LY3298176, a novel dual GIP and GLP-1 receptor agonist for the treatment of type 2 diabetes mellitus: From discovery to clinical proof of concept

Tamer Coskun, Kyle W Sloop, Corina Loghin, Jorge Alsina-Fernandez, Shweta Urva, Krister B Bokvist, Xuewei Cui, Daniel A Briere, Over Cabrera, William C Roell, Uma Kuchibhotla, Julie S Moyers, Charles T Benson, Ruth E Gimeno, David A D'Alessio, Axel Haupt, Tamer Coskun, Kyle W Sloop, Corina Loghin, Jorge Alsina-Fernandez, Shweta Urva, Krister B Bokvist, Xuewei Cui, Daniel A Briere, Over Cabrera, William C Roell, Uma Kuchibhotla, Julie S Moyers, Charles T Benson, Ruth E Gimeno, David A D'Alessio, Axel Haupt

Abstract

Objective: A novel dual GIP and GLP-1 receptor agonist, LY3298176, was developed to determine whether the metabolic action of GIP adds to the established clinical benefits of selective GLP-1 receptor agonists in type 2 diabetes mellitus (T2DM).

Methods: LY3298176 is a fatty acid modified peptide with dual GIP and GLP-1 receptor agonist activity designed for once-weekly subcutaneous administration. LY3298176 was characterised in vitro, using signaling and functional assays in cell lines expressing recombinant or endogenous incretin receptors, and in vivo using body weight, food intake, insulin secretion and glycemic profiles in mice. A Phase 1, randomised, placebo-controlled, double-blind study was comprised of three parts: a single-ascending dose (SAD; doses 0.25-8 mg) and 4-week multiple-ascending dose (MAD; doses 0.5-10 mg) studies in healthy subjects (HS), followed by a 4-week multiple-dose Phase 1 b proof-of-concept (POC; doses 0.5-15 mg) in patients with T2DM (ClinicalTrials.gov no. NCT02759107). Doses higher than 5 mg were attained by titration, dulaglutide (DU) was used as a positive control. The primary objective was to investigate safety and tolerability of LY3298176.

Results: LY3298176 activated both GIP and GLP-1 receptor signaling in vitro and showed glucose-dependent insulin secretion and improved glucose tolerance by acting on both GIP and GLP-1 receptors in mice. With chronic administration to mice, LY3298176 potently decreased body weight and food intake; these effects were significantly greater than the effects of a GLP-1 receptor agonist. A total of 142 human subjects received at least 1 dose of LY3298176, dulaglutide, or placebo. The PK profile of LY3298176 was investigated over a wide dose range (0.25-15 mg) and supports once-weekly administration. In the Phase 1 b trial of diabetic subjects, LY3298176 doses of 10 mg and 15 mg significantly reduced fasting serum glucose compared to placebo (least square mean [LSM] difference [95% CI]: -49.12 mg/dL [-78.14, -20.12] and -43.15 mg/dL [-73.06, -13.21], respectively). Reductions in body weight were significantly greater with the LY3298176 1.5 mg, 4.5 mg and 10 mg doses versus placebo in MAD HS (LSM difference [95% CI]: -1.75 kg [-3.38, -0.12], -5.09 kg [-6.72, -3.46] and -4.61 kg [-6.21, -3.01], respectively) and doses of 10 mg and 15 mg had a relevant effect in T2DM patients (LSM difference [95% CI]: -2.62 kg [-3.79, -1.45] and -2.07 kg [-3.25, -0.88], respectively. The most frequent side effects reported with LY3298176 were gastrointestinal (vomiting, nausea, decreased appetite, diarrhoea, and abdominal distension) in both HS and patients with T2DM; all were dose-dependent and considered mild to moderate in severity.

Conclusions: Based on these results, the pharmacology of LY3298176 translates from preclinical to clinical studies. LY3298176 has the potential to deliver clinically meaningful improvement in glycaemic control and body weight. The data warrant further clinical evaluation of LY3298176 for the treatment of T2DM and potentially obesity.

