REVIEW ARTICLE

review article

Diabetes, Obesity and Metabolism 17: 720–725, 2015. © 2015 John Wiley & Sons Ltd

Inhibiting or antagonizing glucagon: making progress in diabetes care P. J. Lefèbvre1 , N. Paquot1 & A. J. Scheen1,2 1 Division of Diabetes, Nutrition and Metabolic Disorders, Department of Medicine, University of Liège, Liège, Belgium 2 Division of Clinical Pharmacology, Department of Medicine, University of Liège, Liège, Belgium

Absolute or relative hyperglucagonaemia has been recognized for years in all experimental or clinical forms of diabetes. It has been suggested that excess secretion of glucagon by the islet 𝛼 cells is a direct consequence of intra-islet insulin secretory defects. Recent studies have shown that knockout of the glucagon receptor or administration of a monoclonal specific glucagon receptor antibody make insulin-deficient type 1 diabetic rodents thrive without insulin. These observations suggest that glucagon plays an essential role in the pathophysiology of diabetes and that targeting the 𝛼 cell and glucagon are innovative approaches in the management of diabetes. Despite active research and identification of promising compounds, no one selective glucagon antagonist is presently used in the treatment of diabetes. Interestingly, besides insulin, several drugs used today in the management of diabetes appear to exert their effects, in part, by inhibiting glucagon secretion (glucagon-like peptide-1 receptor agonists, dipeptidyl peptidase-4 inhibitors, 𝛼-glucosidase inhibitors and, possibly, sulphonylureas) or glucagon action (metformin). The potential risks associated with total glucagon suppression include 𝛼-cell hyperplasia, increased mass of the pancreas, increased susceptibility to hepatosteatosis and hepatocellular injury and increased risk of hypoglycaemia, and these should be considered in the search and development of new compounds reducing glucagon receptor signalling. More than 40 years after its initial description, hyperglucagonaemia in diabetes can no longer be ignored or minimized, and its correction represents an attractive way to improve diabetes management. Keywords: diabetes, glucagon Date submitted 24 February 2015; date of first decision 28 March 2015; date of final acceptance 27 April 2015

Introduction It has been our privilege to work on glucagon for more than 50 years [1] and to suggest 35 years ago that a search for selective glucagon inhibitors could open a new avenue in diabetes management [2]. In the present concise review, we will recall that hyperglucagonaemia is a constant feature in all forms of diabetes and attempt to explain why. We will briefly review the concept of Unger and Orci [3] that diabetes should be seen as a paracrinopathy of the islets of Langerhans, and recall recent animal studies that have shown that knockout of the glucagon receptor makes insulin-deficient type 1 diabetic rodents thrive without insulin. The effect on glucagon of current antidiabetic drugs will be reviewed, while the search for selective glucagon antagonists will be briefly analysed. Finally, the potential risks associated with total glucagon suppression will be considered. A comprehensive bibliography on glucagon up to 1996 can be found in the three volumes of the Handbook of Experimental Pharmacology that we edited [4,5] and in the proceedings of a more recently held conference published in this journal [6].

Hyperglucagonaemia is a Constant Feature of Diabetes It has been established for years that plasma glucagon levels are increased in all experimental and clinical forms of Correspondence to: Dr Pierre Lefebvre, Department of Medicine, CHU Sart Tilman, B-4000 Liège 1, Belgium. E-mail: [email protected]

diabetes [7]. This disturbance undoubtedly contributes to the hyperglycaemia of the disease and the excessive ketogenesis of diabetic coma [8]. Numerous studies have shown that failure of glucagon suppression contributes to postprandial hyperglycaemia in type 1 [9,10] and type 2 diabetes [11,12]. Impaired glucagon suppression concurs with impaired insulin release to the excessive blood glucose levels in early type 1 diabetes [13], in impaired glucose tolerance [14,15] and in ketosis-prone atypical diabetes [16,17]. Morphological studies have now firmly established that the main abnormality in the islet cell population of human diabetes is a decrease in the 𝛽-cell mass with a relative expansion of the 𝛼-cell mass [18].

Why Hyperglucagonaemia in Diabetes? The mechanisms leading to hyperglucagonaemia in diabetes have long been discussed and include: a direct consequence of intra-islet insulin secretory defects, a resistance of the 𝛼 cell to insulin or a desensitization of the 𝛼 cell to glucose [7]. Unger and Orci have recently proposed that diabetes should be considered as a ‘paracrinopathy’ of the islets of Langerhans [3]. This proposal is based on the concept that the very high concentrations of insulin normally reached inside the stimulated islets exert, directly or by proxy, a major inhibitory effect on glucagon secretion from the neighbouring 𝛽 cells, as previously suggested by us and others [19,20]. Disruption of this mechanism is proposed as a key factor in the pathogenesis of diabetes [21].The concept is supported by recent data on

review article

DIABETES, OBESITY AND METABOLISM

A

C

E Frequency (%)

