1,459
Views
11
CrossRef citations to date
0
Altmetric
Editorial

Glucagon-like peptide-1 receptor agonist therapeutics for total diabetes management: assessment of composite end-points

, , , &
Pages 1267-1270 | Accepted 17 Apr 2015, Published online: 20 May 2015

Abstract

Assessment of the benefits of anti-diabetic drugs for type 2 diabetes requires analysis of composite end-points, taking HbA1c, bodyweight, hypoglycemia and other metabolic parameters into consideration; continuous, optimal glycemic control as well as bodyweight, blood pressure and lipid levels are critical to prevent micro- and macro-vascular complications. Glucagon-like peptide-1 receptor agonists (GLP-1RAs) are now established as an important total treatment strategy for type 2 diabetes, exerting glucose-lowering effects with little hypoglycemia risk and also ameliorating bodyweight, blood pressure and lipid levels, which are therapeutic targets for prevention of complications of the disease. The available data strongly suggest only beneficial effects of GLP-1RAs; however, long-term evaluation of the relevant composite end-points including health-related quality of life and cost-effectiveness remain to be investigated in adequately powered, prospective, controlled clinical trials. In the meantime, healthcare professionals need to be scrupulously attentive for potential, rare adverse events in patients using GLP-1RAs.

The drastic increase of type 2 diabetes (T2DM) is one of the most serious health-related problems today. The number of patients with diabetes, estimated to be 385 million in 2014, is expected to be 592 million by 2035; the global health expenditure for diabetes is now at least US$612 billion, approximately 11% of total medical spending on adults worldwideCitation1. T2DM is a progressive metabolic disorder characterized by deterioration of pancreatic β-cell function and insulin resistance in the liver and peripheral tissues. These factors combine to result in overproduction of hepatic blood glucose, elevation of fasting plasma glucose levels and insufficient glucose uptake by muscle cells that elevate postprandial plasma glucose following mealsCitation2. In addition to that of β-cells, liver and muscle, dysfunction of pancreatic α-cells (hyperglucagonemia), fat cells (accelerated lipolysis) and kidney (increased glucose reabsorption) all can play a role in the development and progression of T2DMCitation1. Because chronic elevation of plasma glucose levels eventually results in microvascular complications (neuropathy, retinopathy, nephropathy, etc.) and macrovascular complications (ischemic heart disease, cerebrovascular disease, peripheral arterial disease etc.)Citation3, therapies to achieve optimal glycemic control together with optimal management of bodyweight, blood pressure and lipid levels have been sought for decades. Evaluation of the benefits of each of the new anti-diabetic drugs on the complications of the disease requires long-term analysis of composite end-points, taking HbA1c, bodyweight, hypoglycemia and other metabolic parameters into considerationCitation4.

Glucagon-like peptide-1 (GLP-1) receptor agonists (RAs) are established as an important total treatment strategy for T2DM, with an efficacy and safety profile distinct from that of other anti-diabetic drugsCitation5. Many studies have shown that GLP-1 RAs exert glucose-lowering effects in Asian T2DM characterized by impaired insulin secretionCitation6–8, but GLP-1 RAs can also ameliorate glycemic control in obese as well as non-obese T2DMCitation5. GLP-1 RAs exert glucose-lowering effects through multiple mechanisms, including enhancement of glucose-induced insulin secretion, suppression of glucagon secretion and delayed gastric emptyingCitation9,Citation10. Since GLP-1 stimulates insulin secretion glucose-dependently, GLP-1 RAs administered alone are thought to have little hypoglycemia riskCitation11. GLP-1 RAs also promote bodyweight reduction, possibly by suppression of appetite through activation of GLP-1 receptors in the brainCitation12,Citation13. GLP-1 RAs reduce blood pressure by enhancing urinary sodium excretion and inducing vasodilation, thereby lowering blood pressure. GLP-1 RAs ameliorate postprandial hyperlipidemia by suppressing production of apolipoprotein B-48 and triglyceride absorptionCitation9,Citation10. GLP-1 RAs also can suppress chronic inflammation by reducing production of inflammatory cytokines and macrophage infiltrationCitation9,Citation10. Thus, GLP-1 RAs exert beneficial effects not only on glycemia but also on bodyweight, blood pressure, lipid levels and chronic inflammation, all of which are therapeutic targets for prevention of complications of the disease, and for these reasons the drugs are often considered an attractive alternative to other treatment regimens.

