Skip to main content
Log in

Miglitol

A Review of its Therapeutic Potential in Type 2 Diabetes Mellitus

  • Adis Drug Evaluation
  • Published:
Drugs Aims and scope Submit manuscript

Summary

Abstract

Miglitol, the first pseudomonosaccharide α-glucosidase inhibitor, smooths postprandial peak plasma glucose levels and thus improves glycaemic control, which is reflected in a reduced glycosylated haemoglobin (HbA1c) level. This oral antihyperglycaemic agent is indicated for the treatment of patients with type 2 diabetes mellitus. Miglitol is generally well tolerated and, unlike the sulphonylurea agents, is not associated with bodyweight gain or hypoglycaemia when administered as monotherapy. The drug is systemically absorbed but is not metabolised and is rapidly excreted via the kidneys.

Clinical trials with miglitol (usually 50 or 100mg 3 times daily) in patients with type 2 diabetes mellitus consistently demonstrated a significant improvement in glycaemic control for periods of 6 to 12 months. There were also marked reductions in postprandial serum insulin levels, although miglitol generally had no effect on fasting insulin levels. In comparative studies miglitol had similar efficacy to acarbose, but at lower therapeutic doses (50 and 100mg 3 times daily, respectively). In addition, although sulphonylurea agents provided superior reductions in HbA1c levels, miglitol provided similar or superior reductions in fasting and postprandial plasma glucose levels. In combination with other oral antidiabetic agents or insulin, miglitol improved glycaemic control in patients in whom metabolic control was suboptimal despite dietary and pharmacological intervention.

Most adverse events associated with miglitol treatment involve disturbances of the gastrointestinal tract (most common effects are flatulence, abdominal pain and diarrhoea). These symptoms are usually dose dependent, mild to moderate in severity, occur at the onset of treatment, decline with time and resolve promptly on discontinuation of the drug or with dosage adjustment. As monotherapy, miglitol is not associated with hypoglycaemia, but concomitant use with other oral antidiabetic agents may necessitate dosage adjustment of the other agents. Miglitol had no significant effects on renal, cardiovascular, respiratory or haematological parameters in long term studies. No dosage adjustments are required in elderly patients, in those with hepatic impairment or in those with mild to moderate renal insufficiency.

Conclusions: In long term, well designed trials miglitol reduces fasting and postprandial plasma glucose levels, thus improving glycaemic control, which is reflected in a reduced HbA1c level in patients with type 2 diabetes mellitus. Most adverse events associated with miglitol involve disturbances of the gastrointestinal tract. This agent is a useful first-line therapy in patients with type 2 diabetes mellitus insufficiently controlled by diet alone and as second-line or as adjuvant therapy in those insufficiently controlled with diet and sulphonylurea agents. Miglitol may prove particularly beneficial in elderly patients and those with hepatic impairment or mild to moderate renal impairment, in whom other oral antidiabetic agents are contraindicated or need to be used with caution.

Pharmacodynamic Properties

Miglitol is a 1-deoxynojirimycin derivative which reversibly inhibits intestinal α-glucosidase enzymes responsible for the digestion of carbohydrates to absorbable monosaccharides. Its structure resembles that of glucose, and unlike acarbose (a pseudotetrasaccharide α-glucosidase inhibitor), it is almost completely absorbed in the upper section of the small intestine. The rank order of its inhibitory activity is sucrase > glucoamylase > isomaltase > lactase > trehalase. Although miglitol delayed carbohydrate absorption in healthy volunteers, there were no significant losses of carbohydrates, protein or fat in the faeces and no significant caloric losses.

Miglitol smooths postprandial glycaemic peaks thereby reducing postprandial peak plasma glucose levels in patients with type 2 diabetes mellitus (average decrease with multiple 50mg doses was 1.8 to 3.0 mmol/L). Short term studies indicated that miglitol had no effect on postprandial serum insulin levels or C-peptide concentrations; however, after 8 weeks’ treatment with miglitol there was a significant reduction in both of these parameters, which was confirmed in long term clinical trials. A few studies also indicate that miglitol reduces fasting triglyceride levels in patients with type 2 diabetes mellitus following 8 weeks of treatment.

Since miglitol is almost completely absorbed, it has been suggested that it may exert extraintestinal effects on glucose homeostasis. Its effect on enhanced glucose uptake following an oral glucose load is unclear and requires confirmation. Miglitol significantly reduced the postprandial increase in gastric inhibitory polypeptide in healthy volunteers and patients with type 2 diabetes mellitus. It also increased peptide tyrosine-tyrosine (PYY) and motilin levels compared with placebo in patients with type 2 diabetes. Animal studies have shown that miglitol (at recommended therapeutic doses) has no effect on lysosomal α-glucosidases and there was no evidence of glycogen storage or changes in liver weight. Miglitol, unlike several other compounds with cationic polarity, e.g. biguanides, had no effect on sodium-dependent small intestine transport of organic solutes, such as, hexoses.