D

B

60 40 20 0 beta wrapping alpha

apha-beta

alpha wrapping beta

Figure 1. Human islet cells were isolated and cultured for 24 h. After a double immunofluorescence for insulin (red) and glucagon (green), islet cells were analysed by confocal microscopy. (A–C) Images showing a cell pair composed of one 𝛼 cell surrounded by one 𝛽 cell, the merged image is shown in (C). The cell pair shown here is representative of cell pairs observed in different human cell preparations from at least 10 different pancreata. (D) One series of consecutive merged images of a cell pair composed of one 𝛼 cell (green) surrounded by a 𝛽 cell (red). Scale bars, 10 μm. All heterologous contacts between 𝛼 and 𝛽 cells were coded according to their type of association: a 𝛽 cell wrapping an 𝛼 cell (beta wrapping alpha), neutral apposition between 𝛼 and 𝛽 cells (alpha-beta) and an 𝛼 cell wrapping a 𝛽 cell (alpha wrapping beta). Results are shown in E as relative frequencies and columns are means ± standard error of the mean (s.e.m.) of five islet cell preparations from five different pancreata. From all heterogenous contacts between 𝛼 and 𝛽 cells, the percentage of 𝛼 cells whose profile was round and perimeter almost completely wrapped by a 𝛽 cell, as in (D), was 38 ± 8 (mean ± s.e.m. of three experiments). Figure from Diabetes 2010; 59: 1202–1210 and reprinted with permission from the American Diabetes Association. Copyright 2010 American Diabetes Association.

the microanatomy of the islets of Langerhans in man [22] and by new data on the intimate relationships between human 𝛽 and 𝛼 cells as shown in Figure 1, reproduced from Bosco et al. [22]. When the association between 𝛼 and 𝛽 cells is assessed in cultured isolated human islet cells, many 𝛼 cells are wrapped by 𝛽 cells and almost never the contrary. This strongly suggests that, in some way, human 𝛼 cells are under the control of 𝛽 cells. As discussed by the authors, this observation reveals a unique plasticity of the 𝛽 cells, which are able to spread around 𝛼 cells, suggesting that this characteristic is intrinsic to 𝛽 cells and not dictated by some islet coercions, such as extracellular matrix or islet vasculature. The molecular mechanism of this extraordinary so-called embracing of 𝛼 cells by 𝛽 cells remains to be elucidated, but the phenomenon in itself must be considered when discussing the paracrinology of the islets and the attractive suggestion that diabetes must be seen as a paracrinopathy in which glucagon cannot be ignored [3,23]. In type 1 diabetes, 𝛼 cells lack constant action of high insulin levels from juxtaposed 𝛽 cells. Replacement with exogenous insulin subcutaneously injected does not approach the paracrine levels of insulin, except with high doses that over-insulinize the peripheral insulin targets, thereby promoting glycaemic volatility [3,21]. In type 2 diabetes, the 𝛼-cell dysfunction may result from the failure of the juxtaposed 𝛽 cells to secrete the first phase of insulin or from the loss of the intra-islet pulsatile secretion of insulin. Observations made in experimental diabetes in minipigs [24], and recently confirmed in human type 2 diabetes [25], are in support of the second mechanism.

Volume 17 No. 8 August 2015

Essential Role for Glucagon in the Pathophysiology of Diabetes? Several recent studies on experimental animal models of diabetes have suggested that glucagon plays an essential role in the pathophysiology of diabetes. Yu et al. [26] have shown that inhibition of glucagon secretion markedly improves experimental diabetes in rodents and that knockout of the glucagon receptor makes insulin-dependent type 1 diabetic rodents thrive without insulin [27]. Interestingly, a recent study found a role for the glucagon-like peptide-1 (GLP-1) receptor in controlling endogenous glucose production in the combined absence of glucagon and insulin in rodents [28]. To determine unambiguously if suppression of glucagon action eliminates manifestations of diabetes, Lee et al. [29] expressed glucagon receptors in livers of glucagon receptor (GcgR)-null (−/−) mice before and after 𝛽-cell destruction by high doses streptozotocin. Wild-type mice developed fatal diabetic ketoacidosis after streptozotocin, whereas GcgR−/− mice remained clinically normal without hyperglycaemia, impaired glucose tolerance or hepatic glycogen depletion. Administration of an adenovirus containing the GcgR cDNA transiently restored hepatic GcgR mRNA expression, while phospho-cAMP response element binding protein (P-CREB) and phosphoenol-pyruvate carboxykinase, markers of glucagon action, rose dramatically and severe hyperglycaemia developed. When GcgR mRNA spontaneously disappeared 7 days later, P-CREB declined and hyperglycaemia disappeared. In this experimental setting, the

doi:10.1111/dom.12480 721

review article metabolic manifestations of diabetes cannot occur without glucagon action, and once present, disappear promptly when glucagon action is abolished. Supporting these data, Wang et al. [30] recently reported that administration of a specific monoclonal antiglucagon receptor antibody completely normalized blood glucose and glycated haemoglobin levels without insulin in murine models of type 1 diabetes. All these experimental observations strongly suggest that targeting the 𝛼 cell and glucagon are innovative approaches in diabetes management.

