Editorial - Ingenta Connect

6 downloads 214 Views 114KB Size Report
We recently reviewed the role of liraglutide (3.0 mg) in obesity treatment [1]. Since then, the Sati- ety and Clinical Adiposity-Liraglutide Evidence in Nondiabetic ...
76 Current Vascular Pharmacology, 2016, Vol. 14, No. 1

Editorial

Editorial Liraglutide and Cardiometabolic Effects: More than Just Another Antiobesity Drug? Niki Katsiki1, Georgios A. Christou2 and Dimitrios N. Kiortsis2,* 1 nd

2 Propedeutic Department of Internal Medicine, Medical School, Aristotle University, Thessaloniki, Greece; 2Laboratory of Physiology, Medical School, University of Ioannina, Ioannina, Greece

We recently reviewed the role of liraglutide (3.0 mg) in obesity treatment [1]. Since then, the Satiety and Clinical Adiposity-Liraglutide Evidence in Nondiabetic and Diabetic people (SCALE) Obesity and Prediabetes study, the largest (n= 3731) randomized controlled trial investigating the use of liraglutide (3.0 mg) in obesity, was published [2]. Briefly, patients were randomly assigned in a 2:1 ratio to liraglutide 3.0 mg once-daily or placebo as an adjunct to diet and exercise [2]. Patients (78.5% women) were non-diabetics (61.2% had prediabetes as defined by the American Diabetes Association [3]) with a mean body mass index (BMI = 38.3 ± 6.4 Kg/m2).

Dimitrios N. Kiortsis

After 56 weeks of treatment, patients on liraglutide lost significantly more weight compared with the placebo group (– 5.4% difference; 95% confidence interval – 5.8 to – 5.0%; p < 0.001) [2]. Similarly, the percentages of patients achieving at least 5% or > 10% weight loss were significantly higher in the liraglutide group (63.2 and 33.1%, respectively) compared with placebo (27.1 and 10.6%, respectively; p < 0.001 for all comparisons). Notably, missing values in the SCALE Obesity and Prediabetes study were imputed with the use of the last-observation-carried-forward (LOCF) method for measurements made after baseline. The LOCF method is one of the most commonly used approaches for dealing with dropout data. In the LOCF method, the outcome variable for a dropout subject is the change from baseline to the last observed response [4]. This assumes that the outcome remains frozen in time after the point a subject drops out. In this aspect, the greater magnitude of dropout in the placebo arm (36%) compared with the liraglutide arm (28%) in the SCALE Obesity and Prediabetes study possibly results in the overestimation of the weight loss effect of liraglutide compared with placebo, given the expected further weight loss of the individuals who withdrew from the study if they had not dropped out. Liraglutide 3.0 mg/day also led to significantly greater improvements in several cardiometabolic parameters [BMI, waist circumference, fasting glucose and insulin, glycated haemoglobin, total cholesterol, serum triglycerides (TG), low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol (HDL-C), systolic and diastolic blood pressure and serum levels of high-sensitivity C-reactive protein (hsCRP), plasminogen activator inhibitor-1 (PAI-1) and adiponectin] [2]. Of note, several continuous variables expressed as mean ± standard deviation (SD) had the SD > mean (e.g. for hsCRP, PAI-1, adiponectin, urinary albumin:creatinine ratio, fibrinogen and insulin). These variables may not be normally distributed (e.g. urinary albumin:creatinine ratio in the placebo group was 3.6±627.2 mg/g) and may have been better expressed as median (range). Apart from LDL-C, small dense LDL (sdLDL) particles are related to increased cardiovascular disease (CVD) risk [5-7]. Liraglutide was reported to decrease sdLDL-C levels [8]. In the SCALE Obesity and Prediabetes study [2], TG levels were significantly reduced and HDL-C levels were significantly increased following liraglutide 3.0 mg/day treatment. This improvement in atherogenic dyslipidaemia may also be accompanied by a fall in the proportion of sdLDL-C to LDL-C [5-7]. However, the authors did not measure sdLDL-C levels. Similarly, postprandial lipaemia (PPL) may be another CVD risk predictor [9,10]. Liraglutide up to 1.8 mg/day was shown to reduce PPL [11, 12]. In the SCALE Obesity and Prediabetes study [2], PPL was not reported. Metabolic syndrome (MetS) is a cluster of hypertension, dyslipidaemia, insulin resistance and/or central obesity associated with increased CVD risk [13, 14]. Based on a joint interim statement of several scientific societies published in 2009 [15], MetS diagnostic criteria include elevated blood pressure, waist circumference, TG and fasting glucose as well as low HDL-C levels. In the SCALE Obesity and Prediabetes study [2], liraglutide 3.0 mg/day significantly improved all these cardiometabolic factors. In this context, it would be useful to assess the impact of liraglutide 3.0 mg/day on MetS prevalence in this study. Non-alcoholic fatty liver disease (NAFLD), the hepatic manifestation of MetS, has been related to increased CVD risk [1618]. Elevated liver enzyme activities were also linked to raised CVD morbidity and mortality [19]. NAFLD treatment, although not yet established, is multifactorial including lifestyle changes, hypolipidaemic, antihypertensive, antiobesity and antidiabetic drugs [20-22]. Liraglutide 0.9-1.8 mg/dl was reported to improve biochemical and/or histological features of NAFLD/ nonalcoholic steatohepatitis (NASH) [23-25]. No data on NAFLD were reported in the SCALE Obesity and Prediabetes study. Elevated serum uric acid (SUA) levels have been associated with increased vascular risk [26, 27]. No data exists for liraglutide effects on SUA levels. Chronic kidney disease (CKD) stage 3, as assessed by estimated glomerular filtration rate (eGFR) 1875-6212/16 $58.00+.00