Ross recently reported a summary of original articles and meta-analyses assessing the efficacy of liraglutide and exenatide, the only available GLP-1 RAs in Canada at the time, using composite end-points including HbA1c <7%, no weight gain and no hypoglycemic episodesCitation14. He found that liraglutide and exenatide showed better achievement of these end-points compared to placebo and oral anti-diabetic drugs, which was expected in accord with the therapeutic characteristics of GLP-1 RAs described above. Ross also noted that the benefits of GLP-1 RA were more prominent in patients with shorter duration T2DM in one report on liraglutide. This is consistent with several reports demonstrating that the HbA1c-lowering effects of liraglutide depend on residual β-cell function such as may be found in patients with shorter duration of the diseaseCitation15–17. Despite the possible amelioration of diabetic complications independently of glycemic control, there are important issues to be considered, including patient health-related quality of life (HRQOL) and cost-effectiveness; GLP-1 RAs involve injections, which often lower HRQOL, and there are considerably higher medical costs in comparison with oral anti-diabetic drugs. Analysis of HRQOL and the medical costs associated with GLP-1 RAs is currently limitedCitation18,Citation19, and delineation of the effects of the various GLP-1 RAs on diabetic complications requires clinical trials with adequately powered, prospective, controlled and relevant diabetic complication end-points.

GLP-1 RAs are generally grouped into short- and long-acting agents depending on their half-lives, which alters the mechanism by which they exert their glucose lowering effectsCitation5,Citation20. While long-acting agents such as liraglutide mainly affect insulin and glucagon secretions to improve both pre- and post-prandial glucose excursions, short-acting agents primarily affect gastric emptying to strongly suppress elevation of post-prandial glucose levels immediately after injection. These very different modes of action suggest that short- and long-acting GLP-1 RAs are distinct anti-diabetic drug categories. Previous studies comparing the achievement of composite end-points found the long-acting liraglutide to be somewhat superior to the short-acting exenatideCitation21,Citation22, although thorough comparison of long- and short-acting awaits more detailed investigation. Accumulating results on short-acting (exenatideCitation23,Citation24 and lixisenatideCitation25–27) and long-acting (liraglutideCitation18,Citation19,Citation28,Citation29, exenatide long-acting releaseCitation30,Citation31, albiglutide, dulaglutide and semaglutide) are becoming available, allowing comparison of achievement of composite end-points by these agents in future. Moreover, long-acting GLP-1 RAs seem to differ in bodyweight reduction according to their molecular weight and permeability across the brain–blood barrier, possibly by affecting their accessibility to GLP-1 receptors in the brainCitation32,Citation33. Although liraglutide therapy involves once daily injection, the other long-acting GLP-1 RAs require injection only once weekly, which might both improve adherence and minimize the reduction in HRQOL. Most importantly, the differences in achievement of composite end-points by the various GLP-1 RAs available need to be considered carefully when selecting which GLP-1 RAs to use for each patient.

Combination of GLP-1 RAs and basal insulin have been recently gaining much attention due to their complementary actionCitation34,Citation35. Add-on of GLP-1 RAs to insulin generally reduces bodyweight gain without increasing hypoglycemic risk, when compared to an increase of insulin dosage. Recent meta-analysis reveals that GLP-1 RA and basal insulin combinations have HbA1c-lowering effects similar to that of basal–bolus insulin therapies, but suppress bodyweight gain and do not increase hypoglycemiaCitation35, and could improve HRQOL by reducing injection frequency. Insulin co-administration could also benefit patients with severely reduced β-cell function who are generally unable to achieve optimal glycemic control by GLP-1 RAs aloneCitation15,Citation36. It remains to be determined how much remaining β-cell function is actually required for GLP-1 RAs in combination with insulin injection to exert optimal glycemic control.