Current Antidiabetic Agents and Glucagon Secretion or Action Old and new antidiabetic agents may exert part of their therapeutic effects via inhibition of glucagon secretion or inhibition of glucagon action.

Insulin The classic inhibitory effect of insulin on glucagon secretion observed in vitro contrasts with the elevated, or relatively elevated, glucagon circulating levels often reported in people with insulin-treated diabetes. In fact, under intensive insulin therapy either by multiple injections or by portable pumps, hyperglucagonaemia is markedly [31] or fully [32] corrected. Perfect substitution of insulin deficiency, including restoration of normal intra-islet pulsatile insulin secretion, would probably represent the best means to normalize the hypersecretion of glucagon [33]. In practice, and as alluded to above, reproduction of high intra-islet insulin concentrations that allow the achievement of sufficient inhibition of glucagon secretion by subcutaneous insulin administration may induce peripheral tissues over-insulinization and excessively increase the risk of hypoglycaemia [3,21].

Sulphonylureas The effects of sulphonylureas on glucagon secretion appear particularly complex. By stimulating intra-islet insulin release, sulphonylureas potentially reduce glucagon secretion, as elegantly analysed by Cooperberg and Cryer [34]. By contrast, in profound insulin deficiency, as in type 1 diabetes lacking C-peptide, sulphonylureas, such as glibenclamide [35] or glimepiride [34,36], stimulate glucagon secretion. Interestingly, in people with type 2 diabetes, with persistent endogenous insulin secretion, oral glibenclamide inhibits the glucagon response to insulin-induced hypoglycaemia [37]. Recent in vitro studies on the effect of sulphonylureas on pancreatic 𝛼 cells have shown the extreme complexity of the mechanisms involved [38]. Most interestingly, studies using islets of Langerhans from donors with type 2 diabetes have shown that low concentrations of tolbutamide (lower than those required to stimulate insulin secretion) restore normal glucose regulation of glucagon secretion (inhibition at high and stimulation at low glucose concentrations) [39]. Clinical studies are needed to evaluate the role of low-dose sulphonylureas precisely targeting glucagon secretion in both type 1 and type 2 diabetes.

722 Lefebvre et al.

DIABETES, OBESITY AND METABOLISM

Biguanides Biguanides, such as metformin [40] or buformin [41], seem to have little or no effect of their own on pancreatic 𝛼-cell function. Interestingly, glucagon signalling in the liver is attenuated by metformin [42].

Glucagon-like Peptide-1 (GLP-1), GLP-1 Receptor Agonists and Dipeptidyl Peptidase-4 Inhibitors As recently reviewed [43], GLP-1 and GLP-1 receptor agonists, such as exenatide, liraglutide, dulaglutide and albiglutide, are now largely used in the treatment of Type 2 diabetes. Regarding GLP-1, Hare et al. [44] elegantly showed that inhibition of glucagon secretion and stimulation of insulin release contribute equally to its glucose-lowering action. Inhibition of glucagon secretion was further demonstrated for exenatide [45,46] and liraglutide [47,48]. This inhibition is most likely explained by a GLP-1 receptor-dependent stimulation of islet somatostatin secretion which, in turn, inhibits glucagon secretion through activation of somatostatin receptor-2 in 𝛼 cells [49–51]. The action of endogenously released GLP-1 is transient because the peptide is rapidly cleared by the kidney and inactivated by cleavage at the N-terminus by dipeptidyl peptidase-4 (DPP-4), a ubiquitous exopeptidase. Numerous DPP-4 inhibitors (vildagliptin, sitagliptin, saxagliptin, linagliptin and alogliptin) have been developed with the aim of prolonging the action of endogenously released GLP-1. It is currently accepted that their efficacy in controlling blood glucose in type 2 diabetes is partly attributable to their inhibitory effect on glucagon secretion [52,53]. Interestingly, despite inhibition of glucagon secretion during hyperglycaemia, gliptins appear not to compromise the glucagon counter-regulatory response to hypoglycaemia [54].

𝛼-Glucosidase Inhibitors The 𝛼-glucosidase inhibitor class of agent includes acarbose, miglitol and voglibose. These cause a significant postprandial increase in active GLP-1 circulating levels, an effect that may contribute to reduced glucagon secretion [55,56].

Thiazolidinediones or Glitazones Thiazolidinediones or glitazones are compounds aimed at reducing insulin resistance in type 2 diabetes, but their use has been limited by cardiovascular side effects and other adverse events [57]. As part of their antidiabetic effect, thiazolidinediones, such as rosiglitazone, have been shown to inhibit glucagon gene transcription in pancreatic 𝛼-cell lines through binding to the nuclear peroxisome proliferator-activated receptor-𝛾 and inhibition of the transcriptional activity of PAX6 that is required for cell-specific activation of the glucagon gene [58]. Clinical studies are needed to assess the importance of this mechanism in man.