Keywords: Glucagon-like peptide-1; Glucose-dependent insulinotropic polypeptide; LY3298176; Obesity; Type 2 diabetes mellitus.

Copyright © 2018 The Authors. Published by Elsevier GmbH.. All rights reserved.

Figures

Figure 1
Figure 1
Discovery and Characterization of LY3298176, a GIP-based Dual Incretin Receptor Agonist. (A) Structure schematic of the dual GIP and GLP-1 receptor agonist, LY3298176. (B) Representative concentration response curves for stimulation of cAMP accumulation by GIP, GLP-1 or LY3298176 in HEK293 cells expressing human GIPR or GLP-1R. (C) cAMP accumulation in human pancreatic ECN90 beta-cells in response to treatment with GLP-1, GIP, the combination of GLP-1 plus GIP, or LY3298176. P < 0.05 using one-way ANOVA versus GLP-1 (#) or GIP (++). (D) cAMP accumulation in human adipocytes. All data are expressed as mean ± SEM.
Figure 2
Figure 2
LY3298176 Enhances Islet Insulin Secretion and Improves Glucose Tolerance in Mice. Glucose-stimulated insulin secretion by GIP, GLP-1, or LY3298176 in isolated islets from wild-type (A), GIPR null (B), and GLP-1R null mice (C). Antagonists to GLP-1R (exendin-4(9-39)) and GIPR (modified GIP(3-30)NH2) were used to assess receptor specificity. P < 0.05 using one-way ANOVA versus vehicle treatment in high glucose (*). Glucose excursions from intraperitoneal glucose tolerance tests in wild-type (D), GIPR null (E), and GLP-1R null (F) mice administered LY3298176 or the selective agonists semaglutide or [d-Ala2]GIP. Glucose was administered 18 h after a single injection of LY3298176 or semaglutide and 1 h following [d-Ala2]GIP; glucose AUC(0–120 min, mg. min/dL) is depicted in the upper right panels of (D, E, and F). Data are presented as Mean ± SEM of 6 mice per group. P < 0.05 using one-way ANOVA versus vehicle (*).
Figure 3
Figure 3
LY3298176 Lowers Body Weight in Obese Mice. The starting body weight of DIO mice was around 45 g. Body weight loss in DIO mice chronically administered semaglutide (A), LA-GIPRA (B), or LY3298176 (C). Metabolic effects of LY3298176 (♦) or semaglutide (▲) compared to vehicle treatment (■) on body weight (D), cumulative food intake (E), and energy expenditure (F) in DIO mice. For body weight and food consumption differences, P < 0.05 using one-way ANOVA repeated measures versus the vehicle (*) or semaglutide (+) treated groups. For energy expenditure, data presented are least square means for each treatment over time, which were adjusted for body weight as indicated in the statistical model as described in the methods. P < 0.05 using ANCOVA versus the vehicle (*) or semaglutide (+) treated groups. All data are expressed as mean ± SEM.
Figure 4
Figure 4
Body Weight Change and Glucose Response to a OGTT in Healthy Subjects (MAD Part) *Statistically significant at a 5% confidence level compared to placebo. (A) Change from baseline in body weight over time. Data presented as LSM (SE) (B) Treatment differences in body weight on Day 29. Data presented as LSM difference (95% CI) for LY3298176 versus placebo (C) Glucose AUC(0–2 h) on Day 23. Data presented as mean ± SD. Subjects in the 5/5/8/10 mg group received 5 mg LY3298176 on Day 1 and Day 8, 8 mg LY3298176 on Day 15, and 10 mg LY3298176 on Day 22. DU = dulaglutide; LY = LY3298176.
Figure 5
Figure 5
Body Weight Change, Glucose and Insulin Response to a OGTT and Fasting Glucose Response in Patients with T2DM (Phase 1 b POC Part) *Statistically significant at 5% significance level compared to placebo (A) Change from baseline in body weight over time. Data presented as LSM (SE) (B) Treatment differences in body weight on Day 29. Data presented as LSM difference (95% CI) for LY3298176 versus placebo (C) Glucose AUC(0–2 h) on Day 23. Data presented as mean ± SD (D) Insulin AUC(0–2 h) on Day 23. Data presented as arithmetic mean ± SD (E) 7-Point SMBG on Days −2 (baseline), 8 (2nd dose), 15 (3rd dose) and 22 (4 t h dose). Data presented as mean ± SD (F) Treatment differences in fasting glucose on Day 29. Data presented as LSM difference (95% CI) for LY3298176 versus placebo. Subjects in the 5/5/10/10 mg group received 5 mg LY3298176 on Day 1 and Day 8, and 10 mg LY3298176 on Day 15 and Day 22. Subjects in the 5/5/10/15 mg group received 5 mg LY3298176 on Day 1 and Day 8, 10 mg LY3298176 on Day 15, and 15 mg LY3298176 on Day 22. LY = LY3298176.