It is now recognized that GLP-1 RAs can ameliorate several pathophysiological deficits of T2DM and can also reduce metabolic risk factors such as bodyweight, blood pressure and dyslipidemia, all of which are associated with the disease. This has established GLP-1 RA as an important treatment strategy for T2DM. However, long-term safety issues need to be carefully monitored. Scrutiny of the data related to acute pancreatitis and pancreatic cancers has not resulted in any recommendation to stop use of incretin therapies in management of T2DMCitation37. Pancreatic cancers take years before becoming metastaticCitation38, but recent investigation of GLP-1 receptor expression profiles in human tissues does not support a causal relationship of GLP-1 receptor activation with the pathogenesis of any of these cancersCitation39. However, careful observation for possible onset of malignancies remains essential. Recently, Koehler et al. reported that GLP-1 receptor activation in a mouse model of colorectal cancer facilitated onset of tumorigenesisCitation40, suggesting possible colorectal cancer risks with use of GLP-1 RAs in special populations, such as those with familial adenomatous polyposis. While there are many expectations of possible beneficial pleiotropic effects of GLP-1 RA therapy such as prevention of dementiaCitation41,Citation42, healthcare professionals need to be scrupulously careful of potential adverse events until results of ongoing large-scale randomized clinical trials clarify these dire possibilities.

Transparency

Declaration of funding

This editorial was written independently and received no professional help. D.Y. has received Grant-in-Aid for Young Scientists (B) from the Japan Society for Science Promotion and Grants for Young Researchers from the Japan Association for Diabetes Education and Care; Y.S. has received grants from the Japan Vascular Disease Research Foundation.

Declaration of financial/other relationships

D.Y. has disclosed that he has received consulting and/or speaker fees from Eli Lilly, MSD, Sanofi, Novo Nordisk, Boehringer Ingelheim, Takeda and Taisho pharmaceutical; he has also received clinical commissioned/joint research grants from Nippon Boehringer Ingelheim, Eli Lilly, and MSD. T.K. has disclosed that he has received consulting and/or speaker fees from Astellas, Boehringer Ingelheim, Sanofi, Novo Nordisk, MSD, Takeda, Kowa, Tanabe Mitsubishi, Kaken Pharm, AstraZeneca, Daiichi-Sankyo, and Kyowa Kirin; he has also received clinical commissioned/joint research grants from Boehringer Ingelheim, Novo Nordisk, Merck, Sharp and Dohme, Takeda, Ono Pharm, Eli Lilly, Teijin, and Sanofi. Y.S. has disclosed that he has received consulting and/or speaker fees from Eli Lilly, Sanofi, Novo Nordisk, Glaxo-Smith-Kline, Taisho Pharmaceutical, Astellas Pharma, BD, Boehringer Ingelheim, Johnson & Johnson and Takeda; he has also received clinical commissioned/joint research grants from Boehringer Ingelheim, Eli Lilly and MSD. H.K. and R.U. have disclosed that they have no significant relationships with or financial interests in any commercial companies related to this study or article.

Acknowledgments

The authors thank Michiko Yamane of Kansai Electric Power Hospital for secretarial assistance.

