Once-daily delayed-release metformin lowers plasma glucose and enhances fasting and postprandial GLP-1 and PYY: results from two randomised trials

Ralph A DeFronzo, John B Buse, Terri Kim, Colleen Burns, Sharon Skare, Alain Baron, Mark Fineman, Ralph A DeFronzo, John B Buse, Terri Kim, Colleen Burns, Sharon Skare, Alain Baron, Mark Fineman

Abstract

Aims/hypothesis: Delayed-release metformin (Metformin DR) was developed to maximise gut-based mechanisms of metformin action by targeting the drug to the ileum. Metformin DR was evaluated in two studies. Study 1 compared the bioavailability and effects on circulating glucose and gut hormones (glucagon-like peptide-1, peptide YY) of Metformin DR dosed twice-daily to twice-daily immediate-release metformin (Metformin IR). Study 2 compared the bioavailability and glycaemic effects of Metformin DR dosages of 1,000 mg once-daily in the morning, 1,000 mg once-daily in the evening, and 500 mg twice-daily.

Methods: Study 1 was a blinded, randomised, crossover study (three × 5 day treatment periods) of twice-daily 500 mg or 1,000 mg Metformin DR vs twice-daily 1,000 mg Metformin IR in 24 participants with type 2 diabetes conducted at two study sites (Celerion Inc.; Tempe, AZ, and Lincoln, NE, USA). Plasma glucose and gut hormones were assessed over 10.25 h at the start and end of each treatment period; plasma metformin was measured over 11 h at the end of each treatment period. Study 2 was a non-blinded, randomised, crossover study (three × 7 day treatment periods) of 1,000 mg Metformin DR once-daily in the morning, 1,000 mg Metformin DR once-daily in the evening, or 500 mg Metformin DR twice-daily in 26 participants with type 2 diabetes performed at a single study site (Celerion, Tempe, AZ). Plasma glucose was assessed over 24 h at the start and end of each treatment period, and plasma metformin was measured over 30 h at the end of each treatment period. Both studies implemented centrally generated computer-based randomisation using a 1:1:1 allocation ratio.

Results: A total of 24 randomised participants were included in study 1; of these, 19 completed the study and were included in the evaluable population. In the evaluable population, all treatments produced similar significant reductions in fasting glucose (median reduction range, -0.67 to -0.81 mmol/l across treatments) and postprandial glucose (Day 5 to baseline AUC0-t ratio = 0.9 for all three treatments) and increases in gut hormones (Day 5 to baseline AUC0-t ratio range: 1.6-1.9 for GLP-1 and 1.4-1.5 for PYY) despite an almost 60% reduction in systemic metformin exposure for 500 mg Metformin DR compared with Metformin IR. A total of 26 randomised participants were included in study 2: 24 had at least one dose of study medication and at least one post-dose pharmacokinetic/pharmacodynamic assessment and were included in the pharmacokinetic/pharmacodynamic intent-to-treat analysis; and 12 completed all treatment periods and were included in the evaluable population. In the evaluable population, Metformin DR administered once-daily in the morning had 28% (90% CI -16%, -39%) lower bioavailability (least squares mean ratio of metformin AUC0-24) compared with either once-daily in the evening or twice-daily, although the glucose-lowering effects were maintained. In both studies, adverse events were primarily gastrointestinal in nature, and indicated similar or improved tolerability for Metformin DR vs Metformin IR; there were no clinically meaningful differences in vital signs, physical examinations or laboratory values.

Conclusions/interpretation: Dissociation of gut hormone release and glucose lowering from plasma metformin exposure provides strong supportive evidence for a distal small intestine-mediated mechanism of action. Directly targeting the ileum with Metformin DR once-daily in the morning may provide maximal metformin efficacy with lower doses and substantially reduce plasma exposure. Metformin DR may minimise the risk of lactic acidosis in those at increased risk from metformin therapy, such as individuals with renal impairment.

Trial registration: Clinicaltrials.gov NCT01677299, NCT01804842 FUNDING: : This study was funded by Elcelyx Therapeutics Inc.

Keywords: Clinical care; Clinical study; Lactate; Mechanisms; Metformin.