References

    1. Andersen A. Glucagon-like peptide 1 in health and disease. Nature Reviews Endocrinology. 2018;14(7):390–403.
    1. Richards P. Identification and characterization of GLP-1 receptor-expressing cells using a new transgenic mouse model. Diabetes. 2014;63(4):1224–1233.
    1. Holst J.J. Truncated glucagon-like peptide I, an insulin-releasing hormone from the distal gut. FEBS Letters. 1987;211(2):169–174.
    1. Kreymann B. Glucagon-like peptide-1 7-36: a physiological incretin in man. Lancet. 1987;2(8571):1300–1304.
    1. Imeryuz N. Glucagon-like peptide-1 inhibits gastric emptying via vagal afferent-mediated central mechanisms. American Journal of Physiology. 1997;273(4 Pt 1):G920–G927.
    1. Nauck M.A. Glucagon-like peptide 1 inhibition of gastric emptying outweighs its insulinotropic effects in healthy humans. American Journal of Physiology. 1997;273(5 Pt 1):E981–E988.
    1. Schirra J. Mechanisms of the antidiabetic action of subcutaneous glucagon-like peptide-1(7-36)amide in non-insulin dependent diabetes mellitus. Journal of Endocrinology. 1998;156(1):177–186.
    1. Turton M.D. A role for glucagon-like peptide-1 in the central regulation of feeding. Nature. 1996;379(6560):69–72.
    1. Holz G.G. cAMP-dependent mobilization of intracellular Ca2+ stores by activation of ryanodine receptors in pancreatic beta-cells. A Ca2+ signaling system stimulated by the insulinotropic hormone glucagon-like peptide-1-(7-37) Journal of Biological Chemistry. 1999;274:14147–14156.
    1. Kang G., Chepurny O.G., Holz G.G. cAMP-regulated guanine nucleotide exchange factor II (Epac2) mediates Ca2+-induced Ca2+ release in INS-1 pancreatic beta-cells. Journal of Physiology. 2001;536:375–385.
    1. Vilsboll T. No reactive hypoglycaemia in Type 2 diabetic patients after subcutaneous administration of GLP-1 and intravenous glucose. Diabetic Medicine. 2001;18(2):144–149.
    1. English W.J., Williams D.B. Metabolic and bariatric surgery: an effective treatment option for obesity and cardiovascular disease. Progress in Cardiovascular Diseases. 2018
    1. Drucker D.J. Mechanisms of action and therapeutic application of glucagon-like peptide-1. Cell Metabolism. 2018;27(4):740–756.
    1. Nauck M.A., Meier J.J. Incretin hormones: their role in health and disease. Diabetes, Obesity and Metabolism. 2018;20(Suppl 1):5–21.
    1. Vollmer K. Predictors of incretin concentrations in subjects with normal, impaired, and diabetic glucose tolerance. Diabetes. 2008;57(3):678–687.
    1. Dupre J. Stimulation of insulin secretion by gastric inhibitory polypeptide in man. The Journal of Cinical Endocrinology and Metabolism. 1973;37:826–828.
    1. Meier J.J. Gastric inhibitory polypeptide (GIP) dose-dependently stimulates glucagon secretion in healthy human subjects at euglycaemia. Diabetologia. 2003;46(6):798–801.
    1. Christensen M.B. Glucose-dependent insulinotropic polypeptide: blood glucose stabilizing effects in patients with type 2 diabetes. The Journal of Cinical Endocrinology and Metabolism. 2014;99(3):E418–E426.
    1. Christensen M. Glucose-dependent insulinotropic polypeptide augments glucagon responses to hypoglycemia in type 1 diabetes. Diabetes. 2015;64(1):72–78.