References

  • Diabetes Atlas 2014 Update. Brussels: International Diabetes Federation, 2014
  • Defronzo RA. Banting Lecture. From the triumvirate to the ominous octet: a new paradigm for the treatment of type 2 diabetes mellitus. Diabetes 2009;58:773-95
  • Forbes JM, Cooper ME. Mechanisms of diabetic complications. Physiol Rev 2013;93:137-88
  • Einarson TR, Garg M, Kaur V, Hemels ME. Composite endpoints in trials of type-2 diabetes. Diabetes Obes Metab 2014;16:492-9
  • Yabe D, Seino Y. Defining the role of GLP-1 receptor agonists for individualized treatment of type 2 diabetes. Expet Rev Endocrinol Metabol 2014;9:659-70
  • Yabe D, Seino Y, Fukushima K, Seino S. β cell dysfunction versus insulin resistance in the pathogenesis of type 2 diabetes in East Asians. Curr Diabetes Rep in press (DOI: 10.1007/s11892-015-0602-9)
  • Yabe D, Kuwata H, Iwasaki M, Seino Y. Why are incretin-based therapies more efficient in East Asians? Perspectives from the pathophysiology of type 2 diabetes and East Asian dietary habits. Eur Med J in press
  • Kim YG, Hahn S, Oh TJ, et al. Differences in the HbA1c-lowering efficacy of glucagon-like peptide-1 analogues between Asians and non-Asians: a systematic review and meta-analysis. Diabetes Obes Metab 2014;16:900-9
  • Drucker DJ. Incretin action in the pancreas: potential promise, possible perils, and pathological pitfalls. Diabetes 2013;62:3316-23
  • Seino Y, Yabe D. Glucose-dependent insulinotropic polypeptide and glucagon-like peptide-1: incretin actions beyond the pancreas. J Diabetes Investig 2013;4:108-30
  • Yabe D, Seino Y. Two incretin hormones GLP-1 and GIP: comparison of their actions in insulin secretion and beta cell preservation. Progr Biophys Mol Biol 2011;107:248-56
  • Secher A, Jelsing J, Baquero AF, et al. The arcuate nucleus mediates GLP-1 receptor agonist liraglutide-dependent weight loss. J Clin Invest 2014;124:4473-88
  • Sisley S, Gutierrez-Aguilar R, Scott M, et al. Neuronal GLP1R mediates liraglutide's anorectic but not glucose-lowering effect. J Clin Invest 2014;124:2456-63
  • Ross SA. A multiplicity of targets: evaluating composite endpoint studies of the GLP-1 receptor agonists in type 2 diabetes. Curr Med Res Opin 2015;31:125-35
  • Usui R, Yabe D, Kuwata H, et al. Retrospective analysis of safety and efficacy of insulin-to-liraglutide switch in Japanese type 2 diabetes: a caution against inappropriate use in patients with reduced beta cell function. J Diabet Invest 2013;4:585-94
  • Meier JJ, Vilsbøll T, Donsmark M, et al. Greater glycaemic control is achieved with the once-daily human GLP-1 analogue liraglutide vs comparators across the continuum of estimated beta cell mass. Diabetologia 2011;54:S1-S566 (Abstract 896)
  • Kondo Y, Satoh S, Nagakura J, et al. Defining criteria for the introduction of liraglutide using the glucagon stimulation test in patients with type 2 diabetes. J Diabetes Investig 2013;4:571-5
  • King AB, Montanya E, Pratley RE, et al. Liraglutide achieves A1C targets more often than sitagliptin or exenatide when added to metformin in patients with type 2 diabetes and a baseline A1C <8.0%. Endocr Pract 2013;19:64-72
  • Langer J, Hunt B, Valentine WJ. Evaluating the short-term cost-effectiveness of liraglutide versus sitagliptin in patients with type 2 diabetes failing metformin monotherapy in the United States. J Manag Care Pharm 2013;19:237-46
  • Meier JJ. GLP-1 receptor agonists for individualized treatment of type 2 diabetes mellitus. Nat Rev Endocrinol 2012;8:728-42
  • Zinman B, Schmidt WE, Moses A, et al. Achieving a clinically relevant composite outcome of an HbA1c of <7% without weight gain or hypoglycaemia in type 2 diabetes: a meta-analysis of the liraglutide clinical trial programme. Diabetes Obes Metab 2012;14:77-82
  • Davies MJ, Kela R, Khunti K. Liraglutide – overview of the preclinical and clinical data and its role in the treatment of type 2 diabetes. Diabetes Obes Metab 2011;13:207-20
  • Gallwitz B, Bohmer M, Segiet T, et al. Exenatide twice daily versus premixed insulin aspart 70/30 in metformin-treated patients with type 2 diabetes: a randomized 26-week study on glycemic control and hypoglycemia. Diabetes Care 2011;34:604-6
