634
Views
13
CrossRef citations to date
0
Altmetric
REVIEW ARTICLE

The GLP‐1 system as a therapeutic target

Pages 314-322 | Published online: 08 Jul 2009

Abstract

Glucagon‐like peptide‐1 (GLP‐1) was first isolated and sequenced twenty years ago. It has been shown to have many effects in man. GLP‐1 stimulates insulin secretion, delays gastric emptying, decreases glucagon levels and reduces appetite, all resulting in a fall in plasma glucose concentrations. More recently, evidence suggests it can stimulate beta cell neogenesis and improve cardiovascular function, and may even be neuroprotective. It is no surprise that this peptide is of increasing interest as a target for the treatment of diabetes. None of the drugs currently available for diabetes are able to achieve targets in all patients and none of them are without side effects. The multiple modes of action of GLP‐1 together with its low propensity for hypoglycaemia appear to give it advantages over currently available treatment modalities. In this review I shall examine the data suggesting that medications modelled on the GLP‐1 system may provide a new therapeutic option for diabetes in the future.

Introduction

It is 90 years since Moore et al. first demonstrated that a substance extracted from the mucosa of the gastrointestinal tract could treat diabetes Citation1. In 1929 Zunz and La Barre named a substance proposed to be released from the gut in response to nutrient ingestion that stimulates insulin secretion ‘incretin’ Citation2. It was not until just over 40 years ago that it was demonstrated that oral ingestion of glucose produced higher insulin levels than intravenous infusion of an equivalent dose of glucose Citation3. It has since been shown that the incretin effect is responsible for at least 50% of meal‐induced insulin release in man Citation4. The incretin effect was defined by Creutzfeldt, who suggested the presence of more than one factor Citation5. The sequence of glucagon‐like peptide‐1 [GLP‐1] in man was first published in 1983 Citation6. It is highly conserved in all species implying an important function Citation7. A number of studies indicated that GLP‐1 is the most potent naturally occurring incretin in man Citation8–9, and has a physiological role in the regulation of postprandial glucose Citation10.

Key messages

  • Current therapeutic strategies for type‐2 diabetes are inadequate, the disease is rapidly increasing in prevalence and new and better drugs are urgently needed.

  • GLP‐1 increases plasma insulin, decreases glucagon, delays gastric emptying and reduces appetite whilst causing little or no hypoglycaemia.

  • Long‐acting GLP‐1 analogues resistant to breakdown by dipeptidyl peptidase IV and antagonists of this enzyme are potentially advantageous treatments for diabetes.

Actions of GLP‐1

Regulation of carbohydrate metabolism

GLP‐1 not only stimulates insulin secretion (insulinotropic), but has several other actions which coordinate to reduce plasma glucose. GLP‐1 stimulates insulin synthesis Citation11, and it appears to increase islet cell mass Citation12, though the mechanism through which this occurs is poorly characterized Citation13. It also inhibits islet apoptosis Citation14. Type‐2 diabetes is associated with a progressive decline in beta cell function Citation15, thus, up‐regulation of the GLP‐1 axis may even slow the progression of diabetes.

GLP‐1 reduces glucagon Citation8, Citation16, which opposes the action of insulin. As the major role of glucagon is in the fasted state Citation17, and levels in diabetes are high Citation18, antagonism via the GLP‐1 system would be expected to be particularly important in controlling fasting glucose in diabetes. It also reduces glucose by delaying gastric emptying Citation19–21, and inhibits gastric acid secretion Citation19. Administration of GLP‐1 with a meal in healthy humans causes a reduction in meal‐related glucose and insulin excursions, implying that delay in gastric emptying is more important than its insulinotropic effect Citation20. GLP‐1 reduces food intake and act as a satiety factor in rats Citation22, it can also cause weight loss Citation23. Further data indicate that it can reduce food intake in man Citation21, Citation24, Citation25, also likely to help ameliorate diabetes.

A further advantage of GLP‐1 as a potential agent to treat diabetes would be if it enhanced glucose disposal independent of insulin. This was suggested Citation17, Citation26, but most studies have failed to reproduce it Citation27, Citation28, and the effect remains controversial. GLP‐1 is glucose‐dependent Citation29, thus, it reduces glucose to a greater extent in the hyper‐ than the normoglycaemic state Citation30. This fact was used to infer that it does not cause fasting hypoglycaemia. In some studies this is the case Citation31, Citation32, whereas in others hypoglycaemia can occur Citation30, Citation33–35, however, apparently only in normal volunteers, not people with type 2 diabetes. Overall it seems that therapeutic agents for type‐2 diabetes based on GLP‐1 will cause little hypoglycaemia, giving it major advantage over insulin or sulphonylureas Citation36.

Cardiovascular action

GLP‐1 has cardiovascular actions though these vary between species. In the rat it increased heart rate and blood pressure Citation37. In a rat model of hypertension, GLP‐1 reduced blood pressure, seemingly via a natriuretic and diuretic action Citation38. GLP‐1 receptor knockout mice have reduced heart rate and increased left ventricular wall thickness Citation39, consistent with a physiological role in cardiovascular metabolism. In the calf, it is chronotropic without effect on blood pressure Citation40. In the dog it appeared to enhance recovery from ischaemic myocardial stunning Citation41.

Cardiovascular effects of GLP‐1 in man also vary. Subcutaneous (s/c) injections in healthy volunteers increased blood pressure Citation34, whereas continuous s/c infusion tended to lower it Citation33. GLP‐1 infusion improved endothelial function in patients with stable coronary artery disease whilst having no effect in normal subjects Citation42. Intravenous (iv) GLP‐1 improved left ventricular function in patients with severe heart failure after large myocardial infarction and successful primary angioplasty Citation43. Given the frequency of endothelial dysfunction in diabetes, it may have beneficial effects long‐term, and may have a short‐term role in certain high risk patients.