© 2016 Bentham Science Publishers

Editorial

Current Vascular Pharmacology, 2016, Vol. 14, No. 1

77

30-60 mL/min/1.73 m2, is related to CVD morbidity and mortality [28-30]. Therefore, in terms of CVD risk, it would be interesting to evaluate if any eGFR changes occurred in the SCALE Obesity and Prediabetes study [2]. However, the significant overestimation of true GFR by the relevant estimated values in obese individuals should be taken into account [31]. The beneficial liraglutide-related effects on glucose control were greater in patients with prediabetes than those without. Furthermore, at the end of the study (56 weeks), fewer patients on liraglutide had prediabetes and type 2 diabetes compared with the placebo group [2]. Statins are known to increase the risk of new-onset diabetes (NOD) [32, 33]. In contrast, ezetimibe and fibrates may improve glucose metabolism [34]. In the SCALE Obesity and Prediabetes study [2], there were patients on lipid-lowering drugs (although the authors do not specify which). Based on the observed liraglutide-related beneficial impact on glucose metabolism, it would have been interesting to also evaluate its effect on statin-induced NOD. It follows that, if liraglutide 3.0 mg was shown to prevent this drug-related NOD, this would have important clinical implications in statin-treated patients. Side effects (mainly nausea and diarrhoea), with mild to moderate severity, were more common in the liraglutide-treated patients compared with placebo. Hypoglycaemias were more frequent in the liraglutide group compared with placebo (11.9 vs. 3.3% for all events; 1.3 vs. 1.0% for spontaneously reported; 3.6 vs. 0.8% for those reported at fasting plasma glucose visit and 8.3 vs. 1.4% for those reported at oral glucose tolerance test visit, respectively) (see appendix) [2]. However, the authors do not report any statistics with regard to hypoglycaemias. We performed Chi-square test (with Yates correction) that showed a significant difference in all hypoglycaemic events between the 2 groups (p < 0.0001). None of these hypoglycaemic events was serious or required third-party assistance. Resting heart rate (HR) was also slightly increased in the liraglutide group [2.5±9.8 vs 0.1±9.5 beats per min (bpm), respectively; p < 0.001]. The upregulation of HR by liraglutide appears to occur within the first few weeks and then reaches a plateau. Glucagon-like peptide-1 (GLP-1) receptors are found in the myocytes of the sinoatrial node, thus possibly explaining the increased HR observed with liraglutide administration [35]. However, studies investigating the direct effect of liraglutide on the automatism of the sinoatrial node are lacking. It should be noted that elevated HR (with no lower threshold reported) has been associated with an increased risk for coronary atherosclerosis, CVD morbidity (especially heart failure) and mortality as well as with total mortality [36, 37]. The reported association between the elevation of HR and CVD events are possibly attributed at least in part to the downregulation of the Parasympathetic Nervous System (PNS) and/or upregulation of the Sympathetic Nervous System (SNS) [37]. Importantly, the presumed mechanism of HR upregulation by liraglutide involves the activation of GLP-1 receptors in the sinoatrial node and thus exposes the heart to a lower cardiovascular stress compared with the modulation of PNS and SNS, which can also lead to proarrhythmia, increased inotropy and elevation of blood pressure [38]. From this point of view the prognostic significance of the upregulation of HR by liraglutide should be differentiated from the one of other antiobesity drugs, which increase HR through adrenergic activation, such as phentermine/topiramate, naltrexone/bupropion and sibutramine. Nevertheless, the elevation of HR by liraglutide may