Skip to main content
Log in

Benefit-Risk Assessment of Obesity Drugs: Focus on Glucagon-like Peptide-1 Receptor Agonists

  • Review Article
  • Published:
Drug Safety Aims and scope Submit manuscript

A Correction to this article was published on 30 April 2019

This article has been updated

Abstract

The prevalence of obesity and related comorbidities is increasing worldwide. Furthermore, clinically meaningful body weight losses has proven difficult to achieve and especially to maintain through sustained lifestyle change in the form of diet and exercise. Pharmacotherapy against obesity is a non-invasive treatment as an adjunct to lifestyle changes, but approved anti-obesity drugs are currently few. This article reviews the major anti-obesity drugs and the benefit-risk profiles of the long-acting glucagon-like peptide-1 receptor agonists (GLP-1 RAs) liraglutide and semaglutide (a modified version of liraglutide with longer half-life and tripled receptor affinity). Generally, GLP-1 RAs are well tolerated and induce significant weight loss and lowering of comorbidities. Studies with liraglutide 3.0 mg/day have shown an average placebo-subtracted weight loss of 5.5 kg (range 4.6–5.9) in 1- to 3-year duration trials. One trial using semaglutide 0.4 mg once daily reported an average weight loss of 11.6% (~ 13.1 kg) after 1 year. Furthermore, semaglutide induced a ~ 6 percentage point larger placebo-subtracted body weight loss in a head-to-head comparison with liraglutide (11.6 vs. 5.5% weight loss, respectively). The safety profiles for both drugs were similar, with transient gastrointestinal disorders being the most commonly reported adverse events. The longest running trial and the most recent trials have not raised any new safety concerns. Long-term trials and post-marketing surveillance is warranted to fully assess both long-term efficacy and safety. Future combinational therapies of mimicked gut hormones involved in regulation of energy homeostasis and/or additional lifestyle change in the form of exercise might further improve efficacy.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Institutional subscriptions

Similar content being viewed by others

Change history

  • 30 April 2019

    In the original publication of this article, the following correction should be noted in Table 1.

References

  1. World Health Organization. Obesity and overweight. 2018. http://www.who.int/en/news-room/fact-sheets/detail/obesity-and-overweight. Accessed 28 Mar 2019.

  2. World Health Organization. Prevalence of obesity among adults, BMI ≥ 30, age-standardized Estimates by WHO region. 2017. http://apps.who.int/gho/data/view.main.REGION2480A?lang=en. Accessed 28 Mar 2019.

  3. Guh DP, Zhang W, Bansback N, Amarsi Z, Birmingham CL, Anis AH. The incidence of co-morbidities related to obesity and overweight: a systematic review and meta-analysis. BMC Public Health. 2009;9:88. https://doi.org/10.1186/1471-2458-9-88.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Pi-Sunyer X. The medical risks of obesity. Postgrad Med. 2009;121(6):21–33. https://doi.org/10.3810/pgm.2009.11.2074.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Finkelstein EA, Trogdon JG, Cohen JW, Dietz W. Annual medical spending attributable to obesity: payer-and service-specific estimates. Health Affairs (Project Hope). 2009;28(5):w822–31. https://doi.org/10.1377/hlthaff.28.5.w822.

    Article  Google Scholar 

  6. Peeters A, Barendregt JJ, Willekens F, Mackenbach JP, Al Mamun A, Bonneux L. Obesity in adulthood and its consequences for life expectancy: a life-table analysis. Ann Intern Med. 2003;138(1):24–32.

    Article  Google Scholar 

  7. Jensen MD, Ryan DH, Apovian CM, Ard JD, Comuzzie AG, Donato KA et al. 2013 AHA/ACC/TOS guideline for the management of overweight and obesity in adults: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines and The Obesity Society. J Am Coll Cardiol. 2014;63(25 Pt B):2985–3023. https://doi.org/10.1016/j.jacc.2013.11.004.

  8. The Look AHEAD Research Group. Eight-year weight losses with an intensive lifestyle intervention: the look AHEAD study. Obesity (Silver Spring, Md). 2014;22(1):5–13. https://doi.org/10.1002/oby.20662.

    Article  PubMed Central  Google Scholar 

  9. Wing RR, Lang W, Wadden TA, Safford M, Knowler WC, Bertoni AG, et al. Benefits of modest weight loss in improving cardiovascular risk factors in overweight and obese individuals with type 2 diabetes. Diabetes Care. 2011;34(7):1481–6. https://doi.org/10.2337/dc10-2415.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Fildes A, Charlton J, Rudisill C, Littlejohns P, Prevost AT, Gulliford MC. Probability of an obese person attaining normal body weight: cohort study using electronic health records. Am J Public Health. 2015;105(9):e54–9. https://doi.org/10.2105/AJPH.2015.302773.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Fried M, Yumuk V, Oppert JM, Scopinaro N, Torres A, Weiner R, et al. Interdisciplinary European guidelines on metabolic and bariatric surgery. Obes Surg. 2014;24(1):42–55. https://doi.org/10.1007/s11695-013-1079-8.