Action on sodium and water balance

Peripheral administration of GLP‐1 inhibited water intake in the rat Citation44. This effect was abolished by destruction of the arcuate nucleus and circumventricular organs Citation45. The area postrema forms part of the circumventricular organs and peripheral administration activates this area Citation46, implying peripheral GLP‐1 may be acting centrally. It also acts as a natriuretic and diuretic in the hypertensive rat Citation38.

As well as a reduction in food intake with GLP‐1 administration, a decrease in water intake in healthy volunteers Citation47 and people with type‐2 diabetes Citation25 has been found. It is natriuretic in healthy human subjects and patients with insulin resistance, suggesting a possible renoprotective effect Citation48.

Neurological function

GLP‐1 may also be neuroprotective. It reduced apoptosis in models of neurodegeneration, Citation49 and reduced levels of amyloid Citation50. It improved spatial learning in rats and restored a learning deficit in GLP‐1 knockout mice: indeed receptor overexpression improved learning and memory Citation51. There are no human data reported, however, a beneficial role in Alzheimer's disease has been proposed Citation50.

Therapeutic principle of GLP‐1

GLP‐1 is rapidly broken down by dipeptidyl peptidase IV (DPP‐IV). The plasma half‐life in man is 1–3 minutes Citation52. Nevertheless, exogenous GLP‐1 can reduce plasma glucose in type‐2 diabetes Citation16, Citation53, Citation9. This has been confirmed using s/c injections Citation54, buccal tablets Citation55, and s/c infusion Citation56–59, with additional effect over metformin Citation56 and glitazones Citation59. All these studies verified that the GLP‐1 system is a target for therapy of type‐2 diabetes, alone or in combination providing impetus to the search for a long‐acting analogue.

GLP‐1 analogues

Exendin‐4

Exendin‐4, isolated from the salivary gland of the Gila monster, is a long acting GLP‐1 receptor agonist Citation60. The half‐life of this peptide in man is about 30 minutes Citation61, making it a superior prospect to GLP‐1 as an anti‐diabetic agent. Exendin‐4 has a longer lasting and more potent glucose‐lowering effect than GLP‐1 in diabetic mice Citation62, rats and monkeys Citation63. It stimulates beta cell replication and islet neogenesis Citation64, Citation65 improving glucose tolerance Citation65, and reduces weight gain in rats Citation66. Exendin‐4 also mimics the neuroprotective effects of GLP‐1 Citation49.

Intravenous infusion of exendin‐4 in healthy humans reduced fasting and postprandial glucose, as well as reducing food intake by 19% Citation61. The dose used in this study may have been close to the maximal tolerated as double this dose caused nausea in some subjects Citation61. A hyperglycaemic clamp was used to show that iv exendin‐4 is a potent insulinotropic agent in type‐2 diabetes, with a long duration of action Citation67. Subcutaneous exendin‐4 caused complete abolition of postprandial glycaemia in people with type‐2 diabetes, plasma glucose dropping or at least failing to rise after a meal in the active group, associated with a potent slowing of gastric emptying Citation68. However, a number of subjects suffered nausea and/or vomiting. A reduction of fasting glucose was also noted, though no one was hypoglycaemic Citation68. Subcutaneous exendin‐4 also reduced postprandial hyperglycaemia in type‐1 diabetes by 90%, via inhibition of gastric emptying and glucagon suppression Citation69.

Twice‐daily s/c exendin‐4 injections for a month in people with type‐2 diabetes caused long‐term glycaemic control, as measured by glycosylated haemoglobin (HbA1c), to improve by 0.8% Citation70. There were no significant side‐effects, aside from short‐lasting nausea at the start of the study with two subjects. There was no hypoglycaemia with the lowest blood glucose being 3.3 mmol/l, but there was no weight loss. Improvement in glycaemic control did not occur for 24 hours, and the authors concluded that exendin‐4 in this dose/form would be unlikely to be a treatment for type‐2 diabetes Citation70. Data from Amylin Pharmaceuticals illustrated its effects combined with metformin and/or sulphonylureas Citation71. Over half the patients were on combined medication. ‘Triple’ therapy with two or three times a day s/c exendin‐4 reduced HbA1c 0.9% in one month, compared with 0.3% in the placebo group. HbA1c indicates average plasma glucose over three months, thus longer trials should have a greater effect. Mild hypoglycaemia did occur, although only in combination with a sulphonylurea: nausea also occurred causing a dropout rate of 3.7% Citation71. A recent report showed that the nausea caused by exendin‐4 can be reduced by dose titration Citation72.

The first study of exendin‐4 for longer than one month has recently been reported by Amylin Citation73. Patients taking sulphonylureas were randomized to one of two doses of s/c exendin‐4 twice a day or placebo for six months. HbA1c decreased by 0.98% with 10 mcg and 0.57% with 5 mcg compared to placebo, the effect was greater in more poorly controlled patients (1.35 and 0.71 respectively if HbA1c>9%). Study withdrawal was 20%–30% in the treatment arms, and 40% in the placebo arm. The latter was partly due to worse glycaemic control as per the protocol, which is likely to have caused a reduction in the measured treatment effect compared with the actual effect. Adverse events causing withdrawal were 3.3% for placebo, 7.2% for low dose and 10.1% for high dose exendin‐4. There was a small weight loss of 1.6 kg over the study period in the high dose group; the placebo group lost 0.6 kg; and reported nausea and hypoglycaemia were more common in the treatment groups Citation73. The improvement in glycaemic control is not unlike that for agents currently used in type‐2 diabetes. Data looking at its effects in combination with metformin or with sulphonylureas and metformin have been presented in abstract form.

In 2002 Eli Lilly went into partnership with Amylin to develop exendin‐4, also known as exenatide. The peptide currently has more published human data than any other targeting the GLP‐1 receptor. A long acting formulation designed for use once a week to once a month is in clinical trials, but no more information is currently available.