Sodium-Glucose Cotransporter Type 2 Inhibitors As recently reviewed [59,60], sodium-glucose cotransporter type 2 inhibitors (canagliflozin, dapagliflozin, empagliflozin and ipragliflozin) are glucose-lowering agents that exert their

Volume 17 No. 8 August 2015

review article

DIABETES, OBESITY AND METABOLISM

therapeutic activity independently of insulin by facilitating glucose excretion through the kidneys. Quite surprisingly, they appear to increase rather than reduce endogenous glucose production [61]. This appears to be driven by increased glucagon secretion [62].

Search for Selective Glucagon Antagonists ‘A search for selective glucagon inhibitors represents an attractive new way in diabetes management’ was the concluding sentence of a review we published more than 35 years ago [2]. At the time of writing the present review, there is still no selective glucagon inhibitor used in the treatment of diabetes and data concerning compounds in clinical development are not in the public domain. In addition to the effects of eliminating glucagon signalling by knocking out the glucagon receptor mentioned above, animal studies have shown that experimental diabetes is markedly improved by immunoneutralization of glucagon by specific glucagon antibodies [63], by the use of antisense oligonucleotides targeting hepatic glucagon expression [64] or by monoclonal glucagon receptor antibodies [30]. Numerous peptide [65–67] and non-peptide glucagon receptor antagonists have been developed, including substituted ureas [68], benzamidine derivatives [69], alkylidene hydrazides [70–72], 𝛽-alanine derivatives [73] and furan-2-carbohyrazides [74], and have been shown to reduce blood glucose in dogs and monkeys, as well as in non-diabetic and diabetic rodents and, for one compound, in humans [75]. Kelly et al. [76] recently reported that short-term oral administration of the small-molecule glucagon receptor antagonist LY2409021 results in substantial reduction of fasting and postprandial glucose, with minimal hypoglycaemia, but reversible dose-dependent increases in aminotransferases, indicating that longer clinical trials are requested to better evaluate benefits and risks.

A Recent Iconoclastic Proposal: Using a Glucagon Receptor Agonist Rather than an Antagonist? Over the recent years, a new paradigm arose with the development of a GLP-1/glucagon receptor co-agonist [85] and even a GLP-1/glucose-dependent insulinotropic polypeptide (GIP)/glucagon receptor tri-agonist [86]. These newly designed peptides showed remarkable effects in rodent models of diabetes and obesity by improving glucose metabolism and inducing weight loss. In this approach, activation of the glucagon receptor drastically contrasts with previous attempts to inhibit glucagon secretion or action, as described above. The theory is that glucagon could have some beneficial activities by increasing energy expenditure [87] and promoting weight loss, whereas its diabetogenic effects might be counteracted by co-activation of the GLP-1 and GIP receptors [88]. These promising results in rodents have been partly confirmed in humans. Indeed, Tan et al. [89] reported that co-administration of GLP-1 during glucagon infusion results in increased energy expenditure and amelioration of hyperglycaemia, while Cegla et al. [90] reported that co-administration of glucagon and GLP-1, at doses which are individually subanorectic, significantly reduce food intake.

Conclusion Long ignored or neglected, glucagon today appears to be a major factor in the pathophysiology of diabetes. Inhibiting glucagon secretion or antagonizing the action of the hormone are now considered innovative approaches in the treatment of diabetes. Several drugs currently used in the treatment of diabetes may exert part of their effects by reducing glucagon secretion or action. Research is active to identify new compounds reducing glucagon secretion or antagonizing glucagon action. Such compounds should provide a therapeutic benefit without the risk of adverse events.

Risks Associated with Glucagon Suppression Glucagon being recognized to be the first line of defence against hypoglycaemia [77], inhibiting or reducing glucagon action may lead to an increased risk of hypoglycaemia in people with insulin-treated diabetes [78]. It has long been known that glucagon induces hypolipidaemic effects in multiple species [79,80] and, more recently, that GcgR−/− mice exhibit significant defects in lipid synthesis, secretion and oxidation [81]. Consequently, marked attenuation of glucagon signalling potentially increases the risk of hepatosteatosis and hepatocellular injury [81]. Furthermore, glucagon receptor signalling is an important regulator of hepatocyte survival [82], but the minimum level of expression of glucagon for this effect in humans has still to be determined. Last, marked inhibition of glucagon signalling in rodents results in islet hyperplasia, increased endocrine cell proliferation and significant increases in pancreatic weight [83]. The relevance of these observations to humans remains to be established, particularly in view of the fact that rodent islet cells appear to have a greater capacity for replication relative to human [84].

Volume 17 No. 8 August 2015

Conflicts of Interest None declared.

