Efficacy and safety of cotadutide, a dual glucagon-like peptide-1 and glucagon receptor agonist, in a randomized phase 2a study of patients with type 2 diabetes and chronic kidney disease

Victoria E R Parker, Thuong Hoang, Heike Schlichthaar, Fraser W Gibb, Barbara Wenzel, Maximillian G Posch, Ludger Rose, Yi-Ting Chang, Marcella Petrone, Lars Hansen, Philip Ambery, Lutz Jermutus, Hiddo J L Heerspink, Rory J McCrimmon, Victoria E R Parker, Thuong Hoang, Heike Schlichthaar, Fraser W Gibb, Barbara Wenzel, Maximillian G Posch, Ludger Rose, Yi-Ting Chang, Marcella Petrone, Lars Hansen, Philip Ambery, Lutz Jermutus, Hiddo J L Heerspink, Rory J McCrimmon

Abstract

Aim: To assess the efficacy, safety and tolerability of cotadutide in patients with type 2 diabetes mellitus and chronic kidney disease.

Materials and methods: In this phase 2a study (NCT03550378), patients with body mass index 25-45 kg/m2 , estimated glomerular filtration rate 30-59 ml/min/1.73 m2 and type 2 diabetes [glycated haemoglobin 6.5-10.5% (48-91 mmol/mol)] controlled with insulin and/or oral therapy combination, were randomized 1:1 to once-daily subcutaneous cotadutide (50-300 μg) or placebo for 32 days. The primary endpoint was plasma glucose concentration assessed using a mixed-meal tolerance test.

Results: Participants receiving cotadutide (n = 21) had significant reductions in the mixed-meal tolerance test area under the glucose concentration-time curve (-26.71% vs. +3.68%, p < .001), more time in target glucose range on continuous glucose monitoring (+14.79% vs. -21.23%, p = .001) and significant reductions in absolute bodyweight (-3.41 kg vs. -0.13 kg, p < .001) versus placebo (n = 20). In patients with baseline micro- or macroalbuminuria (n = 18), urinary albumin-to-creatinine ratios decreased by 51% at day 32 with cotadutide versus placebo (p = .0504). No statistically significant difference was observed in mean change in estimated glomerular filtration rate between treatments. Mild/moderate adverse events occurred in 71.4% of participants receiving cotadutide and 35.0% receiving placebo.

Conclusions: We established the efficacy of cotadutide in this patient population, with significantly improved postprandial glucose control and reduced bodyweight versus placebo. Reductions in urinary albumin-to-creatinine ratios suggest potential benefits of cotadutide on kidney function, supporting further evaluation in larger, longer-term clinical trials.

Conflict of interest statement

VP, TH, YC, MPe, LH, PA and LJ are employees and shareholders of AstraZeneca. TH has stock or stock options with AstraZeneca, GW Pharma and Jazz Pharma. FG has received payment for lectures, presentations, speaker bureaus, manuscript writing or educational events from Eli Lilly and Novo Nordisk. LR has participated in advisory panels for Novo Nordisk and acted as a consultant for and is a member of the Association of Statutory Health Insurance Physicians. HH is a steering committee member and consultant for clinical trials sponsored by AstraZeneca, has received research grants from AstraZeneca for the present manuscript, is an advisor for AbbVie, Bayer, Boehringer Ingelheim, Chinook, CSL Behring, Dimerix, Eli Lilly, Janssen, Gilead, Merck, MundiPharma, Mitsubishi Tanabe, NovoNordisk, Travere Pharmaceuticals, and has received grants from AbbVie, Boehringer Ingelheim, Janssen, and NovoNordisk. RM has received research support from AstraZeneca, royalties or licenses from Elsevier, lecture fees from Sanofi Aventis and NovoNordisk, has participated on the Advisory Board for Sanofi Aventis, is a non‐executive member of NHS Tayside Health Board, and a panel member of MRC Population and Systems Medicine Board. HS, BW and MPo declare that they have no competing interests. Editorial support was provided by Oxford PharmaGenesis, Oxford, UK, and was funded by AstraZeneca.

© 2022 AstraZeneca. Diabetes, Obesity and Metabolism published by John Wiley & Sons Ltd.

Figures

FIGURE 1
FIGURE 1
Study design. Abbreviations: ABPM, ambulatory blood pressure monitoring; CGM, continuous glucose monitoring; n, number of subjects
FIGURE 2
FIGURE 2
Primary and secondary endpoints. (A) Percentage change in MMTT plasma glucose AUC4h from baseline to day 32. (B) 15‐minute mean CGM over time. (C) Proportion of time spent in target interstitial glucose range across 32 days of dosing. (D) Change from baseline to end of dosing in daily insulin dose in participants receiving insulin ≥20 units per day, according to baseline HbA1c (n = 14 in each treatment arm). (E) Percentage change in bodyweight from baseline to day 32 and over 38 days follow‐up. (F) Mean percentage change in UACR over time in subgroup of patients with baseline micro‐ or macroalbuminuria (n = 18). Abbreviations: AUC, area under the curve; CGM, continuous glucose monitoring; HbA1c, glycated haemoglobin; MMTT, mixed‐meal tolerance test; SE, standard error; UACR, urinary albumin‐to‐creatinine ratio