Liraglutide

The other GLP‐1 agonist with considerable published data is the Novo Nordisk product liraglutide. This is an acylated derivative of GLP‐1 that binds to albumin and is resistant to DPP‐IV degradation Citation74. Subcutaneous injection of liraglutide in rats caused weight loss for ten days Citation75. It attenuated diabetes in two models of pre‐diabetes, at least partly via reduction in food intake Citation76, and induced a short term increase in beta cell mass in normal rats Citation77. Liraglutide reduced glucose in diabetic ob/ob and db/db mice, also increasing beta cell mass in the latter Citation78. In pigs, it improved glycaemic control chronically, it stimulated insulin, decreased glucagon levels and delayed gastric emptying Citation79.

The half‐life of liraglutide in man is 11–15 hours Citation80. Injection into people with type‐2 diabetes reduced fasting glucose, and postprandial glucose via stimulation of insulin, reduction of glucagon and delay in gastric emptying Citation81. Subcutaneous liraglutide once daily for a week in people with type‐2 diabetes caused a decrease in average plasma glucose of 2 mmol/L and postprandial glucose after meals of 20 per cent Citation82. Insulin levels were unchanged, though in the presence of reduced plasma glucose this suggests a relative insulinotropic effect, as has been described with GLP‐1 Citation83. It decreased plasma glucagon and glycogenolysis, improved islet cell function and first phase insulin response, but did not affect gastric emptying Citation82. There was no hypoglycaemia but nausea and abdominal pain were experienced by some.

An eight‐week study of once daily s/c liraglutide reduced HbA1c by 0.8% compared with placebo, with no change in gastric emptying, body weight or energy expenditure Citation84. Side‐effects of medication were minor. These latter two studies Citation82, Citation84 used a lower dose of liraglutide than that of the previous Citation81 and it seems likely at higher doses that liraglutide does delay gastric emptying.

Administration of liraglutide for 12 weeks caused a decrease in HbA1c of 0.75% with the highest dose Citation85. There was a tendency for weight loss, with one dose causing significant weight loss of 1.2 kg compared with placebo. There was minor nausea with increase in dose, but no patients withdrew from the study Citation85. Acute studies in man using GLP‐1 and its agonists have generally shown a decrease in food intake and even weight loss Citation21, Citation24, Citation25, Citation47, Citation61, however, the chronic effects are much less impressive. Weight loss of 1 kg versus placebo was found with exenatide administered for six months Citation73 and 1.2 kg with one dose of liraglutide for three months Citation85. I suspect, given the number of studies showing decreased food intake without nausea, that the weight loss is a direct effect, at least at certain doses; however, the metabolic significance may be minor, and dosage is obviously critical. Nevertheless, for prescribers and patients alike the fact that a GLP‐1‐based drug is likely to cause weight loss, however minimal, compared with the well recognized weight gain of insulin and sulphonylureas and the weight stabilization of metformin Citation86 will be crucial in the ability of these drugs to compete in the pharmaceutical market.

Other GLP‐1 agonists

Various strategies have been used to protect GLP‐1 from breakdown by DPP‐IV and thus increase the half‐life. Acylation or modification of one of the first three amino acids of GLP‐1 (7‐36 amide) has been most commonly investigated Citation87. Conjuchem have patented a modified GLP‐1, which binds to albumin called CJC‐1131, this is currently in phase II clinical trials Citation88. Human Genome Sciences have a similar albumin‐bound peptide, Albugon, which has been shown to have a prolonged action in mice Citation89.

DPP‐IV antagonists

An alternative strategy to target the GLP‐1 system, rather than creating DPP‐IV resistant analogues, is to inhibit the enzyme itself. DPP‐IV knockout mice have reduced glycaemic excursion after a glucose challenge Citation90 and are protected from both obesity and insulin resistance, implying DPP‐IV antagonists are a therapeutic option Citation91. The enzyme not only inactivates GLP‐1, but also glucose‐dependent insulinotropic peptide (GIP) Citation92. Knockout of both the GLP‐1 and GIP receptors in a mouse model caused increased glycaemic excursion in response to oral glucose, it also did not respond to DPP‐IV inhibition implying that GLP‐1 and GIP account for most of the incretin action regulated by DPP‐IV Citation93.

However, a note of caution must be sounded in that DPP‐IV, also known as CD26, has many actions. Not only does it have many peptide hormones as substrates, it also stimulates T‐cell proliferation as well as inactivating cytokines such as tumour necrosis factor‐alpha (TNF‐α) Citation94. Activity of the enzyme alters in a number of disease states, in particular dropping in depression Citation95. DPP‐IV structure is highly conserved in evolution like GLP‐1, implying an important physiological role Citation96 and long‐term blockade may have predictable or hitherto unpredictable side‐effects Citation97.

Despite the above, a number of oral DPP‐IV inhibitors have been tested in animals, particularly glucose intolerant models, and in man. Several acute studies have been reported: less data are available on the chronic use of DPP‐IV antagonists. The Novartis compound DPP728 given long‐term to mice improved glucose tolerance with increase in insulin levels Citation98. The Ferring compound FE 999011 improved glycaemia and delayed the onset of diabetes in Zucker fatty rats Citation99. Chronic administration of the Probiodrug P32/98 improved glycaemia in Zucker rats Citation100.

DPP728 has also been shown to be active in people with diet‐controlled type‐2 diabetes Citation101. Two or three times a day oral therapy improved average plasma glucose by 1 mmol/L after four weeks compared with placebo, there was no change in body weight. Tablets were taken ten minutes before food, and were generally well tolerated, the incidence of hypoglycaemia and gastrointestinal side‐effects was low, though one patient did develop transient nephrotic syndrome Citation101.

