Metformin for early comorbid glucose dysregulation and schizophrenia spectrum disorders: a pilot double-blind randomized clinical trial

Sri Mahavir Agarwal, Roshni Panda, Kenya A Costa-Dookhan, Nicole E MacKenzie, Quinn Casuccio Treen, Fernando Caravaggio, Eyesha Hashim, General Leung, Anish Kirpalani, Kelly Matheson, Araba F Chintoh, Caroline K Kramer, Aristotle N Voineskos, Ariel Graff-Guerrero, Gary J Remington, Margaret K Hahn, Sri Mahavir Agarwal, Roshni Panda, Kenya A Costa-Dookhan, Nicole E MacKenzie, Quinn Casuccio Treen, Fernando Caravaggio, Eyesha Hashim, General Leung, Anish Kirpalani, Kelly Matheson, Araba F Chintoh, Caroline K Kramer, Aristotle N Voineskos, Ariel Graff-Guerrero, Gary J Remington, Margaret K Hahn

Abstract

Patients with schizophrenia have exceedingly high rates of metabolic comorbidity including type 2 diabetes and lose 15-20 years of life due to cardiovascular diseases, with early accrual of cardiometabolic disease. In this study, thirty overweight or obese (Body Mass Index (BMI) > 25) participants under 40 years old with schizophrenia spectrum disorders and early comorbid prediabetes or type 2 diabetes receiving antipsychotic medications were randomized, in a double-blind fashion, to metformin 1500 mg/day or placebo (2:1 ratio; n = 21 metformin and n = 9 placebo) for 4 months. The primary outcome measures were improvements in glucose homeostasis (HbA1c, fasting glucose) and insulin resistance (Matsuda index-derived from oral glucose tolerance tests and homeostatic model of insulin resistance (HOMA-IR)). Secondary outcome measures included changes in weight, MRI measures of fat mass and distribution, symptom severity, cognition, and hippocampal volume. Twenty-two patients (n = 14 metformin; n = 8 placebo) completed the trial. The metformin group had a significant decrease over time in the HOMA-IR (p = 0.043) and fasting blood glucose (p = 0.007) vs. placebo. There were no differences between treatment groups in the Matsuda index, HbA1c, which could suggest liver-specific effects of metformin. There were no between group differences in other secondary outcome measures, while weight loss in the metformin arm correlated significantly with decreases in subcutaneous, but not visceral or hepatic adipose tissue. Our results show that metformin improved dysglycemia and insulin sensitivity, independent of weight loss, in a young population with prediabetes/diabetes and psychosis spectrum illness, that is at extremely high risk of early cardiovascular mortality. Trial Registration: This protocol was registered with clinicaltrials.gov (NCT02167620).

Conflict of interest statement

The authors declare no competing interests.

Figures

Fig. 1. Flowchart of study participants.
Fig. 1. Flowchart of study participants.
All participants who were randomized and had received at least 1 dose of metformin or placebo were included in the analyses.
Fig. 2
Fig. 2
Change in insulin sensitivity (HOMA-IR) (A) and fasting glucose levels (B) with metformin compared to placebo at baseline, 8 weeks, and 16 weeks of treatment. Error bars represent ± 1 S.E.
Fig. 3
Fig. 3
Correlation between percentage change in weight and subcutaneous adipose tissue (SAT) volume with metformin treatment.