  • Davies MJ, Donnelly R, Barnett AH, et al. Exenatide compared with long-acting insulin to achieve glycaemic control with minimal weight gain in patients with type 2 diabetes: results of the Helping Evaluate Exenatide in patients with diabetes compared with Long-Acting insulin (HEELA) study. Diabetes Obes Metab 2009;11:1153-62
  • Raccah D, Gourdy P, Sagnard L, Ceriello A. Lixisenatide as add-on to oral anti-diabetic therapy: an effective treatment for glycaemic control with body weight benefits in type 2 diabetes. Diabetes Metab Res Rev 2014;30:742-8
  • Hanefeld M, Berria R, Lin J, et al. Lixisenatide treatment for older patients with type 2 diabetes mellitus uncontrolled on oral antidiabetics: meta-analysis of five randomized controlled trials. Adv Ther 2014;31:861-72
  • Charbonnel B, Bertolini M, Tinahones FJ, et al. Lixisenatide plus basal insulin in patients with type 2 diabetes mellitus: a meta-analysis. J Diabetes Complications 2014;28:880-6
  • Brady EM, Davies MJ, Gray LJ, et al. A randomized controlled trial comparing the GLP-1 receptor agonist liraglutide to a sulphonylurea as add on to metformin in patients with established type 2 diabetes during Ramadan: the Treat 4 Ramadan Trial. Diabetes Obes Metab 2014;16:527-36
  • Li CJ, Li J, Zhang QM, et al. Efficacy and safety comparison between liraglutide as add-on therapy to insulin and insulin dose-increase in Chinese subjects with poorly controlled type 2 diabetes and abdominal obesity. Cardiovasc Diabetol 2012;11:142-9
  • Bergenstal RM, Li Y, Porter TK, et al. Exenatide once weekly improved glycaemic control, cardiometabolic risk factors and a composite index of an HbA1c <7%, without weight gain or hypoglycaemia, over 52 weeks. Diabetes Obes Metab 2013;15:264-71
  • Grimm M, Li Y, Brunell SC, Blase E. Exenatide once weekly versus daily basal insulin as add-on treatment to metformin with or without a sulfonylurea: a retrospective pooled analysis in patients with poor glycemic control. Postgrad Med 2013;125:101-8
  • Pratley RE, Nauck MA, Barnett AH, et al. Once-weekly albiglutide versus once-daily liraglutide in patients with type 2 diabetes inadequately controlled on oral drugs (HARMONY 7): a randomised, open-label, multicentre, non-inferiority phase 3 study. Lancet Diabetes Endocrinol 2014;2:289-97
  • Dungan KM, Povedano ST, Forst T, et al. Once-weekly dulaglutide versus once-daily liraglutide in metformin-treated patients with type 2 diabetes (AWARD-6): a randomised, open-label, phase 3, non-inferiority trial. Lancet 2014;384:1349-57
  • Balena R, Hensley IE, Miller S, Barnett AH. Combination therapy with GLP-1 receptor agonists and basal insulin: a systematic review of the literature. Diabetes Obes Metab 2013;15:485-502
  • Eng C, Kramer CK, Zinman B, Retnakaran R. Glucagon-like peptide-1 receptor agonist and basal insulin combination treatment for the management of type 2 diabetes: a systematic review and meta-analysis. Lancet 2014;384:2228-34
  • Yabe D, Ambos A, Cariou B, et al. Adjunctive lixisenatide treatment improves glycemic control in patients with T2DM irrespective of b-cell function. Diabetes 2014;64(Suppl 1):A253
  • Egan AG, Blind E, Dunder K, et al. Pancreatic safety of incretin-based drugs – FDA and EMA assessment. N Engl J Med 2014;370:794-7
  • Yachida S, Jones S, Bozic I, et al. Distant metastasis occurs late during the genetic evolution of pancreatic cancer. Nature 2010;467:1114-17
  • Waser B, Blank A, Karamitopoulou E, et al. Glucagon-like-peptide-1 receptor expression in normal and diseased human thyroid and pancreas. Mod Pathol 2014;28:391-402
  • Koehler JA, Baggio LL, Yusta B, et al. GLP-1R agonists promote normal and neoplastic intestinal growth through mechanisms requiring Fgf7. Cell Metab 2015;21:379-91
  • McClean PL, Holscher C. Lixisenatide, a drug developed to treat type 2 diabetes, shows neuroprotective effects in a mouse model of Alzheimer's disease. Neuropharmacology 2014;86:241-58
  • Aviles-Olmos I, Dickson J, Kefalopoulou Z, et al. Exenatide and the treatment of patients with Parkinson's disease. J Clin Invest 2013;123:2730-6

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.