Pharmacokinetic Properties

Miglitol 25 to 200mg exhibits nonlinear absorption kinetics. The mean peak plasma concentrations (Cmax) following single oral doses of miglitol 25, 50, 100 and 200mg were 0.78, 1.22, 1.86 and 2.61 mg/L, respectively, and were attained within 2 to 4 hours.

Miglitol was rapidly and almost completely absorbed after an oral dose of 25mg, but with doses ≥50mg (therapeutic dose range) a saturation of absorption became evident. The maximum absorbable dose was 2.4 mg/kg (i.e. 200mg). In animal studies, absorption of miglitol occurred predominantly in the upper small intestine. The volume of distribution at steady state (Vss) was low (0.28 L/kg) in healthy volunteers. Furthermore, there was virtually no binding of miglitol to plasma proteins over a range of 10 μg/L to 10 mg/L as determined in in vitro assays.

Excretion of miglitol occurred very rapidly after both oral and intravenous administration and no metabolism of miglitol was detected. In healthy volunteers, ≈59% of oral miglitol was excreted via the kidneys, with 29% being excreted in the faeces. Miglitol showed a biphasic pattern of elimination; a first phase of 1.8 hours during which >90% of drug was eliminated and a second phase of 2 to 4 hours. The total plasma clearance of radiolabelled drug was 0.103 kg/L · h, which is similar to the glomerular filtration rate. In healthy volunteers, dose-dependent increases in the terminal half-life of the drug indicate that it may accumulate following multiple doses.

Studies in healthy volunteers indicate that miglitol 100mg significantly decreases the bioavailability of glibenclamide, but has no clinically relevant effects on the pharmacokinetics of either (R)- or (S)-warfarin or phenytoin. According to the manufacturer’s prescribing information, miglitol reduces the bioavailability of metformin during concomitant administration but the clinical effects of these agents are synergistic. However, the clinical relevance of this is still to be investigated. Further, concomitant administration of miglitol with propranolol or ranitidine reduced the absorption of these agents, and therefore the dose adjustment of propranolol or ranitidine may be necessary.

Clinical Efficacy

Large, well designed trials of 6 to 12 months’ duration in a total of 1783 patients with type 2 diabetes mellitus insufficiently controlled by diet alone or diet plus sulphonylurea agents have demonstrated that miglitol monotherapy 50 to 100mg 3 times daily (therapeutic range) significantly improves glycaemic control and decreases postprandial serum insulin levels. With miglitol 150 to 300 mg/day, the mean absolute reduction in glycosylated haemoglobin (HbA1c) level was 0.18 to 0.75%, which was associated with a 7.3 to 21.1% reduction in postprandial plasma glucose (PPG) levels (i.e. a mean decrease of 0.86 to 3.21 mmol/L) and a 6.8 to 12.5% reduction in fasting plasma glucose (FPG) levels (i.e. a mean decrease of 0.56 to 1.08 mmol/L).

Miglitol had similar efficacy in both younger adults (30 to <70 years of age) and older adults (>60 years) and in different ethnic groups. Efficacy was maintained for the duration of these studies (up to 1 year). Most of these studies reported a group of patients who withdrew because of a lack of efficacy (poor glycaemic control); nonresponder rates between each treatment group of individual studies were generally similar.

Miglitol provided similar improvements in glycaemic control to those observed with other oral antihyperglycaemic agents. Thus, miglitol 50 or 100mg 3 times daily was at least as effective as acarbose 100mg 3 times daily in improving glycaemic control. In elderly patients (aged >60 years), although there was a more marked reduction in HbA1c with glibenclamide (p < 0.05 vs miglitol), miglitol 50mg 3 times daily provided a superior reduction in PPG level (p < 0.05). Furthermore, there was a significantly higher incidence of hypoglycaemic episodes with glibenclamide treatment (46%) than with miglitol (9%) or placebo (8%).

Approximately 30% of patients responded to treatment (based on changes in HbA1c levels) with miglitol 50mg 3 times daily compared with ≈50% of those in the glibenclamide group and 18% in the placebo group. In addition, after 6 months’ treatment, fewer than 15% of patients receiving miglitol 100mg 3 times daily were in poor control (HbA1c >8%) compared with 14 and 48% in the glibenclamide and placebo groups, respectively.

Miglitol significantly reduced postprandial serum insulin levels. The reduction in postprandial serum insulin levels was superior to that of glibenclamide (p < 0.05). However, miglitol generally had no effect on fasting serum insulin levels. There was also a trend to a decrease in body weight with miglitol treatment during the trial period, although this decrease never reached statistically significant levels. In general, unlike acarbose, miglitol had no significant effects on either fasting or postprandial lipid parameters in monotherapy studies of 6 months’ to 1 year’s duration.

The concomitant use of miglitol with other oral antihyperglycaemic agents or insulin has been investigated in several large, well designed trials in patients with type 2 diabetes mellitus in whom glycaemic control was suboptimal despite dietary and pharmacological intervention. Miglitol provided significant improvements in HbA1c, FPG and PPG levels, irrespective of the concomitant antidiabetic agent used. With 150 and 300 mg/day dosages, there was a significant reduction in HbA1c (mean absolute reduction in HbA1c level 0.20 to 1.6%), PPG levels (mean reduction 10.1 to 26.1%) and FPG levels (0 to 14% reduction), although the reduction in FPG levels failed to reach statistical significance in some studies.