Figures

Fig. 1
Fig. 1
Study 1: plasma metformin concentrations and bioavailability. (a) Steady state plasma metformin concentrations (mean ± SD) by treatment. White circles, 1,000 mg Metformin IR twice-daily; black squares, 1,000 mg Metformin DR twice-daily; black triangles, 500 mg Metformin DR twice-daily. All treatments were administered at t = −1 min (arrow) relative to the standardised breakfast (t = 0 h) and lunch (t = 5 h) (dotted vertical lines). (b, c) Plasma metformin relative bioavailability and exposure at steady state (data are the geometric LS mean and 90% CI for the ratios of twice-daily 1,000 mg Metformin DR [Met DR] and 500 mg Metformin DR to twice-daily 1,000 mg Metformin IR). Evaluable population, n = 19. ****p < 0.0001 vs twice-daily 1,000 mg Metformin IR
Fig. 2
Fig. 2
Study 1: change in plasma glucose and gut hormone levels. (a) Individual (median) changes from baseline to day 5 in FPG concentrations by treatment. (bd) Mean and SEM for plasma glucose at baseline and day 5. (eg) Mean and SEM for plasma GLP-1 active at baseline and day 5. (hj) Mean and SEM for PYY total at baseline and day 5. White symbols, values at baseline; black symbols, values at day 5. Evaluable population, n = 19. All treatments were administered at t = −1 min relative to the standardised breakfast (t = 0 h) and lunch (t = 5 h; dotted vertical lines). Met, metformin
Fig. 3
Fig. 3
Study 2: change in metformin concentrations and bioavailability. (a) Steady state plasma metformin concentrations (mean ± SD) by treatment. Inset: mean plasma metformin concentration from the time of dose administration (t = 0). Black squares, 1,000 mg Metformin DR once-daily am; white squares, 1,000 mg Metformin DR once-daily pm; grey triangles, 500 mg Metformin DR twice-daily. (b, c) Plasma metformin (mean and 90% CI) relative bioavailability and exposure at steady state. (d) 24 h urine excretion (Ae) of metformin (mean and %CV) by treatment at steady state. Evaluable population, n = 12. Meals were administered at t = 0, 3, 12 and 18 h. **p < 0.01, *p < 0.05
Fig. 4
Fig. 4
Study 2: change in plasma glucose. (a, b) Overall plasma glucose and peak exposure at baseline and steady state. White bars, baseline; black bars, treatment. (c) FPG change from baseline at steady state. Data are the geometric LS mean and SEM (a, b) or LS mean and SEM (c) for the evaluable population (n = 12). *p < 0.05, **p < 0.01, ***p < 0.001