References 1. Lefèbvre P. Glucagon’s golden jubilee at the University of Liège. Br J Diabetes Vasc Dis 2012; 12: 278–284. 2. Lefèbvre PJ, Luyckx AS. Glucagon and diabetes: a reappraisal. Diabetologia 1979; 16: 347–354. 3. Unger RH, Orci L. Paracrinology of islets and the paracrinopathy of diabetes. Proc Natl Acad Sci U S A 2010; 107: 16009–16012. 4. Lefèbvre PJ (ed). Glucagon I and II (Handbook of Experimental Pharmacology), Vol. 66, Vols I and II. Berlin: Springer, 1983. 5. Lefèbvre PJ (ed). Glucagon III (Handbook of Experimental Pharmacology), Vol. 123. Berlin: Springer, 1996. 6. Cerasi E, Accili D, Ahrén B, Henquin J-C, Seino S (eds). Pancreatic 𝛼-cells and glucagon – Neglected metabolic actors. Diabetes Obes Metab 2011; 13(Suppl. 1): 166. 7. Lefèbvre PJ. Glucagon and diabetes. In: Lefèbvre PJ, (ed). Glucagon III (Handbook of Experimental Pharmacology), Vol. 123. Berlin: Springer, 1996; 115–131.

doi:10.1111/dom.12480 723

review article

DIABETES, OBESITY AND METABOLISM

8. Edgerton DS, Cherrington AD. Glucagon as a critical factor in the pathology of diabetes. Diabetes 2011; 60: 377–380.

31. Ohneda A, Ishii S, Horigome K, Yamagata S. Glucagon response to arginine after treatment of diabetes mellitus. Diabetes 1975; 24: 811–819.

9. Dinneen S, Alzaid A, Turk D, Rizza R. Failure of glucagon suppression contributes to postprandial hyperglycaemia in IDDM. Diabetologia 1995; 38: 337–343.

32. Kawamori R, Schichiri M, Kikuchi M, Yamasaki Y, Abe H. Perfect normalization of excessive glucagon responses to intravenous arginine in human diabetes mellitus with the artificial beta-cell. Diabetes 1980; 29: 762–765.

10. Fredhelm S, Andersen M-L, Pörksen S et al. The influence of glucagon on postprandial hyperglycemia in children 5 years after onset of type 1 diabetes. Diabetologia 2015; 58: 828–834. 11. Paquot N, Schneiter P, Jéquier E et al. Effects of ingested fructose and infused glucagon on endogenous glucose production in obese NIDDM patients, obese non-diabetic subjects, and healthy subjects. Diabetologia 1996; 39: 580–586. 12. Shah P, Vella A, Basu A, Schwenk WF, Rizza RA. Lack of suppression of glucagon contributes to postprandial hyperglycemia in subjects with Type 2 diabetes mellitus. J Clin Endocrinol Metab 2000; 85: 4053–4059. 13. Greenbaum CJ, Prigeon RL, D’Alessio DA. Impaired 𝛽-cell function, incretin effect, and glucagon suppression in patients with Type 1 diabetes who have normal fasting glucose. Diabetes 2002; 51: 951–957. 14. Ipp E. Impaired glucose tolerance. The irrepressible 𝛼-cell? Diabetes Care 2000; 23: 569–570. 15. Larsson H, Ahrén B. Islet dysfunction in insulin resistance involves impaired insulin secretion and increased glucagon secretion in post-menopausal women with impaired glucose tolerance. Diabetes Care 2000; 23: 650–657. 16. Choukem SP, Sobngwi E, Boudou P et al. Beta- and alpha-cell dysfunction in Africans with ketosis-prone atypical diabetes during near-normoglycemic remissions. Diabetes Care 2013; 36: 118–123. 17. Lefèbvre P. Ketosis-prone atypical diabetes: glucagon there, too. Diabetes Care 2013; 36: 8–10. 18. Henquin JC, Rahier J. Pancreatic alpha cell mass in European subjects with type 2 diabetes. Diabetologia 2011; 54: 1720–1725. 19. Samols E, Weir GC, Bonner-Weir S. Intra-islet insulin– glucagon–somatostatin relationships. In: Lefèbvre PJ, ed. Glucagon, Vol. II. Berlin: Springer, 1983; 133–173. 20. Lefèbvre P, Paolisso G, Scheen AJ. Pulsatility of glucagon. In: Lefèbvre PJ, ed. Glucagon III (Handbook of Experimental Pharmacology). Berlin: Springer, 1996; 105–113. 21. Unger RH, Cherrington AD. Glucagonocentric restructuring of diabetes: a pathophysiologic and therapeutic makeover. J Clin Invest 2012; 122: 4–12. 22. Bosco D, Armanet M, Morel P et al. Unique arrangement of 𝛼- and 𝛽-cells in human islets of Langerhans. Diabetes 2010; 59: 1202–1210. 23. Lefèbvre P. Diabetes as a paracrinopathy of the islets of Langerhans. Eur Endocrinol 2011; 7: 79–82. 24. Meier JJ, Kjems LL, Veldhuis JD et al. Postprandial suppression of glucagon secretion depends on intact pulsatile insulin secretion: further evidence for the intraislet insulin hypothesis. Diabetes 2006; 55: 1051–1556. 25. Menge BA, Grüber L, Jørgensen SM et al. Loss of inverse relationship between pulsatile insulin and glucagon secretion in patients with type 2 diabetes. Diabetes 2011; 60: 2160–2168. 26. Yu X, Park BH, Wang MY et al. Making insulin-deficient type 1 diabetic rodents thrive without insulin. Proc Natl Acad Sci U S A 2008; 105: 14070–14075. 27. Lee Y, Wang M-Y, Du XQ et al. Glucagon receptor knock out prevents insulin-deficient type 1 diabetes in mice. Diabetes 2011; 60: 391–397. 28. Jun LS, Millican RL, Hawkins ED et al. Absence of glucagon and insulin action reveals a role for Glucagon-like Peptide-1 receptor in endogenous glucose production. Diabetes 2015; 64: 819–827. 29. Lee Y, Berglund ED, Wang MY et al. Metabolic manifestations of insulin deficiency do not occur without glucagon action. Proc Natl Acad Sci U S A 2012; 109: 14972–14976. 30. Wang M-Y, Yan H, Shi Z et al. Glucagon receptor antibody suppresses type 1 diabetes phenotype without insulin by disrupting a novel diabetogenic pathway. Proc Natl Acad Sci USA 2015; 112: 2503–2504.