Responder rates in patients receiving concomitant miglitol and sulphonylurea therapy were 27 to 39% compared with 5 to 10% in placebo groups. Improved glycaemic control also appeared more marked during the first 6 months of a 1-year study than during the second 6 months; the placebo-subtracted mean change from baseline in HbA1c levels for these 2 periods was −1.19 and −0.74%, respectively. Similar effects have been reported previously with sulphonylurea and biguanide agents in the United Kingdom Prospective Diabetes Study (UKPDS) and may be a reflection of a progressive loss of pancreatic β cell function associated with the natural progression of this disease. Concomitant use of miglitol in patients using insulin therapy may also reduce the insulin dosage required, although further studies are warranted to confirm this finding.

When used concomitantly with sulphonylurea agents, effects of miglitol on fasting and postprandial serum insulin levels were similar to those observed with miglitol in monotherapy studies, with a significant reduction in postprandial levels (10.1 to 20.5% reduction) but no effect on fasting levels. However, there was no effect on postprandial serum insulin level when used concomitantly with exogenous insulin.

Concomitant therapy with miglitol and sulphonylurea agents also reduced fasting serum triglyceride levels significantly compared with placebo. As with monotherapy trials, adjuvant treatment with miglitol had no significant effects on fasting high density lipoprotein, low density lipoprotein or total cholesterol levels.

Tolerability

The most common adverse events in miglitol-treated patients involve the gastrointestinal system and include flatulence, abdominal pain and diarrhoea. Symptoms were usually mild to moderate in intensity, dose dependent, occurred at the onset of treatment, declined with time and resolved promptly on discontinuation of the drug or with dosage adjustment. Compared with placebo, miglitol showed no significant effects on renal, cardiovascular, respiratory or haematological functions in long term clinical studies (6 months to 1 year). There were no significant differences in the withdrawal rate judged by investigators to be attributable to adverse events between individual treatment groups in clinical studies.

Dosage and Administration

In Europe, the recommended initial dosage of miglitol is 50mg 3 times daily, to be taken orally immediately before each main meal. This should be increased to the recommended maintenance dosage of 100mg 3 times daily after 4 to 12 weeks’ treatment. In the US, the maximum recommended daily dosage is 100mg 3 times daily, with treatment initiated at a dosage of 25mg 3 times daily and gradually increased. The recommended maintenance dose in the US is 50mg 3 times daily. No dosage adjustments are required in elderly patients, in those with hepatic impairment or those with mild to moderate renal insufficiency (creatinine clearance >1.5 L/h).

Miglitol is not indicated for use in patients with diabetic ketoacidosis and is contraindicated in patients with inflammatory bowel disease, colonic ulceration or partial intestinal obstruction, in patients predisposed to intestinal obstruction, in individuals under the age of 18 years and in pregnant or lactating women. Miglitol monotherapy does not induce hypoglycaemia, but may potentiate the hypoglycaemic effects of other antidiabetic drugs, and thus the dosage of these agents may need to be adjusted accordingly. Episodes of hypoglycaemia should be treated with glucose. Miglitol should not be used concomitantly with laxatives, intestinal absorbants or digestive enzyme preparations containing carbohydrate-splitting enzymes. Coadministration of miglitol with propranolol and ranitidine reduces their absorption and therefore dose adjustment of these agents may be necessary.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Institutional subscriptions

Fig. 1
Table I
Table II
Fig. 2
Table III
Table IV
Table V
Fig. 3
Table VI
Fig. 4
Fig. 5

Similar content being viewed by others

References

  1. Zimmerman BR, Espenshade J, Fujimoto WY, et al.The pharmacological treatment of hyperglycemia in NIDDM. Diabetes Care 1996 Jan; 19 Suppl. 1: 54–61

    Google Scholar 

  2. Mandrup-Poulsen T. Diabetes. BMJ 1998 Apr 18; 316: 1221–5

    Article  PubMed  CAS  Google Scholar 

  3. Williams G. Management of non-insulin-dependent diabetes mellitus. Lancet 1994; 343(8889): 95–100

    Article  PubMed  CAS  Google Scholar 

  4. Gaster B, Hirsch IB. The effects of improved glycemic control on complications in type 2 diabetes. Arch Intern Med 1998 Jan 26; 158: 134–40

    Article  PubMed  CAS  Google Scholar 

  5. Scheen AT, Lefèbvre PJ. Antihyperglycaemic agents: drug interactions of clinical importance. Drug Saf 1995 Jan; 12: 32–45

    Article  PubMed  CAS  Google Scholar 

  6. Mahler RJ, Adler ML. Type 2 diabetes mellitus: update on diagnosis, pathophysiology, and treatment. J Clin Endocrinol Metab 1999 Apr; 84: 1165–71

    Article  PubMed  CAS  Google Scholar 

  7. Johnston PS, Feig PU, Coniff RF, et al. Chronic treatment of African-American type 2 diabetic patients with α-glucosidase inhibition. Diabetes Care 1998 Mar; 21: 416–22

