers in patients with type 2 diabetes mellitus. Intern Med. 2013;52(10):1029–1034. doi: 10.2169/internalmedicine.52.8961.
    1. Tang A, Rabasa-Lhoret R, Castel H, et al. 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. 2015;38(7):1339–1346. doi: 10.2337/dc14-2548.
    1. Jonker JT, de Mol P, de Vries ST, et al. Exercise and type 2 diabetes mellitus: changes in tissue-specific fat distribution and cardiac function. Radiology. 2013;269(2):434–442. doi: 10.1148/radiol.13121631.
    1. Dutour A, Abdesselam I, Ancel P, et al. Exenatide decreases liver fat content and epicardial adipose tissue in patients with obesity and type 2 diabetes: a prospective randomized clinical trial using magnetic resonance imaging and spectroscopy. Diabetes Obes Metab. 2016;18(9):882–891. doi: 10.1111/dom.12680.
    1. Iacobellis G, Mohseni M, Bianco SD, Banga PK. Liraglutide causes large and rapid epicardial fat reduction. Obesity (Silver Spring) 2017;25(2):311–316. doi: 10.1002/oby.21718.
    1. Bizino MB, Sala ML, de Heer P, et al. MR of multi-organ involvement in the metabolic syndrome. Magn Reson Imaging Clin N Am. 2015;23(1):41–58. doi: 10.1016/j.mric.2014.09.010.
    1. Bizino MB, Jazet IM, Westenberg JJM, et al. Effect of liraglutide on cardiac function in patients with type 2 diabetes mellitus: randomized placebo-controlled trial. Cardiovasc Diabetol. 2019;18(1):55–12. doi: 10.1186/s12933-019-0857-6.
    1. de Heer P, Bizino MB, Lamb HJ, Webb AG. Parameter optimization for reproducible cardiac (1) H-MR spectroscopy at 3 Tesla. J Magn Reson Imaging. 2016;44(5):1151–1158. doi: 10.1002/jmri.25254.
    1. Pastel E, McCulloch LJ, Ward R, et al. GLP-1 analogue-induced weight loss does not improve obesity-induced AT dysfunction. Clin Sci (Lond) 2017;131(5):343–353. doi: 10.1042/cs20160803.
    1. Drucker DJ. Mechanisms of action and therapeutic application of glucagon-like peptide-1. Cell Metab. 2018;27(4):740–756. doi: 10.1016/j.cmet.2018.03.001.
    1. Lee YS, Park MS, Choung JS, et al. Glucagon-like peptide-1 inhibits adipose tissue macrophage infiltration and inflammation in an obese mouse model of diabetes. Diabetologia. 2012;55(9):2456–2468. doi: 10.1007/s00125-012-2592-3.
    1. Kreier F, Kap YS, Mettenleiter TC, et al. Tracing from fat tissue, liver, and pancreas: a neuroanatomical framework for the role of the brain in type 2 diabetes. Endocrinology. 2006;147(3):1140–1147. doi: 10.1210/en.2005-0667.
    1. Kreier F, Fliers E, Voshol PJ, et al. Selective parasympathetic innervation of subcutaneous and intra-abdominal fat--functional implications. J Clin Invest. 2002;110(9):1243–1250. doi: 10.1172/jci15736.
    1. Kooijman S, Wang Y, Parlevliet ET, et al. Central GLP-1 receptor signalling accelerates plasma clearance of triacylglycerol and glucose by activating brown adipose tissue in mice. Diabetologia. 2015;58(11):2637–2646. doi: 10.1007/s00125-015-3727-0.
    1. Yan Jinhua, Yao Bin, Kuang Hongyu, Yang Xubin, Huang Qin, Hong Tianpei, Li Yushu, Dou Jingtao, Yang Wenying, Qin Guijun, Yuan Huijuan, Xiao Xinhua, Luo Sihui, Shan Zhongyan, Deng Hongrong, Tan Ying, Xu Fen, Xu Wen, Zeng Longyi, Kang Zhuang, Weng Jianping. Liraglutide, Sitagliptin, and Insulin Glargine Added to Metformin: The Effect on Body Weight and Intrahepatic Lipid in Patients With Type 2 Diabetes Mellitus and Nonalcoholic Fatty Liver Disease. Hepatology. 2019;69(6):2414–2426. doi: 10.1002/hep.30320.
