Effect of GLP-1 receptor agonist treatment on body weight in obese antipsychotic-treated patients with schizophrenia: a randomized, placebo-controlled trial

Pelle L Ishøy, Filip K Knop, Brian V Broberg, Nikolaj Bak, Ulrik B Andersen, Niklas R Jørgensen, Jens J Holst, Birte Y Glenthøj, Bjørn H Ebdrup, Pelle L Ishøy, Filip K Knop, Brian V Broberg, Nikolaj Bak, Ulrik B Andersen, Niklas R Jørgensen, Jens J Holst, Birte Y Glenthøj, Bjørn H Ebdrup

Abstract

Aims: Schizophrenia is associated with cardiovascular co-morbidity and a reduced life-expectancy of up to 20 years. Antipsychotics are dopamine D2 receptor antagonists and are the standard of medical care in schizophrenia, but the drugs are associated with severe metabolic side effects such as obesity and diabetes. Glucagon-like peptide-1 receptor agonists (GLP-1RAs) are registered for treatment of both obesity and type 2 diabetes. We investigated metabolic effects of the GLP-1RA, exenatide once-weekly, in non-diabetic, antipsychotic-treated, obese patients with schizophrenia.

Material and methods: Antipsychotic-treated, obese, non-diabetic, schizophrenia spectrum patients were randomized to double-blinded adjunctive treatment with once-weekly subcutaneous exenatide (n = 23) or placebo (n = 22) injections for 3 months. The primary outcome was loss of body weight after treatment and repeated measures analysis of variance was used as statistical analysis.

Results: Between March 2013 and June 2015, 40 patients completed the trial. At baseline, mean body weight was 118.3 ± 16.0 kg in the exenatide group and 111.7 ± 18.0 kg in the placebo group, with no group differences ( P = .23). The exenatide and placebo groups experienced significant ( P = .004), however similar ( P = .98), weight losses of 2.24 ± 3.3 and 2.23 ± 4.4 kg, respectively, after 3 months of treatment.

Conclusions: Treatment with exenatide once-weekly did not promote weight loss in obese, antipsychotic-treated patients with schizophrenia compared to placebo. Our results could suggest that the body weight-lowering effect of GLP-1RAs involves dopaminergic signaling, but blockade of other receptor systems may also play a role. Nevertheless, anti-obesity regimens effective in the general population may not be readily implemented in antipsychotic-treated patients with schizophrenia.

Trial registration: ClinicalTrials.gov NCT01794429.

Keywords: GLP-1 analogue; antidiabetic drug; exenatide; randomized trial schizophrenia.

© 2016 The Authors. Diabetes, Obesity and Metabolism published by John Wiley & Sons Ltd.

Figures

Figure 1
Figure 1
Study diagram of the patient flow according to CONSORT 2010 statement. Of the 65 patients originally referred to the study, 45 entered the subsequent 3‐month treatment period. Five patients withdrew from trial, resulting in a completion of n = 40 (n = 20 exenatide only weekly, n = 20 placebo).
Figure 2
Figure 2
Time lines of mean body mass index (BMI) over a 3‐month period after glucagon‐like peptide‐1 receptor agonist (GLP‐1RA) treatment in obese patients with schizophrenia. Blue line: exenatide group; red line: placebo group. Repeated measures analysis of variance showed no treatment effect of exenatide treatment, Time × Group interaction (P = .98). The x‐axis shows time points for body weight measurements. Horizontal lines on these time points are BMI standard deviations within the groups.