    Article  PubMed  CAS  Google Scholar 

  8. Henry RR. Glucose control and insulin resistance in non-insulin-dependent diabetes mellitus. Ann Intern Med 1996 Jan 1; 124 (Part 2): 97–103

    PubMed  CAS  Google Scholar 

  9. Pickup JC, Crook MA. Is type II diabetes mellitus a disease of the innate immune system? Diabetologia 1998 Oct; 41: 1241–8

    Article  PubMed  CAS  Google Scholar 

  10. Hollander PA. New oral agents for type II diabetes: taking a more aggressive approach to therapy. Postgrad Med 1995 Dec; 98: 110–26

    PubMed  CAS  Google Scholar 

  11. UK Prospective Diabetes Study 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

    Article  Google Scholar 

  12. Diabetes Control and Complications Trial Research Group. The effect of intensive treatment of diabetes on the development and progression of long-term complications in insulin-dependent diabetes mellitus. N Engl J Med 1993; 329: 977–86

    Article  Google Scholar 

  13. Monnier L. The role of blood-glucose lowering agents in the light of the UKPDS. Diabetes Obesity Metab 1999; 1 Suppl. 2: 14–23

    Article  Google Scholar 

  14. Baron AD. Postprandial hyperglycaemia and α-glucosidase inhibitors. Diabetes Res Clin Pract 1998; 40 Suppl.: 51–5

    Article  Google Scholar 

  15. Standl E. Cardiovascular risk in type 2 diabetes. Diabetes Obesity Metab 1999; 1 Suppl. 2: 24–36

    Article  Google Scholar 

  16. Mooradian AD, Thurman JE. Drug therapy of postprandial hyperglycaemia. Drugs 1999 Jan; 57: 19–29

    Article  PubMed  CAS  Google Scholar 

  17. Lefebvre PJ, Scheen AJ. The postprandial state and risk of cardiovascular disease. Diabet Med 1998; 15 Suppl. 4: 63–8

    Article  Google Scholar 

  18. Haffner SM. The importance of hyperglycaemia in the nonfasting state to the development of cardiovascular disease. Endocr Rev 1998; 19: 583–92

    Article  PubMed  CAS  Google Scholar 

  19. Savage PJ. Cardiovascular complications of diabetes mellitus: what we know and what we need to know about their prevention. Ann Intern Med 1996; 124 (Pt 2): 123–6

    PubMed  CAS  Google Scholar 

  20. Colwell JA. Pharmacological strategies to prevent macrovascular disease in NIDDM. Diabetes 1997 Sep; 46 Suppl. 2: 131–4

    Google Scholar 

  21. Wagenknecht LE, Mayer EJ, Rewers M, et al. The Insulin Resistance Atherosclerosis Study (IRAS): objectives, design, and recruitment results. Ann Epidemiol 1995 Nov; 5: 464–72

    Article  PubMed  CAS  Google Scholar 

  22. Pyörälä K, Steiner G. Will correction of dyslipoproteinaemia reduce coronary heart disease risk in patients with non-insulin-dependent diabetes ? Need for trial evidence. Ann Med 1996 Aug; 28: 357–62

    Article  PubMed  Google Scholar 

  23. Fontbonne A, Eschwège E, Cambien F, et al. Hypertriglyceridaemia as a risk factor of coronary heart disease mortality in subjects with impaired glucose tolerance or diabetes. Diabetologia 1989; 32: 300–4

    Article  PubMed  CAS  Google Scholar 

  24. Colwell JA. Intensive insulin therapy in type II diabetes: rationale and collaborative clinical trial results. Diabetes 1996 Jul; 45 Suppl. 3: 87–90

    Google Scholar 

  25. Bischoff H. Pharmacology of α-glucosidase inhibition. Eur J Clin Invest Suppl. 1994; 24(3): 3–10

    CAS  Google Scholar 

  26. Lefebvre PJ, Scheen AJ. Management of non-insulin-dependent diabetes mellitus. Drugs 1992; 44 Suppl. 3: 29–38

    Article  PubMed  Google Scholar 

  27. Balfour JA, Clissold SP. Management of non-insulin-dependent diabetes mellitus: defining the role of metformin. Dis Manage Health Outcomes 1997 Jan; 1: 49–59

    Article  Google Scholar 

  28. Salvatore T, Giugliano D. Pharmacokinetic-pharmacodynamic relationships of acarbose. Clin Pharmacokinet 1996 Feb; 30: 94–106

    Article  PubMed  CAS  Google Scholar 

  29. Feld S. American Association of Clinical Endocrinologists Diabetes Guidelines. Endocr Pract 1999; 6(1): 3–27

    Google Scholar 

  30. Clissold SP, Edwards C. Acarbose: a preliminary review of its pharmacodynamic and pharmacokinetic properties, and therapeutic potential. Drugs 1988; 35: 214–43

    Article  PubMed  CAS  Google Scholar 

  31. Balfour JA, McTavish D. Acarbose: an update of its pharmacology and therapeutic use in diabetes mellitus. Drugs 1993 Dec; 46: 1025–54

