Tolerability of nausea and vomiting and associations with weight loss in a randomized trial of liraglutide in obese, non-diabetic adults

M E J Lean, R Carraro, N Finer, H Hartvig, M L Lindegaard, S Rössner, L Van Gaal, A Astrup, NN8022-1807 Investigators, Luc Van Gaal, Stepan Svacina, Marie Kunesova, Arne Astrup, Bjørn Richelsen, Kjeld Hermansen, Steen Madsbad, Aila Rissanen, Leo Niskanen, Markku Savolainen, Mazin Al Hakim, Guillem Cuatrecasas Cambra, Belen Sadaba, Raffaele Carraro, Basilio Moreno, Stephan Rossner, Martin Ridderstråle, Michael Lean, Nick Finer, Mike Sampson, M E J Lean, R Carraro, N Finer, H Hartvig, M L Lindegaard, S Rössner, L Van Gaal, A Astrup, NN8022-1807 Investigators, Luc Van Gaal, Stepan Svacina, Marie Kunesova, Arne Astrup, Bjørn Richelsen, Kjeld Hermansen, Steen Madsbad, Aila Rissanen, Leo Niskanen, Markku Savolainen, Mazin Al Hakim, Guillem Cuatrecasas Cambra, Belen Sadaba, Raffaele Carraro, Basilio Moreno, Stephan Rossner, Martin Ridderstråle, Michael Lean, Nick Finer, Mike Sampson

Abstract

Background: Liraglutide 3.0 mg, with diet and exercise, produced substantial weight loss over 1 year that was sustained over 2 years in obese non-diabetic adults. Nausea was the most frequent side effect.

Objective: To evaluate routinely collected data on nausea and vomiting among individuals on liraglutide and their influence on tolerability and body weight.

Design: A randomized, placebo-controlled, double-blind 20-week study with an 84-week extension (sponsor unblinded at 20 weeks, open-label after 1 year) in eight European countries (Clinicaltrials.gov: NCT00422058).

Subjects: After commencing a 500-kcal/day deficit diet plus exercise, 564 participants (18-65 years, body mass index (BMI) 30-40 kg m(-2)) were randomly assigned (after a 2-week run-in period) to once-daily subcutaneous liraglutide (1.2, 1.8, 2.4 or 3.0 mg), placebo or open-label orlistat (120 mg × 3 per day). After 1 year, participants on liraglutide/placebo switched to liraglutide 2.4 mg, and subsequently, to liraglutide 3.0 mg (based on 20-week and 1-year results, respectively).

Results: The intention-to-treat population comprised 561 participants (n=90-98 per arm, age 45.9±10.3 years, BMI 34.8±2.7 kg m(-2) (mean±s.d.)). In year 1, more participants reported ⩾1 episode of nausea/vomiting on treatment with liraglutide 1.2-3.0 mg (17-38%) than with placebo or orlistat (both 4%, P⩽0.001). Most episodes occurred during dose escalation (weeks 1-6), with 'mild' or 'moderate' symptoms. Among participants on liraglutide 3.0 mg, 48% reported some nausea and 13% some vomiting, with considerable variation between countries, but only 4 out of 93 (4%) reported withdrawals. The mean 1-year weight loss on treatment with liraglutide 3.0 mg from randomization was 9.2 kg for participants reporting nausea/vomiting episodes, versus 6.3 kg for those with none (a treatment difference of 2.9 kg (95% confidence interval 0.5-5.3); P=0.02). Both weight losses were significantly greater than the respective weight losses for participants on placebo (P<0.001) or orlistat (P<0.05). Quality-of-life scores at 20 weeks improved similarly with or without nausea/vomiting on treatment with liraglutide 3.0 mg.

Conclusion: Transient nausea and vomiting on treatment with liraglutide 3.0 mg was associated with greater weight loss, although symptoms appeared tolerable and did not attenuate quality-of-life improvements. Improved data collection methods on nausea are warranted.

