The Effects of a GLP-1 Analog on Glucose Homeostasis in Type 2 Diabetes Mellitus Quantified by an Integrated Glucose Insulin Model

R M Røge, S Klim, S H Ingwersen, M C Kjellsson, N R Kristensen, R M Røge, S Klim, S H Ingwersen, M C Kjellsson, N R Kristensen

Abstract

In recent years, several glucagon-like peptide-1 (GLP-1)-based therapies for the treatment of type 2 diabetes mellitus (T2DM) have been developed. The aim of this work was to extend the semimechanistic integrated glucose-insulin model to include the effects of a GLP-1 analog on glucose homeostasis in T2DM patients. Data from two trials comparing the effect of steady-state liraglutide vs. placebo on the responses of postprandial glucose and insulin in T2DM patients were used for model development. The effect of liraglutide was incorporated in the model by including a stimulatory effect on insulin secretion. Furthermore, for one of the trials an inhibitory effect on glucose absorption was included to account for a delay in gastric emptying. As other GLP-1 receptor agonists have similar modes of action, it is believed that the model can also be used to describe the effect of other receptor agonists on glucose homeostasis.

Figures

Figure 1
Figure 1
Schematic presentation of the integrated model including glucose, insulin, and liraglutide. Full arrows indicate flows and broken arrows indicate control mechanisms. GC and GP, central and peripheral compartments for glucose; GE2, glucose effect compartment for control of insulin secretion; GA and GT, absorption (GA) and transit (GT) compartment for glucose absorption; I, compartment for insulin distribution; IE, insulin effect compartment for control of glucose elimination; Q, CLG, and CLGI, clearance parameters for glucose; ka, rate constant for glucose absorption; D, glucose dose; CLI, endogenous insulin clearance; kGE2, and kIE, rate constants for the delay of effect compartment concentrations; LC, compartment for liraglutide distribution; LA, depot compartment for liraglutide absorption; kaL, rate constant for liraglutide absorption; CLL, liraglutide clearance.
Figure 2
Figure 2
(Left) Time course for liraglutide concentration. The dots show the geometric mean and the corresponding 95% confidence interval of the data. The solid lines show the geometric mean of the individual predictions (solid line) and the geometric mean of the population predictions (dashed line). (Right) Visual predictive check for the PK model for liraglutide. Solid lines represent median concentrations and dashed lines represent 5th and 95th percentiles. The red lines represent the data and the black lines represent simulations from the model. The shaded area represents the 95% confidence intervals for the model predicted median. Time zero is the time at which the last dose of liraglutide 1.8 mg was administered.
Figure 3
Figure 3
Time courses for plasma glucose and insulin concentrations for trial 1. The dots show the geometric mean and the corresponding 95% confidence interval of the data. The solid lines show the geometric mean of the individual predictions (solid lines) and the geometric mean of the population predictions (dashed lines). Time zero is defined as the time of the first measurement in the MTT, which is 10 minutes before meal ingestion.
Figure 4
Figure 4
Time courses for plasma glucose and insulin concentrations for trial 2. The dots show the geometric mean and the corresponding 95% confidence interval of the data. The solid lines show the geometric mean of the individual predictions (solid lines) and the geometric mean of the population predictions (dashed lines). Time zero is defined as the time of the first measurement in the MTT, which is 15 minutes before meal ingestion.
Figure 5
Figure 5
(Left) Model predicted insulin secretion for the different dose levels for trial 1. (Right) Mean baseline corrected postprandial plasma glucose profiles during the MTTs for trial 1. All placebo data was included in the calculation of the mean, i.e., each subject contributes with three placebo profiles.