References

    1. Reutens AT. Epidemiology of diabetic kidney disease. Med Clin North Am. 2013;97(1):1‐18.
    1. Gorriz JL, Soler MJ, Navarro‐Gonzalez JF, et al. GLP‐1 receptor agonists and diabetic kidney disease: a call of attention to nephrologists. J Clin Med. 2020;9(4):947.
    1. Kovesdy CP, Furth SL, Zoccali C. Obesity and kidney disease: hidden consequences of the epidemic. Can J Kidney Health Dis. 2017;4:2054358117698669.
    1. Cherney DZI, Repetto E, Wheeler DC, et al. Impact of cardio‐renal‐metabolic comorbidities on cardiovascular outcomes and mortality in type 2 diabetes mellitus. Am J Nephrol. 2020;51(1):74‐82.
    1. Gheith O, Farouk N, Nampoory N, Halim MA, Al‐Otaibi T. Diabetic kidney disease: world wide difference of prevalence and risk factors. J Nephropharmacol. 2016;5(1):49‐56.
    1. Adler AI, Stevens RJ, Manley SE, et al. Development and progression of nephropathy in type 2 diabetes: the United Kingdom prospective diabetes study (UKPDS 64). Kidney Int. 2003;63(1):225‐232.
    1. Ahlqvist E, Storm P, Karajamaki A, et al. Novel subgroups of adult‐onset diabetes and their association with outcomes: a data‐driven cluster analysis of six variables. Lancet Diabetes Endocrinol. 2018;6(5):361‐369.
    1. Musso G, Gambino R, Tabibian JH, et al. Association of non‐alcoholic fatty liver disease with chronic kidney disease: a systematic review and meta‐analysis. PLoS Med. 2014;11(7):e1001680.
    1. Musso G, Cassader M, Cohney S, et al. Fatty liver and chronic kidney disease: novel mechanistic insights and therapeutic opportunities. Diabetes Care. 2016;39(10):1830‐1845.
    1. Byrne CD, Targher G. NAFLD as a driver of chronic kidney disease. J Hepatol. 2020;72(4):785‐801.
    1. Sun DQ, Jin Y, Wang TY, et al. MAFLD and risk of CKD. Metabolism. 2021;115:154433.
    1. Tuttle KR, Bakris GL, Bilous RW, et al. Diabetic kidney disease: a report from an ADA consensus conference. Diabetes Care. 2014;37(10):2864‐2883.
    1. Perkovic V, Jardine MJ, Neal B, et al. Canagliflozin and renal outcomes in type 2 diabetes and nephropathy. N Engl J Med. 2019;380(24):2295‐2306.
    1. Heerspink HJL, Stefansson BV, Correa‐Rotter R, et al. Dapagliflozin in patients with chronic kidney disease. N Engl J Med. 2020;383(15):1436‐1446.
    1. Davies MJ, D'Alessio DA, Fradkin J, et al. Management of hyperglycaemia in type 2 diabetes, 2018. A consensus report by the American Diabetes Association (ADA) and the European Association for the Study of diabetes (EASD). Diabetologia. 2018;61(12):2461‐2498.
    1. Mann JFE, Orsted DD, Buse JB. Liraglutide and renal outcomes in type 2 diabetes. N Engl J Med. 2017;377(22):2197‐2198.
    1. Marso SP, Bain SC, Consoli A, et al. Semaglutide and cardiovascular outcomes in patients with type 2 diabetes. N Engl J Med. 2016;375(19):1834‐1844.
    1. Armstrong MJ, Hull D, Guo K, et al. Glucagon‐like peptide 1 decreases lipotoxicity in non‐alcoholic steatohepatitis. J Hepatol. 2016;64(2):399‐408.
    1. Kawanami D, Takashi Y. GLP‐1 receptor agonists in diabetic kidney disease: from clinical outcomes to mechanisms. Front Pharmacol. 2020;11:967.
    1. Henderson SJ, Konkar A, Hornigold DC, et al. Robust anti‐obesity and metabolic effects of a dual GLP‐1/glucagon receptor peptide agonist in rodents and non‐human primates. Diabetes Obes Metab. 2016;18(12):1176‐1190.
    1. Ambery P, Parker VE, Stumvoll M, et al. 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. Lancet. 2018;391(10140):2607‐2618.
    1. Boland ML, Laker RC, Mather K, et al. Resolution of NASH and hepatic fibrosis by the GLP‐1R/GcgR dual‐agonist Cotadutide via modulating mitochondrial function and lipogenesis. Nat Metab. 2020;2(5):413‐431.
    1. Nahra R, Wang T, Gadde KM, et al. Effects of Cotadutide on metabolic and hepatic parameters in adults with overweight or obesity and type 2 diabetes: a 54‐week randomized phase 2b study. Diabetes Care. 2021;44(6):1433‐1442.
    1. Parker V, Robertson D, Hansen L, et al. Cotadutide (MEDI0382), a dual receptor agonist with balanced glucagon‐like peptide‐1 and glucagon activity, modulates hepatic glycogen stores. Presented at: European Association for the Study of Diabetes 55th Annual Meeting; September 18, 2019; Barcelona, Spain.