be an important consideration for the achievement of the target HR in obese patients with coronary artery disease. Other antiobesity drugs have also been shown to increase HR including the combination of phentermine and topiramate extended release [39, 40] (mean increase 1.3 and 1.7 bpm on the 7.5/46 and 15/92 mg dose, respectively [41]) and the 32 mg naltrexone/360 mg bupropion combination (increase up to 2 bpm) [42, 43]. Sibutramine, despite its beneficial metabolic effects [44], was found to raise HR in the Sibutramine Cardiovascular Outcomes (SCOUT) trial (mean differences compared with placebo were from 2.2 to 3.7 bpm) [45]. In this study, sibutramine treatment was also associated with an increased risk for CVD morbidity, a finding that led to its withdrawal from the market. In contrast, lorcaserin has not been reported to affect HR [46]. Hypocaloric diet plus orlistat has been shown to induce greater decrease in HR compared with hypocaloric diet plus placebo, which could be attributed to the greater orlistat-induced weight loss, rather than to a direct effect of orlistat on sinoatrial node automatism [47, 48]. Night-time HR changes may be even more strongly associated with inflammation [49]. The clinical implications of drug-related changes in HR remain to be established. However, for liraglutide, any disadvantage related to the increased HR may be more than compensated for by the beneficial effects described above. In another recent study, 3-months treatment of liraglutide in non-diabetic obese individuals significantly improved binge eating as well as obesity, glucose and lipid parameters but also increased plasma ghrelin levels compared with placebo [50]. Ghrelin is a gut hormone involved in appetite control [51] and its levels may be affected by antidiabetic drugs including liraglutide (at the dose of 1.2 mg/day) [52, 53]. Therefore, it would have been useful to measure ghrelin levels in the recently published SCALE Obesity and Prediabetes study. Overall, the findings of the SCALE Obesity and Prediabetes study are in accordance with both previous and current reviews on the efficacy, safety and tolerability of liraglutide for the management of obesity [54-59]. Importantly, the number of participants (n = 3731) in the SCALE Obesity and Prediabetes study permits to reach relatively firm conclusions regarding the safety of liraglutide 3.0 mg for a treatment period of 56 weeks. Taking into account that antiobesity drugs are not usually be taken for many years, the long term safety of this treatment should be investigated (e.g. by studies and surveillance programmes) for the detection of potential adverse effects after the cessation of liraglutide 3.0 mg treatment, rather than through extended studies using liraglutide 3.0 mg therapy. In conclusion, liraglutide 3.0 mg/day seems a promising antiobesity agent exerting beneficial metabolic effects. Future studies may further explore its impact on several cardiometabolic risk factors such as SUA, eGFR, sdLDL, PPL, MetS and NAFLD as well as on HR. Therefore, its long term efficacy and safety remains to be established.

78 Current Vascular Pharmacology, 2016, Vol. 14, No. 1

Editorial

COMPETING INTEREST This editorial was written independently. The authors did not receive financial or professional help with the preparation of the manuscript. NK has given talks, attended conferences and participated in trials sponsored by Amgen, Astra Zeneca, Libytec, Novo Nordisk, MSD and Novartis. GAC declares that he has no conflict of interest. DNK has given talks, attended conferences and participated in trials sponsored by Amgen, Angelini, MSD, BGP, ELPEN and Unipharma. REFERENCES [1] [2] [3] [4] [5] [6] [7] [8] [9] [10] [11] [12] [13] [14] [15]

[16] [17] [18] [19] [20] [21] [22] [23] [24] [25] [26] [27] [28] [29] [30] [31] [32] [33] [34]