    Article  CAS  PubMed  Google Scholar 

  12. Garvey WT, Mechanick JI, Brett EM, Garber AJ, Hurley DL, Jastreboff AM, et al. American Association of Clinical Endocrinologists and American College of Endocrinology Comprehensive Clinical Practice Guidelines for medical care of patients with obesity. Endocr Pract. 2016;22(Suppl 3):1–203. https://doi.org/10.4158/EP161365.GL.

    Article  PubMed  Google Scholar 

  13. Onakpoya IJ, Heneghan CJ, Aronson JK. Post-marketing withdrawal of anti-obesity medicinal products because of adverse drug reactions: a systematic review. BMC Med. 2016;14(1):191. https://doi.org/10.1186/s12916-016-0735-y.

  14. Haslam D. Weight management in obesity—past and present. Int J Clin Pract. 2016;70(3):206–17. https://doi.org/10.1111/ijcp.12771.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Connolly HM, Crary JL, McGoon MD, Hensrud DD, Edwards BS, Edwards WD, et al. Valvular heart disease associated with fenfluramine-phentermine. N Engl J Med. 1997;337(9):581–8. https://doi.org/10.1056/nejm199708283370901.

    Article  CAS  PubMed  Google Scholar 

  16. US Food and Drug Administration. Full prescribing information Suprenza (phentermine hydrochloride) orally disintegrating tablet. https://www.accessdata.fda.gov/drugsatfda_docs/label/2013/202088s005lbl.pdf. Accesssed 28 Mar 2019.

  17. European Medicines Agency. The European Agency for the Evaluation of Medicinal Products. 1999. https://www.ema.europa.eu/documents/press-release/extraordinary-meeting-finalise-review-anorectic-agents_en.pdf. Accessed 28 Mar 2019.

  18. Van Gaal LF, Rissanen AM, Scheen AJ, Ziegler O, Rossner S. Effects of the cannabinoid-1 receptor blocker rimonabant on weight reduction and cardiovascular risk factors in overweight patients: 1-year experience from the RIO-Europe study. Lancet. 2005;365(9468):1389–97. https://doi.org/10.1016/s0140-6736(05)66374-x.

    Article  PubMed  Google Scholar 

  19. Christensen R, Kristensen PK, Bartels EM, Bliddal H, Astrup A. Efficacy and safety of the weight-loss drug rimonabant: a meta-analysis of randomised trials. Lancet. 2007;370(9600):1706–13. https://doi.org/10.1016/s0140-6736(07)61721-8.

    Article  CAS  PubMed  Google Scholar 

  20. European Medicines Agency. Acomplia. https://www.ema.europa.eu/en/medicines/human/EPAR/acomplia. Accessed 28 Mar 2019.

  21. European Medicines Agency. Zimulti. https://www.ema.europa.eu/medicines/human/EPAR/Zimulti. Accessed 28 Mar 2019.

  22. Rolls BJ, Shide DJ, Thorwart ML, Ulbrecht JS. Sibutramine reduces food intake in non-dieting women with obesity. Obes Res. 1998;6(1):1–11.

    Article  CAS  Google Scholar 

  23. Luque CA, Rey JA. The discovery and status of sibutramine as an anti-obesity drug. Eur J Pharmacol. 2002;440(2):119–28. https://doi.org/10.1016/S0014-2999(02)01423-1.

    Article  CAS  PubMed  Google Scholar 

  24. European Medicines Agency. European Medicines Agency recommends suspension of marketing authorisation for sibutramine. 2010. https://www.ema.europa.eu/news/european-medicines-agency-recommends-suspension-marketing-authorisation-sibutramine?fbclid=IwAR0j-JZEdrJ3qbW2iDOGiVJoiIPZVKuJSZkV5j9a438Lt9h6O6lMP0c34qA. Accessed 28 Mar 2019.

  25. US Food and Drug Administration. FDA Drug Safety Communication: FDA Recommends Against the Continued Use of Meridia (sibutramine). 2010. https://www.fda.gov/Drugs/DrugSafety/ucm228746.htm. Accessed 28 Mar 2019. 