LAF237 (Vildagliptin)

Novartis have published no further data on DPP728, and have concentrated on the longer acting DPP‐IV inhibitor LAF237, also known as vildagliptin. Administration once a day thirty minutes before breakfast for four weeks in people with type‐2 diabetes decreased plasma glucose 1 mmol/L. There was little effect on insulin, but glucagon was inhibited. No significant side effects were noted, and there was no change in body weight Citation102. This group has gone on to perform a longer‐term study of vildagliptin in patients treated with metformin Citation103. After 12 weeks HbA1c dropped 0.7% compared with placebo, after 1 year it was 1.1% in the 58 out of the original 107 patients who continued. Diabetic control at the start of the study was moderate (HbA1c 7.7 or 7.8). There was reduced fasting glucose and glucose area under the curve (AUC) in response to a test meal with vildagliptin, insulin AUC was unchanged indicating a relative insulinotropic effect. No effect on body weight was found and there was a low incidence of hypoglycaemia Citation103. Three of 42 patients treated with vildagliptin for a year required an increase in their anti‐hypertensives; the cause of this is unknown, though this would be consistent with the increase in blood pressure previously found with GLP‐1 Citation34.

Conclusion

Much data has been published on GLP‐1 and its agonists in rats and other species. Potentially important theories have been proposed regarding increase in islet cell mass induced by GLP‐1, slowing of the progression of diabetes or delay in its onset with GLP‐1 and its agonists and even a neuroprotective role. However, these have all just been demonstrated in animals, and are only inferred and have not been reported in man. Nevertheless, several reports have been published in man regarding three first‐generation agents targeting the GLP‐1 system. Exenatide reduces HbA1c and probably causes some weight loss, it produces little hypoglycaemia, though it has to be injected twice a day, and nausea is likely to limit its usefulness in some. There is a hint of a much longer‐acting exendin‐like molecule. Less data are available for liraglutide, which seems to have similar efficacy and also may cause some weight loss. It only has to be injected once a day. Vildagliptin is the only oral compound available. It has similar anti‐hyperglycaemic effect to the other two but without causing weight loss. We await evidence to suggest whether the less specific DPP‐IV antagonism is of long‐term advantage or harm.

Currently, the major compounds used in the treatment of type‐2 diabetes are metformin, which is not tolerated by many and contra‐indicated in many more, sulphonylureas, which cause weight gain and hypoglycaemia, and thiazolidinediones, which appear less efficacious than the others, cause weight gain and have less long‐term data. The alternative is insulin; however, the necessary multiple injections are disliked by patients, and weight gain and hypoglycaemia remain problematic. The prevalence of obesity and thence type‐2 diabetes is increasing rapidly. There is an urgent need for newer drugs that have the activity profile of agents acting on the GLP‐1 system: soon we will see whether any of the agents above fit the bill. Excitingly, we have early evidence suggesting the possibility of advantage in the long term, ranging from cardiovascular to neurological protection, features that would make such drugs even more attractive for the long‐term treatment of type‐2 diabetes.