    Article  PubMed  CAS  Google Scholar 

  32. Donckier J, Williams G. The roles of α-glucosidase inhibitors in diabetes. Eur J Clin Invest 1994 Aug; 24: 522–4

    Article  PubMed  CAS  Google Scholar 

  33. Lembcke B, Folsch UR, Creutzfeldt W. Effect of 1-desoxynojirimycin derivatives on small intestinal disaccharidase activities and on active transport in vitro. Digestion 1985; 31: 120–7

    Article  PubMed  CAS  Google Scholar 

  34. Samulitis BK, Goda T, Lee SM, et al. Inhibitory mechanism of acarbose and 1-deoxynojirimycin derivatives on carbohydrases in rat small intestine. Drugs Exp Clin Res 1987; 13: 517–24

    PubMed  CAS  Google Scholar 

  35. Lembcke B, editor. Therapy principle of absorption delay: the effect principle of α-glucosidase inhibition. Berlin: Springer-Verlag, 1991

    Google Scholar 

  36. Wisselaar HA, van-Dongen JM, Reuser AJ. Effects of N-hydroxyethyl-1-deoxynojirimycin (BAY m 1099) on the activity of neutral- and acid α-glucosidases in human fibroblasts and HepG2 cells. Clin Chim Acta 1989 Jun 15; 182: 41–52

    Article  PubMed  CAS  Google Scholar 

  37. Reuser AJJ, Wisselaar HA. An evaluation of the potential side-effects of α-glucosidase inhibitors used for the management of diabetes mellitus. Eur J Clin Invest 1994 Aug; 24 Suppl. 3: 19–24

    Article  PubMed  CAS  Google Scholar 

  38. Ludolph D, Gross V, Katz NR, et al. Effect of the α-glucosidase inhibitor N-hydroxyethyl-1-deoxynojirimycin (Bay m 1099) on the biosynthesis of liver secretory glycoproteins. Biochem Pharmacol 1989 Aug 1; 38: 2479–86

    Article  PubMed  CAS  Google Scholar 

  39. Ladas SD, Frydas A, Papadopoulos A, et al. Effects of α-glucosidase inhibitors on mouth to caecum transit time in humans. Gut 1992 Sep; 33: 1246–8

    Article  PubMed  CAS  Google Scholar 

  40. Taylor RH, Barker HM, Bowey EA, et al. Regulation of the absorption of dietary carbohydrate in man by two new glycosidase inhibitors. Gut 1986 Dec; 27: 1471–8

    Article  PubMed  CAS  Google Scholar 

  41. Holt PR, Thea D, Yang M-Y, et al. Intestinal and metabolic responses to an α-glucosidase inhibitor in normal volunteers. Metabolism 1988 Dec; 37: 1163–70

    Article  PubMed  CAS  Google Scholar 

  42. Heinz G, Komjati M, Korn A, et al. Reduction of postprandial blood glucose by the α-glucosidase inhibitor Miglitol (BAY m 1099) in type II diabetes. Eur J Clin Pharmacol 1989; 37: 33–6

    PubMed  CAS  Google Scholar 

  43. Joubert PH, Bam WJ, Manyane N. Effect of an α-glucosidase inhibitor (BAY m 1099) on post-prandial blood glucose and insulin in type II diabetics. Eur J Clin Pharmacol 1986; 30: 253–5

    Article  PubMed  CAS  Google Scholar 

  44. Katsilambros N, Philippides P, Toskas A, et al. A double-blind study on the efficacy and tolerance of a new α-glucosidase inhibitor in type-2 diabetics. Arzneimittelforschung 1986 Jul; 36: 1136–8

    PubMed  CAS  Google Scholar 

  45. Kingma PJ, Menheere PPCA, Sels JP, et al. α-Glucosidase inhibition by miglitol in NIDDM patients. Diabetes Care 1992 Apr; 15: 478–83

    Article  PubMed  CAS  Google Scholar 

  46. Scott AR, Tattersall RB. α-Glucosidase inhibition in the treatment of non-insulin-dependent diabetes mellitus. Diabetic Med 1988 Jan; 5: 42–6

    Article  PubMed  CAS  Google Scholar 

  47. Samad AH, Ty-Willing TS, Alberti KGMM, et al. Effects of BAY m 1099, new α-glucosidase inhibitor, on acute metabolic responses and metabolic control in NIDDM over 1 mo. Diabetes Care 1988 Apr; 11: 337–44

    Article  PubMed  CAS  Google Scholar 

  48. Schnack C, Piager RJF, Winkler J, et al. Effects of 8-wk α-glucosidase inhibition on metabolic control, C-peptide secretion, hepatic glucose output, and peripheral insulin sensitivity in poorly controlled type II diabetic patients. Diabetes Care 1989 Sep; 12: 537–43

    Article  PubMed  CAS  Google Scholar 

  49. Johnson AB, Taylor R. Does suppression of postprandial blood glucose excursions by the α-glucosidase inhibitor miglitol improve insulin sensitivity in diet-treated type II diabetic patients? Diabetes Care 1996 Jun; 19: 559–63