  26. James WP, Caterson ID, Coutinho W, Finer N, Van Gaal LF, Maggioni AP, et al. Effect of sibutramine on cardiovascular outcomes in overweight and obese subjects. N Engl J Med. 2010;363(10):905–17. https://doi.org/10.1056/NEJMoa1003114.

    Article  CAS  PubMed  Google Scholar 

  27. US Food and Drug Administration. Center for Drug Evaluation and Research 208524Orig1s000. https://www.accessdata.fda.gov/drugsatfda_docs/nda/2016/208524Orig1s000Approv.pdf. Accessed 28 Mar 2019.

  28. European Medicines Agency. Withdrawal assessment report for Belviq. 2013. https://www.ema.europa.eu/documents/withdrawal-report/withdrawal-assessment-report-belviq_en.pdf. Accessed 28 Mar 2019. 

  29. Tecott LH, Sun LM, Akana SF, Strack AM, Lowenstein DH, Dallman MF, et al. Eating disorder and epilepsy in mice lacking 5-HT2c serotonin receptors. Nature. 1995;374(6522):542–6. https://doi.org/10.1038/374542a0.

    Article  CAS  PubMed  Google Scholar 

  30. Porter RJ, Dhir A, Macdonald RL, Rogawski MA. Chapter 39—mechanisms of action of antiseizure drugs. In: Stefan H, Theodore WH, editors. Handbook of clinical neurology. Amsterdam: Elsevier; 2012. p. 663–81.

    Google Scholar 

  31. European Medicines Agency. Refusal of the marketing authorisation for Qsiva (phentermine / topiramate). 2013. https://www.ema.europa.eu/documents/smop-initial/questions-answers-refusal-marketing-authorisation-qsiva-phentermine/topiramate_en.pdf. Accessed 28 Mar 2019.

  32. Arterburn DE, Crane PK, Veenstra DL. The efficacy and safety of sibutramine for weight loss: a systematic review. Arch Intern Med. 2004;164(9):994–1003. https://doi.org/10.1001/archinte.164.9.994.

    Article  CAS  PubMed  Google Scholar 

  33. Khera R, Murad MH, Chandar AK, Dulai PS, Wang Z, Prokop LJ, et al. Association of pharmacological treatments for obesity with weight loss and adverse events: a systematic review and meta-analysis. JAMA. 2016;315(22):2424–34. https://doi.org/10.1001/jama.2016.7602.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Davidson MH, Hauptman J, DiGirolamo M, et al. Weight control and risk factor reduction in obese subjects treated for 2 years with orlistat: a randomized controlled trial. JAMA. 1999;281(3):235–42. https://doi.org/10.1001/jama.281.3.235.

    Article  CAS  PubMed  Google Scholar 

  35. Apovian CM, Aronne L, Rubino D, Still C, Wyatt H, Burns C, et al. A randomized, phase 3 trial of naltrexone SR/bupropion SR on weight and obesity-related risk factors (COR-II). Obesity (Silver Spring, Md). 2013;21(5):935–43. https://doi.org/10.1002/oby.20309.

    Article  CAS  PubMed Central  Google Scholar 

  36. Agerso H, Jensen LB, Elbrond B, Rolan P, Zdravkovic M. The pharmacokinetics, pharmacodynamics, safety and tolerability of NN2211, a new long-acting GLP-1 derivative, in healthy men. Diabetologia. 2002;45(2):195–202. https://doi.org/10.1007/s00125-001-0719-z.

    Article  CAS  PubMed  Google Scholar 

  37. Madsbad S, Holst JJ. Treatment with GLP-1 receptor agonists. In: Bonora E, DeFronzo R, editors. Diabetes. Epidemiology, genetics, pathogenesis, diagnosis, prevention, and treatment. Cham: Springer International Publishing; 2018. p. 1–45.

    Google Scholar 

  38. Holst JJ. The physiology of glucagon-like peptide 1. Physiol Rev. 2007;87(4):1409–39. https://doi.org/10.1152/physrev.00034.2006.

    Article  CAS  PubMed  Google Scholar 

  39. Torekov SS, Madsbad S, Holst JJ. Obesity—an indication for GLP-1 treatment? Obesity pathophysiology and GLP-1 treatment potential. Obes Rev. 2011;12(8):593–601. https://doi.org/10.1111/j.1467-789X.2011.00860.x.

    Article  CAS  PubMed  Google Scholar 

  40. Madsbad S. The role of glucagon-like peptide-1 impairment in obesity and potential therapeutic implications. Diabetes Obes Metab. 2014;16(1):9–21. https://doi.org/10.1111/dom.12119.