    1. Bankir L, Bouby N, Blondeau B, Crambert G. Glucagon actions on the kidney revisited: possible role in potassium homeostasis. Am J Physiol Renal Physiol. 2016;311(2):F469‐F486.
    1. Nauck MA, Quast DR, Wefers J, Meier JJ. GLP‐1 receptor agonists in the treatment of type 2 diabetes ‐ state‐of‐the‐art. Mol Metab. 2021;46:101102.
    1. Kolterman OG, Kim DD, Shen L, et al. Pharmacokinetics, pharmacodynamics, and safety of exenatide in patients with type 2 diabetes mellitus. Am J Health Syst Pharm. 2005;62(2):173‐181.
    1. Parker VER, Robertson D, Wang T, et al. Efficacy, safety, and mechanistic insights of cotadutide, a dual receptor glucagon‐like peptide‐1 and glucagon agonist. J Clin Endocrinol Metab. 2020;105(3):803‐820.
    1. Mann JFE, Fonseca VA, Poulter NR, et al. Safety of liraglutide in type 2 diabetes and chronic kidney disease. Clin J Am Soc Nephrol. 2020;15(4):465‐473.
    1. Sorli C, Harashima SI, Tsoukas GM, et al. Efficacy and safety of once‐weekly semaglutide monotherapy versus placebo in patients with type 2 diabetes (SUSTAIN 1): a double‐blind, randomised, placebo‐controlled, parallel‐group, multinational, multicentre phase 3a trial. Lancet Diabetes Endocrinol. 2017;5(4):251‐260.
    1. Persson F, Bain SC, Mosenzon O, et al. Changes in albuminuria predict cardiovascular and renal outcomes in type 2 diabetes: a post hoc analysis of the LEADER trial. Diabetes Care. 2021;44(4):1020‐1026.
    1. Mann JFE, Hansen T, Idorn T, et al. Effects of once‐weekly subcutaneous semaglutide on kidney function and safety in patients with type 2 diabetes: a post‐hoc analysis of the SUSTAIN 1‐7 randomised controlled trials. Lancet Diabetes Endocrinol. 2020;8(11):880‐893.
    1. Elnaem MH, Mansour NO, Nahas AF, Baraka MA, Elkalmi R, Cheema E. Renal outcomes associated with the use of non‐insulin antidiabetic pharmacotherapy: a review of current evidence and recommendations. Int J Gen Med. 2020;13:1395‐1409.
    1. van Baar MJB, van der Aart AB, Hoogenberg K, Joles JA, Heerspink HJL, van Raalte DH. The incretin pathway as a therapeutic target in diabetic kidney disease: a clinical focus on GLP‐1 receptor agonists. Ther Adv Endocrinol Metab. 2019;10:2042018819865398.
    1. Jensen EP, Poulsen SS, Kissow H, et al. Activation of GLP‐1 receptors on vascular smooth muscle cells reduces the autoregulatory response in afferent arterioles and increases renal blood flow. Am J Physiol Renal Physiol. 2015;308(8):F867‐F877.
    1. Schlatter P, Beglinger C, Drewe J, Gutmann H. Glucagon‐like peptide 1 receptor expression in primary porcine proximal tubular cells. Regul Pept. 2007;141(1‐3):120‐128.
    1. Wilson PC, Wu H, Kirita Y, et al. The single‐cell transcriptomic landscape of early human diabetic nephropathy. Proc Natl Acad Sci USA. 2019;116(39):19619‐19625.
    1. Muskiet MH, Tonneijck L, Smits MM, et al. Acute renal haemodynamic effects of glucagon‐like peptide‐1 receptor agonist exenatide in healthy overweight men. Diabetes Obes Metab. 2016;18(2):178‐185.
    1. Jensen EP, Moller S, Hviid AV, et al. GLP‐1‐induced renal vasodilation in rodents depends exclusively on the known GLP‐1 receptor and is lost in prehypertensive rats. Am J Physiol Renal Physiol. 2020;318(6):F1409‐F1417.
    1. Selley E, Kun S, Szijarto IA, Kertesz M, Wittmann I, Molnar GA. Vasodilator effect of glucagon: receptorial crosstalk among glucagon, GLP‐1, and receptor for glucagon and GLP‐1. Horm Metab Res. 2016;48(7):476‐483.
    1. Rosic M, Pantovic S, Rosic G, et al. Glucagon effects on ischemic vasodilatation in the isolated rat heart. J Biomed Biotechnol. 2010;2010:231832.
    1. Friedlander G, Blanchet‐Benque F, Nitenberg A, Laborie C, Assan R, Amiel C. Glucagon secretion is essential for aminoacid‐induced hyperfiltration in man. Nephrol Dial Transplant. 1990;5(2):110‐117.
    1. Farah AE. Glucagon and the circulation. Pharmacol Rev. 1983;35(3):181‐217.

Source: PubMed

3
Abonneren