    Article  PubMed  CAS  Google Scholar 

  50. Joubert PH, Foukaridis GN, Bopape ML. Miglitol may have a blood glucose lowering effect unrelated to inhibition of α-glucosidase. Eur J Clin Pharmacol 1987; 31: 723–4

    Article  PubMed  CAS  Google Scholar 

  51. Joubert PH, Venter HL, Foukaridis GN. The effect of miglitol and acarbose after an oral glucose load: a novel hypoglycaemic mechanism? Br J Clin Pharmacol 1990 Sep; 30: 391–6

    Article  PubMed  CAS  Google Scholar 

  52. Sels JPJE, Nauta JJP, Menheere PPCA, et al. Miglitol (Bay m 1099) has no extraintestinal effects on glucose control in healthy volunteers. Br J Clin Pharmacol 1996 Oct; 42: 503–6

    Article  PubMed  CAS  Google Scholar 

  53. Lembcke B, Fölsch UR, Gatzemeier W, et al. Inhibition of glycemic and hormonal responses after repetitive sucrose and starch loads by different doses of the α-glucosidase inhibitor miglitol (BAY m 1099) in man. Pharmacology 1991 Dec; 43: 318–28

    Article  PubMed  CAS  Google Scholar 

  54. Lembcke B, Diederich M, Fölsch UR, et al. Postprandial glycemic control, hormonal effects and carbohydrate malabsorption during long-term administration of the α-glucosidase inhibitor miglitol. Digestion 1990 Sep; 47: 47–55

    Article  PubMed  CAS  Google Scholar 

  55. Requejo F, Uttenthal LO, Bloom SR. Effects of α-glucosidase inhibition and viscous fibre on diabetic control and postprandial gut hormone responses. Diabetic Med 1990 Jul;7: 515–20

    Article  PubMed  CAS  Google Scholar 

  56. Lembcke B, Lamberts R, Wöhler J, et al. Lysosomal storage of glycogen as a sequel of α-glucosidase inhibition by the absorbed deoxynojirimycin derivative emiglitate (BAY o 1248). A drug-induced pattern of hepatic glycogen storage mimicking Pompe’s disease (glycogenosis type II) [in German]. Res Exp Med Berl 1991; 191: 389–404

    Article  PubMed  CAS  Google Scholar 

  57. Hillebrand I, Boehme K, Graefe KH, et al. The effect of new α-glucosidase inhibitors (BAY m 1099 and BAY o 1248) on meal-stimulated increases in glucose and insulin levels in man. Klin Wochenschr 1986 Apr 15; 64: 393–6

    Article  PubMed  CAS  Google Scholar 

  58. Karam JH, Salber PR, Forsham PH. Pancreatic hormones and diabetes mellitus. In: Greenspan FS, editor. Basic and clinical endocrinology. 3rd ed. Morwalk (CT): Apple & Lange, 1991: 592–650

    Google Scholar 

  59. Mathur S, Fonseca V, Dandona P. The effect of the α-glucosidase inhibitor miglitol on insulin sensitivity in non-insulin dependent diabetics [abstract]. Diabetes 1991 May; 40 Suppl. 1: 336

    Google Scholar 

  60. Arends J, Willms BH. Smoothing effect of a new α-glucosidase inhibitor BAY m 1099 on blood glucose profiles of sulfonylurea-treated type II diabetic patients. Horm Metab Res 1986 Nov; 18: 761–4

    Article  PubMed  CAS  Google Scholar 

  61. Ahr H-J, Boberg M, Brendel E, et al. Pharmacokinetics of miglitol: absorption, distribution, metabolism, and excretion following administration to rats, dogs, and man. Arzneimittelforschung 1997 Jun; 47: 734–45

    PubMed  CAS  Google Scholar 

  62. Beneke PC, Müller FO, VanDyk M, et al. The pharmacokinetics of miglitol over a therapeutic dose range [abstract]. S Afr Med J 1991 Dec 7; 80: 622

    Google Scholar 

  63. Sanofi-Synthelabo. Diastabol: product monograph. Paris, France

  64. Johnston PS, Coniff RF, Hoogwerf BJ, et al. Effects of the carbohydrase inhibitor miglitol in sulfonylurea-treated NIDDM patients. Diabetes Care 1994 Jan; 17: 20–9

    Article  PubMed  CAS  Google Scholar 

  65. Segal P, Feig PU, Schernthaner G, et al. The efficacy and safety of miglitol therapy compared with glibenclamide in patients with NIDDM inadequately controlled by diet alone. Diabetes Care 1997 May; 20: 687–91

    Article  PubMed  CAS  Google Scholar 

  66. Bayer Vital GmbH and Company K.G. Multicentre, multinational, double-blind, placebo-controlled clinical study to investigate the efficacy and tolerability of miglitol (Bay m 1099) in type 2 diabetes unsatisfactorily treated with sulphonylurea and metformin. Bayer Vital GmbH and Company K.G. (Germany), 1997. Report no.: PH 26828 (Data on file)