724 Lefebvre et al.

33. Paolisso G, Sgambato S, Torella R et al. Pulsatile insulin delivery is more efficient than continuous infusion in modulating islet cell function in normal subjects and patients with type-1 diabetes. J Clin Endocrinol Metab 1988; 66: 1220–1226. 34. Cooperberg BA, Cryer PE. 𝛽-cell-mediated signalling predominates over direct 𝛼-cell signalling in the regulation of glucagon secretion in humans. Diabetes Care 2009; 32: 2275–2280. 35. Østergard T, Degn C, Gall M-A et al. The insulin secretagogues glibenclamide and repaglinide do not influence growth hormone secretion in humans but stimulate glucagon secretion during profound insulin deficiency. J Clin Endocrinol Metab 2004; 89: 297–302. 36. Ahren B, Foley JE, Ferrannini E et al. Changes in plasma glucagon levels after a 2-year treatment with vildagliptin or glimepiride in patients with type 2 diabetes inadequately controlled with metformin monotherapy. Diabetes Care 2010; 33: 730–732. 37. Lanstedt-Hallin L, Adamson U, Lins PE. Oral glibenclamide suppresses glucagon secretion during insulin-induced hypoglycaemia in patients with type 2 diabetes. J Clin Endocrinol Metab 1999; 84: 3140–3145. 38. Rorsman P, Ramracheya R, Rorsman NJG, Zhang Q. ATP-regulated potassium channels and voltage-gated calcium channels in pancreatic alpha and beta cells: similar functions but reciprocal effects on secretion. Diabetologia 2014; 57: 1749–1761. 39. Zhang Q, Ramracheya R, Lahmann C et al. Role of KATP channels in glucose-regulated glucagon secretion and impaired counterregulation in type 2 diabetes. Cell Metab 2013; 18: 871–882. 40. Carpentier JL, Luyckx AS, Lefèbvre PJ. Influence of metformin on arginineinduced glucagon secretion in human diabetes. Diabete Metab 1975; 1: 23–28. 41. Lefèbvre PJ, Luyckx AS, Mosora F et al. Oxidation of an exogenous glucose load using naturally labelled 13 C-glucose. Effect of butylbiguanide therapy in obese mildly diabetic subjects. Diabetologia 1978; 14: 39–45. 42. Miller RA, Chu Q, Xie J et al. Biguanides suppress hepatic glucagon signalling by decreasing production of cyclic AMP. Nature 2013; 494: 256–260. 43. Nauck M, Meier J. Incretin-based therapies. In: Ferrannini E, Zimmet P, DeFronzo R, Alberti G, eds. International Textbook of Diabetes Mellitus. Oxford: Wiley, 2015, in press. 44. Hare KJ, Visboll T, Asmar M et al. The glucagonostatic and insulinotropic effects of glucagon-like peptide 1 contribute equally to its glucose-lowering action. Diabetes 2010; 59: 1765–1770. 45. DeFronzo RA, Okerson T, Viswanatha P et al. Effect of exenatide versus sitagliptine on postprandial glucose, insulin and glucagon secretion, gastric emptying, and caloric intake: a randomized, cross-over study. Curr Med Res Opin 2008; 24: 2943–2952. 46. Kolterman OG, Buse JB, Fineman MS et al. Synthetic exendin-4 (exenatide) significantly reduces poetprandial and fasting plasma glucose in subjects with type 2 diabetes. J Clin Endocrinol Metab 2003; 88: 3082–3089. 47. Degn KB, Juhl CB, Sturis J et al. One week’s treatment with the long-acting glucagon-like peptide 1 derivative liraglutide (NN2211) markedly improves 24-h glycemia and alpha- and beta-cell function and reduces endogenous glucose release in patients with type 2 diabetes. Diabetes 2004; 53: 1187–1194. 48. Zinman B, Gerich J, Buse JB et al. Efficacy and safety of the human glucagon-like peptide-1 liraglutide in combination with metformin and thiazolidinedione in patients with type 2 diabetes (LEAD-4 Met + TZD). Diabetes Care 2009; 32: 1124–1130. 49. de Heer J, Rasmussen C, Coy DH, Holst JJ. Glucagon-like peptide-1, but not glucose-dependent insulinotropic peptide inhibits glucagon secretion via