    1. Christensen M. Glucose-dependent insulinotropic polypeptide: a bifunctional glucose-dependent regulator of glucagon and insulin secretion in humans. Diabetes. 2011;60:3103–3109.
    1. Pederson R.A., Brown J.C. Interaction of gastric inhibitory polypeptide, glucose, and arginine on insulin and glucagon secretion from the perfused rat pancreas. Endocrinology. 1978;103:610–615.
    1. Yip R.G. Functional GIP receptors are present on adipocytes. Endocrinology. 1998;139(9):4004–4007.
    1. Nyberg J. Immunohistochemical distribution of glucose-dependent insulinotropic polypeptide in the adult rat brain. Journal of Neuroscience Research. 2007;85(10):2099–2119.
    1. Song D.H. Glucose-dependent insulinotropic polypeptide enhances adipocyte development and glucose uptake in part through Akt activation. Gastroenterology. 2007;133(6):1796–1805.
    1. Getty-Kaushik L. Glucose-dependent insulinotropic polypeptide modulates adipocyte lipolysis and reesterification. Obesity. 2006;14(7):1124–1131.
    1. Kim S.J., Nian C., McIntosh C.H. Activation of lipoprotein lipase by glucose-dependent insulinotropic polypeptide in adipocytes. A role for a protein kinase B, LKB1, and AMP-activated protein kinase cascade. Journal of Biological Chemistry. 2007;282(12):8557–8567.
    1. Miyawaki K. Inhibition of gastric inhibitory polypeptide signaling prevents obesity. Nature Medicine. 2002;8(7):738–742.
    1. Eckel R.H., Fujimoto W.Y., Brunzell J.D. Gastric inhibitory polypeptide enhanced lipoprotein lipase activity in cultured preadipocytes. Diabetes. 1979;28(12):1141–1142.
    1. Asmar M. Glucose-dependent insulinotropic polypeptide increases blood flow in adipose tissue of humans by recruiting capillaries. The Journal of Cinical Endocrinology and Metabolism. 2018 [Epub ahead of print]
    1. Kim S.J. GIP-overexpressing mice demonstrate reduced diet-induced obesity and steatosis, and improved glucose homeostasis. PloS One. 2012;7(7)
    1. Finan B. Unimolecular dual incretins maximize metabolic benefits in rodents, monkeys, and humans. Science Translational Medicine. 2013;5(209):209ra151.
    1. NamKoong C. Central administration of GLP-1 and GIP decreases feeding in mice. Biochemical and Biophysical Research Communications. 2017;490(2):247–252.
    1. Hojberg P.V. Near normalisation of blood glucose improves the potentiating effect of GLP-1 on glucose-induced insulin secretion in patients with type 2 diabetes. Diabetologia. 2008;51(4):632–640.
    1. Hojberg P.V. Four weeks of near-normalisation of blood glucose improves the insulin response to glucagon-like peptide-1 and glucose-dependent insulinotropic polypeptide in patients with type 2 diabetes. Diabetologia. 2009;52(2):199–207.
    1. Frias J.P. The sustained effects of a dual GIP/GLP-1 receptor agonist, NNC0090-2746, in patients with type 2 diabetes. Cell Metabolism. 2017;26(2):343–352.e2.
    1. Portron A. Pharmacodynamics, pharmacokinetics, safety and tolerability of the novel dual glucose-dependent insulinotropic polypeptide/glucagon-like peptide-1 agonist RG7697 after single subcutaneous administration in healthy subjects. Diabetes, Obesity and Metabolism. 2017;19(10):1446–1453.