Christou GA, Katsiki N, Kiortsis DN. The current role of liraglutide in the pharmacotherapy of obesity. Curr Vasc Pharmacol 2015 Jun 15. [Epub ahead of print] Pi-Sunyer X, Astrup A, Fujioka K, et al. SCALE Obesity and PrediabetesNN8022-1839 Study Group. A Randomized, Controlled Trial of 3.0 mg of Liraglutide in Weight Management. N Engl J Med 2015; 373: 11-22. American Diabetes Association. Diagnosis and classification of diabetes mellitus. Diabetes Care 2010; 33: S62-9. Kong F, Chen YF, Jin K. A bias correction in testing treatment efficacy under informative dropout in clinical trials. J Biopharm Stat 2009; 19: 9801000. Mikhailidis DP, Elisaf M, Rizzo M, et al. "European panel on low density lipoprotein (LDL) subclasses": a statement on the pathophysiology, atherogenicity and clinical significance of LDL subclasses: executive summary. Curr Vasc Pharmacol 2011; 9: 531-2. Mikhailidis DP, Elisaf M, Rizzo M, et al. "European panel on low density lipoprotein (LDL) subclasses": a statement on the pathophysiology, atherogenicity and clinical significance of LDL subclasses. Curr Vasc Pharmacol 2011; 9: 533-71. Nikolic D, Katsiki N, Montalto G, et al. Lipoprotein subfractions in metabolic syndrome and obesity: clinical significance and therapeutic approaches. Nutrients 2013; 5: 928-48. Ariel D, Kim SH, Abbasi F, et al. Effect of liraglutide administration and a calorie-restricted diet on lipoprotein profile in overweight/obese persons with prediabetes. Nutr Metab Cardiovasc Dis 2014; 24: 1317-22. Stefanutti C, Labbadia G, Athyros VG. Hypertriglyceridaemia, postprandial lipaemia and non-HDL cholesterol. Curr Pharm Des 2014; 20: 6238-48. Kolovou GD, Mikhailidis DP, Kovar J, et al. Assessment and clinical relevance of non-fasting and postprandial triglycerides: an expert panel statement. Curr Vasc Pharmacol 2011; 9: 258-70. Voukali M, Kastrinelli I, Stragalinou S, et al. Study of postprandial lipaemia in type 2 diabetes mellitus: exenatide versus liraglutide. J Diabetes Res 2014; 2014: 304032. Hermansen K, Bækdal TA, Düring M, et al. Liraglutide suppresses postprandial triglyceride and apolipoprotein B48 elevations after a fat-rich meal in patients with type 2 diabetes: a randomized, double-blind, placebo-controlled, cross-over trial. Diabetes Obes Metab 2013; 15: 1040-8. Katsiki N, Athyros VG, Karagiannis A, Mikhailidis DP. Characteristics other than the diagnostic criteria associated with metabolic syndrome: an overview. Curr Vasc Pharmacol. 2014; 12: 627-41. Katsiki N, Athyros VG, Karagiannis A, Mikhailidis DP. Metabolic syndrome and non-cardiac vascular diseases: an update from human studies. Curr Pharm Des 2014; 20: 4944-52. Alberti KG, Eckel RH, Grundy SM, et al. International Diabetes Federation Task Force on Epidemiology and Prevention; Hational Heart, Lung, and Blood Institute; American Heart Association; World Heart Federation; International Atherosclerosis Society; International Association for the Study of Obesity. Harmonizing the metabolic syndrome: a joint interim statement of the International Diabetes Federation Task Force on Epidemiology and Prevention; National Heart, Lung, and Blood Institute; American Heart Association; World Heart Federation; International Atherosclerosis Society; and International Association for the Study of Obesity. Circulation 2009; 