References

    1. Pernicova I, Korbonits M. Metformin – mode of action and clinical implications for diabetes and cancer. Nat Rev Endocrinol. 2014;10:143–156. doi: 10.1038/nrendo.2013.256.
    1. Cusi K, Consoli A, DeFronzo RA. Metabolic effects of metformin on glucose and lactate metabolism in noninsulin-dependent diabetes mellitus. J Clin Endocrinol Metab. 1996;81:4059–4067.
    1. Napolitano A, Miller S, Nicholls AW, et al. Novel gut-based pharmacology of metformin in patients with type 2 diabetes mellitus. PLoS One. 2014;9 doi: 10.1371/journal.pone.0100778.
    1. Duca FA, Cote CD, Rasmussen BA, 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. Buse JB, DeFronzo RA, Rosenstock J, et al. The primary glucose-lowering effect of metformin resides in the gut, not the circulation. Results from short-term pharmacokinetic and 12-week dose-ranging studies. Diabetes Care. 2016;39:198–205. doi: 10.2337/dc15-1531.
    1. Shin NR, Lee JC, Lee HY, et al. An increase in the Akkermansia spp. population induced by metformin treatment improves glucose homeostasis in diet-induced obese mice. Gut. 2014;63:727–735. doi: 10.1136/gutjnl-2012-303839.
    1. Forslund K, Hildebrand F, Nielsen T, et al. Disentangling type 2 diabetes and metformin treatment signatures in the human gut microbiota. Nature. 2015;528:262–266. doi: 10.1038/nature15766.
    1. McCreight LJ, Bailey CJ, Pearson ER. Metformin and the gastrointestinal tract. Diabetologia. 2016;59:426–435. doi: 10.1007/s00125-015-3844-9.
    1. Stepensky D, Friedman M, Raz I, Hoffman A. Pharmacokinetic-pharmacodynamic analysis of the glucose-lowering effect of metformin in diabetic rats reveals first-pass pharmacodynamic effect. Drug Metab Dispos. 2002;30:861–868. doi: 10.1124/dmd.30.8.861.
    1. Stepensky D, Friedman M, Srour W, Raz I, Hoffman A. Preclinical evaluation of pharmacokinetic-pharmacodynamic rationale for oral CR metformin formulation. J Control Release. 2001;71:107–115. doi: 10.1016/S0168-3659(00)00374-6.
    1. Bonora E, Cigolini M, Bosello O, et al. Lack of effect of intravenous metformin on plasma concentrations of glucose, insulin, C-peptide, glucagon and growth hormone in non-diabetic subjects. Curr Med Res Opin. 1984;9:47–51. doi: 10.1185/03007998409109558.
    1. Sum CF, Webster JM, Johnson AB, Catalano C, Cooper BG, Taylor R. The effect of intravenous metformin on glucose metabolism during hyperglycaemia in type 2 diabetes. Diabet Med. 1992;9:61–65. doi: 10.1111/j.1464-5491.1992.tb01716.x.
    1. Graham GG, Punt J, Arora M, et al. Clinical pharmacokinetics of metformin. Clin Pharmacokinet. 2011;50:81–98. doi: 10.2165/11534750-000000000-00000.
    1. Vidon N, Chaussade S, Noel M, Franchisseur C, Huchet B, Bernier JJ. Metformin in the digestive tract. Diabetes Res Clin Pract. 1988;4:223–229. doi: 10.1016/S0168-8227(88)80022-6.
    1. Tucker GT, Casey C, Phillips PJ, Connor H, Ward JD, Woods HF. Metformin kinetics in healthy subjects and in patients with diabetes mellitus. Br J Clin Pharmacol. 1981;12:235–246. doi: 10.1111/j.1365-2125.1981.tb01206.x.
    1. Bailey CJ, Wilcock C, Scarpello JH. Metformin and the intestine. Diabetologia. 2008;51:1552–1553. doi: 10.1007/s00125-008-1053-5.
    1. Jensen JB, Sundelin EI, Jakobsen S et al (2016) [11C]-metformin distribution in the liver and small intestine using dynamic PET in mice demonstrates tissue-specific transporter dependency. Diabetes. doi:10.2337/db16-0032
    1. Jensen JB, Gormsen LC, Sundelin E et al (2015) Organ-specific uptake and elimination of metformin can be determined in vivo in mice and humans by PET-imaging using a novel 11C-metformin tracer. ADA Scientific Sessions pp. 128-LB (Abstract)
    1. Pentikainen PJ, Neuvonen PJ, Penttila A. Pharmacokinetics of metformin after intravenous and oral administration to man. Eur J Clin Pharmacol. 1979;16:195–202. doi: 10.1007/BF00562061.
    1. Lee N, Duan H, Hebert MF, Liang CJ, Rice KM, Wang J. Taste of a pill: organic cation transporter-3 (OCT3) mediates metformin accumulation and secretion in salivary glands. J Biol Chem. 2014;289:27055–27064. doi: 10.1074/jbc.M114.570564.
    1. Wilcock C, Bailey CJ. Accumulation of metformin by tissues of the normal and diabetic mouse. Xenobiotica. 1994;24:49–57. doi: 10.3109/00498259409043220.