References

  • Moore B., Edie E. S., Abram J. H. On the treatment of diabetes mellitus by the acid extraction of duodenal mucous membranes. Biochem J 1906; 1: 28–38
  • Zunz E., La Barre J. Contributions a l'etude des variations physiologiques de la secretion interne du pancreas: relations entre les secretions externe et interne du pancreas. Arch Int Physiol Biochim Biophys 1929; 31: 20–44
  • Mcintyre N., Holdsworth C. D., Turner D. S. New interpretation of oral glucose tolerance. Lancet 1964; 41: 20–1
  • Perley M. J., Kipnis D. M. Plasma insulin responses to oral and intravenous glucose: studies in normal and diabetic subjects. J Clin Invest 1967; 46: 1954–62
  • Creutzfeldt W. The incretin concept today. Diabetologia 1979; 16: 75–85
  • Bell G. I., Sanchez‐Pescador R., Laybourn P. J., Najarian R. C. Exon duplication and divergence in the human preproglucagon gene. Nature 1983; 304: 368–71
  • Lopez L. C., Li W. H., Frazier M. L., Luo C. C., Saunders G. F. Evolution of glucagon genes. Mol Biol Evol 1984; 1: 335–44
  • Kreymann B., Williams G., Ghatei M. A., Bloom S. R. Glucagon‐like peptide‐1 7‐36: a physiological incretin in man. Lancet 1987; 2: 1300–4
  • Nauck M. A., Heimesaat M. M., Orskov C., Holst J. J., Ebert R., Creutzfeldt W. Preserved incretin activity of glucagon‐like peptide 1 [7‐36 amide] but not of synthetic human gastric inhibitory polypeptide in patients with type‐2 diabetes mellitus. J Clin Invest 1993; 91: 301–7
  • Edwards C. M., Todd J. F., Mahmoudi M., Wang Z., Wang R. M., Ghatei M. A., et al. Glucagon‐like peptide 1 has a physiological role in the control of postprandial glucose in humans: studies with the antagonist exendin 9–39. Diabetes 1999; 48: 86–93
  • Drucker D. J., Philippe J., Mojsov S., Chick W. L., Habener J. F. Glucagon‐like peptide 1 stimulates insulin gene expression and increases cyclic AMP levels in a rat islet cell line. Proc Natl Acad Sci U S A 1987; 84: 3434–8
  • De Leon D. D., Deng S., Madani R., Ahima R. S., Drucker D. J., Stoffers D. A. Role of endogenous glucagon‐like peptide‐1 in islet regeneration following partial pancreatectomy. Diabetes 2003; 52: 365–71
  • Wang Q., Brubaker P. L. Glucagon‐like peptide‐1 treatment delays the onset of diabetes in 8 week‐old db/db mice. Diabetologia 2002; 45: 1263–73
  • Farilla L., Bulotta A., Hirshberg B., Li Calzi S., Khoury N., Noushmehr H., et al. Glucagon‐like peptide 1 inhibits cell apoptosis and improves glucose responsiveness of freshly isolated human islets. Endocrinology 2003; 144: 5149–58
  • UK Prospective Diabetes Study (UKPDS) Group. Glycemic control with diet, sulfonylurea, metformin, or insulin in patients with type 2 diabetes mellitus: progressive requirement for multiple therapies (UKPDS 49). JAMA 1999; 281: 2005–12
  • Gutniak M., Orskov C., Holst J. J., Ahren B., Efendic S. Antidiabetogenic effect of glucagon‐like peptide‐1 (7‐36) amide in normal subjects and patients with diabetes mellitus. N Engl J Med 1992; 326: 1316–22
  • Unger R. H. Role of glucagon in the pathogenesis of diabetes: the status of the controversy. Metabolism 1978; 27: 1691–709
  • Unger R. H., Orci L. The essential role of glucagon in the pathogenesis of diabetes mellitus. Lancet 1975; 7897: 14–6
  • Wettergren A., Schjoldager B., Mortensen P. E., Myhre J., Christiansen J., Holst J. J. Truncated GLP‐1 (proglucagon 78‐107‐amide) inhibits gastric and pancreatic functions in man. Dig Dis Sci 1993; 38: 665–73
  • Nauck M. A., Niedereichholz U., Ettler R., Holst J. J., Orskov C., Ritzel R., et al. Glucagon‐like peptide 1 inhibition of gastric emptying outweighs its insulinotropic effects in healthy humans. Am J Physiol 1997; 273: E981–8
  • Naslund E., Gutniak M., Skogar S., Rossner S., Hellstrom P. M. Glucagon‐like peptide 1 increases the period of postprandial satiety and slows gastric emptying in obese men. Am J Clin Nutr 1998; 68: 525–30
  • Turton M. D., O'Shea D., Gunn I., Beak S. A., Edwards C. M., Meeran K., et al. A role for glucagon‐like peptide‐1 in the central regulation of feeding. Nature 1996; 379: 69–72
  • Meeran K., O'Shea D., Edwards C. M., Turton M. D., Heath M. M., Gunn I., et al. Repeated intracerebroventricular administration of glucagon‐like peptide‐1‐(7‐36) amide or exendin‐(9‐39) alters body weight in the rat. Endocrinology 1999; 140: 244–50
  • Flint A., Raben A., Astrup A., Holst J. J. Glucagon‐like peptide 1 promotes satiety and suppresses energy intake in humans. J Clin Invest 1998; 101: 515–20
  • Gutzwiller J. P., Drewe J., Goke B., Schmidt H., Rohrer B., Lareida J., et al. Glucagon‐like peptide‐1 promotes satiety and reduces food intake in patients with diabetes mellitus type 2. Am J Physiol 1999; 276: R1541–4
  • D'Alessio D. A., Kahn S. E., Leusner C. R., Ensinck J. W. Glucagon‐like peptide 1 enhances glucose tolerance both by stimulation of insulin release and by increasing insulin‐independent glucose disposal. J Clin Invest 1994; 93: 2263–6
  • Orskov L., Holst J. J., Moller J., Orskov C., Moller N., Alberti K. G., et al. GLP‐1 does not not acutely affect insulin sensitivity in healthy man. Diabetologia 1996; 39: 1227–32
  • Ahrén B., Larsson H., Holst J. J. Effects of glucagon‐like peptide‐1 on islet function and insulin sensitivity in noninsulin‐dependent diabetes mellitus. J Clin Endocrinol Metab 1997; 82: 473–8
  • Holz G. G 4th., Kuhtreiber W. M., Habener J. F. Pancreatic beta‐cells are rendered glucose‐competent by the insulinotropic hormone glucagon‐like peptide‐1(7‐37). Nature 1993; 361: 362–5
  • Ritzel R., Orskov C., Holst J. J., Nauck M. A. Pharmacokinetic, insulinotropic, and glucagonostatic properties of GLP‐1 [7‐36 amide] after subcutaneous injection in healthy volunteers. Dose‐response‐relationships. Diabetologia 1995; 38: 720–5