References

    1. Campbell JE. Drucker DJ. Pharmacology, physiology, and mechanisms of incretin hormone action. Cell. Metab. 2013;17:819–837.
    1. Vilsbøll T, Agersø H, Krarup T. Holst JJ. Similar elimination rates of glucagon-like peptide-1 in obese type 2 diabetic patients and healthy subjects. J. Clin. Endocrinol. Metab. 2003;88:220–224.
    1. Garber AJ. Novel GLP-1 receptor agonists for diabetes. Expert Opin. Investig. Drugs. 2012;211:45–57.
    1. Madsbad S. Exenatide and liraglutide: different approaches to develop GLP-1 receptor agonists (incretin mimetics) — preclinical and clinical results. Best. Pract. Res. Clin. Endocrinol. Metab. 2009;23:463–477.
    1. Russell-Jones D. Molecular, pharmacological and clinical aspects of liraglutide, a once-daily human GLP-1 analogue. Mol. Cell. Endocrinol. 2009;297:137–140.
    1. Montanya E. Sesti G. A review of efficacy and safety data regarding the use of liraglutide, a once-daily human glucagon-like peptide 1 analogue, in the treatment of type 2 diabetes mellitus. Clin. Ther. 2009;31:2472–2488.
    1. Silber HE, Jauslin PM, Frey N, Gieschke R, Simonsson USH. Karlsson MO. An integrated model for glucose and insulin regulation in healthy volunteers and type 2 diabetic patients following intravenous glucose provocations. J. Clin. Pharmacol. 2007;47:1159–1171.
    1. Silber HE, Frey N. Karlsson MO. An integrated glucose-insulin model to describe oral glucose tolerance test data in healthy volunteers. J. Clin. Pharmacol. 2010;50:246–256.
    1. Jauslin PM, et al. An integrated glucose-insulin model to describe oral glucose tolerance test data in type 2 diabetics. J. Clin. Pharmacol. 2007;47:1244–1255.
    1. Jauslin PM, Frey N. Karlsson MO. Modeling of 24-hour glucose and insulin profiles of patients with type 2 diabetes. J. Clin. Pharmacol. 2011;51:153–164.
    1. Schneck KB, Zhang X, Bauer R, Karlsson MO. Sinha VP. Assessment of glycemic response to an oral glucokinase activator in a proof of concept study: Application of a semi-mechanistic, integrated glucose-insulin-glucagon model. J. Pharmacokinet. Pharmacodyn. 2013;40:67–80.
    1. Røge RM, Klim S, Kristensen NR, Ingwersen SH. Kjellsson MC. Modeling of 24-hour glucose and insulin profiles in patients with type 2 diabetes mellitus treated with biphasic insulin aspart. J. Clin. Pharmacol. 2014
    1. Zhang X, Schneck K, Bue-Valleskey J, Yeo KP, Heathman M. Sinha V. Dose selection using a semi-mechanistic integrated glucose-insulin-glucagon model: designing phase 2 trials for a novel oral glucokinase activator. J. Pharmacokinet. Pharmacodyn. 2013;40:53–65.
    1. Flint A, Kapitza C, Hindsberger C. Zdravkovic M. The once-daily human glucagon-like peptide-1 (GLP-1) analog liraglutide improves postprandial glucose levels in type 2 diabetes patients. Adv. Ther. 2011;28:213–226.
    1. Hermansen K, et al. Liraglutide suppresses postprandial triglyceride and apolipoprotein B48 elevations after a fat-rich meal in patients with type 2 diabetes: a randomized, double-blind, placebo-controlled, cross-over trial. Diabetes Obes. Metab. 2013;15:1040–1048.
    1. Ingwersen SH, et al. Dosing rationale for liraglutide in type 2 diabetes mellitus: a pharmacometric assessment. J. Clin. Pharmacol. 2012;52:815–1823.
    1. Watson E, Jonker DM, Jacobsen LV. Ingwersen SH. Population pharmacokinetics of liraglutide, a once-daily human glucagon-like peptide-1 analog, in healthy volunteers and subjects with type 2 diabetes, and comparison to twice-daily exenatide. J. Clin. Pharmacol. 2010;52:886–894.
    1. Degn KB, et al. One week's treatment with the long-acting glucagon-like peptide 1 derivative liraglutide (NN2211) markedly improves 24-h glycemia and α- and β-cell function and reduces endogenous glucose release in patients with type 2 diabetes. Diabetes. 2004;53:1187–1194.
    1. Landersdorfer CB, He YL. Jusko WJ. Mechanism-based population modelling of the effects of vildagliptin on GLP-1, glucose and insulin in patients with type 2 diabetes. Br. J. Clin. Pharmacol. 2011;73:373–390.
    1. Mager DE, Abernethy DR, Egan JM. Elahi D. Exendin-4 pharmacodynamics: insights from the hyperglycemic clamp technique. J. Pharmacol. Exp. Ther. 2004;311:830–835.
    1. Agersø H. Vicini P. Pharmacodynamics of NN2211, a novel long acting GLP-1 derivative. Eur. J. Pharm. Sci. 2003;19:141–150.
    1. Calbet JAL. MacLean DA. Role of caloric content on gastric emptying in humans. J. Physiol. 1997;498:553–559.
    1. Jenkins DJA, et al. Glycemic index of foods: A physiological basis for carbohydrate exchange. Am. J. Clin. Nutr. 1981;34:362–366.
    1. Gannon MC, Nuttall FQ, Neil BJ. Westphal SA. The insulin and glucose responses to meals of glucose plus various proteins in type II diabetic subjects. Metabolism. 1988;37:1081–1088.
    1. Normand S, et al. Influence of dietary fat on postprandial glucose metabolism (exogenous and endogenous) using intrinsically 13C-enriched durum wheat. Br. J. Nutr. 2001;86:3–11.
    1. Jauslin P, Dubar M, Sebastien B, Laveille C. Gisleskog PO. 1989. Comparison of post-prandial glucose control by two GLP-1 receptor agonists (lixisenatide and liraglutide) in type 2 diabetes. Poster presented at the Population Approach Group Meeting in Alicante, Spain < >.
    1. Beal SL, Sheiner LB. Boeckmann AJ. Icon development solutions. NONMEM Users Guides. Ellicott City, MD: Icon Development Solutions; 2014. –2006.

Source: PubMed

3
Abonner