  67. Bayer Vital GmbH and Company K.G. A multicentre, double-blind, placebo-controlled study, investigating the efficacy and safety of miglitol in combination with metformin as compared to metformin monotherapy in the treatment of patients with non-insulin-dependent diabetes mellitus (type II) inadequately controlled with diet alone. Bayer Vital GmbH and Company K.G (Germany), 1997. Report no.:MMRR-1439 (Data on file)

  68. Bayer Corporation. International multicentre, double-blind, placebo-controlled clinical study to investigate the efficacy and tolerability of miglitol (Bay m 1099) in type 2 diabetics unsatisfactorily treated with metformin. Bayer Corporation (Germany), 1998. Report no. PH-2827/1 (Data on file)

  69. Salvatore T, Scheen AJ, Ferreira Alves de Magalhaes AC, et al. Slight modifications of the pharmacokinetic parameters of glibenclamide after treatment with the α-glucosidase inhibitor miglitol in normal subjects [abstract]. Therapie 1990 Jul–Aug; 45: 365

    Google Scholar 

  70. Schall R, Müller FO, Hundt HKL, et al. Study of the effect of miglitol on the pharmacokinetics and pharmacodynamics of warfarin in healthy males. Arzneimittelforschung 1996 Jan; 46: 41–6

    PubMed  CAS  Google Scholar 

  71. Richardt D, Rosmarin C, Havlik I, et al. No effect of miglitol on the oral bioavailability of single-dose phenytoin in healthy males. Clin Drug Invest 1997 Mar; 13: 171–4

    Article  CAS  Google Scholar 

  72. Mosby. Miglitol; prescribing information. In: Rabbitt KE, editor. Mosby’s GenRx. 9th ed. Missouri: Mosby, Inc., 1999: 1506–8

    Google Scholar 

  73. Standl E, Schernthaner G, Rybka J, et al. Improved glycaemic control with miglitol in type 2 diabetics insufficiently controlled on oral hypoglycaemics [abstract and poster no.839]. Diabetologia 1999; 42 Suppl. 1: 223

    Google Scholar 

  74. Drent ML. Miglitol as single oral hypoglycemic agent in type 2 diabetes [abstract]. Diabetologia 1994; 37 Suppl. 1: 211

    Google Scholar 

  75. Rybka J, Goke B, Sissmann J. European comparative study of 2 α-glucosidase inhibitors, miglitol and acarbose [abstract]. Diabetes 1999 May; 48 Suppl. 1: 101

    Google Scholar 

  76. Pagano G, Marena S, Corgiat-Mansin L, et al. Comparison of miglitol and glibenclamide in diet-treated type 2 diabetic patients [French]. Diabete Metab 1995 Jun; 21: 162–7

    PubMed  CAS  Google Scholar 

  77. Johnston PS, Lebovitz HE, Coniff RF, et al. Advantages of α-glucosidase inhibition as monotherapy in elderly type 2 diabetic patients. J Clin Endocrinol Metab 1998 May; 83: 1515–22

    Article  PubMed  CAS  Google Scholar 

  78. Johnston PS, Feig PU, Coniff RF, et al. Long-term titrated-dose α-glucosidase inhibition in non-insulin-requiring Hispanic NIDDM patients. Diabetes Care 1998 Mar; 21: 409–15

    Article  PubMed  CAS  Google Scholar 

  79. Coniff RF, Johnston PS, Krol A. Long-term effects of the α-glucosidase inhibitor miglitol (BAY m 1099) in NIDDM patients treated with maximal-dose sulfonylurea [abstract]. Diabetes 1996 May; 45 Suppl. 2: 221

    Google Scholar 

  80. Maislos M, Van Gaal L, Segal P. Miglitol in combination with metformin improves glycaemic control in type 2 diabetics [abstract and poster no. 838]. Diabetologia 1999; 42 Suppl. 1: 223

    Google Scholar 

  81. Mitrakou A, Tountas N, Raptis AE, et al. Long-term effectiveness of a new α-glucosidase inhibitor (BAY m 1099-miglitol) in insulin-treated type 2 diabetes mellitus. Diabetic Med 1998 Aug; 15: 657–60

    Article  PubMed  CAS  Google Scholar 

  82. Jay RH, Round JM, Betteridge DJ. Effects of miglitol on glucose control, intermediary metabolism, lipoproteins and clotting factors in type 1 diabetes [abstract]. Diabetic Med 1992 Jul; 9 Suppl. 1: 51A

    Google Scholar 

  83. Dolben J, Luzio S, Smith H, et al. Miglitol: a new α-glucosidase inhibitor decreases post-prandial glucose excursions [abstract]. Br J Clin Pharmacol 1992 Feb; 33: 225P

    Google Scholar 

  84. Bayer Vital GmbH and Company K.G. Efficacy and tolerability of miglitol (Bay m 1099) and acarbose (Bay g 5421) in type II diabetes. Bayer Vital GmbH and Company K.G. (Germany), 1994. Report no.: R 6245. (Data on file)

  85. United Kingdom Prospective Diabetes Study Group. UK Prospective Diabetes Study 16. Overview of 6 years’ therapy of type II diabetes: a progressive disease. Diabetes 1995; 44: 1249–58