  • Qualmann C., Nauck M. A., Holst J. J., Orskov C., Creutzfeldt W. Insulinotropic actions of intravenous glucagon‐like peptide‐1 (GLP‐1) [7‐36 amide] in the fasting state in healthy subjects. Acta Diabetol 1995; 32: 13–6
  • Knop F. K., Vilsboll T., Larsen S., Madsbad S., Holst J. J., Krarup T. No hypoglycemia after subcutaneous administration of glucagon‐like peptide‐1 in lean type 2 diabetic patients and in patients with diabetes secondary to chronic pancreatitis. Diabetes Care 2003; 26: 2581–7
  • Toft‐Nielsen M., Madsbad S., Holst J. J. Exaggerated secretion of glucagon‐like peptide‐1 (GLP‐1) could cause reactive hypoglycaemia. Diabetologia 1998; 41: 1180–6
  • Edwards C. M., Todd J. F., Ghatei M. A., Bloom S. R. Subcutaneous glucagon‐like peptide‐1 (7‐36) amide is insulinotropic and can cause hypoglycaemia in fasted healthy subjects. Clin Sci 1998; 95: 719–24
  • Vilsboll T., Krarup T., Madsbad S., Holst J. J. No reactive hypoglycaemia in Type 2 diabetic patients after subcutaneous administration of GLP‐1 and intravenous glucose. Diabet Med 2001; 18: 144–9
  • UK Prospective Diabetes Study (UKPDS) Group. Intensive blood‐glucose control with sulphonylureas or insulin compared with conventional treatment and risk of complications in patients with type 2 diabetes (UKPDS 33). Lancet 1998; 352: 837–53
  • Barragan J. M., Rodriguez R. E., Blazquez E. Changes in arterial blood pressure and heart rate induced by glucagon‐like peptide‐1‐(7‐36) amide in rats. Am J Physiol 1994; 266: E459–66
  • Yu M., Moreno C., Hoagland K. M., Dahly A., Ditter K., Mistry M., et al. Antihypertensive effect of glucagon‐like peptide 1 in Dahl salt‐sensitive rats. J Hypertens 2003; 21: 1125–35
  • Gros R., You X., Baggio L. L., Kabir M. G., Sadi A. M., Mungrue I. N., et al. Cardiac function in mice lacking the glucagon‐like peptide‐1 receptor. Endocrinology 2003; 144: 2242–52
  • Edwards C. M., Edwards A. V., Bloom S. R. Cardiovascular and pancreatic endocrine responses to glucagon‐like peptide‐1(7‐36) amide in the conscious calf. Exp Physiol 1997; 82: 709–16
  • Nikolaidis L. A., Doverspike A., Hentosz T., Zourelias L., Shen Y. T., Elahi D., et al. Glucagon‐like peptide‐1 limits myocardial stunning following brief coronary occlusion and reperfusion in conscious canines. J Pharmacol Exp Ther 2005; 312: 303–8
  • Nystrom T., Gutniak M. K., Zhang Q., Zhang F., Holst J. J., Ahren B., et al. Effects of glucagon‐like peptide‐1 on endothelial function in type 2 diabetes patients with stable coronary artery disease. Am J Physiol Endocrinol Metab 2004; 287: E1209–15
  • Nikolaidis L. A., Mankad S., Sokos G. G., Miske G., Shah A., Elahi D., et al. Effects of glucagon‐like peptide‐1 in patients with acute myocardial infarction and left ventricular dysfunction after successful reperfusion. Circulation 2004; 109: 962–5
  • Tang‐Christensen M., Larsen P. J., Goke R., Fink‐Jensen A., Jessop D. S., Moller M., et al. Central administration of GLP‐1‐(7‐36) amide inhibits food and water intake in rats. Am J Physiol 1996; 271: R848–56
  • Tang‐Christensen M., Vrang N., Larsen P. J. Glucagon‐like peptide 1(7‐36) amide's central inhibition of feeding and peripheral inhibition of drinking are abolished by neonatal monosodium glutamate treatment. Diabetes 1998; 47: 530–7
  • Yamamoto H., Kishi T., Lee C. E., Choi B. J., Fang H., Hollenberg A. N., et al. Glucagon‐like peptide‐1‐responsive catecholamine neurons in the area postrema link peripheral glucagon‐like peptide‐1 with central autonomic control sites. J Neurosci 2003; 23: 2939–46
  • Gutzwiller J. P., Goke B., Drewe J., Hildebrand P., Ketterer S., Handschin D., et al. Glucagon‐like peptide‐1: a potent regulator of food intake in humans. Gut 1999; 44: 81–6
  • Gutzwiller J. P., Tschopp S., Bock A., Zehnder C. E., Huber A. R., Kreyenbuehl M., et al. Glucagon‐like peptide 1 induces natriuresis in healthy subjects and in insulin‐resistant obese men. J Clin Endocrinol Metab 2004; 89: 3055–61
  • Perry T., Haughey N. J., Mattson M. P., Egan J. M., Greig N. H. Protection and reversal of excitotoxic neuronal damage by glucagon‐like peptide‐1 and exendin‐4. J Pharmacol Exp Ther 2002; 302: 881–8
  • Perry T., Lahiri D. K., Sambamurti K., Chen D., Mattson M. P., Egan J. M., et al. Glucagon‐like peptide‐1 decreases endogenous amyloid‐beta peptide (Abeta) levels and protects hippocampal neurons from death induced by Abeta and iron. J Neurosci Res 2003; 72: 603–12
  • During M. J., Cao L., Zuzga D. S., Francis J. S., Fitzsimons H. L., Jiao X., et al. Glucagon‐like peptide‐1 receptor is involved in learning and neuroprotection. Nat Med 2003; 9: 1173–9
  • Deacon C. F., Knudsen L. B., Madsen K., Wiberg F. C., Jacobsen O., Holst J. J. Dipeptidyl peptidase IV resistant analogues of glucagon‐like peptide‐1 which have extended metabolic stability and improved biological activity. Diabetologia 1998; 41: 271–8
  • Nauck M. A., Kleine N., Orskov C., Holst J. J., Willms B., Creutzfeldt W. Normalization of fasting hyperglycaemia by exogenous glucagon‐like peptide 1 (7‐36 amide) in type 2 (non‐insulin‐dependent) diabetic patients. Diabetologia 1993; 36: 741–4
  • Todd J. F., Edwards C. M., Ghatei M. A., Mather H. M., Bloom S. R. Subcutaneous glucagon‐like peptide‐1 improves postprandial glycaemic control over a 3‐week period in patients with early type 2 diabetes. Clin Sci 1998; 95: 325–9
  • Gutniak M. K., Larsson H., Sanders S. W., Juneskans O., Holst J. J., Ahren B. GLP‐1 tablet in type 2 diabetes in fasting and postprandial conditions. Diabetes Care 1997; 20: 1874–9