References

    1. McGrath J, Saha S, Chant D, Welham J. Schizophrenia: a concise overview of incidence, prevalence, and mortality. Epidemiol Rev. 2008;30(1):67‐76.
    1. Dieset I, Andreassen OA, Haukvik UK. Somatic comorbidity in schizophrenia: some possible biological mechanisms across the life span. Schizophr Bull. 2016;42(6):1316‐1319.
    1. Nordentoft M, Wahlbeck K, Hällgren J, et al. Excess mortality, causes of death and life expectancy in 270,770 patients with recent onset of mental disorders in Denmark, Finland and Sweden. PLoS One. 2013;8(1):e55176.
    1. Saha S, Chant D, Mcgrath J. A systematic review of mortality in schizophrenia. Arch Gen Psychiatry. 2007;64(10):1123‐1131.
    1. Baskin ML, Ard J, Franklin F, Allison DB. Prevalence of obesity in the United States. Obes Rev. 2005;6(1):5‐7.
    1. Allison DB, Newcomer JW, Dunn AL, et al. Obesity among those with mental disorders: a National Institute of Mental Health meeting report. Am J Prev Med. 2009;36(4):341‐350.
    1. Megna JL, Schwartz TL, Siddiqui U, Herrera Rojas M. Obesity in adults with serious and persistent mental illness: a review of postulated mechanisms and current interventions. Ann Clin Psychiatry. 2011;23(2):131‐140.
    1. Howes OD, Kapur S. The dopamine hypothesis of schizophrenia: version III ‐ the final common pathway. Schizophr Bull. 2009;35(3):549‐562.
    1. Wise RA, Rompre PP. Brain dopamine and reward. Annu Rev Psychol. 1989;40:191‐225.
    1. Stice E, Spoor S, Ng J, Zald DH. Relation of obesity to consummatory and anticipatory food reward. Physiol Behav. 2009;97(5):551‐560.
    1. Caravaggio F, Hahn M, Nakajima S, Gerretsen P, Remington G, Graff‐Guerrero A. Reduced insulin‐receptor mediated modulation of striatal dopamine release by basal insulin as a possible contributing factor to hyperdopaminergia in schizophrenia. Med Hypotheses. 2015;85(4):391‐396.
    1. Nielsen MØ, Nielsen MØ, Rostrup E, Wulff S, Glenthøj B, Ebdrup BH. Striatal reward activity and antipsychotic‐associated weight change in patients with schizophrenia undergoing initial treatment. JAMA Psychiatry. 2016;73(2):1‐8.
    1. Buchanan RW, Kreyenbuhl J, Kelly DL, et al. The 2009 schizophrenia PORT psychopharmacological treatment recommendations and summary statements. Schizophr Bull. 2010;36(1):71‐93.
    1. Mukundan A, Faulkner G, Cohn T, Remington G. Antipsychotic switching for people with schizophrenia who have neuroleptic‐induced weight or metabolic problems. Cochrane Database Syst Rev. 2010;(2):1‐129.
    1. Gabriele JM, Dubbert PM, Reeves RR. Efficacy of behavioural interventions in managing atypical antipsychotic weight gain. Obes Rev. 2009;10(4):442‐455.
    1. Maayan L, Vakhrusheva J, Correll CU. Effectiveness of medications used to attenuate antipsychotic‐related weight gain and metabolic abnormalities: a systematic review and meta‐analysis. Neuropsychopharmacology. 2010;35(7):1520‐1530.
    1. Faulkner G, Cohn T, Remington G. Interventions to reduce weight gain in schizophrenia. Schizophr Bull. 2007;33(3):654‐656.
    1. Jarskog LF, Hamer RM, Catellier DJ, et al. Metformin for weight loss and metabolic control in overweight outpatients with schizophrenia and schizoaffective disorder. Am J Psychiatry. 2013;170(9):1032‐1040.
    1. Holst JJ. The physiology of glucagon‐like peptide 1. Physiol Rev. 2007;87(4):1409‐1439.
    1. Ebdrup BH, Knop FK, Ishøy PL, et al. Glucagon‐like peptide‐1 analogs against antipsychotic‐induced weight gain: potential physiological benefits. BMC Med. 2012;10(1):92‐99.
    1. Ishøy PL, Knop FK, Vilsbøll T, Glenthøj BY, Ebdrup BH. Sustained weight loss after treatment with a glucagon‐like Peptide‐1 receptor agonist in an obese patient with schizophrenia and type 2 diabetes. Am J Psychiatry. 2013;170(6):681‐682.
    1. Ishøy PL, Knop FK, Broberg BV, et al. Treatment of antipsychotic‐associated obesity with a GLP‐1 receptor agonist — protocol for an investigator‐ initiated prospective, randomised, intervention study: the TAO study protocol. BMJ Open. 2014;4:e004158.
    1. Moher D, Hopewell S, Schulz KF, et al. CONSORT 2010 explanation and elaboration: updated guidelines for reporting parallel group randomised trials. BMJ. 2010;340:c869.
    1. Gøtzsche PC. Blinding during data analysis and writing of manuscripts. Control Clin Trials. 1996;17(4):285‐290; discussion 290–293.