120: 1640-5. Athyros VG, Tziomalos K, Katsiki N, Doumas M, Karagiannis A, Mikhailidis DP. Cardiovascular risk across the histological spectrum and the clinical manifestations of non-alcoholic fatty liver disease: An update. World J Gastroenterol 2015; 21: 6820-34. Athyros VG, Katsiki N, Karagiannis A. Nonalcoholic fatty liver disease and severity of cardiovascular disease manifestations. Angiology 2013; 64: 572-5. Katsiki N, Athyros VG, Karagiannis A, Wierzbicki AS, Mikhailidis DP. Should we expand the concept of coronary heart disease equivalents? Curr Opin Cardiol 2014; 29: 389-95. Lioudaki E, Ganotakis ES, Mikhailidis DP. Liver enzymes: potential cardiovascular risk markers? Curr Pharm Des 2011; 17: 3632-43. Athyros VG, Katsiki N, Karagiannis A, Mikhailidis DP. Are statins 'IDEAL' for non-alcoholic fatty liver disease? Curr Med Res Opin 2014; 30: 22931. Athyros VG, Katsiki N, Karagiannis A, Mikhailidis DP. Statins and nonalcoholic fatty liver disease: a bright future? Expert Opin Investig Drugs 2013; 22: 1089-93. Katsiki N, Athyros VG, Karagiannis A, Mikhailidis DP. The role of statins in the treatment of type 2 diabetes mellitus: an update. Curr Pharm Des 2014; 20: 3665-74. Eguchi Y, Kitajima Y, Hyogo H, et al. Japan Study Group for NAFLD (JSG-NAFLD). Pilot study of liraglutide effects in non-alcoholic steatohepatitis and non-alcoholic fatty liver disease with glucose intolerance in Japanese patients (LEAN-J). Hepatol Res 2015; 45: 269-78. Armstrong MJ, Houlihan DD, Rowe IA, et al. Safety and efficacy of liraglutide in patients with type 2 diabetes and elevated liver enzymes: individual patient data meta-analysis of the LEAD program. Aliment Pharmacol Ther 2013; 37: 234-42. Olaywi M, Bhatia T, Anand S, Singhal S. Novel anti-diabetic agents in non-alcoholic fatty liver disease: a mini-review. Hepatobiliary Pancreat Dis Int 2013; 12: 584-8. Katsiki N, Karagiannis A, Athyros VG, Mikhailidis DP. Hyperuricaemia: more than just a cause of gout? J Cardiovasc Med (Hagerstown) 2013; 14: 397-402. Rizzo M, Obradovic M, Labudovic-Borovic M, et al. Uric acid metabolism in pre-hypertension and the metabolic syndrome. Curr Vasc Pharmacol. 2014; 12: 572-85. Alani H, Tamimi A, Tamimi N. Cardiovascular co-morbidity in chronic kidney disease: Current knowledge and future research needs. World J Nephrol 2014; 3: 156-68. Athyros VG, Katsiki N, Karagiannis A, Mikhailidis DP. Editorial: should chronic kidney disease be considered as a coronary heart disease equivalent? Curr Vasc Pharmacol 2012; 10: 374-7. Athyros VG, Katsiki N, Karagiannis A, Mikhailidis DP. Stage of chronic kidney disease and severity of coronary heart disease manifestation. Expert Opin Pharmacother 2012; 13: 457-60. Pai MP. Estimating the glomerular filtration rate in obese adult patients for drug dosing. Adv Chronic Kidney Dis 2010; 17: e53-62. Athyros VG, Mikhailidis DP. Pharmacotherapy: statins and new-onset diabetes mellitus--a matter for debate. Nat Rev Endocrinol 2012; 8: 133-4. Katsiki N, Rizzo M, Mikhailidis DP, Mantzoros CS. New-onset diabetes and statins: throw the bath water out, but, please, keep the baby! Metabolism 2015; 64: 471-5. Zafrir B, Jain M. Lipid-lowering therapies, glucose control and incident diabetes: evidence, mechanisms and clinical implications. Cardiovasc Drugs Ther 2014; 28: 361-77.