Figures

Figure 1
Figure 1
Study design. *During 20–52 weeks, participants/investigators remained blinded to liraglutide/placebo treatment, but the sponsor was unblinded; after 1 year, all were unblinded.
Figure 2
Figure 2
(a) The proportion of individuals reporting nausea/vomiting at any time during year 1 (logistic regression analysis). Data are estimated means. Odds ratios (OR) versus placebo/orlistat are shown, together with 95% CI. The proportion of participants reporting nausea (b) and vomiting (c) by country in year 1. (d) Severity of nausea and vomiting episodes reported in year 1. (e) The proportion of individuals with nausea at any time during 2 years of treatment, by week. Safety analysis set.
Figure 3
Figure 3
Weight change at year 1 (a) and year 2 (b) in individuals with or without at least one episode of nausea or vomiting. Mean changes (analysis of covariance) are shown for the intention-to-treat population with the last observation carried forward. Estimated treatment differences (ETD) are shown, together with 95% CI.
Figure 4
Figure 4
Mean (analysis of covariance) changes (positive=improved) in quality-of-life scores (IWQoL-Lite) among individuals on liraglutide 3.0 mg at 20 weeks, in individuals with or without at least one episode of nausea or vomiting (intention-to-treat population with the last observation carried forward). IWQoL-Lite scores ranged from 0 (worst) to 100 (best).