    Article  CAS  PubMed  Google Scholar 

  41. Muscogiuri G, DeFronzo RA, Gastaldelli A, Holst JJ. Glucagon-like Peptide-1 and the Central/Peripheral Nervous System: Crosstalk in Diabetes. Trends Endocrinol Metab. 2017;28(2):88–103. https://doi.org/10.1016/j.tem.2016.10.001.

    Article  CAS  PubMed  Google Scholar 

  42. Iepsen EW, Zhang J, Thomsen HS, Hansen EL, Hollensted M, Madsbad S, et al. Patients with obesity caused by melanocortin-4 receptor mutations can be treated with a glucagon-like peptide-1 receptor agonist. Cell Metab. 2018;28(1):23–32. https://doi.org/10.1016/j.cmet.2018.05.008.

    Article  CAS  PubMed  Google Scholar 

  43. Knudsen LB, Secher A, Hecksher-Sørensen J, Pyke C. Long-acting glucagon-like peptide-1 receptor agonists have direct access to and effects on pro-opiomelanocortin/cocaine- and amphetamine-stimulated transcript neurons in the mouse hypothalamus. Journal of diabetes investigation. 2016;7 Suppl 1(Suppl Suppl 1):56–63. https://doi.org/10.1111/jdi.12463.

  44. Secher A, Jelsing J, Baquero AF, Hecksher-Sorensen J, Cowley MA, Dalboge LS, et al. The arcuate nucleus mediates GLP-1 receptor agonist liraglutide-dependent weight loss. J Clin Investig. 2014;124(10):4473–88. https://doi.org/10.1172/JCI75276.

    Article  CAS  PubMed  Google Scholar 

  45. Wettergren A, Wojdemann M, Meisner S, Stadil F, Holst JJ. The inhibitory effect of glucagon-like peptide-1 (GLP-1) 7-36 amide on gastric acid secretion in humans depends on an intact vagal innervation. Gut. 1997;40(5):597–601.

  46. Kanoski SE, Fortin SM, Arnold M, Grill HJ, Hayes MR. Peripheral and central GLP-1 receptor populations mediate the anorectic effects of peripherally administered GLP-1 receptor agonists, liraglutide and exendin-4. Endocrinology. 2011;152(8):3103–12. https://doi.org/10.1210/en.2011-0174.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Halawi H, Khemani D, Eckert D, O’Neill J, Kadouh H, Grothe K, et al. Effects of liraglutide on weight, satiation, and gastric functions in obesity: a randomised, placebo-controlled pilot trial. Lancet Gastroenterol Hepatol. 2017;2(12):890–9. https://doi.org/10.1016/s2468-1253(17)30285-6.

    Article  PubMed  Google Scholar 

  48. Jelsing J, Vrang N, Hansen G, Raun K, Tang-Christensen M, Knudsen LB. Liraglutide: short-lived effect on gastric emptying—long lasting effects on body weight. Diabetes Obes Metab. 2012;14(6):531–8. https://doi.org/10.1111/j.1463-1326.2012.01557.x.

    Article  CAS  PubMed  Google Scholar 

  49. Iepsen EW, Lundgren J, Dirksen C, Jensen JE, Pedersen O, Hansen T et al. Treatment with a GLP-1 receptor agonist diminishes the decrease in free plasma leptin during maintenance of weight loss. Int J Obes (2005). 2015;39(5):834–41. https://doi.org/10.1038/ijo.2014.177.

  50. Bradley DP, Kulstad R, Racine N, Shenker Y, Meredith M, Schoeller DA. Alterations in energy balance following exenatide administration. Applied physiology, nutrition, and metabolism = Physiologie appliquee, nutrition et metabolisme. 2012;37(5):893–9. https://doi.org/10.1139/h2012-068.

  51. van Can J, Sloth B, Jensen CB, Flint A, Blaak EE, Saris WH. Effects of the once-daily GLP-1 analog liraglutide on gastric emptying, glycemic parameters, appetite and energy metabolism in obese, non-diabetic adults. Int J Obes (2005). 2014;38(6):784–93. https://doi.org/10.1038/ijo.2013.162.

  52. Pannacciulli N, Bunt JC, Koska J, Bogardus C, Krakoff J. Higher fasting plasma concentrations of glucagon-like peptide 1 are associated with higher resting energy expenditure and fat oxidation rates in humans. Am J Clin Nutr. 2006;84(3):556–60. https://doi.org/10.1093/ajcn/84.3.556.

    Article  CAS  PubMed  Google Scholar 

  53. Harder H, Nielsen L, Tu DT, Astrup A. The effect of liraglutide, a long-acting glucagon-like peptide 1 derivative, on glycemic control, body composition, and 24-h energy expenditure in patients with type 2 diabetes. Diabetes Care. 2004;27(8):1915–21.