  • Zander M., Taskiran M., Toft‐Nielsen M. B., Madsbad S., Holst J. J. Additive glucose‐lowering effects of glucagon‐like peptide‐1 and metformin in type 2 diabetes. Diabetes Care 2001; 24: 720–5
  • Zander M., Madsbad S., Madsen J. L., Holst J. J. Effect of 6‐week course of glucagon‐like peptide 1 on glycaemic control, insulin sensitivity, and beta‐cell function in type 2 diabetes: a parallel‐group study. Lancet 2002; 359: 824–30
  • Meneilly G. S., Greig N., Tildesley H., Habener J. F., Egan J. M., Elahi D. Effects of 3 months of continuous subcutaneous administration of glucagon‐like peptide 1 in elderly patients with type 2 diabetes. Diabetes Care 2003; 26: 2835–41
  • Zander M., Christiansen A., Madsbad S., Holst J. J. Additive effects of glucagon‐like peptide 1 and pioglitazone in patients with type 2 diabetes. Diabetes Care 2004; 27: 1910–4
  • Raufman J. P., Singh L., Singh G., Eng J. Truncated glucagon‐like peptide‐1 interacts with exendin receptors on dispersed acini from guinea pig pancreas. Identification of a mammalian analogue of the reptilian peptide exendin‐4. J Biol Chem 1992; 267: 21432–7
  • Edwards C. M., Stanley S. A., Davis R., Brynes A. E., Frost G. S., Seal L. J., et al. Exendin‐4 reduces fasting and postprandial glucose and decreases energy intake in healthy volunteers. Am J Physiol Endocrinol Metab 2001; 281: E155–61
  • Greig N. H., Holloway H. W., De Ore K. A., Jani D., Wang Y., Zhou J., et al. Once daily injection of exendin‐4 to diabetic mice achieves long‐term beneficial effects on blood glucose concentrations. Diabetologia 1999; 42: 45–50
  • Young A. A., Gedulin B. R., Bhavsar S., Bodkin N., Jodka C., Hansen B., et al. Glucose‐lowering and insulin‐sensitizing actions of exendin‐4: studies in obese diabetic (ob/ob, db/db) mice, diabetic fatty Zucker rats, and diabetic rhesus monkeys (Macaca mulatta). Diabetes 1999; 48: 1026–34
  • Xu G., Stoffers D. A., Habener J. F., Bonner‐Weir S. Exendin‐4 stimulates both beta‐cell replication and neogenesis, resulting in increased beta‐cell mass and improved glucose tolerance in diabetic rats. Diabetes 1999; 48: 2270–6
  • Tourrel C., Bailbe D., Meile M. J., Kergoat M., Portha B. Glucagon‐like peptide‐1 and exendin‐4 stimulate beta‐cell neogenesis in streptozotocin‐treated newborn rats resulting in persistently improved glucose homeostasis at adult age. Diabetes 2001; 50: 1562–70
  • Szayna M., Doyle M. E., Betkey J. A., Holloway H. W., Spencer R. G., Greig N. H., et al. Exendin‐4 decelerates food intake, weight gain, and fat deposition in Zucker rats. Endocrinology 2000; 141: 1936–41
  • Egan J. M., Clocquet A. R., Elahi D. The insulinotropic effect of acute exendin‐4 administered to humans: comparison of nondiabetic state to type 2 diabetes. J Clin Endocrinol Metab 2002; 87: 1282–90
  • Kolterman O. G., Buse J. B., Fineman M. S., Gaines E., Heintz S., Bicsak T. A., et al. Synthetic exendin‐4 (exenatide) significantly reduces postprandial and fasting plasma glucose in subjects with type 2 diabetes. J Clin Endocrinol Metab 2003; 88: 3082–9
  • Dupre J., Behme M. T., McDonald T. J. Exendin‐4 normalized postcibal glycemic excursions in type 1 diabetes. J Clin Endocrinol Metab 2004; 89: 3469–73
  • Egan J. M., Meneilly G. S., Elahi D. Effects of 1‐mo bolus subcutaneous administration of exendin‐4 in type 2 diabetes. Am J Physiol Endocrinol Metab 2003; 284: E1072–9
  • Fineman M. S., Bicsak T. A., Shen L. Z., Taylor K., Gaines E., Varns A., et al. Effect on glycemic control of exenatide (synthetic exendin‐4) additive to existing metformin and/or sulfonylurea treatment in patients with type 2 diabetes. Diabetes Care 2003; 26: 2370–7
  • Fineman M. S., Shen L. Z., Taylor K., Kim D. D., Baron A. D. Effectiveness of progressive dose‐escalation of exenatide (exendin‐4) in reducing dose‐limiting side effects in subjects with type 2 diabetes. Diabetes Metab Res Rev 2004; 20: 411–7
  • Buse J. B., Henry R. R., Han J., Kim D. D., Fineman M. S., Baron AD; Exenatide‐113 Clinical Study Group. Effects of exenatide (exendin‐4) on glycemic control over 30 weeks in sulfonylurea‐treated patients with type 2 diabetes. Diabetes Care 2004; 27: 2628–35
  • Knudsen L. B., Nielsen P. F., Huusfeldt P. O., Johansen N. L., Madsen K., Pedersen F. Z., et al. Potent derivatives of glucagon‐like peptide‐1 with pharmacokinetic properties suitable for once daily administration. J Med Chem 2000; 43: 1664–9
  • Larsen P. J., Fledelius C., Knudsen L. B., Tang‐Christensen M. Systemic administration of the long‐acting GLP‐1 derivative NN2211 induces lasting and reversible weight loss in both normal and obese rats. Diabetes 2001; 50: 2530–9
  • Sturis J., Gotfredsen C. F., Romer J., Rolin B., Ribel U., Brand C. L., et al. GLP‐1 derivative liraglutide in rats with beta‐cell deficiencies: influence of metabolic state on beta‐cell mass dynamics. Br J Pharmacol 2003; 140: 123–32
  • Bock T., Pakkenberg B., Buschard K. The endocrine pancreas in non‐diabetic rats after short‐term and long‐term treatment with the long‐acting GLP‐1 derivative NN2211. APMIS 2003; 111: 1117–24
  • Rolin B., Larsen M. O., Gotfredsen C. F., Deacon C. F., Carr R. D., Wilken M., et al. The long‐acting GLP‐1 derivative NN2211 ameliorates glycemia and increases beta‐cell mass in diabetic mice. Am J Physiol Endocrinol Metab 2002; 283: E745–52
  • Ribel U., Larsen M. O., Rolin B., Carr R. D., Wilken M., Sturis J., et al. NN2211: a long‐acting glucagon‐like peptide‐1 derivative with anti‐diabetic effects in glucose‐intolerant pigs. Eur J Pharmacol 2002; 451: 217–25
  • Elbrond B., Jakobsen G., Larsen S., Agerso H., Jensen L. B., Rolan P., et al. Pharmacokinetics, pharmacodynamics, safety, and tolerability of a single‐dose of NN2211, a long‐acting glucagon‐like peptide 1 derivative, in healthy male subjects. Diabetes Care 2002; 25: 1398–404