References

    1. Mayer MA, Hocht C, Puyo A, Taira CA. Recent advances in obesity pharmacotherapy. Curr Clin Pharmacol. 2009;4:53–61.
    1. Murray CJL, Lozano R, Naghavi M, Flaxman AD, Michaud C, Ezzati M, et al. Disability-adjusted life years (DALYs) for 291 diseases and injuries in 21 regions, 1990–2010: a systematic analysis for the Global Burden of Disease Study 2010. Lancet. 2012;380:2197–2223.
    1. Flegal KM, Graubard BI, Williamson DF, Gail MH. Cause-specific excess deaths associated with underweight, overweight, and obesity. J Am Med Assoc. 2007;298:2028–2037.
    1. Kaukua J, Pekkarinen T, Sane T, Mustajoki P. Health-related quality of life in obese outpatients losing weight with very-low-energy diet and behaviour modification: a 2-y follow-up study. Int J Obes Relat Metab Disord. 2003;27:1072–1080.
    1. Hassan MK, Joshi AV, Madhavan SS, Amonkar MM. Obesity and health-related quality of life: a cross-sectional analysis of the US population. Int J Obes Relat Metab Disord. 2003;27:1227–1232.
    1. Counterweight Project Team Evaluation of the Counterweight Programme for obesity management in primary care: a starting point for continuous improvement. Br J Gen Pract. 2008;58:548–554.
    1. Knowler WC, Barrett-Connor E, Fowler SE, Hamman RF, Lachin JM, Walker EA, et al. Reduction in the incidence of type 2 diabetes with lifestyle intervention or metformin. N Engl J Med. 2002;346:393–403.
    1. National Institutes of Health Clinical guidelines on the identification, evaluation, and treatment of overweight and obesity in adults—The Evidence Report. Obes Res. 1998;6 (Suppl 2:51S–209S.
    1. Mertens IL, Van Gaal LF. Overweight, obesity, and blood pressure: the effects of modest weight reduction. Obes Res. 2000;8:270–278.
    1. Neter JE, Stam BE, Kok FJ, Grobbee DE, Geleijnse JM. Influence of weight reduction on blood pressure: a meta-analysis of randomized controlled trials. Hypertension. 2003;42:878–884.
    1. Look AHEAD Research Group. Wing RR. Long-term effects of a lifestyle intervention on weight and cardiovascular risk factors in individuals with type 2 diabetes mellitus: four-year results of the Look AHEAD trial. Arch Intern Med. 2010;170:1566–1575.
    1. Padwal RS, Majumdar SR. Drug treatments for obesity: orlistat, sibutramine, and rimonabant. Lancet. 2007;369:71–77.
    1. Astrup A. Drug management of obesity - efficacy versus safety. N Engl J Med. 2010;363:288–290.
    1. Field BC, Wren AM, Cooke D, Bloom SR. Gut hormones as potential new targets for appetite regulation and the treatment of obesity. Drugs. 2008;68:147–163.
    1. Flint A, Raben A, Astrup A, Holst JJ. Glucagon-like peptide 1 promotes satiety and suppresses energy intake in humans. J Clin Invest. 1998;101:515–520.
    1. Meeran K, O'Shea D, Edwards CM, Turton MD, Heath MM, Gunn I, et al. Repeated intracerebroventricular administration of glucagon-like peptide-1-(7-36) amide or exendin-(9-39) alters body weight in the rat. Endocrinology. 1999;140:244–250.
    1. Gutzwiller JP, Goke B, Drewe J, Hildebrand P, Ketterer S, Handschin D, et al. Glucagon-like peptide-1: a potent regulator of food intake in humans. Gut. 1999;44:81–86.
    1. Zander M, Madsbad S, Madsen JL, Holst JJ. Effect of 6-week course of glucagon-like peptide 1 on glycaemic control, insulin sensitivity, and beta-cell function in type 2 diabetes: a parallel-group study. Lancet. 2002;359:824–830.
    1. Tang-Christensen M, Larsen PJ, Goke R, Fink-Jensen A, Jessop DS, Moller M, et al. Central administration of GLP-1-(7-36) amide inhibits food and water intake in rats. Am J Physiol. 1996;271:R848–R856.
    1. Turton MD, O'Shea D, Gunn I, Beak SA, Edwards CM, Meeran K, et al. A role for glucagon-like peptide-1 in the central regulation of feeding. Nature. 1996;379:69–72.
    1. Williams DL, Baskin DG, Schwartz MW. Evidence that Intestinal Glucagon-Like Peptide-1 Plays a Physiological Role in Satiety. Endocrinology. 2009;150:1680–1687.
    1. Flint A, Raben A, Ersboll AK, Holst JJ, Astrup A. The effect of physiological levels of glucagon-like peptide-1 on appetite, gastric emptying, energy and substrate metabolism in obesity. Int J Obes. 2001;25:781–792.
    1. Garber A, Henry R, Ratner R, Garcia-Hernandez PA, Rodriguez-Pattzi H, Olvera-Alvarez I, et al. Liraglutide versus glimepiride monotherapy for type 2 diabetes (LEAD-3 Mono): a randomised, 52-week, phase III, double-blind, parallel-treatment trial. Lancet. 2009;373:473–481.