    Article  CAS  Google Scholar 

  54. Shalev A, Holst JJ, Keller U. Effects of glucagon-like peptide 1 (7-36 amide) on whole-body protein metabolism in healthy man. Eur J Clin Invest. 1997;27(1):10–6.

    Article  CAS  Google Scholar 

  55. Williams DL, Baskin DG, Schwartz MW. Evidence that intestinal glucagon-like peptide-1 plays a physiological role in satiety. Endocrinology. 2009;150(4):1680–7. https://doi.org/10.1210/en.2008-1045.

    Article  CAS  PubMed  Google Scholar 

  56. Tran KL, Park YI, Pandya S, Muliyil NJ, Jensen BD, Huynh K, et al. Overview of glucagon-like peptide-1 receptor agonists for the treatment of patients with type 2 diabetes. Am Health Drug Benefits. 2017;10(4):178–88.

    PubMed  PubMed Central  Google Scholar 

  57. European Medicines Agency. Ozempic. https://www.ema.europa.eu/en/medicines/human/EPAR/ozempic. Accessed 28 Mar 2019.

  58. US Food and Drug Administration. Full prescribing information saxenda (liraglutide [rDNA origin] injection), solution for subcutaneous use. https://www.accessdata.fda.gov/drugsatfda_docs/label/2014/206321Orig1s000lbl.pdf. Accessed 28 Mar 2019.

  59. O’Neil PM, Birkenfeld AL, McGowan B, Mosenzon O, Pedersen SD, Wharton S, et al. Efficacy and safety of semaglutide compared with liraglutide and placebo for weight loss in patients with obesity: a randomised, double-blind, placebo and active controlled, dose-ranging, phase 2 trial. Lancet. 2018;392(10148):637–49. https://doi.org/10.1016/s0140-6736(18)31773-2.

    Article  CAS  PubMed  Google Scholar 

  60. Guo JJ, Pandey S, Doyle J, Bian B, Lis Y, Raisch DW. A review of quantitative risk-benefit methodologies for assessing drug safety and efficacy-report of the ISPOR risk-benefit management working group. Value Health. 2010;13(5):657–66. https://doi.org/10.1111/j.1524-4733.2010.00725.x.

    Article  PubMed  Google Scholar 

  61. European Medicines Agency. Assessment report for GLP-1 based therapies. 2013. https://www.ema.europa.eu/documents/report/assessment-report-article-53-procedure-glp-1-based-therapies_en.pdf. Accessed 28 Mar 2019. 

  62. Filippatos TD, Panagiotopoulou TV, Elisaf MS. Adverse Effects of GLP-1 Receptor Agonists. Rev Diabetic Stud. 2014;11(3–4):202–30. https://doi.org/10.1900/RDS.2014.11.202.

    Article  PubMed  Google Scholar 

  63. 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(9701):1606–16. https://doi.org/10.1016/s0140-6736(09)61375-1.

    Article  CAS  PubMed  Google Scholar 

  64. 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 (2005). 2012;36(6):843–54. https://doi.org/10.1038/ijo.2011.158.

  65. Wadden TA, Hollander P, Klein S, Niswender K, Woo V, Hale PM et al. Weight maintenance and additional weight loss with liraglutide after low-calorie-diet-induced weight loss: the SCALE Maintenance randomized study. Int J Obes (2005). 2013;37(11):1443–51. https://doi.org/10.1038/ijo.2013.120.

  66. Davies MJ, Bergenstal R, Bode B, Kushner RF, Lewin A, Skjoth TV, et al. Efficacy of liraglutide for weight loss among patients with type 2 diabetes: the SCALE diabetes randomized clinical trial. JAMA. 2015;314(7):687–99. https://doi.org/10.1001/jama.2015.9676.

    Article  CAS  PubMed  Google Scholar 

  67. Pi-Sunyer X, Astrup A, Fujioka K, Greenway F, Halpern A, Krempf M et al. A randomized, controlled trial of 3.0 mg of liraglutide in weight management. N Engl J Med. 2015;373(1):11–22. https://doi.org/10.1056/nejmoa1411892.

  68. Blackman A, Foster GD, Zammit G, Rosenberg R, Aronne L, Wadden T et al. Effect of liraglutide 3.0 mg in individuals with obesity and moderate or severe obstructive sleep apnea: the SCALE Sleep Apnea randomized clinical trial. Int J Obes (2005). 2016;40(8):1310–9. https://doi.org/10.1038/ijo.2016.52.