Volume 17 No. 8 August 2015

DIABETES, OBESITY AND METABOLISM

review article

somatostatin (receptor subtype 2) in the perfused rat pancreas. Diabetologia 2008; 51: 2263–2270.

glucagon-like peptide 1 without severe alpha cell hypertrophy in diet-induced obese mice. Diabetologia 2011; 54: 2381–2391.

50. Rutter GA. Regulating glucagon secretion: somatostatin in the spotlight. Diabetes 2009; 58: 299–301.

70. Ling A, Hong Y, Gonzalez J et al. Identification of alkylidene hydrazides as glucagon receptor antagonists. J Med Chem 2001; 44: 3141–3149.

51. Hauge-Evans AC, King AJ, Carmignac D et al. Somatostatin secreted by islet 𝛿-cells fulfils multiple roles as a paracrine regulator of islet function. Diabetes 2009; 58: 403–411.

71. Ling A, Plewe M, Gonzalez J et al. Human glucagon receptor antagonists based on alkylidene hydrazides. Bioorg Med Chem Lett 2002; 12: 663–666.

52. Meier JJ. GLP-1 receptor agonists for individualized treatment of type-2 diabetes mellitus. Nat Rev Endocrinol 2012; 8: 728–742. 53. Scheen AJ. A review of gliptins for 2014. Expert Opin Pharmacother 2015; 16: 43–62. 54. Farngren J, Persson M, Schweizer A, Foley JE, Ahrén B. Vildagliptin reduces glucagon during hyperglycemia and sustains glucagon counterregulation during hypoglycaemia in type 1 diabetes. J Clin Endocrinol Metab 2012; 97: 3799–3806.

72. Madsen P, Ling A, Plewe M et al. Optimization of alkylidene hydrazide based human glucagon receptor antagonists. Discovery of the highly potent and orally available 3-cyano-4-hydroxybenzoic acid[1-(2,3,5,6-tetramethylbenzyl)1Hindol-4-ylmethylene]hydrazide. J Biol Chem 2002; 45: 5755–5775. 73. Lau J, Behrens C, Sidelman UG et al. New beta-alanine derivatives are orally available glucagon receptor antagonists. J Biol Chem 2007; 50: 113–128. 74. Hasegawa F, Niidome K, Migihashi C et al. Discovery of furan-2carbohydrazides as orally active glucagon receptor antagonists. Bioorg Med Chem Lett 2014; 24: 4266–4270.

55. Zhang MY, Yang JH, Shan CY et al. Effects of 24-week treatment with acarbose on glucagon-like peptide 1 in newly diagnosed type 2 diabetic patients: a preliminary report. Cardiovasc Diabetol 2013; 12: 73.

75. Petersen KF, Sullivan JT. Effect of a novel glucagon receptor antagonist (BAY 27-9955) on glucagon-stimulated glucose production in humans. Diabetologia 2001; 44: 2018–2024.

56. Arakawa M, Ebato C, Mita T et al. Miglitol suppresses the postprandial increase in interleukin 6 and enhances active glucagon-like peptide 1 secretion in viscerally obese subjects. Metabolism 2008; 58: 1299–1306.

76. Kelly RP, Garhyan P, Raddad E et al. Short-term administration of the glucagon receptor antagonist LY2402091 lowers blood glucose in healthy people and in those with type 2 diabetes. Diabetes Obes Metab 2015; 17: 414–422.

57. Soccio RE, Chen ER, Lazar MA. Thiazolidinediones and the promise of insulin sensitization in type 2 diabetes. Cell Metab 2014; 20: 573–591. 58. Krätzner R, Fröhlich F, Lepler K et al. A peroxisome proliferator-activated receptor 𝛾-retinoid X receptor heterodimer physically interacts with the transcriptional activator PAX6 to inhibit glucagon gene transcription. Mol Pharmacol 2008; 73: 506–517. 59. Scheen AJ, Paquot N. Metabolic effects of SGLT2 inhibitors beyond increased glycosuria: a review of the clinical experience. Diabetes Metab 2014; 40: S4–11. 60. Scheen AJ. Pharmacodynamics, efficacy and safety of sodium-glucose co-transporter type 2 (SGLT2) inhibitors for the treatment of type 2 diabetes mellitus. Drugs 2015; 75: 33–59. 61. Ferrannini E, Muscelli E, Frascerra S et al. Metabolic response to sodium-glucose cotransporter 2 inhibition in type 2 diabetic patients. J Clin Invest 2014; 124: 499–508.