    1. Schmitt C. Pharmacodynamics, pharmacokinetics and safety of multiple ascending doses of the novel dual glucose-dependent insulinotropic polypeptide/glucagon-like peptide-1 agonist RG7697 in people with type 2 diabetes mellitus. Diabetes, Obesity and Metabolism. 2017;19(10):1436–1445.
    1. Frias J.P. Efficacy and safety of LY3298176, a novel GIP and GLP-1 receptor dual agonist in patients with type 2 diabetes: a 26-week, randomised, placebo- and active comparator-controlled trial. The Lancet. 2018 [in press]
    1. Lau J. Discovery of the once-weekly glucagon-like peptide-1 (GLP-1) analogue semaglutide. Journal of Medicinal Chemistry. 2015;58(18):7370–7380.
    1. Hinke S.A. Dipeptidyl peptidase IV-resistant [D-Ala2]Glucose-Dependent insulinotropic polypeptide (GIP) improves glucose tolerance in normal and obese diabetic rats. Diabetes. 2002;51(3):652–661.
    1. Auld D.S. Receptor binding assays for HTS and drug discovery. In: Sittampalam G.S., editor. Assay guidance manual. 2004. Bethesda (MD)
    1. Bueno A.B. Positive allosteric modulation of the glucagon-like peptide-1 receptor by diverse electrophiles. Journal of Biological Chemistry. 2016;291(20):10700–10715.
    1. Tschop M.H. A guide to analysis of mouse energy metabolism. Nature Methods. 2011;9(1):57–63.
    1. Speakman J.R., Fletcher Q., Vaanholt L. The '39 steps': an algorithm for performing statistical analysis of data on energy intake and expenditure. Dis Model Mech. 2013;6(2):293–301.
    1. Hansen L.S. N-terminally and C-terminally truncated forms of glucose-dependent insulinotropic polypeptide are high-affinity competitive antagonists of the human GIP receptor. British Journal of Pharmacology. 2016;173(5):826–838.
    1. Patterson J.T. A novel human-based receptor antagonist of sustained action reveals body weight control by endogenous GLP-1. ACS Chemical Biology. 2011;6(2):135–145.
    1. Barrington P. A 5-week study of the pharmacokinetics and pharmacodynamics of LY2189265, a novel, long-acting glucagon-like peptide-1 analogue, in patients with type 2 diabetes. Diabetes, Obesity and Metabolism. 2011;13(5):426–433.
    1. Barrington P. LY2189265, a long-acting glucagon-like peptide-1 analogue, showed a dose-dependent effect on insulin secretion in healthy subjects. Diabetes, Obesity and Metabolism. 2011;13(5):434–438.
    1. Schmidt W.E. Early clinical studies with liraglutide. Int J Clin Pract Suppl. 2010;(167):12–20.
    1. Asmar M. Glucose-dependent insulinotropic polypeptide may enhance fatty acid re-esterification in subcutaneous abdominal adipose tissue in lean humans. Diabetes. 2010;59(9):2160–2163.
    1. Finan B. Reappraisal of GIP pharmacology for metabolic diseases. Trends in Molecular Medicine. 2016;22(5):359–376.
    1. Norregaard P.K. A novel GIP analogue, ZP4165, enhances glucagon-like peptide-1-induced body weight loss and improves glycaemic control in rodents. Diabetes, Obesity and Metabolism. 2018;20(1):60–68.
    1. Lindauer K. Early clinical development of the dual GLP-1/glucagon receptor agonist SAR425899 and establishment of a population PK/PD model. Diabetologia. 2016;59(Suppl 1):1–581.
    1. Ambery P. MEDI0382, a GLP-1 and glucagon receptor dual agonist, in obese or overweight patients with type 2 diabetes: a randomised, controlled, double-blind, ascending dose and phase 2a study. The Lancet. 2018;391(10140):2607–2618.

Source: PubMed

3
Tilaa