  69. le Roux CW, Astrup A, Fujioka K, Greenway F, Lau DCW, Van Gaal L, et al. 3 years of liraglutide versus placebo for type 2 diabetes risk reduction and weight management in individuals with prediabetes: a randomised, double-blind trial. Lancet. 2017;389(10077):1399–409. https://doi.org/10.1016/s0140-6736(17)30069-7.

    Article  PubMed  Google Scholar 

  70. European Medicines Agency. Summary of produce characteristics. https://www.ema.europa.eu/documents/product-information/saxenda-epar-product-information_en.pdf. Accessed 28 Mar 2019. 

  71. Rigato M, Fadini GP. Comparative effectiveness of liraglutide in the treatment of type 2 diabetes. Diab Metabol Syndr Obesi. 2014;7:107–20. https://doi.org/10.2147/DMSO.S37644.

    Article  CAS  Google Scholar 

  72. Marso SP, Daniels GH, Brown-Frandsen K, Kristensen P, Mann JFE, Nauck MA et al. Liraglutide and cardiovascular outcomes in type 2 diabetes. N Engl J Med. 2016;375(4):311–22. https://doi.org/10.1056/nejmoa1603827.

  73. Rompianesi G, Hann A, Komolafe O, Pereira SP, Davidson BR, Gurusamy KS. Serum amylase and lipase and urinary trypsinogen and amylase for diagnosis of acute pancreatitis. Cochrane Database Syst Rev. 2017. https://doi.org/10.1002/14651858.cd012010.pub2.

  74. Egan AG, Blind E, Dunder K, de Graeff PA, Hummer BT, Bourcier T et al. Pancreatic safety of incretin-based drugs—FDA and EMA assessment. N Engl J Med. 2014;370(9):794–7. https://doi.org/10.1056/nejmp1314078.

  75. Steinberg WM, Rosenstock J, Wadden TA, Donsmark M, Jensen CB, DeVries JH. Impact of liraglutide on amylase, lipase, and acute pancreatitis in participants with overweight/obesity and normoglycemia, prediabetes, or type 2 diabetes: secondary analyses of pooled data from the SCALE Clinical Development Program. Diabetes Care. 2017;40(7):839–48. https://doi.org/10.2337/dc16-2684.

    Article  CAS  PubMed  Google Scholar 

  76. Portincasa P, Moschetta A, Palasciano G. Cholesterol gallstone disease. Lancet. 2006;368(9531):230–9. https://doi.org/10.1016/s0140-6736(06)69044-2.

    Article  CAS  PubMed  Google Scholar 

  77. Nexøe-Larsen CC, Sørensen PH, Hausner H, Agersnap M, Baekdal M, Brønden A, et al. Effects of liraglutide on gallbladder emptying: a randomized, placebo-controlled trial in adults with overweight or obesity. Diabetes Obes Metab. 2018;20(11):2557–64. https://doi.org/10.1111/dom.13420.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Smits MM, Tonneijck L, Muskiet MHA, Hoekstra T, Kramer MHH, Diamant M, et al. Biliary effects of liraglutide and sitagliptin, a 12-week randomized placebo-controlled trial in type 2 diabetes patients. Diabetes Obes Metab. 2016;18(12):1217–25. https://doi.org/10.1111/dom.12748.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Madsen LW, Knauf JA, Gotfredsen C, Pilling A, Sjogren I, Andersen S, et al. GLP-1 receptor agonists and the thyroid: C-cell effects in mice are mediated via the GLP-1 receptor and not associated with RET activation. Endocrinology. 2012;153(3):1538–47. https://doi.org/10.1210/en.2011-1864.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Novo Nordisk. CHMP endorses EU label update of Saxenda® based on the LEADER trial. 2017. https://www.novonordisk.com/bin/getPDF.2115243.pdf. Accessed 28 Mar 2019. 

  81. Lau J, Bloch P, Schaffer L, Pettersson I, Spetzler J, Kofoed J, et al. Discovery of the once-weekly glucagon-like peptide-1 (GLP-1) analogue semaglutide. J Med Chem. 2015;58(18):7370–80. https://doi.org/10.1021/acs.jmedchem.5b00726.

    Article  CAS  PubMed  Google Scholar 

  82. US Food and Drug Administration. Center for drug evaluation and research 209637Orig1s000. https://www.accessdata.fda.gov/drugsatfda_docs/nda/2017/209637Orig1s000lbl.pdf. Accessed 28 Mar 2019. 

  83. Nauck MA, Petrie JR, Sesti G, Mannucci E, Courreges JP, Lindegaard ML, et al. A phase 2, randomized, dose-finding study of the novel once-weekly human GLP-1 analog, semaglutide, compared with placebo and open-label liraglutide in patients with type 2 diabetes. Diabetes Care. 2016;39(2):231–41. https://doi.org/10.2337/dc15-0165.