References

    1. Dixon L, et al. Prevalence and correlates of diabetes in national schizophrenia samples. Schizophr. Bull. 2000;26:903–912. doi: 10.1093/oxfordjournals.schbul.a033504.
    1. Rajkumar AP, et al. Endogenous and antipsychotic-related risks for diabetes mellitus in young people with schizophrenia: a Danish population-based cohort study. Am. J. Psychiatry. 2017;174:686–694. doi: 10.1176/appi.ajp.2016.16040442.
    1. Correll CU, et al. Prevalence, incidence and mortality from cardiovascular disease in patients with pooled and specific severe mental illness: a large-scale meta-analysis of 3,211,768 patients and 113,383,368 controls. World Psychiatry. 2017;16:163–180. doi: 10.1002/wps.20420.
    1. Howes OD, et al. A prospective study of impairment in glucose control caused by clozapine without changes in insulin resistance. Am. J. Psychiatry. 2004;161:361–363. doi: 10.1176/appi.ajp.161.2.361.
    1. Oriot P, et al. Insulin sensitivity, adjusted beta-cell function and adiponectinaemia among lean drug-naive schizophrenic patients treated with atypical antipsychotic drugs: a nine-month prospective study. Diabetes Metab. 2008;34:490–496. doi: 10.1016/j.diabet.2008.03.003.
    1. Correll CU, et al. Cardiometabolic risk of second-generation antipsychotic medications during first-time use in children and adolescents. JAMA. 2009;302:1765–1773. doi: 10.1001/jama.2009.1549.
    1. Hahn MK, et al. Acute effects of single-dose olanzapine on metabolic, endocrine, and inflammatory markers in healthy controls. J. Clin. Psychopharmacol. 2013;33:740–746. doi: 10.1097/JCP.0b013e31829e8333.
    1. Kowalchuk C, et al. Antipsychotics and glucose metabolism: how brain and body collide. Am. J. Physiol. Endocrinol. Metab. 2019;316:E1–E15. doi: 10.1152/ajpendo.00164.2018.
    1. Bora E, Akdede BB, Alptekin K. The relationship between cognitive impairment in schizophrenia and metabolic syndrome: a systematic review and meta-analysis. Psychological Med. 2017;47:1030–1040. doi: 10.1017/S0033291716003366.
    1. Cooper SJ, et al. BAP guidelines on the management of weight gain, metabolic disturbances and cardiovascular risk associated with psychosis and antipsychotic drug treatment. J. Psychopharmacol. 2016;30:717–748. doi: 10.1177/0269881116645254.
    1. Faulkner G, Cohn T, Remington G, Irving H. Body mass index, waist circumference and quality of life in individuals with schizophrenia. Schizophrenia Res. 2007;90:174–178. doi: 10.1016/j.schres.2006.10.009.
    1. Nasrallah HA, et al. Low rates of treatment for hypertension, dyslipidemia and diabetes in schizophrenia: data from the CATIE schizophrenia trial sample at baseline. Schizophr. Res. 2006;86:15–22. doi: 10.1016/j.schres.2006.06.026.
    1. Kohen D. Diabetes mellitus and schizophrenia: historical perspective. Br. J. Psychiatry Suppl. 2004;47:S64–S66. doi: 10.1192/bjp.184.47.s64.
    1. Chintoh AF, et al. Insulin resistance and secretion in vivo: effects of different antipsychotics in an animal model. Schizophr. Res. 2009;108:127–133. doi: 10.1016/j.schres.2008.12.012.
    1. Houseknecht KL, et al. Acute effects of atypical antipsychotics on whole-body insulin resistance in rats: implications for adverse metabolic effects. Neuropsychopharmacology. 2007;32:289–297. doi: 10.1038/sj.npp.1301209.
    1. Henderson DC, et al. A double-blind, placebo-controlled trial of rosiglitazone for clozapine-induced glucose metabolism impairment in patients with schizophrenia. Acta Psychiatr. Scand. 2009;119:457–465. doi: 10.1111/j.1600-0447.2008.01325.x.
    1. Larsen JR, et al. Effect of liraglutide treatment on prediabetes and overweight or obesity in clozapine- or olanzapine-treated patients with schizophrenia spectrum disorder: a randomized clinical trial. JAMA Psychiatry. 2017;74:719–728. doi: 10.1001/jamapsychiatry.2017.1220.
    1. Remington GJ, et al. Metformin attenuates olanzapine-induced hepatic, but not peripheral insulin resistance. J. Endocrinol. 2015;227:71–81. doi: 10.1530/JOE-15-0074.
    1. Boyda HN, et al. Differential effects of 3 classes of antidiabetic drugs on olanzapine-induced glucose dysregulation and insulin resistance in female rats. J. Psychiatry Neurosci. 2012;37:407–415. doi: 10.1503/jpn.110140.