    1. Petit JM, Cercueil JP, Loffroy R, et al. Effect of liraglutide therapy on liver fat content in patients with inadequately controlled type 2 diabetes: the Lira-NAFLD Study. J Clin Endocrinol Metab. 2017;102(2):407–415. doi: 10.1210/jc.2016-2775.
    1. Armstrong MJ, Gaunt P, Aithal GP, et al. Liraglutide safety and efficacy in patients with non-alcoholic steatohepatitis (LEAN): a multicentre, double-blind, randomised, placebo-controlled phase 2 study. Lancet. 2016;387(10019):679–690. doi: 10.1016/s0140-6736(15)00803-x.
    1. Cuthbertson DJ, Irwin A, Gardner CJ, et al. Improved glycaemia correlates with liver fat reduction in obese, type 2 diabetes, patients given glucagon-like peptide-1 (GLP-1) receptor agonists. PLoS One. 2012;7(12):e50117. doi: 10.1371/journal.pone.0050117.
    1. Ishii S, Iizuka K, Miller BC, Uyeda K. Carbohydrate response element binding protein directly promotes lipogenic enzyme gene transcription. Proc Natl Acad Sci U S A. 2004;101(44):15597–15602. doi: 10.1073/pnas.0405238101.
    1. Rijzewijk LJ, van der Meer RW, Smit JW, et al. Myocardial steatosis is an independent predictor of diastolic dysfunction in type 2 diabetes mellitus. J Am Coll Cardiol. 2008;52(22):1793–1799. doi: 10.1016/j.jacc.2008.07.062.
    1. Hammer S, Snel M, Lamb HJ, et al. Prolonged caloric restriction in obese patients with type 2 diabetes mellitus decreases myocardial triglyceride content and improves myocardial function. J Am Coll Cardiol. 2008;52(12):1006–1012. doi: 10.1016/j.jacc.2008.04.068.
    1. Wu Lujin, Wang Ke, Wang Wei, Wen Zheng, Wang Peihua, Liu Lei, Wang Dao Wen. Glucagon-like peptide-1 ameliorates cardiac lipotoxicity in diabetic cardiomyopathy via the PPARα pathway. Aging Cell. 2018;17(4):e12763. doi: 10.1111/acel.12763.
    1. Leonardini A, D’Oria R, Incalza MA, et al. GLP-1 Receptor Activation Inhibits Palmitate-Induced Apoptosis via Ceramide in Human Cardiac Progenitor Cells. J Clin Endocrinol Metab. 2017;102(11):4136–4147. doi: 10.1210/jc.2017-00970.
    1. Levelt E, Pavlides M, Banerjee R, et al. Ectopic and visceral fat deposition in lean and obese patients with type 2 diabetes. J Am Coll Cardiol. 2016;68(1):53–63. doi: 10.1016/j.jacc.2016.03.597.
    1. Mahabadi AA, Berg MH, Lehmann N, et al. Association of epicardial fat with cardiovascular risk factors and incident myocardial infarction in the general population: the Heinz Nixdorf Recall Study. J Am Coll Cardiol. 2013;61(13):1388–1395. doi: 10.1016/j.jacc.2012.11.062.
    1. van Eyk HJ, van Schinkel LD, Kantae V, et al. Caloric restriction lowers endocannabinoid tonus and improves cardiac function in type 2 diabetes. Nutr Diabetes. 2018;8(1):6–10. doi: 10.1038/s41387-017-0016-7.
    1. Chau YY, Bandiera R, Serrels A, et al. Visceral and subcutaneous fat have different origins and evidence supports a mesothelial source. Nat Cell Biol. 2014;16(4):367–375. doi: 10.1038/ncb2922.
    1. Marso SP, Daniels GH, Brown-Frandsen K, et al. Liraglutide and cardiovascular outcomes in type 2 diabetes. N Engl J Med. 2016;375(4):311–322. doi: 10.1056/NEJMoa1603827.
    1. Goodman SN, Berlin JA. The use of predicted confidence intervals when planning experiments and the misuse of power when interpreting results. Ann Intern Med. 1994;121(3):200–206. doi: 10.7326/0003-4819-121-3-199408010-00008.
    1. Davies MJ, Bergenstal R, Bode B, et al. Efficacy of liraglutide for weight loss among patients with type 2 diabetes: the SCALE diabetes randomized clinical trial. Jama. 2015;314(7):687–699. doi: 10.1001/jama.2015.9676.

Source: PubMed

3
Abonneren