    Article  CAS  PubMed  Google Scholar 

  84. Novo Nordisk. Strengthen leadership in obesity. 2017. https://www.novonordisk.com/content/dam/Denmark/HQ/investors/irmaterial/cmd/2017/05_Strengthen%20leadership%20in%20obesity.pdf. Accessed 28 Mar 2019.

  85. ClinicalTrials.gov. Semaglutide Effects on Heart Disease and Stroke in Patients With Overweight or Obesity (SELECT). NCT03574597. 2018.https://clinicaltrials.gov/ct2/show/NCT03574597. Accessed 28 Mar 2019. 

  86. Sorli C, Harashima SI, Tsoukas GM, Unger J, Karsbol JD, Hansen T, et al. Efficacy and safety of once-weekly semaglutide monotherapy versus placebo in patients with type 2 diabetes (SUSTAIN 1): a double-blind, randomised, placebo-controlled, parallel-group, multinational, multicentre phase 3a trial. Lancet Diabetes Endocrinol. 2017;5(4):251–60. https://doi.org/10.1016/s2213-8587(17)30013-x.

    Article  CAS  PubMed  Google Scholar 

  87. Marso SP, Bain SC, Consoli A, Eliaschewitz FG, Jodar E, Leiter LA, et al. Semaglutide and cardiovascular outcomes in patients with type 2 diabetes. N Engl J Med. 2016;375(19):1834–44. https://doi.org/10.1056/NEJMoa1607141.

    Article  CAS  PubMed  Google Scholar 

  88. Davies M, Pieber TR, Hartoft-Nielsen ML, Hansen OKH, Jabbour S, Rosenstock J. Effect of oral semaglutide compared with placebo and subcutaneous semaglutide on glycemic control in patients with type 2 diabetes: a randomized clinical trial. JAMA. 2017;318(15):1460–70. https://doi.org/10.1001/jama.2017.14752.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Dicembrini I, Nreu B, Scatena A, Andreozzi F, Sesti G, Mannucci E, et al. Microvascular effects of glucagon-like peptide-1 receptor agonists in type 2 diabetes: a meta-analysis of randomized controlled trials. Acta Diabetol. 2017;54(10):933–41. https://doi.org/10.1007/s00592-017-1031-9.

    Article  CAS  PubMed  Google Scholar 

  90. Hernandez C, Bogdanov P, Corraliza L, Garcia-Ramirez M, Sola-Adell C, Arranz JA, et al. Topical administration of GLP-1 receptor agonists prevents retinal neurodegeneration in experimental diabetes. Diabetes. 2016;65(1):172–87. https://doi.org/10.2337/db15-0443.

    Article  CAS  PubMed  Google Scholar 

  91. Mensberg P, Nyby S, Jorgensen PG, Storgaard H, Jensen MT, Sivertsen J, et al. Near-normalization of glycaemic control with glucagon-like peptide-1 receptor agonist treatment combined with exercise in patients with type 2 diabetes. Diabetes Obes Metab. 2017;19(2):172–80. https://doi.org/10.1111/dom.12797.

    Article  CAS  PubMed  Google Scholar 

  92. Nissen SE, Wolski KE, Prcela L, Wadden T, Buse JB, Bakris G, et al. Effect of Naltrexone-Bupropion on major adverse cardiovascular events in overweight and obese patients with cardiovascular risk factors: a randomized clinical trial. JAMA. 2016;315(10):990–1004. https://doi.org/10.1001/jama.2016.1558.

    Article  CAS  PubMed  Google Scholar 

  93. Bohula EA, Wiviott SD, McGuire DK, Inzucchi SE, Kuder J, Im K, et al. Cardiovascular safety of Lorcaserin in overweight or obese patients. N Engl J Med. 2018;379(12):1107–17. https://doi.org/10.1056/NEJMoa1808721.

    Article  CAS  PubMed  Google Scholar 

  94. Bethel MA, Patel RA, Merrill P, Lokhnygina Y, Buse JB, Mentz RJ, et al. Cardiovascular outcomes with glucagon-like peptide-1 receptor agonists in patients with type 2 diabetes: a meta-analysis. Lancet Diabetes Endocrinol. 2018;6(2):105–13. https://doi.org/10.1016/s2213-8587(17)30412-6.

    Article  PubMed  Google Scholar 

  95. Kaneko M, Narukawa M. Assessment of cardiovascular risk with glucagon-like peptide 1 receptor agonists in patients with type 2 diabetes using an alternative measure to the hazard ratio. Ann Pharmacother. 2018;52(7):632–8. https://doi.org/10.1177/1060028018757407.