    1. Flory J, Lipska K. Metformin in 2019. JAMA. 2019;321:1926–1927. doi: 10.1001/jama.2019.3805.
    1. de Silva VA, et al. Metformin in prevention and treatment of antipsychotic induced weight gain: a systematic review and meta-analysis. BMC Psychiatry. 2016;16:341. doi: 10.1186/s12888-016-1049-5.
    1. Wharton S, et al. Obesity in adults: a clinical practice guideline. Can. Med. Assoc. J. 2020;192:E875. doi: 10.1503/cmaj.191707.
    1. Matsuda M, DeFronzo RA. Insulin sensitivity indices obtained from oral glucose tolerance testing: comparison with the euglycemic insulin clamp. Diabetes Care. 1999;22:1462–1470. doi: 10.2337/diacare.22.9.1462.
    1. Retnakaran R, Qi Y, Goran MI, Hamilton JK. Evaluation of proposed oral disposition index measures in relation to the actual disposition index. Diabet. Med. 2009;26:1198–1203. doi: 10.1111/j.1464-5491.2009.02841.x.
    1. Milne NT, et al. Hippocampal atrophy, asymmetry, and cognition in type 2 diabetes mellitus. Brain Behav. 2018;8:e00741. doi: 10.1002/brb3.741.
    1. Goldenberg R, Punthakee Z. Definition, classification and diagnosis of diabetes, prediabetes and metabolic syndrome. Can. J. Diabetes. 2013;37:S8–S11. doi: 10.1016/j.jcjd.2013.01.011.
    1. American Diabetes, A. 2. Classification and diagnosis of diabetes: standards of medical care in diabetes-2018. Diabetes Care. 2018;41:S13–S27. doi: 10.2337/dc18-S002.
    1. Jordan S, Knight J, Pointon D. Monitoring adverse drug reactions: scales, profiles, and checklists. Int Nurs. Rev. 2004;51:208–221. doi: 10.1111/j.1466-7657.2004.00251.x.
    1. Overall JE, Gorham DR. The brief psychiatric rating scale. Psychological Rep. 1962;10:799–812. doi: 10.2466/pr0.1962.10.3.799.
    1. Addington, D., Addington, J. & Maticka-Tyndale, E. Assessing depression in schizophrenia: the Calgary Depression Scale. Br. J. Psychiatry Suppl.22, 39–44 (1993).
    1. Wallace TM, Levy JC, Matthews DR. Use and abuse of HOMA modeling. Diabetes Care. 2004;27:1487–1495. doi: 10.2337/diacare.27.6.1487.
    1. Carnevale Schianca GP, et al. Comparison between HOMA-IR and ISI-gly in detecting subjects with the metabolic syndrome. Diabetes Metab. Res. Rev. 2006;22:111–117. doi: 10.1002/dmrr.560.
    1. Joshi AA, Hu HH, Leahy RM, Goran MI, Nayak KS. Automatic intra-subject registration-based segmentation of abdominal fat from water-fat MRI. J. Magn. Reson. Imaging. 2013;37:423–430. doi: 10.1002/jmri.23813.
    1. Pipitone, J. et al. Multi-atlas segmentation of the whole hippocampus and subfields using multiple automatically generated templates. Neuroimage10.1016/j.neuroimage.2014.04.054 (2014).
    1. Fedorov A, et al. 3D Slicer as an image computing platform for the Quantitative Imaging Network. Magn. Reson. Imaging. 2012;30:1323–1341. doi: 10.1016/j.mri.2012.05.001.
    1. Agarwal SM, et al. S245. A systematic review and meta-analysis of pharmacological interventions for reduction or prevention of weight gain in schizophrenia. Biol. Psychiatry. 2018;83:S443. doi: 10.1016/j.biopsych.2018.02.1137.
    1. Foretz M, Guigas B, Bertrand L, Pollak M, Viollet B. Metformin: from mechanisms of action to therapies. Cell Metab. 2014;20:953–966. doi: 10.1016/j.cmet.2014.09.018.
    1. Duca FA, et al. Metformin activates a duodenal Ampk-dependent pathway to lower hepatic glucose production in rats. Nat. Med. 2015;21:506–511. doi: 10.1038/nm.3787.
    1. Lv WS, et al. The effect of metformin on food intake and its potential role in hypothalamic regulation in obese diabetic rats. Brain Res. 2012;1444:11–19. doi: 10.1016/j.brainres.2012.01.028.
    1. Derkach K, et al. The evidence of metabolic-improving effect of metformin in Ay/a mice with genetically-induced melanocortin obesity and the contribution of hypothalamic mechanisms to this effect. PloS ONE. 2019;14:e0213779. doi: 10.1371/journal.pone.0213779.
    1. DeFronzo RA, Stonehouse AH, Han J, Wintle ME. Relationship of baseline HbA1c and efficacy of current glucose-lowering therapies: a meta-analysis of randomized clinical trials. Diabet. Med. 2010;27:309–317. doi: 10.1111/j.1464-5491.2010.02941.x.
    1. De Lorenzo A, et al. Adiposity rather than BMI determines metabolic risk. Int J. Cardiol. 2013;166:111–117. doi: 10.1016/j.ijcard.2011.10.006.

Source: PubMed

3
購読する