    1. Weiss W, Gohlisch C, Harsch‐Gladisch C, Tölle M, Zidek W, van der Giet M. Oscillometric estimation of central blood pressure: validation of the Mobil‐O‐Graph in comparison with the SphygmoCor device. Blood Press Monit. 2012;17(3):128‐131.
    1. Luzardo L, Lujambio I, Sottolano M, et al. 24‐h ambulatory recording of aortic pulse wave velocity and central systolic augmentation: a feasibility study. Hypertens Res. 2012;35(10):980‐987.
    1. Kielgast U, Holst JJ, Madsbad S. Antidiabetic actions of endogenous and exogenous GLP‐1 in type 1 diabetic patients with and without residual β‐cell function. Diabetes. 2011;60(5):1599‐1607.
    1. Wewer Albrechtsen NJ, Hartmann B, Veedfald S, et al. Hyperglucagonaemia analysed by glucagon sandwich ELISA: nonspecific interference or truly elevated evels? Diabetologia. 2014;57(9):1919‐1926.
    1. Vilsbøll T, Christensen M, Junker AE, Knop FK, Gluud LL. Effects of glucagon‐like peptide‐1 receptor agonists on weight loss: systematic review and meta‐analyses of randomised controlled trials. BMJ. 2012;344:d7771.
    1. Buse JB, Drucker DJ, Taylor KL, et al. DURATION‐1: exenatide once weekly produces sustained glycemic control and weight loss over 52 weeks. Diabetes Care. 2010;33(6):1255‐1261.
    1. Buse JB, Nauck M, Forst T, et al. Exenatide once weekly versus liraglutide once daily in patients with type 2 diabetes (DURATION‐6): a randomised, open‐label study. Lancet. 2013;381(9861):117‐124.
    1. Davies MJ, Bergenstal R, Bode B, et al. Efficacy of liraglutide for weight loss among patients with type 2 diabetes. JAMA. 2015;314(7):687.
    1. European Medicines Agency. Annex I: summary of product characteristics for Saxenda;URL: . Accessed April 16, 2015.
    1. Anderberg RH, Anefors C, Bergquist F, Nissbrandt H, Skibicka KP. Dopamine signaling in the amygdala, increased by food ingestion and GLP‐1, regulates feeding behavior. Physiol Behav. 2014;136:135‐144.
    1. Weston‐Green K, Huang X‐F, Lian J, Deng C. Effects of olanzapine on muscarinic M3 receptor binding density in the brain relates to weight gain, plasma insulin and metabolic hormone levels. Eur Neuropsychopharmacol. 2012;22(5):364‐373.
    1. He M, Deng C, Huang X‐F. The role of hypothalamic H1 receptor antagonism in antipsychotic‐induced weight gain. CNS Drugs. 2013;27(6):423‐434.
    1. Rasmussen H, Ebdrup BH, Oranje B, Pinborg LH, Knudsen GM, Glenthøj B. Neocortical serotonin2A receptor binding predicts quetiapine associated weight gain in antipsychotic‐naive first‐episode schizophrenia patients. Int J Neuropsychopharmacol. 2014:1‐8.
    1. Astrup A, Carraro R, Finer N, et al. Safety, tolerability and sustained weight loss over 2 years with the once‐daily human GLP‐1 analog, liraglutide. Int . Obes (Lond). 2012;36(6):843‐854.
    1. Ebdrup BH, Knop FK, Madsen A, et al. Glucometabolic hormones and cardiovascular risk markers in antipsychotic‐treated patients. J Clin Psychiatry. 2014;75(9):e899‐e905.
    1. Díaz A, Galli C, Tringler M, Ramírez A, Cabrera Fischer EI. Reference values of pulse wave velocity in healthy people from an urban and rural argentinean population. Int J Hypertens. 2014;2014:653239:1‐7.
    1. Reference Values for Arterial Stiffness’ Collaboration . Determinants of pulse wave velocity in healthy people and in the presence of cardiovascular risk factors: “establishing normal and reference values”. Eur Heart J. 2016;31(19):2338‐2350.
    1. Fineman M, Flanagan S, Taylor K, et al. Pharmacokinetics and pharmacodynamics of exenatide extended‐release after single and multiple dosing. Clin Pharmacokinet. 2011;50(1):65‐74.
    1. Drucker DJ, Buse JB, Taylor K, et al. Exenatide once weekly versus twice daily for the treatment of type 2 diabetes: a randomised, open‐label, non‐inferiority study. Lancet. 2008;372(9645):1240‐1250.
    1. Herrstedt J. Risk‐benefit of antiemetics in prevention and treatment of chemotherapy‐induced nausea and vomiting. Expert Opin Drug Saf. 2004;3(3):231‐248.
    1. Chiu L, Chow R, Popovic M, et al. Efficacy of olanzapine for the prophylaxis and rescue of chemotherapy‐induced nausea and vomiting: a systematic review and meta‐analysis. Support Care Cancer. 2016;24(5):2381‐2392.

Source: PubMed

3
S'abonner