77. Cryer PE. The barrier of hypoglycaemia in diabetes. Diabetes 2008; 57: 3169–3176. 78. Ali S, Drucker DJ. Benefits and limitations of reducing glucagon action for the treatment of type 2 diabetes. Am J Physiol Endocrinol Metab 2009; 296: E415–421. 79. Tiengo A, Nosadini R. Glucagon and lipoprotein metabolism. In: Lefèbvre PJ, ed. Glucagon II (Handbook of Experimental Pharmacology). Berlin: Springer, 1983; 441–451. 80. Guettet C, Rostaqui N, Mathe D, Lecuyer B, Navarro N, Jacotot B. Effect of chronic glucagon administration on lipoprotein composition in normally fed, fasted and cholesterol-fed rats. Lipids 1991; 26: 451–458. 81. Longuet C, Sinclair EM, Malda A. The glucagon receptor is required for adaptive metabolic response to fasting. Cell Metab 2008; 8: 359–371.

62. Bonner C, Kerr-Conte J, Gmyr V et al. Glucose transporter SGLT2 inhibition triggers glucagon secretion in alpha cells (abstract). Diabetologia 2014; 57(Suppl. 1): 251.

82. Sinclair EM, Yusta B, Streurker C et al. Glucagon receptor signalling is essential for control of murine hepatocyte survival. Gastroenterology 2008; 135: 2096–2106.

63. Sørensen H, Brand CL, Neschen S et al. Immunoneutralization of endogenous glucagon reduces hepatic glucose output and improves long-term glycemic control in diabetic ob/ob mice. Diabetes 2006; 55: 2843–2848.

83. Drucker DJ. Incretin action in the pancreas: potential promise, possible perils and pathological pitfalls. Diabetes 2013; 62: 3316–3323.

64. Sloop KW, Cao JX, Siesky AM et al. Hepatic and glucagon-like peptide-1mediated reversal of diabetes by glucagon receptor antisense oligonucleotides inhibitors. J Clin Invest 2004; 113: 1571–1581. 65. Bagger JI, Knop FK, Holst JJ, Visbøll T. Glucagon antagonism as a potential therapeutic target in type 2 diabetes. Diabetes Obes Metab 2011; 13: 965–971. 66. McShane LM, Franklin ZJ, O’Harte FP, Irwin N. Ablation of glucagon receptor signalling by peptide-based glucagon antagonists improves glucose tolerance in high fat fed mice. Peptides 2014; 60: 95–101. 67. O’Harte FP, Franklin ZJ, Irwin N. Two novel glucagon receptor antagonists prove effective therapeutic agents in high-fat-fed and obese diabetic mice. Diabetes Obes Metab 2014; 16: 1214–1222.

84. Parnaud G, Bosco D, Berney T et al. Proliferation of sorted human and rat beta cells. Diabetologia 2008; 51: 91–100. 85. Day JW, Ottaway N, Patterson JT et al. A new glucagon and GLP-1 coagonist eliminates obesity in rodents. Nat Chem Biol 2009; 5: 749–757. 86. Finan B, Yang B, Ottaway N et al. A rationally designed monomeric peptide triagonist corrects obesity and diabetes in rodents. Nat Med 2015; 21: 27–36. 87. Arafat AM, Kaczmarek P, Skrzypski M et al. Glucagon regulates orexin A secretion in humans and rodents. Diabetologia 2014; 57: 2108–2116. 88. Scheen AJ, Paquot N. A new paradigm for treating obesity and diabetes mellitus. Nat Rev Endocrinol 2015; 11: 196–198.

68. Shen DM, Zhang F, Brady EJ et al. Discovery of novel, potent, and orally active spiro-urea human glucagon receptor antagonists. Bioorg Med Chem Lett 2005; 15: 4564–4569.

89. Tan TM, Field BC, McCullough KA et al. Coadministration of glucagon-like peptide-1 during glucagon infusion in humans results in increased energy expenditure and amelioration of hyperglycemia. Diabetes 2013; 62: 1131–1138.

69. Mu J, Jiang G, Brady E et al. Chronic treatment with a glucagon receptor antagonist lowers glucose and moderately raises circulating glucagon and

90. Cegla J, Troke RC, Jones B et al. Coinfusion of low-dose GLP-1 and glucagon in man results in a reduction in food intake. Diabetes 2014; 63: 3711–3720.

Volume 17 No. 8 August 2015

doi:10.1111/dom.12480 725

Inhibiting or antagonizing glucagon: making progress in diabetes care.

Absolute or relative hyperglucagonaemia has been recognized for years in all experimental or clinical forms of diabetes. It has been suggested that ex...
252KB Sizes 0 Downloads 8 Views