    1. Vilsboll T, Zdravkovic M, Le-Thi T, Krarup T, Schmitz O, Courreges JP, et al. Liraglutide, a long-acting human glucagon-like peptide-1 analog, given as monotherapy significantly improves glycemic control and lowers body weight without risk of hypoglycemia in patients with type 2 diabetes. Diabetes Care. 2007;30:1608–1610.
    1. Astrup A, Rossner S, Van Gaal L, Rissanen A, Niskanen L, Al Hakim M, et al. Effects of liraglutide in the treatment of obesity: a randomised, double-blind, placebo-controlled study. Lancet. 2009;374:1606–1616.
    1. Astrup A, Carraro R, Finer N, Harper A, Kunesova M, Lean ME, et al. Safety, tolerability and sustained weight loss over 2 years with the once-daily human GLP-1 analog, liraglutide. Int J Obes (Lond) 2011;36:843–854.
    1. Garber AJ. Incretin-based therapies in the management of type 2 diabetes: rationale and reality in a managed care setting. Am J Manag Care. 2010;16 (7 Suppl:S187–S194.
    1. Isidro ML, Cordido F. Drug Treatment of obesity: Established and emerging therapies. Mini-Rev Med Chem. 2009;9:664–673.
    1. Davies MJ, Kela R, Khunti K. Liraglutide - overview of the preclinical and clinical data and its role in the treatment of type 2 diabetes. Diab Obesity Metabol. 2011;13:207–220.
    1. World Medical Association World Medical Association Declaration of Helsinki: Ethical principles for medical research involving human subjects. JAMA. 2000;284:3043–3045.
    1. Lean ME, James WP. Prescription of diabetic diets in the 1980s. Lancet. 1986;1:723–725.
    1. Kolotkin RL, Crosby RD, Kosloski KD, Williams GR. Development of a brief measure to assess quality of life in obesity. Obes Res. 2001;9:102–111.
    1. Joelson S, Joelson IB, Wallander MA. Geographical variation in adverse event reporting rates in clinical trials. Pharmacoepidemiol Drug Saf. 1997;6 (Suppl 3:S31–S35.
    1. Raisch DW, Troutman WG, Sather MR, Fudala PJ. Variability in the assessment of adverse events in a multicenter clinical trial. Clin Ther. 2001;23:2011–2020.
    1. Barrera JG, Sandoval DA, D'Alessio DA, Seeley RJ. GLP-1 and energy balance: an integrated model of short-term and long-term control. Nat Rev Endocrinol. 2011;7:507–516.
    1. Williams DL. Minireview: Finding the sweet spot: Peripheral versus central glucagon-like peptide 1 action in feeding and glucose homeostasis. Endocrinology. 2009;150:2997–3001.
    1. Horowitz M, Flint A, Jones KL, Hindsberger C, Rasmussen MF, Kapitza C, et al. Effect of the once-daily human GLP-1 analogue liraglutide on appetite, energy intake, energy expenditure and gastric emptying in type 2 diabetes. Diabetes Res Clin Pract. 2012;97:258–266.
    1. Flint A, Kapitza C, Zdravkovic M.The once-daily human GLP-1 analogue liraglutide impacts appetite and energy intake in patients with type 2 diabetes after short-term treatment Diab Obes Metabol 2013. doi: 10.1111/dom.12108
    1. Seimon RV, Lange K, Little TJ, Brennan IM, Pilichiewicz AN, Feltrin KL, et al. Pooled-data analysis identifies pyloric pressures and plasma cholecystokinin concentrations as major determinants of acute energy intake in healthy, lean men. Am J Clin Nutr. 2010;92:61–68.
    1. Nauck MA, Meier JJ. Glucagon-like peptide 1 and its derivatives in the treatment of diabetes. Regul Pept. 2005;128:135–148.
    1. Vilsboll T, Krarup T, Madsbad S, Holst JJ. No reactive hypoglycaemia in Type 2 diabetic patients after subcutaneous administration of GLP-1 and intravenous glucose. Diabet Med. 2001;18:144–149.
    1. Neumiller JJ, Campbell RK. Liraglutide: a once-daily incretin mimetic for the treatment of type 2 diabetes mellitus. Ann Pharmacother. 2009;43:1433–1444.
    1. Orskov C, Poulsen SS. M°ller M, Holst JJ. Glucagon-like peptide I receptors in the subfornical organ and the area postrema are accessible to circulating glucagon-like peptide I. Diabetes. 1996;45:832–835.
    1. Nauck MA, Kemmeries G, Holst JJ, Meier JJ. Rapid tachyphylaxis of the glucagon-like peptide 1-induced deceleration of gastric emptying in humans. Diabetes. 2011;60:1561–1565.
    1. Jelsing J, Vrang N, Hansen G, Ravn K, Tang-Christensen M, Knudsen LB. Liraglutide: Short lived effect on gastric emptying - long lasting effects on body-weight. Diab Obes Metabol. 2012;14:531–538.
    1. Hussain Z. Quigley EMM. Gastrointestinal issues in the assessment and management of the obese patient. Gastroenterol Hepatol. 2007;3:559–569.

Source: PubMed

3
Abonnieren