    1. Proctor WR, Bourdet DL, Thakker DR. Mechanisms underlying saturable intestinal absorption of metformin. Drug Metab Dispos. 2008;36:1650–1658. doi: 10.1124/dmd.107.020180.
    1. Garber AJ, Duncan TG, Goodman AM, Mills DJ, Rohlf JL. Efficacy of metformin in type II diabetes: results of a double-blind, placebo-controlled, dose-response trial. Am J Med. 1997;103:491–497. doi: 10.1016/S0002-9343(97)00254-4.
    1. Hong Y, Rohatagi S, Habtemariam B, Walker JR, Schwartz SL, Mager DE. Population exposure-response modeling of metformin in patients with type 2 diabetes mellitus. J Clin Pharmacol. 2008;48:696–707. doi: 10.1177/0091270008316884.
    1. Inzucchi SE, Bergenstal RM, Buse JB, et al. Management of hyperglycemia in type 2 diabetes: a patient-centered approach. Position statement of the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD) Diabetologia. 2012;55:1577–1596. doi: 10.1007/s00125-012-2534-0.
    1. Owen MR, Doran E, Halestrap AP. Evidence that metformin exerts its anti-diabetic effects through inhibition of complex 1 of the mitochondrial respiratory chain. Biochem J. 2000;348(Pt 3):607–614. doi: 10.1042/bj3480607.
    1. Zhou G, Myers R, Li Y, et al. Role of AMP-activated protein kinase in mechanism of metformin action. J Clin Invest. 2001;108:1167–1174. doi: 10.1172/JCI13505.
    1. Foretz M, Hebrard S, Leclerc J, et al. Metformin inhibits hepatic gluconeogenesis in mice independently of the LKB1/AMPK pathway via a decrease in hepatic energy state. J Clin Invest. 2010;120:2355–2369. doi: 10.1172/JCI40671.
    1. Madiraju A, Erion D, Rahimi Y, et al. Metformin suppresses gluconeogenesis by inhibiting mitochondrial glycerophosphate dehydrogenase. Nature. 2014;510:542–546. doi: 10.1038/nature13270.
    1. Carter D, Howlett HC, Wiernsperger NF, Bailey CJ. Differential effects of metformin on bile salt absorption from the jejunum and ileum. Diabetes Obes Metab. 2003;5:120–125. doi: 10.1046/j.1463-1326.2003.00252.x.
    1. Panossian Z, Drury PL, Cundy T. Reversible severe deterioration of glycaemic control after withdrawal of metformin treatment. Diabetologia. 2012;55:267–269. doi: 10.1007/s00125-011-2351-x.
    1. Migoya EM, Bergeron R, Miller JL, et al. Dipeptidyl peptidase-4 inhibitors administered in combination with metformin result in an additive increase in the plasma concentration of active GLP-1. Clin Pharmacol Ther. 2010;88:801–808. doi: 10.1038/clpt.2010.184.
    1. Purnell JQ, Weyer C. Weight effect of current and experimental drugs for diabetes mellitus: from promotion to alleviation of obesity. Treat Endocrinol. 2003;2:33–47. doi: 10.2165/00024677-200302010-00004.
    1. Schmidt JB, Gregersen NT, Pedersen SD, et al. Effects of PYY3-36 and GLP-1 on energy intake, energy expenditure, and appetite in overweight men. Am J Physiol Endocrinol Metab. 2014;306:E1248–E1256. doi: 10.1152/ajpendo.00569.2013.
    1. Sloth B, Holst JJ, Flint A, Gregersen NT, Astrup A. Effects of PYY1-36 and PYY3-36 on appetite, energy intake, energy expenditure, glucose and fat metabolism in obese and lean subjects. Am J Physiol Endocrinol Metab. 2007;292:E1062–E1068. doi: 10.1152/ajpendo.00450.2006.
    1. Dantas RO, Aben-Athar CG. Aspects of sleep effects on the digestive tract. Arq Gastroenterol. 2002;39:55–59. doi: 10.1590/S0004-28032002000100010.
    1. Glucophage (metformin hydrochloride) and Glucophage XR (extended-release) [package insert] (2009) US prescribing information. Bristol-Myers Squibb, Bristol, NJ, USA. Available from , accessed 19 May 2016
    1. Stang M, Wysowski DK, Butler-Jones D. Incidence of lactic acidosis in metformin users. Diabetes Care. 1999;22:925–927. doi: 10.2337/diacare.22.6.925.
    1. Misbin RI, Green L, Stadel BV, Gueriguian JL, Gubbi A, Fleming GA. Lactic acidosis in patients with diabetes treated with metformin. N Engl J Med. 1998;338:265–266. doi: 10.1056/NEJM199801223380415.
    1. DeFronzo R, Fleming GA, Chen K, Bicsak TA. Metformin-associated lactic acidosis: current perspectives on causes and risk. Metabolism. 2016;65:20–29. doi: 10.1016/j.metabol.2015.10.014.
    1. Jones GC, Sainsbury CA. Comment on ‘A justification for less restrictive guidelines on the use of metformin in stable chronic renal failure’. Diabet Med. 2015;32:287. doi: 10.1111/dme.12626.

Source: PubMed

Подписаться