  • Juhl C. B., Hollingdal M., Sturis J., Jakobsen G., Agerso H., Veldhuis J., et al. Bedtime administration of NN2211, a long‐acting GLP‐1 derivative, substantially reduces fasting and postprandial glycemia in type 2 diabetes. Diabetes 2002; 51: 424–9
  • Degn K. B., Juhl C. B., Sturis J., Jakobsen G., Brock B., Chandramouli V., 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: 1187–94
  • Edwards C. M., Bloom S. R. The incretins‐outdated terminology in man?. Diabetologia 1999; 42: 1148–9
  • Harder H., Nielsen L., Tu D. T., Astrup A. The effect of liraglutide, a long‐acting glucagon‐like peptide 1 derivative, on glycemic control, body composition, and 24‐h energy expenditure in patients with type 2 diabetes. Diabetes Care 2004; 27: 1915–21
  • Madsbad S., Schmitz O., Ranstam J., Jakobsen G., Matthews D. R., NN2211‐1310 International Study Group. Improved glycemic control with no weight increase in patients with type 2 diabetes after once‐daily treatment with the long‐acting glucagon‐like peptide 1 analog liraglutide (NN2211): a 12‐week, double‐blind, randomized, controlled trial. Diabetes Care 2004; 27: 1335–42
  • UK Prospective Diabetes Study (UKPDS) Group. Effect of intensive blood‐glucose control with metformin on complications in overweight patients with type 2 diabetes (UKPDS 34). Lancet 1998; 352: 854–65
  • Green B. D., Gault V. A., O'harte F. P., Flatt P. R. Structurally modified analogues of glucagon‐like peptide‐1 (GLP‐1) and glucose‐dependent insulinotropic polypeptide (GIP) as future antidiabetic agents. Curr Pharm Des 2004; 10: 3651–62
  • Kim J. G., Baggio L. L., Bridon D. P., Castaigne J. P., Robitaille M. F., Jette L., et al. Development and characterization of a glucagon‐like peptide 1‐albumin conjugate: the ability to activate the glucagon‐like peptide 1 receptor in vivo. Diabetes 2003; 52: 751–9
  • Baggio L. L., Huang Q., Brown T. J., Drucker D. J. A recombinant human glucagon‐like peptide (GLP)‐1‐albumin protein (albugon) mimics peptidergic activation of GLP‐1 receptor‐dependent pathways coupled with satiety, gastrointestinal motility, and glucose homeostasis. Diabetes 2004; 53: 2492–500
  • Marguet D., Baggio L., Kobayashi T., Bernard A. M., Pierres M., Nielsen P. F., et al. Enhanced insulin secretion and improved glucose tolerance in mice lacking CD26. Proc Natl Acad Sci U S A 2000; 97: 6874–9
  • Conarello S. L., Li Z., Ronan J., Roy R. S., Zhu L., Jiang G., et al. Mice lacking dipeptidyl peptidase IV are protected against obesity and insulin resistance. Proc Natl Acad Sci U S A 2003; 100: 6825–30
  • Mentlein R., Gallwitz B., Schmidt W. E. Dipeptidyl‐peptidase IV hydrolyses gastric inhibitory polypeptide, glucagon‐like peptide‐1(7‐36)amide, peptide histidine methionine and is responsible for their degradation in human serum. Eur J Biochem 1993; 214: 829–35
  • Hansotia T., Baggio L. L., Delmeire D., Hinke S. A., Yamada Y., Tsukiyama K., et al. Double incretin receptor knockout (DIRKO) mice reveal an essential role for the enteroinsular axis in transducing the glucoregulatory actions of DPP‐IV inhibitors. Diabetes 2004; 53: 1326–35
  • Bauvois B., Sanceau J., Wietzerbin J. Human U937 cell surface peptidase activities: characterization and degradative effect on tumor necrosis factor‐alpha. Eur J Immunol 1992; 22: 923–30
  • Maes M., De Meester I., Scharpe S., Desnyder R., Ranjan R., Meltzer H. Y. Alterations in plasma dipeptidyl peptidase IV enzyme activity in depression and schizophrenia: effects of antidepressants and antipsychotic drugs. Acta Psychiatr Scand 1996; 93: 1–8
  • Darmoul D., Lacasa M., Chantret I., Swallow D. M., Trugnan G. Isolation of a cDNA probe for the human intestinal dipeptidylpeptidase IV and assignment of the gene locus DPP4 to chromosome 2. Ann Hum Genet 1990; 54: 191–7
  • Hildebrandt M., Reutter W., Arck P., Rose M., Klapp B. F. A guardian angel: the involvement of dipeptidyl peptidase IV in psychoneuroendocrine function, nutrition and immune defence. Clin Sci 2000; 99: 93–104
  • Reimer M. K., Holst J. J., Ahren B. Long‐term inhibition of dipeptidyl peptidase IV improves glucose tolerance and preserves islet function in mice. Eur J Endocrinol 2002; 146: 717–27
  • Sudre B., Broqua P., White R. B., Ashworth D., Evans D. M., Haigh R., et al. Chronic inhibition of circulating dipeptidyl peptidase IV by FE 999011 delays the occurrence of diabetes in male zucker diabetic fatty rats. Diabetes 2002; 51: 1461–9
  • Pospisilik J. A., Stafford S. G., Demuth H. U., Brownsey R., Parkhouse W., Finegood D. T., et al. Long‐term treatment with the dipeptidyl peptidase IV inhibitor P32/98 causes sustained improvements in glucose tolerance, insulin sensitivity, hyperinsulinemia, and beta‐cell glucose responsiveness in VDF (fa/fa) Zucker rats. Diabetes 2002; 51: 943–50
  • Ahren B., Simonsson E., Larsson H., Landin‐Olsson M., Torgeirsson H., Jansson P. A., et al. Inhibition of dipeptidyl peptidase IV improves metabolic control over a 4‐week study period in type 2 diabetes. Diabetes Care 2002; 25: 869–75
  • Ahren B., Landin‐Olsson M., Jansson P. A., Svensson M., Holmes D., Schweizer A. Inhibition of dipeptidyl peptidase‐4 reduces glycemia, sustains insulin levels, and reduces glucagon levels in type 2 diabetes. J Clin Endocrinol Metab 2004; 89: 2078–84
  • Ahren B., Gomis R., Standl E., Mills D., Schweizer A. Twelve‐ and 52‐week efficacy of the dipeptidyl peptidase IV inhibitor LAF237 in metformin‐treated patients with type 2 diabetes. Diabetes Care 2004; 27: 2874–80

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.