    Article  Google Scholar 

  86. Bayer AG. A multi-centre, double-blind, placebo-controlled group comparsion study to investigate the efficacy, tolerability and safety of miglitol (Bay m 1099) in three different doses in the treatment of patients with non-insulin dependent diadebtes mellitus (NIDDM) inadequately controlled with maximum daily doses of sulphonylurea and diet. Bayer Corporation (Germany), 1995. Report no. 1313 (Data on file)

  87. Evans J, Krentz AJ. Recent developments and emerging therapies for type 2 diabetes mellitus. Drugs Res Dev 1999; 2(2): 75–94

    Article  CAS  Google Scholar 

  88. Scheen AJ. Drug treatment of non-insulin-dependent diabetes mellitus in the 1990s: achievements and future developments. Drugs 1997 Sep; 54: 355–68

    Article  PubMed  CAS  Google Scholar 

  89. Zimmerman BR. Preventing long term complications. Implications for combination therapy with acarbose. Drugs 1992; 44 Suppl. 3: 54–9

    Article  PubMed  Google Scholar 

  90. UK Prospective Diabetes Study 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

    Article  Google Scholar 

  91. Kuusisto J, Mykkänen L, Pyörälä K, et al. NIDDM and its metabolic control predict coronary heart disease in elderly subjects. Diabetes 1994; 43: 960–7

    Article  PubMed  CAS  Google Scholar 

  92. Hanefeld M, Fischer S, Julius U, et al. Risk factors for myocardial infarction and death in newly detected NIDDM: the Diabetes Intervention Study, 11-year follow-up. Diabetologia 1996; 39: 1577–83

    Article  PubMed  CAS  Google Scholar 

  93. Tominaga M, Eguchi H, Manaka H, et al. Impaired glucose tolerance is a risk factor for cardiovascular disease, but not impaired fasting glucose: the Fungata Diabetes Study. Diabetes Care 1999; 22(6): 920–4

    Article  PubMed  CAS  Google Scholar 

  94. Rabasa-Lhoret R, Chiasson JL. Potential of α-glucosidase inhibitors in elderly patients with diabetes mellitus and impaired glucose tolerance. Drugs Aging 1998 Aug; 13: 131–43

    Article  PubMed  CAS  Google Scholar 

  95. Bressler R, Johnson DG. Oral antidiabetic drug use in the elderly. Drugs Aging 1996 Dec; 9: 418–37

    Article  PubMed  CAS  Google Scholar 

  96. Lardinois CK. Type 2 diabetes: glycemic targets and oral therapies for older patients. Geriatrics 1998 Nov; 53(11): 22–3, 27-8, 33-4, 37-9

    PubMed  CAS  Google Scholar 

  97. Bretzel RG. The United Kingdom Prospective Study (UKPDS) implications for the pharmacotherapy of type 2 diabetes mellitus. Exp Clin Endocrinol Diabetes 1998; 106: 369–72

    Article  PubMed  CAS  Google Scholar 

  98. Mooradian AD. Drug therapy of non-insulin-dependent diabetes mellitus in the elderly. Drugs 1996 Jun; 51: 931–41

    Article  PubMed  CAS  Google Scholar 

  99. Lunt H. Diabetes mellitus in older patients: is tight blood glucose control warranted? Drugs Aging 1996 Jun; 8: 401–7

    Article  PubMed  CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Lesley J. Scott.

Additional information

Various sections of the manuscript reviewed by: B. Göke, Department of Internal Medicine, Inselspital Bern, Bern, Switzerland; P.R. Holt, St.Luke’s Roosevelt Centre, New York, USA; G.D. Johnston, Department of Therapeutics and Pharmacology, The Queen’s University of Belfast, Belfast, Northern Ireland; S. Kumar, Department of Medicine, The University of Birmingham, Birmingham, England; B. Lembcke, Department of Internal Medicine, St. Barbara Hospital, Gladbeck, Germany.

Data Selection

Sources: Medical literature published in any language since 1966 on Miglitol, identified using AdisBase (a proprietary database of Adis International, Auckland, New Zealand), Medline and EMBASE. Additional references were identified from the reference lists of published articles. Bibliographical information, including contributory unpublished data, was also requested from the company developing the drug.

Search strategy: AdisBase search terms were ‘Miglitol’ or ‘BAY-1099’ or ‘BAY M-1099’. Medline search terms were ‘Miglitol’ or ‘BAY-1099’ or ‘BAY M-1099’. EMBASE search terms were ‘Miglitol’ or ‘BAY-1099’ or ‘BAY M-1099’. Searches were last updated 23 Feb 2000.

Selection: Studies in patients with type 2 diabetes mellitus who received miglitol. Inclusion of studies was based mainly on the methods section of the trials. When available, large, well controlled trials with appropriate statistical methodology were preferred. Relevant pharmacodynamic and pharmacokinetic data are also included.

Index terms: miglitol, pharmacodynamics, pharmacokinetics, therapeutic use.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Scott, L.J., Spencer, C.M. Miglitol. Drugs 59, 521–549 (2000). https://doi.org/10.2165/00003495-200059030-00012

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.2165/00003495-200059030-00012

Keywords

Navigation