    Article  CAS  PubMed  Google Scholar 

  96. The Look AHEAD Research Group. Cardiovascular effects of intensive lifestyle intervention in type 2 diabetes. N Engl J Med. 2013;369(2):145–54. https://doi.org/10.1056/NEJMoa1212914.

    Article  CAS  PubMed Central  Google Scholar 

  97. Finkelstein EA, Verghese NR. Incremental cost-effectiveness of evidence-based non-surgical weight loss strategies. Clin Obes. 2019;0(0):e12294. https://doi.org/10.1111/cob.12294.

  98. ClinicalTrials.gov. Saxenda in obesity services (STRIVE Study) (STRIVE). NCT03036800. 2017. https://clinicaltrials.gov/ct2/show/NCT03036800. Accessed 28 Mar 2019.

  99. Basalay MV, Mastitskaya S, Mrochek A, Ackland GL, Del Arroyo AG, Sanchez J, et al. Glucagon-like peptide-1 (GLP-1) mediates cardioprotection by remote ischaemic conditioning. Cardiovasc Res. 2016;112(3):669–76. https://doi.org/10.1093/cvr/cvw216.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Salcedo I, Tweedie D, Li Y, Greig NH. Neuroprotective and neurotrophic actions of glucagon-like peptide-1: an emerging opportunity to treat neurodegenerative and cerebrovascular disorders. Br J Pharmacol. 2012;166(5):1586–99. https://doi.org/10.1111/j.1476-5381.2012.01971.x.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Vinue A, Navarro J, Herrero-Cervera A, Garcia-Cubas M, Andres-Blasco I, Martinez-Hervas S, et al. The GLP-1 analogue lixisenatide decreases atherosclerosis in insulin-resistant mice by modulating macrophage phenotype. Diabetologia. 2017;60(9):1801–12. https://doi.org/10.1007/s00125-017-4330-3.

    Article  CAS  PubMed  Google Scholar 

  102. Holst JJ, Madsbad S, Bojsen-Møller KN, Svane MS, Jørgensen NB, Dirksen C, et al. Mechanisms in bariatric surgery: gut hormones, diabetes resolution, and weight loss. Surg Obes Relat Dis. 2018;14(5):708–14. https://doi.org/10.1016/j.soard.2018.03.003.

    Article  PubMed  PubMed Central  Google Scholar 

  103. Meek CL, Lewis HB, Reimann F, Gribble FM, Park AJ. The effect of bariatric surgery on gastrointestinal and pancreatic peptide hormones. Peptides. 2016;77:28–37. https://doi.org/10.1016/j.peptides.2015.08.013.

    Article  CAS  PubMed  Google Scholar 

  104. Sadry SA, Drucker DJ. Emerging combinatorial hormone therapies for the treatment of obesity and T2DM. Nat Rev Endocrinol. 2013;9(7):425–33. https://doi.org/10.1038/nrendo.2013.47.

    Article  CAS  PubMed  Google Scholar 

  105. Alexiadou K, Anyiam O, Tan T. Cracking the combination: gut hormones for the treatment of obesity and diabetes. J Neuroendocrinol. 2018:e12664. https://doi.org/10.1111/jne.12664.

Download references

Author information

Authors and Affiliations

Authors

Contributions

Conceptualisation and methodology: SST. Writing first draft: RMC. Critically editing, re-writing, analysing and reviewing the manuscript: SST, RMC, and CRJ.

Corresponding author

Correspondence to Signe S. Torekov.

Ethics declarations

Funding

This work was supported by the Tripartite Immunometabolism Consortium [TrIC], Novo Nordisk Foundation; grant number NNF15CC0018486.

Conflict of interest

Rasmus Michael Christensen was supported by the Novo Nordisk Foundation, grant number NNF15CC0018486. Christian Rimer Juhl has no conflicts of interest that are directly relevant to the content of this article. Signe Sørensen Torekov has received research grants from the Novo Nordisk Foundation and Novo Nordisk A/S.

Additional information

The original version of this article was revised: The drug “Orlistat” was incorrectly changed to “Gastrointestinal lipase inhibitor” in Table 1 and the same has been corrected.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Christensen, R.M., Juhl, C.R. & Torekov, S.S. Benefit-Risk Assessment of Obesity Drugs: Focus on Glucagon-like Peptide-1 Receptor Agonists. Drug Saf 42, 957–971 (2019). https://doi.org/10.1007/s40264-019-00812-7

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s40264-019-00812-7

Navigation