Editorial [35] [36] [37] [38] [39] [40] [41] [42] [43] [44] [45] [46] [47] [48] [49] [50] [51] [52] [53] [54] [55] [56] [57] [58] [59]

Current Vascular Pharmacology, 2016, Vol. 14, No. 1

79

Pyke C, Heller RS, Kirk RK, et al. GLP-1 receptor localization in monkey and human tissue: novel distribution revealed with extensively validated monoclonal antibody. Endocrinology 2014; 155: 1280-90. Giannoglou GD, Chatzizisis YS, Zamboulis C, Parcharidis GE, Mikhailidis DP, Louridas GE. Elevated heart rate and atherosclerosis: an overview of the pathogenetic mechanisms. Int J Cardiol 2008; 126: 302-12. Ho JE, Larson MG, Ghorbani A, et al. Long-term cardiovascular risks associated with an elevated heart rate: the Framingham Heart Study. J Am Heart Assoc 2014; 3: e000668. Vaseghi M, Shivkumar K. The role of the autonomic nervous system in sudden cardiac death. Prog Cardiovasc Dis 2008; 50: 404-19. Alfaris N, Minnick AM, Hopkins CM, Berkowitz RI, Wadden TA. Combination phentermine and topiramate extended release in the management of obesity. Expert Opin Pharmacother 2015; 16: 1263-74. Kiortsis DN. A review of the metabolic effects of controlled-release Phentermine/Topiramate. Hormones (Athens) 2013; 12: 507-16. Garvey WT, Ryan DH, Look M, et al. Two-year sustained weight loss and metabolic benefits with controlled-release phentermine/topiramate in obese and overweight adults (SEQUEL): a randomized, placebo-controlled, phase 3 extension study. Am J Clin Nutr 2012; 95: 297-308. Christou GA, Kiortsis DN. The efficacy and safety of the naltrexone/bupropion combination for the treatment of obesity: an update. Hormones (Athens) 2015; 14: 370-5. Katsiki N, Hatzitolios AI, Mikhailidis DP. Naltrexone sustained-release (SR) + bupropion SR combination therapy for the treatment of obesity: 'a new kid on the block'? Ann Med 2011; 43: 249-58. Filippatos TD, Kiortsis DN, Liberopoulos EN, Mikhailidis DP, Elisaf MS. A review of the metabolic effects of sibutramine. Curr Med Res Opin 2005; 21: 457-68. James WP, Caterson ID, Coutinho W, et al. SCOUT Investigators. Effect of sibutramine on cardiovascular outcomes in overweight and obese subjects. N Engl J Med 2010; 363: 905-17. Chan EW, He Y, Chui CS, Wong AY, Lau WC, Wong IC. Efficacy and safety of lorcaserin in obese adults: a meta-analysis of 1-year randomized controlled trials (RCTs) and narrative review on short-term RCTs. Obes Rev 2013; 14: 383-92. Sharma AM, Golay A. Effect of orlistat-induced weight loss on blood pressure and heart rate in obese patients with hypertension. J Hypertens 2002; 20: 1873-8. Kiortsis DN, Filippatos TD, Elisaf MS. The effects of orlistat on metabolic parameters and other cardiovascular risk factors. Diabetes Metab 2005; 31: 15-22. Hartaigh B, Gaksch M, Kienreich K, et al. Associations of daytime, nighttime, and 24-hour heart rate with four distinct markers of inflammation in hypertensive patients: the Styrian Hypertension Study. J Clin Hypertens (Greenwich) 2014; 16: 856-61. Robert SA, Rohana AG, Shah SA, Chinna K, Wan Mohamud WN, Kamaruddin NA. Improvement in binge eating in non-diabetic obese individuals after 3 months of treatment with liraglutide - A pilot study. Obes Res Clin Pract 2015; 9: 301-4. Pinkney J. The role of ghrelin in metabolic regulation. Curr Opin Clin Nutr Metab Care 2014; 17: 497-502. Katsiki N, Mikhailidis DP, Gotzamani-Psarrakou A, Yovos JG, Karamitsos D. Effect of various treatments on leptin, adiponectin, ghrelin and neuropeptide Y in patients with type 2 diabetes mellitus. Expert Opin Ther Targets. 2011; 15: 401-20. Rizzo M, Abate N, Chandalia M, et al. Liraglutide reduces oxidative stress and restores heme oxygenase-1 and ghrelin levels in patients with type 2 diabetes: a prospective pilot study. J Clin Endocrinol Metab 2015; 100: 603-6. Clements JN, Shealy KM. Liraglutide: An injectable option for the management of obesity. Ann Pharmacother 2015; 49(8): 938-44. Fujioka K. Safety and tolerability of medications approved for chronic weight management. Obesity (Silver Spring) 2015; 23: S7-11. Iepsen EW, Torekov SS, Holst JJ. Liraglutide for Type 2 diabetes and obesity: a 2015 update. Expert Rev Cardiovasc Ther 2015; 13: 753-67. Ryan D, Acosta A. GLP-1 receptor agonists: Nonglycemic clinical effects inweight loss and beyond. Obesity (Silver Spring) 2015; 23: 1119-29. Scott LJ. Liraglutide: a review of its use in the management of obesity. Drugs 2015; 75: 899-910. Bray GA. Obesity: Liraglutide-another weapon in the war against obesity? Nat Rev Endocrinol 2015; 11: 569-70.

Dimitrios N Kiortsis Professor of Physiology Laboratory of Physiology, Medical School University of Ioannina 45110 Ioannina, Greece Phone: 0030 2651007551 Fax: 0030 2651007850 E-mail: [email protected]